Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neural Plast ; 2016: 3679545, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057361

RESUMEN

During postnatal development, closure of critical periods coincides with the appearance of extracellular matrix structures, called perineuronal nets (PNN), around various neuronal populations throughout the brain. The absence or presence of PNN strongly correlates with neuronal plasticity. It is not clear how PNN regulate plasticity. The repulsive axon guidance proteins Semaphorin (Sema) 3A and Sema3B are also prominently expressed in the postnatal and adult brain. In the neocortex, Sema3A accumulates in the PNN that form around parvalbumin positive inhibitory interneurons during the closure of critical periods. Sema3A interacts with high-affinity with chondroitin sulfate E, a component of PNN. The localization of Sema3A in PNN and its inhibitory effects on developing neurites are intriguing features and may clarify how PNN mediate structural neural plasticity. In the cerebellum, enhanced neuronal plasticity as a result of an enriched environment correlates with reduced Sema3A expression in PNN. Here, we first review the distribution of Sema3A and Sema3B expression in the rat brain and the biochemical interaction of Sema3A with PNN. Subsequently, we review what is known so far about functional correlates of changes in Sema3A expression in PNN. Finally, we propose a model of how Semaphorins in the PNN may influence local connectivity.


Asunto(s)
Matriz Extracelular/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Semaforina-3A/metabolismo , Animales , Proteínas de la Matriz Extracelular/metabolismo , Ratas
2.
Gene Ther ; 22(10): 767-80, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25938190

RESUMEN

Schwann cells (SCs) in an injured peripheral nerve form pathways for regenerating axons. Although these cells initially support regeneration, SCs lose their pro-regenerative properties following a prolonged period of denervation. Gene transfer to SC can enhance their therapeutic potential. In this article, we compared adeno-associated viral (AAV) vectors based on serotypes 1-9 for their capability to transduce cultured primary rat and human SCs and nerve segments. AAV1 is the best serotype to transduce rat SCs, whereas AAV2 and AAV6 performed equally well in human SCs. Transduction of monolayers of cultured rat and human SCs did not accurately predict the transduction efficiency in nerve segments. Rat nerve segments could be genetically modified equally well by a set of four AAV vectors (AAV1, AAV5, AAV7, AAV9), whereas AAV2 was superior in human nerve segments. The current experiments were undertaken as a first step towards future clinical implementation of ex vivo AAV-based gene therapy in surgical nerve repair. The transduction of rat and human SCs and nerve segments by entirely different AAV serotypes, as documented here, highlights one of the challenges of translating gene therapy from experimental animals to human patients.


Asunto(s)
Dependovirus , Terapia Genética , Vectores Genéticos , Lentivirus , Células de Schwann/fisiología , Transducción Genética/métodos , Animales , Células Cultivadas , Humanos , Traumatismos de los Nervios Periféricos/terapia , Ratas , Células de Schwann/trasplante
3.
Gene Ther ; 21(3): 242-52, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24285216

RESUMEN

Adeno-associated viral (AAV) vectors based on serotype 5 are an efficient means to target dorsal root ganglia (DRG) to study gene function in the primary sensory neurons of the peripheral nervous system. In this study, we have developed a compact AAV dual promoter vector composed of the cytomegalovirus (CMV) and chicken beta-actin (CAG) promoters in a back-to-back configuration with a shared enhancer, and show efficient expression of two proteins simultaneously in DRG neurons. We demonstrate how this is useful for experiments on axonal regeneration, by co-expressing a gene of interest and an axonal marker. Using a farnesylated form of eGFP, which is actively transported along axons, we show superior long-distance labelling of axons of DRG neurons compared with normal eGFP. Additionally, we have efficiently transduced lumbar DRG neurons by injecting the AAV dual promoter vector into the dorsal intrathecal space, which is a less invasive delivery method. In summary, we have developed an AAV dual promoter vector designed for simultaneous expression of a gene of interest and a fluorescent protein to label long-distance axonal projections, which allows specific quantification of axons from transduced neurons after injury.


Asunto(s)
Axones/metabolismo , Dependovirus/genética , Ganglios Espinales/metabolismo , Regiones Promotoras Genéticas , Actinas/genética , Animales , Células Cultivadas , Pollos , Citomegalovirus/genética , Dependovirus/metabolismo , Ganglios Espinales/citología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inyecciones Espinales , Ratas , Ratas Endogámicas F344
4.
Gene Ther ; 21(6): 549-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24694534

RESUMEN

Viral vector-mediated gene transfer of neurotrophic factors is an emerging and promising strategy to promote the regeneration of injured peripheral nerves. Unfortunately, the chronic exposure to neurotrophic factors results in local trapping of regenerating axons or other unwanted side effects. Therefore, tight control of therapeutic gene expression is required. The tetracycline/doxycycline-inducible system is considered to be one of the most promising systems for regulating heterologous gene expression. However, an immune response directed against the transactivator protein rtTA hampers further translational studies. Immunogenic proteins fused with the Gly-Ala repeat of the Epstein-Barr virus Nuclear Antigen-1 protein have been shown to successfully evade the immune system. In this article, we used this strategy to demonstrate that a chimeric transactivator, created by fusing the Gly-Ala repeat with rtTA and embedded in a lentiviral vector (i) retained its transactivator function in vitro, in muscle explants, and in vivo following injection into the rat peripheral nerve, (ii) exhibited a reduced leaky expression, and (iii) had an immune-evasive advantage over rtTA as shown in a novel bioassay for human antigen presentation. The current findings are an important step toward creating a clinically applicable potentially immune-evasive tetracycline-regulatable viral vector system.


Asunto(s)
Vectores Genéticos/farmacología , Nervios Periféricos/efectos de los fármacos , Tetraciclina/farmacología , Animales , Secuencia de Bases , Femenino , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Células HEK293 , Humanos , Técnicas In Vitro , Lentivirus/genética , Datos de Secuencia Molecular , Músculo Esquelético/fisiología , Ratas Wistar , Linfocitos T Citotóxicos/inmunología , Transactivadores/genética , Transactivadores/metabolismo
5.
Mol Psychiatry ; 18(9): 993-1005, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22925833

RESUMEN

Glucocorticoids (GCs) secreted after stress reduce adult hippocampal neurogenesis, a process that has been implicated in cognitive aspects of psychopathology, amongst others. Yet, the exact role of the GC receptor (GR), a key mediator of GC action, in regulating adult neurogenesis is largely unknown. Here, we show that GR knockdown, selectively in newborn cells of the hippocampal neurogenic niche, accelerates their neuronal differentiation and migration. Strikingly, GR knockdown induced ectopic positioning of a subset of the new granule cells, altered their dendritic complexity and increased their number of mature dendritic spines and mossy fiber boutons. Consistent with the increase in synaptic contacts, cells with GR knockdown exhibit increased basal excitability parallel to impaired contextual freezing during fear conditioning. Together, our data demonstrate a key role for the GR in newborn hippocampal cells in mediating their synaptic connectivity and structural as well as functional integration into mature hippocampal circuits involved in fear memory consolidation.


Asunto(s)
Hipocampo/citología , Motivación/genética , Neurogénesis/genética , Neuronas/fisiología , Receptores de Glucocorticoides/deficiencia , Animales , Movimiento Celular/genética , Condicionamiento Clásico/fisiología , Corticosterona/metabolismo , Dendritas/metabolismo , Dendritas/ultraestructura , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Miedo , Vectores Genéticos/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Terminales Presinápticos/metabolismo , ARN Interferente Pequeño/metabolismo , Radioinmunoensayo
6.
Gene Ther ; 19(1): 49-60, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21562590

RESUMEN

The corticospinal tract (CST) is extensively used as a model system for assessing potential therapies to enhance neuronal regeneration and functional recovery following spinal cord injury (SCI). However, efficient transduction of the CST is challenging and remains to be optimised. Recombinant adeno-associated viral (AAV) vectors and integration-deficient lentiviral vectors are promising therapeutic delivery systems for gene therapy to the central nervous system (CNS). In the present study the cellular tropism and transduction efficiency of seven AAV vector serotypes (AAV1, 2, 3, 4, 5, 6, 8) and an integration-deficient lentiviral vector were assessed for their ability to transduce corticospinal neurons (CSNs) following intracortical injection. AAV1 was identified as the optimal serotype for transducing cortical and CSNs with green fluorescent protein (GFP) expression detectable in fibres projecting through the dorsal CST (dCST) of the cervical spinal cord. In contrast, AAV3 and AAV4 demonstrated a low efficacy for transducing CNS cells and AAV8 presented a potential tropism for oligodendrocytes. Furthermore, it was shown that neither AAV nor lentiviral vectors generate a significant microglial response. The identification of AAV1 as the optimal serotype for transducing CSNs should facilitate the design of future gene therapy strategies targeting the CST for the treatment of SCI.


Asunto(s)
Dependovirus/metabolismo , Vectores Genéticos/metabolismo , Lentivirus/metabolismo , Tractos Piramidales/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Recuento de Células , Dependovirus/clasificación , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Lentivirus/genética , Microglía/citología , Microglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Tractos Piramidales/citología , Ratas , Ratas Sprague-Dawley , Serotipificación , Transfección/métodos , Integración Viral
7.
Mol Cell Neurosci ; 46(2): 507-15, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21145973

RESUMEN

Intravitreal injections of recombinant ciliary neurotrophic factor (rCNTF) protect adult rat retinal ganglion cells (RGCs) after injury and stimulate regeneration, an effect enhanced by co-injection with a cAMP analogue (CPT-cAMP). This effect is partly mediated by PKA and associated signaling pathways, but CPT-cAMP also moderates upregulation of suppressor of cytokine signaling (SOCS) pathways after rCNTF injection, which will also enhance the responsiveness of RGCs to this and perhaps other cytokines. We now report that intravitreal injections of CPT-cAMP do not potentiate RGC axonal regeneration when CNTF is expressed via an adeno-associated viral vector (rAAV2), and concomitantly we show that increases in retinal SOCS mRNA expression are less when CNTF is delivered using the vector. We also directly tested the impact of elevated SOCS3 expression on the survival and regeneration of injured adult RGCs by injecting a bicistronic rAAV2-SOCS3-GFP vector into the vitreous of eyes in rats with a peripheral nerve graft sutured onto the cut optic nerve. Overexpression of SOCS3 resulted in an overall reduction in axonal regrowth and almost complete regeneration failure of RGCs transduced with the rAAV2-SOCS3-GFP vector. Furthermore, rAAV2-mediated expression of SOCS3 abolished the normally neurotrophic effects elicited by intravitreal rCNTF injections. In summary, CNTF delivery to the retina using viral vectors may be more effective than bolus rCNTF injections because the gene therapy approach has a less pronounced effect on neuron-intrinsic SOCS repressor pathways. Our new gain of function data using rAAV2-SOCS3-GFP demonstrate the negative impact of enhanced SOCS3 expression on the regenerative potential of mature CNS neurons.


Asunto(s)
Axones/metabolismo , Factor Neurotrófico Ciliar/administración & dosificación , Terapia Genética/métodos , Regeneración Nerviosa/fisiología , Células Ganglionares de la Retina/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Adenoviridae/genética , Animales , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , AMP Cíclico/administración & dosificación , AMP Cíclico/análogos & derivados , Femenino , Expresión Génica , Vectores Genéticos/genética , Inmunohistoquímica , Inyecciones Intravítreas , Microscopía Confocal , Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Traumatismos del Nervio Óptico/fisiopatología , Traumatismos del Nervio Óptico/terapia , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Ratas , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Células Ganglionares de la Retina/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/efectos de los fármacos , Transducción Genética
8.
Exp Neurol ; 355: 114120, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35605669

RESUMEN

Gene therapy is a powerful approach to promote spinal cord regeneration. For a clinical application it is important to restrict therapeutic gene expression to the appropriate time window to limit unwanted side effects. The doxycycline (dox)-inducible system is a widely used regulatable gene expression platform, however, this system depends on a bacterial-derived immunogenic transactivator. The foreign origin of this transactivator prevents reliable regulation of therapeutic gene expression and currently limits clinical translation. The glycine-alanine repeat (GAR) of Epstein-Barr virus nuclear antigen-1 protein inhibits its presentation to cytotoxic T cells, allowing virus-infected cells to evade the host immune system. We developed a chimeric transactivator (GARrtTA) and show that GARrtTA has an immune-evading advantage over "classical" rtTA in vivo. Direct comparison of lentiviral vectors expressing rtTA and GARrtTA in the rat spinal cord shows that the GARrtTA system is inducible for 6 doxycycline-cycles over a 47 week period, whereas with the rtTA-based system luciferase reporter expression declines during the 3rd cycle and is no longer re-inducible, indicating that GARrtTA provides an immune-advantage over rtTA. Immunohistochemistry revealed that GARrtTA expressing cells in the spinal cord appear healthier and survive better than rtTA expressing cells. Characterization of the immune response shows that expression of GARrtTA, in contrast to rtTA, does not recruit cytotoxic T-cells to the transduced spinal cord. This study demonstrates that fusion of the GAR domain to rtTA results in a functional doxycycline-inducible transactivator with a clear immune-advantage over the classical rtTA in vivo.


Asunto(s)
Doxiciclina , Infecciones por Virus de Epstein-Barr , Animales , Doxiciclina/farmacología , Regulación de la Expresión Génica , Terapia Genética/métodos , Herpesvirus Humano 4/genética , Ratas , Médula Espinal , Transactivadores/genética
9.
Gene Ther ; 17(5): 662-71, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20182518

RESUMEN

Neuroinflammation, as defined by activation of local glial cells and production of various inflammatory mediators, is an important feature of many neurological disorders. Expression of pro-inflammatory mediators produced by glial cells in the central nervous system (CNS) is considered to contribute to the neuropathology observed in those diseases. To diminish the production or action of pro-inflammatory mediators, we have used lentiviral (LV) vector-mediated encoding rat interleukin-10 (rIL-10) or rat interleukin-1 receptor antagonist (rIL-1ra) to direct the local, long-term expression of these anti-inflammatory cytokines in the CNS. We have shown that cultured macrophages or astroglia transduced with LV-rIL-10 or LV-rIL-1ra produced far less tumor necrosis factor (TNF)alpha or IL-6, respectively in response to pro-inflammatory stimuli. Moreover, intracerebroventricular (i.c.v.) administration of LV-rIL-10 or LV-rIL-1ra resulted in transduction of glial cells and macrophages and, subsequently reduced TNFalpha, IL-6 and inducible nitric oxide synthase (iNOS) expression in various brain regions induced by inflammatory stimuli, whereas peripheral expression of these mediators remained unaffected. In addition, expression levels of the anti-inflammatory cytokines IL-4 and transforming growth factor-beta were not altered in either brain or pituitary gland. Furthermore, i.c.v. administration of LV-rIL-10 or LV-rIL-1ra given during the remission phase of chronic-relapsing experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, improved the clinical outcome of the relapse phase. Thus, local application of LV vectors expressing anti-inflammatory cytokines could be of therapeutic interest to counteract pro-inflammatory processes in the brain without interfering with the peripheral production of inflammatory mediators.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Terapia Genética/métodos , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-10/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encefalomielitis Autoinmune Experimental/patología , Vectores Genéticos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Interleucina-4/análisis , Interleucina-4/metabolismo , Interleucina-6/análisis , Interleucina-6/metabolismo , Lentivirus , Macrófagos/metabolismo , Masculino , Neuroglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/análisis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Wistar , Transducción Genética , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
10.
Gene Ther ; 16(4): 521-32, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19092858

RESUMEN

Recombinant adeno-associated virus (rAAV) vectors are increasingly being used as tools for gene therapy, and clinical trials have begun in patients with genetically linked retinal disorders. Intravitreal injection is optimal for the transduction of retinal ganglion cells (RGCs), although complete selectivity has not been achieved. There may also be advantages in using intravitreal approaches for the transduction of photoreceptors. Here we compared the cellular tropism and transduction efficiency of rAAV2/1, -2/2, -2/3, -2/4, -2/5, -2/6 and -2/8 in adult rat retina after intravitreal injection. Each vector encoded green fluorescent protein (GFP), and the number, laminar distribution and morphology of transduced GFP(+) cells were determined using fluorescent microscopy. Assessment of transduced cell phenotype was based on cell morphology and immunohistochemistry. rAAV2/2 and rAAV2/6 transduced the greatest number of cells, whereas rAAV2/5 and rAAV2/8 were least efficient. Most vectors primarily transduced RGCs; however, rAAV2/6 had a more diverse tropism profile, with 46% identified as amacrine or bipolar cells, 23% as RGCs and 22% as Müller cells. Müller cells were also frequently transduced by rAAV2/4. The highest photoreceptor transduction was seen after intravitreal rAAV2/3 injection. These data facilitate the design and selection of rAAV vectors to target specific retinal cells, potentially leading to an improved gene therapy for various human retinal pathologies.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Retina/metabolismo , Transducción Genética , Animales , Dependovirus/clasificación , Dependovirus/fisiología , Femenino , Inyecciones Intraoculares , Microscopía Confocal , Ratas , Ratas Wistar , Retina/virología , Serotipificación , Tropismo Viral , Cuerpo Vítreo
11.
Exp Neurol ; 321: 113032, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31398353

RESUMEN

Many studies, using pre-clinical models of SCI, have demonstrated the efficacy of chondroitinase ABC as a treatment for spinal cord injury and this has been confirmed in laboratories worldwide and in several animal models. The aim of this review is report the current state of research in the field and to compare the relative efficacies of these new interventions to improve outcomes in both acute and chronic models of SCI. We also report new methods of chondroitinase delivery and the outcomes of two clinical trials using the enzyme to treat spinal cord injury in dogs and disc herniation in human patients. Recent studies have assessed the outcomes of combining chondroitinase with other strategies known to promote recovery following spinal cord injury and new approaches. Evidence is emerging that one of the most powerful combinations is that of chondroitinase with cell transplants. The particular benefits of each of the different cell types used for these transplant experiments are discussed. Combining chondroitinase with rehabilitation also improves outcomes. Gene therapy is an efficient method of enzyme delivery to the injured spinal cord and circumvents the issue of the enzyme's thermo-instability. Other methods of delivery, such as via nanoparticles or synthetic scaffolds, have shown promise; however, the outcomes from these experiments suggest that these methods of delivery require further optimization to achieve similar levels of efficacy to that obtained by a gene therapy approach. Pre-clinical models have also shown chondroitinase is efficacious in the treatment of other conditions, such as peripheral nerve injury, stroke, coronary reperfusion, Parkinson's disease and certain types of cancer. The wide range of conditions where the benefits of chondroitinase treatment have been demonstrated reflects the complex roles that chondroitin sulphate proteoglycans (its substrate) play in health and disease and warrants the enzyme's further development as a therapy.


Asunto(s)
Condroitina ABC Liasa/uso terapéutico , Animales , Humanos , Traumatismos de la Médula Espinal/terapia
12.
Curr Biol ; 6(9): 1153-8, 1996 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8805367

RESUMEN

BACKGROUND: Growth cones at the tips of growing axons move along predetermined pathways to establish synaptic connections between neurons and their distant targets. To establish their orientation, growth cones continuously sample for, and respond to, guidance information provided by cell surfaces and the extracellular matrix. To identify specific guidance cues, growth cones have sensor molecules on their surface, which are expressed differentially during the temporospatial progress of axon outgrowth, at levels that depend on the pattern of neural activity. However, it has not been elucidated whether a change in gene expression can indeed change the molecular composition and, hence, the function of the sensor apparatus of growth cones. RESULTS: We have constructed adenoviral gene transfer vectors of the chicken growth cone sensor molecules axonin-1 and Ng-CAM. Using these vectors, we initiated the expression of axonin-1 and Ng-CAM in rat dorsal root ganglia explants during ongoing neurite outgrowth. Using specific surface immunodetection at varying time points after infection, we found that axonin-1 and Ng-CAM are transported directly to the growth cone and inserted exclusively in the growth cone membrane and not in the axolemma of the axon shaft. Furthermore, we found that axonin-1 and Ng-CAM do not diffuse retrogradely, suggesting that the sensor molecules are integrated into multimolecular complexes in the growth cone. CONCLUSIONS: During axon outgrowth, the pathway sensor apparatus of the growth cone is continuously updated by newly synthesized sensor molecules that originate directly from the transcription/translation machinery. Changes in the expression of sensor molecules may have a direct impact, therefore, on the exploratory function of the growth cone.


Asunto(s)
Axones , Neuritas , Animales , Axones/metabolismo , Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Embrión de Pollo , Contactina 2 , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Inmunohistoquímica , Ratas
13.
Prog Neurobiol ; 55(4): 399-432, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9654386

RESUMEN

Viral vectors are becoming increasingly important tools to investigate the function of neural proteins and to explore the feasibility of gene therapy to treat diseases of the nervous system. This gene transfer technology is based on the use of a virus as a gene delivery vehicle. In contrast to functional analysis of gene products in transgenic mouse, viral vectors can be applied to transfer genes to somatic, post-mitotic cells of fully developed animals. To date, five viral vector systems are available for gene transfer in the nervous system. These include recombinant and defective herpes viral vectors, adenoviral vectors, adeno-associated viral vectors and lentiviral vectors. Of these vectors herpes and adenoviral vectors are the most common in use. To date, one of the main hurdles in applying these two vector systems is the focal immune response that occurs following intraparenchymal infusion. Despite this limitation, herpes and adenoviral vectors have been used successfully to modify the physiological response to injury in several rodent models of neurodegeneration. The first purpose of this review is to describe the principles of the generation of viral vectors and to discuss the advantages and disadvantages of the viral vector systems currently in use for gene transfer in the nervous system. Secondly, we give an overview of the performance of these vectors following direct infusion in the nervous system and review the results obtained with these vectors in animal models of neurodegeneration and regeneration. The results of these initial studies have provided a framework for future experiments based on gene transfer strategies with viral vectors to study normal physiology and pathology of the nervous system.


Asunto(s)
Adenoviridae , Técnicas de Transferencia de Gen , Herpes Simple , Enfermedades del Sistema Nervioso/terapia , Fenómenos Fisiológicos del Sistema Nervioso , Animales , Humanos
14.
Prog Neurobiol ; 53(6): 627-86, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9447616

RESUMEN

The growth-associated protein B-50 (GAP-43) is a presynaptic protein. Its expression is largely restricted to the nervous system. B-50 is frequently used as a marker for sprouting, because it is located in growth cones, maximally expressed during nervous system development and re-induced in injured and regenerating neural tissues. The B-50 gene is highly conserved during evolution. The B-50 gene contains two promoters and three exons which specify functional domains of the protein. The first exon encoding the 1-10 sequence, harbors the palmitoylation site for attachment to the axolemma and the minimal domain for interaction with G0 protein. The second exon contains the "GAP module", including the calmodulin binding and the protein kinase C phosphorylation domain which is shared by the family of IQ proteins. Downstream sequences of the second and non-coding sequences in the third exon encode species variability. The third exon also contains a conserved domain for phosphorylation by casein kinase II. Functional interference experiments using antisense oligonucleotides or antibodies, have shown inhibition of neurite outgrowth and neurotransmitter release. Overexpression of B-50 in cells or transgenic mice results in excessive sprouting. The various interactions, specified by the structural domains, are thought to underlie the role of B-50 in synaptic plasticity, participating in membrane extension during neuritogenesis, in neurotransmitter release and long-term potentiation. Apparently, B-50 null-mutant mice do not display gross phenotypic changes of the nervous system, although the B-50 deletion affects neuronal pathfinding and reduces postnatal survival. The experimental evidence suggests that neuronal morphology and communication are critically modulated by, but not absolutely dependent on, (enhanced) B-50 presence.


Asunto(s)
Comunicación Celular/fisiología , Proteína GAP-43/fisiología , Sistema Nervioso/citología , Neuronas/fisiología , Neuronas/ultraestructura , Animales , Humanos , Ratones
15.
FASEB J ; 15(14): 2680-8, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11726544

RESUMEN

Ubiquitin-B+1 (UBB+1) is a mutant ubiquitin that accumulates in the neurones of patients with Alzheimer's disease (AD). Here we report on the biochemical and functional differences between ubiquitin and UBB+1 and the effect of the mutant protein on neuronal cells. UBB+1 lacks the capacity to ubiquitinate, and although it is ubiquitinated itself, UBB+1 is not degraded by the ubiquitin-proteasomal system and is quite stable in neuronal cells. Overexpression of UBB+1 in neuroblastoma cells significantly induces nuclear fragmentation and cell death. Our results demonstrate that accumulation of UBB+1 in neurones is detrimental and may contribute to neuronal dysfunction in AD patients.


Asunto(s)
Enfermedad de Alzheimer/patología , Apoptosis/fisiología , Neuronas/citología , Ubiquitina/genética , Adenosina Trifosfato/farmacología , Adenoviridae/genética , Enfermedad de Alzheimer/genética , Western Blotting , Núcleo Celular/metabolismo , Cisteína Endopeptidasas/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Vectores Genéticos/genética , Humanos , Operón Lac/genética , Complejos Multienzimáticos/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Mutación , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ubiquitina/efectos de los fármacos , Ubiquitina/metabolismo
16.
Hum Gene Ther ; 8(9): 1049-63, 1997 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-9189763

RESUMEN

In the present paper, we examined the effect of the adenoviral vector dosage, the role of T cells, and the influence of the presence of replication-competent adenovirus (RCA) in adenoviral vector stocks, on the efficacy of adenoviral vector-directed transgene expression in the facial nucleus of immunocompetent Wistar and athymic nude rats. A small number of motor neurons and glial cells was transduced at low dosages of viral vector (1 x 10(6) pfu) and in the absence of RCA, and transgene-expressing cells persisted throughout the 3-week period of observation. Intraparenchymal infusion of 2 x 10(7) pfu of a recombinant adenoviral vector free of RCA was required for optimal transduction of facial motor neurons. In Wistar rats, a biphasic immune response occurred at higher dosages of the vector (5 x 10(6) and 2 x 10(7) pfu) that was characterized by early infiltration of macrophages and the occurrence of T cells during the second week after injection of the vector. The immune response was associated with the loss of transduced neural cells. In nude rats, administration of an adenoviral vector free of RCA resulted in a macrophage response comparable to that in the Wistar rat and long-term survival of transduced astroglial cells. However, transduced motor neurons degenerated according to a similar time course as observed in Wistar rats. Small amounts of RCA (2 x 10(5) pfu) injected with 2 x 10(7) pfu recombinant viral vector particles resulted in an accelerated T cell response and a rapid elimination of transduced cells within 1 week in Wistar rats, whereas in nude rats transgene expression continued during this period. Taken together, these observations suggest that at the high viral vector loads necessary to achieve optimal transduction of the facial nucleus, T cells play a role in the degeneration of adenoviral vector-transduced astroglial cells. The adverse effects on neurons appear to be due to the observed inflammatory response or to direct adenoviral vector toxicity.


Asunto(s)
Adenoviridae/genética , Astrocitos/virología , Técnicas de Transferencia de Gen , Vectores Genéticos/farmacología , Sistema Nervioso/virología , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Supervivencia Celular/genética , Relación Dosis-Respuesta a Droga , Nervio Facial/virología , Genes Reporteros , Vectores Genéticos/genética , Inmunocompetencia/genética , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Masculino , Neuronas Motoras/virología , Necrosis , Ratas , Ratas Desnudas , Ratas Wistar , Transducción Genética , Transgenes , Replicación Viral
17.
Hum Gene Ther ; 10(11): 1885-91, 1999 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-10446928

RESUMEN

Recombinant adeno-associated virus (rAAV) vectors have become attractive tools for in vivo gene transfer. The production and purification of high-titer rAAV vector stocks for experimental and therapeutic gene transfer continue to undergo improvement. Standard rAAV vector purification protocols include the purification of the vector by cesium chloride (CsCl)-density gradient centrifugation followed by extensive desalination via dialysis against a physiological buffer for in vivo use. These procedures are extremely time consuming and frequently result in a substantial loss of the infectious vector titer. As an alternative to CsCl we have investigated the use of Iodixanol, an X-ray contrast solution, as the density-gradient medium. Purification of rAAV vectors by Iodixanol shortened the centrifugation period to 3 hr and resulted in reproducible concentration and purification of rAAV-vector stocks. We show that injection of rAAV derived from an Iodixanol gradient can be used for in vivo gene transfer applications in the brain and spinal cord without detectable cytopathic effects and directing stable transgene expression for at least 2 months.


Asunto(s)
Centrifugación por Gradiente de Densidad/métodos , Dependovirus/aislamiento & purificación , Técnicas de Transferencia de Gen , Vectores Genéticos/aislamiento & purificación , Sistema Nervioso , Animales , Encéfalo , Cesio/química , Cloruros/química , Medios de Contraste/química , Dependovirus/genética , Dependovirus/fisiología , Inmunohistoquímica , Ratas , Médula Espinal , Ácidos Triyodobenzoicos/química
18.
Eur J Neurosci ; 2(5): 397-407, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-12106027

RESUMEN

The olfactory neuroepithelium exhibits neurogenesis throughout adult life, and in response to lesions, a phenomenon that distinguishes this neural tissue from the rest of the mammalian brain. The newly formed primary olfactory neurons elaborate axons into the olfactory bulb. Thus, denervation and subsequent re-innervation of olfactory bulb neurons may occur throughout life. In this study the authors demonstrate the distribution of the growth-associated phosphoprotein B-50/GAP43 and its mRNA in the olfactory neuroepithelium and olfactory bulb during development and aging. In neonatal rats B-50/GAP43 mRNA was expressed in primary olfactory neurons throughout the olfactory epithelium and in their target neurons in the olfactory bulb, the mitral, juxtaglomerular and tufted cells. In contrast, in adult (7.5 weeks) and aging animals (6 - 18 months of age) B-50/GAP43 mRNA expression was progressively restricted to neurons in the basal region of the neuroepithelium and to some of their target mitral and juxtaglomerular cells in the olfactory bulb. The continuing expression of B-50/GAP43 mRNA in mitral- and juxtaglomerular cells in mature animals is thought to be related to their capacity to respond to continuously changing input from the primary olfactory neurons present in the olfactory neuroepithelium.

19.
Eur J Neurosci ; 2(6): 487-499, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-12106019

RESUMEN

The hypothesis that B-50/GAP43, a membrane-associated phosphoprotein, is involved in process outgrowth has been tested by studying the developmental pattern of expression of B-50/GAP43 mRNA and protein during mouse neuroembryogenesis. B-50/GAP43 mRNA is first detectable at embryonic day 8.5 (E8.5) in the presumptive acoustico-facialis ganglion. Subsequently, both B-50/GAP43 mRNA and protein were co-expressed in a series of neural structures: in the ventral neural tube (from E9.5) and dorsal root ganglia (from E10.5), in the marginal layer of the neuroepithelium surrounding the brain vesicles and in the cranial ganglia (from E9.5), in the autonomic nervous system (from E10.5), in the olfactory neuroepithelium and in the mesenteric nervous system (from E11.5), in a continuum of brain regions (from E12.5) and in the retina (from E13.5). Immunoreactive fibers were always seen arising from these regions when they expressed B-50/GAP43 mRNA. The spatial and temporal pattern of B-50/GAP43 expression demonstrates that this protein is absent from neuroblasts and consistently appears in neurons committed to fiber outgrowth. The expression of the protein in immature neurons is independent of their embryological origin. Our detailed study of B-50/GAP43 expression during mouse neuroembryogenesis supports the view that this protein is involved in a process common to all neurons elaborating fibers.

20.
J Comp Neurol ; 422(4): 579-93, 2000 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-10861527

RESUMEN

Geniculate (gustatory) and trigeminal (somatosensory) afferents take different routes to the tongue during rat embryonic development. To learn more about the mechanisms controlling neurite outgrowth and axon guidance, we are studying the roles of diffusible factors. We previously profiled the in vitro sensitivity of trigeminal axons to neurotrophins and target-derived diffusible factors and now report on these properties for geniculate axons. GDNF, BDNF, and NT-4, but not NT-3 or NGF, stimulate geniculate axon outgrowth during the ages investigated, embryonic days 12-14. Sensitivity to effective neurotrophins is developmentally regulated and different from that of the trigeminal ganglion. In vitro coculture studies revealed that geniculate axons were repelled by branchial arch explants that were previously shown to be repellent to trigeminal axons (Rochlin and Farbman [1998] J Neurosci 18:6840-6852). In addition, some branchial arch explants and untransfected COS7 cells repelled geniculate but not trigeminal axons. Sema3A, a ligand for neuropilin-1, is effective in repelling geniculate and trigeminal axons, and antineuropilin-1, but not antineuropilin-2, completely blocks the repulsion by arch explants that repel axon outgrowth from both ganglia. Sema3A mRNA is concentrated in branchial arch epithelium at the appropriate time to mediate the repulsion. In Sema3A knockout mice, geniculate and trigeminal afferents explore medial regions of the immature tongue and surrounding territories not explored in heterozygotes, supporting our previous hypothesis that Sema3A-based repulsion mediates the early restriction of sensory afferents away from midline structures.


Asunto(s)
Axones/efectos de los fármacos , Ganglio Geniculado/efectos de los fármacos , Factores de Crecimiento Nervioso/farmacología , Lengua/inervación , Ganglio del Trigémino/efectos de los fármacos , Animales , Axones/fisiología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células COS , Células Cultivadas , Ganglio Geniculado/embriología , Ganglio Geniculado/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial , Ratones , Ratones Noqueados , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Neurotrofina 3/farmacología , Ratas , Ratas Sprague-Dawley , Lengua/embriología , Ganglio del Trigémino/embriología , Ganglio del Trigémino/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA