Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
Adv Drug Deliv Rev ; 64(12): 1320-30, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22429662

RESUMO

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.


Assuntos
Doenças Ósseas/terapia , Regeneração Óssea/genética , Terapia Genética/métodos , Animais , Doenças Ósseas/patologia , Proteínas Morfogenéticas Ósseas/administração & dosagem , Proteínas Morfogenéticas Ósseas/efeitos adversos , Proteínas Morfogenéticas Ósseas/uso terapêutico , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Modelos Animais de Doenças , Vetores Genéticos/genética , Humanos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Fusão Vertebral/métodos , Engenharia Tecidual/métodos
4.
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
5.
J Tissue Eng Regen Med ; 5(5): 384-93, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20740691

RESUMO

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.


Assuntos
Engenharia Genética/métodos , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Fosfatase Alcalina/metabolismo , Animais , Reatores Biológicos , Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular , Fibrinogênio/química , Hidrogéis , Camundongos , Camundongos Endogâmicos C3H , Osteogênese , Polietilenoglicóis/química
6.
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
7.
Biomacromolecules ; 11(6): 1516-26, 2010 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-20462241

RESUMO

Stimuli responsive or "smart" hydrogels are of interest for tissue-engineering applications, featuring the advantages of minimally invasive application. Currently, these materials have yet to be used as a biological replacement in restoring the function of damaged tissues or organs. The aim of this study was to demonstrate the advantages of thermoresponsive, peptide-containing hydrogels as a supportive matrix for genetically engineered stem cells. We used injectable hydrogels, enabling cell delivery to the desired site and providing adequate scaffolding postimplantation. Thermoresponsive hydrogels were developed based on amphiphilic block copolymers of polyethylene-oxide and polypropylene-oxide end-capped with methacrylate or maleimide entities and further reacted with RGD-containing peptides. Cell metabolic activity and survival within those hydrogels was studied, illustrating that the stable peptide-polymer conjugate is required for prolonged cell support. The unique polymer characteristics, combined with its enhanced cell interactions, suggest the potential use of these biomaterials in various tissue engineering applications.


Assuntos
Proteína Morfogenética Óssea 2/genética , Osso e Ossos/citologia , Hidrogéis/química , Células-Tronco Mesenquimais/citologia , Oligopeptídeos/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Osso e Ossos/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , DNA/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Microscopia Eletrônica de Varredura , Reologia , Solubilidade , Espectrometria por Raios X , Propriedades de Superfície , Temperatura , Viscosidade
8.
Biomaterials ; 30(27): 4639-48, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19540585

RESUMO

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.


Assuntos
Fluorocarbonos/farmacologia , Hidrogéis/química , Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Oxigênio/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Implantes Experimentais , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Osteocalcina/genética , Osteocalcina/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Rádio (Anatomia)/efeitos dos fármacos , Rádio (Anatomia)/patologia , Fusão Vertebral , Tetraciclina/farmacologia , Cicatrização/efeitos dos fármacos
9.
J Clin Invest ; 118(2): 439-44, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18246194

RESUMO

Tendons and ligaments are unique forms of connective tissue that are considered an integral part of the musculoskeletal system. The ultimate function of tendon is to connect muscles to bones and to conduct the forces generated by muscle contraction into movements of the joints, whereas ligaments connect bone to bone and provide joint stabilization. Unfortunately, the almost acellular and collagen I-rich structure of tendons and ligaments makes them very poorly regenerating tissues. Injured tendons and ligaments are considered a major clinical challenge in orthopedic and sports medicine. This Review discusses the several factors that might serve as molecular targets that upon activation can enhance or lead to tendon neoformation.


Assuntos
Regeneração , Traumatismos dos Tendões/metabolismo , Tendões/fisiologia , Animais , Biomarcadores/metabolismo , Linhagem Celular , Humanos , Camundongos , Morfogênese , Tendões/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA