Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 426
Filtrar
1.
J Neurosci ; 41(46): 9483-9502, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789560

RESUMEN

Understanding how neurons of the striatum are formed and integrate into complex synaptic circuits is essential to provide insight into striatal function in health and disease. In this review, we summarize our current understanding of the development of striatal neurons and associated circuits with a focus on their embryonic origin. Specifically, we address the role of distinct types of embryonic progenitors, found in the proliferative zones of the ganglionic eminences in the ventral telencephalon, in the generation of diverse striatal interneurons and projection neurons. Indeed, recent evidence would suggest that embryonic progenitor origin dictates key characteristics of postnatal cells, including their neurochemical content, their location within striatum, and their long-range synaptic inputs. We also integrate recent observations regarding embryonic progenitors in cortical and other regions and discuss how this might inform future research on the ganglionic eminences. Last, we examine how embryonic progenitor dysfunction can alter striatal formation, as exemplified in Huntington's disease and autism spectrum disorder, and how increased understanding of embryonic progenitors can have significant implications for future research directions and the development of improved therapeutic options.SIGNIFICANCE STATEMENT This review highlights recently defined novel roles for embryonic progenitor cells in shaping the functional properties of both projection neurons and interneurons of the striatum. It outlines the developmental mechanisms that guide neuronal development from progenitors in the embryonic ganglionic eminences to progeny in the striatum. Where questions remain open, we integrate observations from cortex and other regions to present possible avenues for future research. Last, we provide a progenitor-centric perspective onto both Huntington's disease and autism spectrum disorder. We suggest that future investigations and manipulations of embryonic progenitor cells in both research and clinical settings will likely require careful consideration of their great intrinsic diversity and neurogenic potential.


Asunto(s)
Cuerpo Estriado/embriología , Vías Nerviosas/embriología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Animales , Humanos
2.
Science ; 372(6542)2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33958447

RESUMEN

Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.


Asunto(s)
Atlas como Asunto , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Neurogénesis/genética , ARN Largo no Codificante/genética , Análisis de la Célula Individual , Factores de Transcripción/genética , Feto , Neuronas GABAérgicas/metabolismo , Humanos , RNA-Seq , Transcripción Genética
3.
Cell Rep ; 35(4): 109041, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33910016

RESUMEN

Synaptic circuits in the brain are precisely organized, but the processes that govern this precision are poorly understood. Here, we explore how distinct embryonic neural progenitor pools in the lateral ganglionic eminence contribute to neuronal diversity and synaptic circuit connectivity in the mouse striatum. In utero labeling of Tα1-expressing apical intermediate progenitors (aIP), as well as other progenitors (OP), reveals that both progenitors generate direct and indirect pathway spiny projection neurons (SPNs) with similar electrophysiological and anatomical properties and are intermingled in medial striatum. Subsequent optogenetic circuit-mapping experiments demonstrate that progenitor origin significantly impacts long-range excitatory input strength, with medial prefrontal cortex preferentially driving aIP-derived SPNs and visual cortex preferentially driving OP-derived SPNs. In contrast, the strength of local inhibitory inputs among SPNs is controlled by birthdate rather than progenitor origin. Combined, these results demonstrate distinct roles for embryonic progenitor origin in shaping neuronal and circuit properties of the postnatal striatum.


Asunto(s)
Cuerpo Estriado/embriología , Células Madre/metabolismo , Telencéfalo/embriología , Animales , Ratones
4.
Mol Neurobiol ; 58(8): 3729-3744, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33821423

RESUMEN

Specification of the progenitors' regional identity is a pivotal step during development of the cerebral cortex and basal ganglia. The molecular mechanisms underlying progenitor regionalization, however, are poorly understood. Here we showed that the transcription factor Vax1 was highly expressed in the developing subpallium. In its absence, the RNA-Seq analysis, in situ RNA hybridization, and immunofluorescence staining results showed that the cell proliferation was increased in the subpallium, but the neuronal differentiation was blocked. Moreover, the dLGE expands ventrally, and the vLGE, MGE, and septum get smaller. Finally, overexpressed VAX1 in the LGE progenitors strongly inhibits Gsx2 expression. Taken together, our findings show that Vax1 is crucial for subpallium regionalization by repressing Gsx2.


Asunto(s)
Cuerpo Estriado/embriología , Cuerpo Estriado/metabolismo , Globo Pálido/embriología , Globo Pálido/metabolismo , Proteínas de Homeodominio/biosíntesis , Neuropéptidos/biosíntesis , Animales , Cuerpo Estriado/citología , Globo Pálido/citología , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuropéptidos/genética
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.
Cell Rep ; 29(6): 1419-1428.e5, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693884

RESUMEN

One long-standing model of striatal function divides the striatum into compartments called striosome and matrix. While some anatomical evidence suggests that these populations represent distinct striatal pathways with differing inputs and outputs, functional investigation has been limited by the methods for identifying and manipulating these populations. Here, we utilize hs599CreER mice as a new tool for targeting striosome projection neurons and testing their functional connectivity. Extending anatomical work, we demonstrate that striosome neurons receive greater synaptic input from prelimbic cortex, whereas matrix neurons receive greater input from primary motor cortex. We also identify functional differences in how striosome and matrix neurons process excitatory input, providing the first electrophysiological method for delineating striatal output neuron subtypes. Lastly, we provide the first functional demonstration that striosome neurons are the predominant striatal output to substantia nigra pars compacta dopamine neurons. These results identify striosome and matrix as functionally distinct striatal pathways.


Asunto(s)
Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/fisiología , Corteza Motora/fisiología , Vías Nerviosas/fisiología , Neurogénesis , Corteza Prefrontal/fisiología , Animales , Cuerpo Estriado/embriología , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Ratones , Ratones Transgénicos , Corteza Motora/citología , Corteza Motora/metabolismo , Neurogénesis/efectos de los fármacos , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Sustancia Negra/citología , Sustancia Negra/metabolismo , Sustancia Negra/fisiología
7.
Sci Rep ; 9(1): 1976, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760829

RESUMEN

Neuropsychiatric disease has polygenic determinants but is often precipitated by environmental pressures, including adverse perinatal events. However, the way in which genetic vulnerability and early-life adversity interact remains obscure. We hypothesised that the extreme environmental stress of prematurity would promote neuroanatomic abnormality in individuals genetically vulnerable to psychiatric disorders. In 194 unrelated infants (104 males, 90 females), born before 33 weeks of gestation (mean gestational age 29.7 weeks), we combined Magnetic Resonance Imaging with a polygenic risk score (PRS) for five psychiatric pathologies to test the prediction that: deep grey matter abnormalities frequently seen in preterm infants are associated with increased polygenic risk for psychiatric illness. The variance explained by the PRS in the relative volumes of four deep grey matter structures (caudate nucleus, thalamus, subthalamic nucleus and lentiform nucleus) was estimated using linear regression both for the full, mixed ancestral, cohort and a subsample of European infants. Psychiatric PRS was negatively associated with lentiform volume in the full cohort (ß = -0.24, p = 8 × 10-4) and a European subsample (ß = -0.24, p = 8 × 10-3). Genetic variants associated with neuropsychiatric disease increase vulnerability to abnormal lentiform development after perinatal stress and are associated with neuroanatomic changes in the perinatal period.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Sustancia Gris/embriología , Enfermedades del Prematuro/genética , Enfermedades del Prematuro/psicología , Trastornos Mentales/genética , Herencia Multifactorial/genética , Mapeo Encefálico , Núcleo Caudado/anomalías , Núcleo Caudado/embriología , Cuerpo Estriado/anomalías , Cuerpo Estriado/embriología , Europa (Continente) , Femenino , Sustancia Gris/anomalías , Humanos , Recién Nacido , Recien Nacido Prematuro/psicología , Imagen por Resonancia Magnética , Masculino , Núcleo Subtalámico/anomalías , Núcleo Subtalámico/embriología , Tálamo/anomalías , Tálamo/embriología
8.
Cereb Cortex ; 29(5): 2115-2124, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29688344

RESUMEN

DACH1 is the human homolog of the Drosophila dachshund gene, which is involved in the development of the eye, nervous system, and limbs in the fly. Here, we systematically investigate DACH1 expression patterns during human neurodevelopment, from 5 to 21 postconceptional weeks. By immunodetection analysis, we found that DACH1 is highly expressed in the proliferating neuroprogenitors of the developing cortical ventricular and subventricular regions, while it is absent in the more differentiated cortical plate. Single-cell global transcriptional analysis revealed that DACH1 is specifically enriched in neuroepithelial and ventricular radial glia cells of the developing human neocortex. Moreover, we describe a previously unreported DACH1 expression in the human striatum, in particular in the striatal medium spiny neurons. This finding qualifies DACH1 as a new striatal projection neuron marker, together with PPP1R1B, BCL11B, and EBF1. We finally compared DACH1 expression profile in human and mouse forebrain, where we observed spatio-temporal similarities in its expression pattern thus providing a precise developmental description of DACH1 in the 2 mammalian species.


Asunto(s)
Cuerpo Estriado/embriología , Cuerpo Estriado/metabolismo , Proteínas del Ojo/metabolismo , Neocórtex/embriología , Neocórtex/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Feto Abortado/embriología , Feto Abortado/metabolismo , Células Ependimogliales/metabolismo , Edad Gestacional , Humanos , Ventrículos Laterales/embriología , Ventrículos Laterales/metabolismo , Células-Madre Neurales/metabolismo , Células Neuroepiteliales/metabolismo , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Especificidad de la Especie
9.
Int J Dev Neurosci ; 66: 1-8, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29183706

RESUMEN

Dopamine is widely involved in behaviors related to motor activity, cognition, motivation, and reward. Dopamine signal is transduced through the dopamine receptor gene family. The dopamine D1 receptor (D1R) is highly expressed in the striatum, and is responsible for regulating the motor function. Recently, we have reported that the knockdown (KD) mice in which D1R was conditionally eliminated at adult stage, displayed a hypoactivity in the home cage than wild type mice; however, conventional D1R knockout (KO) mice show hyperactive phenotypes. In order to assess whether the difference in the time of eliminating D1R expression affects the behavioral phenotypes, we generated D1R KD mice at the postnatal and adult stages, and compared their motor function with D1R KO mice. Consequently, D1R KD at postnatal and adult stages resulted in severe locomotive defects compared with D1R KO mice. These results suggested that D1R has versatile functions, and the knockdown timing greatly influences the normal motor activity in the adolescent to adult stages.


Asunto(s)
Envejecimiento , Regulación del Desarrollo de la Expresión Génica/genética , Hipercinesia/genética , Receptores de Dopamina D1/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Antibacterianos/farmacología , Cuerpo Estriado/embriología , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Doxiciclina/farmacología , Embrión de Mamíferos , Conducta Exploratoria/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipercinesia/fisiopatología , Locomoción/efectos de los fármacos , Locomoción/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Destreza Motora/fisiología , Receptores de Dopamina D1/genética
10.
J Comp Neurol ; 525(13): 2805-2819, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472858

RESUMEN

The striatum is the major component of the basal ganglia and is well known to play a key role in the control of motor function via balanced output from the indirect (iSPNs) and direct pathway striatal projection neurons (dSPNs). Little is known, however, about the molecular genetic mechanisms that control the formation of the iSPNs versus dSPNs. We show here that the SoxE family member, Sox8, is co-expressed with the dSPN markers, Isl1 and Ebf1, in the developing striatum. Moreover, dSPNs, as marked by Isl1-cre fate map, express Sox8 in the embryonic striatum and Sox8-EGFP BAC transgenic mice specifically reveal the direct pathway axons during development. These EGFP+ axons are first observed to reach their midbrain target, the substantia nigra pars reticulata (SNr), at E14 in the mouse with a robust connection observed already at birth. The selective expression of EGFP in dSPNs of Sox8-EGFP BAC mice is maintained at postnatal timepoints. Sox8 is known to be expressed in oligodendrocyte precursor cells (OPCs) together with other SoxE factors and we show here that the EGFP signal co-localizes with the OPC markers throughout the brain. Finally, we show that Sox8-EGFP BAC mice can be used to interrogate the altered dSPN development in Isl1 conditional mutants including aberrant axonal projections detected already at embryonic timepoints. Thus, Sox8 represents an early and specific marker of embryonic dSPNs and the Sox8-EGFP BAC transgenic mice are an excellent tool to study the development of basal ganglia circuitry.


Asunto(s)
Cuerpo Estriado , Regulación del Desarrollo de la Expresión Génica/fisiología , Vías Nerviosas/fisiología , Neuronas/metabolismo , Factores de Transcripción SOXE/metabolismo , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/metabolismo , Embrión de Mamíferos , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Mol Cell Neurosci ; 80: 44-51, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28213137

RESUMEN

Recent studies have shown that the LIM-homeodomain transcription factor Isl1 is required for the survival and differentiation of direct pathway striatonigral neurons during embryonic development. The downstream effectors of Isl1 in these processes are presently unknown. We show here that Foxo1, a transcription factor that has been implicated in cell survival, is expressed in striatal projection neurons (SPNs) that derive from the Isl1 lineage (i.e. direct pathway SPNs). Moreover, Isl1 conditional knockouts (cKOs) show a severe loss of Foxo1 expression at E15.5 with a modest recovery by E18.5. Although Foxo1 is enriched in the direct pathway SPNs at embryonic stages, it is expressed in both direct and indirect pathway SPNs at postnatal time points as evidenced by co-localization with EGFP in both Drd1-EGFP and Drd2-EGFP BAC transgenic mice. Foxo1 was not detected in striatal interneurons as marked by the transcription factor Nkx2.1. Conditional knockout of Foxo1 using Dlx5/6-CIE mice results in reduced expression of the SPN marker Darpp-32, as well as in the direct pathway SPN markers Ebf1 and Zfp521 within the embryonic striatum at E15.5. However, this phenotype improves in the conditional mutants by E18.5. Interestingly, the Foxo family members, Foxo3 and Foxo6, remain expressed at late embryonic stages in the Foxo1 cKOs unlike the Isl1 cKOs where Foxo1/3/6 as well as the Foxo1/3 target Bach2 are all reduced. Taken together, these findings suggest that Foxo-regulated pathways are downstream of Isl1 in the survival and/or differentiation of direct pathway SPNs.


Asunto(s)
Cuerpo Estriado/citología , Proteína Forkhead Box O1/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas con Homeodominio LIM/metabolismo , Neuronas/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Diferenciación Celular/fisiología , Cuerpo Estriado/embriología , Cuerpo Estriado/metabolismo , Embrión de Mamíferos , Proteína Forkhead Box O1/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Vías Nerviosas/fisiología , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/genética
12.
J Comp Neurol ; 525(4): 794-817, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27532901

RESUMEN

The striatum, the largest nucleus of the basal ganglia controlling motor and cognitive functions, can be characterized by a labyrinthine mosaic organization of striosome/matrix compartments. It is unclear how striosome/matrix mosaic formation is spatially and temporally controlled at the cellular level during striatal development. Here, by combining in vivo electroporation and brain slice cultures, we set up a prospective experimental system in which we differentially labeled striosome and matrix cells from the time of birth and followed their distributions and migratory behaviors. Our results showed that, at an initial stage of striosome/matrix mosaic formation, striosome cells were mostly stationary, whereas matrix cells actively migrated in multiple directions regardless of the presence of striosome cells. The mostly stationary striosome cells were still able to associate to form patchy clusters via attractive interactions. Our results suggest that the restricted migratory capability of striosome cells may allow them to cluster together only when they happen to be located in close proximity to each other and are not separated by actively migrating matrix cells. The way in which the mutidirectionally migrating matrix cells intermingle with the mostly stationary striosome cells may therefore determine the topographic features of striosomes. At later stages, the actively migrating matrix cells began to repulse the patchy clusters of striosomes, presumably enhancing the striosome cluster formation and the segregation and eventual formation of dichotomous homogeneous striosome/matrix compartments. Overall, our study reveals temporally distinct migratory behaviors of striosome/matrix cells, which may underlie the sequential steps of mosaic formation in the developing striatum. J. Comp. Neurol. 525:794-817, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Neurogénesis/fisiología , Neuronas/citología , Animales , Movimiento Celular/fisiología , Inmunohistoquímica , Ratones , Ratones Endogámicos ICR , Imagen de Lapso de Tiempo
13.
Neuroscience ; 333: 320-30, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27475250

RESUMEN

Human Dopamine- and cAMP-regulated phosphoprotein of molecular weight 32kDa (DARPP-32, also known as PPP1R1B) gene codes for different transcripts that are mainly translated into two DARPP-32 protein isoforms, full length (fl)-DARPP-32 and truncated (t)-DARPP. The t-DARPP lacks the first 36 residues at the N-terminal, which alters its function. In the central nervous system, fl-DARPP-32 is highly expressed in GABAergic striatal medium spiny neurons (MSNs), where it integrates dopaminergic and glutamatergic input signaling. However, no information about human DARPP-32 isoform expression during MSNs maturation is available. In this study, our aim is to determine the expression of the two DARPP-32 isoforms in human fetal and adult striatal samples. We show that DARPP-32 isoform expression is differentially regulated during human striatal development, with the t-DARPP isoform being virtually absent from whole ganglionic eminence (WGE) and highly induced in the adult striatum (in both caudate and putamen). We next compared the four most common anti-DARPP-32 antibodies used in human specimens, to study their recognition of the two isoforms in fetal and adult human striatal samples by western blot and immunohistochemistry. The four antibodies specifically identify the fl-DARPP-32 in both fetal and adult samples, while t-DARPP form was only detected in adult striatal samples. In addition, the lack of t-DARPP recognition in human adult striatum by the antibody generated against the full-length domain produces in turn different efficacy by immunohistochemical analysis. In conclusion, our results show that expression of human DARPP-32 protein isoforms depends on the striatal neurodevelopmental stage with t-DARPP being specific for the human adult striatum.


Asunto(s)
Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Femenino , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido
14.
Mol Cell Neurosci ; 75: 50-62, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27370937

RESUMEN

Over the past decades, studies in both Huntington's disease animal models and pilot clinical trials have demonstrated that replacement of degenerated striatum and repair of circuitries by grafting fetal striatal primordium is feasible, safe and may counteract disease progression. However, a better comprehension of striatal ontogenesis is required to assess the fetal graft regenerative potential. During neuronal development, neurotrophins exert pleiotropic actions in regulating cell fate and synaptic plasticity. In this regard, brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF2) are crucially implicated in the control of fate choice of striatal progenitor cells. In this study, we intended to refine the functional features of human striatal precursor (HSP) cells isolated from ganglionic eminence of 9-12week old human fetuses, by studying with electrophysiological methods the effect of BDNF and FGF2 on the membrane biophysical properties and the voltage-dependent Ca(2+) currents. These features are particularly relevant to evaluate neuronal cell functioning and can be considered reliable markers of the developmental phenotype of human striatal primordium. Our results have demonstrated that BDNF and FGF2 induced membrane hyperpolarization, increased the membrane capacitance and reduced the resting total and specific conductance values, suggesting a more efficient control of resting ionic fluxes. Moreover, the treatment with both neurotrophins enhanced N-type Ca(2+) current amplitude and reduced L- and T-type ones. Overall, our data indicate that BDNF and FGF2 may help HSP cells to attain a more functionally mature phenotype.


Asunto(s)
Potenciales de Acción , Factor Neurotrófico Derivado del Encéfalo/farmacología , Canales de Calcio/metabolismo , Cuerpo Estriado/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células-Madre Neurales/fisiología , Neurogénesis , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Humanos , Células-Madre Neurales/efectos de los fármacos
15.
Amino Acids ; 48(8): 1913-27, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27129463

RESUMEN

Transplantation of neural progenitor cells (NPCs) is a promising experimental therapy for Huntington's disease (HD). The variables responsible for the success of this approach, including selection of the optimal developmental stage of the grafted cells, are however largely unknown. Supporting cellular energy metabolism by creatine (Cr) supplementation is a clinically translatable method for improving cell transplantation strategies. The present study aims at investigating differences between early (E14) and late (E18) developmental stages of rat striatal NPCs in vitro. NPCs were isolated from E14 and E18 embryos and cultured for 7 days with or without Cr [5 mM]. Chronic treatment significantly increased the percentage of GABA-immunoreactive neurons as compared to untreated controls, both in the E14 (170.4 ± 4.7 %) and the E18 groups (129.3 ± 9.3 %). This effect was greater in E14 cultures (p < 0.05). Similarly, short-term treatment for 24 h resulted in increased induction (p < 0.05) of the GABA-ergic phenotype in E14 (163.0 ± 10.4 %), compared to E18 cultures (133.3 ± 9.5 %). Total neuronal cell numbers and general viability were not affected by Cr (p > 0.05). Protective effects of Cr against a metabolic insult were equal in E14 and E18 NPCs (p > 0.05). Cr exposure promoted morphological differentiation of GABA-ergic neurons, including neurite length in both groups (p < 0.05), but the number of branching points was increased only in the E18 group (p < 0.05). Our results demonstrate that the role of Cr as a GABA-ergic differentiation factor depends on the developmental stage of striatal NPCs, while Cr-mediated neuroprotection is not significantly influenced. These findings have potential implications for optimizing future cell replacement strategies in HD.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cuerpo Estriado/embriología , Creatina/farmacología , Embrión de Mamíferos/embriología , Neuronas GABAérgicas/metabolismo , Células-Madre Neurales/metabolismo , Animales , Cuerpo Estriado/citología , Embrión de Mamíferos/citología , Femenino , Neuronas GABAérgicas/citología , Células-Madre Neurales/citología , Ratas , Ratas Wistar
16.
Exp Neurol ; 282: 9-18, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27154297

RESUMEN

Identifying the steps involved in striatal development is important both for understanding the striatum in health and disease, and for generating protocols to differentiate striatal neurons for regenerative medicine. The most prominent neuronal subtype in the adult striatum is the medium spiny projection neuron (MSN), which constitutes more than 85% of all striatal neurons and classically expresses DARPP-32. Through a microarray study of genes expressed in the whole ganglionic eminence (WGE: the developing striatum) in the mouse, we identified the gene encoding the transcription factor Forkhead box protein P1 (FoxP1) as the most highly up-regulated gene, thus providing unbiased evidence for the association of FoxP1 with MSN development. We also describe the expression of FoxP1 in the human fetal brain over equivalent gestational stages. FoxP1 expression persisted through into adulthood in the mouse brain, where it co-localised with all striatal DARPP-32 positive projection neurons and a small population of DARPP-32 negative cells. There was no co-localisation of FoxP1 with any interneuron markers. FoxP1 was detectable in primary fetal striatal cells following dissection, culture, and transplantation into the adult lesioned striatum, demonstrating its utility as an MSN marker for transplantation studies. Furthermore, DARPP-32 expression was absent from FoxP1 knock-out mouse WGE differentiated in vitro, suggesting that FoxP1 is important for the development of DARPP-32-positive MSNs. In summary, we show that FoxP1 labels MSN precursors prior to the expression of DARPP-32 during normal development, and in addition suggest that FoxP1 labels a sub-population of MSNs that are not co-labelled by DARPP-32. We demonstrate the utility of FoxP1 to label MSNs in vitro and following neural transplantation, and show that FoxP1 is required for DARPP-32 positive MSN differentiation in vitro.


Asunto(s)
Diferenciación Celular/fisiología , Cuerpo Estriado , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células-Madre Neurales/fisiología , Neuronas/citología , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Animales , Animales Recién Nacidos , Proteínas Portadoras/metabolismo , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Cuerpo Estriado/crecimiento & desarrollo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Embrión de Mamíferos , Endodesoxirribonucleasas , Feto/citología , Factores de Transcripción Forkhead/genética , Técnicas In Vitro , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/trasplante , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Estadísticas no Paramétricas
17.
J Neurosci ; 36(5): 1711-22, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26843651

RESUMEN

Although comprehensively described during early neuronal development, the role of DNA methylation/demethylation in neuronal lineage and subtype specification is not well understood. By studying two distinct neuronal progenitors as they differentiate to principal neurons in mouse hippocampus and striatum, we uncovered several principles governing neuronal DNA methylation during brain development. (1) The program consists of three stages: an initial genome-wide methylation during progenitor proliferation is followed by loss of methylation during the transition of regional progenitors to "young" hippocampal/striatal neurons, which is then reversed by gain in methylation during maturation to subtype-specific neurons. (2) At the first two stages, gain and loss of methylation are limited to CpGs, whereas during the third maturation stage, methylation also occurs at non-CpG sites in both lineages. (3) Methylation/demethylation, similar to transcription, are initially highly similar in the two lineages, whereas diversification in methylation and transcription during maturation creates subtype-specific methylation differences. (4) Initially, methylation targets all genomic locations, whereas later, during early and late differentiation, the preferred targets are intronic/intergenic sequences with enhancer-like activity. (5) Differentially methylated genes are enriched in sequential neurodevelopmental functions (such as progenitor proliferation, migration, neuritogenesis, and synaptic transmission); upregulated genes represent current and consecutive stage-specific functions, and downregulated genes represent preceding functions that are no longer required. The main conclusion of our work is that the neuronal methylation/demethylation program is predominantly developmental with minimal lineage specificity, except in the final stage of development when neuron subtype-specific differences also emerge. SIGNIFICANCE STATEMENT: Our work is the first to describe a set of relatively simple rules that govern DNA methylation and demethylation in neuronal development in vivo. By dividing neurodevelopment to three major stages and applying rules to each of them, we created a matrix that comprehensively describes DNA methylation/demethylation events in two neuronal lineages, with a total of 10 cell types spanning the entire neurodevelopment. Beyond increasing our understanding of the epigenetic regulation of normal development, our work will be useful in deciphering how environmental perturbations, such as gestational toxins, drugs, stress, infection, and offspring neglect/maltreatment, interfere with the developmental methylation program.


Asunto(s)
Linaje de la Célula/fisiología , Cuerpo Estriado/fisiología , Metilación de ADN/fisiología , Hipocampo/fisiología , Neuronas/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Células Cultivadas , Cuerpo Estriado/embriología , Drosophila , Femenino , Hipocampo/embriología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Embarazo
18.
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
19.
Brain Struct Funct ; 221(6): 2905-17, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26163006

RESUMEN

In the mammalian telencephalon, part of the progenitor cells transition from multipolar to bipolar morphology as they invade the mantle zone. This associates with changing patterns of radial migration. However, the molecules implicated in these morphology transitions are not well known. In the present work, we analyzed the function of FoxP2 protein in this process during telencephalic development in vertebrates. We analyzed the expression of FoxP2 protein and its relation with cell morphology and migratory patterns in mouse and chicken developing striatum. We observed FoxP2 protein expressed in a gradient from the subventricular zone to the mantle layer in mice embryos. In the FoxP2 low domain cells showed multipolar migration. In the striatal mantle layer where FoxP2 protein expression is higher, cells showed locomoting migration and bipolar morphology. In contrast, FoxP2 showed a high and homogenous expression pattern in chicken striatum, thus bipolar morphology predominated. Elevation of FoxP2 in the striatal subventricular zone by in utero electroporation promoted bipolar morphology and impaired multipolar radial migration. In mouse cerebral cortex we obtained similar results. FoxP2 promotes transition from multipolar to bipolar morphology by means of gradiental expression in mouse striatum and cortex. Together these results indicate a role of FoxP2 differential expression in cell morphology control of the vertebrate telencephalon.


Asunto(s)
Proteínas Aviares/fisiología , Movimiento Celular , Factores de Transcripción Forkhead/fisiología , Proteínas Represoras/fisiología , Telencéfalo/citología , Telencéfalo/embriología , Animales , Proteínas Aviares/metabolismo , Células Cultivadas , Embrión de Pollo , Pollos , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Cuerpo Estriado/metabolismo , Factores de Transcripción Forkhead/metabolismo , Ventrículos Laterales/citología , Ventrículos Laterales/embriología , Ventrículos Laterales/metabolismo , Ratones , Neuritas/metabolismo , Neuritas/fisiología , Proteínas Represoras/metabolismo , Telencéfalo/metabolismo , Vertebrados/embriología
20.
Neuroscience ; 314: 134-44, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26642808

RESUMEN

Fibroblast growth factors (FGFs) regulate development and maintenance, and reduce vulnerability of neurons. FGF-2 is essential for survival of midbrain dopaminergic (DA) neurons and is responsible for their dysplasia and disease-related degeneration. We previously reported that FGF-2 is involved in adequate forebrain (FB) target innervation by these neurons in an organotypic co-culture model. It remains unclear, how this ex-vivo phenotype relates to the in vivo situation, and which FGF-related signaling pathway is involved in this process. Here, we demonstrate that lack of FGF-2 results in an increased volume of the striatal target area in mice. We further add evidence that the low molecular weight (LMW) FGF-2 isoform is responsible for this phenotype, as this isoform is predominantly expressed in the embryonic ventral midbrain (VM) as well as in postnatal striatum (STR) and known to act via canonical transmembrane FGF receptor (FGFR) activation. Additionally, we confirm that the phenotype with an enlarged FB-target area by DA neurons can be mimicked in an ex-vivo explant model by inhibiting the canonical FGFR signaling, which resulted in decreased extracellular signal-regulated kinase (ERK) activation, while AKT activation remained unchanged.


Asunto(s)
Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Neuronas Dopaminérgicas/citología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Sustancia Negra/citología , Sustancia Negra/metabolismo , Animales , Cuerpo Estriado/embriología , Neuronas Dopaminérgicas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas/citología , Vías Nerviosas/embriología , Vías Nerviosas/metabolismo , Prosencéfalo , Isoformas de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Sustancia Negra/embriología , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...