Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Tissue Eng Regen Med ; 11(10): 2699-2709, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27225726

RESUMEN

To improve the clinical performance of vascular prostheses, which is inacceptably low for implants with small diameters (< 6 mm), biofunctionalization of synthetic implants by endothelialization has become a major, although still unreached, aim. In order to be able to recruit native endothelial progenitor cells (EPCs) to luminal implant surfaces from the blood stream, we generated monoclonal antibodies against the EPC-specific vascular endothelial growth factor receptor 2 (VEGFR-2). Employing the very efficient genetic immunization strategy, > 10 000 hybridoma clones were generated. Screening with various deletion mutants of VEGFR-2, 49 highly-specific monoclonal antibodies (mAbs) covering all seven Ig domains of VEGFR-2 were selected. mAb 9H10 was characterized in detail. Once immobilized on synthetic surfaces, mAb 9H10 allowed, within min, nearly 100-fold enrichment of VEGFR-2-expressing cells under continuous flow conditions. Cell trapping was cell-type specific and essentially not affected by competing VEGFR-2-negative cells. To exclude that the antibody would adversely modify receptor responses, four different in vitro assays were employed. Cell proliferation, angiogenic tube formation, acetylated low-density lipoprotein uptake and VEGFR-2 phosphorylation remained unaffected, suggesting that the antibody did not interfere with the receptor functioning of human umbilical vascular endothelial cells. The molecular and cellular characteristics make the selected monoclonal antibody a very promising tool for the biofunctionalization of vascular implants. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Prótesis Vascular , Linfocitos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunización , Fosforilación , Dominios Proteicos , Especificidad de la Especie , Sus scrofa , Factor A de Crecimiento Endotelial Vascular
2.
Carbohydr Polym ; 143: 25-34, 2016 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-27083340

RESUMEN

Polyethylene glycol (PEG) is a widely used modification for drug delivery systems. It reduces undesired interaction with biological components, aggregation of complexes and serves as a hydrophilic linker of ligands for targeted drug delivery. However, PEGylation can also lead to undesired changes in physicochemical characteristics of chitosan/siRNA nanoplexes and hamper gene silencing. To address this conflicting issue, PEG-chitosan copolymers were synthesized with stepwise increasing degrees of PEG substitution (1.5% to 8.0%). Subsequently formed PEG-chitosan/siRNA nanoplexes were characterized physicochemically and biologically. The results showed that small ratios of chitosan PEGylation did not affect nanoplex stability and density. However, higher PEGylation ratios reduced nanoplex size and charge, as well as cell uptake and final siRNA knockdown efficiency. Therefore, we recommend fine-tuning of PEGylation ratios to generate PEG-chitosan/siRNA delivery systems with maximum bioactivity. The degree of PEGylation for chitosan/siRNA nanoplexes should be kept low in order to maintain optimal nanoplex efficiency.


Asunto(s)
Quitosano/análogos & derivados , Polietilenglicoles/química , ARN Nuclear Pequeño/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular , Quitosano/síntesis química , Quitosano/química , Portadores de Fármacos , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Concentración de Iones de Hidrógeno , Indicadores y Reactivos , Nanopartículas , Oxazinas/química , Tamaño de la Partícula , Polietilenglicoles/síntesis química , ARN Nuclear Pequeño/química , ARN Nuclear Pequeño/genética , Solubilidad , Xantenos/química
3.
Mol Pharm ; 12(9): 3146-57, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26181293

RESUMEN

Excessive extracellular matrix formation in organs and tissues arises from an imbalance between the synthesis and degradation of matrix proteins, especially collagen. This condition interferes with proper wound healing and regeneration, and to date, no specific treatment is available. In the present study, we propose a targeted drug delivery system consisting of cell-specific immunoliposomes (ILs) loaded with deferoxamine (DFO) as an antifibrotic drug. ILs were functionalized with polyethylene glycol (PEG) to improve the steric stability and prolong their half-life. In addition, a single-chain Fv (scFv) antibody fragment that specifically targets fibroblast activation protein (FAP) was incorporated. An in vitro fibrosis model was employed to test this construct. This model consisted of highly activated pro-fibrotic fibroblasts with 2- to 6-fold induction of selected fibrosis markers: cell/matrix deposited collagen I, total soluble collagen, and α smooth muscle actin. The activation was accompanied by a significant and cell-specific elevation of FAP expression and activity, thereby confirming that FAP is an adequate target for antifibrotic drug delivery. Purified anti-FAP scFv was shown to bind specifically to these cells without influencing the FAP enzymatic activity. DFO was demonstrated to have a dose-dependent antifibrotic activity as quantified by collagen deposition. Specific binding and intracellular uptake of DiI-labeled ILs into the activated fibroblasts were shown by flow cytometry and microscopy. Finally, DFO-loaded ILs targeted to FAP caused a significant reduction in the collagen deposition, whereas no effect was observed using liposomes that lacked the targeting antibody fragment. These results suggest that the FAP-specific scFv-conjugated liposomes have considerable potential for cell-specific targeting applicable as a therapy for excessive collagen deposition during fibrosis. In general, through liposome encapsulation, bioactive molecules, such as DFO, that have broad effects and poor cell penetration can be converted into cell-specific composites for targeted drug delivery.


Asunto(s)
Deferoxamina/administración & dosificación , Fibroblastos/efectos de los fármacos , Fibrosarcoma/tratamiento farmacológico , Gelatinasas/antagonistas & inhibidores , Liposomas/química , Pulmón/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Anticuerpos de Cadena Única/administración & dosificación , Células Cultivadas , Sistemas de Liberación de Medicamentos , Endopeptidasas , Fibroblastos/inmunología , Fibroblastos/patología , Fibrosarcoma/inmunología , Fibrosarcoma/patología , Gelatinasas/inmunología , Semivida , Humanos , Técnicas para Inmunoenzimas , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/inmunología , Liposomas/inmunología , Pulmón/inmunología , Pulmón/patología , Proteínas de la Membrana/inmunología , Polietilenglicoles/química , Serina Endopeptidasas/inmunología , Sideróforos/administración & dosificación , Anticuerpos de Cadena Única/inmunología
4.
Cell Tissue Res ; 362(1): 9-20, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25920589

RESUMEN

Lesions of the central nervous system elicit inflammatory responses that counteract the regeneration of neurites. Microglia and infiltrating macrophages that were activated by trauma have been identified as cellular sources of inhibitory factors. We examine cultured macrophage (RAW264.7) and neuronal (PC12) cell lines to ascertain the potential modulators of the inflammatory impact on neurons. By exposing quiescent macrophages to lipopolysaccharide (LPS) and interferon γ (IFN-γ), cells can be transformed into an activated M1 phenotype. Neurite extension was induced in PC12 cells by culturing them in the presence of nerve growth factor. Neurite outgrowth was quantified by analyzing immunofluorescence and phase contrast microscopy images. Activated macrophages significantly reduced neurite extension. Macrophage activation by LPS/IFN-γ induced a 1000-fold increase in tumor necrosis factor alpha (TNF-α) secretion, as quantified by enzyme-linked immunosorbent assays (ELISA). Recombinant TNF-α inhibited neurite formation at concentrations as low as 0.016 ng/ml. In contrast, the masking of TNF-α with specific functional antibodies abrogated neurite growth inhibition by activated macrophages. Taken together, these results indicated that TNF-α is a key component of inhibitory macrophage action. The transfection of PC12 neurons with microRNA-124 (miR-124) counteracted the inhibition of neurites mediated by both recombinant TNF-α and macrophages. miR-124 did not stimulate neurite formation per se, nor was cell viability affected. These data suggest that miR-124 might be a valuable tool for desensitizing neurons to a repulsive inflammatory environment.


Asunto(s)
Inflamación/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Neuritas/metabolismo , Animales , Inflamación/genética , Inflamación/patología , Ratones , MicroARNs/genética , Neuritas/patología , Células PC12 , Células RAW 264.7 , Ratas , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
PLoS One ; 9(1): e86740, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24475174

RESUMEN

In the field of regenerative medicine we aim to develop implant matrices for specific tissue needs. By combining two per se, cell-permissive gel systems with enzymatic crosslinkers (gelatin/transglutaminase and fibrinogen/thrombin) to generate a blend (technical term: quattroGel), an unexpected cell-selectivity evolved. QuattroGels were porous and formed cavities in the cell diameter range, possessed gelation kinetics in the minute range, viscoelastic properties and a mechanical strength appropriate for general cell adhesion, and restricted diffusion. Cell proliferation of endothelial cells, chondrocytes and fibroblasts was essentially unaffected. In contrast, on quattroGels neither endothelial cells formed vascular tubes nor did primary neurons extend neurites in significant amounts. Only chondrocytes differentiated properly as judged by collagen isoform expression. The biophysical quattroGel characteristics appeared to leave distinct cell processes such as mitosis unaffected and favored differentiation of sessile cells, but hampered differentiation of migratory cells. This cell-type selectivity is of interest e.g. during articular cartilage or invertebral disc repair, where pathological innervation and angiogenesis represent adverse events in tissue engineering.


Asunto(s)
Células/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Medicina Regenerativa/instrumentación , Medicina Regenerativa/métodos , Andamios del Tejido/química , Sustancias Viscoelásticas/química , Análisis de Varianza , Animales , Fenómenos Biomecánicos , Bromodesoxiuridina , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Reactivos de Enlaces Cruzados/metabolismo , Cartilla de ADN/genética , Difusión , Perfilación de la Expresión Génica , Humanos , Cinética , Ratones , Concentración Osmolar , Ratas , Medicina Regenerativa/tendencias , Reología , Ingeniería de Tejidos/métodos
6.
J Control Release ; 168(3): 289-97, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23562632

RESUMEN

Binding, stabilizing and promoting cellular uptake of siRNA are all critical efforts in creating matrices for the localized delivery of siRNA molecules to target cells. In this study, we describe the generation of chitosan imidazole/siRNA nanoplexes (NPs) embedded in nano scope polyelectrolyte multilayers (PEMs) composed of hyaluronic acid and chitosan for sustained and localized drug delivery. Regular PEM build-up, successful integration of NPs and controlled release under physiological conditions were shown. Biological efficacy was evaluated in neuronal cell culture concerning cell adhesion, viability, NPs uptake and gene silencing. The additionally shown biological functionalization of neuronal implants possesses potential for future applications in the field of regenerative medicine and treatment of spinal cord injuries.


Asunto(s)
Quitosano/química , Ácido Hialurónico/química , Imidazoles/química , Nanoestructuras/administración & dosificación , Prótesis e Implantes , ARN Interferente Pequeño/administración & dosificación , Animales , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Nanoestructuras/química , Neuronas , Células PC12 , ARN Interferente Pequeño/química , Ratas , Proteína de Unión al GTP rhoA/genética
7.
Circulation ; 125(5): 685-96, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22223428

RESUMEN

BACKGROUND: CXCR4-positive bone marrow cells (BMCs) are critically involved in cardiac repair mechanisms contributing to preserved cardiac function. Stromal cell-derived factor-1 (SDF-1) is the most prominent BMC homing factor known to augment BMC engraftment, which is a limiting step of stem cell-based therapy. After myocardial infarction, SDF-1 expression is rapidly upregulated and promotes myocardial repair. METHODS AND RESULTS: We have established a bifunctional protein consisting of an SDF-1 domain and a glycoprotein VI (GPVI) domain with high binding affinity to the SDF-1 receptor CXCR4 and extracellular matrix proteins that become exposed after tissue injury. SDF1-GPVI triggers chemotaxis of CXCR4-positive cells, preserves cell survival, enhances endothelial differentiation of BMCs in vitro, and reveals proangiogenic effects in ovo. In a mouse model of myocardial infarction, administration of the bifunctional protein leads to enhanced recruitment of BMCs, increases capillary density, reduces infarct size, and preserves cardiac function. CONCLUSIONS: These results indicate that administration of SDF1-GPVI may be a promising strategy to treat myocardial infarction to promote myocardial repair and to preserve cardiac function.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Quimiocina CXCL12/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Ataque Isquémico Transitorio/terapia , Infarto del Miocardio/terapia , Glicoproteínas de Membrana Plaquetaria/farmacología , Proteínas Recombinantes de Fusión/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/uso terapéutico , Colágeno/metabolismo , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/fisiopatología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Neovascularización Fisiológica/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/uso terapéutico , Unión Proteica , Receptores CXCR4/metabolismo , Resultado del Tratamiento
8.
Exp Biol Med (Maywood) ; 236(6): 692-700, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21558092

RESUMEN

Telomerase reverse transcriptase (TERT) is fundamental in determining the life span by regulating telomere length of chromosomes. To address the question whether the enhancement of the proliferative potential hampers cell differentiation, we generated TERT-over-expressing endothelial cells (ECs) and analyzed in vitro their (1) barrier function; (2) low-density lipoprotein uptake; (3) expression pattern of six selected marker proteins; (4) angiogenic potential in four assays; and (5) inflammatory responses. In contrast to investigations with focus on other cell parameters, we demonstrate that immortalization of ECs by over-expression of TERT resulted in different angiogenic and inflammatory behavior in comparison to cells with low native telomerase levels.


Asunto(s)
Células Endoteliales/enzimología , Células Endoteliales/fisiología , Telomerasa/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Lipoproteínas LDL/metabolismo , Neovascularización Fisiológica , Telomerasa/genética
9.
Ann Anat ; 193(4): 341-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21489766

RESUMEN

Different bioartificial tubes were recommended for peripheral nerve reconstruction in the past. In order to replace autologous nerve grafts this materials are still under review in different animal studies. Most of them are dealing with the rodent peripheral nerves. One very popular animal model to study different materials is the rat median nerve model. With its easy excess, simple behavioral tests and reliable long term results it is attractive to many scientists in this field. This review gives an overview about the past, current and future options in this model for bioartificial nerve tubes. It summarizes the evolution of successful implantation of different materials across short nerve gaps and demonstrates the obstacles arising from long nerve gaps and the problems associated to them.


Asunto(s)
Órganos Bioartificiales , Regeneración Tisular Dirigida/instrumentación , Nervio Mediano/cirugía , Regeneración Nerviosa/fisiología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Regeneración Tisular Dirigida/métodos , Nervio Mediano/lesiones , Microcirugia/métodos , Modelos Animales , Ratas
10.
Neurosci Lett ; 494(1): 14-8, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21352894

RESUMEN

The manipulation of gene expression by RNA interference could play a key role in future neurotherapies, for example in the development of biohydrid implants to bridge nerve and spinal cord lesion gaps. Such resorbable biomaterial prostheses could serve as growth substrates together with specific siRNA to foster neuronal regeneration. To the best of our knowledge, we are the first to biofunctionalize neuronal prostheses with siRNA. We analyzed neuronal and Schwann cell responses to scrambled siRNA coated polydioxanone polymer filaments designed to imitate pro-regenerative bands of Büngner for oriented axonal regrowth. With a view to future clinical applications we were especially interested in potentially detrimental side effects. We employed a variety of in vitro methods, including a novel impedance electrode microchamber assay, fluorescence and scanning electron microscopy, metabolic labeling and RT-PCR. We found that the application of chitosan/siRNA nanoparticles (1) did not affect glial cell motility or (2) axonal growth in contrast to other formulations, (3) only slightly reduced proliferation, and (4) did not induce inflammatory responses that might hamper axonal regeneration. The data suggest that chitosan/siRNA nanoparticle-coated polymer filaments are suitable for use in biohybrid implants with no significant side effects on neuronal and glial cells.


Asunto(s)
Axones/fisiología , Neuronas/fisiología , ARN Interferente Pequeño/administración & dosificación , Análisis de Varianza , Animales , Materiales Biocompatibles , Quitosano , Inmunohistoquímica , Nanopartículas , ARN Interferente Pequeño/genética , Ratas , Ratas Endogámicas Lew , Células de Schwann/fisiología
11.
Neurosci Lett ; 484(2): 118-22, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20723580

RESUMEN

Nerve guide implants approved for human application in the peripheral nervous system generally fail to bridge lesion gaps longer than 2 cm and cannot match the clinical performance of autologous nerve transplants. Since current synthetic implants are simply hollow tubes, we aim to recreate the native microarchitecture of nerves inside the tubular implants. Most importantly, in the regenerating nerve, dedifferentiated Schwann cells align to form thousands of long glial strands, which act as guiding structures for the regrowing axons. In order to artificially induce the formation of Schwann cell strands, 28 µm thick, endless poly-p-dioxanone filaments (PDO) were synthesized with longitudinal grooves. A polycationic coating on the PDO filaments rendered the polymer surface cell-permissive and induced the growth of highly oriented Schwann cells with polarized expression of N-cadherin at cell-cell contact sites. In vitro cell proliferation on three-dimensional PDO filaments was significantly increased in comparison to planar PDO substrates. Time lapse video recordings revealed high Schwann cell motility, which is advantageous for the repopulation of cell-free implants after implantation. In a pilot study we employed a novel microsurgical technique in vivo. All axon fascicles were selectively dissected from sciatic rat nerves, and the remaining epineural tube was filled with hundreds of PDO filaments. Histological analysis 6 weeks postoperatively showed no fibrosis or encapsulation but instead longitudinal cell alignment and axonal regrowth. The data suggest that the addition of microstructured PDO filaments to the lumen of synthetic tubular implants might significantly improve performance.


Asunto(s)
Bioingeniería/métodos , Regeneración Nerviosa/fisiología , Células de Schwann/fisiología , Neuropatía Ciática/fisiopatología , Neuropatía Ciática/cirugía , Animales , Materiales Biocompatibles/uso terapéutico , Bromodesoxiuridina/metabolismo , Movimiento Celular/fisiología , Proliferación Celular , Dioxanos/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Indoles , Polímeros/uso terapéutico , Ratas , Ratas Endogámicas Lew , Células de Schwann/trasplante , Factores de Tiempo , Grabación en Video/métodos
12.
Nano Lett ; 10(10): 3933-9, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20795625

RESUMEN

Microstructured 20 µm thick polymer filaments used as nerve implants were loaded with chitosan/siRNA nanoparticles to promote nerve regeneration and ensure local delivery of nanotherapeutics. The stable nanoparticles were rapidly internalized by cells and did not affect cell viability. Target mRNA was successfully reduced by 65-75% and neurite outgrowth was enhanced even in an inhibitory environment. This work, thus, supports the application of nanobiofunctionalized implants as a novel approach for spinal cord and nerve repair.


Asunto(s)
Quitosano/química , Nanopartículas/química , Neuronas/citología , Prótesis e Implantes , ARN Interferente Pequeño/administración & dosificación , Animales , Línea Celular , Nanopartículas/ultraestructura , Regeneración Nerviosa , Neuritas/metabolismo , Neuronas/metabolismo , Prótesis e Implantes/ultraestructura , ARN Interferente Pequeño/genética , Transfección , Proteína de Unión al GTP rhoA/genética
13.
J Biomed Mater Res B Appl Biomater ; 94(2): 372-379, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20578223

RESUMEN

To more effectively manage the substantial bleeding encountered during surgical procedures in oto-rhino-laryngology, we developed a novel hemostatic sponge made of pharmaceutical grade, chemically cross-linked gelatin. The sponge is characterized by a high pore density, reduced ligaments, and a high nanoscale roughness of lamella surfaces in the matrix. In vitro blood uptake assays revealed a very rapid absorption of human blood, which was two to three times faster than that measured with comparative hemostyptic devices. In an in vitro hemorrhage model using human veins, the novel gelatin sponge matrix induced hemostasis less than a minute after bleeding was induced. The sponge was shown to bring about rapid hemostasis when it was administered in a young patient suffering from acute bleeding of a pharyngeal angiofibroma, even though the patient had been treated with an anticoagulant because of a transient ischemic attack. As the gelatin matrix of the sponge is biocompatible and resorbable, the hemostyptic device could be left in place and was shown to be resorbed within 2 weeks. We hypothesize that the excellent hemostatic performance of the sponge might be linked to enhanced capillary effects in conjunction with optimized anchoring of fibrinogen on the nano-rough material surface, as suggested by scanning electron microscopy. The novel gelatin sponge appears to be a promising hemostatic matrix, which could be of great benefit for patients suffering from epistaxis and other acute injuries resulting in severe bleeding.


Asunto(s)
Esponja de Gelatina Absorbible/uso terapéutico , Hemostasis , Absorción , Angiofibroma/irrigación sanguínea , Angiofibroma/patología , Sangre , Pérdida de Sangre Quirúrgica/prevención & control , Niño , Hemorragia/prevención & control , Hemostasis Quirúrgica/métodos , Humanos , Masculino , Venas
14.
J Neurosci Methods ; 189(2): 205-9, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20398700

RESUMEN

A novel epineural tube implantation paradigm in the adult rat was designed for the analysis of regulatory cell interactions in peripheral nerves and for the development of therapeutic implants. The aim was to allow the integration of synthetic regenerative structures and cells into the nerve interior while preserving an outer nerve tissue layer with a supportive vasculature. The microsurgical technique allowed us to remove the interfascicular epineurium, leaving behind an epineural tube with an intact tissue wall of about 0.1-0.2mm. The resulting tube was filled with hundreds of bioengineered bands of Büngner which were composed of resorbable polymer filaments seeded with Schwann cells. Alternatively, purified cells to be analyzed or different types of growth matrices were injected into the epineural tube. Such manipulations will allow to generate and investigate concentration gradients of biological factors or to analyse cell-matrix interactions under defined conditions in a supportive in vivo environment. Our current aim is to evaluate bioengineered neural implants. In summary, a microsurgical in vivo paradigm has been developed to address multiple aspects of peripheral nerve regeneration.


Asunto(s)
Implantes Absorbibles , Regeneración Tisular Dirigida/métodos , Microcirugia/métodos , Procedimientos Neuroquirúrgicos/métodos , Nervios Periféricos/cirugía , Andamios del Tejido , Animales , Materiales Biocompatibles , Femenino , Geles , Regeneración Nerviosa , Polímeros , Ratas , Ratas Endogámicas Lew , Células de Schwann/trasplante
15.
Cell Mol Life Sci ; 67(14): 2451-65, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20352467

RESUMEN

By counteracting the shortening of chromosome telomeres, telomerase reverse transcriptase (hTERT) prevents senescence and age-related cell death. Embryonic cells display a high telomerase activity that declines rapidly with cell differentiation. Conversely, de-differentiated tumor cells tend to re-express telomerase. In view of the controversial data on the reciprocal correlation between cell proliferation and differentiation, we questioned whether telomerase overexpression and the resulting immortalization would affect the functional phenotype of human endothelial cells. Our comparative analysis addressed (1) distinct cell adhesion to different ECM-proteins analyzed on miniaturized multisubstrate arrays (MSA), (2) protein expression of diverse markers, (3) the uptake of DiI-Ac-LDL, (4) the inflammatory response based on upregulation of ICAM-1, (5) tube formation, and (6) the barrier properties of cell monolayers in transfilter cultures. Our results, based on some 40 data sets, demonstrate that immortalization of primary endothelial cells by hTERT maintains the typical endothelial characteristics without any sign of functional de-differentiation.


Asunto(s)
Desdiferenciación Celular , Células Endoteliales/citología , Células Endoteliales/fisiología , Telomerasa/metabolismo , Carbocianinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Transformación Celular Neoplásica , Células Cultivadas , Clonación Molecular , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/inmunología , Lipoproteínas LDL/metabolismo , Permeabilidad , Proteínas/metabolismo , Telomerasa/genética , Transducción Genética , Factor de Necrosis Tumoral alfa/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de von Willebrand/metabolismo
16.
Biomaterials ; 30(29): 5251-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19632717

RESUMEN

Peripheral human nerves fail to regenerate across longer tube implants (>2 cm), most likely because implants lack the microarchitecture of native nerves, including bands of Büngner. Bands of Büngner comprise longitudinally aligned Schwann cell strands that guide selectively regrowing axons. We aim to optimize tubular implants by integrating artificial bands of Büngner. Three principle strategies for inducing the formation of bands of Büngner were investigated: (a) an aligned extracellular matrix, (b) polarizing differentiation factors, and (c) microstructured biomaterial filaments. In vitro oriented collagen and a combination of differentiation factors (NGF, neuregulin-1, TGF-beta) induced Schwann cell alignment to some extent. The most pronounced Schwann cell alignment was evident on ultrathin, endless poly-epsilon-caprolactone (PCL) filaments with longitudinal microgrooves. Precoated PCL filaments proved to be non-cytotoxic, displayed good cell attachment, and supported Schwann cell proliferation as well as guided axonal outgrowth. In vitro on PCL filaments Schwann cells displayed a polarized expression of the cell adhesion molecule L1 similar to that seen in vivo in bands of Büngner after sciatic nerve crush in adult rats. In summary, the integration of bioengineered bands of Büngner based on microstructured polymer filaments in nerve conduits promises to be the most valuable approach to initiating a more efficient regeneration across longer nerve lesions.


Asunto(s)
Regeneración Tisular Dirigida/métodos , Regeneración Nerviosa/fisiología , Células de Schwann/fisiología , Células de Schwann/trasplante , Neuropatía Ciática/patología , Neuropatía Ciática/cirugía , Ingeniería de Tejidos/métodos , Animales , Femenino , Ratas , Ratas Endogámicas Lew , Células de Schwann/citología , Resultado del Tratamiento
17.
Eur J Cell Biol ; 88(5): 285-300, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19246119

RESUMEN

In order to reveal non-neuronal cell interactions after peripheral nerve lesions, we began to analyze the impact of sciatic nerve fibroblasts on Schwann cells in vitro. Both cell types are considered to have opposite effects on axonal regeneration. Few data are available on how repulsive nerve fibroblasts affect neuritotrophic Schwann cells and thus might indirectly influence axonal regrowth. Using different culture systems in conjunction with time-lapse video recording, metabolic labeling, pharmacological intervention, RNAi knockdown, Western blotting and RT-PCR analysis, we found that nerve fibroblasts differentially modify the various responses of Schwann cells. In the presence of collagen type IV and heparan sulfate proteoglycan but not of laminin, diffusible fibroblast factors slow down Schwann cell proliferation. In contrast, fibroblast factors increase the migratory activity of Schwann cells without being chemoattractive. One pro-migratory fibroblast factor turned out to be neuregulin. The pro-migratory activity of nerve fibroblasts and of recombinant neuregulin-1beta1 can be counteracted by neuregulin-specific pharmacological intervention and by neuregulin RNA interference. We show for the first time that nerve fibroblasts play antagonistic and agonistic roles for Schwann cells in a context-dependent manner. The data shed light on cellular mechanisms and have implications for some neuro-tissue engineering strategies.


Asunto(s)
Comunicación Celular/fisiología , Fibroblastos/citología , Células de Schwann/citología , Nervio Ciático/citología , Animales , Axones/fisiología , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Silenciador del Gen , Regeneración Nerviosa , Neurregulina-1/biosíntesis , Neurregulina-1/genética , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , ARN Interferente Pequeño , Ratas , Ratas Endogámicas Lew , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Transducción de Señal
18.
Cell Transplant ; 17(5): 543-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18714673

RESUMEN

The quantity of therapeutic gene products released from genetically engineered cells can be controlled externally at different levels. The widely used approach of controlling expression, however, generally has the disadvantage that chemical substances must be applied for stimulation. An alternative strategy aims at controlling gene products at posttranslational levels such as secretion. The secretion of a therapeutic agent can be regulated if the agent is targeted to the regulated secretory pathway and stored in the secretory granules until its release. In this article we address the question of whether the release of beta-endorphin, an opioid with a potent analgesic effect, could be induced by electrically stimulating stably transfected Neuro-2a cells. Throughout this study we used the human proopiomelanocortin (POMC) gene, which is the precursor molecule for human beta-endorphin. We analyzed its subcellular localization and found it in the regulated secretory pathway in Neuro-2a cells. Using electrical field stimulation we were able to identify a stimulation pattern that significantly increased the release of beta-endorphin-immunoreactive material, although to a limited extent. This result indicates that electrical stimulation of secretion could be used to manipulate the amount of a therapeutic agent released from transplanted cells.


Asunto(s)
Neuronas/metabolismo , Vías Secretoras/fisiología , betaendorfina/metabolismo , Línea Celular , Trasplante de Células/métodos , Estimulación Eléctrica , Ingeniería Genética , Humanos , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Procesamiento Proteico-Postraduccional , Ingeniería de Tejidos , betaendorfina/genética
19.
Biomaterials ; 28(36): 5536-43, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17889331

RESUMEN

To overcome limitations on regeneration in the nervous system and other organs caused by insufficient blood supply, we have developed a gelatin sponge material which stimulates blood vessel formation, i.e. angiogenesis. Controlled chemical cross-linking was employed to slow down enzymatic degradation of the gelatin matrix. Four different in vitro assays using L929 fibroblasts and purified endothelial cells indicated that the sponge material did not release toxic components, but provided a permissive substratum for cell attachment, cell migration and pronounced cell proliferation, all of which are crucial for the formation of vasculature. Two in vivo models were employed to directly monitor the pro-angiogenic impact of the sponge material. Implantation of gelatin sponges onto the chorioallantoic membrane of fertilized chicken eggs induced robust attraction of endothelial cells and formation of blood vessels. Angiogenesis inside gelatin implants occurred more than 200 times faster than in a commercial collagen sponge. Similarly, after subcutaneous implantation of tube-like sponges into mice, an increasing immigration of cells and subsequent formation of functional vasculature became evident. Immunocytochemistry revealed no fibronection accumulation and no scarring. In summary, our matrix based on cross-linked gelatin promises to be a valuable component of future implants, improving neuronal and non-neuronal regeneration by concomitant pro-angiogenic stimulation.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Gelatina/química , Gelatina/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Bovinos , Células Cultivadas , Pollos , Gelatina/metabolismo , Ensayo de Materiales , Ratones , Modelos Animales
20.
Restor Neurol Neurosci ; 25(2): 131-41, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17726272

RESUMEN

PURPOSE: Recently we successfully used a conduit of epsilon-caprolactone-co-trimethylene carbonate filled with Schwann cells (SC) across a 20 mm gap in a rat median nerve. In this study we applied the tubes with SC across a 40 mm gap in order to analyse the regenerative potential of the tubes in long nerve defects. METHODS: To augment the nerve defect a cross-chest procedure was used and the tubes were implanted with injected isogeneic SCs inside (group 3). Both ulnar nerves were used for a 40 mm autograft (group 2). For control group non-operated animals were used (group 1). The grasping test, histology (S-100, PAM), electrophysiology, and the muscle weight were used to assess regeneration. RESULTS: After 12 months, grasping was seen only in three animals of group 3 (3.6 g [95% CI: 0 to 7.6 g]). However, in group 2 all rats had a partial functional regeneration (42.8 g [95% CI: 39.1 to 46.6 g]). The grasping force of the non-operated animals (group 1) was 240.9 g [95% CI: 237.2 to 244.7 g] at the time. Histology from group 3 confirmed an irregular arrangement of fibres in contrast to more organized structures in group 2. Electrophysiology in group 3 displayed potentials only in the three animals with functional regeneration. In group 2 all animals exhibited potentials. A significant decrease of muscle weight was observed in groups 2 and 3, most prominent in the latter. CONCLUSION: Regeneration was not successful across the 40 mm gap using the applied tube in combination with SC. For future experiments further consideration should be taken in optimizing the cellular and material components that are critical for a successful application to overcome very large nerve gaps.


Asunto(s)
Órganos Bioartificiales , Regeneración Nerviosa/fisiología , Vías Nerviosas , Células de Schwann/fisiología , Potenciales de Acción , Animales , Axones/ultraestructura , Electrofisiología , Diseño de Equipo , Femenino , Pie , Miembro Anterior , Nervio Mediano/fisiología , Nervio Mediano/cirugía , Nervio Mediano/ultraestructura , Músculo Esquelético/anatomía & histología , Músculo Esquelético/inervación , Músculo Esquelético/fisiología , Fibras Nerviosas/ultraestructura , Tamaño de los Órganos , Desempeño Psicomotor , Ratas , Ratas Endogámicas Lew , Tiempo de Reacción , Trasplante Autólogo , Nervio Cubital/fisiología , Nervio Cubital/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...