Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.809
Filtrar
1.
Cells ; 13(14)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-39056758

RESUMEN

Autism spectrum disorders (ASDs) are complex neurodevelopmental conditions characterized by deficits in social interaction and communication, as well as repetitive behaviors. Although the etiology of ASD is multifactorial, with both genetic and environmental factors contributing to its development, a strong genetic basis is widely recognized. Recent research has identified numerous genetic mutations and genomic rearrangements associated with ASD-characterizing genes involved in brain development. Alterations in developmental programs are particularly harmful during critical periods of brain development. Notably, studies have indicated that genetic disruptions occurring during the second trimester of pregnancy affect cortical development, while disturbances in the perinatal and early postnatal period affect cerebellar development. The developmental defects must be viewed in the context of the role of the cerebellum in cognitive processes, which is now well established. The present review emphasizes the genetic complexity and neuropathological mechanisms underlying ASD and aims to provide insights into the cerebellar involvement in the disorder, focusing on recent advances in the molecular landscape governing its development in humans. Furthermore, we highlight when and in which cerebellar neurons the ASD-associated genes may play a role in the development of cortico-cerebellar circuits. Finally, we discuss improvements in protocols for generating cerebellar organoids to recapitulate the long period of development and maturation of this organ. These models, if generated from patient-induced pluripotent stem cells (iPSC), could provide a valuable approach to elucidate the contribution of defective genes to ASD pathology and inform diagnostic and therapeutic strategies.


Asunto(s)
Trastorno del Espectro Autista , Cerebelo , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Cerebelo/patología , Cerebelo/crecimiento & desarrollo , Animales
2.
JCI Insight ; 9(11)2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38855871

RESUMEN

Human cytomegalovirus (HCMV) infection in infants infected in utero can lead to a variety of neurodevelopmental disorders. However, mechanisms underlying altered neurodevelopment in infected infants remain poorly understood. We have previously described a murine model of congenital HCMV infection in which murine CMV (MCMV) spreads hematogenously and establishes a focal infection in all regions of the brain of newborn mice, including the cerebellum. Infection resulted in disruption of cerebellar cortical development characterized by reduced cerebellar size and foliation. This disruption was associated with altered cell cycle progression of the granule cell precursors (GCPs), which are the progenitors that give rise to granule cells (GCs), the most abundant neurons in the cerebellum. In the current study, we have demonstrated that MCMV infection leads to prolonged GCP cell cycle, premature exit from the cell cycle, and reduced numbers of GCs resulting in cerebellar hypoplasia. Treatment with TNF-α neutralizing antibody partially normalized the cell cycle alterations of GCPs and altered cerebellar morphogenesis induced by MCMV infection. Collectively, our results argue that virus-induced inflammation altered the cell cycle of GCPs resulting in a reduced numbers of GCs and cerebellar cortical hypoplasia, thus providing a potential mechanism for altered neurodevelopment in fetuses infected with HCMV.


Asunto(s)
Ciclo Celular , Cerebelo , Infecciones por Citomegalovirus , Modelos Animales de Enfermedad , Animales , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/patología , Ratones , Cerebelo/virología , Cerebelo/patología , Cerebelo/crecimiento & desarrollo , Cerebelo/anomalías , Femenino , Citomegalovirus , Células-Madre Neurales/virología , Muromegalovirus/fisiología , Animales Recién Nacidos , Humanos , Neuronas/virología , Factor de Necrosis Tumoral alfa/metabolismo , Discapacidades del Desarrollo , Malformaciones del Sistema Nervioso
3.
Int J Mol Sci ; 25(11)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38891784

RESUMEN

The central nervous system of Pacific salmon retains signs of embryonic structure throughout life and a large number of neuroepithelial neural stem cells (NSCs) in the proliferative areas of the brain, in particular. However, the adult nervous system and neurogenesis studies on rainbow trout, Oncorhynchus mykiss, are limited. Here, we studied the localization of glutamine synthetase (GS), vimentin (Vim), and nestin (Nes), as well as the neurons formed in the postembryonic period, labeled with doublecortin (DC), under conditions of homeostatic growth in adult cerebellum and brainstem of Oncorhynchus mykiss using immunohistochemical methods and Western Immunoblotting. We observed that the distribution of vimentin (Vim), nestin (Nes), and glutamine synthetase (GS), which are found in the aNSPCs of both embryonic types (neuroepithelial cells) and in the adult type (radial glia) in the cerebellum and the brainstem of trout, has certain features. Populations of the adult neural stem/progenitor cells (aNSPCs) expressing GS, Vim, and Nes have different morphologies, localizations, and patterns of cluster formation in the trout cerebellum and brainstem, which indicates the morphological and, obviously, functional heterogeneity of these cells. Immunolabeling of PCNA revealed areas in the cerebellum and brainstem of rainbow trout containing proliferating cells which coincide with areas expressing Vim, Nes, and GS. Double immunolabeling revealed the PCNA/GS PCNA/Vim coexpression patterns in the neuroepithelial-type cells in the PVZ of the brainstem. PCNA/GS coexpression in the RG was detected in the submarginal zone of the brainstem. The results of immunohistochemical study of the DC distribution in the cerebellum and brainstem of trout have showed a high level of expression of this marker in various cell populations. This may indicate: (i) high production of the adult-born neurons in the cerebellum and brainstem of adult trout, (ii) high plasticity of neurons in the cerebellum and brainstem of trout. We assume that the source of new cells in the trout brain, along with PVZ and SMZ, containing proliferating cells, may be local neurogenic niches containing the PCNA-positive and silent (PCNA-negative), but expressing NSC markers, cells. The identification of cells expressing DC, Vim, and Nes in the IX-X cranial nerve nuclei of trout was carried out.


Asunto(s)
Tronco Encefálico , Cerebelo , Células-Madre Neurales , Neurogénesis , Plasticidad Neuronal , Oncorhynchus mykiss , Animales , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/crecimiento & desarrollo , Cerebelo/metabolismo , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Tronco Encefálico/metabolismo , Tronco Encefálico/citología , Vimentina/metabolismo , Neuronas/metabolismo , Neuronas/citología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo
4.
Sci Rep ; 14(1): 13030, 2024 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844772

RESUMEN

Digital media (DM) takes an increasingly large part of children's time, yet the long-term effect on brain development remains unclear. We investigated how individual effects of DM use (i.e., using social media, playing video games, or watching television/videos) on the development of the cortex (i.e., global cortical surface area), striatum, and cerebellum in children over 4 years, accounting for both socioeconomic status and genetic predisposition. We used a prospective, multicentre, longitudinal cohort of children from the Adolescent Brain and Cognitive Development Study, aged 9.9 years when entering the study, and who were followed for 4 years. Annually, children reported their DM usage through the Youth Screen Time Survey and underwent brain magnetic resonance imaging scans every 2 years. Quadratic-mixed effect modelling was used to investigate the relationship between individual DM usage and brain development. We found that individual DM usage did not alter the development of cortex or striatum volumes. However, high social media usage was associated with a statistically significant change in the developmental trajectory of cerebellum volumes, and the accumulated effect of high-vs-low social media users on cerebellum volumes over 4 years was only ß = - 0.03, which was considered insignificant. Nevertheless, the developmental trend for heavy social media users was accelerated at later time points. This calls for further studies and longer follow-ups on the impact of social media on brain development.


Asunto(s)
Encéfalo , Imagen por Resonancia Magnética , Juegos de Video , Humanos , Niño , Masculino , Femenino , Encéfalo/crecimiento & desarrollo , Encéfalo/diagnóstico por imagen , Estudios Longitudinales , Juegos de Video/efectos adversos , Medios de Comunicación Sociales , Estudios Prospectivos , Desarrollo Infantil , Adolescente , Cerebelo/crecimiento & desarrollo , Cerebelo/diagnóstico por imagen
5.
eNeuro ; 11(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38777609

RESUMEN

The Cre-lox system is an indispensable tool in neuroscience research for targeting gene deletions to specific cellular populations. Here we assess the utility of several transgenic Cre lines, along with a viral approach, for targeting cerebellar Purkinje cells (PCs) in mice. Using a combination of a fluorescent reporter line (Ai14) to indicate Cre-mediated recombination and a floxed Dystroglycan line (Dag1flox ), we show that reporter expression does not always align precisely with loss of protein. The commonly used Pcp2Cre line exhibits a gradual mosaic pattern of Cre recombination in PCs from Postnatal Day 7 (P7) to P14, while loss of Dag1 protein is not complete until P30. Ptf1aCre drives recombination in precursor cells that give rise to GABAergic neurons in the embryonic cerebellum, including PCs and molecular layer interneurons. However, due to its transient expression in precursors, Ptf1aCre results in stochastic loss of Dag1 protein in these neurons. NestinCre , which is often described as a "pan-neuronal" Cre line for the central nervous system, does not drive Cre-mediated recombination in PCs. We identify a Calb1Cre line that drives efficient and complete recombination in embryonic PCs, resulting in loss of Dag1 protein before the period of synaptogenesis. AAV8-mediated delivery of Cre at P0 results in gradual transduction of PCs during the second postnatal week, with loss of Dag1 protein not reaching appreciable levels until P35. These results characterize several tools for targeting conditional deletions in cerebellar PCs at different developmental stages and illustrate the importance of validating the loss of protein following recombination.


Asunto(s)
Integrasas , Ratones Transgénicos , Células de Purkinje , Animales , Células de Purkinje/metabolismo , Integrasas/genética , Ratones , Recombinación Genética , Alelos , Eliminación de Gen , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Ratones Endogámicos C57BL , Factores de Transcripción
6.
Transgenic Res ; 33(3): 99-117, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38684589

RESUMEN

Golli-myelin basic proteins, encoded by the myelin basic protein gene, are widely expressed in neurons and oligodendrocytes in the central nervous system. Further, prior research has shown that Golli-myelin basic protein is necessary for myelination and neuronal maturation during central nervous system development. In this study, we established Golli-myelin basic protein-floxed mice to elucidate the cell-type-specific effects of Golli-myelin basic protein knockout through the generation of conditional knockout mice (Golli-myelin basic proteinsfl/fl; E3CreN), in which Golli-myelin basic proteins were specifically deleted in cerebellar granule neurons, where Golli-myelin basic proteins are expressed abundantly in wild-type mice. To investigate the role of Golli-myelin basic proteins in cerebellar granule neurons, we further performed histopathological analyses of these mice, with results indicating no morphological changes or degeneration of the major cellular components of the cerebellum. Furthermore, behavioral analysis showed that Golli-myelin basic proteinsfl/fl; E3CreN mice were healthy and did not display any abnormal behavior. These results suggest that the loss of Golli-myelin basic proteins in cerebellar granule neurons does not lead to cerebellar perturbations or behavioral abnormalities. This mouse model could therefore be employed to analyze the effect of Golli-myelin basic protein deletion in specific cell types of the central nervous system, such as other neuronal cells and oligodendrocytes, or in lymphocytes of the immune system.


Asunto(s)
Cerebelo , Ratones Noqueados , Proteína Básica de Mielina , Neuronas , Animales , Neuronas/metabolismo , Ratones , Cerebelo/metabolismo , Cerebelo/crecimiento & desarrollo , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo
7.
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
8.
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
9.
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
10.
Cerebellum ; 23(4): 1626-1641, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38123901

RESUMEN

Cells configure their metabolism in a synchronized and timely manner to meet their energy demands throughout development and adulthood. Transitions of developmental stages are coupled to metabolic shifts, such that glycolysis is highly active during cell proliferation, whereas oxidative phosphorylation prevails in postmitotic states. In the cerebellum, metabolic transitions are remarkable given its protracted developmental timelines. Such distinctive feature, along with its high neuronal density and metabolic demands, make the cerebellum highly vulnerable to metabolic insults. Despite the expansion of metabolomic approaches to uncover biological mechanisms, little is known about the role of metabolism on cerebellar development and maintenance. To illuminate the intricate connections between metabolism, physiology, and cerebellar disorders, we examined here the impact of metabolism on cerebellar growth, maturation, and adulthood through the lens of inborn errors of metabolism.


Asunto(s)
Cerebelo , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Animales , Humanos
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA