Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cereb Cortex ; 28(6): 1946-1958, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28449024

ABSTRACT

The neocortex of primates, including humans, contains more abundant and diverse inhibitory neurons compared with rodents, but the molecular foundations of these observations are unknown. Through integrative gene coexpression analysis, we determined a consensus transcriptional profile of GABAergic neurons in mid-gestation human neocortex. By comparing this profile to genes expressed in GABAergic neurons purified from neonatal mouse neocortex, we identified conserved and distinct aspects of gene expression in these cells between the species. We show here that the calcium-binding protein secretagogin (SCGN) is robustly expressed by neocortical GABAergic neurons derived from caudal ganglionic eminences (CGE) and lateral ganglionic eminences during human but not mouse brain development. Through electrophysiological and morphometric analyses, we examined the effects of SCGN expression on GABAergic neuron function and form. Forced expression of SCGN in CGE-derived mouse GABAergic neurons significantly increased total neurite length and arbor complexity following transplantation into mouse neocortex, revealing a molecular pathway that contributes to morphological differences in these cells between rodents and primates.


Subject(s)
GABAergic Neurons/metabolism , Neocortex/embryology , Neurogenesis/physiology , Secretagogins/metabolism , Animals , Humans , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Neurites/metabolism , Transcriptome
2.
Cereb Cortex ; 28(11): 3797-3815, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29028947

ABSTRACT

The postnatal functions of the Dlx1&2 transcription factors in cortical interneurons (CINs) are unknown. Here, using conditional Dlx1, Dlx2, and Dlx1&2 knockouts (CKOs), we defined their roles in specific CINs. The CKOs had dendritic, synaptic, and survival defects, affecting even PV+ CINs. We provide evidence that DLX2 directly drives Gad1, Gad2, and Vgat expression, and show that mutants had reduced mIPSC amplitude. In addition, the mutants formed fewer GABAergic synapses on excitatory neurons and had reduced mIPSC frequency. Furthermore, Dlx1/2 CKO had hypoplastic dendrites, fewer excitatory synapses, and reduced excitatory input. We provide evidence that some of these phenotypes were due to reduced expression of GRIN2B (a subunit of the NMDA receptor), a high confidence Autism gene. Thus, Dlx1&2 coordinate key components of CIN postnatal development by promoting their excitability, inhibitory output, and survival.


Subject(s)
Cerebral Cortex/growth & development , GABAergic Neurons/physiology , Homeodomain Proteins/physiology , Interneurons/physiology , Synapses/physiology , Transcription Factors/physiology , gamma-Aminobutyric Acid/biosynthesis , Animals , Cerebral Cortex/cytology , Female , GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Glutamate Decarboxylase/metabolism , Homeodomain Proteins/genetics , Interneurons/cytology , Male , Mice, Knockout , Miniature Postsynaptic Potentials , Transcription Factors/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 109(34): 13829-34, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22753490

ABSTRACT

Inhibitory interneurons regulate the responses of cortical circuits. In auditory cortical areas, inhibition from these neurons narrows spectral tuning and shapes response dynamics. Acute disruptions of inhibition expand spectral receptive fields. However, the effects of long-term perturbations of inhibitory circuitry on auditory cortical responses are unknown. We ablated ~30% of dendrite-targeting cortical inhibitory interneurons after the critical period by studying mice with a conditional deletion of Dlx1. Following the loss of interneurons, baseline firing rates rose and tone-evoked responses became less sparse in auditory cortex. However, contrary to acute blockades of inhibition, the sizes of spectral receptive fields were reduced, demonstrating both higher thresholds and narrower bandwidths. Furthermore, long-latency responses at the edge of the receptive field were absent. On the basis of changes in response dynamics, the mechanism for the reduction in receptive field size appears to be a compensatory loss of cortico-cortically (CC) driven responses. Our findings suggest chronic conditions that feature changes in inhibitory circuitry are not likely to be well modeled by acute network manipulations, and compensation may be a critical component of chronic neuronal conditions.


Subject(s)
Acoustic Stimulation , Auditory Cortex/physiology , Homeodomain Proteins/genetics , Interneurons/physiology , Neural Inhibition/physiology , Neurons/physiology , Transcription Factors/genetics , Action Potentials/physiology , Animals , Dendrites/metabolism , Electroencephalography/methods , Female , Male , Mice , Mice, Knockout , Models, Genetic , Neurons/drug effects , Phenotype , Time Factors
4.
J Neurosci ; 33(43): 16897-914, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24155296

ABSTRACT

Here we have provided evidence that nitric oxide-cyclic GMP (NO-cGMP) signaling regulates neurite length and migration of immature neurons derived from the medial ganglionic eminence (MGE). Dlx1/2(-/-) and Lhx6(-/-) mouse mutants, which exhibit MGE interneuron migration defects, have reduced expression of the gene encoding the α subunit of a soluble guanylate cyclase (Gucy1A3). Furthermore, Dlx1/2(-/-) mouse mutants have reduced expression of NO synthase 1 (NOS1). Gucy1A3(-/-) mice have a transient reduction in cortical interneuron number. Pharmacological inhibition of soluble guanylate cyclase and NOS activity rapidly induces neurite retraction of MGE cells in vitro and in slice culture and robustly inhibits cell migration from the MGE and caudal ganglionic eminence. We provide evidence that these cellular phenotypes are mediated by activation of the Rho signaling pathway and inhibition of myosin light chain phosphatase activity.


Subject(s)
Cell Movement , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Interneurons/metabolism , Median Eminence/cytology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cells, Cultured , Guanylate Cyclase/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interneurons/physiology , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Median Eminence/metabolism , Mice , Mutation , Myosin Light Chains/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurites/physiology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Soluble Guanylyl Cyclase , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Development ; 137(23): 4101-10, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21062867

ABSTRACT

Polarized radial glia are crucial to the formation of the cerebral cortex. They serve as neural progenitors and as guides for neuronal placement in the developing cerebral cortex. The maintenance of polarized morphology is essential for radial glial functions, but the extent to which the polarized radial glial scaffold is static or dynamic during corticogenesis remains an open question. The developmental dynamics of radial glial morphology, inter-radial glial interactions during corticogenesis, and the role of the cell polarity complexes in these activities remain undefined. Here, using real-time imaging of cohorts of mouse radial glia cells, we show that the radial glial scaffold, upon which the cortex is constructed, is highly dynamic. Radial glial cells within the scaffold constantly interact with one another. These interactions are mediated by growth cone-like endfeet and filopodia-like protrusions. Polarized expression of the cell polarity regulator Cdc42 in radial glia regulates glial endfeet activities and inter-radial glial interactions. Furthermore, appropriate regulation of Gsk3 activity is required to maintain the overall polarity of the radial glia scaffold. These findings reveal dynamism and interactions among radial glia that appear to be crucial contributors to the formation of the cerebral cortex. Related cell polarity determinants (Cdc42, Gsk3) differentially influence radial glial activities within the evolving radial glia scaffold to coordinate the formation of cerebral cortex.


Subject(s)
Cell Communication , Cell Polarity , Cerebral Cortex/embryology , Glycogen Synthase Kinase 3/metabolism , Neuroglia/cytology , Neuroglia/enzymology , cdc42 GTP-Binding Protein/metabolism , Animals , Biological Assay , Cell Shape , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Glycogen Synthase Kinase 3 beta , Growth Cones/metabolism , Integrases/metabolism , Intermediate Filament Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nestin , Pseudopodia/enzymology
6.
Proc Natl Acad Sci U S A ; 107(12): 5622-7, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212127

ABSTRACT

Neuregulin-1 (NRG1) and Disrupted-in-Schizophrenia-1 (DISC1) are promising susceptibility factors for schizophrenia. Both are multifunctional proteins with roles in a variety of neurodevelopmental processes, including progenitor cell proliferation, migration, and differentiation. Here, we provide evidence linking these factors together in a single pathway, which is mediated by ErbB receptors and PI3K/Akt. We show that signaling by NRG1 and NRG2, but not NRG3, increase expression of an isoform of DISC1 in vitro. Receptors ErbB2 and ErbB3, but not ErbB4, are responsible for transducing this effect, and PI3K/Akt signaling is also required. In NRG1 knockout mice, this DISC1 isoform is selectively reduced during neurodevelopment. Furthermore, a similar decrease in DISC1 expression is seen in beta-site amyloid precursor protein cleaving enzyme-1 (BACE1) knockout mice, in which NRG1/Akt signaling is reportedly impaired. In contrast to neuronal DISC1 that was reported and characterized, expression of DISC1 in other types of cells in the brain has not been addressed. Here we demonstrate that DISC1, like NRG and ErbB proteins, is expressed in neurons, astrocytes, oligodendrocytes, microglia, and radial progenitors. These findings may connect NRG1, ErbBs, Akt, and DISC1 in a common pathway, which may regulate neurodevelopment and contribute to susceptibility to schizophrenia.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Nerve Tissue Proteins/metabolism , Neuregulin-1/metabolism , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/genetics , Astrocytes/metabolism , Brain/metabolism , Cell Line , Cells, Cultured , Humans , Mice , Mice, Knockout , Microglia/metabolism , Nerve Tissue Proteins/genetics , Neuregulin-1/deficiency , Neuregulin-1/genetics , Neurogenesis , Neurons/metabolism , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schizophrenia/etiology , Signal Transduction
7.
Development ; 136(17): 2965-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19666823

ABSTRACT

The radial glial cells serve as neural progenitors and as a migratory guide for newborn neurons in the developing cerebral cortex. These functions require appropriate organization and proliferation of the polarized radial glial scaffold. Here, we demonstrate in mice that the myristoylated alanine-rich C-kinase substrate protein (MARCKS), a prominent cellular substrate for PKC, modulates radial glial placement and expansion. Loss of MARCKS results in ectopic collection of mitotically active radial progenitors away from the ventricular zone (VZ) in the upper cerebral wall. Apical restriction of key polarity complexes [CDC42, beta-catenin (CTNNB1), N-cadherin (CDH2), myosin IIB (MYOIIB), aPKCzeta, LGL, PAR3, pericentrin, PROM1] is lost. Furthermore, the radial glial scaffold in Marcks null cortex is compromised, with discontinuous, non-radial processes apparent throughout the cerebral wall and deformed, bulbous, unbranched end-feet at the basal ends. Further, the density of radial processes within the cerebral cortex is reduced. These deficits in radial glial development culminate in aberrant positioning of neurons and disrupted cortical lamination. Genetic rescue experiments demonstrate, surprisingly, that phosphorylation of MARCKS by PKC is not essential for the role of MARCKS in radial glial cell development. By contrast, the myristoylation domain of MARCKS needed for membrane association is essential for MARCKS function in radial glia. The membrane-associated targeting of MARCKS and the resultant polarized distribution of signaling complexes essential for apicobasal polarity may constitute a critical event in the appropriate placement, proliferation and organization of polarized radial glial scaffold in the developing cerebral cortex.


Subject(s)
Cell Proliferation , Cerebral Cortex , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neuroglia , Stem Cells , Animals , Biomarkers/metabolism , Cell Membrane/metabolism , Cell Polarity , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Myristoylated Alanine-Rich C Kinase Substrate , Neuroglia/cytology , Neuroglia/physiology , Protein Kinase C/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/cytology , Stem Cells/physiology
8.
Proc Natl Acad Sci U S A ; 106(18): 7595-600, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19383784

ABSTRACT

Cortical GABAergic interneurons, most of which originate in the ganglionic eminences, take distinct tangential migratory trajectories into the developing cerebral cortex. However, the ligand-receptor systems that modulate the tangential migration of distinct groups of interneurons into the emerging cerebral wall remain unclear. Here, we show that netrin-1, a diffusible guidance cue expressed along the migratory routes traversed by GABAergic interneurons, interacts with alpha3beta1 integrin to promote interneuronal migration. In vivo analysis of interneuron-specific alpha3beta1 integrin, netrin-1-deficient mice (alpha3(lox/-)Dlx5/6-CIE, netrin-1(-/-)) reveals specific deficits in the patterns of interneuronal migration along the top of the developing cortical plate, resulting in aberrant interneuronal positioning throughout the cerebral cortex and hippocampus of conditional alpha3(lox/-)Dlx5/6-CIE, netrin-1(-/-) mice. These results indicate that specific guidance mechanisms, such as netrin-1-alpha3beta1 integrin interactions, modulate distinct routes of interneuronal migration and the consequent positioning of groups of cortical interneurons in the developing cerebral cortex.


Subject(s)
Cell Movement , Cerebral Cortex/growth & development , Integrin alpha3beta1/metabolism , Interneurons/physiology , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Movement/genetics , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Interneurons/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Netrin-1 , Tumor Suppressor Proteins/genetics , gamma-Aminobutyric Acid/metabolism
9.
J Neurophysiol ; 105(5): 1984-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21325686

ABSTRACT

Dlx transcription factors are important in the differentiation of GABAergic interneurons. In mice lacking Dlx1, early steps in interneuron development appear normal. Beginning at ∼ 1 mo of age, primarily dendrite-innervating interneuron subtypes begin to undergo apoptosis in Dlx1(-/-) mice; this is accompanied by a reduction in GABAergic transmission and late-onset epilepsy. The reported reduction of synaptic inhibition is greater than might be expected given that interneuron loss is relatively modest in Dlx1(-/-) mice. Here we report that voltage-clamp recordings of CA1 interneurons in hippocampal slices prepared from Dlx1(-/-) animals older than postnatal day 30 (>P30) revealed a significant reduction in excitatory postsynaptic current (EPSC) amplitude. No changes in EPSCs onto interneurons were observed in cells recorded from younger animals (P9-12). Current-clamp recordings from interneurons at these early postnatal ages showed that interneurons in Dlx1(-/-) mutants were immature and more excitable, although membrane properties normalized by P30. Terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling, caspase-3, and NeuN staining did not reveal frank cell damage or loss in area CA3 of hippocampal sections from adult Dlx1(-/-) mice. Delayed interneuron maturation may lead to interneuron hyperexcitability, followed by a compensatory reduction in the strength of excitatory transmission onto interneurons. This reduced excitation onto surviving interneurons, coupled with the loss of a significant fraction of GABAergic inputs to excitatory neurons starting at P30, may underlie cortical dysrhythmia and seizures previously observed in adult Dlx1(-/-) mice.


Subject(s)
CA1 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials/physiology , Gene Deletion , Glutamic Acid/physiology , Interneurons/physiology , Transcription Factors/deficiency , Age Factors , Animals , Hippocampus/physiology , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Neural Pathways/physiology , Transcription Factors/genetics , gamma-Aminobutyric Acid/physiology
10.
Glia ; 56(9): 935-41, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18381651

ABSTRACT

Extracellular matrix associated Sparc-like 1 (SC1/SPARCL1) can influence the function of astroglial cells in the developing and mature central nervous system (CNS). To examine SC1's significance in the CNS, we generated a BAC transgenic mouse model in which Sc1 is expressed in radial glia and their astrocyte derivatives using the astroglial-specific Blbp (Brain-lipid binding protein; [Feng et al., (1994) Neuron 12:895-908]) regulatory elements. Characterization of these Blbf-Sc1 transgenic mice show elevated Sc1 transcript and protein in an astroglial selective pattern throughout the CNS. This model provides a novel in vivo system for evaluating the role of SC1 in brain development and function, in general, and for understanding SC1's significance in the fate and function of astroglial cells, in particular.


Subject(s)
Astrocytes/physiology , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Central Nervous System/physiology , Chromosomes, Artificial, Bacterial/physiology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Animals , Astrocytes/metabolism , Calcium-Binding Proteins/physiology , Central Nervous System/cytology , Central Nervous System/metabolism , Chromosomes, Artificial, Bacterial/genetics , Extracellular Matrix Proteins/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Animal
11.
Neuron ; 84(5): 940-53, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25467980

ABSTRACT

Little is known about genetic mechanisms that regulate the ratio of cortical excitatory and inhibitory neurons. We show that NPAS1 and NPAS3 transcription factors (TFs) are expressed in progenitor domains of the mouse basal ganglia (subpallium, MGE, and CGE). NPAS1(-/-) mutants had increased proliferation, ERK signaling, and expression of Arx in the MGE and CGE. NPAS1(-/-) mutants also had increased neocortical inhibition (sIPSC and mIPSC) and generated an excess of somatostatin(+) (SST) (MGE-derived) and vasoactive intestinal polypeptide(+) (VIP) (CGE-derived) neocortical interneurons, but had a normal density of parvalbumin(+) (PV) (MGE-derived) interneurons. In contrast, NPAS3(-/-) mutants showed decreased proliferation and ERK signaling in progenitors of the ganglionic eminences and had fewer SST(+) and VIP(+) interneurons. NPAS1 repressed activity of an Arx enhancer, and Arx overexpression resulted in increased proliferation of CGE progenitors. These results provide insights into genetic regulation of cortical interneuron numbers and cortical inhibitory tone.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/cytology , Gene Expression Regulation, Developmental/physiology , Interneurons/classification , Interneurons/physiology , Nerve Tissue Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Autistic Disorder/genetics , Autistic Disorder/pathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation/genetics , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Dev Cell ; 31(6): 677-89, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25535916

ABSTRACT

Coordinated migration of distinct classes of neurons to appropriate positions leads to the formation of functional neuronal circuitry in the cerebral cortex. The two major classes of cortical neurons, interneurons and projection neurons, utilize distinctly different modes (radial versus tangential) and routes of migration to arrive at their final positions in the cerebral cortex. Here, we show that adenomatous polyposis coli (APC) modulates microtubule (MT) severing in interneurons to facilitate tangential mode of interneuron migration, but not the glial-guided, radial migration of projection neurons. APC regulates the stability and activity of the MT-severing protein p60-katanin in interneurons to promote the rapid remodeling of neuronal processes necessary for interneuron migration. These findings reveal how severing and restructuring of MTs facilitate distinct modes of neuronal migration necessary for laminar organization of neurons in the developing cerebral cortex.


Subject(s)
Adenosine Triphosphatases/metabolism , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Gene Expression Regulation, Developmental , Interneurons/metabolism , Microtubules/metabolism , Neurons/physiology , Alleles , Animals , Cell Differentiation , Cell Movement , Cerebral Cortex/metabolism , Cytoskeleton/metabolism , Gene Deletion , Green Fluorescent Proteins/metabolism , Katanin , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neurons/metabolism , Time Factors
13.
PLoS One ; 8(6): e67679, 2013.
Article in English | MEDLINE | ID: mdl-23826333

ABSTRACT

Synaptogenesis has been extensively studied along with dendritic spine development in glutamatergic pyramidal neurons, however synapse development in cortical interneurons, which are largely aspiny, is comparatively less well understood. Dact1, one of 3 paralogous Dact (Dapper/Frodo) family members in mammals, is a scaffold protein implicated in both the Wnt/ß-catenin and the Wnt/Planar Cell Polarity pathways. We show here that Dact1 is expressed in immature cortical interneurons. Although Dact1 is first expressed in interneuron precursors during proliferative and migratory stages, constitutive Dact1 mutant mice have no major defects in numbers or migration of these neurons. However, cultured cortical interneurons derived from these mice have reduced numbers of excitatory synapses on their dendrites. We selectively eliminated Dact1 from mouse cortical interneurons using a conditional knock-out strategy with a Dlx-I12b enhancer-Cre allele, and thereby demonstrate a cell-autonomous role for Dact1 during postsynaptic development. Confirming this cell-autonomous role, we show that synapse numbers in Dact1 deficient cortical interneurons are rescued by virally-mediated re-expression of Dact1 specifically targeted to these cells. Synapse numbers in these neurons are also rescued by similarly targeted expression of the Dact1 binding partner Dishevelled-1, and partially rescued by expression of Disrupted in Schizophrenia-1, a synaptic protein genetically implicated in susceptibility to several major mental illnesses. In sum, our results support a novel cell-autonomous postsynaptic role for Dact1, in cooperation with Dishevelled-1 and possibly Disrupted in Schizophrenia-1, in the formation of synapses on cortical interneuron dendrites.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , GABAergic Neurons/metabolism , Interneurons/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Prosencephalon/cytology , Prosencephalon/growth & development , Animals , Cell Movement , Dendrites/metabolism , Dishevelled Proteins , GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Interneurons/cytology , Intracellular Signaling Peptides and Proteins/genetics , Lentivirus/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Nerve Tissue Proteins/metabolism , Neural Inhibition , Prosencephalon/metabolism , Protein Binding , RNA-Binding Proteins , Synapses/metabolism
14.
Nat Commun ; 2: 446, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21863013

ABSTRACT

Microtubule cytoskeletal dynamics of cortical progenitors and astroglial cells have critical roles in the emergence of normal functional organization of cerebral cortex and in disease processes such as tumorigenesis. However, tools to efficiently visualize these events are lacking. Here we describe a mouse genetic model to efficiently visualize and analyse radial progenitors, their astroglial progeny, and the microtubule cytoskeleton of these cells in the developing and adult brain. Using this tool, we demonstrate altered microtubule organization and capture dynamics in adenomatous polyposis coli-deficient radial progenitors. Further, using multiphoton microscopy, we show the utility of this tool in real-time imaging of astrocytes in living mouse brain and the short-term stable nature of astrocytes in cerebral cortex. Thus, this model will help explore the dynamics of radial progenitor/astrocyte development or dysfunction and the influence of microtubule functions during these events.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Genetic Techniques , Mice/metabolism , Microtubules/chemistry , Stem Cells/metabolism , Animals , Astrocytes/chemistry , Astrocytes/cytology , Brain/cytology , Brain/growth & development , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice/genetics , Mice/growth & development , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microtubules/metabolism , Stem Cells/chemistry , Stem Cells/cytology
15.
Neuron ; 69(1): 61-76, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21220099

ABSTRACT

CXCL12/CXCR4 signaling is critical for cortical interneuron migration and their final laminar distribution. No information is yet available on CXCR7, a newly defined CXCL12 receptor. Here we demonstrated that CXCR7 regulated interneuron migration autonomously, as well as nonautonomously through its expression in immature projection neurons. Migrating cortical interneurons coexpressed Cxcr4 and Cxcr7, and Cxcr7(-/-) and Cxcr4(-/-) mutants had similar defects in interneuron positioning. Ectopic CXCL12 expression and pharmacological blockade of CXCR4 in Cxcr7(-/-) mutants showed that both receptors were essential for responding to CXCL12 during interneuron migration. Furthermore, live imaging revealed that Cxcr4(-/-) and Cxcr7(-/-) mutants had opposite defects in interneuron motility and leading process morphology. In vivo inhibition of Gα(i/o) signaling in migrating interneurons phenocopied the interneuron lamination defects of Cxcr4(-/-) mutants. On the other hand, CXCL12 stimulation of CXCR7, but not CXCR4, promoted MAP kinase signaling. Thus, we suggest that CXCR4 and CXCR7 have distinct roles and signal transduction in regulating interneuron movement and laminar positioning.


Subject(s)
Cell Movement/physiology , Interneurons/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Chemokine CXCL12/metabolism , Immunohistochemistry , Interneurons/physiology , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Receptors, CXCR/deficiency , Receptors, CXCR/genetics , Receptors, CXCR4/deficiency , Receptors, CXCR4/genetics
16.
Neuron ; 61(1): 42-56, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19146812

ABSTRACT

Radial glia are highly polarized cells that serve as neuronal progenitors and as scaffolds for neuronal migration during construction of the cerebral cortex. How radial glial cells establish and maintain their morphological polarity is unknown. Using conditional gene targeting in mice, we demonstrate that adenomatous polyposis coli (APC) serves an essential function in the maintenance of polarized radial glial scaffold during brain development. In the absence of APC, radial glial cells lose their polarity and responsiveness to the extracellular polarity maintenance cues, such as neuregulin-1. Elimination of APC further leads to marked instability of the radial glial microtubule cytoskeleton. The resultant changes in radial glial function and loss of APC in radial glial progeny lead to defective generation and migration of cortical neurons, severely disrupted cortical layer formation, and aberrant axonal tract development. Thus, APC is an essential regulator of radial glial polarity and is critical for the construction of cerebral cortex in mammals.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Polarity , Cerebral Cortex , Neuroglia , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Movement/physiology , Cell Proliferation , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Mice, Knockout , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neurogenesis/physiology , Neuroglia/cytology , Neuroglia/physiology , beta Catenin/genetics , beta Catenin/metabolism
17.
Development ; 131(24): 6023-31, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15537685

ABSTRACT

We show that alpha3 integrin mutation disrupts distinct aspects of neuronal migration and placement in the cerebral cortex. The preplate develops normally in alpha3 integrin mutant mice. However, time lapse imaging of migrating neurons in embryonic cortical slices indicates retarded radial and tangential migration of neurons, but not ventricular zone-directed migration. Examination of the actin cytoskeleton of alpha3 integrin mutant cortical cells reveals aberrant actin cytoskeletal dynamics at the leading edges. Deficits are also evident in the ability of developing neurons to probe their cellular environment with filopodial and lamellipodial activity. Calbindin or calretinin positive upper layer neurons as well as the deep layer neurons of alpha3 integrin mutant mice expressing EGFP were misplaced. These results suggest that alpha3beta1 integrin deficiency impairs distinct patterns of neuronal migration and placement through dysregulated actin dynamics and defective ability to search and respond to migration modulating cues in the developing cortex.


Subject(s)
Actins/metabolism , Cell Movement/physiology , Cerebral Cortex/embryology , Integrin alpha3beta1/metabolism , Neurons/cytology , Animals , Cerebral Cortex/metabolism , Mice , Mice, Neurologic Mutants , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL