Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.802
Filtrar
1.
Sci Adv ; 10(17): eade1650, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669326

RESUMEN

While the kinesin-2 motors KIF3A and KIF3B have essential roles in ciliogenesis and Hedgehog (HH) signal transduction, potential role(s) for another kinesin-2 motor, KIF17, in HH signaling have yet to be explored. Here, we investigated the contribution of KIF17 to HH-dependent cerebellar development, where Kif17 is expressed in both HH-producing Purkinje cells and HH-responding cerebellar granule neuron progenitors (CGNPs). Germline Kif17 deletion in mice results in cerebellar hypoplasia due to reduced CGNP proliferation, a consequence of decreased HH pathway activity mediated through decreased Sonic HH (SHH) protein. Notably, Purkinje cell-specific Kif17 deletion partially phenocopies Kif17 germline mutants. Unexpectedly, CGNP-specific Kif17 deletion results in the opposite phenotype-increased CGNP proliferation and HH target gene expression due to altered GLI transcription factor processing. Together, these data identify KIF17 as a key regulator of HH-dependent cerebellar development, with dual and opposing roles in HH-producing Purkinje cells and HH-responding CGNPs.


Asunto(s)
Cerebelo , Cerebelo/anomalías , Proteínas Hedgehog , Cinesinas , Malformaciones del Sistema Nervioso , Células de Purkinje , Animales , Cinesinas/metabolismo , Cinesinas/genética , Cerebelo/metabolismo , Cerebelo/crecimiento & desarrollo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Ratones , Células de Purkinje/metabolismo , Transducción de Señal , Proliferación Celular , Ratones Noqueados , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Discapacidades del Desarrollo
2.
J Neurosci ; 44(17)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38527808

RESUMEN

Throughout life, the cerebellum plays a central role in the coordination and optimization of movements, using cellular plasticity to adapt a range of behaviors. Whether these plasticity processes establish a fixed setpoint during development, or continuously adjust behaviors throughout life, is currently unclear. Here, by spatiotemporally manipulating the activity of protein phosphatase 2B (PP2B), an enzyme critical for cerebellar plasticity in male and female mice, we examined the consequences of disrupted plasticity on the performance and adaptation of the vestibulo-ocular reflex (VOR). We find that, in contrast to Purkinje cell (PC)-specific deletion starting early postnatally, acute pharmacological as well as adult-onset genetic deletion of PP2B affects all forms of VOR adaptation but not the level of VOR itself. Next, we show that PC-specific genetic deletion of PP2B in juvenile mice leads to a progressive loss of the protein PP2B and a concurrent change in the VOR, in addition to the loss of adaptive abilities. Finally, re-expressing PP2B in adult mice that lack PP2B expression from early development rescues VOR adaptation but does not affect the performance of the reflex. Together, our results indicate that chronic or acute, genetic, or pharmacological block of PP2B disrupts the adaptation of the VOR. In contrast, only the absence of plasticity during cerebellar development affects the setpoint of VOR, an effect that cannot be corrected after maturation of the cerebellum. These findings suggest that PP2B-dependent cerebellar plasticity is required during a specific period to achieve the correct setpoint of the VOR.


Asunto(s)
Cerebelo , Plasticidad Neuronal , Reflejo Vestibuloocular , Animales , Reflejo Vestibuloocular/fisiología , Plasticidad Neuronal/fisiología , Ratones , Cerebelo/crecimiento & desarrollo , Cerebelo/fisiología , Masculino , Femenino , Células de Purkinje/fisiología , Adaptación Fisiológica/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados
3.
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
4.
Science ; 381(6662): 1112-1119, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37676945

RESUMEN

The cerebellum contains most of the neurons in the human brain and exhibits distinctive modes of development and aging. In this work, by developing our single-cell three-dimensional (3D) genome assay-diploid chromosome conformation capture, or Dip-C-into population-scale (Pop-C) and virus-enriched (vDip-C) modes, we resolved the first 3D genome structures of single cerebellar cells, created life-spanning 3D genome atlases for both humans and mice, and jointly measured transcriptome and chromatin accessibility during development. We found that although the transcriptome and chromatin accessibility of cerebellar granule neurons mature in early postnatal life, 3D genome architecture gradually remodels throughout life, establishing ultra-long-range intrachromosomal contacts and specific interchromosomal contacts that are rarely seen in neurons. These results reveal unexpected evolutionarily conserved molecular processes that underlie distinctive features of neural development and aging across the mammalian life span.


Asunto(s)
Senescencia Celular , Cerebelo , Ensamble y Desensamble de Cromatina , Genoma , Neuronas , Animales , Humanos , Ratones , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Neuronas/metabolismo , Imagenología Tridimensional , Análisis de la Célula Individual , Atlas como Asunto
5.
J Cell Physiol ; 238(3): 533-548, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36649308

RESUMEN

Medulloblastoma (MB) is the most common malignant pediatric brain tumor. Current treatment modalities are not completely effective and can lead to severe neurological and cognitive adverse effects. In addition to urgently needing better treatment approaches, new diagnostic and prognostic biomarkers are required to improve the therapy outcomes of MB patients. The RNA-binding proteins, LIN28A and LIN28B, are known to regulate invasive phenotypes in many different cancer types. However, the expression and function of these proteins in MB had not been studied to date. This study identified the expression of LIN28A and LIN28B in MB patient samples and cell lines and assessed the effect of LIN28 inhibition on MB cell growth, metabolism and stemness. LIN28B expression was significantly upregulated in MB tissues compared to normal brain tissues. This upregulation, which was not observed in other brain tumors, was specific for the aggressive MB subgroups and correlated with patient survival and metastasis rates. Functionally, pharmacological inhibition of LIN28 activity concentration-dependently reduced LIN28B expression, as well as the growth of D283 MB cells. While LIN28 inhibition did not affect the levels of intracellular ATP, it reduced the expression of the stemness marker CD133 in D283 cells and the sphere formation of CHLA-01R cells. LIN28B, which is highly expressed in the human cerebellum during the first few months after birth, subsequently decreased with age. The results of this study highlight the potential of LIN28B as a diagnostic and prognostic marker for MB and open the possibility to utilize LIN28 as a pharmacological target to suppress MB cell growth and stemness.


Asunto(s)
Neoplasias Cerebelosas , Regulación Neoplásica de la Expresión Génica , Meduloblastoma , Niño , Humanos , Neoplasias Cerebelosas/diagnóstico , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Meduloblastoma/diagnóstico , Meduloblastoma/genética , Meduloblastoma/metabolismo , Meduloblastoma/patología , Línea Celular Tumoral , Adenosina Trifosfato/metabolismo , Recién Nacido , Lactante , Preescolar , Envejecimiento/metabolismo , Pronóstico
6.
Biol Open ; 11(8)2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35876806

RESUMEN

Formation of the mouse cerebellum is initiated in the embryo and continues for a few weeks after birth. Double-mutant mice lacking platelet-derived growth factor C (PDGF-C) and that are heterozygous for platelet-derived growth factor receptor alpha (Pdgfc-/-; PdgfraGFP/+) develop cerebellar hypoplasia and malformation with loss of cerebellar lobes in the posterior vermis. This phenotype is similar to those observed in Foxc1 mutant mice and in a human neuroimaging pattern called Dandy Walker malformation. Pdgfc-Pdgfra mutant mice also display ependymal denudation in the fourth ventricle and gene expression changes in cerebellar meninges, which coincide with the first visible signs of cerebellar malformation. Here, we show that PDGF-C/PDGFRα signalling is a critical component in the network of molecular and cellular interactions that take place between the developing meninges and neural tissues, and which are required to build a fully functioning cerebellum.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Malformaciones del Sistema Nervioso , Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal , Animales , Síndrome de Dandy-Walker/diagnóstico , Síndrome de Dandy-Walker/genética , Humanos , Linfocinas , Ratones , Malformaciones del Sistema Nervioso/genética
7.
Sci Rep ; 12(1): 2355, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35149757

RESUMEN

The relationship between gait speed and working memory is well-understood in older adults. However, it remains to be determined whether this relationship also exists in younger adults; and there is little known regarding the possible neural mechanism underlying the association between gait speed and working memory. The aims of this study are to determine if there is: (1) an association between gait speed and working memory performance; and (2) a mediating role of cerebellar subregion volume in the correlation between gait speed and working memory in healthy younger adults. 1054 younger adults (28.7 ± 3.6 years) from the Human Connectome Project were included in the analyses. A four-meter gait test was used to assess gait speed. The 2-back task was used to measure working memory performance [accuracy and response time (RT)]. T1-weighted structural MRI data (obtained using Siemens 3 T MRI scanner) was used to assess cerebellar subregion volumes. Linear regression and mediation analysis were used to examine the relationships between the variables after controlling for age, sex, and education. There was no association between gait speed and 2-back working memory performance in younger adults. Greater Crus I and whole cerebellar volumes were associated with better 2-back working memory accuracy. Greater VIIIa volume was associated with faster gait speed. Greater Crus 1 and VIIIa volumes were also associated with higher fluid cognition. The present study suggests that specific subregions of the cerebellar volumes are distinctively associated with gait speed and working memory performance in healthy younger adults.


Asunto(s)
Cerebelo/fisiología , Marcha , Memoria a Corto Plazo , Adulto , Cerebelo/diagnóstico por imagen , Cerebelo/crecimiento & desarrollo , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Tamaño de los Órganos , Tiempo de Reacción , Velocidad al Caminar , Adulto Joven
8.
Dev Med Child Neurol ; 64(2): 192-199, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34416027

RESUMEN

AIM: To assess the relationship between neonatal brain development and injury with early motor outcomes in infants with critical congenital heart disease (CCHD). METHOD: Neonatal brain magnetic resonance imaging was performed after open-heart surgery with cardiopulmonary bypass. Cortical grey matter (CGM), unmyelinated white matter, and cerebellar volumes, as well as white matter motor tract fractional anisotropy and mean diffusivity were assessed. White matter injury (WMI) and arterial ischaemic stroke (AIS) with corticospinal tract (CST) involvement were scored. Associations with motor outcomes at 3, 9, and 18 months were corrected for repeated cardiac surgery. RESULTS: Fifty-one infants (31 males, 20 females) were included prospectively. Median age at neonatal surgery and postoperative brain magnetic resonance imaging was 7 days (interquartile range [IQR] 5-11d) and 15 days (IQR 12-21d) respectively. Smaller CGM and cerebellar volumes were associated with lower fine motor scores at 9 months (CGM regression coefficient=0.51, 95% confidence interval [CI]=0.15-0.86; cerebellum regression coefficient=3.08, 95% CI=1.07-5.09) and 18 months (cerebellum regression coefficient=2.08, 95% CI=0.47-5.12). The fractional anisotropy and mean diffusivity of white matter motor tracts were not related with motor scores. WMI was related to lower gross motor scores at 9 months (mean difference -0.8SD, 95% CI=-1.5 to -0.2). AIS with CST involvement increased the risk of gross motor problems and muscle tone abnormalities. Cerebral palsy (n=3) was preceded by severe ischaemic brain injury. INTERPRETATION: Neonatal brain development and injury are associated with fewer favourable early motor outcomes in infants with CCHD.


Asunto(s)
Lesiones Encefálicas , Parálisis Cerebral , Desarrollo Infantil/fisiología , Discapacidades del Desarrollo , Cardiopatías Congénitas/cirugía , Accidente Cerebrovascular Isquémico , Destreza Motora/fisiología , Tractos Piramidales , Lesiones Encefálicas/diagnóstico por imagen , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Cerebelo/diagnóstico por imagen , Cerebelo/crecimiento & desarrollo , Cerebelo/patología , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/patología , Parálisis Cerebral/diagnóstico por imagen , Parálisis Cerebral/patología , Parálisis Cerebral/fisiopatología , Discapacidades del Desarrollo/diagnóstico por imagen , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/patología , Discapacidades del Desarrollo/fisiopatología , Femenino , Sustancia Gris/diagnóstico por imagen , Sustancia Gris/crecimiento & desarrollo , Sustancia Gris/patología , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/diagnóstico por imagen , Humanos , Lactante , Recién Nacido , Accidente Cerebrovascular Isquémico/diagnóstico por imagen , Accidente Cerebrovascular Isquémico/patología , Accidente Cerebrovascular Isquémico/fisiopatología , Imagen por Resonancia Magnética , Masculino , Estudios Prospectivos , Tractos Piramidales/diagnóstico por imagen , Tractos Piramidales/crecimiento & desarrollo , Tractos Piramidales/patología , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/crecimiento & desarrollo , Sustancia Blanca/patología
9.
Food Chem Toxicol ; 159: 112751, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34871666

RESUMEN

Recent studies showed a possible association between perfluorooctane sulfonate (PFOS) and developmental disabilities. We previously found the specific effects of PFOS exposure on learning and memory, however, its effect on the other developmental disabilities such as motor and social deficits remains unclear. We examined the effect of early lactational PFOS exposure on motor coordination, social activity, and anxiety in male mice. We orally administered a PFOS solution to dams from postnatal day 1-14. At 10 weeks old, we conducted a behavior test battery to evaluate motor performance, social activity, and anxiety, followed by electrophysiology and Western blot analysis. PFOS-exposed mice displayed impaired motor coordination. Whole-cell patch-clamp recordings from Purkinje cells revealed that the short-term and long-term plasticity at parallel fiber-Purkinje cell synapses are affected by PFOS exposure. Western blot analysis indicated that PFOS exposure increased syntaxin binding protein 1 (Munc18-1) and glutamate metabotropic receptor 1 (mGluR1) protein levels, which may be associated with the change in neurotransmitter release from parallel fibers and the level of long-term depression, respectively. The present study demonstrates that lactational PFOS exposure may have disrupted the pre- and postsynaptic plasticity at parallel fiber-Purkinje cell synapses, causing profound, long-lasting abnormal effects on the cerebellar function.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Cerebelo/efectos de los fármacos , Exposición Dietética , Fluorocarburos/toxicidad , Exposición Materna , Neurotoxinas/toxicidad , Animales , Ansiedad , Conducta Animal/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Cerebelo/fisiopatología , Femenino , Lactancia , Masculino , Ratones , Desempeño Psicomotor/efectos de los fármacos
10.
Elife ; 102021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34730085

RESUMEN

Synaptic transmission, connectivity, and dendritic morphology mature in parallel during brain development and are often disrupted in neurodevelopmental disorders. Yet how these changes influence the neuronal computations necessary for normal brain function are not well understood. To identify cellular mechanisms underlying the maturation of synaptic integration in interneurons, we combined patch-clamp recordings of excitatory inputs in mouse cerebellar stellate cells (SCs), three-dimensional reconstruction of SC morphology with excitatory synapse location, and biophysical modeling. We found that postnatal maturation of postsynaptic strength was homogeneously reduced along the somatodendritic axis, but dendritic integration was always sublinear. However, dendritic branching increased without changes in synapse density, leading to a substantial gain in distal inputs. Thus, changes in synapse distribution, rather than dendrite cable properties, are the dominant mechanism underlying the maturation of neuronal computation. These mechanisms favor the emergence of a spatially compartmentalized two-stage integration model promoting location-dependent integration within dendritic subunits.


Asunto(s)
Cerebelo/fisiología , Interneuronas/fisiología , Transmisión Sináptica/fisiología , Animales , Cerebelo/crecimiento & desarrollo , Femenino , Interneuronas/metabolismo , Masculino , Ratones
11.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34740966

RESUMEN

Cerebellar Purkinje neurons integrate information transmitted at excitatory synapses formed by granule cells. Although these synapses are considered essential sites for learning, most of them appear not to transmit any detectable electrical information and have been defined as silent. It has been proposed that silent synapses are required to maximize information storage capacity and ensure its reliability, and hence to optimize cerebellar operation. Such optimization is expected to occur once the cerebellar circuitry is in place, during its maturation and the natural and steady improvement of animal agility. We therefore investigated whether the proportion of silent synapses varies over this period, from the third to the sixth postnatal week in mice. Selective expression of a calcium indicator in granule cells enabled quantitative mapping of presynaptic activity, while postsynaptic responses were recorded by patch clamp in acute slices. Through this approach and the assessment of two anatomical features (the distance that separates adjacent planar Purkinje dendritic trees and the synapse density), we determined the average excitatory postsynaptic potential per synapse. Its value was four to eight times smaller than responses from paired recorded detectable connections, consistent with over 70% of synapses being silent. These figures remained remarkably stable across maturation stages. According to the proposed role for silent synapses, our results suggest that information storage capacity and reliability are optimized early during cerebellar maturation. Alternatively, silent synapses may have roles other than adjusting the information storage capacity and reliability.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Animales , Señalización del Calcio , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Células de Purkinje/fisiología , Sinapsis/fisiología
12.
Nat Commun ; 12(1): 5702, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34588434

RESUMEN

Regulation of chromatin plays fundamental roles in the development of the brain. Haploinsufficiency of the chromatin remodeling enzyme CHD7 causes CHARGE syndrome, a genetic disorder that affects the development of the cerebellum. However, how CHD7 controls chromatin states in the cerebellum remains incompletely understood. Using conditional knockout of CHD7 in granule cell precursors in the mouse cerebellum, we find that CHD7 robustly promotes chromatin accessibility, active histone modifications, and RNA polymerase recruitment at enhancers. In vivo profiling of genome architecture reveals that CHD7 concordantly regulates epigenomic modifications associated with enhancer activation and gene expression of topologically-interacting genes. Genome and gene ontology studies show that CHD7-regulated enhancers are associated with genes that control brain tissue morphogenesis. Accordingly, conditional knockout of CHD7 triggers a striking phenotype of cerebellar polymicrogyria, which we have also found in a case of CHARGE syndrome. Finally, we uncover a CHD7-dependent switch in the preferred orientation of granule cell precursor division in the developing cerebellum, providing a potential cellular basis for the cerebellar polymicrogyria phenotype upon loss of CHD7. Collectively, our findings define epigenomic regulation by CHD7 in granule cell precursors and identify abnormal cerebellar patterning upon CHD7 depletion, with potential implications for our understanding of CHARGE syndrome.


Asunto(s)
Síndrome CHARGE/genética , Cerebelo/crecimiento & desarrollo , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Polimicrogiria/genética , Animales , Síndrome CHARGE/patología , División Celular/genética , Cerebelo/patología , Ensamble y Desensamble de Cromatina , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos , Epigénesis Genética , Código de Histonas , Humanos , Lactante , Ratones , Ratones Noqueados , Mutación , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Polimicrogiria/patología , RNA-Seq
13.
Cell Rep ; 36(7): 109522, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34407407

RESUMEN

Neuro-vascular communication is essential to synchronize central nervous system development. Here, we identify angiopoietin/Tie2 as a neuro-vascular signaling axis involved in regulating dendritic morphogenesis of Purkinje cells (PCs). We show that in the developing cerebellum Tie2 expression is not restricted to blood vessels, but it is also present in PCs. Its ligands angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are expressed in neural cells and endothelial cells (ECs), respectively. PC-specific deletion of Tie2 results in reduced dendritic arborization, which is recapitulated in neural-specific Ang1-knockout and Ang2 full-knockout mice. Mechanistically, RNA sequencing reveals that Tie2-deficient PCs present alterations in gene expression of multiple genes involved in cytoskeleton organization, dendritic formation, growth, and branching. Functionally, mice with deletion of Tie2 in PCs present alterations in PC network functionality. Altogether, our data propose Ang/Tie2 signaling as a mediator of intercellular communication between neural cells, ECs, and PCs, required for proper PC dendritic morphogenesis and function.


Asunto(s)
Angiopoyetina 2/metabolismo , Dendritas/metabolismo , Morfogénesis , Células de Purkinje/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal , Angiopoyetina 1/metabolismo , Animales , Cerebelo/irrigación sanguínea , Cerebelo/crecimiento & desarrollo , Eliminación de Gen , Regulación de la Expresión Génica , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Especificidad de Órganos
14.
Science ; 373(6558)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34446581

RESUMEN

Organ development is orchestrated by cell- and time-specific gene regulatory networks. In this study, we investigated the regulatory basis of mouse cerebellum development from early neurogenesis to adulthood. By acquiring snATAC-seq (single-nucleus assay for transposase accessible chromatin using sequencing) profiles for ~90,000 cells spanning 11 stages, we mapped cerebellar cell types and identified candidate cis-regulatory elements (CREs). We detected extensive spatiotemporal heterogeneity among progenitor cells and a gradual divergence in the regulatory programs of cerebellar neurons during differentiation. Comparisons to vertebrate genomes and snATAC-seq profiles for ∼20,000 cerebellar cells from the marsupial opossum revealed a shared decrease in CRE conservation during development and differentiation as well as differences in constraint between cell types. Our work delineates the developmental and evolutionary dynamics of gene regulation in cerebellar cells and provides insights into mammalian organ development.


Asunto(s)
Evolución Biológica , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Neuronas/fisiología , Elementos Reguladores de la Transcripción , Animales , Cerebelo/embriología , Cromatina/genética , Cromatina/metabolismo , ADN Intergénico , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis , Zarigüeyas/genética
15.
Sci Rep ; 11(1): 17108, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429472

RESUMEN

Exercise has emerged as an intervention that may mitigate age-related resting state functional connectivity and sensorimotor decline. Here, 42 healthy older adults rested or completed 3 sets of high-intensity interval exercise for a total of 23 min, then immediately practiced an implicit motor task with their non-dominant hand across five separate sessions. Participants completed resting state functional MRI before the first and after the fifth day of practice; they also returned 24-h and 35-days later to assess short- and long-term retention. Independent component analysis of resting state functional MRI revealed increased connectivity in the frontoparietal, the dorsal attentional, and cerebellar networks in the exercise group relative to the rest group. Seed-based analysis showed strengthened connectivity between the limbic system and right cerebellum, and between the right cerebellum and bilateral middle temporal gyri in the exercise group. There was no motor learning advantage for the exercise group. Our data suggest that exercise paired with an implicit motor learning task in older adults can augment resting state functional connectivity without enhancing behaviour beyond that stimulated by skilled motor practice.


Asunto(s)
Envejecimiento/fisiología , Conectoma , Entrenamiento de Intervalos de Alta Intensidad/métodos , Aprendizaje , Destreza Motora , Anciano , Cerebelo/crecimiento & desarrollo , Cerebelo/fisiología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad
16.
Nat Neurosci ; 24(10): 1392-1401, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34400844

RESUMEN

Compromised placental function or premature loss has been linked to diverse neurodevelopmental disorders. Here we show that placenta allopregnanolone (ALLO), a progesterone-derived GABA-A receptor (GABAAR) modulator, reduction alters neurodevelopment in a sex-linked manner. A new conditional mouse model, in which the gene encoding ALLO's synthetic enzyme (akr1c14) is specifically deleted in trophoblasts, directly demonstrated that placental ALLO insufficiency led to cerebellar white matter abnormalities that correlated with autistic-like behavior only in male offspring. A single injection of ALLO or muscimol, a GABAAR agonist, during late gestation abolished these alterations. Comparison of male and female human preterm infant cerebellum also showed sex-linked myelination marker alteration, suggesting similarities between mouse placental ALLO insufficiency and human preterm brain development. This study reveals a new role for a placental hormone in shaping brain regions and behaviors in a sex-linked manner. Placental hormone replacement might offer novel therapeutic opportunities to prevent later neurobehavioral disorders.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Glándulas Endocrinas/fisiología , Placenta/fisiología , Pregnanolona/deficiencia , Pregnanolona/fisiología , Conducta Social , Aldehído Reductasa/genética , Animales , Trastorno del Espectro Autista/etiología , Cerebelo/fisiología , Femenino , Agonistas del GABA/farmacología , Moduladores del GABA , Eliminación de Gen , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Muscimol/farmacología , Embarazo , Receptores de GABA-A/fisiología , Caracteres Sexuales , Trofoblastos/metabolismo , Sustancia Blanca/patología
17.
Alcohol Clin Exp Res ; 45(7): 1408-1423, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34060105

RESUMEN

BACKGROUND: The developing hippocampus and cerebellum, unique among brain regions, exhibit a secondary surge in neurogenesis during the third trimester of pregnancy. Ethanol (EtOH) exposure during this period is results in a loss of tissue volume and associated neurobehavioral deficits. However, mechanisms that link EtOH exposure to teratology in these regions are not well understood. We therefore analyzed transcriptomic adaptations to EtOH exposure to identify mechanistic linkages. METHODS: Hippocampi and cerebella were microdissected at postnatal day (P)10, from control C57BL/6J mouse pups, and pups treated with 4 g/kg of EtOH from P4 to P9. RNA was isolated and RNA-seq analysis was performed. We compared gene expression in EtOH- and vehicle-treated control neonates and performed biological pathway-overrepresentation analysis. RESULTS: While EtOH exposure resulted in the general induction of genes associated with the S-phase of mitosis in both cerebellum and hippocampus, overall there was little overlap in differentially regulated genes and associated biological pathways between these regions. In cerebellum, EtOH additionally induced gene expression associated with the G2/M-phases of the cell cycle and sonic hedgehog signaling, while in hippocampus, EtOH-induced the pathways for ribosome biogenesis and protein translation. Moreover, EtOH inhibited the transcriptomic identities associated with inhibitory interneuron subpopulations in the hippocampus, while in the cerebellum there was a more pronounced inhibition of transcripts across multiple oligodendrocyte maturation stages. CONCLUSIONS: These data indicate that during the delayed neurogenic period, EtOH may stimulate the cell cycle, but it otherwise results in widely divergent molecular effects in the hippocampus and cerebellum. Moreover, these data provide evidence for region- and cell-type-specific vulnerability, which may contribute to the pathogenic effects of developmental EtOH exposure.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Cerebelo/crecimiento & desarrollo , Etanol/efectos adversos , Hipocampo/crecimiento & desarrollo , Neurogénesis/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Animales , Apoptosis/genética , Ciclo Celular/genética , Cerebelo/metabolismo , Etanol/administración & dosificación , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , ARN Mensajero/análisis
18.
Neurosci Lett ; 758: 136013, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34111510

RESUMEN

Preterm infants often suffer from impaired postnatal brain development, and glutamate excitotoxicity is identified as a pivotal mechanism of hyperoxia-induced neurological abnormality. We aimed to investigate the effect of short time hyperoxia on glutamate homeostasis and glutamate transporters expressions in immature brain. Six-day-old (P6) rat pups were exposed to 80% oxygen for 24 h (the hyperoxia group) or placed in atmospheric air (the control group). The concentrations of glutamate and γ-aminobutyric acid (GABA) in immature cerebrum and cerebellum at P7, P14 and P21 were determined by ELISA. The mRNA levels of glutamate transporters including excitatory amino acid transporter 1 (EAAT1), EAAT2, EAAT3, vesicular glutamate transporter 1 (VGLUT1) and VGLUT2 in brain were determined by qPCR. Glutamate accumulation was induced by hyperoxia both in immature cerebrum and cerebellum at P7 but got gradually attenuated at P14 and P21, as evidenced by the changes of glutamate and GABA concentrations. Hyperoxia also induced sustained glutamatic oxidative stress in both cerebrum and cerebellum, as GSH (reduced glutathione) levels in the hyperoxia group were constantly higher than the control group at three examined time-points. Furthermore, at P7, the expressions of all glutamate transporters decreased in both cerebrum and cerebellum except that of EAAT1. At P21, VGLUT2 in cerebrum and EAAT1, EAAT3 and VGLUT2 in cerebellum still displayed significant decrease in expression levels upon hyperoxia stimulation. Taken together, our results indicate that hyperoxia induces glutamate accumulation in brain of rat pups, which is associated with increased oxidative stress and decreased expressions of glutamate transporters.


Asunto(s)
Cerebelo/metabolismo , Cerebro/metabolismo , Hiperoxia/patología , Enfermedades del Prematuro/patología , Animales , Animales Recién Nacidos , Cerebelo/crecimiento & desarrollo , Cerebelo/patología , Cerebro/crecimiento & desarrollo , Cerebro/patología , Modelos Animales de Enfermedad , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Ácido Glutámico/metabolismo , Humanos , Hiperoxia/etiología , Recién Nacido , Recien Nacido Prematuro/crecimiento & desarrollo , Recien Nacido Prematuro/metabolismo , Enfermedades del Prematuro/etiología , Masculino , Estrés Oxidativo , Oxígeno/administración & dosificación , Oxígeno/efectos adversos , Ratas , Factores de Tiempo , Proteínas de Transporte Vesicular de Glutamato/metabolismo
19.
EMBO J ; 40(14): e105712, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34057742

RESUMEN

During development, neural progenitors are in proliferative and immature states; however, the molecular machinery that cooperatively controls both states remains elusive. Here, we report that cyclin D1 (CCND1) directly regulates both proliferative and immature states of cerebellar granule cell progenitors (GCPs). CCND1 not only accelerates cell cycle but also upregulates ATOH1 protein, an essential transcription factor that maintains GCPs in an immature state. In cooperation with CDK4, CCND1 directly phosphorylates S309 of ATOH1, which inhibits additional phosphorylation at S328 and consequently prevents S328 phosphorylation-dependent ATOH1 degradation. Additionally, PROX1 downregulates Ccnd1 expression by histone deacetylation of Ccnd1 promoter in GCPs, leading to cell cycle exit and differentiation. Moreover, WNT signaling upregulates PROX1 expression in GCPs. These findings suggest that WNT-PROX1-CCND1-ATOH1 signaling cascade cooperatively controls proliferative and immature states of GCPs. We revealed that the expression and phosphorylation levels of these molecules dynamically change during cerebellar development, which are suggested to determine appropriate differentiation rates from GCPs to GCs at distinct developmental stages. This study contributes to understanding the regulatory mechanism of GCPs as well as neural progenitors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Ciclina D1/metabolismo , Gránulos Citoplasmáticos/metabolismo , Fosforilación/fisiología , Células Madre/metabolismo , Animales , Ciclo Celular/genética , Diferenciación Celular/fisiología , División Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Proteínas Hedgehog/metabolismo , Ratones , Neurogénesis/fisiología , Transducción de Señal/fisiología , Factores de Transcripción
20.
Nat Commun ; 12(1): 2265, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859199

RESUMEN

Nerve-glia (NG2) glia or oligodendrocyte precursor cells (OPCs) are distributed throughout the gray and white matter and generate myelinating cells. OPCs in white matter proliferate more than those in gray matter in response to platelet-derived growth factor AA (PDGF AA), despite similar levels of its alpha receptor (PDGFRα) on their surface. Here we show that the type 1 integral membrane protein neuropilin-1 (Nrp1) is expressed not on OPCs but on amoeboid and activated microglia in white but not gray matter in an age- and activity-dependent manner. Microglia-specific deletion of Nrp1 compromised developmental OPC proliferation in white matter as well as OPC expansion and subsequent myelin repair after acute demyelination. Exogenous Nrp1 increased PDGF AA-induced OPC proliferation and PDGFRα phosphorylation on dissociated OPCs, most prominently in the presence of suboptimum concentrations of PDGF AA. These findings uncover a mechanism of regulating oligodendrocyte lineage cell density that involves trans-activation of PDGFRα on OPCs via Nrp1 expressed by adjacent microglia.


Asunto(s)
Enfermedades Desmielinizantes/patología , Microglía/fisiología , Neuropilina-1/metabolismo , Células Precursoras de Oligodendrocitos/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Remielinización , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Cuerpo Calloso/citología , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Humanos , Lisofosfatidilcolinas/administración & dosificación , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Modelos Animales , Vaina de Mielina/metabolismo , Neuropilina-1/genética , Oligodendroglía/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Cultivo Primario de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...