Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Ther ; 19(1): 53-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20859259

RESUMO

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.


Assuntos
Regeneração Óssea/genética , Eletroporação/métodos , Fraturas não Consolidadas/terapia , Terapia Genética/métodos , Fator 2 de Diferenciação de Crescimento/genética , Osteogênese/genética , Células-Tronco/fisiologia , Animais , Colágeno/administração & dosagem , Feminino , Fraturas não Consolidadas/patologia , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Fator 2 de Diferenciação de Crescimento/biossíntese , Luciferases/biossíntese , Luciferases/genética , Camundongos , Camundongos Endogâmicos C3H , Plasmídeos/genética , Cicatrização/genética
2.
Mol Pharm ; 8(5): 1592-601, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21834548

RESUMO

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.


Assuntos
Células-Tronco Adultas/transplante , Proteína Morfogenética Óssea 6/uso terapêutico , Regeneração Óssea , Técnicas de Transferência de Genes , Traumatismos da Coluna Vertebral/terapia , Coluna Vertebral/fisiologia , Células-Tronco Adultas/metabolismo , Animais , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Células Cultivadas , Fibrina/química , Genes Reporter , Hidrogel de Polietilenoglicol-Dimetacrilato , Osteocalcina/genética , Regiões Promotoras Genéticas , Radiografia , Distribuição Aleatória , Ratos , Ratos Nus , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Traumatismos da Coluna Vertebral/diagnóstico por imagem , Traumatismos da Coluna Vertebral/metabolismo , Traumatismos da Coluna Vertebral/patologia , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/patologia , Gordura Subcutânea Abdominal/citologia , Suínos , Porco Miniatura , Cauda , Ubiquitina/genética
3.
NMR Biomed ; 23(6): 554-62, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20175140

RESUMO

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.


Assuntos
Degeneração do Disco Intervertebral/patologia , Disco Intervertebral/patologia , Imageamento por Ressonância Magnética/métodos , Animais , Colágeno/metabolismo , Feminino , Humanos , Disco Intervertebral/metabolismo , Ratos , Ratos Wistar
4.
J Clin Invest ; 116(4): 940-52, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16585960

RESUMO

Tissue regeneration requires the recruitment of adult stem cells and their differentiation into mature committed cells. In this study we describe what we believe to be a novel approach for tendon regeneration based on a specific signalling molecule, Smad8, which mediates the differentiation of mesenchymal stem cells (MSCs) into tendon-like cells. A biologically active Smad8 variant was transfected into an MSC line that coexpressed the osteogenic gene bone morphogenetic protein 2 (BMP2). The engineered cells demonstrated the morphological characteristics and gene expression profile of tendon cells both in vitro and in vivo. In addition, following implantation in an Achilles tendon partial defect, the engineered cells were capable of inducing tendon regeneration demonstrated by double quantum filtered MRI. The results indicate what we believe to be a novel mechanism in which Smad8 inhibits the osteogenic pathway in MSCs known to be induced by BMP2 while promoting tendon differentiation. These findings may have considerable importance for the therapeutic replacement of tendons or ligaments and for engineering other tissues in which BMP plays a pivotal developmental role.


Assuntos
Tendão do Calcâneo/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Proteína Smad8/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Biomarcadores , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular , Células Cultivadas , Feminino , Histocitoquímica , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C3H , Microscopia de Contraste de Fase , Estrutura Terciária de Proteína , Ratos , Ratos Nus , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Smad8/genética , Células-Tronco , Engenharia Tecidual , Ativação Transcricional , Transfecção , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia
5.
Stem Cells ; 26(4): 1056-64, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18218819

RESUMO

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.


Assuntos
Tecido Adiposo/transplante , Fusão Vertebral/métodos , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Tecido Adiposo/citologia , Tecido Adiposo/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco/citologia , Células-Tronco/fisiologia , Suínos
6.
Methods Mol Biol ; 455: 261-72, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18463824

RESUMO

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.


Assuntos
Osso e Ossos/citologia , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Animais , Osso e Ossos/metabolismo , Humanos , Imuno-Histoquímica/métodos , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Osteocalcina/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
7.
J Biomech ; 40(2): 399-411, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16524583

RESUMO

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.


Assuntos
Substitutos Ósseos , Osso e Ossos/fisiologia , Nanotecnologia , Células-Tronco , Engenharia Tecidual , Fenômenos Biomecânicos , Substitutos Ósseos/química , Osso e Ossos/patologia , Microscopia de Força Atômica
8.
J Bone Miner Res ; 21(12): 1935-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17002559

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Osteogênese/fisiologia , Fator de Crescimento Transformador beta/biossíntese , Regulação para Cima/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Linhagem Celular , Condrogênese/fisiologia , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator de Crescimento Transformador beta/genética
9.
Tissue Eng ; 12(11): 3021-34, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17518618

RESUMO

Angiogenesis is mandatory for reperfusion of viable tissues, and lack of vascularization may cause ischemia. The increasing disparity between the demand and availability of adequate substitutes for small-diameter human blood vessels has prompted an intensive search for artificial materials or biological allograft tissues, both of which usually fail in the long term. The objective of this study was to pioneer a novel model for in vivo guided angiogenesis based on a specific design process of a filamentous polymeric scaffold with endothelial cells in a 3-dimensional culture system. To our knowledge, this is the first report of an in vivo guided angiogenesis approach based on a 2-step model, composed of endothelial cells and a filamentous polymeric scaffold framework. Endothelial cells that had been cultured on a specifically designed filamentous polymeric scaffold within a regulated dynamic tissue culture system were shown in vivo to induce guided angiogenesis. Cells seeded on a biodegradable polymeric scaffold were implanted into mice. On day 28 after implantation, analysis revealed a guided angiogenic process along the path of the implanted polymeric scaffold as well as initial evidence for early maturation of engineered vessels, allowing red blood cells to flow through the forming lumina of new vessels as the polymer degraded. The authors conclude that in vivo guided angiogenesis can be achieved by combining endothelial cells with biodegradable filamentous polymeric scaffolds and that this model can lay the cornerstone for vascular engineering and future development of clinically available protocols aimed to treat life-threatening cardiovascular conditions.


Assuntos
Células Endoteliais/citologia , Neovascularização Fisiológica , Polímeros/química , Engenharia Tecidual/métodos , Actinas/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Reatores Biológicos , Neoplasias Encefálicas/patologia , Varredura Diferencial de Calorimetria , Caproatos/química , Células Cultivadas , Células Endoteliais/química , Células Endoteliais/transplante , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Luciferases/metabolismo , Medições Luminescentes , Imageamento por Ressonância Magnética , Teste de Materiais , Camundongos , Camundongos Nus , Modelos Biológicos , Peso Molecular , Músculo Liso/metabolismo , Técnicas de Cultura de Órgãos , Polietilenoglicóis/química , Especificidade por Substrato , Fatores de Tempo , Estanho
10.
Tissue Eng ; 12(4): 877-89, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16674300

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Regeneração Óssea/fisiologia , Terapia Genética , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual/métodos , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/metabolismo , Regeneração Óssea/genética , Cálcio/análise , Sobrevivência Celular , Células Cultivadas , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/metabolismo , Fator 2 de Diferenciação de Crescimento , Fatores de Diferenciação de Crescimento , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Transgenes , Transplante Heterólogo
11.
J Biomed Opt ; 11(6): 064010, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17212533

RESUMO

This study explores a novel method to quantify in vivo soft tissue biomechanics from endoscopic confocal fluorescence microscope images of externally loaded biological tissues. A custom algorithm based on normalized cross-correlation is used to track fluorescently labeled cells within soft tissue structures as they deform. Cellular displacements are subsequently reduced to tissue strains by deriving the spatial gradient of the spline smoothed cellular displacement field. The relative performance of the tracking method is verified using a synthetic dataset with known underlying deformation. In biological application of the method, tissue strains are measured in the Achilles tendon of an anesthetized mouse. Over repeated trials, structural strain in the tendon (i.e., the relative change in distance between cells located at view field extremes) is 20.3+/-3.1%, thus establishing the reproducibility of the loading protocol. Analysis of local tendon tissue strains reveal primary engineering strains in the tissue to range from 5 to 55%, signifying a highly inhomogeneous strain state, with complex relative motions of neighboring tendon substructures. In summary, the current work establishes a baseline for a promising experimental method, and demonstrates its technical feasibility.


Assuntos
Endoscopia/métodos , Interpretação de Imagem Assistida por Computador/métodos , Micromanipulação/métodos , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Tendões/citologia , Tendões/fisiologia , Animais , Fenômenos Biomecânicos/métodos , Movimento Celular/fisiologia , Tamanho Celular , Elasticidade , Feminino , Camundongos , Camundongos Endogâmicos C3H , Microscopia de Vídeo/métodos , Estresse Mecânico
12.
J Neurosurg Spine ; 3(1): 47-52, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16122022

RESUMO

OBJECT: The authors hypothesized that spinal fusion can be achieved and monitored by using cell-mediated gene therapy. Mesenchymal stem cells (MSCs) genetically engineered to express recombinant human bone morphogenetic protein-2 (rhBMP-2) conditionally, were implanted into the paraspinal muscles of mice to establish spinal fusion. The goal was to demonstrate an MSC-based gene therapy platform in which controlled gene expression is used to obtain spinal fusion in a murine model. METHODS: Mesenchymal stem cells expressing the rhBMP-2 gene were injected into the paravertebral muscle in mice. Bone formation in the paraspinal region was longitudinally followed by performing micro-computerized tomography scanning, histological studies, and an analysis of osteocalcin expression to demonstrate the presence of engrafted engineered MSCs. The minimal period of rhBMP-2 expression by the engineered MSCs required to induce fusion was determined. The results of this study demonstrate that genetically engineered MSCs induce bone formation in areas adjacent to and touching the posterior elements of the spine. This newly formed bone fuses the spine, as demonstrated by radiological and histological studies. The authors demonstrate that injected cells induce active osteogenesis at the site of implantation for up to 4 weeks postinjection. They found that a 7-day induction of rhBMP-2 expression in genetically engineered MSCs was sufficient to form new bone tissue, although the quantity of this bone increased as longer expression periods were implemented. CONCLUSIONS: After their injection genetically engineered MSCs can efficiently form new bone in the paraspinal muscle of the mouse to obtain spinal fusion. The extent and quantity of this newly formed bone can be monitored by controlling the duration of rhBMP-2 gene expression.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Engenharia Genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Osteogênese/fisiologia , Fusão Vertebral/métodos , Fator de Crescimento Transformador beta/fisiologia , Animais , Proteína Morfogenética Óssea 2 , Estudos de Viabilidade , Feminino , Camundongos , Camundongos Endogâmicos C3H , Modelos Animais , Fatores de Tempo
13.
J Bone Miner Res ; 18(3): 570-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12619943

RESUMO

Monitoring gene expression in vivo, noninvasively, is a critical issue in effective gene therapy systems. To date, there are no adequate molecular imaging techniques, which quantitatively monitor gene expression in vivo in skeletal development and repair. The aim of this study was to monitor gene expression in skeletal development and repair, using a real-time molecular imaging system, which quantitatively and noninvasively detects bioluminescence in vivo. Our experimental model consisted of transgenic mice harboring the luciferase marker gene under the regulation of the human osteocalcin (hOC) promoter. A new light detection cooled charge coupled device (CCCD) camera was applied to monitor luciferase expression. In vitro, mesenchymal stem cells (MSCs) isolated from bone marrow of transgenic mice exhibited hOC promoter regulation, detected by luciferase expression that correlated with their osteogenic differentiation. During development from 1 week to 1.5 years, transgenic mice exhibited transgene expression in a wide spectrum of skeletal organs, including calvaria, vertebra, tail, and limbs, reaching a peak at 1 week in most of the skeletal organs. In two skeletal repair models, bone fracture and marrow ablation, the noninvasive CCCD system revealed a peak of luciferase expression at 6 days postsurgery. All quantitative, noninvasive, real-time CCCD measurements correlated with a luciferase biochemical assay and luciferase immunohistochemistry, which demonstrated luciferase expression in hypertrophic chondrocytes and trabecular osteoblasts. Our studies show for the first time (1) the CCCD detection system is a reliable quantitative gene detection tool for the skeleton in vivo, (2) expression of luciferase regulated by the hOC promoter is significantly decreased with age in most skeletal sites, and (3) the dynamics of hOC regulation during mice skeletal development and repair in real time, quantitatively and noninvasively.


Assuntos
Desenvolvimento Ósseo , Remodelação Óssea , Expressão Gênica , Animais , Sequência de Bases , Primers do DNA , Humanos , Luciferases , Medições Luminescentes , Camundongos , Camundongos Transgênicos , Osteocalcina/genética
14.
Biofactors ; 20(2): 71-84, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15322331

RESUMO

The functional roles of BMP type IA and IB receptors mediating differentiation into the osteogenic and chondrogenic lineage were investigated in the mesenchymal progenitor line C3H10T1/2 in vitro. The capacity of type IA and IB BMP receptors was assessed by the forced expression of the wild-type (wtBMPR-IA or IB) and of the kinase-deficient, dominant-negative form (dnBMPR-IA or -IB) in parental C3H10T1/2 progenitors as well as in C3H10T1/2 progenitors which recombinantly express BMP2 (C3H10T1/2-BMP2) or GDF5 (C3H10T1/2-GDF5). Consistent with the higher endogenous expression rate of BMPR-IA in comparison with BMPR-IB, BMPR-IA plays the dominant role in BMP2-mediated osteo-/chondrogenic development. BMPR-IB moderately influences osteogenic and hardly chondrogenic development. BMPR-IB seems to be unable to efficiently activate downstream signaling pathways upon forced expression. However, a mutation conferring constitutive activity to the BMPR-IB receptor indicates that this receptor possesses the capacity to activate downstream signaling cascades. These results suggest that in mesenchymal progenitors C3H10T1/2 BMPR-IA is responsible for the initiation of the osteogenic as well as chondrogenic development and that BMPR-IA and -IB receptor pathways are well separated in this mesenchymal progenitor line and may not substitute each other. In addition this indicates that type IB and IA BMP receptors may transmit different signals during the specification and differentiation of mesenchymal lineages.


Assuntos
Diferenciação Celular/fisiologia , Condrócitos/citologia , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Animais , Proteína Morfogenética Óssea 2 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Linhagem Celular , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Endogâmicos C3H , Osteogênese , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/análise , Receptores de Fatores de Crescimento/genética , Proteínas Recombinantes de Fusão , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/fisiologia
15.
J Orthop Res ; 30(12): 1932-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22696396

RESUMO

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.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Proteína Smad8/metabolismo , Engenharia Tecidual/métodos , Tendão do Calcâneo/patologia , Animais , Fenômenos Biomecânicos , Módulo de Elasticidade , Feminino , Fibrina/metabolismo , Engenharia Genética/métodos , Camundongos , Camundongos Endogâmicos C3H , Traumatismos dos Tendões/patologia , Tendões/patologia , Cicatrização
16.
J Tissue Eng Regen Med ; 6(10): e43-50, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22941779

RESUMO

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.


Assuntos
Proteína Morfogenética Óssea 2/biossíntese , Regeneração Óssea , Transplante Ósseo , Dependovirus , Vetores Genéticos , Osteogênese , Animais , Proteína Morfogenética Óssea 2/genética , Feminino , Camundongos , Camundongos Transgênicos , Procedimentos de Cirurgia Plástica/métodos , Transplante Homólogo
17.
J Biomech ; 43(12): 2315-20, 2010 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-20471652

RESUMO

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.


Assuntos
Fraturas não Consolidadas/terapia , Terapia Genética/métodos , Transplante de Células-Tronco Mesenquimais , Animais , Fenômenos Biomecânicos , Densidade Óssea , Proteína Morfogenética Óssea 2/genética , Regeneração Óssea , Remodelação Óssea , Modelos Animais de Doenças , Módulo de Elasticidade , Feminino , Análise de Elementos Finitos , Fraturas não Consolidadas/diagnóstico por imagem , Fraturas não Consolidadas/fisiopatologia , Engenharia Genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Fraturas do Rádio/diagnóstico por imagem , Fraturas do Rádio/fisiopatologia , Fraturas do Rádio/terapia , Proteínas Recombinantes/genética , Microtomografia por Raio-X
18.
Tissue Eng Part B Rev ; 16(1): 13-20, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20143927

RESUMO

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.


Assuntos
Regeneração Óssea/fisiologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Modelos Animais , Animais , Consolidação da Fratura/fisiologia , Resultado do Tratamento
19.
Tissue Eng Part A ; 16(12): 3679-86, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20618082

RESUMO

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.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fusão Vertebral , Animais , Fenômenos Biomecânicos , Linhagem Celular , Feminino , Imuno-Histoquímica , Células-Tronco Mesenquimais/fisiologia , Camundongos , Osteogênese/genética , Osteogênese/fisiologia , Coluna Vertebral/citologia , Coluna Vertebral/cirurgia , Microtomografia por Raio-X
20.
Tissue Eng Part C Methods ; 15(3): 485-91, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19366317

RESUMO

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.


Assuntos
Tecnologia de Fibra Óptica/instrumentação , Aumento da Imagem/instrumentação , Microscopia Confocal/instrumentação , Regeneração/fisiologia , Tendões/citologia , Tendões/fisiologia , Animais , Módulo de Elasticidade , Desenho de Equipamento , Análise de Falha de Equipamento , Feminino , Camundongos , Camundongos Endogâmicos C3H , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA