Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 983
Filter
1.
Sci Data ; 11(1): 586, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839806

ABSTRACT

Sex bias is known in the prevalence/pathology of neurodevelopmental disorders. Sex-dependent differences of the certain brain areas are known to emerge perinatally through the exposure to sex hormones, while gene expression patterns in the rodent embryonic brain does not seem to be completely the same between male and female. To investigate potential sex differences in gene expression and cortical organization during the embryonic period in mice, we conducted a comprehensive analysis of gene expression for the telencephalon at embryonic day (E) 11.5 (a peak of neural stem cell expansion) and E14.5 (a peak of neurogenesis) using bulk RNA-seq data. As a result, our data showed the existence of notable sex differences in gene expression patterns not obviously at E11.5, but clearly at E14.5 when neurogenesis has become its peak. These data can be useful for exploring potential contribution of genes exhibiting sex differences to the divergence in brain development. Additionally, our data underscore the significance of studying the embryonic period to gain a deeper understanding of sex differences in brain development.


Subject(s)
Telencephalon , Transcriptome , Animals , Telencephalon/embryology , Telencephalon/metabolism , Mice , Female , Male , Neurogenesis/genetics , Sex Characteristics
2.
Article in English | MEDLINE | ID: mdl-38906412

ABSTRACT

DARPP-32 (dopamine and cAMP-regulated phosphoprotein Mr. 32 kDa) is a phosphoprotein that is modulated by multiple receptors integrating intracellular pathways and playing roles in various physiological functions. It is regulated by dopaminergic receptors through the cAMP/protein kinase A (PKA) pathway, which modulates the phosphorylation of threonine 34 (Thr34). When phosphorylated at Thr34, DARPP-32 becomes a potent protein phosphatase-1 (PP1) inhibitor. Since dopamine is involved in the development of GABAergic neurons and DARPP-32 is expressed in the developing brain, it is possible that DARPP-32 has a role in GABAergic neuronal development. We cloned the zebrafish darpp-32 gene (ppp1r1b) gene and observed that it is evolutionarily conserved in its inhibitory domain (Thr34 and surrounding residues) and the docking motif (residues 7-11 (KKIQF)). We also characterized darpp-32 protein expression throughout the 5 days post-fertilization (dpf) zebrafish larval brain by immunofluorescence and demonstrated that darpp-32 is mainly expressed in regions that receive dopaminergic projections (pallium, subpallium, preoptic region, and hypothalamus). We demonstrated that dopamine acutely suppressed darpp-32 activity by reducing the levels of p-darpp-32 in the 5dpf zebrafish larval brain. In addition, the knockdown of darpp-32 resulted in a decrease in the number of GABAergic neurons in the subpallium of the 5dpf larval brain, with a concomitant increase in the number of DAergic neurons. Finally, we demonstrated that darpp-32 downregulation during development reduced the motor behavior of 5dpf zebrafish larvae. Thus, our observations suggest that darpp-32 is an evolutionarily conserved regulator of dopamine receptor signaling and is required for the formation of GABAergic neurons in the developing telencephalon.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32 , Dopamine , GABAergic Neurons , Telencephalon , Zebrafish , Animals , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , GABAergic Neurons/metabolism , Telencephalon/metabolism , Telencephalon/embryology , Dopamine/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Animals, Genetically Modified , Gene Expression Regulation, Developmental/physiology
3.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682303

ABSTRACT

This paper investigates the effect of altering the protein expression dynamics of the bHLH transcription factor Her6 at the single-cell level in the embryonic zebrafish telencephalon. Using a homozygote endogenous Her6:Venus reporter and 4D single-cell tracking, we show that Her6 oscillates in neural telencephalic progenitors and that the fusion of protein destabilisation (PEST) domain alters its expression dynamics, causing most cells to downregulate Her6 prematurely. However, counterintuitively, oscillatory cells increase, with some expressing Her6 at high levels, resulting in increased heterogeneity of Her6 expression in the population. These tissue-level changes appear to be an emergent property of coupling between single-cells, as revealed by experimentally disrupting Notch signalling and by computationally modelling alterations in Her6 protein stability. Despite the profound differences in the single-cell Her6 dynamics, the size of the telencephalon is only transiently altered and differentiation markers do not exhibit significant differences early on; however, a small increase is observed at later developmental stages. Our study suggests that cell coupling provides a compensation strategy, whereby an almost normal phenotype is maintained even though single-cell gene expression dynamics are abnormal, granting phenotypic robustness.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Phenotype , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Developmental , Telencephalon/metabolism , Telencephalon/embryology , Single-Cell Analysis , Signal Transduction , Receptors, Notch/metabolism , Receptors, Notch/genetics , Cell Differentiation
4.
Science ; 382(6667): eadf3786, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824652

ABSTRACT

During early telencephalic development, intricate processes of regional patterning and neural stem cell (NSC) fate specification take place. However, our understanding of these processes in primates, including both conserved and species-specific features, remains limited. Here, we profiled 761,529 single-cell transcriptomes from multiple regions of the prenatal macaque telencephalon. We deciphered the molecular programs of the early organizing centers and their cross-talk with NSCs, revealing primate-biased galanin-like peptide (GALP) signaling in the anteroventral telencephalon. Regional transcriptomic variations were observed along the frontotemporal axis during early stages of neocortical NSC progression and in neurons and astrocytes. Additionally, we found that genes associated with neuropsychiatric disorders and brain cancer risk might play critical roles in the early telencephalic organizers and during NSC progression.


Subject(s)
Neural Stem Cells , Neurogenesis , Telencephalon , Animals , Female , Pregnancy , Macaca , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurons/physiology , Telencephalon/cytology , Telencephalon/embryology , Neurogenesis/genetics , Galanin-Like Peptide/metabolism , Gene Expression Regulation, Developmental , Mental Disorders/genetics , Nervous System Diseases/genetics , Brain Neoplasms/genetics
5.
Anat Rec (Hoboken) ; 306(2): 298-310, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36104941

ABSTRACT

Cell migration from the olfactory placode (OP) is a well-known phenomenon wherein various cell types, such as gonadotropin-releasing hormone (GnRH)-producing neurons, migrate toward the telencephalon (TEL) during early embryonic development. However, the spatial relationship between early migratory cells and the forebrain is unclear. We examined the early development of whole-mount chick embryos to observe the three-dimensional spatial relationship among OP-derived migratory neurons, olfactory nerve (ON), and TEL. Migratory neurons that express highly polysialylated neural cell adhesion molecule (PSA-NCAM) emerge from the OP and spread over a relatively wide TEL area at the Hamburger and Hamilton (HH) stage 17. Most migratory neurons form a cellular cord between the olfactory pit and rostral TEL within HH18-20. The earliest axons from the olfactory epithelium (OE) were detected along this neuronal cord using DiI-labeling at HH21. Furthermore, a few PSA-NCAM-positive neurons were dispersed around the cellular cord and over the lateral TEL at HH18. A long cellular cord branch extending to the lateral TEL was transiently observed within HH18-24. These results suggest a novel migratory route of OP-derived neurons during the early developmental stages. Following GFP vector introduction into the OP of HH13-15 embryos, labeled neurons were detected around and within the lateral TEL at HH23 and HH27. At HH36, labeled cells were observed in the rostral-lateral TEL, including the olfactory bulb (OB) region. GFP-labeled and calretinin-positive neurons were detected in the OB, suggesting that early OP-derived neurons enter the forebrain and function as interneurons in the OB.


Subject(s)
Neurons , Olfactory Bulb , Telencephalon , Animals , Chick Embryo , Axons , Cell Movement , Neurons/metabolism , Olfactory Bulb/embryology , Olfactory Nerve/embryology , Prosencephalon/embryology , Telencephalon/embryology
6.
Science ; 374(6573): eabj6641, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34882453

ABSTRACT

Genetic variation confers susceptibility to neurodevelopmental disorders by affecting the development of specific cell types. Changes in cortical and striatal γ-aminobutyric acid­expressing (GABAergic) neurons are common in autism and schizophrenia. In this study, we used single-cell RNA sequencing to characterize the emergence of cell diversity in the human ganglionic eminences, the transitory structures of the human fetal brain where striatal and cortical GABAergic neurons are generated. We identified regional and temporal diversity among progenitor cells underlying the generation of a variety of projection neurons and interneurons. We found that these cells are specified within the human ganglionic eminences by transcriptional programs similar to those previously identified in rodents. Our findings reveal an evolutionarily conserved regulatory logic controlling the specification, migration, and differentiation of GABAergic neurons in the human telencephalon.


Subject(s)
Interneurons/physiology , Neurogenesis , Telencephalon/embryology , Transcriptome , Animals , Gene Expression Regulation, Developmental , Humans , Mice , Neural Stem Cells/physiology , RNA-Seq , Single-Cell Analysis , Telencephalon/cytology , gamma-Aminobutyric Acid/metabolism
7.
Cell Rep ; 35(4): 109041, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33910016

ABSTRACT

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


Subject(s)
Corpus Striatum/embryology , Stem Cells/metabolism , Telencephalon/embryology , Animals , Mice
8.
Elife ; 102021 04 19.
Article in English | MEDLINE | ID: mdl-33871356

ABSTRACT

The forebrain hemispheres are predominantly separated during embryogenesis by the interhemispheric fissure (IHF). Radial astroglia remodel the IHF to form a continuous substrate between the hemispheres for midline crossing of the corpus callosum (CC) and hippocampal commissure (HC). Deleted in colorectal carcinoma (DCC) and netrin 1 (NTN1) are molecules that have an evolutionarily conserved function in commissural axon guidance. The CC and HC are absent in Dcc and Ntn1 knockout mice, while other commissures are only partially affected, suggesting an additional aetiology in forebrain commissure formation. Here, we find that these molecules play a critical role in regulating astroglial development and IHF remodelling during CC and HC formation. Human subjects with DCC mutations display disrupted IHF remodelling associated with CC and HC malformations. Thus, axon guidance molecules such as DCC and NTN1 first regulate the formation of a midline substrate for dorsal commissures prior to their role in regulating axonal growth and guidance across it.


Subject(s)
Astrocytes/metabolism , Corpus Callosum/metabolism , DCC Receptor/metabolism , Telencephalon/metabolism , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Animals , COS Cells , Cell Line, Tumor , Cell Movement , Cell Shape , Chlorocebus aethiops , Corpus Callosum/embryology , DCC Receptor/genetics , Gene Expression Regulation, Developmental , Genotype , Gestational Age , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Mutation , Netrin-1/genetics , Netrin-1/metabolism , Phenotype , Signal Transduction , Telencephalon/embryology
9.
Dev Biol ; 476: 137-147, 2021 08.
Article in English | MEDLINE | ID: mdl-33775695

ABSTRACT

The MAPK pathway is a major growth signal that has been implicated during the development of progenitors, neurons, and glia in the embryonic brain. Here, we show that the MAPK pathway plays an important role in the generation of distinct cell types from progenitors in the ventral telencephalon. Our data reveal that phospho-p44/42 (called p-ERK1/2) and the ETS transcription factor Etv5, both downstream effectors in the MAPK pathway, show a regional bias in expression during ventral telencephalic development, with enriched expression in the dorsal region of the LGE and ventral region of the MGE at E13.5 and E15.5. Interestingly, expression of both factors becomes more uniform in ventricular zone (VZ) progenitors by E18.5. To gain insight into the role of MAPK activity during progenitor cell development, we used a cre inducible constitutively active MEK1 allele (RosaMEK1DD/+) in combination with a ventral telencephalon enriched cre (Gsx2e-cre) or a dorsal telencephalon enriched cre (Emx1cre/+). Sustained MEK/MAPK activity in the ventral telencephalon (Gsx2e-cre; RosaMEK1DD/+) expanded dorsal lateral ganglionic eminence (dLGE) enriched genes (Gsx2 and Sp8) and oligodendrocyte progenitor cell (OPC) markers (Olig2, Pdgfrα, and Sox10), and also reduced markers in the ventral (v) LGE domain (Isl1 and Foxp1). Activation of MEK/MAPK activity in the dorsal telencephalon (Emx1cre/+; RosaMEK1DD/+) did not initially activate the expression of dLGE or OPC genes at E15.5 but ectopic expression of Gsx2 and OPC markers were observed at E18.5. These results support the idea that MAPK activity as readout by p-ERK1/2 and Etv5 expression is enriched in distinct subdomains of ventral telencephalic progenitors during development. In addition, sustained activation of the MEK/MAPK pathway in the ventral or dorsal telencephalon influences dLGE and OPC identity from progenitors.


Subject(s)
Cell Differentiation/physiology , MAP Kinase Signaling System/physiology , Telencephalon/metabolism , Animals , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Ganglia/metabolism , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , MAP Kinase Kinase 1/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , SOXE Transcription Factors/genetics , Telencephalon/embryology , Telencephalon/physiology , Transcription Factors/metabolism
10.
Cereb Cortex ; 31(7): 3536-3550, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33704445

ABSTRACT

The purpose of the study was to investigate the interrelation of the signal intensities and thicknesses of the transient developmental zones in the cingulate and neocortical telencephalic wall, using T2-weighted 3 T-magnetic resonance imaging (MRI) and histological scans from the same brain hemisphere. The study encompassed 24 postmortem fetal brains (15-35 postconceptional weeks, PCW). The measurements were performed using Fiji and NDP.view2. We found that T2w MR signal-intensity curves show a specific regional and developmental stage profile already at 15 PCW. The MRI-histological correlation reveals that the subventricular-intermediate zone (SVZ-IZ) contributes the most to the regional differences in the MRI-profile and zone thicknesses, growing by a factor of 2.01 in the cingulate, and 1.78 in the neocortical wall. The interrelations of zone or wall thicknesses, obtained by both methods, disclose a different rate and extent of shrinkage per region (highest in neocortical subplate and SVZ-IZ) and stage (highest in the early second half of fetal development), distorting the zones' proportion in histological sections. This intrasubject, slice-matched, 3 T correlative MRI-histological study provides important information about regional development of the cortical wall, critical for the design of MRI criteria for prenatal brain monitoring and early detection of cortical or other brain pathologies in human fetuses.


Subject(s)
Fetus/embryology , Limbic Lobe/embryology , Neocortex/embryology , Telencephalon/embryology , Brain/diagnostic imaging , Brain/embryology , Brain/pathology , Fetus/diagnostic imaging , Fetus/pathology , Gestational Age , Humans , Lateral Ventricles/diagnostic imaging , Lateral Ventricles/embryology , Lateral Ventricles/pathology , Limbic Lobe/diagnostic imaging , Limbic Lobe/pathology , Magnetic Resonance Imaging , Neocortex/diagnostic imaging , Neocortex/pathology , Organ Size , Telencephalon/diagnostic imaging , Telencephalon/pathology
11.
Sci Rep ; 11(1): 3856, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594090

ABSTRACT

Genetic evidence indicates that haploinsufficiency of ARID1B causes intellectual disability (ID) and autism spectrum disorder (ASD), but the neural function of ARID1B is largely unknown. Using both conditional and global Arid1b knockout mouse strains, we examined the role of ARID1B in neural progenitors. We detected an overall decrease in the proliferation of cortical and ventral neural progenitors following homozygous deletion of Arid1b, as well as altered cell cycle regulation and increased cell death. Each of these phenotypes was more pronounced in ventral neural progenitors. Furthermore, we observed decreased nuclear localization of ß-catenin in Arid1b-deficient neurons. Conditional homozygous deletion of Arid1b in ventral neural progenitors led to pronounced ID- and ASD-like behaviors in mice, whereas the deletion in cortical neural progenitors resulted in minor cognitive deficits. This study suggests an essential role for ARID1B in forebrain neurogenesis and clarifies its more pronounced role in inhibitory neural progenitors. Our findings also provide insights into the pathogenesis of ID and ASD.


Subject(s)
Autism Spectrum Disorder/etiology , Intellectual Disability/etiology , Neurogenesis , Telencephalon/embryology , Transcription Factors/physiology , Animals , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/physiology , Pregnancy , Telencephalon/metabolism , beta Catenin/metabolism
12.
Molecules ; 27(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35011481

ABSTRACT

Although tetrabromobisphenol A (TBBPA) has been well proven to disturb TH signaling in both in vitro and in vivo assays, it is still unclear whether TBBPA can affect brain development due to TH signaling disruption. Here, we employed the T3-induced Xenopus metamorphosis assay (TIXMA) and the spontaneous metamorphosis assay to address this issue. In the TIXMA, 5-500 nmol/L TBBPA affected T3-induced TH-response gene expression and T3-induced brain development (brain morphological changes, cell proliferation, and neurodifferentiation) at premetamorphic stages in a complicated biphasic concentration-response manner. Notably, 500 nmol/L TBBPA treatment alone exerted a stimulatory effect on tadpole growth and brain development at these stages, in parallel with a lack of TH signaling activation, suggesting the involvement of other signaling pathways. As expected, at the metamorphic climax, we observed inhibitory effects of 50-500 nmol/L TBBPA on metamorphic development and brain development, which was in agreement with the antagonistic effects of higher concentrations on T3-induced brain development at premetamorphic stages. Taken together, all results demonstrate that TBBPA can disturb TH signaling and subsequently interfere with TH-dependent brain development in Xenopus; meanwhile, other signaling pathways besides TH signaling could be involved in this process. Our study improves the understanding of the effects of TBBPA on vertebrate brain development.


Subject(s)
Brain/drug effects , Brain/embryology , Organogenesis/drug effects , Polybrominated Biphenyls/adverse effects , Thyroid Hormones/metabolism , Animals , Brain/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Environmental Pollutants/adverse effects , Gene Expression Regulation, Developmental/drug effects , Neurogenesis/drug effects , Telencephalon/drug effects , Telencephalon/embryology , Telencephalon/pathology , Triiodothyronine/metabolism , Xenopus laevis
13.
Genes Dev ; 35(1-2): 157-174, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33334823

ABSTRACT

How homeodomain proteins gain sufficient specificity to control different cell fates has been a long-standing problem in developmental biology. The conserved Gsx homeodomain proteins regulate specific aspects of neural development in animals from flies to mammals, and yet they belong to a large transcription factor family that bind nearly identical DNA sequences in vitro. Here, we show that the mouse and fly Gsx factors unexpectedly gain DNA binding specificity by forming cooperative homodimers on precisely spaced and oriented DNA sites. High-resolution genomic binding assays revealed that Gsx2 binds both monomer and homodimer sites in the developing mouse ventral telencephalon. Importantly, reporter assays showed that Gsx2 mediates opposing outcomes in a DNA binding site-dependent manner: Monomer Gsx2 binding represses transcription, whereas homodimer binding stimulates gene expression. In Drosophila, the Gsx homolog, Ind, similarly represses or stimulates transcription in a site-dependent manner via an autoregulatory enhancer containing a combination of monomer and homodimer sites. Integrating these findings, we test a model showing how the homodimer to monomer site ratio and the Gsx protein levels defines gene up-regulation versus down-regulation. Altogether, these data serve as a new paradigm for how cooperative homeodomain transcription factor binding can increase target specificity and alter regulatory outcomes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Animals , Drosophila Proteins/genetics , Genome/genetics , Genome-Wide Association Study , Homeodomain Proteins/genetics , Mice , Protein Binding , Telencephalon/embryology
14.
Acta Neuropathol Commun ; 8(1): 208, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33256853

ABSTRACT

Alcohol affects multiple neurotransmitter systems, notably the GABAergic system and has been recognised for a long time as particularly damaging during critical stages of brain development. Nevertheless, data from the literature are most often derived from animal or in vitro models. In order to study the production, migration and cortical density disturbances of GABAergic interneurons upon prenatal alcohol exposure, we performed immunohistochemical studies by means of the proliferation marker Ki67, GABA and calretinin antibodies in the frontal cortical plate of 17 foetal and infant brains antenatally exposed to alcohol, aged 15 weeks' gestation to 22 postnatal months and in the ganglionic eminences and the subventricular zone of the dorsal telencephalon until their regression, i.e., 34 weeks' gestation. Results were compared with those obtained in 17 control brains aged 14 weeks of gestation to 35 postnatal months. We also focused on interneuron vascular migration along the cortical microvessels by confocal microscopy with double immunolabellings using Glut1, GABA and calretinin. Semi-quantitative and quantitative analyses of GABAergic and calretininergic interneuron density allowed us to identify an insufficient and delayed production of GABAergic interneurons in the ganglionic eminences during the two first trimesters of the pregnancy and a delayed incorporation into the laminar structures of the frontal cortex. Moreover, a mispositioning of GABAergic and calretininergic interneurons persisted throughout the foetal life, these cells being located in the deep layers instead of the superficial layers II and III. Moreover, vascular migration of calretininergic interneurons within the cortical plate was impaired, as reflected by low numbers of interneurons observed close to the cortical perforating vessel walls that may in part explain their abnormal intracortical distribution. Our results are globally concordant with those previously obtained in mouse models, in which alcohol has been shown to induce an interneuronopathy by affecting interneuron density and positioning within the cortical plate, and which could account for the neurological disabilities observed in children with foetal alcohol disorder spectrum.


Subject(s)
Alcohol Drinking , Brain/embryology , Calbindin 2/metabolism , Fetal Alcohol Spectrum Disorders/metabolism , Fetus/embryology , Interneurons/metabolism , Ki-67 Antigen/metabolism , Prenatal Exposure Delayed Effects/metabolism , gamma-Aminobutyric Acid/metabolism , Alcoholism , Binge Drinking , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Movement , Female , Fetal Alcohol Spectrum Disorders/pathology , Fetus/metabolism , Fetus/pathology , Frontal Lobe/embryology , Frontal Lobe/metabolism , Frontal Lobe/pathology , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Humans , Infant , Infant, Newborn , Interneurons/pathology , Male , Pregnancy , Pregnancy Complications , Pregnancy Trimester, Second , Prenatal Exposure Delayed Effects/pathology , Telencephalon/embryology , Telencephalon/metabolism , Telencephalon/pathology
15.
Nat Commun ; 11(1): 5709, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33177537

ABSTRACT

Dorsal-ventral patterning of the mammalian telencephalon is fundamental to the formation of distinct functional regions including the neocortex and ganglionic eminence. While Bone morphogenetic protein (BMP), Wnt, and Sonic hedgehog (Shh) signaling are known to determine regional identity along the dorsoventral axis, how the region-specific expression of these morphogens is established remains unclear. Here we show that the Polycomb group (PcG) protein Ring1 contributes to the ventralization of the mouse telencephalon. Deletion of Ring1b or both Ring1a and Ring1b in neuroepithelial cells induces ectopic expression of dorsal genes, including those for BMP and Wnt ligands, as well as attenuated expression of the gene for Shh, a key morphogen for ventralization, in the ventral telencephalon. We observe PcG protein-mediated trimethylation of histone 3 at lysine-27 and binding of Ring1B at BMP and Wnt ligand genes specifically in the ventral region. Furthermore, forced activation of BMP or Wnt signaling represses Shh expression. Our results thus indicate that PcG proteins suppress BMP and Wnt signaling in a region-specific manner and thereby allow proper Shh expression and development of the ventral telencephalon.


Subject(s)
Gene Expression Regulation, Developmental , Polycomb Repressive Complex 1/metabolism , Telencephalon/embryology , Animals , Body Patterning , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Histones/genetics , Histones/metabolism , Lysine/metabolism , Mice, Knockout , Mice, Transgenic , Polycomb Repressive Complex 1/genetics , Telencephalon/abnormalities , Transcription Factors/genetics , Wnt Signaling Pathway/genetics
16.
EMBO J ; 39(21): e105479, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32985705

ABSTRACT

Structural integrity and cellular homeostasis of the embryonic stem cell niche are critical for normal tissue development. In the telencephalic neuroepithelium, this is controlled in part by cell adhesion molecules and regulators of progenitor cell lineage, but the specific orchestration of these processes remains unknown. Here, we studied the role of microRNAs in the embryonic telencephalon as key regulators of gene expression. By using the early recombiner Rx-Cre mouse, we identify novel and critical roles of miRNAs in early brain development, demonstrating they are essential to preserve the cellular homeostasis and structural integrity of the telencephalic neuroepithelium. We show that Rx-Cre;DicerF/F mouse embryos have a severe disruption of the telencephalic apical junction belt, followed by invagination of the ventricular surface and formation of hyperproliferative rosettes. Transcriptome analyses and functional experiments in vivo show that these defects result from upregulation of Irs2 upon loss of let-7 miRNAs in an apoptosis-independent manner. Our results reveal an unprecedented relevance of miRNAs in early forebrain development, with potential mechanistic implications in pediatric brain cancer.


Subject(s)
Homeostasis , Insulin Receptor Substrate Proteins/metabolism , MicroRNAs/metabolism , Repressor Proteins/metabolism , Telencephalon/embryology , Telencephalon/metabolism , Adherens Junctions , Animals , Apoptosis , Cell Proliferation , Humans , Insulin Receptor Substrate Proteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , PAX6 Transcription Factor/metabolism , Repressor Proteins/genetics , Stem Cells/metabolism , Telencephalon/cytology , Transcription Factors/metabolism
17.
Cell ; 182(3): 754-769.e18, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32610082

ABSTRACT

To discover regulatory elements driving the specificity of gene expression in different cell types and regions of the developing human brain, we generated an atlas of open chromatin from nine dissected regions of the mid-gestation human telencephalon, as well as microdissected upper and deep layers of the prefrontal cortex. We identified a subset of open chromatin regions (OCRs), termed predicted regulatory elements (pREs), that are likely to function as developmental brain enhancers. pREs showed temporal, regional, and laminar differences in chromatin accessibility and were correlated with gene expression differences across regions and gestational ages. We identified two functional de novo variants in a pRE for autism risk gene SLC6A1, and using CRISPRa, demonstrated that this pRE regulates SCL6A1. Additionally, mouse transgenic experiments validated enhancer activity for pREs proximal to FEZF2 and BCL11A. Thus, this atlas serves as a resource for decoding neurodevelopmental gene regulation in health and disease.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental/genetics , Prefrontal Cortex/embryology , Telencephalon/embryology , Animals , Autistic Disorder/genetics , Cell Line , Chromatin Immunoprecipitation Sequencing , Euchromatin/genetics , GABA Plasma Membrane Transport Proteins/genetics , Gene Ontology , Genetic Predisposition to Disease , Gestational Age , Humans , Mice , Mice, Transgenic , Nucleotide Motifs , Point Mutation , Prefrontal Cortex/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Spatio-Temporal Analysis , Telencephalon/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Sci Rep ; 10(1): 8315, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32433594

ABSTRACT

Neuronal migration is essential for constructing functional neural networks. Two posterior septal (PS) nuclei, the triangular septal nucleus and bed nuclei of the anterior commissure, are involved in fear and anxiety. During development, glutamatergic PS neurons undergo long-distance rostrodorsal migration from the thalamic eminence (TE) of the diencephalon, then settle in the caudalmost telencephalon. However, the developmental behavior of PS neurons and the guidance structures facilitating their migration remain unknown. We previously demonstrated the migration of PS neurons along the fornix, a major efferent pathway from the hippocampal formation. Here, we show that the postcommissural fornix is essential for PS neuron migration which is largely confined to its axonal tract, which grows in the opposite direction as PS neuron migration. Fornical axons reach the TE prior to initiation of PS neuron rostrodorsal migration. Ectopic expression of Semaphorin 3 A in the dorsomedial cortex resulted in defective fornix formation. Furthermore, loss of the postcommissural fornix stalled PS neuron migration resulting in abnormal accumulation near their origin. This suggests that PS neurons utilize the postcommissural fornix as a permissive corridor during migration beyond the diencephalic-telencephalic boundary. This axonal support is essential for the functional organization of the heterogeneous septal nuclear complex.


Subject(s)
Cell Movement , Diencephalon/cytology , Hippocampus/physiology , Neurons/cytology , Telencephalon/cytology , Animals , Diencephalon/embryology , Electroporation , Female , Hippocampus/cytology , Hippocampus/embryology , Mice , Pregnancy , Semaphorin-3A/metabolism , Telencephalon/embryology
19.
PLoS Biol ; 18(5): e3000705, 2020 05.
Article in English | MEDLINE | ID: mdl-32401820

ABSTRACT

Modeling the processes of neuronal progenitor proliferation and differentiation to produce mature cortical neuron subtypes is essential for the study of human brain development and the search for potential cell therapies. We demonstrated a novel paradigm for the generation of vascularized organoids (vOrganoids) consisting of typical human cortical cell types and a vascular structure for over 200 days as a vascularized and functional brain organoid model. The observation of spontaneous excitatory postsynaptic currents (sEPSCs), spontaneous inhibitory postsynaptic currents (sIPSCs), and bidirectional electrical transmission indicated the presence of chemical and electrical synapses in vOrganoids. More importantly, single-cell RNA-sequencing analysis illustrated that vOrganoids exhibited robust neurogenesis and that cells of vOrganoids differentially expressed genes (DEGs) related to blood vessel morphogenesis. The transplantation of vOrganoids into the mouse S1 cortex resulted in the construction of functional human-mouse blood vessels in the grafts that promoted cell survival in the grafts. This vOrganoid culture method could not only serve as a model to study human cortical development and explore brain disease pathology but also provide potential prospects for new cell therapies for nervous system disorders and injury.


Subject(s)
Cell Culture Techniques , Neurogenesis , Organoids/blood supply , Telencephalon/embryology , Animals , Embryonic Stem Cells , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells , Mice, Inbred NOD , Mice, SCID , Organoids/metabolism , Organoids/transplantation
20.
Development ; 147(7)2020 04 10.
Article in English | MEDLINE | ID: mdl-32122989

ABSTRACT

The Gsx2 homeodomain transcription factor promotes neural progenitor identity in the lateral ganglionic eminence (LGE), despite upregulating the neurogenic factor Ascl1. How this balance in maturation is maintained is unclear. Here, we show that Gsx2 and Ascl1 are co-expressed in subapical progenitors that have unique transcriptional signatures in LGE ventricular zone (VZ) cells. Moreover, whereas Ascl1 misexpression promotes neurogenesis in dorsal telencephalic progenitors, the co-expression of Gsx2 with Ascl1 inhibits neurogenesis. Using luciferase assays, we found that Gsx2 reduces the ability of Ascl1 to activate gene expression in a dose-dependent and DNA binding-independent manner. Furthermore, Gsx2 physically interacts with the basic helix-loop-helix (bHLH) domain of Ascl1, and DNA-binding assays demonstrated that this interaction interferes with the ability of Ascl1 to bind DNA. Finally, we modified a proximity ligation assay for tissue sections and found that Ascl1-Gsx2 interactions are enriched within LGE VZ progenitors, whereas Ascl1-Tcf3 (E-protein) interactions predominate in the subventricular zone. Thus, Gsx2 contributes to the balance between progenitor maintenance and neurogenesis by physically interacting with Ascl1, interfering with its DNA binding and limiting neurogenesis within LGE progenitors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/embryology , Cell Proliferation , Homeodomain Proteins/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/metabolism , Cell Proliferation/genetics , Cells, Cultured , Drosophila , Embryo, Mammalian , Female , Ganglia/cytology , Ganglia/embryology , Homeodomain Proteins/genetics , Homeostasis/genetics , Male , Mice , Mice, Transgenic , Protein Binding , Telencephalon/cytology , Telencephalon/embryology
SELECTION OF CITATIONS
SEARCH DETAIL