Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 16: 1253954, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37829513

RESUMEN

Introduction: Brain connectivity requires correct axonal guidance to drive axons to their appropriate targets. This process is orchestrated by guidance cues that exert attraction or repulsion to developing axons. However, the intricacies of the cellular machinery responsible for the correct response of growth cones are just being unveiled. Netrin-1 is a bifunctional molecule involved in axon pathfinding and cell migration that induces repulsion during postnatal cerebellar development. This process is mediated by UNC5 homolog receptors located on external granule layer (EGL) tracts. Methods: Biochemical, imaging and cell biology techniques, as well as syntaxin-1A/B (Stx1A/B) knock-out mice were used in primary cultures and brain explants. Results and discussion: Here, we demonstrate that this response is characterized by enhanced membrane internalization through macropinocytosis, but not clathrin-mediated endocytosis. We show that UNC5A, UNC5B, and UNC5C receptors form a protein complex with the t-SNARE syntaxin-1. By combining botulinum neurotoxins, an shRNA knock-down strategy and Stx1 knock-out mice, we demonstrate that this SNARE protein is required for Netrin1-induced macropinocytosis and chemorepulsion, suggesting that Stx1 is crucial in regulating Netrin-1-mediated axonal guidance.

2.
Hum Mol Genet ; 28(6): 961-971, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30476097

RESUMEN

Spastic paraplegia gene 11(SPG11)-linked hereditary spastic paraplegia is a complex monogenic neurodegenerative disease that in addition to spastic paraplegia is characterized by childhood onset cognitive impairment, thin corpus callosum and enlarged ventricles. We have previously shown impaired proliferation of SPG11 neural progenitor cells (NPCs). For the delineation of potential defect in SPG11 brain development we employ 2D culture systems and 3D human brain organoids derived from SPG11 patients' iPSC and controls. We reveal that an increased rate of asymmetric divisions of NPCs leads to proliferation defect, causing premature neurogenesis. Correspondingly, SPG11 organoids appeared smaller than controls and had larger ventricles as well as thinner germinal wall. Premature neurogenesis and organoid size were rescued by GSK3 inhibititors including the Food and Drug Administration-approved tideglusib. These findings shed light on the neurodevelopmental mechanisms underlying disease pathology.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Neurogénesis/genética , Proteínas/genética , Alelos , Biomarcadores , Corteza Cerebral/fisiopatología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Genotipo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mutación , Organoides , Fenotipo , beta Catenina
3.
J Immunol ; 198(10): 3878-3885, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28411187

RESUMEN

Microglia cells fulfill key homeostatic functions and essentially contribute to host defense within the CNS. Altered activation of microglia, in turn, has been implicated in neuroinflammatory and neurodegenerative diseases. In this study, we identify the nuclear receptor (NR) Nr4a1 as key rheostat controlling the activation threshold and polarization of microglia. In steady-state microglia, ubiquitous neuronal-derived stress signals such as ATP induced expression of this NR, which contributed to the maintenance of a resting and noninflammatory microglia phenotype. Global and microglia-specific deletion of Nr4a1 triggered the spontaneous and overwhelming activation of microglia and resulted in increased cytokine and NO production as well as in an accelerated and exacerbated form of experimental autoimmune encephalomyelitis. Ligand-induced activation of Nr4a1 accordingly ameliorated the course of this disease. Our current data thus identify Nr4a1 as regulator of microglia activation and potentially new target for the treatment of inflammatory CNS diseases such as multiple sclerosis.


Asunto(s)
Sistema Nervioso Central/inmunología , Microglía/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Adenosina Trifosfato/farmacología , Animales , Células Cultivadas , Enfermedades del Sistema Nervioso Central/terapia , Citocinas/biosíntesis , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Encefalomielitis Autoinmune Experimental/prevención & control , Regulación de la Expresión Génica , Humanos , Inflamación , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Esclerosis Múltiple/terapia , Enfermedades Neurodegenerativas/terapia , Óxido Nítrico/biosíntesis , Óxido Nítrico/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia
4.
Schizophr Bull ; 43(1): 214-225, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27613806

RESUMEN

Few studies have addressed likely gene × gene (ie, epistatic) interactions in mediating risk for schizophrenia. Using a preclinical genetic approach, we investigated whether simultaneous disruption of the risk factors Neuregulin-1 (NRG1) and Disrupted-in-schizophrenia 1 (DISC1) would produce a disease-relevant phenotypic profile different from that observed following disruption to either gene alone. NRG1 heterozygotes exhibited hyperactivity and disruption to prepulse inhibition, both reversed by antipsychotic treatment, and accompanied by reduced striatal dopamine D2 receptor protein expression, impaired social cognition, and altered glutamatergic synaptic protein expression in selected brain areas. Single gene DISC1 mutants demonstrated a disruption in social cognition and nest-building, altered brain 5-hydroxytryptamine levels and hippocampal ErbB4 expression, and decreased cortical expression of the schizophrenia-associated microRNA miR-29b. Co-disruption of DISC1 and NRG1, indicative of epistasis, evoked an impairment in sociability and enhanced self-grooming, accompanied by changes in hypothalamic oxytocin/vasopressin gene expression. The findings indicate specific behavioral correlates and underlying cellular pathways downstream of main effects of DNA variation in the schizophrenia-associated genes NRG1 and DISC1.


Asunto(s)
Conducta Animal , Encéfalo/metabolismo , Endofenotipos , Epistasis Genética , Proteínas del Tejido Nervioso/metabolismo , Neurregulina-1/metabolismo , Trastornos Psicóticos , Esquizofrenia , Anfetaminas/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Aseo Animal , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Comportamiento de Nidificación , Neurregulina-1/genética , Oxitocina/metabolismo , Inhibición Prepulso/genética , Agitación Psicomotora/genética , Trastornos Psicóticos/genética , Trastornos Psicóticos/metabolismo , Trastornos Psicóticos/fisiopatología , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Conducta Social , Vasopresinas/metabolismo
5.
PLoS One ; 11(7): e0158686, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27392094

RESUMEN

The transmembrane semaphorin, Sema6A, has important roles in axon guidance, cell migration and neuronal connectivity in multiple regions of the nervous system, mediated by context-dependent interactions with plexin receptors, PlxnA2 and PlxnA4. Here, we demonstrate that Sema6A can also signal cell-autonomously, in two modes, constitutively, or in response to higher-order clustering mediated by either PlxnA2-binding or chemically induced multimerisation. Sema6A activation stimulates recruitment of Abl to the cytoplasmic domain of Sema6A and phos¡phorylation of this cytoplasmic tyrosine kinase, as well as phosphorylation of additional cytoskeletal regulators. Sema6A reverse signaling affects the surface area and cellular complexity of non-neuronal cells and aggregation and neurite formation of primary neurons in vitro. Sema6A also interacts with PlxnA2 in cis, which reduces binding by PlxnA2 of Sema6A in trans but not vice versa. These experiments reveal the complex nature of Sema6A biochemical functions and the molecular logic of the context-dependent interactions between Sema6A and PlxnA2.


Asunto(s)
Semaforinas/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Citoplasma/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Ratones , Proteínas de Microfilamentos , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/genética , Fosforilación/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Multimerización de Proteína , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Semaforinas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
Ann Neurol ; 79(5): 826-840, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26971897

RESUMEN

OBJECTIVE: Mutations in the spastic paraplegia gene 11 (SPG11), encoding spatacsin, cause the most frequent form of autosomal-recessive complex hereditary spastic paraplegia (HSP) and juvenile-onset amyotrophic lateral sclerosis (ALS5). When SPG11 is mutated, patients frequently present with spastic paraparesis, a thin corpus callosum, and cognitive impairment. We previously delineated a neurodegenerative phenotype in neurons of these patients. In the current study, we recapitulated early developmental phenotypes of SPG11 and outlined their cellular and molecular mechanisms in patient-specific induced pluripotent stem cell (iPSC)-derived cortical neural progenitor cells (NPCs). METHODS: We generated and characterized iPSC-derived NPCs and neurons from 3 SPG11 patients and 2 age-matched controls. RESULTS: Gene expression profiling of SPG11-NPCs revealed widespread transcriptional alterations in neurodevelopmental pathways. These include changes in cell-cycle, neurogenesis, cortical development pathways, in addition to autophagic deficits. More important, the GSK3ß-signaling pathway was found to be dysregulated in SPG11-NPCs. Impaired proliferation of SPG11-NPCs resulted in a significant diminution in the number of neural cells. The decrease in mitotically active SPG11-NPCs was rescued by GSK3 modulation. INTERPRETATION: This iPSC-derived NPC model provides the first evidence for an early neurodevelopmental phenotype in SPG11, with GSK3ß as a potential novel target to reverse the disease phenotype. Ann Neurol 2016;79:826-840.

7.
Biol Open ; 4(5): 608-21, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25882555

RESUMEN

According to the general understanding, the chondrocyte lineage terminates with the elimination of late hypertrophic cells by apoptosis in the growth plate. However, recent cell tracking studies have shown that murine hypertrophic chondrocytes can survive beyond "terminal" differentiation and give rise to a progeny of osteoblasts participating in endochondral bone formation. The question how chondrocytes convert into osteoblasts, however, remained open. Following the cell fate of hypertrophic chondrocytes by genetic lineage tracing using BACCol10;Cre induced YFP-reporter gene expression we show that a progeny of Col10Cre-reporter labelled osteoprogenitor cells and osteoblasts appears in the primary spongiosa and participates - depending on the developmental stage - substantially in trabecular, endosteal, and cortical bone formation. YFP(+) trabecular and endosteal cells isolated by FACS expressed Col1a1, osteocalcin and runx2, thus confirming their osteogenic phenotype. In searching for transitory cells between hypertrophic chondrocytes and trabecular osteoblasts we identified by confocal microscopy a novel, small YFP(+)Osx(+) cell type with mitotic activity in the lower hypertrophic zone at the chondro-osseous junction. When isolated from growth plates by fractional enzymatic digestion, these cells termed CDOP (chondrocyte-derived osteoprogenitor) cells expressed bone typical genes and differentiated into osteoblasts in vitro. We propose the Col10Cre-labeled CDOP cells mark the initiation point of a second pathway giving rise to endochondral osteoblasts, alternative to perichondrium derived osteoprogenitor cells. These findings add to current concepts of chondrocyte-osteocyte lineages and give new insight into the complex cartilage-bone transition process in the growth plate.

8.
PLoS One ; 9(8): e103976, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25133820

RESUMEN

Swiprosin-1/EFhd2 (EFhd2) is a cytoskeletal Ca2+ sensor protein strongly expressed in the brain. It has been shown to interact with mutant tau, which can promote neurodegeneration, but nothing is known about the physiological function of EFhd2 in the nervous system. To elucidate this question, we analyzed EFhd2-/-/lacZ reporter mice and showed that lacZ was strongly expressed in the cortex, the dentate gyrus, the CA1 and CA2 regions of the hippocampus, the thalamus, and the olfactory bulb. Immunohistochemistry and western blotting confirmed this pattern and revealed expression of EFhd2 during neuronal maturation. In cortical neurons, EFhd2 was detected in neurites marked by MAP2 and co-localized with pre- and post-synaptic markers. Approximately one third of EFhd2 associated with a biochemically isolated synaptosome preparation. There, EFhd2 was mostly confined to the cytosolic and plasma membrane fractions. Both synaptic endocytosis and exocytosis in primary hippocampal EFhd2-/- neurons were unaltered but transport of synaptophysin-GFP containing vesicles was enhanced in EFhd2-/- primary hippocampal neurons, and notably, EFhd2 inhibited kinesin mediated microtubule gliding. Therefore, we found that EFhd2 is a neuronal protein that interferes with kinesin-mediated transport.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Cinesinas/metabolismo , Neuritas/metabolismo , Animales , Transporte Axonal , Células Cultivadas , Hipocampo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas , Sinaptosomas/metabolismo
9.
Hum Mol Genet ; 23(18): 4859-74, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24794856

RESUMEN

Hereditary spastic paraplegias are a group of inherited motor neuron diseases characterized by progressive paraparesis and spasticity. Mutations in the spastic paraplegia gene SPG11, encoding spatacsin, cause an autosomal-recessive disease trait; however, the precise knowledge about the role of spatacsin in neurons is very limited. We for the first time analyzed the expression and function of spatacsin in human forebrain neurons derived from human pluripotent stem cells including lines from two SPG11 patients and two controls. SPG11 patients'-derived neurons exhibited downregulation of specific axonal-related genes, decreased neurite complexity and accumulation of membranous bodies within axonal processes. Altogether, these data point towards axonal pathologies in human neurons with SPG11 mutations. To further corroborate spatacsin function, we investigated human pluripotent stem cell-derived neurons and mouse cortical neurons. In these cells, spatacsin was located in axons and dendrites. It colocalized with cytoskeletal and synaptic vesicle (SV) markers and was present in synaptosomes. Knockdown of spatacsin in mouse cortical neurons evidenced that the loss of function of spatacsin leads to axonal instability by downregulation of acetylated tubulin. Finally, time-lapse assays performed in SPG11 patients'-derived neurons and spatacsin-silenced mouse neurons highlighted a reduction in the anterograde vesicle trafficking indicative of impaired axonal transport. By employing SPG11 patient-derived forebrain neurons and mouse cortical neurons, this study provides the first evidence that SPG11 is implicated in axonal maintenance and cargo trafficking. Understanding the cellular functions of spatacsin will allow deciphering mechanisms of motor cortex dysfunction in autosomal-recessive hereditary spastic paraplegia.


Asunto(s)
Axones/metabolismo , Neuronas/metabolismo , Prosencéfalo/citología , Proteínas/metabolismo , Paraplejía Espástica Hereditaria/patología , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/patología , Células Madre Pluripotentes/metabolismo , Prosencéfalo/metabolismo , Proteínas/genética , Paraplejía Espástica Hereditaria/genética , Tubulina (Proteína)/metabolismo
10.
Eur J Neurosci ; 36(9): 3152-64, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22946563

RESUMEN

Directed cell migration and axonal guidance are essential steps in neural development that share many molecular mechanisms. The guidance of developing axons and migrating neurons is likely to depend on the precise control of plasmalemma turnover in selected regions of leading edges and growth cones, respectively. Previous results provided evidence of a signaling mechanism that couples chemotropic deleted in colorectal cancer (DCC)/Netrin-1 axonal guidance and exocytosis through Syntaxin1(Sytx1)/TI-VAMP SNARE proteins. Here we studied whether Netrin-1-dependent neuronal migration relies on a similar SNARE mechanism. We show that migrating neurons in the lower rhombic lip (LRL) express several SNARE proteins, and that DCC co-associates with Sytx1 and TI-VAMP in these cells. We also demonstrate that cleavage of Sytx1 by botulinum toxin C1 (BoNT/C1) abolishes Netrin-1-dependent chemoattraction of migrating neurons, and that interference of Sytx1 functions with shRNAs or Sytx1-dominant negatives disrupts Netrin-1-dependent chemoattraction of LRL neurons. These findings indicate that a Sytx1/DCC interaction is required for Netrin-1 guidance of migrating neurons, thereby highlighting a relationship between guidance signaling and SNARE proteins that regulate membrane turnover.


Asunto(s)
Cerebelo/metabolismo , Quimiotaxis , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo , Sintaxina 1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Toxinas Botulínicas/farmacología , Cerebelo/citología , Cerebelo/embriología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/genética , Receptor DCC , Regulación del Desarrollo de la Expresión Génica , Ratones , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Netrina-1 , ARN Interferente Pequeño , Receptores de Superficie Celular/genética , Transducción de Señal , Sintaxina 1/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
11.
J Neurosci ; 31(41): 14463-80, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21994363

RESUMEN

Directed cell migration and axonal guidance are essential steps in neural development. Both processes are controlled by specific guidance cues that activate the signaling cascades that ultimately control cytoskeletal dynamics. Another essential step in migration and axonal guidance is the regulation of plasmalemma turnover and exocytosis in leading edges and growth cones. However, the cross talk mechanisms linking guidance receptors and membrane exocytosis are not understood. Netrin-1 is a chemoattractive cue required for the formation of commissural pathways. Here, we show that the Netrin-1 receptor deleted in colorectal cancer (DCC) forms a protein complex with the t-SNARE (target SNARE) protein Syntaxin-1 (Sytx1). This interaction is Netrin-1 dependent both in vitro and in vivo, and requires specific Sytx1 and DCC domains. Blockade of Sytx1 function by using botulinum toxins abolished Netrin-1-dependent chemoattraction of axons in mouse neuronal cultures. Similar loss-of-function experiments in the chicken spinal cord in vivo using dominant-negative Sytx1 constructs or RNAi led to defects in commissural axon pathfinding reminiscent to those described in Netrin-1 and DCC loss-of-function models. We also show that Netrin-1 elicits exocytosis at growth cones in a Sytx1-dependent manner. Moreover, we demonstrate that the Sytx1/DCC complex associates with the v-SNARE (vesicle SNARE) tetanus neurotoxin-insensitive vesicle-associated membrane protein (TI-VAMP) and that knockdown of TI-VAMP in the commissural pathway in the spinal cord results in aberrant axonal guidance phenotypes. Our data provide evidence of a new signaling mechanism that couples chemotropic Netrin-1/DCC axonal guidance and Sytx1/TI-VAMP SNARE proteins regulating membrane turnover and exocytosis.


Asunto(s)
Exocitosis/fisiología , Conos de Crecimiento/fisiología , Factores de Crecimiento Nervioso/metabolismo , Neuronas/citología , Receptores de Superficie Celular/metabolismo , Proteínas SNARE/metabolismo , Transducción de Señal/genética , Proteínas Supresoras de Tumor/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Axones/fisiología , Compuestos de Boro/metabolismo , Toxinas Botulínicas Tipo A/farmacología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Chlorocebus aethiops , Complemento C1/farmacología , Receptor DCC , Embrión de Mamíferos , Exocitosis/efectos de los fármacos , Exocitosis/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Conos de Crecimiento/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipocampo/citología , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Munc18/genética , Proteínas Munc18/metabolismo , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Netrina-1 , Fármacos Neuromusculares/farmacología , Técnicas de Cultivo de Órganos , Receptores de Superficie Celular/genética , Proteínas SNARE/genética , Transducción de Señal/efectos de los fármacos , Resonancia por Plasmón de Superficie/métodos , Toxina Tetánica/farmacología , Transfección/métodos , Proteínas Supresoras de Tumor/genética , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
12.
Nature ; 467(7319): 1118-22, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20877282

RESUMEN

Cell-cell signalling of semaphorin ligands through interaction with plexin receptors is important for the homeostasis and morphogenesis of many tissues and is widely studied for its role in neural connectivity, cancer, cell migration and immune responses. SEMA4D and Sema6A exemplify two diverse vertebrate, membrane-spanning semaphorin classes (4 and 6) that are capable of direct signalling through members of the two largest plexin classes, B and A, respectively. In the absence of any structural information on the plexin ectodomain or its interaction with semaphorins the extracellular specificity and mechanism controlling plexin signalling has remained unresolved. Here we present crystal structures of cognate complexes of the semaphorin-binding regions of plexins B1 and A2 with semaphorin ectodomains (human PLXNB1(1-2)-SEMA4D(ecto) and murine PlxnA2(1-4)-Sema6A(ecto)), plus unliganded structures of PlxnA2(1-4) and Sema6A(ecto). These structures, together with biophysical and cellular assays of wild-type and mutant proteins, reveal that semaphorin dimers independently bind two plexin molecules and that signalling is critically dependent on the avidity of the resulting bivalent 2:2 complex (monomeric semaphorin binds plexin but fails to trigger signalling). In combination, our data favour a cell-cell signalling mechanism involving semaphorin-stabilized plexin dimerization, possibly followed by clustering, which is consistent with previous functional data. Furthermore, the shared generic architecture of the complexes, formed through conserved contacts of the amino-terminal seven-bladed ß-propeller (sema) domains of both semaphorin and plexin, suggests that a common mode of interaction triggers all semaphorin-plexin based signalling, while distinct insertions within or between blades of the sema domains determine binding specificity.


Asunto(s)
Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Semaforinas/química , Semaforinas/metabolismo , Transducción de Señal , Animales , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Sitios de Unión , Moléculas de Adhesión Celular/genética , Comunicación Celular , Cristalografía por Rayos X , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Semaforinas/genética , Relación Estructura-Actividad
13.
J Neurosci ; 28(14): 3683-8, 2008 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-18385326

RESUMEN

Advances in viral gene therapy have opened new possibilities for treating a range of motor neuron diseases, but these have not yet been translated into clinically applicable therapies because of difficulties in delivery to susceptible/damaged neurons, ambiguities in the identity of gene(s) implicated, and a paucity of means to quantify any physiological improvement. Most of these hurdles can be overcome by using the neuromuscular paralysis induced by botulinum neurotoxin type A (BoNT/A) as a prototype disease. Furthermore, because human botulism, occasionally fatal, causes prolonged muscle disablement as a result of the intraneuronal persistence of the toxin's SNAP-25 (S25)-cleaving protease, development of a genetic approach could lead to a potential treatment for this debilitating disease. Adeno-associated viral delivery of a cleavage-resistant S25 gene (S25-R198T) to chromaffin cells in vitro yielded exocytotically active S25-R198T that diminished subsequent blockade by BoNT/A of evoked catecholamine release. Evaluation in vivo, by administering this virus into rat spinal cord before injecting BoNT/A, showed a decreased inhibition of acetylcholine release as reflected in elevated retention of neuromuscular transmission. A similar, although smaller, protection of synaptic transmission from the toxin was seen after peripherally injecting the therapeutic virus. Such therapy also curtailed nerve sprouting normally induced by BoNT/A. This first demonstration of the utility of a DNA-based therapy for botulism paves the way for further advances in its treatment and for application to genetic disorders of motor neurons.


Asunto(s)
Toxinas Botulínicas Tipo A/farmacología , Fármacos Neuromusculares/farmacología , Unión Neuromuscular/efectos de los fármacos , Parálisis Seudobulbar , Proteína 25 Asociada a Sinaptosomas/metabolismo , Animales , Botulismo/tratamiento farmacológico , Catecolaminas/metabolismo , Células Cultivadas , Células Cromafines , Dependovirus/fisiología , Endocitosis/efectos de los fármacos , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Mutación/fisiología , Unión Neuromuscular/fisiología , Parálisis Seudobulbar/inducido químicamente , Parálisis Seudobulbar/tratamiento farmacológico , Parálisis Seudobulbar/patología , Ratas , Serina/genética , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/farmacología , Treonina/genética
14.
Mol Cell Neurosci ; 28(4): 599-612, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15797708

RESUMEN

The development of the nervous system (NS) requires the coordinated migration of multiple waves of neurons and subsequent processes of neurite maturation, both involving selective guidance mechanisms. In Caenorhabditis elegans, unc-53 codes for a new multidomain protein involved in the directional migration of a subset of cells. We describe here the first functional characterization of the mouse homologue, mouse Neuron navigator 1 (mNAV1), whose expression is largely restricted to the NS during development. EGFP-mNAV1 associates with microtubules (MTs) plus ends present in the growth cone through a new microtubule-binding (MTB) domain. Moreover, its overexpression in transfected cells leads to MT bundling. The abolition of mNAV1 causes loss of directionality in the leading processes of pontine-migrating cells, providing evidence for a role of mNAV1 in mediating Netrin-1-induced directional migration.


Asunto(s)
Movimiento Celular/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Factores de Crecimiento Nervioso/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Desarrollo Fetal/fisiología , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas/química
15.
Mol Cell Neurosci ; 20(2): 169-80, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12093152

RESUMEN

Syntaxin 1 and synaptobrevin/VAMP play an essential role in synaptic vesicle exocytosis. Two isoforms for each of these proteins, syntaxins 1A and 1B and synaptobrevin/VAMPs 1 and 2, have been found in nerve endings. Morphological and biochemical studies have revealed a characteristic colocalization and selective interactions patterns of syntaxin 1 and synaptobrevin/VAMP isoforms in nervous and endocrine systems. Moreover, studies in vitro with recombinant proteins have shown characteristic interaction patterns for each syntaxin 1-synaptobrevin/VAMP pair. The cytosolic protein Munc-18a modulates neurotransmission by inhibiting the binding of synaptobrevin/VAMP and SNAP-25 to syntaxin 1A. In the present study, several binding assays were used to demonstrate that Munc-18a significantly binds both isoforms of syntaxin 1 (syntaxins 1A and 1B). Moreover, the coexpression of Munc-18a and syntaxin 1A or syntaxin 1B in 29.3 T cells revealed syntaxin 1-dependent localization of Munc-18a in the plasma membrane. By using the three-hybrid system, we showed the inhibitory role of Munc-18a in the formation of syntaxin 1-synaptobrevin/VAMP complexes regardless of the isoforms. Since Munc-18a can bind both isoforms of syntaxin 1, the present data suggest that this protein is a general modulator of the formation of different SNARE complexes in the nerve endings.


Asunto(s)
Antígenos de Superficie/metabolismo , Exocitosis/fisiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Terminales Presinápticos/metabolismo , Proteínas/metabolismo , Membranas Sinápticas/metabolismo , Transmisión Sináptica/fisiología , Proteínas de Transporte Vesicular , Animales , Sitios de Unión/fisiología , Citosol/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas Munc18 , Unión Proteica/fisiología , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología , Ratas , Proteínas SNARE , Sintaxina 1 , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...