Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Mater Today Bio ; 20: 100641, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37179535

RESUMEN

Collagen type I lacks affinity for growth factors (GFs) and yet it is clinically used to deliver bone morphogenic protein 2 (BMP-2), a potent osteogenic growth factor. To mitigate this lack of affinity, supra-physiological concentrations of BMP-2 are loaded in collagen sponges leading to uncontrolled BMP-2 leakage out of the material. This has led to important adverse side effects such as carcinogenesis. Here, we design recombinant dual affinity protein fragments, produced in E. Coli, which contain two regions, one that spontaneously binds to collagen and a second one that binds BMP-2. By adding the fragment to collagen sponges, BMP-2 is sequestered enabling solid phase presentation of BMP-2. We demonstrate osteogenesis in vivo with ultra-low doses of BMP-2. Our protein technology enhances the biological activity of collagen without using complex chemistries or changing the manufacturing of the base material and so opens a pathway to clinical translation.

2.
Biomaterials ; 266: 120450, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33096376

RESUMEN

Mesenchymal stem cells are the focus of intense research in bone development and regeneration. The potential of microparticles as modulating moieties of osteogenic response by utilizing their architectural features is demonstrated herein. Topographically textured microparticles of varying microscale features are produced by exploiting phase-separation of a readily soluble sacrificial component from polylactic acid. The influence of varying topographical features on primary human mesenchymal stem cell attachment, proliferation and markers of osteogenesis is investigated. In the absence of osteoinductive supplements, cells cultured on textured microparticles exhibit notably increased expression of osteogenic markers relative to conventional smooth microparticles. They also exhibit varying morphological, attachment and proliferation responses. Significantly altered gene expression and metabolic profiles are observed, with varying histological characteristics in vivo. This study highlights how tailoring topographical design offers cell-instructive 3D microenvironments which allow manipulation of stem cell fate by eliciting the desired downstream response without use of exogenous osteoinductive factors.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre , Ingeniería de Tejidos
3.
Biomaterials ; 252: 120104, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32422492

RESUMEN

Extracellular matrix (ECM)-derived matrices such as Matrigel are used to culture numerous cell types in vitro as they recapitulate ECM properties that support cell growth, organisation, migration and differentiation. These ECM-derived matrices contain various growth factors which make them highly bioactive. However, they suffer lot-to-lot variability, undefined composition and lack of controlled physical properties. There is a need to develop rationally designed biomaterials that can also recapitulate ECM roles. Here, we report the development of fibronectin (FN)-based 3D hydrogels of controlled stiffness and degradability that incorporate full-length FN to enable solid-phase presentation of growth factors in a physiological manner. We demonstrate, in vitro and in vivo, the effect of incorporating vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP2) in these hydrogels to enhance angiogenesis and bone regeneration, respectively. These hydrogels represent a step-change in the design of well-defined, reproducible, synthetic microenvironments for 3D cell culture that incorporate growth factors to achieve functional effects.


Asunto(s)
Fibronectinas , Hidrogeles , Matriz Extracelular , Péptidos y Proteínas de Señalización Intercelular , Factor A de Crecimiento Endotelial Vascular
4.
Macromol Biosci ; 17(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28547877

RESUMEN

Polyelectrolyte multilayer (PEM) coatings on biomaterials are applied to tailor adhesion, growth, and function of cells on biomedical implants. Here, biogenic and synthetic polyelectrolytes (PEL) are used for layer-by-layer assembly to study the osteogenic activity of PEM with human osteosarcoma MG-63 cells in a comparative manner. Formation of PEM is achieved with biogenic PEL fibrinogen (FBG) and poly-l-lysine (PLL) as well as biotinylated chondroitin sulfate (BCS) and avidin (AVI), while poly(allylamine hydrochloride) (PAH) and polystyrene sulfonate (PSS) represent a fully synthetic PEM used as a reference system here. Surface plasmon resonance measurements show highest layer mass for FBG/PLL and similar for PSS/PAH and BCS/AVI systems, while water contact angle and zeta potential measurements indicate larger differences for PSS/PAH and FBG/PLL but not for BCS/AVI multilayers. All PEM systems support cell adhesion and growth and promote osteogenic differentiation as well. However, FBG/PLL layers are superior regarding MG-63 cell adhesion during short-term culture, while the BCS/AVI system increases alkaline phosphatase activity in long-term culture. Particularly, a multilayer system based on affinity interaction like BCS/AVI may be useful for controlled presentation of biotinylated growth factors to promote growth and differentiation of cells for biomedical applications.


Asunto(s)
Neoplasias Óseas/metabolismo , Osteogénesis/efectos de los fármacos , Osteosarcoma/metabolismo , Polielectrolitos , Avidina/química , Avidina/farmacología , Neoplasias Óseas/patología , Línea Celular Tumoral , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacología , Fibrinógeno/química , Fibrinógeno/farmacología , Humanos , Osteosarcoma/patología , Poliaminas/química , Poliaminas/farmacología , Polielectrolitos/química , Polielectrolitos/farmacología , Polilisina/química , Polilisina/farmacología
5.
Biomaterials ; 126: 61-74, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28279265

RESUMEN

We have engineered polymer-based microenvironments that promote vasculogenesis both in vitro and in vivo through synergistic integrin-growth factor receptor signalling. Poly(ethyl acrylate) (PEA) triggers spontaneous organization of fibronectin (FN) into nanonetworks which provide availability of critical binding domains. Importantly, the growth factor binding (FNIII12-14) and integrin binding (FNIII9-10) regions are simultaneously available on FN fibrils assembled on PEA. This material platform promotes synergistic integrin/VEGF signalling which is highly effective for vascularization events in vitro with low concentrations of VEGF. VEGF specifically binds to FN fibrils on PEA compared to control polymers (poly(methyl acrylate), PMA) where FN remains in a globular conformation and integrin/GF binding domains are not simultaneously available. The vasculogenic response of human endothelial cells seeded on these synergistic interfaces (VEGF bound to FN assembled on PEA) was significantly improved compared to soluble administration of VEGF at higher doses. Early onset of VEGF signalling (PLCγ1 phosphorylation) and both integrin and VEGF signalling (ERK1/2 phosphorylation) were increased only when VEGF was bound to FN nanonetworks on PEA, while soluble VEGF did not influence early signalling. Experiments with mutant FN molecules with impaired integrin binding site (FN-RGE) confirmed the role of the integrin binding site of FN on the vasculogenic response via combined integrin/VEGF signalling. In vivo experiments using 3D scaffolds coated with FN and VEGF implanted in the murine fat pad demonstrated pro-vascularization signalling by enhanced formation of new tissue inside scaffold pores. PEA-driven organization of FN promotes efficient presentation of VEGF to promote vascularization in regenerative medicine applications.


Asunto(s)
Microambiente Celular , Integrinas/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Ingeniería de Tejidos/métodos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibronectinas/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Ratones Endogámicos C57BL , Mutación/genética , Fosfolipasa C gamma/metabolismo , Fosforilación , Unión Proteica
6.
Biomaterials ; 113: 170-175, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816000

RESUMEN

Degradable hydrogels to deliver bioactive proteins represent an emerging platform for promoting tissue repair and vascularization in various applications. However, implanting these biomaterials requires invasive surgery, which is associated with complications such as inflammation, scarring, and infection. To address these shortcomings, we applied microfluidics-based polymerization to engineer injectable poly(ethylene glycol) microgels of defined size and crosslinked with a protease degradable peptide to allow for triggered release of proteins. The release rate of proteins covalently tethered within the microgel network was tuned by modifying the ratio of degradable to non-degradable crosslinkers, and the released proteins retained full bioactivity. Microgels injected into the dorsum of mice were maintained in the subcutaneous space and degraded within 2 weeks in response to local proteases. Furthermore, controlled release of VEGF from degradable microgels promoted increased vascularization compared to empty microgels or bolus injection of VEGF. Collectively, this study motivates the use of microgels as a viable method for controlled protein delivery in regenerative medicine applications.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Preparaciones de Acción Retardada/metabolismo , Hidrogeles/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Péptido Hidrolasas/metabolismo , Polietilenglicoles/metabolismo , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Inductores de la Angiogénesis/farmacología , Animales , Preparaciones de Acción Retardada/química , Hidrogeles/química , Dispositivos Laboratorio en un Chip , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Polimerizacion , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Sci Adv ; 2(8): e1600188, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27574702

RESUMEN

Growth factors (GFs) are powerful signaling molecules with the potential to drive regenerative strategies, including bone repair and vascularization. However, GFs are typically delivered in soluble format at supraphysiological doses because of rapid clearance and limited therapeutic impact. These high doses have serious side effects and are expensive. Although it is well established that GF interactions with extracellular matrix proteins such as fibronectin control GF presentation and activity, a translation-ready approach to unlocking GF potential has not been realized. We demonstrate a simple, robust, and controlled material-based approach to enhance the activity of GFs during tissue healing. The underlying mechanism is based on spontaneous fibrillar organization of fibronectin driven by adsorption onto the polymer poly(ethyl acrylate). Fibrillar fibronectin on this polymer, but not a globular conformation obtained on control polymers, promotes synergistic presentation of integrin-binding sites and bound bone morphogenetic protein 2 (BMP-2), which enhances mesenchymal stem cell osteogenesis in vitro and drives full regeneration of a nonhealing bone defect in vivo at low GF concentrations. This simple and translatable technology could unlock the full regenerative potential of GF therapies while improving safety and cost-effectiveness.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Fibronectinas/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Osteogénesis/efectos de los fármacos , Medicina Regenerativa , Resinas Acrílicas/química , Resinas Acrílicas/uso terapéutico , Sitios de Unión , Proteína Morfogenética Ósea 2/química , Regeneración Ósea/efectos de los fármacos , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Fibronectinas/química , Fibronectinas/genética , Humanos , Integrinas/genética , Integrinas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/genética , Polímeros/uso terapéutico
8.
Biomaterials ; 63: 137-45, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26100343

RESUMEN

Metal implants are widely used to provide structural support and stability in current surgical treatments for bone fractures, spinal fusions, and joint arthroplasties as well as craniofacial and dental applications. Early implant-bone mechanical fixation is an important requirement for the successful performance of such implants. However, adequate osseointegration has been difficult to achieve especially in challenging disease states like osteoporosis due to reduced bone mass and strength. Here, we present a simple coating strategy based on passive adsorption of FN7-10, a recombinant fragment of human fibronectin encompassing the major cell adhesive, integrin-binding site, onto 316-grade stainless steel (SS). FN7-10 coating on SS surfaces promoted α5ß1 integrin-dependent adhesion and osteogenic differentiation of human mesenchymal stem cells. FN7-10-coated SS screws increased bone-implant mechanical fixation compared to uncoated screws by 30% and 45% at 1 and 3 months, respectively, in healthy rats. Importantly, FN7-10 coating significantly enhanced bone-screw fixation by 57% and 32% at 1 and 3 months, respectively, and bone-implant ingrowth by 30% at 3 months compared to uncoated screws in osteoporotic rats. These coatings are easy to apply intra-operatively, even to implants with complex geometries and structures, facilitating the potential for rapid translation to clinical settings.


Asunto(s)
Tornillos Óseos , Materiales Biocompatibles Revestidos/química , Fibronectinas/química , Oseointegración , Acero Inoxidable/química , Animales , Adhesión Celular , Diferenciación Celular , Línea Celular , Materiales Biocompatibles Revestidos/metabolismo , Femenino , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Osteoblastos/citología , Osteogénesis , Osteoporosis/patología , Osteoporosis/terapia , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
9.
Semin Ophthalmol ; 30(4): 316-20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24475917

RESUMEN

A 66-year-old woman with breast cancer presented with a painless mass in the left orbit. MRI revealed a well-defined intraconal mass in the temporal quadrant of the orbit. Fifteen months later, a further MRI indicated the mass had grown, displacing the left optic nerve and making contact with the lateral rectus muscle, suggesting its possible intramuscular origin. Despite the clinical and radiological characteristics of the lesion and its slow growth, a PET/CT study was developed because of the history of malignant disease. No metabolic activity of the mass or malignant lesion in other locations was observed. After surgical excision, histopathological examination revealed an abundant myxoid matrix with few spindle-shaped cells and no signs of malignancy. The cells were immunopositive for CD34, positive for S-100 protein, and negative for EMA, actin, and CD57. A diagnosis was made of a nerve sheath myxoma. The orbital location of these tumors is extremely rare.


Asunto(s)
Neurotecoma/diagnóstico , Músculos Oculomotores/inervación , Neoplasias Orbitales/diagnóstico , Anciano , Antígenos CD34/metabolismo , Biomarcadores de Tumor/metabolismo , Femenino , Fluorodesoxiglucosa F18 , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Proteínas de Neoplasias/metabolismo , Neurotecoma/metabolismo , Neurotecoma/cirugía , Neoplasias Orbitales/metabolismo , Neoplasias Orbitales/cirugía , Tomografía de Emisión de Positrones , Proteínas S100/metabolismo , Tomografía Computarizada por Rayos X
10.
Integr Biol (Camb) ; 4(5): 531-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22395101

RESUMEN

A family of polymer substrates which consists of a vinyl backbone chain with the side groups -COO(CH(2))(x)H, with x = 1, 2, 4, was prepared. Substrates with similar chemical groups but decreasing stiffness, characterized by their elastic modulus at 37 °C, as well as surface mobility, characterized by the glass transition temperature, were obtained. We have investigated whether these subtle variations in polymer chemistry lead to alterations in fibronectin (FN) adsorption and mesenchymal stem cell response. The same FN density was adsorbed on every substrate (∼450 ng cm(-2)) although the supramolecular organization of the protein at the material interface, as obtained with AFM, was different for x = 1 and the other two surfaces (x = 2, 4). Consequently, this allows one to investigate the effect of physical properties of the matrix on stem cell differentiation after ruling out any influence of protein activity. Cell adhesion was quantified by calculating the size distribution of focal adhesions. Mesenchymal stem cell differentiation to the osteoblastic lineage was determined by quantifying protein levels for osteocalcin, osteopontin and Runx2, in the absence of any additional osteogenic soluble factors in the culture media, but as a direct effect of material properties. The findings indicate the potential to modulate skeletal progenitor cell commitment to the osteoblastic lineage through surface mobility of the underlying material surface.


Asunto(s)
Membrana Celular/fisiología , Fluidez de la Membrana/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Adhesión Celular/fisiología , Células Cultivadas , Humanos , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA