Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Proc Natl Acad Sci U S A ; 121(16): e2317783121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38588430

ABSTRACT

GABAergic inhibitory interneurons, originating from the embryonic ventral forebrain territories, traverse a convoluted migratory path to reach the neocortex. These interneuron precursors undergo sequential phases of tangential and radial migration before settling into specific laminae during differentiation. Here, we show that the developmental trajectory of FoxG1 expression is dynamically controlled in these interneuron precursors at critical junctures of migration. By utilizing mouse genetic strategies, we elucidate the pivotal role of precise changes in FoxG1 expression levels during interneuron specification and migration. Our findings underscore the gene dosage-dependent function of FoxG1, aligning with clinical observations of FOXG1 haploinsufficiency and duplication in syndromic forms of autism spectrum disorders. In conclusion, our results reveal the finely tuned developmental clock governing cortical interneuron development, driven by temporal dynamics and the dose-dependent actions of FoxG1.


Subject(s)
Cerebral Cortex , Neocortex , Mice , Animals , Cerebral Cortex/metabolism , Cell Movement/physiology , Neurogenesis/physiology , Interneurons/physiology , Biomarkers/metabolism , GABAergic Neurons/physiology
2.
Development ; 145(1)2018 01 09.
Article in English | MEDLINE | ID: mdl-29229772

ABSTRACT

During forebrain development, a telencephalic organizer called the cortical hem is crucial for inducing hippocampal fate in adjacent cortical neuroepithelium. How the hem is restricted to its medial position is therefore a fundamental patterning issue. Here, we demonstrate that Foxg1-Lhx2 interactions are crucial for the formation of the hem. Loss of either gene causes a region of the cortical neuroepithelium to transform into hem. We show that FOXG1 regulates Lhx2 expression in the cortical primordium. In the absence of Foxg1, the presence of Lhx2 is sufficient to suppress hem fate, and hippocampal markers appear selectively in Lhx2-expressing regions. FOXG1 also restricts the temporal window in which loss of Lhx2 results in a transformation of cortical primordium into hem. Therefore, Foxg1 and Lhx2 form a genetic hierarchy in the spatiotemporal regulation of cortical hem specification and positioning, and together ensure the normal development of this hippocampal organizer.


Subject(s)
Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation, Developmental/physiology , Hippocampus/embryology , LIM-Homeodomain Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Telencephalon/embryology , Transcription Factors/biosynthesis , Animals , Forkhead Transcription Factors/genetics , LIM-Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Transcription Factors/genetics
3.
J Neurosci ; 35(37): 12869-89, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377473

ABSTRACT

Neurogliaform (RELN+) and bipolar (VIP+) GABAergic interneurons of the mammalian cerebral cortex provide critical inhibition locally within the superficial layers. While these subtypes are known to originate from the embryonic caudal ganglionic eminence (CGE), the specific genetic programs that direct their positioning, maturation, and integration into the cortical network have not been elucidated. Here, we report that in mice expression of the transcription factor Prox1 is selectively maintained in postmitotic CGE-derived cortical interneuron precursors and that loss of Prox1 impairs the integration of these cells into superficial layers. Moreover, Prox1 differentially regulates the postnatal maturation of each specific subtype originating from the CGE (RELN, Calb2/VIP, and VIP). Interestingly, Prox1 promotes the maturation of CGE-derived interneuron subtypes through intrinsic differentiation programs that operate in tandem with extrinsically driven neuronal activity-dependent pathways. Thus Prox1 represents the first identified transcription factor specifically required for the embryonic and postnatal acquisition of CGE-derived cortical interneuron properties. SIGNIFICANCE STATEMENT: Despite the recognition that 30% of GABAergic cortical interneurons originate from the caudal ganglionic eminence (CGE), to date, a specific transcriptional program that selectively regulates the development of these populations has not yet been identified. Moreover, while CGE-derived interneurons display unique patterns of tangential and radial migration and preferentially populate the superficial layers of the cortex, identification of a molecular program that controls these events is lacking.Here, we demonstrate that the homeodomain transcription factor Prox1 is expressed in postmitotic CGE-derived cortical interneuron precursors and is maintained into adulthood. We found that Prox1 function is differentially required during both embryonic and postnatal stages of development to direct the migration, differentiation, circuit integration, and maintenance programs within distinct subtypes of CGE-derived interneurons.


Subject(s)
Cerebral Cortex/cytology , GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Interneurons/cytology , Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Tumor Suppressor Proteins/physiology , Animals , Biomarkers , Calbindin 2/analysis , Cell Adhesion Molecules, Neuronal/analysis , Cell Lineage , Cell Movement , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Extracellular Matrix Proteins/analysis , GABAergic Neurons/metabolism , Gene Expression Profiling , Interneurons/classification , Interneurons/metabolism , Mice , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Reelin Protein , Serine Endopeptidases/analysis , Tumor Suppressor Proteins/deficiency , Vasoactive Intestinal Peptide/analysis
4.
J Neurosci ; 34(17): 5788-99, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760839

ABSTRACT

The olfactory bulb (OB) is one of the two major loci in the mammalian brain where newborn neurons are constantly integrated into the neural circuit during postnatal life. Newborn neurons are generated from neural stem cells in the subventricular zone (SVZ) of the lateral ventricle and migrate to the OB through the rostral migratory stream. The majority of these newborn neurons differentiate into inhibitory interneurons, such as granule cells and periglomerular cells. It has been reported that prolonged supply of newborn neurons leads to continuous addition/turnover of the interneuronal populations and contributes to functional integrity of the OB circuit. However, it is not still clear how and to what extent postnatal-born neurons contribute to OB neural circuit formation, and the functional role of postnatal neurogenesis in odor-related behaviors remains elusive. To address this question, here by using genetic strategies, we first determined the unique integration mode of newly born interneurons during postnatal development of the mouse OB. We then manipulated these interneuron populations and found that continuous postnatal neurogenesis in the SVZ-OB plays pivotal roles in flexible olfactory associative learning and memory.


Subject(s)
Association Learning/physiology , Nerve Net/physiology , Neurogenesis/physiology , Neurons/physiology , Olfactory Bulb/physiology , Olfactory Perception/physiology , Animals , Cerebral Ventricles/cytology , Cerebral Ventricles/physiology , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Dentate Gyrus/physiology , Interneurons/cytology , Interneurons/physiology , Male , Mice , Mice, Transgenic , Nerve Net/cytology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurons/cytology , Odorants , Olfactory Bulb/cytology , Olfactory Bulb/growth & development
5.
J Neurosci ; 32(19): 6688-98, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22573691

ABSTRACT

During early postnatal development, neuronal networks successively produce various forms of spontaneous patterned activity that provide key signals for circuit maturation. Initially, in both rodent hippocampus and neocortex, coordinated activity emerges in the form of synchronous plateau assemblies (SPAs) that are initiated by sparse groups of gap-junction-coupled oscillating neurons. Subsequently, SPAs are replaced by synapse-driven giant depolarizing potentials (GDPs). Whether these sequential changes in mechanistically distinct network activities correlate with modifications in single-cell properties is unknown. To determine this, we studied the morphophysiological fate of single SPA cells as a function of development. We focused on CA3 GABAergic interneurons, which are centrally involved in generating GDPs in the hippocampus. As the network matures, GABAergic neurons are engaged more in GDPs and less in SPAs. Using inducible genetic fate mapping, we show that the individual involvement of GABAergic neurons in SPAs is correlated to their temporal origin. In addition, we demonstrate that the SPA-to-GDP transition is paralleled by a remarkable maturation in the morphophysiological properties of GABAergic neurons. Compared with those involved in GDPs, interneurons participating in SPAs possess immature intrinsic properties, receive synaptic inputs spanning a wide amplitude range, and display large somata as well as membrane protrusions. Thus, a developmental switch in the morphophysiological properties of GABAergic interneurons as they progress from SPAs to GDPs marks the emergence of synapse-driven network oscillations.


Subject(s)
Hippocampus/growth & development , Interneurons/cytology , Interneurons/physiology , Nerve Net/growth & development , Animals , Animals, Newborn , Female , Gene Knock-In Techniques , Hippocampus/cytology , Male , Mice , Mice, Transgenic , Nerve Net/cytology , Organ Culture Techniques
6.
Cereb Cortex ; 21(4): 845-52, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20732898

ABSTRACT

It is of considerable interest to determine how diverse subtypes of γ-aminobutyric acidergic (GABAergic) interneurons integrate into the functional network of the cerebral cortex. Using inducible in vivo genetic fate mapping approaches, we found that interneuron precursors arising from the medial ganglionic eminence (MGE) and caudal ganglionic eminence (CGE) at E12.5, respectively, populate deep and superficial cortical layers in a complementary manner in the mature cortex. These age-matched populations initiate tangential migration into the cortex simultaneously, migrate above and below the cortical plate in a similar ratio, and complete their entrance into the cortical plate by P1. Surprisingly, while these 2 interneuron populations show a comparable layer distribution at P1, they subsequently segregate into distinct cortical layers. In addition, the initiation of the radial sorting within each lineage coincided well with the upregulation of the potassium/chloride cotransporter KCC2. Moreover, layer sorting of a later born (E16.5) CGE-derived population occurred with a similar time course to the earlier born E12.5 cohorts, further suggesting that this segregation step is controlled in a subtype specific manner. We conclude that radial sorting within the early postnatal cortex is a key mechanism by which the layer-specific integration of GABAergic interneurons into the emerging cortical network is achieved.


Subject(s)
Cell Lineage , Cell Movement/physiology , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Interneurons/cytology , Neurogenesis/physiology , Animals , Cell Differentiation/physiology , Immunohistochemistry , Interneurons/metabolism , Mice , Mice, Transgenic , gamma-Aminobutyric Acid/metabolism
7.
Mol Brain ; 15(1): 37, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35484559

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) /p35 is involved in many developmental processes of the central nervous system. Cdk5/p35 is also implicated in synaptic plasticity, learning and memory. Several lines of conditional Cdk5 knockout mice (KO) have been generated and have shown different outcomes for learning and memory. Here, we present our analysis of p35 conditional KO mice (p35cKO) in hippocampal pyramidal neurons or forebrain GABAergic neurons using electrophysiological and behavioral methods. In the fear conditioning task, CamKII-p35cKO mice showed impaired memory retention. Furthermore, NMDAR-dependent long-term depression (LTD) induction by low-frequency stimuli in hippocampal slices from CamkII-p35cKO mice was impaired compared to that in control mice. In contrast, Dlx-p35cKO mice showed no abnormalities in behavioral tasks and electrophysiological analysis in their hippocampal slices. These results indicated that Cdk5/p35 in excitatory neurons is important for the hippocampal synaptic plasticity and associative memory retention.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cyclin-Dependent Kinase 5 , Nerve Tissue Proteins/metabolism , Animals , GABAergic Neurons , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology
8.
J Neurosci ; 30(6): 2165-76, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20147544

ABSTRACT

GABAergic interneurons critically regulate cortical computation through exquisite spatiotemporal control over excitatory networks. Precision of this inhibitory control requires a remarkable diversity within interneuron populations that is largely specified during embryogenesis. Although interneurons expressing the neuronal isoform of nitric oxide synthase (nNOS) constitute the largest hippocampal interneuron cohort their origin and specification remain unknown. Thus, as neurogliaform cells (NGC) and Ivy cells (IvC) represent the main nNOS(+) interneurons, we investigated their developmental origins. Although considered distinct interneuron subtypes, NGCs and IvCs exhibited similar neurochemical and electrophysiological signatures, including NPY expression and late spiking. Moreover, lineage analyses, including loss-of-function experiments and inducible fate-mapping, indicated that nNOS(+) IvCs and NGCs are both derived from medial ganglionic eminence (MGE) progenitors under control of the transcription factor Nkx2-1. Surprisingly, a subset of NGCs lacking nNOS arises from caudal ganglionic eminence (CGE) progenitors. Thus, while nNOS(+) NGCs and IvCs arise from MGE progenitors, a CGE origin distinguishes a discrete population of nNOS(-) NGCs.


Subject(s)
Hippocampus/cytology , Interneurons/cytology , Interneurons/physiology , Nitric Oxide Synthase Type I/biosynthesis , Action Potentials , Animals , Cell Lineage , Cell Polarity , Hippocampus/enzymology , Immunohistochemistry , Interneurons/enzymology , Male , Mice , Mice, Transgenic , Neuropeptide Y/biosynthesis , Nuclear Proteins/physiology , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/physiology , Telencephalon/cytology , Thyroid Nuclear Factor 1 , Transcription Factors/physiology , Vasoactive Intestinal Peptide/biosynthesis
9.
J Neurosci ; 30(5): 1582-94, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-20130169

ABSTRACT

By combining an inducible genetic fate mapping strategy with electrophysiological analysis, we have systematically characterized the populations of cortical GABAergic interneurons that originate from the caudal ganglionic eminence (CGE). Interestingly, compared with medial ganglionic eminence (MGE)-derived cortical interneuron populations, the initiation [embryonic day 12.5 (E12.5)] and peak production (E16.5) of interneurons from this embryonic structure occurs 3 d later in development. Moreover, unlike either pyramidal cells or MGE-derived cortical interneurons, CGE-derived interneurons do not integrate into the cortex in an inside-out manner but preferentially (75%) occupy superficial cortical layers independent of birthdate. In contrast to previous estimates, CGE-derived interneurons are both considerably greater in number (approximately 30% of all cortical interneurons) and diversity (comprised by at least nine distinct subtypes). Furthermore, we found that a large proportion of CGE-derived interneurons, including the neurogliaform subtype, express the glycoprotein Reelin. In fact, most CGE-derived cortical interneurons express either Reelin or vasoactive intestinal polypeptide. Thus, in conjunction with previous studies, we have now determined the spatial and temporal origins of the vast majority of cortical interneuron subtypes.


Subject(s)
Cell Lineage/genetics , Cerebral Cortex/cytology , Genetic Techniques , Interneurons/cytology , Animals , Body Patterning/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation/genetics , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Extracellular Matrix Proteins/metabolism , Genetic Markers , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Prosencephalon/cytology , Prosencephalon/physiology , Reelin Protein , Serine Endopeptidases/metabolism
10.
Nat Commun ; 12(1): 3773, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145239

ABSTRACT

Abnormalities in GABAergic inhibitory circuits have been implicated in the aetiology of autism spectrum disorder (ASD). ASD is caused by genetic and environmental factors. Several genes have been associated with syndromic forms of ASD, including FOXG1. However, when and how dysregulation of FOXG1 can result in defects in inhibitory circuit development and ASD-like social impairments is unclear. Here, we show that increased or decreased FoxG1 expression in both excitatory and inhibitory neurons results in ASD-related circuit and social behavior deficits in our mouse models. We observe that the second postnatal week is the critical period when regulation of FoxG1 expression is required to prevent subsequent ASD-like social impairments. Transplantation of GABAergic precursor cells prior to this critical period and reduction in GABAergic tone via Gad2 mutation ameliorates and exacerbates circuit functionality and social behavioral defects, respectively. Our results provide mechanistic insight into the developmental timing of inhibitory circuit formation underlying ASD-like phenotypes in mouse models.


Subject(s)
Autism Spectrum Disorder/genetics , Brain/growth & development , Forkhead Transcription Factors/genetics , GABAergic Neurons/cytology , Nerve Tissue Proteins/genetics , Social Behavior , Animals , Brain/physiology , Disease Models, Animal , GABAergic Neurons/transplantation , Glutamate Decarboxylase/genetics , Mice
11.
J Neurosci ; 29(50): 15933-46, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016109

ABSTRACT

In the developing mammalian basal telencephalon, neural progenitors from the subpallium generate the majority of inhibitory medium spiny neurons (MSNs) in the striatum, while both pallial- and subpallial-derived progenitors contribute to excitatory and inhibitory neuronal diversity in the amygdala. Using a combination of approaches, including genetic fate mapping, cell birth dating, cell migration assays, and electrophysiology, we find that cells derived from the Emx1 lineage contribute to two distinct neuronal populations in the mature basal forebrain: inhibitory MSNs in the striatum and functionally distinct subclasses of excitatory neurons in the amygdala. Our cell birth-dating studies reveal that these two populations are born at different times during early neurogenesis, with the amygdala population born before the MSNs. In the striatum, Emx1-lineage neurons represent a unique subpopulation of MSNs: they are disproportionately localized to the dorsal striatum, are found in dopamine receiving, reelin-positive patches, and are born throughout striatal neurogenesis. In addition, our data suggest that a subpopulation of these Emx1-lineage cells originate in the pallium and subsequently migrate to the developing striatum and amygdala. Our intersectional fate-mapping analysis further reveals that Emx1-lineage cells that coexpress Dlx exclusively generate MSNs but do not contribute to the excitatory neurons in the amygdala. Thus, both the timing of neurogenesis and differential combinatorial gene expression appear to be key determinants of striatal versus amygdala fate decisions of Emx1-lineage cells.


Subject(s)
Amygdala/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Corpus Striatum/physiology , Homeodomain Proteins/physiology , Stem Cells/physiology , Transcription Factors/physiology , Amygdala/cytology , Amygdala/embryology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Corpus Striatum/cytology , Corpus Striatum/embryology , Female , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/classification , Neurons/cytology , Neurons/physiology , Pregnancy , Reelin Protein , Stem Cells/classification , Stem Cells/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics
12.
Cereb Cortex ; 19 Suppl 1: i1-10, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19363146

ABSTRACT

Ventral telencephalic progenitors expressing the homeodomain transcription factor Nkx6-2 have been shown to give rise to a multitude of cortical interneuron subtypes usually associated with origin in either the medial ganglionic eminence or the caudal ganglionic eminence. The function of Nkx6-2 in directing the fate of those progenitors has, however, not been thoroughly analyzed. We used a combination of genetic inducible fate mapping and in vivo loss-of-function to analyze the requirement of Nkx6-2 in determining the fate of cortical interneurons. We have found that interneuron subtypes are born with a characteristic temporal pattern. Furthermore, we extend the characterization of interneurons from the Nkx6-2 lineage through the application of electrophysiological methods. Analysis of these populations in Nkx6-2 null mice suggests that there is a small and partially penetrant loss of delayed non-fast spiking somatostatin/calretinin double positive cortical interneurons in the absence of Nkx6-2 gene function.


Subject(s)
Homeodomain Proteins/metabolism , Interneurons/cytology , Interneurons/physiology , Neocortex/cytology , Neocortex/growth & development , Transcription Factors/metabolism , Animals , Mice , Mice, Knockout , Tissue Distribution
13.
Neurosci Res ; 138: 26-32, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30227162

ABSTRACT

GABAergic interneurons in the neocortex play pivotal roles in the feedforward and feedback inhibition that control higher order information processing and thus, malfunction in the inhibitory circuits often leads to neurodevelopmental disorders. Very interestingly, a large diversity of morphology, synaptic targeting specificity, electrophysiological properties and molecular expression profiles are found in cortical interneurons, which originate within the distantly located embryonic ganglionic eminences. Here, I will review the still ongoing effort to understand the developmental trajectories of GABAergic cortical interneuron subtypes.


Subject(s)
GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/physiology , Interneurons/physiology , Neocortex/growth & development , Neurogenesis/physiology , Animals
14.
Nat Commun ; 10(1): 3581, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31395862

ABSTRACT

The bimodal requisite for a genetic program and external stimuli is a key feature of sensory circuit formation. However, the contribution of cell-intrinsic codes to directing sensory-specific circuits remains unknown. Here, we identify the earliest molecular program that preselects projection neuron types in the sensory neocortex. Mechanistically, Foxg1 binds to an H3K4me1-enriched enhancer site to repress COUP-TFI, where ectopic acquisition of Foxg1 in layer 4 cells transforms local projection neurons to callosal projection neurons with pyramidal morphologies. Removal of Foxg1 in long-range projection neurons, in turn, derepresses COUP-TFI and activates a layer 4 neuron-specific program. The earliest segregation of projection subtypes is achieved through repression of Foxg1 in layer 4 precursors by early growth response genes, the major targets of the transforming growth factor-ß signaling pathway. These findings describe the earliest cortex-intrinsic program that restricts neuronal connectivity in sensory circuits, a fundamental step towards the acquisition of mammalian perceptual behavior.


Subject(s)
Gene Expression Regulation/physiology , Neocortex/physiology , Nerve Net/physiology , Neurons/metabolism , Perception/physiology , Animals , COUP Transcription Factor I/metabolism , Early Growth Response Transcription Factors/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Male , Mice , Mice, Knockout , Neocortex/cytology , Nerve Net/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
15.
J Neurosci ; 27(29): 7786-98, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17634372

ABSTRACT

Inhibitory GABAergic interneurons of the mouse neocortex are a highly heterogeneous population of neurons that originate from the ventral telencephalon and migrate tangentially up into the developing cortical plate. The majority of cortical interneurons arise from a transient embryonic structure known as the medial ganglionic eminence (MGE), but how the remarkable diversity is specified in this region is not known. We have taken a genetic fate mapping strategy to elucidate the temporal origins of cortical interneuron subtypes within the MGE. We used an inducible form of Cre under the regulation of Olig2, a basic helix-loop-helix transcription factor highly expressed in neural progenitors of the MGE. We observe that the physiological subtypes of cortical interneurons are, to a large degree, unique to their time point of generation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement/physiology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Interneurons/physiology , Nerve Tissue Proteins/metabolism , Stem Cells/physiology , Action Potentials/physiology , Age Factors , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Patterning/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Interneurons/classification , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Patch-Clamp Techniques/methods , Telencephalon/cytology
16.
Curr Opin Neurobiol ; 16(5): 577-84, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16971113

ABSTRACT

Recent advances in molecular genetics have produced many novel strategies for directing the expression of both functional and regulatory elements in transgenic mice. With the application of such approaches, the specific populations that comprise CNS networks can be both visualized and manipulated. Transgenic methods now range from the use of specific enhancer elements and large genomic regions assembled using BACs and PACs, to the use of gene targeting to a specific locus. In addition, the advent of transactivators and site-specific recombinases has provided unprecedented spatial and temporal control for directing expression in the CNS using a combination of appropriate alleles. As a result, the promise of being able to use transgenics to target specific neuronal populations is now being realized.


Subject(s)
Central Nervous System/physiology , Gene Expression Profiling/methods , Gene Expression , Neurons/physiology , Animals , Mice , Mice, Transgenic , Transgenes
17.
PLoS One ; 13(5): e0198268, 2018.
Article in English | MEDLINE | ID: mdl-29847590

ABSTRACT

Loss or gain of copy number of the gene encoding the transcription factor methyl-CpG-binding protein 2 (MeCP2) leads to neurodevelopmental disorders (Rett and MeCP2 duplication syndrome), indicating that precisely regulated MeCP2 expression during development is critical for mental health. Consistent with this idea, MeCP2 null mutants exhibit synaptic regression in the dorsal lateral geniculate nucleus (dLGN), the visual relay center in the thalamus, a phenotype resembling that of animals reared in the dark during the visual sensitive period. It remains unclear how MeCP2 expression is regulated during circuit formation and maturation, especially in excitatory and inhibitory populations of neurons. We found that, concomitant with the initiation of the dark-rearing sensitive period, MeCP2 protein levels were elevated in glutamatergic but not GABAergic neurons of the dLGN. Moreover, MeCP2 expression in glutamatergic populations was selectively reduced by dark-rearing. Therefore, we propose that visual experience-dependent MeCP2 induction in glutamatergic populations is essential for synaptic maturation within the dLGN.


Subject(s)
Gene Expression Regulation , Methyl-CpG-Binding Protein 2/metabolism , Thalamus/cytology , Visual Cortex/cytology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Up-Regulation , Visual Cortex/physiology
18.
J Neurosci ; 24(14): 3672-82, 2004 Apr 07.
Article in English | MEDLINE | ID: mdl-15071116

ABSTRACT

Neuronal subtype specification depends on multiple transcription factors such as basic helix-loop-helix (bHLH) factors. However, transcription factor codes for most neurons remain to be determined. Here, we report identification of a novel mouse bHLH factor, termed Heslike, that has Hes1-like bHLH domain and transcriptional repressor activity. Heslike is coexpressed with the bHLH factor Mash1 in brain regions that give rise to GABAergic neurons. In the mesencephalon and the caudal diencephalon, coexpression of Heslike and Mash1 is initially restricted to small regions but expanded dorsally from embryonic day 9.5 onward, and this expansion of coexpression is followed by GABAergic neurogenesis. Misexpression of Heslike in mouse embryos generates ectopic GABAergic neurons only from the Mash1(+) region. In contrast, in the mesencephalon and the caudal diencephalon of Mash1-null mice, GABAergic neurons are almost completely missing and, instead, other neurons are generated, although Heslike is still expressed. Furthermore, coexpression of Heslike and Mash1 significantly promotes formation of GABAergic neurons, compared with each gene alone, in neural precursor cell culture. Thus, Heslike or Mash1 alone is not sufficient, but their coexpression may be important for generation of GABAergic neurons. These results suggest that combinations of distinct bHLH factors promote formation of distinct neuronal subtypes, thereby increasing neuronal diversity.


Subject(s)
Brain/metabolism , Helix-Loop-Helix Motifs/genetics , Neurons/metabolism , Repressor Proteins/genetics , gamma-Aminobutyric Acid/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Brain/cytology , Brain/embryology , Cell Lineage/genetics , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Diencephalon/cytology , Diencephalon/embryology , Diencephalon/metabolism , Gene Expression Regulation, Developmental/physiology , Humans , Mesencephalon/cytology , Mesencephalon/embryology , Mesencephalon/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Neurons/cytology , Rats , Repressor Proteins/biosynthesis , Repressor Proteins/isolation & purification , Sequence Alignment , Stem Cells/cytology , Stem Cells/metabolism , Takifugu , Transcription Factors/biosynthesis , Transcription Factors/genetics , Zebrafish
19.
Neuron ; 74(6): 1045-58, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22726835

ABSTRACT

Pyramidal cells of the cerebral cortex are born in the ventricular zone and migrate through the intermediate zone to enter into the cortical plate. In the intermediate zone, these migrating precursors move tangentially and initiate the extension of their axons by transiently adopting a characteristic multipolar morphology. We observe that expression of the forkhead transcription factor FoxG1 is dynamically regulated during this transitional period. By utilizing conditional genetic strategies, we show that the downregulation of FoxG1 at the beginning of the multipolar cell phase induces Unc5D expression, the timing of which ultimately determines the laminar identity of pyramidal neurons. In addition, we demonstrate that the re-expression of FoxG1 is required for cells to transit out of the multipolar cell phase and to enter into the cortical plate. Thus, the dynamic expression of FoxG1 during migration within the intermediate zone is essential for the proper assembly of the cerebral cortex.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/cytology , Forkhead Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Pyramidal Cells/cytology , Animals , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Mice , Pyramidal Cells/metabolism
20.
Neurosci Res ; 73(1): 85-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22343123

ABSTRACT

Reporters of Cre and/or Flp activity are important for defining the spatial and temporal extent of Cre/Flp-mediated recombination. Here, we describe R26-CAG-LF-mTFP1, a multifunctional fluorescent reporter mouse that strongly expresses mTFP1 (bright teal fluorescent protein) after Cre- and Flp-mediated recombination. To meet the need for single recombinase-mediated reporter expression, we generated derivatives of R26-CAG-LF-mTFP1. The germline excision of the Frt-flanked stop cassette in R26-CAG-LF-mTFP1 generated a Cre-dependent reporter (R26-CAG-LoxP-mTFP1). Similarly, R26-CAG-FRT-mTFP1, in which the loxP-flanked stop cassette was excised in the germline, requires only Flp to activate mTFP1 expression.


Subject(s)
DNA Nucleotidyltransferases/physiology , Genes, Reporter/physiology , Green Fluorescent Proteins/physiology , Integrases/physiology , Proteins/physiology , Recombination, Genetic/physiology , Animals , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Untranslated
SELECTION OF CITATIONS
SEARCH DETAIL