Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Tissue Eng Regen Med ; 6(10): e43-50, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22941779

RESUMEN

Bone autografts are considered the gold standard for cranioplasty, although they lead to co-morbidity. Bone allografts are more easily obtained but have low osteogenic potential and fail to integrate into healthy bone. Previously, we showed that, by coating long-bone allografts with freeze-dried recombinant adeno-associated virus (rAAV) vector encoding for an osteogenic gene, enhanced osteogenesis and bone integration were achieved. In this study our aim was to evaluate the bone repair potential of calvarial autografts and allografts coated with either single-stranded rAAV2 vector (SS-rAAV-BMP2) or self-complementary pseudotyped vector (SC-rAAV-BMP2) encoding for bone morphogenetic protein (BMP)2 in a murine cranioplasty model. The grafts were implanted into critical defects in the calvariae of osteocalcin/luciferase (Oc/Luc) transgenic mice, which allowed longitudinal monitoring of osteogenic activity using bioluminescence imaging (BLI). Our results showed that the bioluminescent signal of the SC-rAAV-BMP2-coated allografts was 40% greater than that of the SS-rAAV-BMP2-coated allografts (p<0.05) and that the bioluminescent signal of the SS-rAAV-BMP2-coated allografts was not significantly different from the signals of the autografts or uncoated allografts. Micro-computed tomography (µCT) confirmed the significant increase in osteogenesis in the SC-rAAV-BMP2 group compared with the SS-rAAV-BMP2 group (p<0.05), indicating a significant difference in bone formation when compared with the other grafts tested. In addition, histological analysis revealed extensive remodelling of the autografts. Collectively, these results demonstrate the feasibility of craniofacial regeneration using SC-rAAV-BMP2-coated allografts, which may be an attractive therapeutic solution for repair of severe craniofacial bone defects.


Asunto(s)
Proteína Morfogenética Ósea 2/biosíntesis , Regeneración Ósea , Trasplante Óseo , Dependovirus , Vectores Genéticos , Osteogénesis , Animales , Proteína Morfogenética Ósea 2/genética , Femenino , Ratones , Ratones Transgénicos , Procedimientos de Cirugía Plástica/métodos , Trasplante Homólogo
2.
J Orthop Res ; 30(12): 1932-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22696396

RESUMEN

Tendon tissue regeneration is an important goal for orthopedic medicine. We hypothesized that implantation of Smad8/BMP2-engineered MSCs in a full-thickness defect of the Achilles tendon (AT) would induce regeneration of tissue with improved biomechanical properties. A 2 mm defect was created in the distal region of murine ATs. The injured tendons were then sutured together or given implants of genetically engineered MSCs (GE group), non-engineered MSCs (CH3 group), or fibrin gel containing no cells (FG group). Three weeks later the mice were killed, and their healing tendons were excised and processed for histological or biomechanical analysis. A biomechanical analysis showed that tendons that received implants of genetically engineered MSCs had the highest effective stiffness (>70% greater than natural healing, p < 0.001) and elastic modulus. There were no significant differences in either ultimate load or maximum stress among the treatment groups. Histological analysis revealed a tendon-like structure with elongated cells mainly in the GE group. ATs that had been implanted with Smad8/BMP2-engineered stem cells displayed a better material distribution and functional recovery than control groups. While additional study is required to determine long-term effects of GE MSCs on tendon healing, we conclude that genetically engineered MSCs may be a promising therapeutic tool for accelerating short-term functional recovery in the treatment of tendon injuries.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , Proteína Smad8/metabolismo , Ingeniería de Tejidos/métodos , Tendón Calcáneo/patología , Animales , Fenómenos Biomecánicos , Módulo de Elasticidad , Femenino , Fibrina/metabolismo , Ingeniería Genética/métodos , Ratones , Ratones Endogámicos C3H , Traumatismos de los Tendones/patología , Tendones/patología , Cicatrización de Heridas
3.
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
4.
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
5.
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
6.
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
7.
Tissue Eng Part B Rev ; 16(1): 13-20, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20143927

RESUMEN

While various problems with bone healing remain, the greatest clinical change is the absence of an effective approach to manage large segmental defects in limbs and craniofacial bones caused by trauma or cancer. Thus, nontraditional forms of medicine, such as gene therapy, have been investigated as a potential solution. The use of osteogenic genes has shown great potential in bone regeneration and fracture healing. Several methods for gene delivery to the fracture site have been described. The majority of them include a cellular component as the carrying vector, an approach known as cell-mediated gene therapy. Yet, the complexity involved with cell isolation and culture emphasizes the advantages of direct gene delivery as an alternative strategy. Here we review the various approaches of direct gene delivery for bone repair, the choice of animal models, and the various outcome measures required to evaluate the efficiency and safety of each technique. Special emphasis is given to noninvasive, quantitative, in vivo monitoring of gene expression and biodistribution in live animals. Research efforts should aim at inducing a transient, localized osteogenic gene expression within a fracture site to generate an effective therapeutic approach that would eventually lead to clinical use.


Asunto(s)
Regeneración Ósea/fisiología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Modelos Animales , Animales , Curación de Fractura/fisiología , Resultado del Tratamiento
8.
NMR Biomed ; 23(6): 554-62, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20175140

RESUMEN

In order to investigate intervertebral disc (IVD) degeneration and repair, a quantitative non-invasive tool is needed. Various MRI methods including qCPMG, which yields dipolar echo relaxation time (T(DE)), magnetization transfer contrast (MTC), and (1)H and (2)H double quantum filtered (DQF) MRI were used in the present work to monitor changes in rat IVD after ablation of the nucleus pulposus (NP), serving as a model of severe IVD degeneration. In the intact IVD, a clear distinction between the annulus fibrosus (AF) and the NP is obtained on T(2) and T(DE) weighted images as well as on MTC maps, reflecting the high concentration of ordered collagen fibers in the AF. After ablation of the NP, the distinction between the compartments is lost. T(2) and T(DE) relaxation times are short throughout the disc and MTC is high. (1)H and (2)H DQF signal, which in intact discs is obtained only for the AF, is now observable throughout the tissue. These results indicate that after ablation, there is an ingression of collagen fibers from the AF into the area that was previously occupied by the NP, as was confirmed by histology.


Asunto(s)
Degeneración del Disco Intervertebral/patología , Disco Intervertebral/patología , Imagen por Resonancia Magnética/métodos , Animales , Colágeno/metabolismo , Femenino , Humanos , Disco Intervertebral/metabolismo , Ratas , Ratas Wistar
9.
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
10.
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
11.
Tissue Eng Part C Methods ; 15(3): 485-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19366317

RESUMEN

This work advances fibered confocal microscopy (FCM) as a functional imaging platform for in vivo assessment of tissue mechanics. Building on our earlier studies demonstrating proof of principle and introducing an analytical framework for FCM image processing, here we present data that improve and validate several critical aspects of FCM. Specifically, we have considerably reduced the invasiveness of the imaging procedure, and verified that endoscopic imaging through a transcutaneous access point does not induce functional changes in passive ankle joint biomechanics. We have also verified that periodic (weekly) measurements on uninjured tendons are reproducible. Importantly, we have further proven that the method can sensitively detect and quantify compromised tendon mechanics in injured tendons. These incremental but essential developments further push FCM measurement of tissue mechanics from a novel concept to a usable tool that fills an important niche by functionally imaging living tissue at the highest available spatial resolution of any currently available in vivo imaging method. It is expected that functional FCM imaging will eventually enable accelerated screening of preclinical therapies, and allow researchers to quantifiably relate implanted cell behavior with resulting changes in tissue structure and function.


Asunto(s)
Tecnología de Fibra Óptica/instrumentación , Aumento de la Imagen/instrumentación , Microscopía Confocal/instrumentación , Regeneración/fisiología , Tendones/citología , Tendones/fisiología , Animales , Módulo de Elasticidad , Diseño de Equipo , Análisis de Falla de Equipo , Femenino , Ratones , Ratones Endogámicos C3H , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Estrés Mecánico
12.
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
13.
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
14.
Methods Mol Biol ; 455: 261-72, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18463824

RESUMEN

Monitoring gene expression in vitro and in vivo, is crucial when analyzing osteogenesis and developing effective bone gene therapy protocols. Until recently, molecular analytical tools were only able to detect protein expression either in vitro or in vivo. These systems include histology and immunohistochemistry, fluorescent imaging, PET (micro-PET), CT (micro-CT), and bioluminescent imaging. The last is the only system to date that can enable efficient quantitative monitoring of gene expression both in vitro and in vivo. Effective bioluminescent imaging in bone can be achieved by using transgenic mice harboring the luciferase reporter gene, downstream of an osteogenesis specific promoter. The aim of this chapter is to comprehensively describe the various protocols needed for the detection of bioluminescence in bone development and repair.


Asunto(s)
Huesos/citología , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Animales , Huesos/metabolismo , Humanos , Inmunohistoquímica/métodos , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Osteocalcina/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
15.
Stem Cells ; 26(4): 1056-64, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18218819

RESUMEN

Multiple factors alter intervertebral disc volume, structure, shape, composition, and biomechanical properties, often leading to low back pain. Spinal fusion is frequently performed to treat this problem. We recently published results of our investigation of a novel system of in vivo bone formation, in which we used nonvirally nucleofected human mesenchymal stem cells that overexpress a bone morphogenetic protein gene. We hypothesized that primary porcine adipose tissue-derived stem cells (ASCs) nucleofected with plasmid containing recombinant human bone morphogenetic protein-6 (rhBMP-6) could induce bone formation and achieve spinal fusion in vivo. Primary ASCs were isolated from freshly harvested porcine adipose tissue. Overexpression of rhBMP-6 was achieved ex vivo by using a nucleofection technique. Transfection efficiency was monitored by assessing a parallel transfection involving an enhanced green fluorescent protein reporter gene and flow cytometry analysis. rhBMP-6 protein secreted by the cells was measured by performing an enzyme-linked immunosorbent assay. Genetically engineered cells were injected into the lumbar paravertebral muscle in immunodeficient mice. In vivo bone formation was monitored by a quantitative microcomputed tomography (muCT). The animals were euthanized 5 weeks postinjection, and spinal fusion was evaluated using in vitro muCT and histological analysis. We found formation of a large bone mass adjacent to the lumbar area, which produced posterior spinal fusion of two to four vertebrae. Our data demonstrate that efficient bone formation and spinal fusion can be achieved using ex vivo, nonvirally transfected primary ASCs. These results could pave the way to a novel biological solution for spine treatment.


Asunto(s)
Tejido Adiposo/trasplante , Fusión Vertebral/métodos , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/métodos , Tejido Adiposo/citología , Tejido Adiposo/fisiología , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre/citología , Células Madre/fisiología , Porcinos
16.
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
17.
J Biomech ; 40(2): 399-411, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-16524583

RESUMEN

Stem cell-based gene therapy and tissue engineering have been shown to be an efficient method for the regeneration of critical-sized bone defects. Despite being an area of active research over the last decade, no knowledge of the intrinsic ultrastructural and nanomechanical properties of such bone tissue exists. In this study, we report the nanomechanical properties of engineered bone tissue derived from genetically modified mesenchymal stem cells (MSCs) overexpressing the rhBMP2 gene, grown in vivo in the thigh muscle of immunocompetent mice for 4 weeks, compared to femoral bone adjacent to the transplantation site. The two types of bone had similar mineral contents (61 and 65 wt% for engineered and femoral bone, respectively), overall microstructures showing lacunae and canaliculi (both measured by back-scattered electron microscopy), chemical compositions (measured by energy dispersive X-ray analysis), and nanoscale topographical morphologies (measured by tapping-mode atomic force microscopy imaging or TMAFM). Nanoindentation experiments revealed that the small length scale mechanical properties were statistically different with the femoral bone (indented parallel to the bone long axis) being stiffer and harder (apparent elastic modulus, E approximately 27.3+/-10.5 GPa and hardness, H approximately 1.0+/-0.7G Pa) than the genetically engineered bone (E approximately 19.8+/-5.6 GPa, H approximately 0.9+/-0.4G Pa). TMAFM imaging showed clear residual indents characteristic of viscoelastic plastic deformation for both types of bone. However, fine differences in the residual indent area (smaller for the engineered bone), pile up (smaller for the engineered bone), and fracture mechanisms (microcracks for the engineered bone) were observed with the genetically engineered bone behaving more brittle than the femoral control.


Asunto(s)
Sustitutos de Huesos , Huesos/fisiología , Nanotecnología , Células Madre , Ingeniería de Tejidos , Fenómenos Biomecánicos , Sustitutos de Huesos/química , Huesos/patología , Microscopía de Fuerza Atómica
18.
J Bone Miner Res ; 21(12): 1935-45, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17002559

RESUMEN

UNLABELLED: A bioinformatics-based analysis of endochondral bone formation model detected several genes upregulated in this process. Among these genes the dickkopf homolog 3 (Dkk3) was upregulated and further studies showed that its expression affects in vitro and in vivo osteogenesis. This study indicates a possible role of Dkk3 in regulating bone formation. INTRODUCTION: Endochondral bone formation is a complex biological process involving numerous chondrogenic, osteogenic, and angiogenic proteins, only some of which have been well studied. Additional key genes may have important roles as well. We hypothesized that to identify key genes and signaling pathways crucial for bone formation, a comprehensive gene discovery strategy should be applied to an established in vivo model of osteogenesis. MATERIALS AND METHODS: We used in vivo implanted C3H10T1/2 cells that had been genetically engineered to express human bone morphogenetic protein-2 (BMP2) in a tetracycline-regulated system that controls osteogenic differentiation. Oligonucleotide microarray data from the implants (n = 4 repeats) was analyzed using coupled two-way clustering (CTWC) and statistical methods. For studying the effects of dickkopf homolog 3 (Dkk3) in chondrogenesis and osteogenesis, C3H10T1/2 mesenchymal progenitors were used. RESULTS: The CTWC revealed temporal expression of Dkk3 with other chondrogenesis-, osteogenesis-, and Wnt-related genes. Quantitative RT-PCR confirmed the expression of Dkk3 in the implants. C3H10T1/2 cells that expressed Dkk3 in the presence of BMP2 displayed lower levels of alkaline phosphatase and collagen I mRNA expression than control C3H10T1/2 cells that did not express Dkk3. Interestingly, the levels of collagen II mRNA expression, Alcian blue staining, and glucose aminoglycans (GAGs) production were not influenced by Dkk3 expression. In vivo microCT and bioluminescence imaging revealed that co-expression of Dkk3 and BMP2 by implanted C3H10T1/2 cells induced the formation of significantly lower quantities of bone than cells expressing only BMP2. CONCLUSIONS: A bioinformatics analysis enabled the identification of Dkk3 as a pivotal gene with a novel function in endochondral bone formation. Our results showed that Dkk3 might have inhibitory effects on osteogenesis, but no effect on chondrogenesis, indicating that Dkk3 plays a regulatory role in endochondral bone formation. Further mechanistic studies are required to reveal the mechanism of action of Dkk3 in endochondral bone formation.


Asunto(s)
Proteínas Morfogenéticas Óseas/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Osteogénesis/fisiología , Factor de Crecimiento Transformador beta/biosíntesis , Regulación hacia Arriba/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Condrogénesis/fisiología , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factor de Crecimiento Transformador beta/genética
19.
Tissue Eng ; 12(4): 877-89, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16674300

RESUMEN

There are several gene therapy approaches to tissue regeneration. Although usually efficient, virusbased approaches may elicit an immune response against the viral proteins. An alternative approach, nonviral transfer, is safer, and can be controlled and reproduced. We hypothesized that in vivo bone formation could be achieved using human mesenchymal stem cells (hMSCs) nonvirally transfected with the human bone morphogenetic protein-2 (hBMP-2) or -9 (hBMP-9) gene. Human MSCs were transfected using nucleofection, a unique electropermeabilization-based technique. Postnucleofection, cell viability was 53.6 +/- 2.5% and gene delivery efficiency was 51% to 88% (mean 68.2 +/- 4.1%), as demonstrated by flow cytometry in enhanced green fluorescent protein (EGFP)-nucleofected hMSCs. Transgene expression lasted longer than 14 days and was very low 21 days postnucleofection. Both hBMP-2- and hBMP-9-nucleofected hMSCs in culture demonstrated a significant increase in calcium deposition compared with EGFP-nucleofected hMSCs. Human BMP-2- and hBMP-9-nucleofected hMSCs transplanted in ectopic sites in NOD/SCID mice induced bone formation 4 weeks postinjection. We conclude that in vivo bone formation can be achieved by using nonvirally nucleofected hMSCs. This could lead to a breakthrough in the field of regenerative medicine, in which safer, nonviral therapeutic strategies present a very attractive alternative.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Regeneración Ósea/fisiología , Terapia Genética , Células Madre Mesenquimatosas/metabolismo , Ingeniería de Tejidos/métodos , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/metabolismo , Regeneración Ósea/genética , Calcio/análisis , Supervivencia Celular , Células Cultivadas , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/metabolismo , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Transgenes , Trasplante Heterólogo
20.
Stem Cells ; 24(7): 1728-37, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16601078

RESUMEN

The culture expansion of human mesenchymal stem cells (hMSCs) may alter their characteristics and is a costly and time-consuming stage. This study demonstrates for the first time that immunoisolated noncultured CD105-positive (CD105(+)) hMSCs are multipotent in vitro and exhibit the capacity to form bone in vivo. hMSCs are recognized as promising tools for bone regeneration. However, the culture stage is a limiting step in the clinical setting. To establish a simple, efficient, and fast method for applying these cells for bone formation, a distinct population of CD105(+) hMSCs was isolated from bone marrow (BM) by using positive selection based on the expression of CD105 (endoglin). The immunoisolated CD105(+) cell fraction represented 2.3% +/- 0.45% of the mononuclear cells (MNCs). Flow cytometry analysis of freshly immunoisolated CD105(+) cells revealed a purity of 79.7% +/- 3.2%. In vitro, the CD105(+) cell fraction displayed significantly more colony-forming units-fibroblasts (CFU-Fs; 6.3 +/- 1.4) than unseparated MNCs (1.1 +/- 0.3; p < .05). Culture-expanded CD105(+) cells expressed CD105, CD44, CD29, CD90, and CD106 but not CD14, CD34, CD45, or CD31 surface antigens, and these cells were able to differentiate into osteogenic, chondrogenic, and adipogenic lineages. In addition, freshly immunoisolated CD105(+) cells responded in vivo to recombinant bone morphogenetic protein-2 by differentiating into chondrocytes and osteoblasts. Genetic engineering of freshly immunoisolated CD105(+) cells was accomplished using either adenoviral or lentiviral vectors. Based on these findings, it is proposed that noncultured BM-derived CD105(+) hMSCs are osteogenic cells that can be genetically engineered to induce tissue generation in vivo.


Asunto(s)
Antígenos CD/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Receptores de Superficie Celular/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/aislamiento & purificación , Cartílago/citología , Endoglina , Femenino , Citometría de Flujo , Ingeniería Genética , Humanos , Inmunoensayo , Masculino , Trasplante de Células Madre Mesenquimatosas , Persona de Mediana Edad , Receptores de Superficie Celular/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...