Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 347
Filtrar
1.
Nat Commun ; 12(1): 5916, 2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34625548

RESUMEN

Microglia are brain resident macrophages that play vital roles in central nervous system (CNS) development, homeostasis, and pathology. Microglia both remodel synapses and engulf apoptotic cell corpses during development, but whether unique molecular programs regulate these distinct phagocytic functions is unknown. Here we identify a molecularly distinct microglial subset in the synapse rich regions of the zebrafish (Danio rerio) brain. We found that ramified microglia increased in synaptic regions of the midbrain and hindbrain between 7 and 28 days post fertilization. In contrast, microglia in the optic tectum were ameboid and clustered around neurogenic zones. Using single-cell mRNA sequencing combined with metadata from regional bulk sequencing, we identified synaptic-region associated microglia (SAMs) that were highly enriched in the hindbrain and expressed multiple candidate synapse modulating genes, including genes in the complement pathway. In contrast, neurogenic associated microglia (NAMs) were enriched in the optic tectum, had active cathepsin activity, and preferentially engulfed neuronal corpses. These data reveal that molecularly distinct phagocytic programs mediate synaptic remodeling and cell engulfment, and establish the zebrafish hindbrain as a model for investigating microglial-synapse interactions.


Asunto(s)
Mesencéfalo/citología , Microglía/citología , Neurogénesis/genética , Rombencéfalo/citología , Colículos Superiores/citología , Transcriptoma , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/inmunología , Catepsina B/genética , Catepsina B/inmunología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/inmunología , Microglía/inmunología , Neurogénesis/inmunología , Neuronas/citología , Neuronas/inmunología , Fagocitosis , Rombencéfalo/crecimiento & desarrollo , Rombencéfalo/inmunología , Análisis de la Célula Individual , Colículos Superiores/crecimiento & desarrollo , Colículos Superiores/inmunología , Sinapsis/inmunología , Sinapsis/metabolismo , Sinapsis/ultraestructura , Pez Cebra , Proteínas de Pez Cebra/inmunología
2.
PLoS One ; 16(8): e0256207, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34403440

RESUMEN

Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Larva/genética , Vaina de Mielina/genética , Tiroxina/deficiencia , Triyodotironina/deficiencia , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Antígenos/genética , Antígenos/metabolismo , Embrión no Mamífero , Desarrollo Embrionario , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ácido Yopanoico/farmacología , Larva/citología , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Rombencéfalo/citología , Rombencéfalo/efectos de los fármacos , Rombencéfalo/crecimiento & desarrollo , Rombencéfalo/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Triyodotironina/farmacología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Elife ; 102021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522480

RESUMEN

The ubiquitous presence of inhibitory interneurons in the thalamus of primates contrasts with the sparsity of interneurons reported in mice. Here, we identify a larger than expected complexity and distribution of interneurons across the mouse thalamus, where all thalamic interneurons can be traced back to two developmental programmes: one specified in the midbrain and the other in the forebrain. Interneurons migrate to functionally distinct thalamocortical nuclei depending on their origin: the abundant, midbrain-derived class populates the first and higher order sensory thalamus while the rarer, forebrain-generated class is restricted to some higher order associative regions. We also observe that markers for the midbrain-born class are abundantly expressed throughout the thalamus of the New World monkey marmoset. These data therefore reveal that, despite the broad variability in interneuron density across mammalian species, the blueprint of the ontogenetic organisation of thalamic interneurons of larger-brained mammals exists and can be studied in mice.


Asunto(s)
Linaje de la Célula , Interneuronas , Tálamo/crecimiento & desarrollo , Animales , Callithrix , Movimiento Celular , Femenino , Neuronas GABAérgicas , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Masculino , Mesencéfalo/crecimiento & desarrollo , Ratones , Ratones Transgénicos , Prosencéfalo/crecimiento & desarrollo , Tálamo/citología
4.
STAR Protoc ; 1(2): 100065, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-33111103

RESUMEN

Here, we describe a xeno-free, feeder-free, and chemically defined protocol for the generation of ventral midbrain dopaminergic (vmDA) progenitors from human pluripotent stem cells (hPSCs). This simple-to-follow protocol results in high yields of cryopreservable dopamine neurons across multiple hPSC lines. Wnt signaling is the critical component of the differentiation and can be finely adjusted in a line-dependent manner to enhance production of dopamine neurons for the purposes of transplantation, studying development and homeostasis, disease modeling, drug discovery, and drug development. For complete details on the use and execution of this protocol, please refer to Gantner et al. (2020) and Niclis et al. (2017a).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/crecimiento & desarrollo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Medios de Cultivo/metabolismo , Dopamina , Neuronas Dopaminérgicas/citología , Humanos , Mesencéfalo/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
5.
J Neurosci ; 40(43): 8262-8275, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-32928885

RESUMEN

A subset of adult ventral tegmental area dopamine (DA) neurons expresses vesicular glutamate transporter 2 (VGluT2) and releases glutamate as a second neurotransmitter in the striatum, while only few adult substantia nigra DA neurons have this capacity. Recent work showed that cellular stress created by neurotoxins such as MPTP and 6-hydroxydopamine can upregulate VGluT2 in surviving DA neurons, suggesting the possibility of a role in cell survival, although a high level of overexpression could be toxic to DA neurons. Here we examined the level of VGluT2 upregulation in response to neurotoxins and its impact on postlesional plasticity. We first took advantage of an in vitro neurotoxin model of Parkinson's disease and found that this caused an average 2.5-fold enhancement of Vglut2 mRNA in DA neurons. This could represent a reactivation of a developmental phenotype because using an intersectional genetic lineage-mapping approach, we find that >98% of DA neurons have a VGluT2+ lineage. Expression of VGluT2 was detectable in most DA neurons at embryonic day 11.5 and was localized in developing axons. Finally, compatible with the possibility that enhanced VGluT2 expression in DA neurons promotes axonal outgrowth and reinnervation in the postlesional brain, we observed that DA neurons in female and male mice in which VGluT2 was conditionally removed established fewer striatal connections 7 weeks after a neurotoxin lesion. Thus, we propose here that the developmental expression of VGluT2 in DA neurons can be reactivated at postnatal stages, contributing to postlesional plasticity of dopaminergic axons.SIGNIFICANCE STATEMENT A small subset of dopamine neurons in the adult, healthy brain expresses vesicular glutamate transporter 2 (VGluT2) and thus releases glutamate as a second neurotransmitter in the striatum. This neurochemical phenotype appears to be plastic as exposure to neurotoxins, such as 6-OHDA or MPTP, that model certain aspects of Parkinson's disease pathophysiology, boosts VGluT2 expression in surviving dopamine neurons. Here we show that this enhanced VGluT2 expression in dopamine neurons drives axonal outgrowth and contributes to dopamine neuron axonal plasticity in the postlesional brain. A better understanding of the neurochemical changes that occur during the progression of Parkinson's disease pathology will aid the development of novel therapeutic strategies for this disease.


Asunto(s)
Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/biosíntesis , Animales , Animales Recién Nacidos , Axones/fisiología , Linaje de la Célula/genética , Supervivencia Celular/genética , Cuerpo Estriado/embriología , Cuerpo Estriado/crecimiento & desarrollo , Femenino , Intoxicación por MPTP/genética , Intoxicación por MPTP/metabolismo , Mesencéfalo/embriología , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/fisiología , Ratones , Ratones Noqueados , Vías Nerviosas/embriología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/fisiología , Neurotoxinas/toxicidad , Embarazo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética
6.
Cells ; 9(6)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32570916

RESUMEN

Parkinson's Disease (PD) is a neurodegenerative disorder affecting the motor system. It is primarily due to substantial loss of midbrain dopamine (mDA) neurons in the substantia nigra pars compacta and to decreased innervation to the striatum. Although existing drug therapy available can relieve the symptoms in early-stage PD patients, it cannot reverse the pathogenic progression of PD. Thus, regenerating functional mDA neurons in PD patients may be a cure to the disease. The proof-of-principle clinical trials showed that human fetal graft-derived mDA neurons could restore the release of dopamine neurotransmitters, could reinnervate the striatum, and could alleviate clinical symptoms in PD patients. The invention of human-induced pluripotent stem cells (hiPSCs), autologous source of neural progenitors with less ethical consideration, and risk of graft rejection can now be generated in vitro. This advancement also prompts extensive research to decipher important developmental signaling in differentiation, which is key to successful in vitro production of functional mDA neurons and the enabler of mass manufacturing of the cells required for clinical applications. In this review, we summarize the biology and signaling involved in the development of mDA neurons and the current progress and methodology in driving efficient mDA neuron differentiation from pluripotent stem cells.


Asunto(s)
Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/fisiología , Mesencéfalo/citología , Enfermedad de Parkinson/patología , Animales , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Levodopa/uso terapéutico , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/fisiología , Modelos Neurológicos , Regeneración Nerviosa/fisiología , Neurogénesis/fisiología , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/terapia , Investigación Biomédica Traslacional
7.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326436

RESUMEN

Calcium homeostasis is a cellular process required for proper cell function and survival, maintained by the coordinated action of several transporters, among them members of the Na+/Ca2+-exchanger family, such as SLC8A3. Transforming growth factor beta (TGF-ß) signaling defines neuronal development and survival and may regulate the expression of channels and transporters. We investigated the regulation of SLC8A3 by TGF-ß in a conditional knockout mouse with deletion of TGF-ß signaling from Engrailed 1-expressing cells, i.e., in cells from the midbrain and rhombomere 1, and elucidated the underlying molecular mechanisms. The results show that SLC8A3 is significantly downregulated in developing dopaminergic and dorsal raphe serotonergic neurons in mutants and that low SLC8A3 abundance prevents the expression of the anti-apoptotic protein Bcl-xL. TGF-ß signaling affects SLC8A3 via the canonical and p38 signaling pathway and may increase the binding of Smad4 to the Slc8a3 promoter. Expression of the lipid peroxidation marker malondialdehyde (MDA) was increased following knockdown of Slc8a3 expression in vitro. In neurons lacking TGF-ß signaling, the number of MDA- and 4-hydroxynonenal (4-HNE)-positive cells was significantly increased, accompanied with increased cellular 4-HNE abundance. These results suggest that TGF-ß contributes to the regulation of SLC8A3 expression in developing dopaminergic and dorsal raphe serotonergic neurons, thereby preventing oxidative stress.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/metabolismo , Neurogénesis/genética , Estrés Oxidativo/genética , Neuronas Serotoninérgicas/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Aldehídos/metabolismo , Animales , Apoptosis/genética , Calcio/metabolismo , Línea Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Neuronas Dopaminérgicas/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis , Humanos , Inmunohistoquímica , Malondialdehído/metabolismo , Mesencéfalo/efectos de los fármacos , Mesencéfalo/crecimiento & desarrollo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Neuronas Serotoninérgicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad4/metabolismo , Intercambiador de Sodio-Calcio/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Proteína bcl-X/metabolismo
8.
Neurotoxicology ; 75: 148-157, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31545971

RESUMEN

Manganese (Mn) is essential for neuronal health but neurotoxic in excess. Mn levels vary across brain regions and neurodevelopment. While Mn requirements during infanthood and childhood are significantly higher than in adulthood, the relative vulnerability to excess extracellular Mn across human neuronal developmental time and between distinct neural lineages is unknown. Neurological disease is associated with changes in brain Mn homeostasis and pathology associated with Mn neurotoxicity is not uniform across brain regions. For example, mutations associated with Huntington's disease (HD) decrease Mn bioavailability and increase resistance to Mn cytotoxicity in human and mouse striatal neuronal progenitors. Here, we sought to compare the differences in Mn cytotoxicity between control and HD human-induced pluripotent stem cells (hiPSCs)-derived neuroprogenitor cells (NPCs) and maturing neurons. We hypothesized that there would be differences in Mn sensitivity between lineages and developmental stages. However, we found that the different NPC lineage specific media substantially influenced Mn cytotoxicity in the hiPSC derived human NPCs and did so consistently even in a non-human cell line. This limited the ability to determine which human neuronal sub-types were more sensitive to Mn. Nonetheless, we compared within neuronal subtypes and developmental stage the sensitivity to Mn cytotoxicity between control and HD patient derived neuronal lineages. Consistent with studies in other striatal model systems the HD genotype was associated with resistance to Mn cytotoxicity in human striatal NPCs. In addition, we report an HD genotype-dependent resistance to Mn cytotoxicity in cortical NPCs and hiPSCs. Unexpectedly, the HD genotype conferred increased sensitivity to Mn in early post-mitotic midbrain neurons but had no effect on Mn sensitivity in midbrain NPCs or post-mitotic cortical neurons. Overall, our data suggest that sensitivity to Mn cytotoxicity is influenced by HD genotype in a human neuronal lineage type and stage of development dependent manner.


Asunto(s)
Encéfalo/efectos de los fármacos , Enfermedad de Huntington/metabolismo , Manganeso/toxicidad , Neuronas/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/crecimiento & desarrollo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Enfermedad de Huntington/complicaciones , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/crecimiento & desarrollo , Neuronas/metabolismo
9.
Hear Res ; 380: 166-174, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31306931

RESUMEN

Human subcortical auditory processing is sexually dimorphic. The prevailing view - that sex differences arise from cochlear differences - remains unproven, and the extent to which these differences reflect distinct auditory processes is unknown. To determine the origin of subcortical sex differences, we mapped their emergence onto the peripheral-to-central maturation of the auditory system in 516 participants (250 female) across three age groups: 3-5, 14-15, and 22-26 years. To examine whether these sex differences arise from distinct processes, we compared developmental trajectories of each evoked-response component and tested their ability to predict a participant's sex and age. We find that some subcortical sex differences emerge well after the cochlea is mature and that each measure uniquely contributes to predicting participant demographics, indicating that sex differences arise from multiple central auditory processes.


Asunto(s)
Vías Auditivas/crecimiento & desarrollo , Percepción Auditiva , Potenciales Evocados Auditivos del Tronco Encefálico , Audición , Mesencéfalo/crecimiento & desarrollo , Estimulación Acústica , Adolescente , Desarrollo del Adolescente , Adulto , Factores de Edad , Niño , Desarrollo Infantil , Preescolar , Femenino , Humanos , Masculino , Tiempo de Reacción , Caracteres Sexuales , Factores Sexuales , Adulto Joven
10.
J Neurosci ; 39(34): 6656-6667, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31300520

RESUMEN

The growth of axons corresponding to different neuronal subtypes is governed by unique expression profiles of molecules on the growth cone. These molecules respond to extracellular cues either locally though cell adhesion interactions or over long distances through diffusible gradients. Here, we report that that the cell adhesion molecule ALCAM (CD166) can act as an extracellular substrate to selectively promote the growth of murine midbrain dopamine (mDA) neuron axons through a trans-heterophilic interaction with mDA-bound adhesion molecules. In mixed-sex primary midbrain cultures, the growth-promoting effect of ALCAM was abolished by neutralizing antibodies for components of the Semaphorin receptor complex Nrp1, Chl1, or L1cam. The ALCAM substrate was also found to modulate the response of mDA neurites to soluble semaphorins in a context-specific manner by abolishing the growth-promoting effect of Sema3A but inducing a branching response in the presence of Sema3C. These findings identify a previously unrecognized guidance mechanism whereby cell adhesion molecules act in trans to modulate the response of axonal growth cones to soluble gradients to selectively orchestrate the growth and guidance of mDA neurons.SIGNIFICANCE STATEMENT The mechanisms governing the axonal connectivity of midbrain dopamine (mDA) neurons during neural development have remained rather poorly understood relative to other model systems for axonal growth and guidance. Here, we report a series of novel interactions between proteins previously not identified in the context of mDA neuronal growth. Significantly, the results suggest a previously unrecognized mechanism involving the convergence in signaling between local, adhesion and long-distance, soluble cues. A better understanding of the molecules and mechanisms involved in establishment of the mDA system is important as a part of ongoing efforts to understand the consequence of conditions that may result from aberrant connectivity and also for cell replacement strategies for Parkinson's disease.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/fisiología , Axones/fisiología , Moléculas de Adhesión Celular/fisiología , Neuronas Dopaminérgicas/fisiología , Mesencéfalo/citología , Mesencéfalo/crecimiento & desarrollo , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Semaforinas/fisiología , Animales , Anticuerpos Bloqueadores/farmacología , Femenino , Conos de Crecimiento , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/fisiología
11.
Development ; 146(14)2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332038

RESUMEN

Developmental control of long-range neuronal connections in the mammalian midbrain remains unclear. We explored the mechanisms regulating target selection of the developing superior colliculus (SC). The SC is a midbrain center that directs orienting behaviors and defense responses. We discovered that a transcription factor, Rorß, controls establishment of axonal projections from the SC to two thalamic nuclei: the dorsal lateral geniculate nucleus (dLGN) and the lateral posterior nucleus (LP). A genetic strategy used to visualize SC circuits revealed that in control animals Rorß+ neurons abundantly innervate the dLGN but barely innervate the LP. The opposite phenotype was observed in global and conditional Rorb mutants: projections to the dLGN were strongly decreased, and projections to the LP were increased. Furthermore, overexpression of Rorb in the wild type showed increased projections to the dLGN and decreased projections to the LP. In summary, we identified Rorß as a key developmental mediator of colliculo-thalamic innervation. Such regulation could represent a general mechanism orchestrating long-range neuronal connections in the mammalian brain.


Asunto(s)
Axones/fisiología , Mesencéfalo/embriología , Mesencéfalo/crecimiento & desarrollo , Neuronas/metabolismo , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Vías Visuales/metabolismo , Animales , Animales Recién Nacidos , Embrión de Mamíferos , Femenino , Cuerpos Geniculados/fisiología , Células HEK293 , Humanos , Masculino , Mesencéfalo/citología , Ratones , Ratones Transgénicos , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Embarazo , Colículos Superiores/fisiología
12.
Cerebellum ; 17(5): 685-691, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29663194

RESUMEN

An inherent asymmetry exists between the two centrosomes of a dividing cell. One centrosome is structurally more mature (mother centrosome) than the other (daughter centrosome). Post division, one daughter cell inherits the mother centrosome while the other daughter cell inherits the daughter centrosome. Remarkably, the kind of centrosome inherited is associated with cell fate in several developmental contexts such as in radial glial progenitors in the developing mouse cortex, Drosophila neuroblast divisions and in Drosophila male germline stem cells. However, the role of centrosome inheritance in granule neuron progenitors in the developing cerebellum has not been investigated. Here, we show that mother and daughter centrosomes do exist in these progenitors, and the amount of pericentriolar material (PCM) each centrosome possesses is different. However, we failed to observe any correlation between the fate adopted by the daughter cell and the nature of centrosome it inherited.


Asunto(s)
Centrosoma/fisiología , Cerebelo/crecimiento & desarrollo , Células-Madre Neurales/fisiología , Neuronas/fisiología , Animales , Tronco Encefálico/citología , Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/metabolismo , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Proteínas de Choque Térmico/metabolismo , Inmunohistoquímica , Mesencéfalo/citología , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Ratones , Mitosis/fisiología
13.
Proc Natl Acad Sci U S A ; 114(46): 12273-12278, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29087938

RESUMEN

Fetal infection with Zika virus (ZIKV) can lead to congenital Zika virus syndrome (cZVS), which includes cortical malformations and microcephaly. The aspects of cortical development that are affected during virus infection are unknown. Using organotypic brain slice cultures generated from embryonic mice of various ages, sites of ZIKV replication including the neocortical proliferative zone and radial columns, as well as the developing midbrain, were identified. The infected radial units are surrounded by uninfected cells undergoing apoptosis, suggesting that programmed cell death may limit viral dissemination in the brain and may constrain virus-associated injury. Therefore, a critical aspect of ZIKV-induced neuropathology may be defined by death of uninfected cells. All ZIKV isolates assayed replicated efficiently in early and midgestation cultures, and two isolates examined replicated in late-gestation tissue. Alteration of neocortical cytoarchitecture, such as disruption of the highly elongated basal processes of the radial glial progenitor cells and impairment of postmitotic neuronal migration, were also observed. These data suggest that all lineages of ZIKV tested are neurotropic, and that ZIKV infection interferes with multiple aspects of neurodevelopment that contribute to the complexity of cZVS.


Asunto(s)
Mesencéfalo/virología , Neocórtex/virología , Tropismo Viral , Replicación Viral/fisiología , Virus Zika/fisiología , Animales , Apoptosis , Embrión de Mamíferos , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/patología , Ratones , Microtomía , Neocórtex/crecimiento & desarrollo , Neocórtex/patología , Células-Madre Neurales/patología , Células-Madre Neurales/virología , Neurogénesis/genética , Neuroglía/patología , Neuroglía/virología , Neuronas/patología , Neuronas/virología , Filogenia , Técnicas de Cultivo de Tejidos , Virus Zika/clasificación , Virus Zika/patogenicidad
14.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1900-1912, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28779972

RESUMEN

Thyroid hormones play a crucial role in midbrain dopaminergic (DA) neuron development. However, the underlying molecular mechanisms remain largely unknown. In this study, we revealed that thyroid hormone treatment evokes significant calcium entry through canonical transient receptor potential (TRPC) channels in ventral midbrain neural stem cells and this calcium signaling is essential for thyroid hormone-dependent DA neuronal differentiation. We also found that TRPC1 is the dominant TRPC channel expressed in ventral midbrain neural stem cells which responds to thyroid hormone. In addition, thyroid hormone increases TRPC1 expression through its receptor alpha 1 during DA neuron differentiation, and, importantly, produces calcium signals by activating TRPC1 channels. In vivo and in vitro gene silencing experiments indicate that TRPC1-mediated calcium signaling is required for thyroid hormone-dependent DA neuronal differentiation. Finally, we confirmed that the activation of OTX2, a determinant of DA neuron development and the expression of which is induced by thyroid hormone, is dependent on TRPC1-mediated calcium signaling. These data revealed the molecular mechanisms of how thyroid hormone regulates DA neuron development from ventral midbrain neural stem cells, particularly endowing a novel physiological relevance to TRPC1 channels.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Neurogénesis/genética , Factores de Transcripción Otx/genética , Canales Catiónicos TRPC/genética , Hormonas Tiroideas/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/genética , Diferenciación Celular/genética , Dopamina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factores de Transcripción Otx/metabolismo , Canales Catiónicos TRPC/metabolismo
15.
Cell Tissue Res ; 370(2): 211-225, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28799057

RESUMEN

The temporal dynamic expression of Sonic Hedgehog (SHH) and signaling during early midbrain dopaminergic (mDA) neuron development is one of the key players in establishing mDA progenitor diversity. However, whether SHH signaling is also required during later developmental stages and in mature mDA neurons is less understood. We study the expression of SHH receptors Ptch1 and Gas1 (growth arrest-specific 1) and of the transcription factors Gli1, Gli2 and Gli3 in mouse midbrain during embryonic development [embryonic day (E) 12.5 onwards)], in newborn and adult mice using in situ hybridization and immunohistochemistry. Moreover, we examine the expression and regulation of dopaminergic neuronal progenitor markers, midbrain dopaminergic neuronal markers and markers of the SHH signaling pathway in undifferentiated and butyric acid-treated (differentiated) MN9D cells in the presence or absence of exogenous SHH in vitro by RT-PCR, immunoblotting and immunocytochemistry. Gli1 was expressed in the lateral mesencephalic domains, whereas Gli2 and Gli3 were expressed dorsolaterally and complemented by ventrolateral expression of Ptch1. Co-localization with tyrosine hydroxylase could not be observed. GAS1 was exclusively expressed in the dorsal mesencephalon at E11.5 and co-localized with Ki67. In contrast, MN9D cells expressed all the genes investigated and treatment of the cells with butyric acid significantly upregulated their expression. The results suggest that SHH is only indirectly involved in the differentiation and survival of mDA neurons and that the MN9D cell line is a valuable model for investigating early development but not the differentiation and survival of mDA neurons.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Mesencéfalo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Línea Celular , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Proteínas Hedgehog/análisis , Inmunohistoquímica , Hibridación in Situ , Mesencéfalo/química , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
16.
Mech Dev ; 146: 10-30, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28549975

RESUMEN

Brd2 is a member of the bromodomain-extraterminal domain (BET) family of proteins and functions as an acetyl-histone-directed transcriptional co-regulator and recruitment scaffold in chromatin modification complexes affecting signal-dependent transcription. While Brd2 acts as a protooncogene in mammalian blood, developmental studies link it to regulation of neuronal apoptosis and epilepsy, and complete knockout of the gene is invariably embryonic lethal. In Drosophila, the Brd2 homolog acts as a maternal effect factor necessary for segment formation and identity and proper expression of homeotic loci, including Ultrabithorax and engrailed. To test the various roles attributed to Brd2 in a single developmental system representing a non-mammalian vertebrate, we conducted a phenotypic characterization of Brd2a deficient zebrafish embryos produced by morpholino knockdown and corroborated by Crispr-Cas9 disruption and small molecule inhibitor treatments. brd2aMO morphants exhibit reduced hindbrain with an ill-defined midbrain-hindbrain boundary (MHB) region; irregular notochord, neural tube, and somites; and abnormalities in ventral trunk and ventral nerve cord interneuron positioning. Using whole mount TUNEL and confocal microscopy, we uncover a significant decrease, then a dramatic increase, of p53-independent cell death at the start and end of segmentation, respectively. In contrast, using qualitative and quantitative analyses of BrdU incorporation, phosphohistone H3-tagging, and flow cytometry, we detect little effect of Brd2a knockdown on overall proliferation levels in embryos. RNA in situ hybridization shows reduced or absent expression of homeobox gene eng2a and paired box gene pax2a, in the hindbrain domain of the MHB region, and an overabundance of pax2a-positive kidney progenitors, in knockdowns. Together, these results suggest an evolutionarily conserved role for Brd2 in the proper formation and/or patterning of segmented tissues, including the vertebrate CNS, where it acts as a bi-modal regulator of apoptosis, and is necessary, directly or indirectly, for proper expression of genes that pattern the MHB and/or regulate differentiation in the anterior hindbrain.


Asunto(s)
Morfogénesis/genética , Tubo Neural/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/genética , Transcripción Genética , Proteínas de Pez Cebra/genética , Animales , Apoptosis/genética , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Mesencéfalo/crecimiento & desarrollo , Morfolinos/genética , Proteínas del Tejido Nervioso/genética , Rombencéfalo/crecimiento & desarrollo , Somitos/crecimiento & desarrollo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
17.
BMC Dev Biol ; 17(1): 5, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28407732

RESUMEN

BACKGROUND: Cranial neural crest cells (NCCs) are a unique embryonic cell type which give rise to a diverse array of derivatives extending from neurons and glia through to bone and cartilage. Depending on their point of origin along the antero-posterior axis cranial NCCs are rapidly sorted into distinct migratory streams that give rise to axial specific structures. These migratory streams mirror the underlying segmentation of the brain with NCCs exiting the diencephalon and midbrain following distinct paths compared to those exiting the hindbrain rhombomeres (r). The genetic landscape of cranial NCCs arising at different axial levels remains unknown. RESULTS: Here we have used RNA sequencing to uncover the transcriptional profiles of mouse cranial NCCs arising at different axial levels. Whole transcriptome analysis identified over 120 transcripts differentially expressed between NCCs arising anterior to r3 (referred to as r1-r2 migratory stream for simplicity) and the r4 migratory stream. Eight of the genes differentially expressed between these populations were validated by RT-PCR with 2 being further validated by in situ hybridisation. We also explored the expression of the Neuropilins (Nrp1 and Nrp2) and their co-receptors and show that the A-type Plexins are differentially expressed in different cranial NCC streams. CONCLUSIONS: Our analyses identify a large number of genes differentially regulated between cranial NCCs arising at different axial levels. This data provides a comprehensive description of the genetic landscape driving diversity of distinct cranial NCC streams and provides novel insight into the regulatory networks controlling the formation of specific skeletal elements and the mechanisms promoting migration along different paths.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Cresta Neural/citología , Cresta Neural/crecimiento & desarrollo , Análisis de Secuencia de ARN/métodos , Animales , Movimiento Celular , Diencéfalo/citología , Diencéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Mesencéfalo/citología , Mesencéfalo/crecimiento & desarrollo , Ratones , Proteínas del Tejido Nervioso/genética , Neuropilina-1/genética , Neuropilina-2/genética , Rombencéfalo/citología , Rombencéfalo/crecimiento & desarrollo
18.
Cereb Cortex ; 27(11): 5095-5115, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334187

RESUMEN

Auditory-evoked potentials are classically defined as the summations of synchronous firing along the auditory neuraxis. Converging evidence supports a model whereby timing jitter in neural coding compromises listening and causes variable scalp-recorded potentials. Yet the intrinsic noise of human scalp recordings precludes a full understanding of the biological origins of individual differences in listening skills. To delineate the mechanisms contributing to these phenomena, in vivo extracellular activity was recorded from inferior colliculus in guinea pigs to speech in quiet and noise. Here we show that trial-by-trial timing jitter is a mechanism contributing to auditory response variability. Identical variability patterns were observed in scalp recordings in human children, implicating jittered timing as a factor underlying reduced coding of dynamic speech features and speech in noise. Moreover, intertrial variability in human listeners is tied to language development. Together, these findings suggest that variable timing in inferior colliculus blurs the neural coding of speech in noise, and propose a consequence of this timing jitter for human behavior. These results hint both at the mechanisms underlying speech processing in general, and at what may go awry in individuals with listening difficulties.


Asunto(s)
Percepción Auditiva/fisiología , Variación Biológica Individual , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Mesencéfalo/fisiología , Estimulación Acústica , Animales , Preescolar , Estudios de Cohortes , Electroencefalografía , Femenino , Cobayas , Humanos , Inteligencia , Masculino , Mesencéfalo/crecimiento & desarrollo , Microelectrodos , Modelos Animales , Ruido , Caracteres Sexuales , Habla
19.
Cell Rep ; 17(12): 3233-3245, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009292

RESUMEN

Neural circuits involving midbrain dopaminergic (DA) neurons regulate reward and goal-directed behaviors. Although local GABAergic input is known to modulate DA circuits, the mechanism that controls excitatory/inhibitory synaptic balance in DA neurons remains unclear. Here, we show that DA neurons use autocrine transforming growth factor ß (TGF-ß) signaling to promote the growth of axons and dendrites. Surprisingly, removing TGF-ß type II receptor in DA neurons also disrupts the balance in TGF-ß1 expression in DA neurons and neighboring GABAergic neurons, which increases inhibitory input, reduces excitatory synaptic input, and alters phasic firing patterns in DA neurons. Mice lacking TGF-ß signaling in DA neurons are hyperactive and exhibit inflexibility in relinquishing learned behaviors and re-establishing new stimulus-reward associations. These results support a role for TGF-ß in regulating the delicate balance of excitatory/inhibitory synaptic input in local microcircuits involving DA and GABAergic neurons and its potential contributions to neuropsychiatric disorders.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Aprendizaje Inverso/fisiología , Factor de Crecimiento Transformador beta1/genética , Animales , Dendritas/genética , Dendritas/fisiología , Neuronas Dopaminérgicas/fisiología , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Regulación de la Expresión Génica , Humanos , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Ratones , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal/genética , Sinapsis/genética , Sinapsis/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
20.
Neuroscience ; 339: 219-234, 2016 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-27717810

RESUMEN

Coordinated activity in different sets of widely-projecting neurochemical systems characterize waking (W) and sleep (S). How and when this coordination is achieved during development is not known. We used embryos and newborns of a precocial bird species (chickens) to assess developmental activation in different neurochemical systems using cFos expression, which has been extensively employed to examine cellular activation during S and W in adult mammals. Similarly to adult mammals, newborn awake chicks showed significantly higher cFos expression in W-active hypocretin/orexin (H/O), serotonergic Dorsal Raphe, noradrenergic Locus Coeruleus and cholinergic Laterodorsal and Pedunculopontine Tegmental (Ch-LDT/PT) neurons when compared to sleeping chicks. cFos expression was significantly correlated both between these systems, and with the amount of W. S-active melanin-concentrating hormone (MCH) neurons showed very low cFos expression with no difference between sleeping and awake chicks, possibly due to the very short duration of S episodes. In embryonic chicks, cFos expression was low or absent across all five systems at embryonic day (E) 12. Unexpectedly, a strong activation was seen at E16 in H/O neurons. The highest activation of Ch-LDT/PT (also S-active) and MCH neurons was seen at E20. These data suggest that maturation of arousal systems is achieved soon after hatching, while S-control networks are active in late chick embryos.


Asunto(s)
Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Sueño/fisiología , Vigilia/fisiología , Acetilcolina/metabolismo , Animales , Proteínas Aviares/metabolismo , Western Blotting , Embrión de Pollo , Pollos , Hormonas Hipotalámicas/metabolismo , Inmunohistoquímica , Melaninas/metabolismo , Mesencéfalo/citología , Microscopía Fluorescente , Neuronas/citología , Norepinefrina/metabolismo , Orexinas/metabolismo , Hormonas Hipofisarias/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Distribución Aleatoria , Serotonina/metabolismo , Privación de Sueño/metabolismo , Privación de Sueño/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA