Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 246
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(22): e2201355119, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35613048

RESUMEN

Area-specific axonal projections from the mammalian thalamus shape unique cellular organization in target areas in the adult neocortex. How these axons control neurogenesis and early neuronal fate specification is poorly understood. By using mutant mice lacking the majority of thalamocortical axons, we show that these axons are required for the production and specification of the proper number of layer 4 neurons in primary sensory areas by the neonatal stage. Part of these area-specific roles is played by the thalamus-derived molecule, VGF. Our work reveals that extrinsic cues from sensory thalamic projections have an early role in the formation of cortical cytoarchitecture by enhancing the production and specification of layer 4 neurons.


Asunto(s)
Axones , Tipificación del Cuerpo , Corteza Cerebral , Neurogénesis , Tálamo , Animales , Axones/fisiología , Corteza Cerebral/embriología , Corteza Cerebral/ultraestructura , Ratones , Ratones Mutantes , Vías Nerviosas , Neurogénesis/genética , Neurogénesis/fisiología , Neuronas/fisiología , Tálamo/embriología , Tálamo/ultraestructura
2.
Sci Rep ; 10(1): 18443, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33116204

RESUMEN

Riboflavin transporter 3 (RFVT3), encoded by the SLC52A3 gene, is important for riboflavin homeostasis in the small intestine, kidney, and placenta. Our previous study demonstrated that Slc52a3 knockout (Slc52a3-/-) mice exhibited neonatal lethality and metabolic disorder due to riboflavin deficiency. Here, we investigated the influence of Slc52a3 gene disruption on brain development using Slc52a3-/- embryos. Slc52a3-/- mice at postnatal day 0 showed hypoplasia of the brain and reduced thickness of cortical layers. At embryonic day 13.5, the formation of Tuj1+ neurons and Tbr2+ intermediate neural progenitors was significantly decreased; no significant difference was observed in the total number and proliferative rate of Pax6+ radial glia. Importantly, the hypoplastic phenotype was rescued upon riboflavin supplementation. Thus, it can be concluded that RFVT3 contributes to riboflavin homeostasis in embryos and that riboflavin itself is required during embryonic development of the cerebral cortex in mice.


Asunto(s)
Corteza Cerebral/embriología , Proteínas de Transporte de Membrana/deficiencia , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Deficiencia de Riboflavina/embriología , Animales , Corteza Cerebral/patología , Ratones , Ratones Noqueados , Células-Madre Neurales/patología , Neuronas/patología , Deficiencia de Riboflavina/patología
3.
Nature ; 586(7828): 281-286, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32968276

RESUMEN

'Dysbiosis' of the maternal gut microbiome, in response to challenges such as infection1, altered diet2 and stress3 during pregnancy, has been increasingly associated with abnormalities in brain function and behaviour of the offspring4. However, it is unclear whether the maternal gut microbiome influences neurodevelopment during critical prenatal periods and in the absence of environmental challenges. Here we investigate how depletion and selective reconstitution of the maternal gut microbiome influences fetal neurodevelopment in mice. Embryos from antibiotic-treated and germ-free dams exhibited reduced brain expression of genes related to axonogenesis, deficient thalamocortical axons and impaired outgrowth of thalamic axons in response to cell-extrinsic factors. Gnotobiotic colonization of microbiome-depleted dams with a limited consortium of bacteria prevented abnormalities in fetal brain gene expression and thalamocortical axonogenesis. Metabolomic profiling revealed that the maternal microbiome regulates numerous small molecules in the maternal serum and the brains of fetal offspring. Select microbiota-dependent metabolites promoted axon outgrowth from fetal thalamic explants. Moreover, maternal supplementation with these metabolites abrogated deficiencies in fetal thalamocortical axons. Manipulation of the maternal microbiome and microbial metabolites during pregnancy yielded adult offspring with altered tactile sensitivity in two aversive somatosensory behavioural tasks, but no overt differences in many other sensorimotor behaviours. Together, our findings show that the maternal gut microbiome promotes fetal thalamocortical axonogenesis, probably through signalling by microbially modulated metabolites to neurons in the developing brain.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Disbiosis/microbiología , Feto/embriología , Feto/metabolismo , Microbioma Gastrointestinal/fisiología , Madres , Animales , Axones/metabolismo , Encéfalo/citología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Simulación por Computador , Disbiosis/sangre , Disbiosis/patología , Femenino , Feto/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/microbiología , Complicaciones del Embarazo/patología , Análisis de Componente Principal , Tálamo/citología , Tálamo/embriología , Tálamo/metabolismo
4.
Dev Neurosci ; 42(5-6): 208-216, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33684917

RESUMEN

Thalamus is an important sensory relay station: afferent sensory information, except olfactory signals, is transmitted by thalamocortical axons (TCAs) to the cerebral cortex. The pathway choice of TCAs depends on diverse diffusible or substrate-bound guidance cues in the environment. Not only classical guidance cues (ephrins, slits, semaphorins, and netrins), morphogens, which exerts patterning effects during early embryonic development, can also help axons navigate to their targets at later development stages. Here, expression analyses reveal that morphogen Fibroblast growth factor (FGF)-3 is expressed in the chick ventral diencephalon, hypothalamus, during the pathfinding of TCAs. Then, using in vitro analyses in chick explants, we identify a concentration-dependent effect of FGF3 on thalamic axons: attractant 100 ng/mL FGF3 transforms to a repellent at high concentration 500 ng/mL. Moreover, inhibition of FGF3 guidance functions indicates that FGF3 signaling is necessary for the correct navigation of thalamic axons. Together, these studies demonstrate a direct effect for the member of FGF7 subfamily, FGF3, in the axonal pathfinding of TCAs.


Asunto(s)
Orientación del Axón/fisiología , Factor 3 de Crecimiento de Fibroblastos/metabolismo , Hipotálamo/metabolismo , Vías Nerviosas/embriología , Animales , Corteza Cerebral/embriología , Embrión de Pollo , Tálamo/embriología
5.
Neuron ; 103(6): 980-1004, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31557462

RESUMEN

Adult cortical areas consist of specialized cell types and circuits that support unique higher-order cognitive functions. How this regional diversity develops from an initially uniform neuroepithelium has been the subject of decades of seminal research, and emerging technologies, including single-cell transcriptomics, provide a new perspective on area-specific molecular diversity. Here, we review the early developmental processes that underlie cortical arealization, including both cortex intrinsic and extrinsic mechanisms as embodied by the protomap and protocortex hypotheses, respectively. We propose an integrated model of serial homology whereby intrinsic genetic programs and local factors establish early transcriptomic differences between excitatory neurons destined to give rise to broad "proto-regions," and activity-dependent mechanisms lead to progressive refinement and formation of sharp boundaries between functional areas. Finally, we explore the potential of these basic developmental processes to inform our understanding of the emergence of functional neural networks and circuit abnormalities in neurodevelopmental disorders.


Asunto(s)
Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/fisiología , Neuronas/citología , Animales , Aprendizaje Profundo , Humanos , Interneuronas/citología , Interneuronas/metabolismo , Inhibición Neural , Neurogénesis/genética , Neuronas/metabolismo , Análisis de la Célula Individual , Tálamo/embriología
6.
Nutrients ; 11(5)2019 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-31121888

RESUMEN

The pharmacological properties of Eleutherococcus senticosus leaf have not been clarified although it is taken as a food item. In this study, the effects of water extract of Eleutherococcus senticosus leaves on memory function were investigated in normal mice. Oral administration of the extract for 17 days significantly enhanced object recognition memory. Compounds absorbed in blood and the brain after oral administration of the leaf extract were detected by LC-MS/MS analyses. Primarily detected compounds in plasma and the cerebral cortex were ciwujianoside C3, eleutheroside M, ciwujianoside B, and ciwujianoside A1. Pure compounds except for ciwujianoside A1 were administered orally for 17 days to normal mice. Ciwujianoside C3, eleutheroside M, and ciwujianoside B significantly enhanced object recognition memory. These results demonstrated that oral administration of the leaf extract of E. senticosus enhances memory function, and that active ingredients in the extract, such as ciwujianoside C3, eleutheroside M, and ciwujianoside B, were able to penetrate and work in the brain. Those three compounds as well as the leaf extract had dendrite extension activity against primary cultured cortical neurons. The effect might relate to memory enhancement.


Asunto(s)
Encéfalo/efectos de los fármacos , Eleutherococcus/química , Memoria/efectos de los fármacos , Extractos Vegetales/farmacología , Hojas de la Planta/química , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Corteza Cerebral/química , Corteza Cerebral/embriología , Corteza Cerebral/ultraestructura , Dendritas/efectos de los fármacos , Dendritas/fisiología , Glicósidos/análisis , Glicósidos/farmacocinética , Glicósidos/farmacología , Masculino , Ratones , Extractos Vegetales/análisis , Extractos Vegetales/farmacocinética , Saponinas/análisis , Saponinas/farmacocinética , Saponinas/farmacología
7.
Brain Dev ; 41(7): 567-576, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30954358

RESUMEN

BACKGROUNDS: Metabotropic glutamate receptors, besides ionotropic receptors, mediate the complicated effect of glutamate on neurogenesis. Previous studies showed that metabotropic glutamate receptor 4 (mGluR4) regulated the proliferation and differentiation of neural stem/progenitor cells in vitro. However, little is known about the expression pattern of mGluR4 on prenatal central nervous system in vivo, especially the human being. METHODS: The normal brain tissues of human fetus were collected and divided into 4 groups according to the gestational age: 9-11 W, 14-16 W, 22-24 W and 32-36 W. Then the expression of mGluR4 was evaluated at mRNA and protein levels by means of PCR or immunohistochemistry method, respectively. The type of cell expressing mGluR4 was further investigated using double-labeling immunofluorescence. RESULTS: RT-PCR showed that the mRNA of mGluR4 could be detected in frontal lobe from 9 W to 32 W and real-time PCR quantificationally demonstrated the mRNA increased with development. Similarly, immnoreactivity was found in all layers of frontal lobe, VZ/SVZ. The intensity scores analysis showed that the staining became stronger and the range extended gradually with development. The double-labeling immunofluorescence showed that mGluR4 was present in neural stem/progenitor cells (nestin-positive cells after 9 W), young neurons (DCX-positive cells after 9 W), mature neurons (NeuN-positive cells in cortex after 32 W), as well as typical astrocytes (GFAP-positive cells in medulla after 32 W). CONCLUSION: These results supply an important evidence that mGluR4 is expressed in prenatal human cerebrum, and main kinds of cells related to neurogenesis are involved in its expression.


Asunto(s)
Encéfalo/embriología , Lóbulo Frontal/embriología , Receptores de Glutamato Metabotrópico/metabolismo , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Femenino , Desarrollo Fetal/genética , Lóbulo Frontal/citología , Lóbulo Frontal/metabolismo , Ácido Glutámico/metabolismo , Humanos , Inmunohistoquímica , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Embarazo , Receptores de Glutamato Metabotrópico/genética
8.
Cereb Cortex ; 29(4): 1706-1718, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30668846

RESUMEN

The current model, based on rodent data, proposes that thalamocortical afferents (TCA) innervate the subplate towards the end of cortical neurogenesis. This implies that the laminar identity of cortical neurons is specified by intrinsic instructions rather than information of thalamic origin. In order to determine whether this mechanism is conserved in the primates, we examined the growth of thalamocortical (TCA) and corticofugal afferents in early human and monkey fetal development. In the human, TCA, identified by secretagogin, calbindin, and ROBO1 immunoreactivity, were observed in the internal capsule of the ventral telencephalon as early as 7-7.5 PCW, crossing the pallial/subpallial boundary (PSB) by 8 PCW before the calretinin immunoreactive corticofugal fibers do. Furthermore, TCA were observed to be passing through the intermediate zone and innervating the presubplate of the dorsolateral cortex, and already by 10-12 PCW TCAs were occupying much of the cortex. Observations at equivalent stages in the marmoset confirmed that this pattern is conserved across primates. Therefore, our results demonstrate that in primates, TCAs innervate the cortical presubplate at earlier stages than previously demonstrated by acetylcholinesterase histochemistry, suggesting that pioneer thalamic afferents may contribute to early cortical circuitry that can participate in defining cortical neuron phenotypes.


Asunto(s)
Corteza Cerebral/embriología , Neuronas Aferentes/citología , Tálamo/embriología , Vías Aferentes/citología , Vías Aferentes/embriología , Vías Aferentes/metabolismo , Animales , Callithrix , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Humanos , Neuronas Aferentes/metabolismo , Roedores , Tálamo/citología , Tálamo/metabolismo
9.
Brain Struct Funct ; 223(5): 2361-2375, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29492654

RESUMEN

Sonic Hedgehog (Shh) plays an instrumental role in brain development, fine-tuning processes such as cell proliferation, patterning, and fate specification. Although, mutations in the SHH pathway in humans are associated with various neurodevelopmental disorders, ranging from holoprosencephaly to schizophrenia, its expression pattern in the developing human brain is not well established. We now determined the previously not reported wide expression of SHH in the human fetal cerebral cortex during most of the gestation period (10-40 gestational weeks). This spatiotemporal distribution puts Shh in a position to influence the fundamental processes involved in corticogenesis. SHH expression increased during development, shifting from progenitor cells in the proliferative zones to neurons, both glutamatergic and GABAergic, and astrocytes in upper cortical compartments. Importantly, the expression of its downstream effectors and complementary receptors revealed evolutionary differences in SHH-pathway gene expression between humans and rodents.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Proteínas Hedgehog/metabolismo , Factores de Edad , Encéfalo/embriología , Encéfalo/metabolismo , Corteza Cerebral/citología , Femenino , Feto , Edad Gestacional , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato Descarboxilasa/metabolismo , Proteínas Hedgehog/genética , Humanos , Antígeno Ki-67/metabolismo , Masculino , Neuroglía/metabolismo , Neuronas/metabolismo , Factor de Transcripción PAX6/metabolismo , ARN Mensajero/metabolismo , Proteínas de Dominio T Box/metabolismo , Ácido gamma-Aminobutírico/metabolismo
10.
Mol Cell Endocrinol ; 471: 42-50, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28554804

RESUMEN

Fetal exposure to synthetic glucocorticoids reprograms distinct neural circuits in the developing brain, often in a sex-specific manner, via mechanisms that remain poorly understood. To reveal whether such reprogramming is associated with select molecular signatures, we characterized the transcriptome of primary, embryonic mouse cerebral cortical and hypothalamic neural progenitor/stem cells derived from individual male and female embryos exposed to the synthetic glucocorticoid, dexamethasone. Gene expression profiling by RNA-Seq identified differential expression of common and unique genes based upon brain region, sex, and/or dexamethasone exposure. These gene expression datasets provide a unique resource that will inform future studies examining the molecular mechanisms responsible for region- and sex-specific reprogramming of the fetal brain brought about by in utero exposure to excess glucocorticoids.


Asunto(s)
Corteza Cerebral/embriología , Dexametasona/farmacología , Embrión de Mamíferos/citología , Hipotálamo/embriología , Células-Madre Neurales/metabolismo , Caracteres Sexuales , Transcriptoma/genética , Animales , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Transcriptoma/efectos de los fármacos
11.
Neuromolecular Med ; 19(2-3): 271-285, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28523591

RESUMEN

We have demonstrated that arginine-rich and poly-arginine peptides possess potent neuroprotective properties with arginine content and peptide positive charge being particularly critical for neuroprotective efficacy. In addition, the presence of other amino acids within arginine-rich peptides, as well as chemical modifications, peptide length and cell-penetrating properties also influence the level of neuroprotection. Against this background, we have examined the neuroprotective efficacy of arginine-rich protamine peptides, a cyclic (R12-c) poly-arginine peptide and a R22 poly-arginine peptide, as well as arginine peptides containing tryptophan or other amino acids (phenylalanine, tyrosine, glycine or leucine) in in vitro glutamic acid excitotoxicity and in vivo rat permanent middle cerebral artery occlusion models of stroke. In vitro studies demonstrated that protamine and poly-arginine peptides (R12-c, R22) were neuroprotective. Arginine-tryptophan-containing peptides were highly neuroprotective, with R12W8a being the most potent arginine-rich peptide identified in our laboratory. Peptides containing phenylalanine or tyrosine substituted in place of tryptophan in R12W8a were also highly neuroprotective, whereas leucine, and in particular glycine substitutions, decreased peptide efficacy. In vivo studies with protamine administered intravenously at 1000 nmol/kg 30 min after MCAO significantly reduced infarct volume and cerebral oedema by 22.5 and 38.6%, respectively. The R12W8a peptide was highly toxic when administered intravenously at 300 or 100 nmol/kg and ineffective at reducing infarct volume when administered at 30 nmol/kg 30 min after MCAO, unlike R18 (30 nmol/kg), which significantly reduced infarct volume by 20.4%. However, both R12W8a and R18 significantly reduced cerebral oedema by 19.8 and 42.2%, respectively. Protamine, R12W8a and R18 also reduced neuronal glutamic acid-induced calcium influx. These findings further highlight the neuroprotective properties of arginine-rich peptides and support the view that they represent a new class of neuroprotective agent.


Asunto(s)
Ácido Glutámico/toxicidad , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Aminoácidos/farmacología , Animales , Arginina/química , Astrocitos/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Evaluación Preclínica de Medicamentos , Técnicas In Vitro , Infarto de la Arteria Cerebral Media/patología , Masculino , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacología , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Protaminas/química , Ratas , Ratas Sprague-Dawley , Triptófano/química
12.
Mol Neurobiol ; 54(6): 4414-4420, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27349434

RESUMEN

The diencephalon is the primary relay network transmitting sensory information to the anterior forebrain. During development, distinct progenitor domains in the diencephalon give rise to the pretectum (p1), the thalamus and epithalamus (p2), and the prethalamus (p3), respectively. Shh plays a significant role in establishing the progenitor domains. However, the upstream events influencing the expression of Shh are largely unknown. Here, we show that Barhl2 homeobox gene is expressed in the p1 and p2 progenitor domains and the in zona limitans intrathalamica (ZLI) and regulates the acquisition of identity of progenitor cells in the developing diencephalon. Targeted deletion of Barhl2 results in the ablation of Shh expression in the dorsal portion of ZLI and causes thalamic p2 progenitors to take the fate of p1 progenitors and form pretectal neurons. Moreover, loss of Barhl2 leads to the absence of thalamocortical axon projections, the loss of habenular afferents and efferents, and a gross diminution of the pineal gland. Thus, by acting upstream of Shh signaling pathway, Barhl2 plays a crucial role in patterning the progenitor domains and establishing the positional identities of progenitor cells in the diencephalon.


Asunto(s)
Tipificación del Cuerpo , Diencéfalo/embriología , Diencéfalo/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Axones/metabolismo , Biomarcadores/metabolismo , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Tálamo/embriología , Tálamo/metabolismo
13.
Neuromolecular Med ; 19(1): 136-146, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27567921

RESUMEN

Reelin is an extracellular glycoprotein which contributes to synaptic plasticity and function of memory in the adult brain. It has been indicated that the Reelin signaling cascade participates in Alzheimer's disease (AD). Besides the neurons, glial cells such as astrocytes also express Reelin protein. While functional loss of astrocytes has been reported to be associated with AD, dysfunction of astrocytic Reelin signaling pathway has not received much attention. Therefore, we investigated the effects of α-boswellic acid (ABA) as one of the major component of Boswellia serrata resin on primary fetal human astrocytes under a stress paradigm as a possible model for AD through study on Reelin cascade. For this aim, we used streptozotocin (STZ), in which from an outlook generates Alzheimer's hallmarks in astrocytes, and assayed Reelin expression, Tau and Akt phosphorylation as well as reactive oxygen species (ROS) generation and apoptosis in the presences of ABA. Our results indicated that while STZ (100 µM) down-regulated the expression of Reelin, ABA (25 µM) up-regulated its expression (p < 0.01) for 24 h. ABA efficiently reduced hyperphosphorylated Tau (Ser404) in STZ-treated astrocytes (p < 0.01). Furthermore, STZ-induced apoptosis by increasing cleaved caspase three (p < 0.01) and ROS generation (p < 0.01), a further pathological hallmark of Tauopathy. On the other hand, ABA decreased ROS generation and promoted proliferation of astrocytes through elevating Survivin expression (p < 0.01). These results showed that ABA could be considered as a potent therapeutic agent for prevention and decreasing the progression of Alzheimer's hallmarks in astrocytes; however, more in vivo studies would be needed.


Asunto(s)
Astrocitos/efectos de los fármacos , Moléculas de Adhesión Celular Neuronal/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Triterpenos Pentacíclicos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Serina Endopeptidasas/biosíntesis , Proteínas tau/metabolismo , Apoptosis/efectos de los fármacos , Astrocitos/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipotálamo/citología , Hipotálamo/embriología , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Estrés Oxidativo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética , Estreptozocina/farmacología , Survivin
14.
Mol Cell Neurosci ; 77: 87-94, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27989735

RESUMEN

Deficits in neuronal migration during development in the central nervous system may contribute to psychiatric diseases. The ligand neuregulin1 (NRG1) and its receptor ErbB4 are genes conferring susceptibility to schizophrenia, playing a key role in the control of neuronal migration both during development and adulthood. Several NRG1 and ErbB4 isoforms were identified, which deeply differ in their characteristics. Here we focused on the four ErbB4 isoforms and the two NRG1 isoforms differing in their EGF-like domain, namely α and ß. We hypothesized that these isoforms, which are differently regulated in schizophrenic patients, could play different roles in neuronal migration. Our hypothesis was strengthened by the observation that both NRG1α and NRG1ß and the four ErbB4 isoforms are expressed in the medial and lateral ganglionic eminences and in the cortex during development in rat. We analysed in vitro the signal transduction pathways activated by the different ErbB4 isoforms following the treatment with soluble recombinant NRG1α or NRG1ß and the ability to stimulate migration. Our data show that two ErbB4 isoforms, namely JMa-cyt2 and JMb-cyt1, following NRG1α and NRG1ß treatment, strongly activate AKT phosphorylation, conferring high migratory activity to neuronal progenitors, thus demonstrating that both NRG1α and NRG1ß can play a role in neuronal migration.


Asunto(s)
Movimiento Celular , Células-Madre Neurales/metabolismo , Neurregulina-1/metabolismo , Receptor ErbB-4/metabolismo , Animales , Línea Celular , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Hipotálamo/citología , Hipotálamo/embriología , Hipotálamo/metabolismo , Células-Madre Neurales/fisiología , Neurregulina-1/genética , Neurogénesis , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptor ErbB-4/genética
15.
Elife ; 52016 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-27935475

RESUMEN

Neocortical GABAergic interneuron migration and thalamo-cortical axon (TCA) pathfinding follow similar trajectories and timing, suggesting they may be interdependent. The mechanisms that regulate the radial dispersion of neocortical interneurons are incompletely understood. Here we report that disruption of TCA innervation, or TCA-derived glutamate, affected the laminar distribution of GABAergic interneurons in mouse neocortex, resulting in abnormal accumulation in deep layers of interneurons that failed to switch from tangential to radial orientation. Expression of the KCC2 cotransporter was elevated in interneurons of denervated cortex, and KCC2 deletion restored normal interneuron lamination in the absence of TCAs. Disruption of interneuron NMDA receptors or pharmacological inhibition of calpain also led to increased KCC2 expression and defective radial dispersion of interneurons. Thus, although TCAs are not required to guide the tangential migration of GABAergic interneurons, they provide crucial signals that restrict interneuron KCC2 levels, allowing coordinated neocortical invasion of TCAs and interneurons.


Asunto(s)
Axones/fisiología , Movimiento Celular , Corteza Cerebral/embriología , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Vías Nerviosas/embriología , Tálamo/embriología , Animales , Ratones
16.
Cereb Cortex ; 26(7): 3323-34, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27170656

RESUMEN

Celsr3 and Fzd3 regulate the development of reciprocal thalamocortical projections independently of their expression in cortical or thalamic neurons. To understand this cell non autonomous mechanism further, we tested whether Celsr3 and Fzd3 could act via Isl1-positive guidepost cells. Isl1-positive cells appear in the forebrain at embryonic day (E) 9.5-E10.5 and, from E12.5, they form 2 contingents in ventral telencephalon and prethalamus. In control mice, corticothalamic axons run in the ventral telencephalic corridor in close contact with Isl1-positive cells. When Celsr3 or Fzd3 is inactivated in Isl1-expressing cells, corticofugal fibers stall and loop in the ventral telencephalic corridor of high Isl1 expression, and thalamic axons fail to cross the diencephalon-telencephalon junction (DTJ). At E12.5, before thalamic and cortical axons emerge, pioneer projections from Isl1-positive cells cross the DTJ from both sides in control but not mutant embryos. These early projections appear to act like a bridge to guide later growing thalamic axons through the DTJ. Our data suggest that Celsr3 and Fzd3 orchestrate the formation of a scaffold of pioneer neurons and their axons. This scaffold extends from prethalamus to ventral telencephalon and subcortex, and steers reciprocal corticothalamic fibers.


Asunto(s)
Axones/metabolismo , Cadherinas/metabolismo , Corteza Cerebral/embriología , Receptores Frizzled/metabolismo , Receptores de Superficie Celular/metabolismo , Tálamo/embriología , Animales , Cadherinas/genética , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Receptores Frizzled/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones Transgénicos , Proyección Neuronal/fisiología , ARN Mensajero/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Receptores de Superficie Celular/genética , Tálamo/citología , Tálamo/metabolismo , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
J Vis Exp ; (107): e53303, 2016 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-26862715

RESUMEN

In utero electroporation is a widely used technique for fast and efficient spatiotemporal manipulation of various genes in the rodent central nervous system. Overexpression of desired genes is just as possible as shRNA mediated loss-of-function studies. Therefore it offers a wide range of applications. The feasibility to target particular cells in a distinct area further increases the range of potential applications of this very useful method. For efficiently targeting specific regions knowledge about the subtleties, such as the embryonic stage, the voltage to apply and most importantly the position of the electrodes, is indispensable. Here, we provide a detailed protocol that allows for specific and efficient in utero electroporation of several regions of the C57BL/6 mouse central nervous system. In particular it is shown how to transfect regions the develop into the retrosplenial cortex, the motor cortex, the somatosensory cortex, the piriform cortex, the cornu ammonis 1-3, the dentate gyrus, the striatum, the lateral septal nucleus, the thalamus and the hypothalamus. For this information about the appropriate embryonic stage, the appropriate voltage for the corresponding embryonic stage is provided. Most importantly an angle-map, which indicates the appropriate position of the positive pole, is depicted. This standardized protocol helps to facilitate efficient in utero electroporation, which might also lead to a reduced number of animals.


Asunto(s)
Corteza Cerebral/embriología , Cuerpo Estriado/embriología , Electroporación/métodos , Hipocampo/embriología , Hipotálamo/embriología , Preñez , Núcleos Septales/embriología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo , Tálamo/embriología
18.
J Comp Neurol ; 524(10): 2080-92, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-26587807

RESUMEN

Deciphering the molecular basis for guiding specific aspects of neocortical development remains a challenge because of the complexity of histogenic events and the vast array of protein interactions mediating these events. The Eph family of receptor tyrosine kinases is implicated in a number of neurodevelopmental activities. Eph receptors have been known to be capable of responding to several ephrin ligands within their subgroups, often eliciting similar downstream effects. However, several recent studies have indicated specificity between receptor-ligand pairs within each subfamily, the functional relevance of which is not defined. Here we show that a receptor of the EphA subfamily, EphA4, has effects distinct from those of its close relative, EphA7, in the developing brain. Both EphA4 and EphA7 interact similarly with corresponding ligands expressed in the developing neocortex. However, only EphA7 shows strong interaction with ligands in the somatosensory thalamic nuclei; EphA4 affects only cortical neuronal migration, with no visible effects on the guidance of corticothalamic (CT) axons, whereas EphA7 affects both cortical neuronal migration and CT axon guidance. Our data provide new evidence that Eph receptors in the same subfamily are not simply interchangeable but are functionally specified through selective interactions with distinct ligands in vivo. J. Comp. Neurol. 524:2080-2092, 2016. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Corteza Cerebral , Vías Nerviosas/fisiología , Receptor EphA4/metabolismo , Receptor EphA7/metabolismo , Tálamo , Animales , Animales Recién Nacidos , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Receptor EphA4/genética , Receptor EphA7/genética , Tálamo/citología , Tálamo/embriología , Tálamo/crecimiento & desarrollo , Tálamo/metabolismo
19.
Cereb Cortex ; 26(7): 3219-36, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26209842

RESUMEN

Disabled-1 (Dab1) is an essential intracellular protein in the Reelin pathway. It has a nuclear localization signal (NLS; hereafter referred to as "NLS1") and 2 nuclear export signals, and shuttles between the nucleus and the cytoplasm. In this study, we found that Dab1 has an additional unidentified NLS, and that the Dab1 NLS1 mutant could translocate to the nucleus in an unconventional ATP/temperature-dependent and cytoplasmic factor/RanGTP gradient-independent manner. Additional mutations in the NLS1 mutant revealed that K(67) and K(69) are important for the nuclear transport. Furthermore, an excess of the intracellular domain of the Reelin receptors inhibited the nuclear translocation of Dab1. An in utero electroporation study showed that a large amount of Dab1 in the cytoplasm in migrating neurons inhibited the migration, and that forced transport of Dab1 into the nucleus attenuated this inhibitory effect. In addition, rescue experiments using yotari, an autosomal recessive mutant of dab1, revealed that cells expressing Dab1 NLS1 mutant tend to distribute at more superficial positions than those expressing wild-type Dab1. Taken together, these findings suggest that Dab1 has at least 2 NLSs, and that the regulation of the subcellular localization of Dab1 is important for the proper migration of excitatory neurons.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Movimiento Celular/fisiología , Corteza Cerebral/metabolismo , Citoplasma/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Corteza Cerebral/embriología , Corteza Cerebral/patología , Citoplasma/patología , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ratones Endogámicos ICR , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Receptores de Superficie Celular/metabolismo , Receptores de LDL/metabolismo , Receptores Notch/metabolismo , Proteína Reelina , Temperatura , Proteína de Unión al GTP ran/metabolismo
20.
J Mol Neurosci ; 58(2): 277-86, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26547318

RESUMEN

Maternal nutrients play critical roles in modulating epigenetic events and exert long-term influences on the progeny's health. Folic acid (FA) supplementation during pregnancy has decreased the incidence of neural tube defects in newborns, but the influence of high doses of maternal FA supplementation on infants' brain development is unclear. The present study was aimed at investigating the effects of a high dose of gestational FA on the expression of genes in the cerebral hemispheres (CHs) of 1-day-old pups. One week prior to mating and throughout the entire period of gestation, female C57BL/6J mice were fed a diet, containing FA at either 2 mg/kg (control diet (CD)) or 20 mg/kg (high maternal folic acid (HMFA)). At postnatal day 1, pups from different dams were sacrificed and CH tissues were collected. Quantitative RT-PCR and Western blot analysis confirmed sex-specific alterations in the expression of several genes that modulate various cellular functions (P < 0.05) in pups from the HMFA group. Genomic DNA methylation analysis showed no difference in the level of overall methylation in pups from the HMFA group. These findings demonstrate that HMFA supplementation alters offsprings' CH gene expression in a sex-specific manner. These changes may influence infants' brain development.


Asunto(s)
Trastorno Autístico/genética , Corteza Cerebral/efectos de los fármacos , Ácido Fólico/farmacología , Impresión Genómica , Efectos Tardíos de la Exposición Prenatal/genética , Fenómenos Fisiologicos de la Nutrición Prenatal/genética , Complejo Vitamínico B/farmacología , Animales , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Metilación de ADN , Suplementos Dietéticos , Femenino , Ácido Fólico/administración & dosificación , Predisposición Genética a la Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Factores Sexuales , Complejo Vitamínico B/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA