Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.804
Filter
1.
Sci Rep ; 14(1): 13030, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844772

ABSTRACT

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.


Subject(s)
Brain , Magnetic Resonance Imaging , Video Games , Humans , Child , Male , Female , Brain/growth & development , Brain/diagnostic imaging , Longitudinal Studies , Video Games/adverse effects , Social Media , Prospective Studies , Child Development , Adolescent , Cerebellum/growth & development , Cerebellum/diagnostic imaging
2.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38777609

ABSTRACT

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.


Subject(s)
Integrases , Mice, Transgenic , Purkinje Cells , Animals , Purkinje Cells/metabolism , Integrases/genetics , Mice , Recombination, Genetic , Alleles , Gene Deletion , Cerebellum/growth & development , Cerebellum/metabolism , Mice, Inbred C57BL , Transcription Factors
3.
Sci Adv ; 10(17): eade1650, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669326

ABSTRACT

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.


Subject(s)
Cerebellum , Cerebellum/abnormalities , Hedgehog Proteins , Kinesins , Nervous System Malformations , Purkinje Cells , Animals , Kinesins/metabolism , Kinesins/genetics , Cerebellum/metabolism , Cerebellum/growth & development , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , Purkinje Cells/metabolism , Signal Transduction , Cell Proliferation , Mice, Knockout , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Developmental Disabilities
4.
J Neurosci ; 44(17)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38527808

ABSTRACT

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.


Subject(s)
Cerebellum , Neuronal Plasticity , Reflex, Vestibulo-Ocular , Animals , Reflex, Vestibulo-Ocular/physiology , Neuronal Plasticity/physiology , Mice , Cerebellum/growth & development , Cerebellum/physiology , Male , Female , Purkinje Cells/physiology , Adaptation, Physiological/physiology , Mice, Inbred C57BL , Mice, Knockout
5.
Nature ; 625(7996): 788-796, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029793

ABSTRACT

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.


Subject(s)
Cerebellum , Evolution, Molecular , Mammals , Neurogenesis , Animals , Humans , Mice , Cell Lineage/genetics , Cerebellum/cytology , Cerebellum/embryology , Cerebellum/growth & development , Fetus/cytology , Fetus/embryology , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Opossums/embryology , Opossums/growth & development , Purkinje Cells/cytology , Purkinje Cells/metabolism , Single-Cell Gene Expression Analysis , Species Specificity , Transcriptome , Mammals/embryology , Mammals/growth & development
6.
Science ; 381(6662): 1112-1119, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37676945

ABSTRACT

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.


Subject(s)
Cellular Senescence , Cerebellum , Chromatin Assembly and Disassembly , Genome , Neurons , Animals , Humans , Mice , Cerebellum/cytology , Cerebellum/growth & development , Neurons/metabolism , Imaging, Three-Dimensional , Single-Cell Analysis , Atlases as Topic
7.
J Cell Physiol ; 238(3): 533-548, 2023 03.
Article in English | MEDLINE | ID: mdl-36649308

ABSTRACT

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.


Subject(s)
Cerebellar Neoplasms , Gene Expression Regulation, Neoplastic , Medulloblastoma , Child , Humans , Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellum/growth & development , Cerebellum/metabolism , Medulloblastoma/diagnosis , Medulloblastoma/genetics , Medulloblastoma/metabolism , Medulloblastoma/pathology , Cell Line, Tumor , Adenosine Triphosphate/metabolism , Infant, Newborn , Infant , Child, Preschool , Aging/metabolism , Prognosis
8.
Biol Open ; 11(8)2022 08 15.
Article in English | MEDLINE | ID: mdl-35876806

ABSTRACT

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.


Subject(s)
Cerebellum/growth & development , Nervous System Malformations , Platelet-Derived Growth Factor/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Signal Transduction , Animals , Dandy-Walker Syndrome/diagnosis , Dandy-Walker Syndrome/genetics , Humans , Lymphokines , Mice , Nervous System Malformations/genetics
9.
Sci Rep ; 12(1): 2355, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149757

ABSTRACT

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.


Subject(s)
Cerebellum/physiology , Gait , Memory, Short-Term , Adult , Cerebellum/diagnostic imaging , Cerebellum/growth & development , Female , Humans , Magnetic Resonance Imaging , Male , Organ Size , Reaction Time , Walking Speed , Young Adult
10.
Food Chem Toxicol ; 159: 112751, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34871666

ABSTRACT

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.


Subject(s)
Alkanesulfonic Acids/toxicity , Cerebellum/drug effects , Dietary Exposure , Fluorocarbons/toxicity , Maternal Exposure , Neurotoxins/toxicity , Animals , Anxiety , Behavior, Animal/drug effects , Cerebellum/growth & development , Cerebellum/physiopathology , Female , Lactation , Male , Mice , Psychomotor Performance/drug effects
11.
Dev Med Child Neurol ; 64(2): 192-199, 2022 02.
Article in English | MEDLINE | ID: mdl-34416027

ABSTRACT

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.


Subject(s)
Brain Injuries , Cerebral Palsy , Child Development/physiology , Developmental Disabilities , Heart Defects, Congenital/surgery , Ischemic Stroke , Motor Skills/physiology , Pyramidal Tracts , Brain Injuries/diagnostic imaging , Brain Injuries/pathology , Brain Injuries/physiopathology , Cerebellum/diagnostic imaging , Cerebellum/growth & development , Cerebellum/pathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Cerebral Palsy/diagnostic imaging , Cerebral Palsy/pathology , Cerebral Palsy/physiopathology , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/etiology , Developmental Disabilities/pathology , Developmental Disabilities/physiopathology , Female , Gray Matter/diagnostic imaging , Gray Matter/growth & development , Gray Matter/pathology , Heart Defects, Congenital/complications , Heart Defects, Congenital/diagnostic imaging , Humans , Infant , Infant, Newborn , Ischemic Stroke/diagnostic imaging , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Magnetic Resonance Imaging , Male , Prospective Studies , Pyramidal Tracts/diagnostic imaging , Pyramidal Tracts/growth & development , Pyramidal Tracts/pathology , White Matter/diagnostic imaging , White Matter/growth & development , White Matter/pathology
12.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34740966

ABSTRACT

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.


Subject(s)
Cerebellum/growth & development , Animals , Calcium Signaling , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Purkinje Cells/physiology , Synapses/physiology
13.
Elife ; 102021 11 03.
Article in English | MEDLINE | ID: mdl-34730085

ABSTRACT

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.


Subject(s)
Cerebellum/physiology , Interneurons/physiology , Synaptic Transmission/physiology , Animals , Cerebellum/growth & development , Female , Interneurons/metabolism , Male , Mice
14.
Nat Commun ; 12(1): 5702, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588434

ABSTRACT

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.


Subject(s)
CHARGE Syndrome/genetics , Cerebellum/growth & development , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Polymicrogyria/genetics , Animals , CHARGE Syndrome/pathology , Cell Division/genetics , Cerebellum/pathology , Chromatin Assembly and Disassembly , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Enhancer Elements, Genetic , Epigenesis, Genetic , Histone Code , Humans , Infant , Mice , Mice, Knockout , Mutation , Neural Stem Cells/metabolism , Neurons/metabolism , Polymicrogyria/pathology , RNA-Seq
15.
Cell Rep ; 36(7): 109522, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34407407

ABSTRACT

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.


Subject(s)
Angiopoietin-2/metabolism , Dendrites/metabolism , Morphogenesis , Purkinje Cells/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Angiopoietin-1/metabolism , Animals , Cerebellum/blood supply , Cerebellum/growth & development , Gene Deletion , Gene Expression Regulation , Integrases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Organ Specificity
16.
Nat Neurosci ; 24(10): 1392-1401, 2021 10.
Article in English | MEDLINE | ID: mdl-34400844

ABSTRACT

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.


Subject(s)
Cerebellum/growth & development , Endocrine Glands/physiology , Placenta/physiology , Pregnanolone/deficiency , Pregnanolone/physiology , Social Behavior , Aldehyde Reductase/genetics , Animals , Autism Spectrum Disorder/etiology , Cerebellum/physiology , Female , GABA Agonists/pharmacology , GABA Modulators , Gene Deletion , Humans , Infant , Infant, Newborn , Male , Mice , Muscimol/pharmacology , Pregnancy , Receptors, GABA-A/physiology , Sex Characteristics , Trophoblasts/metabolism , White Matter/pathology
17.
Science ; 373(6558)2021 08 27.
Article in English | MEDLINE | ID: mdl-34446581

ABSTRACT

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.


Subject(s)
Biological Evolution , Cerebellum/cytology , Cerebellum/growth & development , Neurons/physiology , Regulatory Elements, Transcriptional , Animals , Cerebellum/embryology , Chromatin/genetics , Chromatin/metabolism , DNA, Intergenic , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Male , Mice , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Opossums/genetics
18.
Sci Rep ; 11(1): 17108, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34429472

ABSTRACT

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.


Subject(s)
Aging/physiology , Connectome , High-Intensity Interval Training/methods , Learning , Motor Skills , Aged , Cerebellum/growth & development , Cerebellum/physiology , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged
19.
Neurosci Lett ; 758: 136013, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34111510

ABSTRACT

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.


Subject(s)
Cerebellum/metabolism , Cerebrum/metabolism , Hyperoxia/pathology , Infant, Premature, Diseases/pathology , Animals , Animals, Newborn , Cerebellum/growth & development , Cerebellum/pathology , Cerebrum/growth & development , Cerebrum/pathology , Disease Models, Animal , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamic Acid/metabolism , Humans , Hyperoxia/etiology , Infant, Newborn , Infant, Premature/growth & development , Infant, Premature/metabolism , Infant, Premature, Diseases/etiology , Male , Oxidative Stress , Oxygen/administration & dosage , Oxygen/adverse effects , Rats , Time Factors , Vesicular Glutamate Transport Proteins/metabolism
20.
Alcohol Clin Exp Res ; 45(7): 1408-1423, 2021 07.
Article in English | MEDLINE | ID: mdl-34060105

ABSTRACT

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.


Subject(s)
Animals, Newborn/growth & development , Cerebellum/growth & development , Ethanol/adverse effects , Hippocampus/growth & development , Neurogenesis/drug effects , Transcriptome/drug effects , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cerebellum/metabolism , Ethanol/administration & dosage , Female , Gene Expression Regulation, Developmental/drug effects , Hippocampus/metabolism , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Pregnancy , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...