Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Biomaterials ; 32(26): 6183-93, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21645919

RESUMO

A promising strategy to accelerate joint implant integration and reduce recovery time and failure rates is to deliver a combination of certain growth factors to the integration site. There is a need to control the quantity of growth factors delivered at different times during the healing process to maximize efficacy. Polyelectrolyte multilayer (PEM) films, built using the layer-by-layer (LbL) technique, are attractive for releasing controlled amounts of potent growth factors over a sustained period. Here, we present PEM films that sequester physiological amounts of osteogenic rhBMP-2 (recombinant human bone morphogenetic protein-2) and angiogenic rhVEGF165 (recombinant human vascular endothelial growth factor) in different ratios in a degradable [poly(ß-amino ester)/polyanion/growth factor/polyanion] LbL tetralayer repeat architecture where the biologic load scaled linearly with the number of tetralayers. No burst release of either growth factor was observed as the films degraded. The release of rhBMP-2 was sustained over a period of 2 weeks, while rhVEGF165 eluted from the film over the first 8 days. Both growth factors retained their efficacy, as quantified with relevant in vitro assays. rhBMP-2 initiated a dose dependent differentiation cascade in MC3T3-E1S4 pre-osteoblasts while rhVEGF165 upregulated HUVEC proliferation, and accelerated closure of a scratch in HUVEC cell cultures in a dose dependent manner. In vivo, the mineral density of ectopic bone formed de novo by rhBMP-2/rhVEGF165 PEM films was approximately 33% higher than when only rhBMP-2 was introduced, with a higher trabecular thickness, which would indicate a decrease in the risk of osteoporotic fracture. Bone formed throughout the scaffold when both growth factors were released, which suggests more complete remodeling due to an increased local vascular network. This study demonstrates a promising approach to delivering precise doses of multiple growth factors for a variety of implant applications where control over spatial and temporal release profile of the biologic is desired.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Polímeros/química , Fator de Crescimento Transformador beta/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Proteína Morfogenética Óssea 2/química , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos , Polieletrólitos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/química , Fator A de Crescimento do Endotélio Vascular/química
3.
Biomaterials ; 32(5): 1446-53, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084117

RESUMO

Drug eluting coatings that can direct the host tissue response to implanted medical devices have the potential to ameliorate both the medical and financial burden of complications from implantation. However, because many drugs useful in this arena are biologic in nature, a paucity of delivery strategies for biologics, including growth factors, currently limits the control that can be exerted on the implantation environment. Layer-by-Layer (LbL) polyelectrolyte multilayer films are highly attractive as ultrathin biologic reservoirs, due to the capability to conformally coat difficult geometries, the use of aqueous processing likely to preserve fragile protein function, and the tunability of incorporation and release profiles. Herein, we describe the first LbL films capable of microgram-scale release of the biologic Bone Morphogenetic Protein 2 (BMP-2), which is capable of directing the host tissue response to create bone from native progenitor cells. Ten micrograms of BMP-2 are released over a period of two weeks in vitro; less than 1% is released in the first 3 h (compared with commercial collagen matrices which can release up to 60% of BMP-2, too quickly to induce differentiation). BMP-2 released from LbL films retains its ability to induce bone differentiation in MC3T3 E1S4 pre-osteoblasts, as measured by induction of alkaline phosphatase and stains for calcium (via Alizarin Red) and calcium matrix (via Von Kossa). In vivo, BMP-2 film coated scaffolds were compared with film coated scaffolds lacking BMP-2. BMP-2 coatings implanted intramuscularly were able to initiate host progenitor cells to differentiate into bone, which matured and expanded from four to 9 weeks as measured by MicroCT and histology. Such LbL films represent new steps towards controlling and tuning host response to implanted medical devices, which may ultimately increase the success of implanted devices, provide alternative new approaches toward bone wound healing, and lay the foundation for development of a multi-therapeutic release coating.


Assuntos
Engenharia Tecidual/métodos , Animais , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Osso e Ossos/citologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X
4.
Biomacromolecules ; 11(8): 2053-9, 2010 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-20690713

RESUMO

Fibroblast growth factor 2 (FGF-2) is a potent mediator of stem cell differentiation and proliferation. Although FGF-2 has a well-established role in promoting bone tissue formation, flaws in its delivery have limited its clinical utility. Polyelectrolyte multilayer films represent a novel system for FGF-2 delivery that has promise for local, precisely controlled, and sustained release of FGF-2 from surfaces of interest, including medical implants and tissue engineering scaffolds. In this work, the loading and release of FGF-2 from synthetic hydrolytically degradable multilayer thin films of various architectures is explored; drug loading was tunable using at least three parameters (number of nanolayers, counterpolyanion, and type of degradable polycation) and yielded values of 7-45 ng/cm(2) of FGF-2. Release time varied between 24 h and approximately five days. FGF-2 released from these films retained in vitro activity, promoting the proliferation of MC3T3 preosteoblast cells. The use of biologically derived counterpolyanions heparin sulfate and chondroitin sulfate in the multilayer structures enhanced FGF-2 activity. The control over drug loading and release kinetics inform future in vivo bone and tissue regeneration models for the exploration of clinical relevance of LbL growth factor delivery films.


Assuntos
Eletrólitos/química , Fator 2 de Crescimento de Fibroblastos/química , Células 3T3 , Animais , Ensaio de Imunoadsorção Enzimática , Hidrólise , Camundongos
5.
J Control Release ; 131(3): 228-34, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-18721835

RESUMO

Layer-by-layer (LbL) films have multiple features that make them attractive for drug delivery, including the potential to sequentially deliver growth factors from implantable medical devices or tissue engineering scaffolds. To date, however, characterization has been lacking for protein delivery from such films. Here, LbL polyelectrolyte films constructed with the model protein lysozyme and a hydrolytically degradable and biocompatible synthetic polycation are characterized. Milligram/cm(2) scale linear or power law release profiles can be achieved over 2 to 34 days, and control over loading and release are demonstrated through parameters such as tuning the degradability of the synthetic polycation, changing the number of layers used, or changing the polysaccharide polyanion. Functionality is maintained at nearly 100%, underscoring mild processing conditions apt to preserve fragile protein function. LbL films thus have promise as a tool for exploring protein modulation of the interaction between implanted surfaces and the cells they contact.


Assuntos
Materiais Revestidos Biocompatíveis/química , Preparações de Ação Retardada/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Muramidase/química , Polímeros/química , Acetatos/química , Soluções Tampão , Condroitina/química , Heparina/química , Concentração de Íons de Hidrogênio , Hidrólise , Estrutura Molecular , Nanotecnologia/métodos , Fosfatos/química , Polieletrólitos , Polietilenoimina/química , Polímeros/síntese química , Cloreto de Sódio/química , Ácidos Sulfônicos/química , Propriedades de Superfície
6.
Nat Biotechnol ; 23(7): 879-84, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15965465

RESUMO

One of the major obstacles in engineering thick, complex tissues such as muscle is the need to vascularize the tissue in vitro. Vascularization in vitro could maintain cell viability during tissue growth, induce structural organization and promote vascularization upon implantation. Here we describe the induction of endothelial vessel networks in engineered skeletal muscle tissue constructs using a three-dimensional multiculture system consisting of myoblasts, embryonic fibroblasts and endothelial cells coseeded on highly porous, biodegradable polymer scaffolds. Analysis of the conditions for induction and stabilization of the vessels in vitro showed that addition of embryonic fibroblasts increased the levels of vascular endothelial growth factor expression in the construct and promoted formation and stabilization of the endothelial vessels. We studied the survival and vascularization of the engineered muscle implants in vivo in three different models. Prevascularization improved the vascularization, blood perfusion and survival of the muscle tissue constructs after transplantation.


Assuntos
Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Animais , Vasos Sanguíneos/fisiologia , Células Cultivadas , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Fibroblastos/fisiologia , Humanos , Camundongos , Mioblastos Esqueléticos/fisiologia
7.
Oncogene ; 23(33): 5643-53, 2004 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-15122324

RESUMO

It is known that excess amounts of Ski, or any member of its proto-oncoprotein family, causes disruption of the transforming growth factor beta signal transduction pathway, thus causing oncogenic transformation of cells. Previous studies indicate that Ski is a relatively unstable protein whose expression levels can be regulated by ubiquitin-mediated proteolysis. Here, we investigate the mechanism by which the stability of Ski is regulated. We show that the steady-state levels of Ski protein are controlled post-translationally by cell cycle-dependent proteolysis, wherein Ski is degraded during the interphase of the cell cycle but is relatively stable during mitosis. Furthermore, we demonstrate that the ubiquitin-conjugating enzyme Cdc34 mediates cell cycle-dependent Ski degradation both in vitro and in vivo. Overexpression of dominant-negative Cdc34 stabilizes Ski and enhances its ability to antagonize TGF-beta signaling. Our data suggest that regulated proteolysis of Ski is one of the key mechanisms that control the threshold levels of this proto-oncoprotein, and thus prevents epithelial cells from becoming TGF-beta resistant.


Assuntos
Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Animais , Linhagem Celular , Células Epiteliais , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Vison , Ligação Proteica , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Enzimas de Conjugação de Ubiquitina , Ubiquitina-Proteína Ligases/metabolismo , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA