Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
J Neurosci ; 32(15): 5151-64, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22496561

RESUMEN

Ischemic stroke causes transient increase of neural stem and progenitor cell (NSPC) proliferation in the subventricular zone (SVZ), and migration of newly formed neuroblasts toward the damaged area where they mature to striatal neurons. The molecular mechanisms regulating this plastic response, probably involved in structural reorganization and functional recovery, are poorly understood. The adaptor protein LNK suppresses hematopoietic stem cell self-renewal, but its presence and role in the brain are poorly understood. Here we demonstrate that LNK is expressed in NSPCs in the adult mouse and human SVZ. Lnk(-/-) mice exhibited increased NSPC proliferation after stroke, but not in intact brain or following status epilepticus. Deletion of Lnk caused increased NSPC proliferation while overexpression decreased mitotic activity of these cells in vitro. We found that Lnk expression after stroke increased in SVZ through the transcription factors STAT1/3. LNK attenuated insulin-like growth factor 1 signaling by inhibition of AKT phosphorylation, resulting in reduced NSPC proliferation. Our findings identify LNK as a stroke-specific, endogenous negative regulator of NSPC proliferation, and suggest that LNK signaling is a novel mechanism influencing plastic responses in postischemic brain.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Células-Madre Neurales/fisiología , Accidente Cerebrovascular/patología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antimetabolitos , Bromodesoxiuridina , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Electroporación , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Masculino , Proteínas de la Membrana , Ratones , Ratones Noqueados , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Recuperación de la Función , Retroviridae/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/fisiología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/fisiología , Factores de Transcripción/metabolismo , Transfección/métodos
2.
J Neurosci ; 28(47): 12477-88, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020040

RESUMEN

Inflammation influences several steps of adult neurogenesis, but whether it regulates the functional integration of the new neurons is unknown. Here, we explored, using confocal microscopy and whole-cell patch-clamp recordings, whether a chronic inflammatory environment affects the morphological and electrophysiological properties of new dentate gyrus granule cells, labeled with a retroviral vector encoding green fluorescent protein. Rats were exposed to intrahippocampal injection of lipopolysaccharide, which gave rise to long-lasting microglia activation. Inflammation caused no changes in intrinsic membrane properties, location, dendritic arborization, or spine density and morphology of the new cells. Excitatory synaptic drive increased to the same extent in new and mature cells in the inflammatory environment, suggesting increased network activity in hippocampal neural circuitries of lipopolysaccharide-treated animals. In contrast, inhibitory synaptic drive was more enhanced by inflammation in the new cells. Also, larger clusters of the postsynaptic GABA(A) receptor scaffolding protein gephyrin were found on dendrites of new cells born in the inflammatory environment. We demonstrate for the first time that inflammation influences the functional integration of adult-born hippocampal neurons. Our data indicate a high degree of synaptic plasticity of the new neurons in the inflammatory environment, which enables them to respond to the increase in excitatory input with a compensatory upregulation of activity and efficacy at their afferent inhibitory synapses.


Asunto(s)
Hipocampo/patología , Inflamación/patología , Inflamación/fisiopatología , Neurogénesis/fisiología , Neuronas/fisiología , Análisis de Varianza , Animales , Proteínas de Unión al Calcio/metabolismo , Espinas Dendríticas/fisiología , Relación Dosis-Respuesta en la Radiación , Ectodisplasinas/metabolismo , Estimulación Eléctrica , Electroencefalografía/métodos , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Excitadores/efectos de la radiación , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Inflamación/inducido químicamente , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Potenciales Postsinápticos Inhibidores/efectos de la radiación , Lipopolisacáridos , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Proteínas de Microfilamentos , Microscopía Confocal/métodos , Neurogénesis/efectos de los fármacos , Neuronas/citología , Técnicas de Placa-Clamp/métodos , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Tetrodotoxina/farmacología , Factores de Tiempo
3.
J Neurosci ; 26(10): 2738-44, 2006 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-16525053

RESUMEN

Hyperactivation of the cyclin-dependent kinase 5 (cdk5), triggered by proteolytic conversion of its neuronal activator, p35, to a more potent byproduct, p25, has been implicated in Alzheimer's disease (AD), amyotrophic lateral sclerosis, and Niemann-Pick type C disease (NPC). This mechanism is thought to lead to the development of neuropathological hallmarks, i.e., hyperphosphorylated cytoskeletal proteins, neuronal inclusions, and neurodegeneration, that are common to all three diseases. This pathological ensemble is recapitulated in a single model, the npc-1 (npc(-/-)) mutant mouse. Previously, we showed that pharmacological cdk inhibitors dramatically reduced hyperphosphorylation, lesion formation, and locomotor defects in npc(-/-) mice, suggesting that cdk activity is required for NPC pathogenesis. Here, we used genetic ablation of the p35 gene to examine the specific involvement of p35, p25, and hence cdk5 activation in NPC neuropathogenesis. We found that lack of p35/p25 does not slow the onset or progression or improve the neuropathology of NPC. Our results provide direct evidence that p35/p25-mediated cdk5 deregulation is not essential for NPC pathology and suggest that similar pathology in AD may also be cdk5 independent.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neuronas/patología , Enfermedades de Niemann-Pick/metabolismo , Enfermedades de Niemann-Pick/patología , Fosfotransferasas/fisiología , Proteínas tau/metabolismo , Factores de Edad , Animales , Conducta Animal , Western Blotting/métodos , Encéfalo/metabolismo , Encéfalo/patología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Inmunohistoquímica/métodos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Actividad Motora/fisiología , Proteínas de Neurofilamentos/metabolismo , Neuronas/metabolismo , Proteína Niemann-Pick C1 , Proteínas Nucleares/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Fosforilación , Fosfotransferasas/deficiencia , Proteínas/genética , Proteínas/metabolismo , Pérdida de Peso/fisiología , Región del Complejo T del Genoma
4.
J Neurosci ; 26(38): 9703-12, 2006 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-16988041

RESUMEN

Tumor necrosis factor-alpha (TNF-alpha) is a proinflammatory cytokine, acting through the TNF-R1 and TNF-R2 receptors. The two receptors have been proposed to mediate distinct TNF-alpha effects in the CNS, TNF-R1 contributing to neuronal damage and TNF-R2 being neuroprotective. Whether TNF-alpha and its receptors play any role for neurogenesis in the adult brain is unclear. Here we used mouse models with loss of TNF-R1 and TNF-R2 function to establish whether signaling through these receptors could influence hippocampal neurogenesis in vivo under basal conditions, as well as after status epilepticus (SE), which is associated with inflammation and elevated TNF-alpha levels. Notably, in the intact brain, the number of new, mature hippocampal neurons was elevated in TNF-R1(-/-) and TNF-R1/R2(-/-) mice, whereas no significant changes were detected in TNF-R2(-/-) mice. Also after SE, the TNF-R1(-/-) and TNF-R1/R2(-/-) mice produced more new neurons. In contrast, the TNF-R2(-/-) mice showed reduced SE-induced neurogenesis. Cell proliferation in the dentate subgranular zone was elevated in TNF-R1(-/-) and TNF-R1/R2(-/-) mice both under basal conditions and after SE. The TNF-R2(-/-) mice either showed no change or minor decrease of cell proliferation. TNF-R1 and TNF-R2 receptors were expressed by hippocampal progenitors, as assessed with reverse transcription-PCR on sorted or cultured cells and immunocytochemistry on cultures. Our data reveal differential actions of TNF-R1 and TNF-R2 signaling in adult hippocampal neurogenesis and identify for the first time TNF-R1 as a negative regulator of neural progenitor proliferation in both the intact and pathological brain.


Asunto(s)
Proliferación Celular , Inhibidores de Crecimiento/fisiología , Hipocampo/citología , Hipocampo/fisiología , Neuronas/citología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Células Madre/citología , Células Madre/fisiología , Animales , Células Cultivadas , Inhibidores de Crecimiento/genética , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología
5.
J Cereb Blood Flow Metab ; 28(9): 1574-87, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18493257

RESUMEN

Stroke induced by middle cerebral artery occlusion leads to transiently increased progenitor proliferation in the subventricular zone (SVZ) and long-lasting striatal neurogenesis in adult rodents. Tumor necrosis factor-alpha (TNF-alpha) is upregulated in stroke-damaged brain. Whether TNF-alpha and its receptors influence SVZ progenitor proliferation after stroke is unclear. Here we show that the increased proliferation 1 week after stroke occurred concomitantly with elevated microglia numbers and TNF-alpha and TNF receptor-1 (TNF-R1) gene expression in the SVZ of wild-type mice. TNF receptor-1 was expressed on sorted SVZ progenitor cells from nestin-green fluorescent protein reporter mice. In animals lacking TNF-R1, stroke-induced SVZ cell proliferation and neuroblast formation were enhanced. In contrast, deletion of TNF-R1 did not alter basal or status epilepticus-stimulated cell proliferation in SVZ. Addition of TNF-alpha reduced the size and numbers of SVZ neurospheres through a TNF-R1-dependent mechanism without affecting cell survival. Our results provide the first evidence that TNF-R1 is a negative regulator of stroke-induced SVZ progenitor proliferation. Blockade of TNF-R1 signaling might be a novel strategy to promote the proliferative response in SVZ after stroke.


Asunto(s)
Proliferación Celular , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Células Madre/citología , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Supervivencia Celular , Infarto de la Arteria Cerebral Media , Ventrículos Laterales , Ratones , Ratones Noqueados
6.
Neurobiol Dis ; 23(1): 23-35, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16531049

RESUMEN

Partial and generalized status epilepticus (pSE and gSE) trigger the same level of progenitor cell proliferation in adult dentate gyrus, but survival of new neurons is poor after gSE. Here, we show markedly elevated levels of prostaglandin E2 (PGE2) and brain-derived neurotrophic factor (BDNF) in rat hippocampal formation at 7 days following pSE but not gSE. Administration of the cyclooxygenase (COX) inhibitor flurbiprofen for 1 week, starting at day 8 post-SE, abated PGE2 and decreased BDNF levels, but did not affect survival of new neurons 4 weeks later. Thus, high PGE2 and BDNF levels induced by pSE are probably not of major importance for survival of new neurons during the first days after formation. We propose that they modulate other aspects of synaptic and cellular plasticity, and thereby may influence epileptogenesis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Dinoprostona/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/citología , Estado Epiléptico/fisiopatología , Animales , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Proliferación Celular , Supervivencia Celular , Inhibidores de la Ciclooxigenasa/farmacología , Flurbiprofeno/farmacología , Inmunohistoquímica , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Convulsiones/fisiopatología , Células Madre/citología , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA