Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.499
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nature ; 625(7996): 788-796, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029793

RESUMEN

The expansion of the neocortex, a hallmark of mammalian evolution1,2, was accompanied by an increase in cerebellar neuron numbers3. However, little is known about the evolution of the cellular programmes underlying the development of the cerebellum in mammals. In this study we generated single-nucleus RNA-sequencing data for around 400,000 cells to trace the development of the cerebellum from early neurogenesis to adulthood in human, mouse and the marsupial opossum. We established a consensus classification of the cellular diversity in the developing mammalian cerebellum and validated it by spatial mapping in the fetal human cerebellum. Our cross-species analyses revealed largely conserved developmental dynamics of cell-type generation, except for Purkinje cells, for which we observed an expansion of early-born subtypes in the human lineage. Global transcriptome profiles, conserved cell-state markers and gene-expression trajectories across neuronal differentiation show that cerebellar cell-type-defining programmes have been overall preserved for at least 160 million years. However, we also identified many orthologous genes that gained or lost expression in cerebellar neural cell types in one of the species or evolved new expression trajectories during neuronal differentiation, indicating widespread gene repurposing at the cell-type level. In sum, our study unveils shared and lineage-specific gene-expression programmes governing the development of cerebellar cells and expands our understanding of mammalian brain evolution.


Asunto(s)
Cerebelo , Evolución Molecular , Mamíferos , Neurogénesis , Animales , Humanos , Ratones , Linaje de la Célula/genética , Cerebelo/citología , Cerebelo/embriología , Cerebelo/crecimiento & desarrollo , Feto/citología , Feto/embriología , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Zarigüeyas/embriología , Zarigüeyas/crecimiento & desarrollo , Células de Purkinje/citología , Células de Purkinje/metabolismo , Análisis de Expresión Génica de una Sola Célula , Especificidad de la Especie , Transcriptoma , Mamíferos/embriología , Mamíferos/crecimiento & desarrollo
2.
Cell ; 154(1): 118-33, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827678

RESUMEN

The CACNA1A gene, encoding the voltage-gated calcium channel subunit α1A, is involved in pre- and postsynaptic Ca(2+) signaling, gene expression, and several genetic neurological disorders. We found that CACNA1A coordinates gene expression using a bicistronic mRNA bearing a cryptic internal ribosomal entry site (IRES). The first cistron encodes the well-characterized α1A subunit. The second expresses a transcription factor, α1ACT, which coordinates expression of a program of genes involved in neural and Purkinje cell development. α1ACT also contains the polyglutamine (polyQ) tract that, when expanded, causes spinocerebellar ataxia type 6 (SCA6). When expressed as an independent polypeptide, α1ACT-bearing an expanded polyQ tract-lacks transcription factor function and neurite outgrowth properties, causes cell death in culture, and leads to ataxia and cerebellar atrophy in transgenic mice. Suppression of CACNA1A IRES function in SCA6 may be a potential therapeutic strategy.


Asunto(s)
Canales de Calcio/genética , Ataxias Espinocerebelosas/genética , Factores de Transcripción/genética , Animales , Canales de Calcio/metabolismo , Muerte Celular , Línea Celular Tumoral , Cerebelo/embriología , Cerebelo/fisiopatología , Regulación de la Expresión Génica , Humanos , Ratones , Neuritas/metabolismo , Péptidos/genética , Células de Purkinje/metabolismo , Ratas , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/fisiopatología , Factores de Transcripción/metabolismo , Transcripción Genética
3.
Nature ; 609(7929): 1012-1020, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36131015

RESUMEN

Medulloblastoma, a malignant childhood cerebellar tumour, segregates molecularly into biologically distinct subgroups, suggesting that a personalized approach to therapy would be beneficial1. Mouse modelling and cross-species genomics have provided increasing evidence of discrete, subgroup-specific developmental origins2. However, the anatomical and cellular complexity of developing human tissues3-particularly within the rhombic lip germinal zone, which produces all glutamatergic neuronal lineages before internalization into the cerebellar nodulus-makes it difficult to validate previous inferences that were derived from studies in mice. Here we use multi-omics to resolve the origins of medulloblastoma subgroups in the developing human cerebellum. Molecular signatures encoded within a human rhombic-lip-derived lineage trajectory aligned with photoreceptor and unipolar brush cell expression profiles that are maintained in group 3 and group 4 medulloblastoma, suggesting a convergent basis. A systematic diagnostic-imaging review of a prospective institutional cohort localized the putative anatomical origins of group 3 and group 4 tumours to the nodulus. Our results connect the molecular and phenotypic features of clinically challenging medulloblastoma subgroups to their unified beginnings in the rhombic lip in the early stages of human development.


Asunto(s)
Linaje de la Célula , Neoplasias Cerebelosas , Meduloblastoma , Metencéfalo , Animales , Neoplasias Cerebelosas/clasificación , Neoplasias Cerebelosas/embriología , Neoplasias Cerebelosas/patología , Cerebelo/embriología , Humanos , Meduloblastoma/clasificación , Meduloblastoma/embriología , Meduloblastoma/patología , Metencéfalo/embriología , Ratones , Neuronas/patología , Estudios Prospectivos
4.
Nature ; 609(7929): 1021-1028, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36131014

RESUMEN

Medulloblastoma (MB) comprises a group of heterogeneous paediatric embryonal neoplasms of the hindbrain with strong links to early development of the hindbrain1-4. Mutations that activate Sonic hedgehog signalling lead to Sonic hedgehog MB in the upper rhombic lip (RL) granule cell lineage5-8. By contrast, mutations that activate WNT signalling lead to WNT MB in the lower RL9,10. However, little is known about the more commonly occurring group 4 (G4) MB, which is thought to arise in the unipolar brush cell lineage3,4. Here we demonstrate that somatic mutations that cause G4 MB converge on the core binding factor alpha (CBFA) complex and mutually exclusive alterations that affect CBFA2T2, CBFA2T3, PRDM6, UTX and OTX2. CBFA2T2 is expressed early in the progenitor cells of the cerebellar RL subventricular zone in Homo sapiens, and G4 MB transcriptionally resembles these progenitors but are stalled in developmental time. Knockdown of OTX2 in model systems relieves this differentiation blockade, which allows MB cells to spontaneously proceed along normal developmental differentiation trajectories. The specific nature of the split human RL, which is destined to generate most of the neurons in the human brain, and its high level of susceptible EOMES+KI67+ unipolar brush cell progenitor cells probably predisposes our species to the development of G4 MB.


Asunto(s)
Diferenciación Celular , Neoplasias Cerebelosas , Meduloblastoma , Metencéfalo , Diferenciación Celular/genética , Linaje de la Célula , Neoplasias Cerebelosas/clasificación , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Cerebelo/embriología , Cerebelo/patología , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Proteínas Hedgehog/metabolismo , Histona Demetilasas , Humanos , Antígeno Ki-67/metabolismo , Meduloblastoma/clasificación , Meduloblastoma/genética , Meduloblastoma/patología , Metencéfalo/embriología , Metencéfalo/patología , Proteínas Musculares , Mutación , Factores de Transcripción Otx/deficiencia , Factores de Transcripción Otx/genética , Proteínas Represoras , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción
5.
Nature ; 572(7767): 67-73, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31043743

RESUMEN

Study of the origin and development of cerebellar tumours has been hampered by the complexity and heterogeneity of cerebellar cells that change over the course of development. Here we use single-cell transcriptomics to study more than 60,000 cells from the developing mouse cerebellum and show that different molecular subgroups of childhood cerebellar tumours mirror the transcription of cells from distinct, temporally restricted cerebellar lineages. The Sonic Hedgehog medulloblastoma subgroup transcriptionally mirrors the granule cell hierarchy as expected, while group 3 medulloblastoma resembles Nestin+ stem cells, group 4 medulloblastoma resembles unipolar brush cells, and PFA/PFB ependymoma and cerebellar pilocytic astrocytoma resemble the prenatal gliogenic progenitor cells. Furthermore, single-cell transcriptomics of human childhood cerebellar tumours demonstrates that many bulk tumours contain a mixed population of cells with divergent differentiation. Our data highlight cerebellar tumours as a disorder of early brain development and provide a proximate explanation for the peak incidence of cerebellar tumours in early childhood.


Asunto(s)
Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Evolución Molecular , Feto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Transcripción Genética , Animales , Neoplasias Cerebelosas/clasificación , Cerebelo/citología , Cerebelo/embriología , Cerebelo/metabolismo , Niño , Femenino , Feto/citología , Glioma/clasificación , Glioma/genética , Glioma/patología , Humanos , Meduloblastoma/clasificación , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factores de Tiempo , Transcriptoma/genética
6.
Turk J Med Sci ; 54(1): 1-15, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38812654

RESUMEN

Background/aim: This study aims to determine the possible embryotoxic effects of propofol on the cerebellum and spinal cord using fertile chicken eggs. Materials and methods: A total of 430 fertile eggs were divided into 5 groups: control, saline, 2.5 mg.kg-1, 12.5 mg.kg-1, and 37.5 mg.kg-1 propofol. Injections were made immediately before incubation via the air chamber. On the 15th, 18th, and 21st day of incubation, 6 embryos from each group were evaluated. Serial paraffin sections taken from the cerebellum and spinal cord were stained with hematoxylin-eosin, Kluver-Barrera, toluidine blue, and periodic acid-Schiff's reaction. The outer granular layer and total cortex thickness were measured, and the linear density of the Purkinje cells was determined. The ratios of the substantia grisea surface area to the total surface area of the spinal cord were calculated. The transverse and longitudinal diameters of the canalis centralis were also assessed. Results: No structural malformation was observed in any embryos examined macroscopically. No significant difference was observed between the groups in terms of development and histologic organization of the cerebellum and spinal cord. However, on the 15th, 18th, and 21st day, the outer granular layer (p < 0.001 for all days) and the total cortex thickness (p < 0.01, p < 0.001, and p < 0.001, respectively) decreased significantly in different propofol dose groups in varying degrees in the cerebellum. Similarly, in the spinal cord, there were significant changes in the ratios of the substantia grisea surface area to the total surface area (p < 0.01 and p < 0.001, respectively). Conclusion: It was concluded that the in-ovo-administered propofol given immediately before incubation has adverse effects on the developing cerebellum and spinal cord. Therefore, it is important for anesthesiologists always to remain vigilant when treating female patients of childbearing age.


Asunto(s)
Cerebelo , Propofol , Médula Espinal , Animales , Propofol/toxicidad , Propofol/administración & dosificación , Cerebelo/efectos de los fármacos , Cerebelo/patología , Cerebelo/embriología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/embriología , Embrión de Pollo/efectos de los fármacos , Anestésicos Intravenosos/toxicidad , Anestésicos Intravenosos/administración & dosificación
7.
Genome Res ; 30(5): 749-756, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32430339

RESUMEN

Dimension-reduction methods, such as t-SNE or UMAP, are widely used when exploring high-dimensional data describing many entities, for example, RNA-seq data for many single cells. However, dimension reduction is commonly prone to introducing artifacts, and we hence need means to see where a dimension-reduced embedding is a faithful representation of the local neighborhood and where it is not. We present Sleepwalk, a simple but powerful tool that allows the user to interactively explore an embedding, using color to depict original or any other distances from all points to the cell under the mouse cursor. We show how this approach not only highlights distortions but also reveals otherwise hidden characteristics of the data, and how Sleepwalk's comparative modes help integrate multisample data and understand differences between embedding and preprocessing methods. Sleepwalk is a versatile and intuitive tool that unlocks the full power of dimension reduction and will be of value not only in single-cell RNA-seq but also in any other area with matrix-shaped big data.


Asunto(s)
RNA-Seq/métodos , Programas Informáticos , Animales , Cerebelo/embriología , Cerebelo/metabolismo , Expresión Génica , Ratones , Análisis de la Célula Individual
8.
Development ; 147(21)2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32376680

RESUMEN

Sonic hedgehog (Shh) signaling is essential for proliferation of cerebellar granule neuron progenitors (CGNPs) and its mis-regulation is linked to various disorders, including the cerebellar cancer medulloblastoma (MB). We recently identified RNF220, a ubiquitin E3 ligase promoting K63-linked polyubiquitylation and nuclear exportation of Gli transcription factors, as an Shh/Gli regulator involved in ventral neural patterning. Here, we report that RNF220 is required for the proliferation of CGNPs and Daoy cells (an Shh-grouped MB cell line), working as a positive regulator of Shh signaling. Mechanistic investigation demonstrated that RNF220 promotes Shh target gene expression by targeting the PRC2 component EED, and alters levels of epigenetic modification marks on Shh target promoters. We provided evidence that RNF220+/-; Ptch1+/- mice showed lower spontaneous MB occurrence compared with Ptch1+/- mice. Furthermore, in human clinical MB samples, RNF220 expression correlated well with that of GAB1, an Shh-group MB marker. Our findings provide new insights into the epigenetic regulation of Shh signaling and identify RNF220 as a potential new diagnostic marker and therapeutic target for Shh-group MB.


Asunto(s)
Cerebelo/embriología , Progresión de la Enfermedad , Epigénesis Genética , Proteínas Hedgehog/genética , Meduloblastoma/genética , Meduloblastoma/patología , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Cerebelo/patología , Gránulos Citoplasmáticos/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Lisina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
9.
PLoS Genet ; 16(4): e1008630, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32298260

RESUMEN

The cerebellum is a pivotal centre for the integration and processing of motor and sensory information. Its extended development into the postnatal period makes this structure vulnerable to a variety of pathologies, including neoplasia. These properties have prompted intensive investigations that reveal not only developmental mechanisms in common with other regions of the neuraxis but also unique strategies to generate neuronal diversity. How the phenotypically distinct cell types of the cerebellum emerge rests on understanding how gene expression differences arise in a spatially and temporally coordinated manner from initially homogeneous cell populations. Increasingly sophisticated fate mapping approaches, culminating in genetic-induced fate mapping, have furthered the understanding of lineage relationships between early- versus later-born cells. Tracing the developmental histories of cells in this way coupled with analysis of gene expression patterns has provided insight into the developmental genetic programmes that instruct cellular heterogeneity. A limitation to date has been the bulk analysis of cells, which blurs lineage relationships and obscures gene expression differences between cells that underpin the cellular taxonomy of the cerebellum. This review emphasises recent discoveries, focusing mainly on single-cell sequencing in mouse and parallel human studies that elucidate neural progenitor developmental trajectories with unprecedented resolution. Complementary functional studies of neural repair after cerebellar injury are challenging assumptions about the stability of postnatal cellular identities. The result is a wealth of new information about the developmental mechanisms that generate cerebellar neural diversity, with implications for human evolution.


Asunto(s)
Cerebelo/citología , Cerebelo/embriología , Morfogénesis , Análisis de la Célula Individual , Animales , Diferenciación Celular , Linaje de la Célula , Cerebelo/crecimiento & desarrollo , Humanos , Neuronas/clasificación , Neuronas/citología
10.
Genes Dev ; 28(21): 2407-20, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25367036

RESUMEN

Cerebellar development is regulated by a coordinated spatiotemporal interplay between granule neuron progenitors (GNPs), Purkinje neurons, and glia. Abnormal development can trigger motor deficits, and more recent data indicate important roles in aspects of memory, behavior, and autism spectrum disorders (ASDs). Germline mutation in the NF1 tumor suppressor gene underlies Neurofibromatosis type 1, a complex disease that enhances susceptibility to certain cancers and neurological disorders, including intellectual deficits and ASD. The NF1 gene encodes for neurofibromin, a RAS GTPase-activating protein, and thus negatively regulates the RAS signaling pathway. Here, using mouse models to direct conditional NF1 ablation in either embryonic cerebellar progenitors or neonatal GNPs, we show that neurofibromin is required for appropriate development of cerebellar folia layering and structure. Remarkably, neonatal administration of inhibitors of the ERK pathway reversed the morphological defects. Thus, our findings establish a critical cell-autonomous role for the NF1-RAS-ERK pathway in the appropriate regulation of cerebellar development and provide a basis for using neonatal ERK inhibitor-based therapies to treat NF1-induced cerebellar disorders.


Asunto(s)
Cerebelo/embriología , Sistema de Señalización de MAP Quinasas/fisiología , Neurofibromina 1/metabolismo , Células Madre/citología , Células Madre/fisiología , Proteínas ras/fisiología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Eliminación de Gen , Ratones , Neurofibromina 1/genética , Neuronas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Células Madre/efectos de los fármacos
11.
Dev Biol ; 457(1): 150-162, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31586559

RESUMEN

Yap/Taz are well-established downstream effectors of the Hippo pathway, known to regulate organ size by directing proliferation and apoptosis. Although the functions of Yap/Taz have been extensively studied, little is known about their role in brain development. Here, through genetic ablation, we show that Yap/Taz are required for cerebellar morphogenesis. Yap/Taz deletion in neural progenitors causes defects in secondary fissure formation, leading to abnormal folia development. Although they seemed very likely to serve an important function in the development of cerebellar granule cell precursors, Yap/Taz are dispensable for their proliferation. Furthermore, Yap/Taz loss does not rescue the medulloblastoma phenotype caused by constitutively active Smoothened. Importantly, Yap/Taz are highly expressed in radial glia and play a crucial role in establishing the radial scaffold and cellular polarity of neural progenitors during embryogenesis. We found that Yap/Taz are necessary to establish and maintain junctional integrity of cerebellar neuroepithelium as prominent junction proteins are not maintained at the apical junction in the absence of Yap/Taz. Our study identifies a novel function of Yap/Taz in cerebellar foliation and finds that they are required to establish the radial glia scaffold and junctional stability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cerebelo/embriología , Organogénesis , Transactivadores/metabolismo , Animales , Proliferación Celular , Cerebelo/citología , Cerebelo/metabolismo , Células Ependimogliales , Ratones , Tamaño de los Órganos , Células Madre/metabolismo , Proteínas Señalizadoras YAP
12.
Neurobiol Dis ; 150: 105236, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33383187

RESUMEN

Development of the forebrain critically depends on the Sonic Hedgehog (Shh) signaling pathway, as illustrated in humans by the frequent perturbation of this pathway in holoprosencephaly, a condition defined as a defect in the formation of midline structures of the forebrain and face. The Shh pathway requires functional primary cilia, microtubule-based organelles present on virtually every cell and acting as cellular antennae to receive and transduce diverse chemical, mechanical or light signals. The dysfunction of cilia in humans leads to inherited diseases called ciliopathies, which often affect many organs and show diverse manifestations including forebrain malformations for the most severe forms. The purpose of this review is to provide the reader with a framework to understand the developmental origin of the forebrain defects observed in severe ciliopathies with respect to perturbations of the Shh pathway. We propose that many of these defects can be interpreted as an imbalance in the ratio of activator to repressor forms of the Gli transcription factors, which are effectors of the Shh pathway. We also discuss the complexity of ciliopathies and their relationships with forebrain disorders such as holoprosencephaly or malformations of cortical development, and emphasize the need for a closer examination of forebrain defects in ciliopathies, not only through the lens of animal models but also taking advantage of the increasing potential of the research on human tissues and organoids.


Asunto(s)
Encéfalo/anomalías , Cilios/genética , Ciliopatías/embriología , Anomalías Craneofaciales/embriología , Proteínas Hedgehog/fisiología , Prosencéfalo/embriología , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Encéfalo/embriología , Cerebelo/anomalías , Cerebelo/embriología , Trastornos de la Motilidad Ciliar/embriología , Trastornos de la Motilidad Ciliar/genética , Ciliopatías/genética , Anomalías Craneofaciales/genética , Discapacidades del Desarrollo/genética , Encefalocele/embriología , Encefalocele/genética , Anomalías del Ojo/embriología , Anomalías del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Holoprosencefalia/embriología , Holoprosencefalia/genética , Humanos , Enfermedades Renales Quísticas/embriología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Poliquísticas/embriología , Enfermedades Renales Poliquísticas/genética , Retina/anomalías , Retina/embriología , Retinitis Pigmentosa/embriología , Retinitis Pigmentosa/genética , Transducción de Señal , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética , Proteína Gli3 con Dedos de Zinc/genética
13.
Biochem Biophys Res Commun ; 534: 59-66, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33310189

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder caused by genetic and environmental factors. Among the environmental factors, maternal infection is known as one of the principal risk factors for ASD. On the other hand, postmortem studies suggested the relationship of oxidative stress with ASD etiology. However, the role of oxidative stress in the development of ASD remains unclear. Here, we report the involvement of NOX1/NADPH oxidase, an enzyme generating reactive oxygen species (ROS), in behavioral and anatomical abnormalities in a maternal immune activation (MIA) model. In the MIA model of gestational polyinosinic-polycytidylic acid (poly(I:C)) exposure, increased serum levels of IL-6 were observed in both wild-type (WT) and Nox1-deficient mice (Nox1KO). Following the comparable induction of MIA in the two genotypes, impairment of social preference and defects in motor coordination were observed in WT offspring but not in offspring deficient in Nox1. MIA up-regulated NOX1 mRNA in the cerebral cortex and cerebellum of the fetus but not in the adult offspring. Although the development of cortical neurons was unaffected by MIA in either genotype, the dropout of Purkinje cells in lobule VII of MIA-affected offspring was significantly ameliorated in Nox1KO. Taken together, these results suggested that NOX1/NADPH oxidase plays an essential role in some behavioral phenotypes observed in ASD, possibly by promoting the loss of Purkinje cells in the cerebellum.


Asunto(s)
Trastorno del Espectro Autista/etiología , Conducta Animal/fisiología , NADPH Oxidasa 1/genética , Células de Purkinje/patología , Animales , Trastorno del Espectro Autista/inmunología , Cerebelo/embriología , Corteza Cerebral/embriología , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 1/metabolismo , Poli I-C/inmunología , Poli I-C/farmacología , Embarazo
14.
Development ; 145(2)2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343636

RESUMEN

During development, precerebellar neurons migrate tangentially from the dorsal hindbrain to the floor plate. Their axons cross it but their cell bodies stop their ventral migration upon reaching the midline. It has previously been shown that Slit chemorepellents and their receptors, Robo1 and Robo2, might control the migration of precerebellar neurons in a repulsive manner. Here, we have used a conditional knockout strategy in mice to test this hypothesis. We show that the targeted inactivation of the expression of Robo1 and Robo2 receptors in precerebellar neurons does not perturb their migration and that they still stop at the midline. The selective ablation of the expression of all three Slit proteins in floor-plate cells has no effect on pontine neurons and only induces the migration of a small subset of inferior olivary neurons across the floor plate. Likewise, we show that the expression of Slit proteins in the facial nucleus is dispensable for pontine neuron migration. Together, these results show that Robo1 and Robo2 receptors act non-cell autonomously in migrating precerebellar neurons and that floor-plate signals, other than Slit proteins, must exist to prevent midline crossing.


Asunto(s)
Movimiento Celular/fisiología , Cerebelo/embriología , Glicoproteínas/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Receptores Inmunológicos/fisiología , Animales , Cerebelo/citología , Femenino , Glicoproteínas/deficiencia , Glicoproteínas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neurogénesis/fisiología , Embarazo , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Transducción de Señal , Proteínas Roundabout
15.
Acta Neuropathol ; 142(4): 761-776, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34347142

RESUMEN

Dandy-Walker malformation (DWM) and Cerebellar vermis hypoplasia (CVH) are commonly recognized human cerebellar malformations diagnosed following ultrasound and antenatal or postnatal MRI. Specific radiological criteria are used to distinguish them, yet little is known about their differential developmental disease mechanisms. We acquired prenatal cases diagnosed as DWM and CVH and studied cerebellar morphobiometry followed by histological and immunohistochemical analyses. This was supplemented by laser capture microdissection and RNA-sequencing of the cerebellar rhombic lip, a transient progenitor zone, to assess the altered transcriptome of DWM vs control samples. Our radiological findings confirm that the cases studied fall within the accepted biometric range of DWM. Our histopathological analysis points to reduced foliation and inferior vermian hypoplasia as common features in all examined DWM cases. We also find that the rhombic lip, a dorsal stem cell zone that drives the growth and maintenance of the posterior vermis is specifically disrupted in DWM, with reduced proliferation and self-renewal of the progenitor pool, and altered vasculature, all confirmed by transcriptomics analysis. We propose a unified model for the developmental pathogenesis of DWM. We hypothesize that rhombic lip development is disrupted through either aberrant vascularization and/or direct insult which causes reduced proliferation and failed expansion of the rhombic lip progenitor pool leading to disproportionate hypoplasia and dysplasia of the inferior vermis. Timing of insult to the developing rhombic lip (before or after 14 PCW) dictates the extent of hypoplasia and distinguishes DWM from CVH.


Asunto(s)
Cerebelo/anomalías , Síndrome de Dandy-Walker/embriología , Síndrome de Dandy-Walker/patología , Desarrollo Fetal/fisiología , Feto/patología , Malformaciones del Sistema Nervioso/embriología , Malformaciones del Sistema Nervioso/patología , Estudios de Casos y Controles , Cerebelo/embriología , Cerebelo/patología , Discapacidades del Desarrollo/patología , Humanos , Recién Nacido
16.
PLoS Biol ; 16(9): e2005513, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30260948

RESUMEN

The morphological, molecular, and functional heterogeneity of astrocytes is under intense scrutiny, but how this diversity is ontogenetically achieved remains largely unknown. Here, by quantitative in vivo clonal analyses and proliferation studies, we demonstrate that the major cerebellar astrocyte types emerge according to an unprecedented and remarkably orderly developmental program comprising (i) a time-dependent decline in both clone size and progenitor multipotency, associated with clone allocation first to the hemispheres and then to the vermis(ii) distinctive clonal relationships among astrocyte types, revealing diverse lineage potentials of embryonic and postnatal progenitors; and (iii) stereotyped clone architectures and recurrent modularities that correlate to layer-specific dynamics of postnatal proliferation/differentiation. In silico simulations indicate that the sole presence of a unique multipotent progenitor at the source of the whole astrogliogenic program is unlikely and rather suggest the involvement of additional committed components.


Asunto(s)
Astrocitos/citología , Cerebelo/citología , Animales , Animales Recién Nacidos , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Tamaño de la Célula , Cerebelo/embriología , Células Clonales , Simulación por Computador , Femenino , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , Sustancia Blanca/citología
17.
PLoS Biol ; 16(6): e2004880, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29879109

RESUMEN

N6-methyladenosine (m6A) RNA methylation is the most abundant modification on mRNAs and plays important roles in various biological processes. The formation of m6A is catalyzed by a methyltransferase complex including methyltransferase-like 3 (METTL3) as a key factor. However, the in vivo functions of METTL3 and m6A modification in mammalian development remain unclear. Here, we show that specific inactivation of Mettl3 in mouse nervous system causes severe developmental defects in the brain. Mettl3 conditional knockout (cKO) mice manifest cerebellar hypoplasia caused by drastically enhanced apoptosis of newborn cerebellar granule cells (CGCs) in the external granular layer (EGL). METTL3 depletion-induced loss of m6A modification causes extended RNA half-lives and aberrant splicing events, consequently leading to dysregulation of transcriptome-wide gene expression and premature CGC death. Our findings reveal a critical role of METTL3-mediated m6A in regulating the development of mammalian cerebellum.


Asunto(s)
Adenosina/análogos & derivados , Cerebelo/embriología , Metiltransferasas/metabolismo , ARN Mensajero/genética , Adenosina/metabolismo , Empalme Alternativo/genética , Animales , Apoptosis/genética , Células Cultivadas , Cerebelo/anomalías , Cerebelo/patología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Regulación de la Expresión Génica/genética , Metilación , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Estabilidad del ARN/genética , ARN Mensajero/metabolismo
18.
Ultrasound Obstet Gynecol ; 57(4): 614-623, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32196791

RESUMEN

OBJECTIVE: To construct international ultrasound-based standards for fetal cerebellar growth and Sylvian fissure maturation. METHODS: Healthy, well nourished pregnant women, enrolled at < 14 weeks' gestation in the Fetal Growth Longitudinal Study (FGLS) of INTERGROWTH-21st , an international multicenter, population-based project, underwent serial three-dimensional (3D) fetal ultrasound scans every 5 ± 1 weeks until delivery in study sites located in Brazil, India, Italy, Kenya and the UK. In the present analysis, only those fetuses that underwent developmental assessment at 2 years of age were included. We measured the transcerebellar diameter and assessed Sylvian fissure maturation using two-dimensional ultrasound images extracted from available 3D fetal head volumes. The appropriateness of pooling data from the five sites was assessed using variance component analysis and standardized site differences. For each Sylvian fissure maturation score (left or right side), mean gestational age and 95% CI were calculated. Transcerebellar diameter was modeled using fractional polynomial regression, and goodness of fit was assessed. RESULTS: Of those children in the original FGLS cohort who had developmental assessment at 2 years of age, 1130 also had an available 3D ultrasound fetal head volume. The sociodemographic characteristics and pregnancy/perinatal outcomes of the study sample confirmed the health and low-risk status of the population studied. In addition, the fetuses had low morbidity and adequate growth and development at 2 years of age. In total, 3016 and 2359 individual volumes were available for transcerebellar-diameter and Sylvian-fissure analysis, respectively. Variance component analysis and standardized site differences showed that the five study populations were sufficiently similar on the basis of predefined criteria for the data to be pooled to produce international standards. A second-degree fractional polynomial provided the best fit for modeling transcerebellar diameter; we then estimated gestational-age-specific 3rd , 50th and 97th smoothed centiles. Goodness-of-fit analysis comparing empirical centiles with smoothed centile curves showed good agreement. The Sylvian fissure increased in maturation with advancing gestation, with complete overlap of the mean gestational age and 95% CIs between the sexes for each development score. No differences in Sylvian fissure maturation between the right and left hemispheres were observed. CONCLUSION: We present, for the first time, international standards for fetal cerebellar growth and Sylvian fissure maturation throughout pregnancy based on a healthy fetal population that exhibited adequate growth and development at 2 years of age. © 2020 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.


Asunto(s)
Cerebelo/embriología , Acueducto del Mesencéfalo/embriología , Desarrollo Fetal , Gráficos de Crecimiento , Ultrasonografía Prenatal , Adulto , Brasil , Cerebelo/crecimiento & desarrollo , Acueducto del Mesencéfalo/crecimiento & desarrollo , Preescolar , Femenino , Edad Gestacional , Humanos , India , Lactante , Recién Nacido , Italia , Kenia , Estudios Longitudinales , Masculino , Embarazo , Resultado del Embarazo , Estándares de Referencia , Reino Unido
19.
Ultrasound Obstet Gynecol ; 58(6): 864-874, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33942916

RESUMEN

OBJECTIVES: To describe the prenatal neuroimaging spectrum of rhombencephalosynapsis (RES) and criteria for its classification according to the severity of vermian anomaly. METHODS: In this multicenter retrospective study of fetuses with RES between 2002 and 2020, the medical records and brain ultrasound and magnetic resonance images were evaluated comprehensively to determine the severity of the vermian anomaly and the presence of associated brain findings. RES was classified, according to the pattern of vermian agenesis and the extent of the fusion of the hemispheres, as complete RES (complete absence of the vermis) or partial RES (further classified according to the part of the vermis that was missing and, consequently, the region of hemispheric fusion, as anterior, posterior, severe or mixed RES). Findings were compared between cases with complete and those with partial RES. RESULTS: Included in the study were 62 fetuses with a gestational age ranging between 12 and 37 weeks. Most had complete absence of the vermis (complete RES, 77.4% of cases), a 'round-shaped' cerebellum on axial views (72.6%) and a transverse cerebellar diameter (TCD) < 3rd centile (87.1%). Among the 22.6% of cases with partial RES, 6.5% were classified as severe partial, 6.5% as partial anterior, 8.1% as partial mixed and 1.6% as partial posterior. Half of these cases presented with normal or nearly normal cerebellar morphology and 28.5% had a TCD within the normal limits. Infratentorially, the fourth ventricle was abnormal in 88.7% of cases overall, and anomalies of the midbrain and pons were frequent (93.5% and 77.4%, respectively). Ventriculomegaly was observed in 80.6% of all cases, being more severe in cases with complete RES than in those with partial RES, with high rates of parenchymal and septal disruption. CONCLUSIONS: This study provides prenatal neuroimaging criteria for the diagnosis and classification of RES, and identification of related features, using ultrasound and magnetic resonance imaging. According to our findings, a diagnosis of RES should be considered in fetuses with a small TCD (severe cerebellar hypoplasia) and/or a round-shaped cerebellum on axial views, during the second or third trimester, especially when associated with ventriculomegaly. Partial RES is more common than previously thought, but presents an extreme diagnostic challenge, especially in cases with normal or nearly-normal cerebellar morphobiometric features. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.


Asunto(s)
Anomalías Múltiples/diagnóstico por imagen , Vermis Cerebeloso/anomalías , Cerebelo/anomalías , Anomalías del Ojo/diagnóstico por imagen , Enfermedades Renales Quísticas/diagnóstico por imagen , Malformaciones del Sistema Nervioso/diagnóstico por imagen , Neuroimagen , Diagnóstico Prenatal/métodos , Retina/anomalías , Rombencéfalo/anomalías , Anomalías Múltiples/embriología , Adulto , Vermis Cerebeloso/diagnóstico por imagen , Vermis Cerebeloso/embriología , Cerebelo/diagnóstico por imagen , Cerebelo/embriología , Anomalías del Ojo/embriología , Femenino , Edad Gestacional , Humanos , Enfermedades Renales Quísticas/embriología , Imagen por Resonancia Magnética , Imagen Multimodal , Malformaciones del Sistema Nervioso/embriología , Embarazo , Retina/diagnóstico por imagen , Retina/embriología , Estudios Retrospectivos , Rombencéfalo/diagnóstico por imagen , Rombencéfalo/embriología , Índice de Severidad de la Enfermedad , Ultrasonografía Prenatal
20.
Dev Biol ; 455(2): 393-408, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31323192

RESUMEN

The cerebellum and the cerebellum-like structure in the mesencephalic tectum in zebrafish contain multiple cell types, including principal cells (i.e., Purkinje cells and type I neurons) and granule cells, that form neural circuits in which the principal cells receive and integrate inputs from granule cells and other neurons. It is largely unknown how these cells are positioned and how neural circuits form. While Reelin signaling is known to play an important role in cell positioning in the mammalian brain, its role in the formation of other vertebrate brains remains elusive. Here we found that zebrafish with mutations in Reelin or in the Reelin-signaling molecules Vldlr or Dab1a exhibited ectopic Purkinje cells, eurydendroid cells (projection neurons), and Bergmann glial cells in the cerebellum, and ectopic type I neurons in the tectum. The ectopic Purkinje cells and type I neurons received aberrant afferent fibers in these mutants. In wild-type zebrafish, reelin transcripts were detected in the internal granule cell layer, while Reelin protein was localized to the superficial layer of the cerebellum and the tectum. Laser ablation of the granule cell axons perturbed the localization of Reelin, and the mutation of both kif5aa and kif5ba, which encode major kinesin I components in the granule cells, disrupted the elongation of granule cell axons and the Reelin distribution. Our findings suggest that in zebrafish, (1) Reelin is transported from the granule cell soma to the superficial layer by axonal transport; (2) Reelin controls the migration of neurons and glial cells from the ventricular zone; and (3) Purkinje cells and type I neurons attract afferent axons during the formation of the cerebellum and the cerebellum-like structure.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Cerebelo/embriología , Proteínas de la Matriz Extracelular/fisiología , Proteínas del Tejido Nervioso/fisiología , Serina Endopeptidasas/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Sistemas CRISPR-Cas , Moléculas de Adhesión Celular Neuronal/genética , Movimiento Celular , Cerebelo/citología , Proteínas de la Matriz Extracelular/genética , Cinesinas/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Células de Purkinje/citología , Proteína Reelina , Serina Endopeptidasas/genética , Transducción de Señal , Pez Cebra/anatomía & histología , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA