Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Am J Hum Genet ; 111(3): 473-486, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38354736

ABSTRACT

Disease-associated variants identified from genome-wide association studies (GWASs) frequently map to non-coding areas of the genome such as introns and intergenic regions. An exclusive reliance on gene-agnostic methods of genomic investigation could limit the identification of relevant genes associated with polygenic diseases such as Alzheimer disease (AD). To overcome such potential restriction, we developed a gene-constrained analytical method that considers only moderate- and high-risk variants that affect gene coding sequences. We report here the application of this approach to publicly available datasets containing 181,388 individuals without and with AD and the resulting identification of 660 genes potentially linked to the higher AD prevalence among Africans/African Americans. By integration with transcriptome analysis of 23 brain regions from 2,728 AD case-control samples, we concentrated on nine genes that potentially enhance the risk of AD: AACS, GNB5, GNS, HIPK3, MED13, SHC2, SLC22A5, VPS35, and ZNF398. GNB5, the fifth member of the heterotrimeric G protein beta family encoding Gß5, is primarily expressed in neurons and is essential for normal neuronal development in mouse brain. Homozygous or compound heterozygous loss of function of GNB5 in humans has previously been associated with a syndrome of developmental delay, cognitive impairment, and cardiac arrhythmia. In validation experiments, we confirmed that Gnb5 heterozygosity enhanced the formation of both amyloid plaques and neurofibrillary tangles in the brains of AD model mice. These results suggest that gene-constrained analysis can complement the power of GWASs in the identification of AD-associated genes and may be more broadly applicable to other polygenic diseases.


Subject(s)
Alzheimer Disease , GTP-Binding Protein beta Subunits , Mice , Humans , Animals , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Genome-Wide Association Study , Neurofibrillary Tangles/metabolism , Phenotype , Genomics , Amyloid beta-Peptides/genetics , Brain/metabolism , Solute Carrier Family 22 Member 5/genetics , Solute Carrier Family 22 Member 5/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism
2.
Cereb Cortex ; 31(4): 1914-1926, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33290502

ABSTRACT

GluN3A subunits endow N-Methyl-D-Aspartate receptors (NMDARs) with unique biophysical, trafficking, and signaling properties. GluN3A-NMDARs are typically expressed during postnatal development, when they are thought to gate the refinement of neural circuits by inhibiting synapse maturation, and stabilization. Recent work suggests that GluN3A also operates in adult brains to control a variety of behaviors, yet a full spatiotemporal characterization of GluN3A expression is lacking. Here, we conducted a systematic analysis of Grin3a (gene encoding mouse GluN3A) mRNA expression in the mouse brain by combining high-sensitivity colorimetric and fluorescence in situ hybridization with labeling for neuronal subtypes. We find that, while Grin3a mRNA expression peaks postnatally, significant levels are retained into adulthood in specific brain regions such as the amygdala, medial habenula, association cortices, and high-order thalamic nuclei. The time-course of emergence and down-regulation of Grin3a expression varies across brain region, cortical layer of residence, and sensory modality, in a pattern that correlates with previously reported hierarchical gradients of brain maturation and functional specialization. Grin3a is expressed in both excitatory and inhibitory neurons, with strong mRNA levels being a distinguishing feature of somatostatin interneurons. Our study provides a comprehensive map of Grin3a distribution across the murine lifespan and paves the way for dissecting the diverse functions of GluN3A in health and disease.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Neurons/metabolism , Prosencephalon/growth & development , Prosencephalon/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Age Factors , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, N-Methyl-D-Aspartate/genetics
3.
Development ; 142(7): 1267-78, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25804737

ABSTRACT

Medial ganglionic eminence (MGE)-derived GABAergic cortical interneurons (cINs) consist of multiple subtypes that are involved in many cortical functions. They also have a remarkable capacity to migrate, survive and integrate into cortical circuitry after transplantation into postnatal cortex. These features have engendered considerable interest in generating distinct subgroups of interneurons from pluripotent stem cells (PSCs) for the study of interneuron fate and function, and for the development of cell-based therapies. Although advances have been made, the capacity to generate highly enriched pools of subgroup fate-committed interneuron progenitors from PSCs has remained elusive. Previous studies have suggested that the two main MGE-derived interneuron subgroups--those expressing somatostatin (SST) and those expressing parvalbumin (PV)--are specified in the MGE from Nkx2.1-expressing progenitors at higher or lower levels of sonic hedgehog (Shh) signaling, respectively. To further explore the role of Shh and other factors in cIN fate determination, we generated a reporter line such that Nkx2.1-expressing progenitors express mCherry and postmitotic Lhx6-expressing MGE-derived interneurons express GFP. Manipulations of Shh exposure and time in culture influenced the subgroup fates of ESC-derived interneurons. Exposure to higher Shh levels, and collecting GFP-expressing precursors at 12 days in culture, resulted in the strongest enrichment for SST interneurons over those expressing PV, whereas the strongest enrichment for PV interneurons was produced by lower Shh and by collecting mCherry-expressing cells after 17 days in culture. These findings confirm that fate determination of cIN subgroups is crucially influenced by Shh signaling, and provide a system for the further study of interneuron fate and function.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Hedgehog Proteins/metabolism , Interneurons/metabolism , Parvalbumins/metabolism , Signal Transduction , Somatostatin/metabolism , Action Potentials , Animals , Body Patterning , Cell Line , Cell Separation , Cerebral Cortex/cytology , Embryonic Stem Cells/metabolism , GABAergic Neurons/metabolism , Genes, Reporter , Green Fluorescent Proteins/metabolism , Median Eminence/cytology , Mice , Mitosis , Stem Cell Transplantation , Telencephalon/embryology , Telencephalon/metabolism , Time Factors
4.
Front Cell Neurosci ; 18: 1334244, 2024.
Article in English | MEDLINE | ID: mdl-38419656

ABSTRACT

Introduction: Enhancer of zeste homolog 2 (Ezh2) is responsible for trimethylation of histone 3 at lysine 27 (H3K27me3), resulting in repression of gene expression. Here, we explore the role of Ezh2 in forebrain GABAergic interneuron development. Methods: We removed Ezh2 in the MGE by generating Nkx2-1Cre;Ezh2 conditional knockout mice. We then characterized changes in MGE-derived interneuron fate and electrophysiological properties in juvenile mice, as well as alterations in gene expression, chromatin accessibility and histone modifications in the MGE. Results: Loss of Ezh2 increases somatostatin-expressing (SST+) and decreases parvalbumin-expressing (PV+) interneurons in the forebrain. We observe fewer MGE-derived interneurons in the first postnatal week, indicating reduced interneuron production. Intrinsic electrophysiological properties in SST+ and PV+ interneurons are normal, but PV+ interneurons display increased axonal complexity in Ezh2 mutant mice. Single nuclei multiome analysis revealed differential gene expression patterns in the embryonic MGE that are predictive of these cell fate changes. Lastly, CUT&Tag analysis revealed that some genomic loci are particularly resistant or susceptible to shifts in H3K27me3 levels in the absence of Ezh2, indicating differential selectivity to epigenetic perturbation. Discussion: Thus, loss of Ezh2 in the MGE alters interneuron fate, morphology, and gene expression and regulation. These findings have important implications for both normal development and potentially in disease etiologies.

5.
Neurobiol Dis ; 53: 36-48, 2013 May.
Article in English | MEDLINE | ID: mdl-23201207

ABSTRACT

GABAergic interneurons of the cerebral cortex (cINs) play crucial roles in many aspects of cortical function. The diverse types of cINs are classified into subgroups according to their morphology, intrinsic physiology, neurochemical markers and synaptic targeting. Recent advances in mouse genetics, imaging and electrophysiology techniques have greatly advanced our efforts to understand the role of normal cIN function and its dysfunction in neuropsychiatric disorders. In schizophrenia (SCZ), a wealth of data suggests that cIN function is perturbed, and that interneuron dysfunction may underlie key symptoms of the disease. In this review, we discuss the link between cINs and SCZ, focusing on the evidence for GABAergic signaling deficits from both SCZ patients and mouse models.


Subject(s)
Cerebral Cortex/pathology , GABAergic Neurons/pathology , Interneurons/pathology , Neural Inhibition/physiology , Schizophrenia/pathology , Animals , Cerebral Cortex/physiology , GABAergic Neurons/physiology , Humans , Interneurons/physiology , Schizophrenia/physiopathology , gamma-Aminobutyric Acid/physiology
6.
STAR Protoc ; 4(1): 101944, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36520627

ABSTRACT

Efficient protocols to generate single-cell and single-nuclei suspensions are critical for the burgeoning field of single-cell/single-nuclei sequencing. Here we describe procedures to generate single-cell and single-nuclei suspensions from embryonic and adult mouse brains. This protocol can be modified for any brain region and/or neural cell type. For complete details on the use and execution of this protocol, please refer to Lee et al. (2022),1 Rhodes et al. (2022),2 Mahadevan et al. (2021),3 Ekins et al. (2020),4 and Wester et al. (2019).5.


Subject(s)
Cell Nucleus , Neurons , Animals , Mice , Suspensions , Brain
7.
Nat Genet ; 55(2): 280-290, 2023 02.
Article in English | MEDLINE | ID: mdl-36717694

ABSTRACT

How enhancers activate their distal target promoters remains incompletely understood. Here we dissect how CTCF-mediated loops facilitate and restrict such regulatory interactions. Using an allelic series of mouse mutants, we show that CTCF is neither required for the interaction of the Sox2 gene with distal enhancers, nor for its expression. Insertion of various combinations of CTCF motifs, between Sox2 and its distal enhancers, generated boundaries with varying degrees of insulation that directly correlated with reduced transcriptional output. However, in both epiblast and neural tissues, enhancer contacts and transcriptional induction could not be fully abolished, and insertions failed to disrupt implantation and neurogenesis. In contrast, Sox2 expression was undetectable in the anterior foregut of mutants carrying the strongest boundaries, and these animals fully phenocopied loss of SOX2 in this tissue. We propose that enhancer clusters with a high density of regulatory activity can better overcome physical barriers to maintain faithful gene expression and phenotypic robustness.


Subject(s)
Chromatin , Enhancer Elements, Genetic , Mice , Animals , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism
8.
Elife ; 112022 02 17.
Article in English | MEDLINE | ID: mdl-35175194

ABSTRACT

The ventricular zone (VZ) of the nervous system contains radial glia cells that were originally considered relatively homogenous in their gene expression, but a detailed characterization of transcriptional diversity in these VZ cells has not been reported. Here, we performed single-cell RNA sequencing to characterize transcriptional heterogeneity of neural progenitors within the VZ and subventricular zone (SVZ) of the ganglionic eminences (GEs), the source of all forebrain GABAergic neurons. By using a transgenic mouse line to enrich for VZ cells, we characterize significant transcriptional heterogeneity, both between GEs and within spatial subdomains of specific GEs. Additionally, we observe differential gene expression between E12.5 and E14.5 VZ cells, which could provide insights into temporal changes in cell fate. Together, our results reveal a previously unknown spatial and temporal genetic diversity of VZ cells in the ventral forebrain that will aid our understanding of initial fate decisions in the forebrain.


Subject(s)
Neural Stem Cells , Prosencephalon , Animals , Cell Differentiation/physiology , Lateral Ventricles , Mice , Mice, Transgenic , Neural Stem Cells/metabolism
9.
Nat Commun ; 13(1): 4196, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35858915

ABSTRACT

A comprehensive characterization of epigenomic organization in the embryonic mouse forebrain will enhance our understanding of neurodevelopment and provide insight into mechanisms of neurological disease. Here we collected single-cell chromatin accessibility profiles from four distinct neurogenic regions of the embryonic mouse forebrain using single nuclei ATAC-Seq (snATAC-Seq). We identified thousands of differentially accessible peaks, many restricted to distinct progenitor cell types or brain regions. We integrated snATAC-Seq and single cell transcriptome data to characterize changes of chromatin accessibility at enhancers and promoters with associated transcript abundance. Multi-modal integration of histone modifications (CUT&Tag and CUT&RUN), promoter-enhancer interactions (Capture-C) and high-order chromatin structure (Hi-C) extended these initial observations. This dataset reveals a diverse chromatin landscape with region-specific regulatory mechanisms and genomic interactions in distinct neurogenic regions of the embryonic mouse brain and represents an extensive public resource of a 'ground truth' epigenomic landscape at this critical stage of neurogenesis.


Subject(s)
Chromatin , Epigenome , Animals , Chromatin/genetics , Histone Code , Mice , Prosencephalon , Regulatory Sequences, Nucleic Acid
10.
Nat Commun ; 12(1): 2471, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931636

ABSTRACT

In vertebrates, motor control relies on cholinergic neurons in the spinal cord that have been extensively studied over the past hundred years, yet the full heterogeneity of these neurons and their different functional roles in the adult remain to be defined. Here, we develop a targeted single nuclear RNA sequencing approach and use it to identify an array of cholinergic interneurons, visceral and skeletal motor neurons. Our data expose markers for distinguishing these classes of cholinergic neurons and their rich diversity. Specifically, visceral motor neurons, which provide autonomic control, can be divided into more than a dozen transcriptomic classes with anatomically restricted localization along the spinal cord. The complexity of the skeletal motor neurons is also reflected in our analysis with alpha, gamma, and a third subtype, possibly corresponding to the elusive beta motor neurons, clearly distinguished. In combination, our data provide a comprehensive transcriptomic description of this important population of neurons that control many aspects of physiology and movement and encompass the cellular substrates for debilitating degenerative disorders.


Subject(s)
Cholinergic Neurons/cytology , Interneurons/cytology , Motor Neurons/cytology , Single-Cell Analysis/methods , Solitary Nucleus/metabolism , Spinal Cord/metabolism , Transcriptome/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Female , In Situ Hybridization , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/physiology , RNA-Seq , Spinal Cord/cytology , Spinal Cord/physiology
11.
Front Mol Neurosci ; 14: 712609, 2021.
Article in English | MEDLINE | ID: mdl-34630033

ABSTRACT

Medial ganglionic eminence (MGE)-derived parvalbumin (PV)+, somatostatin (SST)+and Neurogliaform (NGFC)-type cortical and hippocampal interneurons, have distinct molecular, anatomical, and physiological properties. However, the molecular mechanisms regulating their maturation remain poorly understood. Here, via single-cell transcriptomics, we show that the obligate NMDA-type glutamate receptor (NMDAR) subunit gene Grin1 mediates transcriptional regulation of gene expression in specific subtypes of MGE-derived interneurons, leading to altered subtype abundances. Notably, MGE-specific early developmental Grin1 loss results in a broad downregulation of diverse transcriptional, synaptogenic and membrane excitability regulatory programs in the juvenile brain. These widespread gene expression abnormalities mirror aberrations that are typically associated with neurodevelopmental disorders. Our study hence provides a road map for the systematic examination of NMDAR signaling in interneuron subtypes, revealing potential MGE-specific genetic targets that could instruct future therapies of psychiatric disorders.

12.
J Neurosci ; 29(47): 14855-63, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19940181

ABSTRACT

Partial decussation of sensory pathways allows neural inputs from both sides of the body to project to the same target region where these signals will be integrated. Here, to better understand mechanisms of eye-specific targeting, we studied how retinal ganglion cell (RGC) axons terminate in their thalamic target, the dorsal lateral geniculate nucleus (dLGN), when crossing at the optic chiasm midline is altered. In models with gain- and loss-of-function of EphB1, the receptor that directs the ipsilateral projection at the optic chiasm, misrouted RGCs target the appropriate retinotopic zone in the opposite dLGN. However, in EphB1(-/-) mice, the misrouted axons do not intermingle with normally projecting RGC axons and segregate instead into a distinct patch. We also revisited the role of retinal activity on eye-specific targeting by blocking correlated waves of activity with epibatidine into both eyes. We show that, in wild-type mice, retinal waves are necessary during the first postnatal week for both proper distribution and eye-specific segregation of ipsilateral axons in the mature dLGN. Moreover, in EphB1(-/-) mice, refinement of ipsilateral axons is perturbed in control conditions and is further impaired after epibatidine treatment. Finally, retinal waves are required for the formation of the segregated patch of misrouted axons in EphB1(-/-) mice. These findings implicate molecular determinants for targeting of eye-specific zones that are independent of midline guidance cues and that function in concert with correlated retinal activity to sculpt retinogeniculate projections.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Geniculate Bodies/abnormalities , Growth Cones/ultrastructure , Receptor, EphB1/genetics , Retina/abnormalities , Visual Pathways/abnormalities , Action Potentials/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cues , Functional Laterality/genetics , Geniculate Bodies/metabolism , Growth Cones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/genetics , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/genetics , Vision, Ocular/physiology , Visual Pathways/metabolism
13.
J Neurosci ; 29(11): 3463-74, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295152

ABSTRACT

At the optic chiasm, retinal ganglion cell (RGC) axons make the decision to either avoid or traverse the midline, a maneuver that establishes the binocular pathways. In mice, the ipsilateral retinal projection arises from RGCs in the peripheral ventrotemporal (VT) crescent of the retina. These RGCs express the guidance receptor EphB1, which interacts with ephrin-B2 on radial glia cells at the optic chiasm to repulse VT axons away from the midline and into the ipsilateral optic tract. However, because VT RGCs express more than one EphB receptor, the sufficiency and specificity of the EphB1 receptor in directing the ipsilateral projection is unclear. In this study, we use in utero retinal electroporation to demonstrate that ectopic EphB1 expression can redirect RGCs with a normally crossed projection to an ipsilateral trajectory. Moreover, EphB1 is specifically required for rerouting RGC projections ipsilaterally, because introduction of the highly similar EphB2 receptor is much less efficient in redirecting RGC fibers, even when expressed at higher surface levels. Introduction of EphB1-EphB2 chimeric receptors into RGCs reveals that both extracellular and juxtamembrane domains of EphB1 are required to efficiently convert RGC projections ipsilaterally. Together, these data describe for the first time functional differences between two highly similar Eph receptors at a decision point in vivo, with EphB1 displaying unique properties that efficiently drives the uncrossed retinal projection.


Subject(s)
Receptor, EphB1/physiology , Retina/embryology , Retina/physiology , Visual Pathways/embryology , Visual Pathways/physiology , Animals , Female , Functional Laterality/genetics , Mice , Mice, Inbred C57BL , Point Mutation/genetics , Pregnancy , Receptor, EphB1/genetics , Recombinant Fusion Proteins/physiology , Retina/cytology , Retinal Ganglion Cells/physiology
14.
Elife ; 92020 11 05.
Article in English | MEDLINE | ID: mdl-33150866

ABSTRACT

Type I lissencephaly is a neuronal migration disorder caused by haploinsuffiency of the PAFAH1B1 (mouse: Pafah1b1) gene and is characterized by brain malformation, developmental delays, and epilepsy. Here, we investigate the impact of Pafah1b1 mutation on the cellular migration, morphophysiology, microcircuitry, and transcriptomics of mouse hippocampal CA1 parvalbumin-containing inhibitory interneurons (PV+INTs). We find that WT PV+INTs consist of two physiological subtypes (80% fast-spiking (FS), 20% non-fast-spiking (NFS)) and four morphological subtypes. We find that cell-autonomous mutations within interneurons disrupts morphophysiological development of PV+INTs and results in the emergence of a non-canonical 'intermediate spiking (IS)' subset of PV+INTs. We also find that now dominant IS/NFS cells are prone to entering depolarization block, causing them to temporarily lose the ability to initiate action potentials and control network excitation, potentially promoting seizures. Finally, single-cell nuclear RNAsequencing of PV+INTs revealed several misregulated genes related to morphogenesis, cellular excitability, and synapse formation.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Classical Lissencephalies and Subcortical Band Heterotopias/pathology , Hippocampus/cytology , Interneurons/metabolism , Microtubule-Associated Proteins/metabolism , Parvalbumins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Electrophysiological Phenomena , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics
15.
J Neurosci ; 28(23): 5910-9, 2008 Jun 04.
Article in English | MEDLINE | ID: mdl-18524895

ABSTRACT

The navigation of retinal axons to ipsilateral and contralateral targets in the brain depends on the decision to cross or avoid the midline at the optic chiasm, a critical guidance maneuver that establishes the binocular visual pathway. Previous work has identified a specific guidance receptor, EphB1, that mediates the repulsion of uncrossed axons away from its ligand, ephrinB2, at the optic chiasm midline (Williams et al., 2003), and a transcription factor Zic2, that, like EphB1, is required for formation of the ipsilateral retinal projection (Herrera et al., 2003). Although the reported similarities in localization implicated that Zic2 regulates EphB1 (Herrera et al., 2003; Williams et al., 2003; Pak et al., 2004), whether Zic2 drives expression of EphB1 protein has not been elucidated. Here we show that EphB1 protein is expressed in the growth cones of axons from ventrotemporal (VT) retina that project ipsilaterally and that repulsion by ephrinB2 is determined by the presence of this receptor on growth cones. Moreover, ectopic delivery of Zic2 into explants from non-VT retina induces expression of EphB1 mRNA and protein. The upregulated EphB1 receptor protein is localized to growth cones and is functional, because it is sufficient to change retinal ganglion cell axon behavior from extension onto, to avoidance of, ephrinB2 substrates. Our results demonstrate that Zic2 upregulates EphB1 expression and define a link between a transcription factor and expression of a guidance receptor protein essential for axon guidance at the vertebrate midline.


Subject(s)
Axons/physiology , Cell Movement/physiology , Ephrin-B2/metabolism , Gene Expression Regulation, Developmental/physiology , Receptor, EphB1/biosynthesis , Retinal Ganglion Cells/physiology , Transcription Factors/physiology , Zinc Fingers/physiology , Animals , Cells, Cultured , Female , Mice , Mice, Mutant Strains , Pregnancy , Receptor, EphB1/genetics , Receptor, EphB1/physiology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
16.
Neuron ; 102(5): 960-975.e6, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31027966

ABSTRACT

Neocortical circuits consist of stereotypical motifs that must self-assemble during development. Recent evidence suggests that the subtype identity of both excitatory projection neurons (PNs) and inhibitory interneurons (INs) is important for this process. We knocked out the transcription factor Satb2 in PNs to induce those of the intratelencephalic (IT) type to adopt a pyramidal tract (PT)-type identity. Loss of IT-type PNs selectively disrupted the lamination and circuit integration of INs derived from the caudal ganglionic eminence (CGE). Strikingly, reprogrammed PNs demonstrated reduced synaptic targeting of CGE-derived INs relative to controls. In control mice, IT-type PNs targeted neighboring CGE INs, while PT-type PNs did not in deep layers, confirming this lineage-dependent motif. Finally, single-cell RNA sequencing revealed that major CGE IN subtypes were conserved after loss of IT PNs, but with differential transcription of synaptic proteins and signaling molecules. Thus, IT-type PNs influence CGE-derived INs in a non-cell-autonomous manner during cortical development.


Subject(s)
Cell Lineage , Interneurons/metabolism , Neocortex/embryology , Synapses/metabolism , Animals , Cell Movement , Gene Expression , Gene Knockout Techniques , Interneurons/cytology , Matrix Attachment Region Binding Proteins/genetics , Mice , Neural Inhibition/physiology , Neural Pathways/embryology , Neurons/cytology , Neurons/metabolism , Pyramidal Tracts/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Telencephalon/cytology , Transcription Factors/genetics
17.
Neuron ; 103(5): 853-864.e4, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31257105

ABSTRACT

GABAergic interneurons have many important functions in cortical circuitry, a reflection of their cell diversity. The developmental origins of this diversity are poorly understood. Here, we identify rostral-caudal regionality in Wnt exposure within the interneuron progenitor zone delineating the specification of the two main interneuron subclasses. Caudally situated medial ganglionic eminence (MGE) progenitors receive high levels of Wnt signaling and give rise to somatostatin (SST)-expressing cortical interneurons. By contrast, parvalbumin (PV)-expressing basket cells originate mostly from the rostral MGE, where Wnt signaling is attenuated. Interestingly, rather than canonical signaling through ß-catenin, signaling via the non-canonical Wnt receptor Ryk regulates interneuron cell-fate specification in vivo and in vitro. Indeed, gain of function of Ryk intracellular domain signaling regulates SST and PV fate in a dose-dependent manner, suggesting that Ryk signaling acts in a graded fashion. These data reveal an important role for non-canonical Wnt-Ryk signaling in establishing the correct ratios of cortical interneuron subtypes.


Subject(s)
Cerebral Cortex/embryology , GABAergic Neurons/metabolism , Interneurons/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Receptor Protein-Tyrosine Kinases/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , GABAergic Neurons/cytology , Interneurons/cytology , Mice , Mouse Embryonic Stem Cells , Neural Stem Cells/cytology , Parvalbumins/metabolism , Somatostatin/metabolism
18.
Mol Cell Biol ; 25(13): 5699-711, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15964824

ABSTRACT

The Krüppel-like transcription factors (KLFs) are important regulators of cell proliferation and differentiation in several different organ systems. The mouse Klf7 gene is strongly active in postmitotic neuroblasts of the developing nervous system, and the corresponding protein stimulates transcription of the cyclin-dependent kinase inhibitor p21waf/cip gene. Here we report that loss of KLF7 activity in mice leads to neonatal lethality and a complex phenotype which is associated with deficits in neurite outgrowth and axonal misprojection at selected anatomical locations of the nervous system. Affected axon pathways include those of the olfactory and visual systems, the cerebral cortex, and the hippocampus. In situ hybridizations and immunoblots correlated loss of KLF7 activity in the olfactory epithelium with significant downregulation of the p21waf/cip and p27kip1 genes. Cotransfection experiments extended the last finding by documenting KLF7's ability to transactivate a reporter gene construct driven by the proximal promoter of p27kip1. Consistent with emerging evidence for a role of Cip/Kip proteins in cytoskeletal dynamics, we also documented p21waf/cip and p27kip1 accumulation in the cytoplasm of differentiating olfactory sensory neurons. KLF7 activity might therefore control neuronal morphogenesis in part by optimizing the levels of molecules that promote axon outgrowth.


Subject(s)
Central Nervous System/embryology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Morphogenesis , Neurons/metabolism , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Axons/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Central Nervous System/growth & development , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21 , Embryo Loss/genetics , Gene Expression Regulation, Developmental , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Kruppel-Like Transcription Factors , Mice , Mice, Knockout , Mice, Transgenic , Neurons/cytology , Olfactory Mucosa/cytology , Promoter Regions, Genetic , Retina/cytology , Tissue Distribution , Transcription, Genetic
19.
J Exp Neurosci ; 12: 1179069518758656, 2018.
Article in English | MEDLINE | ID: mdl-29511360

ABSTRACT

The mammalian brain develops from a simple sheet of neuroepithelial cells into an incredibly complex structure containing billions of neurons with trillions of synapses. Understanding how intrinsic genetic programs interact with environmental cues to generate neuronal diversity and proper connectivity is one of the most daunting challenges in developmental biology. We recently explored this issue in forebrain GABAergic inhibitory interneurons, an extremely diverse population of neurons that are classified into distinct subtypes based on morphology, neurochemical markers, and electrophysiological properties. Immature interneurons were harvested from one brain region and transplanted into a different region, allowing us to assess how challenging cells in a new environment affected their fate. Do these grafted cells adopt characteristics of the host environment or retain features from the donor environment? We found that the proportion of interneuron subgroups is determined by the host region, but some interneuron subtypes maintain features attributable to the donor environment. In this commentary, I expound on potential mechanisms that could underlie these observations and explore the implications of these findings in a greater context of developmental neuroscience.

20.
J Vis Exp ; (136)2018 06 08.
Article in English | MEDLINE | ID: mdl-29939182

ABSTRACT

Neuronal fate determination and maturation requires an intricate interplay between genetic programs and environmental signals. However, disentangling the roles of intrinsic vs. extrinsic mechanisms that regulate this differentiation process is a conundrum for all developmental neurobiologists. This issue is magnified for GABAergic interneurons, an incredibly heterogeneous cell population that is born from transient embryonic structures and undergo a protracted migratory phase to disperse throughout the telencephalon. To explore how different brain environments affect interneuron fate and maturation, we developed a protocol for harvesting fluorescently labeled immature interneuron precursors from specific brain regions in newborn mice (P0-P2). At this age, interneuron migration is nearly complete and these cells are residing in their final resting environments with relatively little synaptic integration. Following collection of single cell solutions via flow cytometry, these interneuron precursors are transplanted into P0-P2 wildtype postnatal pups. By performing both homotopic (e.g., cortex-to-cortex) or heterotopic (e.g., cortex-to-hippocampus) transplantations, one can assess how challenging immature interneurons in new brain environments affects their fate, maturation, and circuit integration. Brains can be harvested in adult mice and assayed with a wide variety of posthoc analysis on grafted cells, including immunohistochemical, electrophysiological and transcriptional profiling. This general approach provides investigators with a strategy to assay how distinct brain environments can influence numerous aspects of neuron development and identify if specific neuronal characteristics are primarily driven by hardwired genetic programs or environmental cues.


Subject(s)
Brain/pathology , Hippocampus/metabolism , Interneurons/metabolism , Animals , Cell Differentiation , Hippocampus/pathology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL