Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Dev Cell ; 58(14): 1221-1236.e7, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37290446

ABSTRACT

In developing brains, activity-dependent remodeling facilitates the formation of precise neuronal connectivity. Synaptic competition is known to facilitate synapse elimination; however, it has remained unknown how different synapses compete with one another within a post-synaptic cell. Here, we investigate how a mitral cell in the mouse olfactory bulb prunes all but one primary dendrite during the developmental remodeling process. We find that spontaneous activity generated within the olfactory bulb is essential. We show that strong glutamatergic inputs to one dendrite trigger branch-specific changes in RhoA activity to facilitate the pruning of the remaining dendrites: NMDAR-dependent local signals suppress RhoA to protect it from pruning; however, the subsequent neuronal depolarization induces neuron-wide activation of RhoA to prune non-protected dendrites. NMDAR-RhoA signals are also essential for the synaptic competition in the mouse barrel cortex. Our results demonstrate a general principle whereby activity-dependent lateral inhibition across synapses establishes a discrete receptive field of a neuron.


Subject(s)
Dendrites , Olfactory Bulb , Dendrites/physiology , Olfactory Bulb/physiology , Synapses/physiology , Neurons , Cell Differentiation
2.
Sci Rep ; 12(1): 17145, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229519

ABSTRACT

mRNA vaccines for SARS-CoV-2 have been widely used and saving millions of people in the world. How efficiently proteins are produced from exogenous mRNAs in the embryonic brain, however, is less known. Here we show that protein expression occurs highly efficiently in neural stem cells, in a very narrow time window after mRNA electroporation in the embryonic mouse brain, where plasmids have been successfully transfected. Protein expression is detected 1 h and 12 h after the electroporation of mRNAs and plasmids, respectively. The delivery of exogenous mRNAs may be useful for not only vaccines but also functional analysis in the brain.


Subject(s)
COVID-19 Vaccines , COVID-19 , Animals , Brain/metabolism , COVID-19/genetics , Electroporation , Humans , Mice , Plasmids , RNA, Messenger/genetics , RNA, Messenger/metabolism , SARS-CoV-2
3.
Front Neuroanat ; 15: 669631, 2021.
Article in English | MEDLINE | ID: mdl-34054439

ABSTRACT

Schizophrenia is a devastating neuropsychiatric disease with a globally 1% life-long prevalence. Clinical studies have linked Zswim6 mutations to developmental and neurological diseases, including schizophrenia. Zswim6's function remains largely unknown. Given the involvement of Zswim6 in schizophrenia and schizophrenia as a neurodevelopmental disease, it is important to understand the spatiotemporal expression pattern of Zswim6 in the developing brain. Here, we performed a comprehensive analysis of the spatiotemporal expression pattern of Zswim6 in the mouse forebrain by in situ hybridization with radioactive and non-radioactive-labeled riboprobes. Zswim6 mRNA was detected as early as E11.5 in the ventral forebrain. At E11.5-E13.5, Zswim6 was highly expressed in the lateral ganglionic eminence (LGE). The LGE consisted of two progenitor populations. Dlx+;Er81+ cells in dorsal LGE comprised progenitors of olfactory bulb interneurons, whereas Dlx+;Isl1+ progenitors in ventral LGE gave rise to striatal projection neurons. Zswim6 was not colocalized with Er81 in the dorsal LGE. In the ventral LGE, Zswim6 was colocalized with striatal progenitor marker Nolz-1. Zswim6 was highly expressed in the subventricular zone (SVZ) of LGE in which progenitors undergo the transition from proliferation to differentiation. Double labeling showed that Zswim6 was not colocalized with proliferation marker Ki67 but was colocalized with differentiation marker Tuj1 in the SVZ, suggesting Zswim6 expression in early differentiating neurons. Zswim6 was also expressed in the adjacent structures of medial and caudal ganglionic eminences (MGE, CGE) that contained progenitors of cortical interneurons. At E15.5 and E17.5, Zswim6 was expressed in several key brain regions that were involved in the pathogenesis of schizophrenia, including the striatum, cerebral cortex, hippocampus, and medial habenular nucleus. Zswim6 was persistently expressed in the postnatal brain. Cell type analysis indicated that Zswim6 mRNA was colocalized with D1R-expressing striatonigral and D2R-expressing striatopallidal neurons of the adult striatum with a higher colocalization in striatopallidal neurons. These findings are of particular interest as striatal dopamine D2 receptors are known to be involved in the pathophysiology of schizophrenia. In summary, the comprehensive analysis provides an anatomical framework for the study of Zswim6 function and Zswim6-associated neurological disorders.

4.
J Comp Neurol ; 528(14): 2404-2419, 2020 10.
Article in English | MEDLINE | ID: mdl-32144752

ABSTRACT

GABAergic interneurons play an essential role in modulating cortical networks. The progenitor domains of cortical interneurons are localized in developing ventral forebrain, including the medial ganglionic eminence (MGE), caudal ganglionic eminence (CGE), preoptic area (POA), and preoptic hypothalamic border domain (POH). Here, we characterized the expression pattern of Zswim5, an MGE-enriched gene in the mouse forebrain. At E11.5-E13.5, prominent Zswim5 expression was detected in the subventricular zone (SVZ) of MGE, POA, and POH, but not CGE of ventral telencephalon where progenitors of cortical interneurons resided. At E15.5 and E17.5, Zswim5 expression remained in the MGE/pallidum primordium and ventral germinal zone. Zswim5 mRNA was markedly decreased after birth and was absent in the adult forebrain. Interestingly, the Zswim5 expression pattern resembled the tangential migration pathways of cortical interneurons. Zswim5-positive cells in the MGE appeared to migrate from the MGE through the SVZ of LGE to overlying neocortex. Indeed, Zswim5 was co-localized with Nkx2.1 and Lhx6, markers of progenitors and migratory cortical interneurons. Double labeling showed that Ascl1/Mash1-positive cells co-expressed Zswim5. Zswim5 expressing cells contained none or at most low levels of Ki67 but co-expressed Tuj1 in the SVZ of MGE. These results suggest that Zswim5 is immediately upregulated as progenitors exiting cell cycle become postmitotic. Given that recent studies have elucidated that the cell fate of cortical interneurons is determined shortly after becoming postmitotic, the timing of Zswim5 expression in early postmitotic interneurons suggests a potential role of Zswim5 in regulation of neurogenesis and tangential migration of cortical interneurons.


Subject(s)
Interneurons/metabolism , Neurogenesis/physiology , Prosencephalon/metabolism , Transcription Factors/metabolism , Animals , Cell Movement/physiology , Mice , Neural Stem Cells/metabolism , Prosencephalon/cytology , Transcriptome , Zinc Fingers/physiology
5.
Adv Exp Med Biol ; 1218: 93-101, 2020.
Article in English | MEDLINE | ID: mdl-32060873

ABSTRACT

Notch signaling is required for maintaining neural stem cells (NSCs) in the developing brain. NSCs have potential to give rise to many neuronal types in the early telencephalon, and the potential decreases as embryonic development proceeds. Nepro, which encodes a unique nucleolar protein and is activated downstream of Notch, is essential for maintaining NSCs in the early telencephalon. Nepro is also expressed at basal levels and required for maintaining the preimplantation embryo, by repressing mitochondria-associated p53 apoptotic signaling. Notch signaling also controls dendritic complexity in mitral cells, major projection neurons in the olfactory bulb, showing that many steps of neural development involve Notch signaling.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Neurogenesis , Neurons/cytology , Neurons/metabolism , Receptors, Notch/metabolism
6.
Dev Growth Differ ; 61(2): 141-149, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30675906

ABSTRACT

Selective cell death by apoptosis plays important roles in organogenesis. Apoptotic cells are observed in the developmental and homeostatic processes of several ectodermal organs, such as hairs, feathers, and mammary glands. In chick feather development, apoptotic events have been observed during feather morphogenesis, but have not been investigated during early feather bud formation. Previously, we have reported a method for generating feather buds on a bioengineered skin from dissociated skin epithelial and mesenchymal cells in three-dimensional culture. During the development of the bioengineered skin, epithelial cavity formation by apoptosis was observed in the epithelial tissue. In this study, we examined the selective epithelial cell death during the bioengineered skin development. Histological analyses suggest that the selective epithelial cell death in the bioengineered skin was induced by caspase-3-related apoptosis. The formation of feather buds of the bioengineered skin was disturbed by the treatment with a pan-caspase inhibitor. The pan-caspase inhibitor treatment suppressed the rearrangement of the epithelial layer and the formation of dermal condensation, which are thought to be essential step to form feather buds. The suppression of the formation of feather buds on the pan-caspase inhibitor-treated skin was partially compensated by the addition of a GSK-3ß inhibitor, which activates Wnt/ß-catenin signaling. These results suggest that the epithelial cell death is involved in the formation of feather buds of the bioengineered skin. These observations also suggest that caspase activities and Wnt/ß-catenin signaling may contribute to the formation of epithelial and mesenchymal components in the bioengineered skin.


Subject(s)
Cell Death , Epithelial Cells/cytology , Feathers/cytology , Feathers/growth & development , Skin/cytology , Tissue Engineering , Animals , Cells, Cultured , Chickens , Skin/growth & development
7.
Dev Growth Differ ; 60(5): 291-299, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29785739

ABSTRACT

A Merkel cell-neurite complex is a touch receptor composed of specialized epithelial cells named Merkel cells and peripheral sensory nerves in the skin. Merkel cells are found in touch-sensitive skin components including whisker follicles. The nerve fibers that innervate Merkel cells of a whisker follicle extend from the maxillary branch of the trigeminal ganglion. Whiskers as a sensory organ attribute to the complicated architecture of the Merkel cell-neurite complex, and therefore it is intriguing how the structure is formed. However, observing the dynamic process of the formation of a Merkel cell-neurite complex in whiskers during embryonic development is still difficult. In this study, we tried to develop an organotypic co-culture method of a whisker pad and a trigeminal ganglion explant to form the Merkel cell-neurite complex in vitro. We initially developed two distinct culture methods of a single whisker row and a trigeminal ganglion explant, and then combined them. By dissecting and cultivating a single row from a whisker pad, the morphogenesis of whisker follicles could be observed under a microscope. After the co-cultivation of the whisker row with a trigeminal ganglion explant, a Merkel cell-neurite complex composed of Merkel cells, which were positive for both cytokeratin 8 and SOX2, Neurofilament-H-positive trigeminal nerve fibers and Schwann cells expressing Nestin, SOX2 and SOX10 was observed via immunohistochemical analyses. These results suggest that the process for the formation of a Merkel cell-neurite complex can be observed under a microscope using our organotypic co-culture method.


Subject(s)
Embryo, Mammalian/cytology , Merkel Cells/metabolism , Neurites/metabolism , Trigeminal Nerve/embryology , Vibrissae/embryology , Animals , Coculture Techniques , Embryo, Mammalian/metabolism , Immunohistochemistry , In Situ Hybridization , Merkel Cells/cytology , Mice , Trigeminal Ganglion
8.
Dev Growth Differ ; 59(9): 701-712, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29124740

ABSTRACT

The anterior part of the embryonic telencephalon gives rise to several brain regions that are important for animal behavior, including the frontal cortex (FC) and the olfactory bulb. The FC plays an important role in decision-making behaviors, such as social and cognitive behavior, and the olfactory bulb is involved in olfaction. Here, we show the organizing activity of fibroblast growth factor 8 (Fgf8) in the regionalization of the anterior telencephalon, specifically the FC and the olfactory bulb. Misexpression of Fgf8 in the most anterior part of the mouse telencephalon at embryonic day 11.5 (E11.5) by ex utero electroporation resulted in a lateral shift of dorsal FC subdivision markers and a lateral expansion of the dorsomedial part of the FC, the future anterior cingulate and prelimbic cortex. Fgf8-transfected brains had lacked ventral FC, including the future orbital cortex, which was replaced by the expanded olfactory bulb. The olfactory region occupied a larger area of the FC when transfection efficiency of Fgf8 was higher. These results suggest that Fgf8 regulates the proportions of the FC and olfactory bulb in the anterior telencephalon and has a medializing effect on the formation of FC subdivisions.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Telencephalon/metabolism , Animals , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Mice , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Telencephalon/embryology
9.
PLoS Genet ; 12(12): e1006514, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28027303

ABSTRACT

Mitral cells (MCs) of the mammalian olfactory bulb have a single primary dendrite extending into a single glomerulus, where they receive odor information from olfactory sensory neurons (OSNs). Molecular mechanisms for controlling dendritic arbors of MCs, which dynamically change during development, are largely unknown. Here we found that MCs displayed more complex dendritic morphologies in mouse mutants of Maml1, a crucial gene in Notch signaling. Similar phenotypes were observed by conditionally misexpressing a dominant negative form of MAML1 (dnMAML1) in MCs after their migration. Conversely, conditional misexpression of a constitutively active form of Notch reduced their dendritic complexity. Furthermore, the intracellular domain of Notch1 (NICD1) was localized to nuclei of MCs. These findings suggest that Notch signaling at embryonic stages is involved in the dendritic complexity of MCs. After the embryonic misexpression of dnMAML1, many MCs aberrantly extended dendrites to more than one glomerulus at postnatal stages, suggesting that Notch signaling is essential for proper formation of olfactory circuits. Moreover, dendrites in cultured MCs were shortened by Jag1-expressing cells. Finally, blocking the activity of Notch ligands in OSNs led to an increase in dendritic complexity as well as a decrease in NICD1 signals in MCs. These results demonstrate that the dendritic complexity of MCs is controlled by their presynaptic partners, OSNs.


Subject(s)
Nuclear Proteins/genetics , Olfactory Bulb/metabolism , Olfactory Receptor Neurons/metabolism , Receptor, Notch1/genetics , Transcription Factors/genetics , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation, Developmental , Jagged-1 Protein/biosynthesis , Jagged-1 Protein/genetics , Mice , Nuclear Proteins/biosynthesis , Olfactory Bulb/growth & development , Olfactory Receptor Neurons/cytology , Receptor, Notch1/biosynthesis , Signal Transduction/genetics , Transcription Factors/biosynthesis
10.
eNeuro ; 3(5)2016.
Article in English | MEDLINE | ID: mdl-27785463

ABSTRACT

The glomerular map in the olfactory bulb (OB) is the basis for odor recognition. Once established during development, the glomerular map is stably maintained throughout the life of an animal despite the continuous turnover of olfactory sensory neurons (OSNs). However, traumatic damage to OSN axons in the adult often leads to dysosmia, a qualitative and quantitative change in olfaction in humans. A mouse model of dysosmia has previously indicated that there is an altered glomerular map in the OB after the OSN axon injury; however, the underlying mechanisms that cause the map distortion remain unknown. In this study, we examined how the glomerular map is disturbed and how the odor information processing in the OB is affected in the dysosmia model mice. We found that the anterior-posterior coarse targeting of OSN axons is disrupted after OSN axon injury, while the local axon sorting mechanisms remained. We also found that the connectivity of mitral/tufted cell dendrites is reduced after injury, leading to attenuated odor responses in mitral/tufted cells. These results suggest that existing OSN axons are an essential scaffold for maintaining the integrity of the olfactory circuit, both OSN axons and mitral/tufted cell dendrites, in the adult.


Subject(s)
Axons/pathology , Dendrites/pathology , Olfaction Disorders/pathology , Olfactory Bulb/pathology , Olfactory Receptor Neurons/pathology , Animals , Axons/metabolism , Dendrites/metabolism , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Olfaction Disorders/etiology , Olfaction Disorders/metabolism , Olfactory Bulb/metabolism , Olfactory Receptor Neurons/metabolism , Smell/physiology
11.
Dev Growth Differ ; 57(7): 529-38, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26178919

ABSTRACT

We generated knockout (KO) mice of Nepro, which has been shown to be necessary to maintain neural progenitor cells downstream of Notch in the mouse developing neocortex by using knockdown experiments, to explore its function in embryogenesis. Nepro KO embryos were morphologically indistinguishable from wild type (WT) embryos until the morula stage but failed in blastocyst formation, and many cells of the KO embryos resulted in apoptosis. We found that Nepro was localized in the nucleolus at the blastocyst stage. The number of nucleolus precursor bodies (NPBs) and nucleoli per nucleus was significantly higher in Nepro KO embryos compared with WT embryos later than the 2-cell stage. Furthermore, at the morula stage, whereas 18S rRNA and ribosomal protein S6 (rpS6), which are components of the ribosome, were distributed to the cytoplasm in WT embryos, they were mainly localized in the nucleoli in Nepro KO embryos. In addition, in Nepro KO embryos, the amount of the mitochondria-associated p53 protein increased, and Cytochrome c was distributed in the cytoplasm. These findings indicate that Nepro is a nucleolus-associated protein, and its loss leads to the apoptosis before blastocyst formation in mice.


Subject(s)
Blastocyst/metabolism , Cell Nucleolus/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis , Cell Nucleolus/chemistry , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/deficiency , Repressor Proteins/deficiency
12.
J Neurosci Methods ; 214(2): 170-6, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23357027

ABSTRACT

In vivo electroporation has been widely used to transfect foreign genes into neural progenitors and analyze the function of genes of interest in the developing nervous system. However, it has not been thoroughly examined in the conditional regulation of exogenous genes in postmitotic neurons. Here we show that the combination of in vivo electroporation and the newest version of the tetracycline (Tet)-controlled gene regulatory (Tet-On) system efficiently induced gene expression in various types of neurons in mouse embryonic and postnatal tissues. In pyramidal neurons of the cerebral cortex, tetracycline-responsive element (TRE)-driven gene expression was induced in the presence of doxycycline (Dox). The induction occurred in a dose-dependent manner. The Dox-dependent induction was also observed in cerebellar Purkinje cells and spinal cord neurons. Moreover, the TRE-driven inducible expression of mammalian Barh1 (Mbh1) mimicked the phenotype of the ubiquitous expression of Mbh1 in the spinal cord. These results indicate that the combination of the Tet-On system and in vivo electroporation is useful for analyzing gene function specifically in postmitotic neurons.


Subject(s)
Brain/metabolism , Electroporation/methods , Gene Expression , Gene Transfer Techniques , Neurons/metabolism , Tetracycline/administration & dosage , Animals , Brain/drug effects , Mice , Mice, Inbred ICR , Neurons/drug effects
13.
Adv Exp Med Biol ; 727: 61-70, 2012.
Article in English | MEDLINE | ID: mdl-22399339

ABSTRACT

The Notch pathway is essential for maintaining neural progenitor cells (NPCs) in the developing brain. Activation of the pathway is sufficient to maintain NPCs, whereas loss-of-function mutations in the critical components of the pathway cause precocious neuronal differentiation and NPC depletion. Hairy and Enhancer of split (Hes)-type transcription factors have long been thought to be the only Notch effectors for the maintenance of NPCs. Recently, a novel nuclear protein, Nepro, has been identified as another critical effector of Notch. The Notch pathway is bifurcated into Nepro and Hes-type proteins in the early development of the neocortex. The combination of Nepro and Hes-type proteins is necessary and sufficient for maintaining NPCs downstream of Notch.


Subject(s)
Neocortex/cytology , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Humans , Mice , Neurogenesis
14.
Methods Enzymol ; 477: 37-50, 2010.
Article in English | MEDLINE | ID: mdl-20699135

ABSTRACT

Electroporation combined with surgery is a quick and highly efficient method to transfect nucleic acids into various embryonic tissues in a spatiotemporally restricted manner. Forceps-type electrodes facilitate transfection by delivering electric pulses from outside of the embryo. Many electroporated embryos survive in the pregnant mouse, are born, and are reared. The developing central nervous system (CNS) is a good target for transfection, because there are many neural progenitors adjacent to the ventricle, into which nucleic acids are relatively easily injected. The expression of transfected genes persists in neurons for months.


Subject(s)
Electroporation/methods , Embryo, Mammalian/metabolism , Transfection/methods , Animals , Central Nervous System/embryology , Female , Gene Expression Regulation, Developmental , Mice , Pregnancy
15.
Dev Biol ; 344(2): 1026-34, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20599893

ABSTRACT

During development, commissural neurons in the spinal cord project their axons across the ventral midline, floor plate, via multiple interactions among temporally controlled molecular guidance cues and receptors. The transcriptional regulation of commissural axon-associated receptors, however, is not well characterized. Spinal dorsal cells are transfated into commissural neurons by misexpression of Mbh1, a Bar-class homeobox gene. We examined the function of another Bar-class homeobox gene, Mbh2, and how Mbh1 and Mbh2 modulate expression of the receptors, leading to midline crossing of axons. Misexpression of Mbh1 and Mbh2 showed the same effects in the spinal cord. The competence of spinal dorsal cells to become commissural neurons was dependent on the embryonic stage, during which misexpression of the Mbh genes was able to activate guidance receptor genes such as Rig1 and Nrp2. Misexpression of Lhx2, which has been recently shown to be involved in Rig1 expression, activated Rig1 but not Nrp2, and was less effective in generating commissural neurons. Moreover, expression of Lhx2 was activated by and required the Mbh genes. These findings have revealed a transcriptional cascade, in which Lhx2-dependent and -independent pathways leading to expression of guidance receptors branch downstream of the Mbh genes.


Subject(s)
Neurons/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Axons/metabolism , Axons/physiology , Embryo, Mammalian , Genes , Mammals/genetics , Mammals/metabolism , Mice , Mice, Inbred ICR
16.
Development ; 136(23): 3889-93, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19906856

ABSTRACT

In the developing neocortex, neural progenitor cells (NPCs) produce projection neurons of the six cortical layers in a temporal order. Over the course of cortical neurogenesis, maintenance of NPCs is essential for the generation of distinct types of neurons at the required time. Notch signaling plays a pivotal role in the maintenance of NPCs by inhibiting neuronal differentiation. Although Hairy and Enhancer-of-split (Hes)-type proteins are central to Notch signaling, it remains unclear whether other essential effectors take part in the pathway. In this study, we identify Nepro, a gene expressed in the developing mouse neocortex at early stages that encodes a 63 kDa protein that has no known structural motif except a nuclear localization signal. Misexpression of Nepro inhibits neuronal differentiation only in the early neocortex. Furthermore, knockdown of Nepro by siRNA causes precocious differentiation of neurons. Expression of Nepro is activated by the constitutively active form of Notch but not by Hes genes. Nepro represses expression of proneural genes without affecting the expression of Hes genes. Finally, we show that the combination of Nepro and Hes maintains NPCs even when Notch signaling is blocked. These results indicate that Nepro is involved in the maintenance of NPCs in the early neocortex downstream of Notch.


Subject(s)
Gene Expression , Genes , Neocortex/cytology , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/physiology , Electroporation , Embryo, Mammalian , Expressed Sequence Tags , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred ICR , Molecular Sequence Data , Neocortex/embryology , Nerve Tissue Proteins/genetics , Neurogenesis , Neurons/cytology , Neurons/physiology , Nuclear Localization Signals , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptors, Notch/genetics , Repressor Proteins/genetics , Stem Cells/cytology , Stem Cells/physiology
17.
Dev Biol ; 322(2): 345-54, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18723012

ABSTRACT

Cerebellar granule cells (CGCs) are the most abundant neuronal type in the mammalian brain, and their differentiation is regulated by the basic helix-loop-helix gene, Math1. However, little is known about downstream genes of Math1 and their functions in the cerebellum. To investigate them, we have here established an electroporation-based in vivo gene transfer method in the developing mouse cerebellum. Misexpression of Math1 ectopically induced expression of Bar-class homeobox genes, Mbh1 and Mbh2, which are expressed by CGCs. Conversely, their expression was repressed in CGCs by knockdown of Math1. These findings, taken together with chromatin immunoprecipitation assays, suggest that Math1 directly regulates the Mbh genes in CGCs. Furthermore, a dominant-negative form of the Mbh proteins disrupted proper formation of the external granule layer and differentiation of CGCs, whereas misexpression of the Mbh genes ectopically induced expression of a CGC marker in nonneuronal cells, indicating that the Mbh proteins are required for the differentiation of CGCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebellum/embryology , Microfilament Proteins/metabolism , Neurons/cytology , Nuclear Proteins/metabolism , Animals , Antigens, Differentiation/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/physiology , Cerebellum/cytology , Cerebellum/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred ICR , Mice, Knockout , Microfilament Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/metabolism , Transcription, Genetic
18.
FEBS Lett ; 582(18): 2811-5, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18638478

ABSTRACT

The transcription factor Sox2 is expressed at high levels in neural stem and progenitor cells. Here, we inactivated Sox2 specifically in the developing brain by using Cre-loxP system. Although mutant animals did not survive after birth, analysis of late gestation embryos revealed that loss of Sox2 causes enlargement of the lateral ventricles and a decrease in the number of neurosphere-forming cells. However, although their neurogenic potential is attenuated, Sox2-deficient neural stem cells retain their multipotency and self-renewal capacity. We found that expression level of Sox3 is elevated in Sox2 null developing brain, probably mitigating the effects of loss of Sox2.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/physiology , Embryonic Stem Cells/cytology , HMGB Proteins/physiology , Lateral Ventricles/embryology , Neurons/cytology , Transcription Factors/physiology , Animals , Cell Differentiation/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Gene Silencing , HMGB Proteins/genetics , High Mobility Group Proteins/biosynthesis , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Neurons/metabolism , Receptors, Notch/metabolism , SOXB1 Transcription Factors , Signal Transduction/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
19.
J Biol Chem ; 281(19): 13374-13381, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16547000

ABSTRACT

Sox2 is expressed at high levels in neuroepithelial stem cells and persists in neural stem/progenitor cells throughout adulthood. We showed previously that the Sox2 regulatory region 2 (SRR2) drives strong expression in these cells. Here we generated transgenic mouse strains with the beta-geo reporter gene under the control of the SRR2 in order to examine the spatiotemporal function of this regulatory region. We show that the SRR2 functions specifically in neural stem/progenitor cells. However, unlike Nestin 2nd intronic enhancer, the SRR2 shows strong regional specificity functioning only in restricted areas of the telencephalon but not in any other portions of the central nervous system such as the spinal cord. We also show by in vitro clonogenic assay that at least some of these SRR2-functioning cells possess the hallmark properties of neural stem cells. In adult brains, we could detect strong beta-geo expression in the subventricular zone of the lateral ventricle and along the rostral migrating stream where actively dividing cells reside. Chromatin immunoprecipitation assays reveal interactions of POU and Sox factors with SRR2 in neural stem/progenitor cells. Our data also suggest that the specific recruitment of these proteins to the SRR2 in the telencephalon defines the spatiotemporal activity of the enhancer in the developing nervous system.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/physiology , Neurons/metabolism , Stem Cells/metabolism , Telencephalon/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , POU Domain Factors/metabolism , SOXB1 Transcription Factors , Telencephalon/cytology , Telencephalon/growth & development , Trans-Activators/genetics
20.
Development ; 133(6): 1113-23, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16501169

ABSTRACT

Nuclei are aggregates of neurons distributed in the central nervous system and are fundamental functional units that share anatomical and physiological features. Despite their importance, the cellular basis that leads to nucleogenesis is only poorly understood. Using exo utero electroporation with an enhanced yellow fluorescent protein (EYFP) gene, we show that the precerebellar neurons derived from the lower rhombic lip (lRL) undergo multiple migration steps to form nuclei. After the unilateral transfer of EYFP to the lRL of embryonic day 12.5 mice, EYFP-labelled neurons migrate tangentially from the lRL in two distinct streams, one towards the ventral metencephalon and the other towards the ventral myelencephalon. These neurons cross the ventral midline and then become radially directed. Labelled neurons in the tangential migratory streams form contralateral clusters in the external cuneate nucleus (ECN) and lateral reticular nucleus (LRN) in the myelencephalon, and bilateral clusters in the pontine grey nucleus (PGN) and reticulotegmental nucleus (RTN) in the metencephalon. Before forming the clusters, EYFP-labelled neurons begin to migrate radially towards the ventricle in close apposition to nestin-positive radial fibres, and then they aggregate as they detach from the fibres. Inhibition of cadherin function in ECN and LRN progenitors caused ipsilateral formation of the ECN and LRN, implying that the transition of their migration from tangential to radial involves a cell-intrinsic mechanism. These observations suggest that nucleogenesis of precerebellar neurons is a result of multi-phasic migration, and that ventricle-directed radial glia-guided migration is a key step for nucleogenesis.


Subject(s)
Cell Differentiation , Cell Movement , Cell Nucleus/physiology , Cerebellum/cytology , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Neurons/cytology , Animals , Animals, Newborn , Cadherins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Shape , Cerebellum/embryology , Cerebellum/metabolism , Electroporation , Genes, Reporter , Mice , Mice, Inbred ICR , Microscopy, Electron , Neurons/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...