Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 9: 253, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26217181

RESUMEN

We derived adult neural stem/progenitor cells (NSPCs) from the sub-ventricular zone of male and female mice to examine direct responses to principal sex hormones. In the presence of epidermal growth factor (EGF) and fibroblast growth factor-2 (FGF2) NSPCs of both sexes expressed nestin and sox2, and could be maintained as neurospheres without addition of any sex hormones. The reverse was not observed; neither testosterone (T), 17ß-estradiol (E2) nor progesterone (P4) was able to support neurosphere growth in the absence of EGF and FGF2. Ten nanomolar T, E2 or P4 induced nestin(+) cell proliferation within 20 min and enhanced neurosphere growth over 7 days irrespective of sex, which was abolished by Erk inhibition with 20 µM U0126. Maintaining neurospheres with each sex hormone did not affect subsequent neuronal differentiation. However, 10 nM T, E2 or P4 added during differentiation increased ßIII tubulin(+) neuron production with E2 being more potent compared to T and P4 in both sexes. Androgen receptor (AR) inhibition with 20 µM flutamide but not aromatase inhibition with 10 µM letrozole reduced basal and T-induced neurosphere growth in females, while only concurrent inhibition of AR and aromatase produced the same effect in males. This sex-specific effect was supported by higher aromatase expression in male neurospheres compared to females measured by Western blot and green fluorescent protein (GFP) reporter. Ten micromolar menadione induced oxidative stress, impaired neurosphere growth and up-regulated aromatase expression in both sexes. However, under oxidative stress letrozole significantly exacerbated impaired neurosphere growth in males only. While both E2 and T could prevent oxidative stress-induced growth reduction in both sexes, the effects of T were dependent on innate aromatase activity. We show for the first time that intrinsic androgen and estrogen signaling may impact the capacity of NSPCs to produce neural progenitors under pathological conditions of oxidative stress.

2.
Neurobiol Dis ; 69: 248-62, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24825316

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by a tandem repeat mutation in the huntingtin gene. Lifestyle factors, such as lack of activity may contribute to the variability in the age of disease onset. Therefore, better understanding of environmental modifiers may uncover potential therapeutic approaches to delay disease onset and progression. Recent data suggest that HD patients and transgenic mouse models show a dysregulated stress response. In this present study, we elevated stress hormone levels through oral corticosterone (CORT) treatment and assessed its impact on the development of motor impairment and cognitive deficits using the R6/1 transgenic mouse model of HD. We found that CORT consumption did not alter rotarod performance of R6/1 HD or wild-type (WT) littermates. However, the onset of hippocampal-dependent Y-maze deficits was accelerated in male R6/1 mice by 5days of CORT treatment, whereas short term memory of WT and female R6/1 mice was unaffected. We then further investigated the male HD susceptibility to CORT by measuring TrkB activation, BDNF and glucocorticoid receptor expression as well as the level of cell proliferation in the hippocampus. CORT treatment increased the levels of phosphorylated TrkB in male R6/1 mice only. There were no effects of CORT on hippocampal BDNF protein or mRNA levels; nor on expression of the glucocorticoid receptors in any group. Hippocampal cell proliferation was decreased in male R6/1 mice and this was further reduced in CORT-drinking male R6/1 mice. Female mice (WT and R6/1) appeared to be protected from the impacts of CORT treatment in all our hippocampal measures. Overall, our data demonstrate that treatment with corticosterone is able to modulate the onset of HD symptomatology. We present the first evidence of a male-specific vulnerability to stress impacting on the development of short-term memory deficits in HD. More generally, we found that female mice were protected from the detrimental effects of CORT treatment on a variety of hippocampus-based measures. Hippocampal plasticity and memory in HD may be more susceptible to the impacts of stress in a sex-dependent manner. We propose clinical investigations of stress as a key environmental modifier of HD symptom onset.


Asunto(s)
Corticosterona/metabolismo , Enfermedad de Huntington/fisiopatología , Trastornos de la Memoria/fisiopatología , Memoria a Corto Plazo/fisiología , Edad de Inicio , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones Transgénicos , Actividad Motora/fisiología , ARN Mensajero/metabolismo , Receptor trkB/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Caracteres Sexuales
3.
Mol Cell Neurosci ; 54: 93-107, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23384443

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disorder affecting a range of cellular and molecular functions in the brain. Deficits in adult hippocampal neurogenesis (AHN) have been documented in the R6/1 mouse model of HD. Here we examined basal and running-induced neuronal precursor proliferation in adult female and male R6/1 HD mice. We further tested whether sequential delivery of voluntary running followed by environmental enrichment could synergistically enhance functional AHN in female R6/1 HD mice. R6/1 HD mice engaged in significantly reduced levels of voluntary running, with males showing a more severe deficit. Basal neural precursor proliferation in the hippocampal sub-granular zone remained unchanged between female and male R6/1 HD mice and neither sex significantly responded to running-induced proliferation. While discrete provision of running wheels and enriched environments doubled AHN in adult female R6/1 HD mice it did not reflect the significant 3-fold increase in female wildtypes. Nevertheless, triple-label c-Fos/BrdU/NeuN immunofluorescence and confocal microscopy provided evidence that the doubling of AHN in female R6/1 HD mice was functional. Intrinsic cellular dysfunction mediated by protein aggregates containing mutant huntingtin (mHtt) did not appear to coincide with AHN deficits. In the hippocampus of female R6/1 HD mice, proliferating precursors and 6 week old adult-generated neurons were devoid of mHtt immuno-reactive aggregates, as were endothelial, microglial and astroglial cells populating the neurogenic niche. Serum transforming growth factor-ß concentrations remained unaltered in female R6/1 HD mice as did the hippocampal levels of proliferating microglia and glial fibrillarly acidic protein expression. Examining the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis showed no change in base-line serum GH between genotypes. However, despite a reduced distance, acute running increases serum GH in both female wildtype and R6/1 HD mice. Serum IGF-1 levels were increased in female R6/1 HD mice compared to wildtypes during daytime inactive period, while hippocampal levels of the IGF-1 receptor remained unchanged. Running induced Akt phosphorylation in the hippocampus of female wildtype mice, which was not reflected in R6/1 HD mice. Total Akt levels were decreased in the hippocampus of both control and running R6/1 HD mice. Our results show adult-generated hippocampal neurons in female R6/1 HD mice express c-Fos and that running and Akt signaling deficits may mediate reduced basal and running-induced AHN levels.


Asunto(s)
Hipocampo/patología , Enfermedad de Huntington/metabolismo , Neurogénesis , Esfuerzo Físico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Proliferación Celular , Proteínas de Unión al ADN , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hormona del Crecimiento/sangre , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Proteína Huntingtina , Enfermedad de Huntington/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Carrera , Caracteres Sexuales , Factor de Crecimiento Transformador beta/sangre
4.
Neural Plast ; 2012: 874387, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22830053

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by a tandem repeat expansion encoding a polyglutamine tract in the huntingtin protein. HD involves progressive psychiatric, cognitive, and motor symptoms, the selective pathogenesis of which remains to be mechanistically elucidated. There are a range of different brain regions, including the cerebral cortex and striatum, known to be affected in HD, with evidence for hippocampal dysfunction accumulating in recent years. In this review we will focus on hippocampal abnormalities, in particular, deficits of adult neurogenesis. We will discuss potential molecular mechanisms mediating disrupted hippocampal neurogenesis, and how this deficit of cellular plasticity may in turn contribute to specific cognitive and affective symptoms that are prominent in HD. The generation of transgenic animal models of HD has greatly facilitated our understanding of disease mechanisms at molecular, cellular, and systems levels. Transgenic HD mice have been found to show progressive behavioral changes, including affective, cognitive, and motor abnormalities. The discovery, in multiple transgenic lines of HD mice, that adult hippocampal neurogenesis and synaptic plasticity is disrupted, may help explain specific aspects of cognitive and affective dysfunction. Furthermore, these mouse models have provided insight into potential molecular mediators of adult neurogenesis deficits, such as disrupted serotonergic and neurotrophin signaling. Finally, a number of environmental and pharmacological interventions which are known to enhance adult hippocampal neurogenesis have been found to have beneficial affective and cognitive effects in mouse models, suggesting common molecular targets which may have therapeutic utility for HD and related diseases.


Asunto(s)
Trastornos del Conocimiento/patología , Hipocampo/crecimiento & desarrollo , Hipocampo/patología , Enfermedad de Huntington/patología , Trastornos del Humor/patología , Neurogénesis/fisiología , Animales , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/complicaciones , Enfermedad de Huntington/psicología , Ratones , Ratones Transgénicos , Trastornos del Humor/etiología , Trastornos del Humor/psicología , Plasticidad Neuronal/fisiología
5.
Neurobiol Learn Mem ; 97(2): 261-70, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22261461

RESUMEN

Huntington's disease (HD) is a neurodegenerative condition characterised by progressive motor, psychological and cognitive decline. R6/1 HD transgenic mice model the clinical hippocampal-dependent cognitive deficits observed in patients. Cholinergic and GABAergic septohippocampal projections play important roles in hippocampal-dependent cognition. The current study examined neuronal activity of cholinergic and GABAergic septohippocampal projections in response to arousal elicited during differing behavioural states. The different behavioural states examined were; home cage (controls), acute exploration of a novel enriched environment and either spontaneous wakefulness (dark phase) or spontaneous sleep (light phase). We employed triple-label immunohistochemistry using c-Fos as an indirect marker of neuron activation and parvalbumin and choline acetyltransferase (ChAT) to label GABAergic and cholinergic neurons in the basal forebrain, respectively. The Y-maze was used to assess short-term hippocampal-dependent memory independently during either the dark or light phase and revealed a memory deficit in R6/1 HD mice compared to wild types that was particularly prominent during the dark phase. Three-way ANOVA of basal forebrain cholinergic and GABAergic activity through co-expression of c-Fos revealed overt responses to differing behavioural states. Both genotypes increased cholinergic neuron activity in response to exploring a novel enriched environment and also an increase during the dark phase compared to the light phase. Novel enriched environment exploration caused a larger response of GABAergic neuron activity in R6/1 HD mice, which also failed to increase the activity of GABAergic neurons during the dark phase compared to the light phase as observed for wildtype mice. Basal levels of c-Fos-positive cells were greatly increased in the hippocampal granule cell layer of R6/1 HD mice during both circadian phases. The differential activation of septohippocampal cholinergic and GABAergic neurons in R6/1 HD mice in response to differing behavioural states may be associated with impaired hippocampal-dependent short-term memory.


Asunto(s)
Conducta Animal/fisiología , Neuronas Colinérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Enfermedad de Huntington/metabolismo , Animales , Cognición/fisiología , Modelos Animales de Enfermedad , Ambiente , Conducta Exploratoria/fisiología , Enfermedad de Huntington/genética , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratones , Ratones Transgénicos , Actividad Motora/fisiología
6.
Neural Regen Res ; 7(28): 2227-39, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-25538744

RESUMEN

Interest surrounds the role of sex-hormones in regulating brain function outside of reproductive behaviour. Declining androgen production in aging males has been associated with cognitive impairment, depression and increased risk of developing Alzheimer's disease. Indication for testosterone replacement therapy is based on biochemically determined low circulating testosterone combined with manifest symptoms. However, which aspects of age-related cognitive decline are attributable to low circulating testosterone remain ambiguous. Studies examining cognition in aging men receiving testosterone replacement therapy have yielded equivocal results. The exact role of testosterone in maintaining cognitive function and the underlying neural mechanisms are largely unknown, though it would appear to be domain specific. Clarity in this area will provide clinical direction toward addressing an increasing healthcare burden of mental health decline coincident with increasing longevity. The premise that androgens contribute to maintaining aspects of mental health in aging men by preserving hippocampal neurogenesis will be used as a forum in this review to discuss current knowledge and the need for further studies to better define testosterone replacement strategies for aging male health.

7.
Hippocampus ; 22(2): 309-19, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21080410

RESUMEN

Schizophrenia is a devastating psychiatric illness with a complex pathophysiology. We have recently documented schizophrenia-like endophenotypes in phospholipase C-ß1 knockout (PLC-ß1(-/-)) mice, including deficits in prepulse inhibition, hyperlocomotion, and cognitive impairments. PLC-ß1 signals via multiple G-protein coupled receptor pathways implicated in neural cellular plasticity; however, adult neurogenesis has yet to be explored in this knockout model. In this study, we employed PLC-ß1(-/-) mice to elucidate possible correlates between aberrant adult hippocampal neurogenesis (AHN) and schizophrenia-like behaviors. Using stereology and bromodeoxyuridine (BrdU) immunohistochemistry we demonstrated a significant increase in the density of adult-generated cells in the granule cell layer (GCL) of adult PLC-ß1(-/-) mice compared with wild-type littermates. Cellular phenotype analysis using confocal microscopy revealed these cells to be mature granule neurons expressing NeuN and calbindin. Increased neuronal survival occurred concomitant with reduced caspase-3(+) cells in the GCL of PLC-ß1(-/-) mice. Stereological analysis of Ki67(+) cells in the subgranular zone suggested that neural precursor proliferation is unchanged in PLC-ß1(-/-) mice. We further showed aberrant migration of mature granule neurons within the GCL of adult PLC-ß1(-/-) mice with excessive adult-generated mature neurons residing in the middle and outer GCL. PLC-ß1(-/-) mice exhibited specific behavioral deficits in location recognition, a measure of hippocampal-dependent memory, but not novel object recognition. Overall, we have shown that PLC-ß1(-/-) mice have a threefold increase in net AHN, and have provided further evidence to suggest a specific deficit in hippocampal-dependent cognition. We propose that abnormal cellular plasticity in these mice may contribute to their schizophrenia-like behavioral endophenotypes.


Asunto(s)
Movimiento Celular , Hipocampo/patología , Neurogénesis , Neuronas/patología , Fosfolipasa C beta/deficiencia , Esquizofrenia/patología , Células Madre Adultas/patología , Animales , Movimiento Celular/genética , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Células-Madre Neurales/patología , Fosfolipasa C beta/genética , Esquizofrenia/enzimología , Esquizofrenia/genética
8.
Mol Cell Neurosci ; 38(4): 537-47, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18586515

RESUMEN

Erythropoietin (EPO) enhances neurogenesis, neuroprotection and regeneration. Here, we examined the effects of EPO on axonal and dendritic growth in a model of neuronal polarization. EPO did not effect survival or the polarized morphology of hippocampal neurons but its effect on neurite outgrowth depended upon the stage of polarization. When added to neurons in the process of establishing polarity (0-2 days in vitro (DIV)), it enhanced axonal and dendritic growth, while EPO added to early polarized cultures at 3-4 DIV promoted the growth of axons but not dendrites. EPO stimulated the phosphorylation of Akt at serine-473 and co-incubation of the Akt/PI-3 kinase pathway inhibitor LY294002 with EPO abolished its effects on Akt phosphorylation and axonal growth. However, while Leukemia Inhibitory Factor (LIF) similarly stimulated phosphorylation of Akt, it had no effect on axonal or dendritic growth, indicating that AKT phosphorylation is necessary but not sufficient for neurite outgrowth in hippocampal neurons.


Asunto(s)
Axones/fisiología , Polaridad Celular/fisiología , Eritropoyetina/fisiología , Modelos Neurológicos , Neuronas/citología , Neuronas/fisiología , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo , Proteínas Recombinantes
9.
Hippocampus ; 18(10): 1034-50, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18566962

RESUMEN

Adult hippocampal neurogenesis (AHN) is modulated by a variety of factors through effects on the proliferation-differentiation-survival regulatory axis. We have employed growth hormone receptor knockout (GH-R-/-) and suppressor of cytokine signaling-2 transgenic (SOCS-2 Tg) mice as models of altered GH-signaling to assess their affects on basal and exercised-induced hippocampal neurogenesis. Assessment of proliferation 24-h after 7-days of bromodeoxyuridine (BrdU) labeling with or without voluntary running showed that the density of BrdU(+) cells in the subgranular zone remained unchanged between genotypes in control housing, while running induced significant increases in BrdU-labeled cells in WT, GH-R-/-, and SOCS-2 Tg mice. The proportion of BrdU/doublecortin and BrdU/S100beta cells did not vary between genotype or running conditions at this time-point. Assessment of cell survival 28-days after BrdU labeling showed that SOCS-2 Tg animals had significantly higher BrdU(+) cell densities in the granule cell layer compared to WT and GH-R-/- animals in control housing and after voluntary running. There were no differences in cell survival between WT and GH-R-/- mice with or without running. Mature phenotype analysis showed similar proportions of BrdU/NeuN and BrdU/S100beta in all groups. While SOCS-2 Tg mice had similar social interaction behaviors and sensorimotor gating, they appeared to be less anxious with heightened basal locomotor activity and showed enhanced performance in the Morris watermaze test. Overall, our data indicated that mice over-expressing SOCS-2 showed increased survival of neurons generated during AHN, which correlated with improved performance in a hippocampal-dependent cognitive task. Furthermore, voluntary running increased AHN in WT, SOCS-2 Tg, and serum-IGF-1-deficient GH-R-/- mice.


Asunto(s)
Hormona del Crecimiento/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Neurogénesis/fisiología , Neuronas/citología , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Supervivencia Celular , Cognición/fisiología , Proteína Doblecortina , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Actividad Motora/fisiología , Neuronas/metabolismo , Receptores de Somatotropina/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
10.
Stem Cells ; 26(1): 173-81, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17901403

RESUMEN

Ongoing production of neurons in adult brain is restricted to specialized neurogenic niches. Deregulated expression of genes controlling homeostasis of neural progenitor cell division and/or their microenvironment underpins a spectrum of brain pathologies. Using conditional gene deletion, we show that the proto-oncogene c-myb regulates neural progenitor cell proliferation and maintains ependymal cell integrity in mice. These two cellular compartments constitute the neurogenic niche in the adult brain. Brains devoid of c-Myb showed enlarged ventricular spaces, ependymal cell abnormalities, and reduced neurogenesis. Neural progenitor cells lacking c-Myb showed a reduced intrinsic proliferative capacity and reduction of Sox-2 and Pax-6 expression. These data point to an important role for c-Myb in the neurogenic niche of the adult brain.


Asunto(s)
Células Madre Adultas/citología , Encéfalo/citología , Genes myb , Neuronas/citología , Neuronas/metabolismo , Células Madre Adultas/metabolismo , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Recuento de Células , Diferenciación Celular/fisiología , Proliferación Celular , Proteínas de Unión al ADN/biosíntesis , Proteínas del Ojo/biosíntesis , Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Microscopía Electrónica de Rastreo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/biosíntesis , ARN Mensajero/análisis , Proteínas Represoras/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1 , Transactivadores/biosíntesis
11.
J Neurochem ; 102(6): 1953-1965, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17555554

RESUMEN

Erythropoietin is a primary regulator of erythropoiesis in the hematopoietic system. More recently erythropoietin has been shown to play a role in neurogenesis and provide neurotrophic support to injured CNS tissue. Here the effects of large systemic doses of erythropoietin on basal levels of adult hippocampal neurogenesis in mice were examined. A 7-day period of recombinant human erythropoietin (rhEPO) administration increased the number of bromodeoxyuridine [BrdU(+)] cells in the sub-granular zone (SGZ) by 30%. Analysis of cell phenotype revealed an increase in mitotically active doublecortin(+) neuronal progenitor cells and glial fibrillary acidic protein(+) SGZ radial astrocytes/stem cells but not mature S100beta(+) astrocytes. These effects appeared to be mediated, in part, by mitogen-activated protein kinase signaling and potentially regulated by suppressor of cytokine signaling-3. Hippocampal levels of phosphorylated extracellular signal-related kinase 42/44 and suppressor of cytokine signaling-3 were increased 2-6 h after a single systemic rhEPO injection. However, rhEPO had no observed effect on the long-term survival of new born cells in the SGZ, with similar numbers of BrdU(+) cells and BrdU(+)/NeuN(+) co-labeled cells after 4 weeks. Therefore, systemically delivered rhEPO transiently increased adult hippocampal neurogenesis without any apparent long-term effects.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Eritropoyetina/farmacología , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Células Madre/efectos de los fármacos , Factores de Edad , Animales , Bromodesoxiuridina , Recuento de Células , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Eritropoyetina/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/metabolismo , Inyecciones Intraperitoneales , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Células Madre/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
12.
Brain Res ; 1067(1): 138-45, 2006 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-16360125

RESUMEN

Neuronal differentiation of neural progenitor cells is regulated by a variety of growth and transcription factors, that not only regulate cell fate of the progenitor cells but that can also regulate neuronal morphology. Suppressor of cytokine signaling-2 (SOCS2) is an intracellular regulator of Growth Hormone (GH) signaling that is expressed in neural stem cells and neurons during development and is required to overcome the inhibitory effects of GH on neuronal differentiation. SOCS2 also promotes neurite outgrowth, however, whether the mechanism by which SOCS2 regulates neuronal differentiation and neurite outgrowth is the same is not clear. Furthermore, whether the over-expression of SOCS2 has physiological in addition to morphological effects is unknown. To address these questions, we differentiated adult neural progenitor cells derived from wildtype C57BL/6 or SOCS2 over-expressing transgenic mice (SOCS2Tg) in the presence or absence of GH and determined effects on neuronal differentiation and morphology. Compared to wildtype cells, differentiation of SOCS2Tg neurospheres resulted in increased neurogenesis, which was not inhibited by GH. The neurons derived from these cells appeared more complex, with increased neurite outgrowth and number. GH did not, however, have any effect on neurite outgrowth of wildtype or SOCS2Tg neurons. Furthermore, basic electrophysiological analysis of wildtype and SOCS2Tg neurons derived from the neurospheres showed that they were both of an immature electrophysiological neuronal phenotype, indicating that although SOCS2 expression can regulate neuronal morphology, it appears to have little effect on neuronal ion channel expression.


Asunto(s)
Neuronas/citología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Diferenciación Celular , Electrofisiología , Hormona del Crecimiento/fisiología , Canales Iónicos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuritas/fisiología , Neuritas/ultraestructura , Neuronas/fisiología , Proteínas Supresoras de la Señalización de Citocinas/genética
13.
Eur J Neurosci ; 19(8): 2069-79, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15090034

RESUMEN

Recently we have shown that growth hormone (GH) inhibits neuronal differentiation and that this process is blocked by suppressor of cytokine signalling-2 (SOCS2). Here we examine several cortical and subcortical neuronal populations in GH hyper-responsive SOCS2 null (-/-) mice and GH non-responsive GH receptor null (GHR-/-) mice. While SOCS2-/- mice showed a 30% decrease in density of NeuN positive neurons in cortex compared to wildtype, GHR-/- mice showed a 25% increase even though brain size was decreased. Interneuron sub-populations were variably affected, with a slight decrease in cortical parvalbumin expressing interneurons in SOCS2-/- mice and an increase in cortical calbindin and calretinin and striatal cholinergic neuron density in GHR-/- mice. Analysis of glial cell numbers in cresyl violet or glial fibrillary acidic protein (GFAP) stained sections of cortex showed that the neuron : glia ratio was increased in GHR-/- mice and decreased in SOCS2-/- mice. The astrocytes in GHR-/- mice appeared smaller, while they were larger in SOCS2-/- mice. Neuronal soma size also varied in the different genotypes, with smaller striatal cholinergic neurons in GHR-/- mice. While the size of layer 5 pyramidal neurons was not significantly different from wildtype, SOCS2-/- neurons were larger than GHR-/- neurons. In addition, primary dendritic length was similar in all genotypes but dendritic branching of pyramidal neurons in the cortex appeared sparser in GHR-/- and SOCS2-/- mice. These results suggest that GH, possibly regulated by SOCS2, has multiple effects on central nervous system (CNS) development and maturation, regulating the number and size of multiple neuronal and glial cell types.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/deficiencia , Hormona del Crecimiento/deficiencia , Transactivadores/deficiencia , Animales , Recuento de Células , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/genética , Hormona del Crecimiento/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Proteínas Represoras/genética , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA