Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Dev Neurosci ; 39(6): 507-518, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29073621

RESUMEN

Prenatal exposure to the antiepileptic, mood-stabilizing drug, valproic acid (VPA), increases the incidence of autism spectrum disorders (ASDs); in utero administration of VPA to pregnant rodents induces ASD-like behaviors such as repetitive, stereotyped activity, and decreased socialization. In both cases, males are more affected than females. We previously reported that VPA, administered to pregnant mice at gestational day 12.5, rapidly induces a transient, 6-fold increase in BDNF (brain-derived neurotrophic factor) protein and mRNA in the fetal brain. Here, we investigate sex differences in the induction of Bdnf expression by VPA as well as the underlying epigenetic mechanisms. We found no sex differences in the VPA stimulation of total brain Bdnf mRNA as indicated by probing for the BDNF protein coding sequence (exon 9); however, stimulation of individual transcripts containing two of the nine 5'-untranslated exons (5'UTEs) in Bdnf (exons 1 and 4) by VPA was greater in female fetal brains. These Bdnf transcripts have been associated with different cell types or subcellular compartments within neurons. Since VPA is a histone deacetylase inhibitor, covalent histone modifications at Bdnf 5'UTEs in the fetal brain were analyzed by chromatin immunoprecipitation. VPA increased the acetylation of multiple H3 and H4 lysine residues in the vicinity of exons 1, 2, 4, and 6; minimal differences between the sexes were observed. H3 lysine 4 trimethylation (H3K4me3) at those exons was also stimulated by VPA. Moreover, the VPA-induced increase in H3K4me3 at exons 1, 4, and 6 was significantly greater in females than in males, i.e., sexually dimorphic stimulation of H3K4me3 by VPA correlated with Bdnf transcripts containing exons 1 and 4, but not 6. Neither H3K27me3 nor cytosine methylation at any of the 117 CpGs in the vicinity of the transcription start sites of exons 1, 4, and 6 was affected by VPA. Thus, of the 6 epigenetic marks analyzed, only H3K4me3 can account for the sexually dimorphic expression of Bdnf transcripts induced by VPA in the fetal brain. Preferential expression of exon 1- and exon 4-Bdnf transcripts in females may contribute to sex differences in ASDs by protecting females from the adverse effects of genetic variants or environmental factors such as VPA on the developing brain.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Caracteres Sexuales , Ácido Valproico/farmacología , Animales , Trastorno del Espectro Autista/inducido químicamente , Encéfalo/crecimiento & desarrollo , Inmunoprecipitación de Cromatina/métodos , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Femenino , Ratones Endogámicos C57BL , Neuronas/metabolismo
2.
Mol Cell Neurosci ; 59: 57-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24480134

RESUMEN

Human fetal exposure to valproic acid (VPA), a widely-used anti-epileptic and mood-stabilizing drug, leads to an increased incidence of behavioral and intellectual impairments including autism; VPA administration to pregnant rats and mice at gestational days 12.5 (E12.5) or E13.5 leads to autistic-like symptoms in the offspring and is widely used as an animal model for autism. We report here that this VPA administration protocol transiently increased both BDNF mRNA and BDNF protein levels 5-6-fold in the fetal mouse brain. VPA exposure in utero induced smaller increases in the expression of mRNA encoding the other neurotrophins, NT3 (2.5-fold) and NT4 (2-fold). Expression of the neurotrophin receptors, trkA, trkB and trkC were minimally affected, while levels of the low-affinity neurotrophin receptor, p75(NTR), doubled. Of the nine 5'-untranslated exons of the mouse BDNF gene, only expression of exons I, IV and VI was stimulated by VPA in utero. In light of the well-established role of BDNF in regulating neurogenesis and the laminar fate of postmitotic neurons in the developing cortex, an aberrant increase in BDNF expression in the fetal brain may contribute to VPA-induced cognitive disorders by altering brain development.


Asunto(s)
Trastorno Autístico/inducido químicamente , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Feto/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Trastorno Autístico/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Feto/metabolismo , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Ácido Valproico/toxicidad
3.
Res Sq ; 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38260618

RESUMEN

There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 255 genes that have been reported to play fundamental roles in fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The set of genes meeting these criteria provides potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.

4.
bioRxiv ; 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37205520

RESUMEN

There is an increased incidence of autism among the children of women who take the anti-epileptic, mood stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNAseq data ob-tained from E12.5 fetal mouse brains 3 hours after VPA administration revealed that VPA significant-ly increased or decreased the expression of approximately 7,300 genes. No significant sex differ-ences in VPA-induced gene expression were observed. Expression of genes associated with neu-rodevelopmental disorders (NDDs) such as autism as well as neurogenesis, axon growth and syn-aptogenesis, GABAergic, glutaminergic and dopaminergic synaptic transmission, perineuronal nets, and circadian rhythms was dysregulated by VPA. Moreover, expression of 399 autism risk genes was significantly altered by VPA as was expression of 252 genes that have been reported to play fundamental roles in the development of the nervous system but are not otherwise linked to autism. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity in the postnatal and adult brain. The set of genes meeting these criteria pro-vides potential targets for future hypothesis-driven approaches to elucidating the proximal underly-ing causes of defective brain connectivity in NDDs such as autism.

5.
Neuron ; 51(1): 21-8, 2006 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-16815329

RESUMEN

Imbalances in neurotrophins or their high-affinity Trk receptors have long been reported in neurodegenerative diseases. However, a molecular link between these gene products and neuronal cell death has not been established. In the trisomy 16 (Ts16) mouse there is increased apoptosis in the cortex, and hippocampal neurons undergo accelerated cell death that cannot be rescued by administration of brain-derived neurotrophic factor (BDNF). Ts16 neurons have normal levels of the TrkB tyrosine kinase receptor but an upregulation of the TrkB.T1 truncated receptor isoform. Here we show that restoration of the physiological level of the TrkB.T1 receptor by gene targeting rescues Ts16 cortical cell and hippocampal neuronal death. Moreover, it corrects resting Ca2+ levels and restores BDNF-induced intracellular signaling mediated by full-length TrkB in Ts16 hippocampal neurons. These data provide a direct link between neuronal cell death and abnormalities in Trk neurotrophin receptor levels.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/metabolismo , Degeneración Nerviosa/genética , Neuronas/metabolismo , Receptor trkB/genética , Trisomía/genética , Animales , Apoptosis/genética , Encéfalo/citología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Marcación de Gen/métodos , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Peso Molecular , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Recuperación de la Función/genética , Transducción de Señal/genética , Trisomía/fisiopatología
6.
J Neurosci ; 29(40): 12702-10, 2009 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-19812345

RESUMEN

cAMP can stimulate the transcription of many activity-dependent genes via activation of the transcription factor, cAMP response element-binding protein (CREB). However, in mouse cortical neuron cultures, prior to synaptogenesis, neither cAMP nor dopamine, which acts via cAMP, stimulated CREB-dependent gene transcription when NR2B-containing NMDA receptors (NMDARs) were blocked. Stimulation of transcription by cAMP was potentiated by inhibitors of excitatory amino acid uptake, suggesting a role for extracellular glutamate or aspartate in cAMP-induced transcription. Aspartate was identified as the extracellular messenger: enzymatic scavenging of l-aspartate, but not glutamate, blocked stimulation of CREB-dependent gene transcription by cAMP; moreover, cAMP induced aspartate but not glutamate release. Together, these results suggest that cAMP acts via an autocrine or paracrine pathway to release aspartate, which activates NR2B-containing NMDARs, leading to Ca(2+) entry and activation of transcription. This cAMP/aspartate/NMDAR signaling pathway may mediate the effects of transmitters such as dopamine on axon growth and synaptogenesis in developing neurons or on synaptic plasticity in mature neural networks.


Asunto(s)
Ácido Aspártico/metabolismo , Comunicación Autocrina/fisiología , AMP Cíclico/metabolismo , Dopamina/metabolismo , Neuronas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transcripción Genética/fisiología , Secuencia de Aminoácidos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Calcio/metabolismo , Canales de Calcio/metabolismo , Técnicas de Cultivo de Célula , Corteza Cerebral/embriología , Corteza Cerebral/fisiología , Colforsina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Datos de Secuencia Molecular , Plasticidad Neuronal/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-fos/genética , Activación Transcripcional/fisiología
7.
Glia ; 57(8): 828-34, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19031446

RESUMEN

Astrocytes participate in signaling via Ca(2+) transients that spread from cell to cell across a multicellular syncytium. The effect, if any, of these Ca(2+) waves on the transcription of Ca(2+)/cAMP-regulatory element binding protein (CREB)-dependent genes is not known. We report here that, unlike neurons, increasing intracellular Ca(2+) in cultured mouse cortical astrocytes failed to activate CREB-dependent transcription, even though CREB was phosphorylated at serine 133. In contrast, both CREB phosphorylation and CREB-dependent transcription were robustly stimulated by increasing cAMP. The failure of Ca(2+)-activated transcription in astrocytes was correlated with the absence of CaMKIV, a Ca(2+)-dependent protein kinase required for Ca(2+)-stimulated gene transcription in neurons. The inability of Ca(2+) to signal via CaMKIV may insulate CREB-dependent gene transcription in astrocytes from activation by Ca(2+) waves.


Asunto(s)
Astrocitos/metabolismo , Proteína de Unión a CREB/metabolismo , Calcio/metabolismo , Adenosina Trifosfato/farmacología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Proteína de Unión a CREB/genética , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Quelantes/farmacología , AMP Cíclico/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Inhibidores Enzimáticos/farmacología , Ionomicina/farmacología , Ionóforos/farmacología , Ratones , Ratones Endogámicos C57BL , Ácido Ocadaico/farmacología , Transfección/métodos
8.
J Neurosci ; 22(7): 2571-8, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11923422

RESUMEN

The neurotrophin, brain derived neurotrophic factor (BDNF), exerts multiple effects on the development and maintenance of the nervous system, including regulating synaptic plasticity and promoting neuron survival. Here we report the selective failure of BDNF-dependent survival in cultured hippocampal neurons from the trisomy 16 (Ts16) mouse, an animal model of Down syndrome. This failure is accompanied by overexpression of a truncated, kinase-deficient isoform (T1) of the BDNF receptor tyrosine receptor kinase B (trkB). Adenovirus-mediated introduction of exogenous full-length trkB into Ts16 neurons fully restored BDNF-dependent survival, whereas exogenous truncated trkB expression in normal, euploid neurons reproduced the Ts16 BDNF signaling failure. Thus, the failure of Ts16 neurons to respond to BDNF is caused by dysregulation of trkB isoform expression. Such a neurotrophin signaling defect could contribute to developmental and degenerative disorders of the nervous system.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas/metabolismo , Trisomía/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/metabolismo , Inmunohistoquímica , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Receptor trkB/metabolismo , Receptor trkC/biosíntesis , Transducción de Señal/efectos de los fármacos
9.
Mol Cell Neurosci ; 35(3): 447-55, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17553693

RESUMEN

Brain-derived neurotrophic factor (BDNF) signaling through its receptor, trkB, is essential for the proper development and function of the nervous system. Here we identify a novel regulatory element designated TCaRE3 (TRKB Ca(2+) response element 3) required for CREB-dependent TRKB transcription in neurons. TCaRE3-inactivating mutations abolished both Ca(2+)- and cAMP-stimulated TRKB expression, despite the presence of upstream CREs. TCaRE3 mutations also reduced basal expression by at least 80%. Electrophoretic mobility shift assays revealed the presence of a neuronal nuclear factor able to bind TCaRE3 in a sequence-specific manner and we have identified krüppel-like factor 7 (KLF7) as a candidate TCaRE3 transcription factor. Importantly, despite limited overall sequence homology between the promoter regions of the human and mouse TRKB genes, TCaRE3 exhibits 100% sequence identity. Mutation analysis of the human TRKB promoter region demonstrated that the role of TCaRE3 is also conserved, suggesting that the functional interaction between CREB bound to the CREs and KLF7 bound to TCaRE3 is essential for the proper regulation of TRKB in neurons.


Asunto(s)
Calcio/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptor trkB/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Inmunoprecipitación de Cromatina/métodos , Embrión de Mamíferos , Humanos , Ratones , Mutación/fisiología , Neuronas/metabolismo , Regiones Promotoras Genéticas/fisiología , Unión Proteica/fisiología , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transducción de Señal , Transcripción Genética , Transfección/métodos
10.
J Neurochem ; 103(2): 761-70, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17666045

RESUMEN

Cyclic AMP response element binding protein (CREB) functions as an activity-dependent transcription factor in the nervous system. Increases in intracellular Ca(2+) due to neuronal activity lead to the phosphorylation and subsequent activation of CREB. Although phosphorylation of CREB at Ser-133 is necessary for the stimulation of transcriptional activity, it is not sufficient. Here we demonstrate that in mouse cortical neurons, inhibition of the Ca(2+)-dependent protein phosphatase calcineurin by FK506 or cyclosporine A blocks CREB-dependent gene expression induced by depolarization without inhibiting depolarization-induced Ca(2+) influx or CREB Ser-133 phosphorylation. Over-expression of a constitutively-active allele of the transducer of regulated CREB activity could not bypass the requirement for calcineurin activity. Stimulation of a CRE-luciferase reporter gene by depolarization was sensitive to FK506 throughout the entire time course of the transcriptional response, revealing that calcineurin activity is required to maintain CREB-dependent transcription. Stimulation of CRE-luciferase expression by forskolin and 8-Br-cAMP also required calcineurin activity. These results suggest that calcineurin functions as a critical determinant in shaping genome responses to CREB activation in cortical neurons.


Asunto(s)
Calcineurina/metabolismo , Corteza Cerebral/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Neuronas/metabolismo , Transcripción Genética/fisiología , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Inhibidores de la Calcineurina , Calcio/metabolismo , Polaridad Celular/fisiología , Células Cultivadas , Corteza Cerebral/citología , AMP Cíclico/fisiología , Genes Reporteros/efectos de los fármacos , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Fosforilación , ARN/biosíntesis , ARN/genética , Receptor trkB/biosíntesis , Receptor trkB/genética , Tacrolimus/farmacología , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
11.
J Bioenerg Biomembr ; 38(1): 43-7, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16786428

RESUMEN

Isolated brain mitochondria are a heterogeneous mixture from different cell types and these subsets may have differing sensitivities to Ca2+-induced membrane permeability transition (MPT) and to inhibition of the MPT by cyclosporin A (CsA). This study tested the hypothesis that mitochondria within primary cultures of astrocytes and neurons exhibit different energy-dependent Ca2+ uptake capacities and different degrees to which CsA increases their uptake capacity. Astrocytes and neurons were suspended in a cytosol-like medium containing respiratory substrates, ATP, and Mg2+ in the presence of digitonin to selectively permeabilize the plasma membrane. Uptake of added Ca2+ by mitochondria within the cells was measured by Calcium Green 5N fluorescent monitoring of the medium [Ca2+]. Permeabilized astrocytes had a fourfold higher Ca2+ uptake capacity, relative to neurons and a twofold higher content based on relative contents of mitochondria assessed by measurements of mitochondrial DNA and cytochrome oxidase subunit 1 protein. In astrocytes the Ca2+ uptake capacity was increased twofold by preincubation with 2-5 microM CsA, while in neurons CsA had no effect. Similar results were obtained using measurements of the effects of added Ca2+ on mitochondrial membrane potential. FK506, a drug similar to CsA but without MPT inhibitory activity, had no effect on either cell type. These results are consistent with the presence of a calcium-induced MPT in astrocytes, even in the presence of ATP, and indicate that the MPT in cerebellar granule neurons is resistant to CsA inhibition. Some of the protective effects of CsA in vivo may therefore be mediated by preservation of mitochondrial functional integrity within astrocytes.


Asunto(s)
Astrocitos/efectos de los fármacos , Calcio/fisiología , Ciclosporina/farmacología , Inmunosupresores/farmacología , Mitocondrias/fisiología , Neuronas/efectos de los fármacos , Animales , Astrocitos/metabolismo , Permeabilidad de la Membrana Celular , Células Cultivadas , Cerebelo/citología , Corteza Cerebral/citología , Digitonina/farmacología , Complejo IV de Transporte de Electrones/metabolismo , Transporte Iónico , Potenciales de la Membrana , Membranas Mitocondriales/fisiología , Neuronas/metabolismo , Compuestos Orgánicos/química , Permeabilidad , Ratas , Ratas Sprague-Dawley , Tacrolimus/farmacología
12.
Eur J Neurosci ; 21(12): 3251-61, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16026463

RESUMEN

GABA(A) receptor activation during brain development is a critical source of excitation. This is due to the positive equilibrium potential for chloride relative to resting membrane potential, resulting in membrane depolarization sufficient to open voltage sensitive calcium channels. The gonadal steroid estradiol has pronounced trophic effects on the developing hippocampus, promoting cell survival and synaptogenesis. In the current study, we investigated the effect of estradiol on GABA(A) receptor-mediated calcium transients in cultured neonatal hippocampal neurons, from Sprague-Dawley rats, using the calcium sensitive dye, Fura-2-AM. Treatment of hippocampal neurons with physiological levels of estradiol significantly increased the peak amplitude of calcium transients, increased the number of cells responding to the GABA(A) agonist muscimol with membrane depolarization, and delayed the rate of clearance of free intracellular calcium. These effects were significantly attenuated by pretreatment with the oestrogen receptor antagonist ICI-182,780. This suggests that estradiol, via its action on the oestrogen receptor, prolongs the developmental duration of depolarizing GABA. Estradiol likely maintains GABA-mediated excitation by promoting increased protein levels of the active/phosphorylated form of the chloride cotransporter Na+K+2CL- and L-type voltage sensitive calcium channels containing the alpha1C subunit. We propose that a component of the trophic effects of estradiol on hippocampal development results from enhanced calcium influx subsequent to GABA(A) receptor activation.


Asunto(s)
Estradiol/administración & dosificación , Hipocampo/citología , Hipocampo/enzimología , Neuronas/efectos de los fármacos , Receptores de GABA-A/fisiología , Animales , Bicuculina/farmacología , Western Blotting/métodos , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Diagnóstico por Imagen/métodos , Diltiazem/farmacología , Maleato de Dizocilpina/farmacología , Esquema de Medicación , Interacciones Farmacológicas , Embrión de Mamíferos , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Fura-2/análogos & derivados , Fura-2/metabolismo , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Ácido Glutámico/farmacología , Muscimol/farmacología , Neuronas/metabolismo , Nimodipina/farmacología , Cloruro de Potasio/farmacología , Embarazo , Quinoxalinas/farmacología , Ratas , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12 , Simportadores/metabolismo , Factores de Tiempo , Cotransportadores de K Cl
13.
J Neurosci Res ; 73(1): 89-94, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12815712

RESUMEN

Genes from the Down syndrome (DS) critical region of human chromosome 21, which contribute to the pathology of DS, are also found on mouse chromosome 16. Several animal models of DS with triplication of genes from the DS critical region have been generated, including mouse trisomy 16 (Ts16) and a partial trisomic mouse, Ts65Dn. Using computer-assisted imaging of fura-2 fluorescence, we found an elevation of intracellular cytoplasmic calcium in cortical astrocytes from neonatal Ts65Dn mouse brain, similar to that observed previously in embryonic Ts16 astrocytes. Furthermore, astrocytes from both Ts65Dn and Ts16 cortex fail to respond to the anti-proliferative actions of glutamate. These results suggest that defective regulation of cell proliferation and cellular calcium can result from triplication of DS critical region genes.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Calcio/metabolismo , Síndrome de Down/metabolismo , Síndrome de Down/patología , Animales , Animales Recién Nacidos , División Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Ácido Glutámico/farmacología , Homeostasis , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Embarazo , Trisomía
14.
J Biol Chem ; 278(42): 40744-8, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-12900419

RESUMEN

The neurotrophin brain-derived neurotrophic factor (BDNF), via activation of its receptor, tyrosine receptor kinase B (trkB), regulates a wide variety of cellular processes in the nervous system, including neuron survival and synaptic plasticity. Although the expression of BDNF is known to be Ca2+-dependent, the regulation of trkB expression has not been extensively studied. Here we report that depolarization of cultured mouse cortical neurons increased the expression of the full-length, catalytically active isoform of trkB without affecting expression of the truncated isoform. This increase in protein expression was accompanied by increased levels of transcripts encoding full-length, but not truncated, trkB. Depolarization also regulated transcription of the gene, TRKB, via entry of Ca2+ through voltage-gated Ca2+ channels and subsequent activation of Ca2+-responsive elements in the two TRKB promoters. Using transient transfection of neurons with TRKB promoter-luciferase constructs, we found that Ca2+ inhibited the upstream promoter P1 but activated the downstream promoter P2. Ca2+-dependent stimulation of TRKB expression requires two adjacent, non-identical CRE sites located within P2. The coordinated regulation of BDNF and trkB by Ca2+ may play a role in activity-dependent survival and synaptic plasticity by enhancing BDNF signaling in electrically active neurons.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Calcio/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Receptor trkB/metabolismo , Animales , Sitios de Unión , Células Cultivadas , AMP Cíclico/metabolismo , Regulación hacia Abajo , Luciferasas/metabolismo , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Plásmidos/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Transfección
15.
Dev Neurosci ; 26(2-4): 255-65, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15711065

RESUMEN

During embryonic development of the mammalian cerebral cortex, the generation of the marginal zone (MZ) and subplate (SP) precedes that of the cortical plate (CP). MZ and SP neurons are believed to play a 'pioneering' role in directing the organization of the CP and the specificity of connections between the CP and other brain regions. Here we report that this sequential order of neurogenesis is disrupted in the trisomy 16 (Ts16) mouse, a potential animal model of Down syndrome. Bromodeoxyuridine labeling was used to establish the date of generation of postmitotic SP and CP neurons in the somatosensory cortex. As has been previously reported, most SP neurons in euploid (control) cortex were generated on embryonic day 12.5 (E12.5), and production of CP neurons began a day later. In contrast, in the Ts16 cortex, few SP neurons were born on E12.5 and most were generated on E13.5 and E14.5 when CP neurons were also being produced. Thus, in the Ts16 cortex, many CP neurons are born and arrive at their destinations before the normal complement of SP neurons is present. This disruption of the temporal sequence of SP and CP generation may, therefore, interfere with the pioneering functions of the SP during cortical neurogenesis and may alter the connectivity of the cortex. Indeed, using lipophilic membrane tracers to label axonal projections, we found very little thalamocortical innervation of the Ts16 SP at an age when there is extensive innervation of the euploid SP.


Asunto(s)
Diferenciación Celular/genética , Corteza Cerebral/anomalías , Malformaciones del Sistema Nervioso/genética , Neuronas/metabolismo , Células Madre/metabolismo , Trisomía/genética , Animales , Carbocianinas , Movimiento Celular/genética , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/fisiopatología , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/fisiopatología , Vías Nerviosas/anomalías , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Células Madre/citología , Tálamo/anomalías , Tálamo/citología , Tálamo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA