Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38895210

RESUMO

Mitochondria-ER membrane contact sites (MERCS) represent a fundamental ultrastructural feature underlying unique biochemistry and physiology in eukaryotic cells. The ER protein PDZD8 is required for the formation of MERCS in many cell types, however, its tethering partner on the outer mitochondrial membrane (OMM) is currently unknown. Here we identified the OMM protein FKBP8 as the tethering partner of PDZD8 using a combination of unbiased proximity proteomics, CRISPR-Cas9 endogenous protein tagging, Cryo-Electron Microscopy (Cryo-EM) tomography, and correlative light-EM (CLEM). Single molecule tracking revealed highly dynamic diffusion properties of PDZD8 along the ER membrane with significant pauses and capture at MERCS. Overexpression of FKBP8 was sufficient to narrow the ER-OMM distance, whereas independent versus combined deletions of these two proteins demonstrated their interdependence for MERCS formation. Furthermore, PDZD8 enhances mitochondrial complexity in a FKBP8-dependent manner. Our results identify a novel ER-mitochondria tethering complex that regulates mitochondrial morphology in mammalian cells.

2.
J Cell Biol ; 223(5)2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38506728

RESUMO

The development of multicellular organisms depends on spatiotemporally controlled differentiation of numerous cell types and their maintenance. To generate such diversity based on the invariant genetic information stored in DNA, epigenetic mechanisms, which are heritable changes in gene function that do not involve alterations to the underlying DNA sequence, are required to establish and maintain unique gene expression programs. Polycomb repressive complexes represent a paradigm of epigenetic regulation of developmentally regulated genes, and the roles of these complexes as well as the epigenetic marks they deposit, namely H3K27me3 and H2AK119ub, have been extensively studied. However, an emerging theme from recent studies is that not only the autonomous functions of the Polycomb repressive system, but also crosstalks of Polycomb with other epigenetic modifications, are important for gene regulation. In this review, we summarize how these crosstalk mechanisms have improved our understanding of Polycomb biology and how such knowledge could help with the design of cancer treatments that target the dysregulated epigenome.


Assuntos
Repressão Epigenética , Genes Controladores do Desenvolvimento , Proteínas do Grupo Polycomb , Diferenciação Celular , Proteínas de Drosophila , Epigênese Genética , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Humanos , Animais
3.
Nat Commun ; 14(1): 6420, 2023 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828010

RESUMO

Identification of factors that regulate chromatin condensation is important for understanding of gene regulation. High-mobility group AT-hook (HMGA) proteins 1 and 2 are abundant nonhistone chromatin proteins that play a role in many biological processes including tissue stem-progenitor cell regulation, but the nature of their protein function remains unclear. Here we show that HMGA2 mediates direct condensation of polynucleosomes and forms droplets with nucleosomes. Consistently, most endogenous HMGA2 localized to transposase 5- and DNase I-inaccessible chromatin regions, and its binding was mostly associated with gene repression, in mouse embryonic neocortical cells. The AT-hook 1 domain was necessary for chromatin condensation by HMGA2 in vitro and in cellulo, and an HMGA2 mutant lacking this domain was defective in the ability to maintain neuronal progenitors in vivo. Intrinsically disordered regions of other proteins could substitute for the AT-hook 1 domain in promoting this biological function of HMGA2. Taken together, HMGA2 may regulate neural cell fate by its chromatin condensation activity.


Assuntos
Cromatina , Regulação da Expressão Gênica , Camundongos , Animais , Nucleossomos , Células-Tronco , Diferenciação Celular/genética
4.
Aging Cell ; 22(9): e13925, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37476844

RESUMO

Neurons decline in their functionality over time, and age-related neuronal alterations are associated with phenotypes of neurodegenerative diseases. In nonneural tissues, an infolded nuclear shape has been proposed as a hallmark of aged cells and neurons with infolded nuclei have also been reported to be associated with neuronal activity. Here, we performed time-lapse imaging in the visual cortex of Nex-Cre;SUN1-GFP mice. Nuclear infolding was observed within 10 min of stimulation in young nuclei, while the aged nuclei were already infolded pre-stimulation and showed reduced dynamics of the morphology. In young nuclei, the depletion of the stimuli restored the nucleus to a spherical shape and reduced the dynamic behavior, suggesting that nuclear infolding is a reversible process. We also found the aged nucleus to be stiffer than the young one, further relating to the age-associated loss of nuclear shape dynamics. We reveal temporal changes in the nuclear shape upon external stimulation and observe that these morphological dynamics decrease with age.


Assuntos
Neurônios , Córtex Visual , Camundongos , Animais , Córtex Visual/fisiologia
5.
Front Neurosci ; 17: 1149603, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37456993

RESUMO

Adult neural stem cells (NSCs) in the mouse subventricular zone (SVZ) serve as a lifelong reservoir for newborn olfactory bulb neurons. Recent studies have identified a slowly dividing subpopulation of embryonic neural stem-progenitor cells (NPCs) as the embryonic origin of adult NSCs. Yet, little is known about how these slowly dividing embryonic NPCs are maintained until adulthood while other NPCs are extinguished by the completion of brain development. The extracellular matrix (ECM) is an essential component of stem cell niches and thus a key determinant of stem cell fate. Here we investigated tissue inhibitors of metalloproteinases (TIMPs)-regulators of ECM remodeling-for their potential roles in the establishment of adult NSCs. We found that Timp2, Timp3, and Timp4 were expressed at high levels in slowly dividing NPCs compared to rapidly dividing NPCs. Deletion of TIMP3 reduced the number of adult NSCs and neuroblasts in the lateral SVZ. In addition, overexpression of TIMP3 in the embryonic NPCs suppressed neuronal differentiation and upregulated the expression levels of Notch signaling relating genes. These results thus suggest that TIMP3 keeps the undifferentiated state of embryonic NPCs, leading to the establishment and maintenance of adult NSCs.

6.
Nat Commun ; 12(1): 6562, 2021 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-34772946

RESUMO

Quiescent neural stem cells (NSCs) in the adult mouse brain are the source of neurogenesis that regulates innate and adaptive behaviors. Adult NSCs in the subventricular zone are derived from a subpopulation of embryonic neural stem-progenitor cells (NPCs) that is characterized by a slower cell cycle relative to the more abundant rapid cycling NPCs that build the brain. Yet, how slow cell cycle can cause the establishment of adult NSCs remains largely unknown. Here, we demonstrate that Notch and an effector Hey1 form a module that is upregulated by cell cycle arrest in slowly dividing NPCs. In contrast to the oscillatory expression of the Notch effectors Hes1 and Hes5 in fast cycling progenitors, Hey1 displays a non-oscillatory stationary expression pattern and contributes to the long-term maintenance of NSCs. These findings reveal a novel division of labor in Notch effectors where cell cycle rate biases effector selection and cell fate.


Assuntos
Células-Tronco Adultas/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Neurogênese/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/citologia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Células-Tronco Embrionárias , Expressão Gênica , Ventrículos Laterais/metabolismo , Camundongos , Sistema Nervoso , Neurogênese/genética , Receptor Notch1 , Proteínas Repressoras/metabolismo
7.
STAR Protoc ; 2(3): 100763, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34467231

RESUMO

Dynamic changes in histone modifications mediated by Polycomb group proteins can be indicative of the transition of gene repression mode during development. Here, we present methods for the isolation of mouse neocortical neural progenitor-stem cells (NPCs) and their culture, followed by chromatin immunoprecipitation quantitative PCR (ChIP-qPCR) techniques to examine changes in histone H2A ubiquitination patterns at various developmental stages. This protocol can be applied for both in vitro NPCs and NPCs directly isolated from mouse neocortices. For complete details on the use and execution of this protocol, please refer to (Tsuboi et al., 2018).


Assuntos
Imunoprecipitação da Cromatina/métodos , Histonas/metabolismo , Neocórtex/citologia , Neocórtex/embriologia , Células-Tronco Neurais/metabolismo , Animais , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Células Cultivadas , Imunoprecipitação da Cromatina/instrumentação , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células-Tronco Neurais/citologia , Gravidez , Reação em Cadeia da Polimerase em Tempo Real/métodos , Ubiquitinação
8.
STAR Protoc ; 2(2): 100540, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34041504

RESUMO

The embryonic mammalian neocortex includes neural progenitors and neurons at various stages of differentiation. The regulatory mechanisms underlying multiple aspects of neocortical development-including cell division, neuronal fate commitment, neuronal migration, and neuronal differentiation-have been explored using in utero electroporation and virus infection. Here, we describe a protocol for investigation of the effects of genetic manipulation on neural development through direct isolation of neural progenitors and neurons from the mouse embryonic neocortex by fluorescence-activated cell sorting. For complete details on the use and execution of this protocol, please refer to Tsuboi et al. (2018) and Sakai et al. (2019).


Assuntos
Citometria de Fluxo/métodos , Neocórtex , Células-Tronco Neurais/citologia , Animais , Embrião de Mamíferos/citologia , Camundongos , Camundongos Transgênicos , Neocórtex/citologia , Neocórtex/embriologia , Neurônios/citologia
9.
Drug Discov Ther ; 15(2): 55-65, 2021 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-33678755

RESUMO

Immature neurons undergo morphological and physiological maturation in order to establish neuronal networks. During neuronal maturation, a large number of genes change their transcriptional levels, and these changes may be mediated by chromatin modifiers. In this study, we found that the level of Ezh1, a component of Polycomb repressive complex 2 (PRC2), increases during neuronal maturation in mouse neocortical culture. In addition, conditional knockout of Ezh1 in post-mitotic excitatory neurons leads to downregulation of a set of genes related to neuronal maturation. Moreover, the locus encoding Cpg15/Neuritin (Nrn1), which is regulated by neuronal activity and implicated in stabilization and maturation of excitatory synapses, is a direct target of Ezh1 in cortical neurons. Together, these results suggest that elevated expression of Ezh1 contributes to maturation of cortical neurons.


Assuntos
Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Córtex Cerebral/citologia , Depressão/genética , Depressão/metabolismo , Regulação para Baixo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/genética , Neurônios/fisiologia , Complexo Repressor Polycomb 2/sangue , Complexo Repressor Polycomb 2/genética
10.
Stem Cells ; 39(7): 929-944, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33609411

RESUMO

Lysosomes have recently been implicated in regulation of quiescence in adult neural stem cells (NSCs). Whether lysosomes regulate the differentiation of neural stem-progenitor cells (NPCs) in the embryonic brain has remained unknown, however. We here show that lysosomes are more abundant in rapidly dividing NPCs than in differentiating neurons in the embryonic mouse neocortex and ganglionic eminence. The genes for TFEB and TFE3, master regulators of lysosomal biosynthesis, as well as other lysosome-related genes were also expressed at higher levels in NPCs than in differentiating neurons. Anatomic analysis revealed accumulation of lysosomes at the apical and basal endfeet of NPCs. Knockdown of TFEB and TFE3, or that of the lysosomal transporter Slc15a4, resulted in premature differentiation of neocortical NPCs. Conversely, forced expression of an active form of TFEB (TFEB-AA) suppressed neuronal differentiation of NPCs in association with upregulation of NPC-related genes. These results together point to a previously unappreciated role for TFEB and TFE3, and possibly for lysosomes, in maintenance of the undifferentiated state of embryonic NPCs. We further found that lysosomes are even more abundant in an NPC subpopulation that rarely divides and includes the embryonic origin of adult NSCs than in the majority of NPCs that divide frequently for construction of the embryonic brain, and that overexpression of TFEB-AA also suppressed the cell cycle of neocortical NPCs. Our results thus also implicate lysosomes in establishment of the slowly dividing, embryonic origin of adult NSCs.


Assuntos
Neocórtex , Células-Tronco Neurais , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular/fisiologia , Lisossomos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo
11.
Sci Rep ; 11(1): 613, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436697

RESUMO

The lateral ventricles of the adult mammalian brain are lined by a single layer of multiciliated ependymal cells, which generate a flow of cerebrospinal fluid through directional beating of their cilia as well as regulate neurogenesis through interaction with adult neural stem cells. Ependymal cells are derived from a subset of embryonic neural stem-progenitor cells (NPCs, also known as radial glial cells) that becomes postmitotic during the late embryonic stage of development. Members of the Geminin family of transcriptional regulators including GemC1 and Mcidas play key roles in the differentiation of ependymal cells, but it remains largely unclear what extracellular signals regulate these factors and ependymal differentiation during embryonic and early-postnatal development. We now show that the levels of Smad1/5/8 phosphorylation and Id1/4 protein expression-both of which are downstream events of bone morphogenetic protein (BMP) signaling-decline in cells of the ventricular-subventricular zone in the mouse lateral ganglionic eminence in association with ependymal differentiation. Exposure of postnatal NPC cultures to BMP ligands or to a BMP receptor inhibitor suppressed and promoted the emergence of multiciliated ependymal cells, respectively. Moreover, treatment of embryonic NPC cultures with BMP ligands reduced the expression level of the ependymal marker Foxj1 and suppressed the emergence of ependymal-like cells. Finally, BMP ligands reduced the expression levels of Gemc1 and Mcidas in postnatal NPC cultures, whereas the BMP receptor inhibitor increased them. Our results thus implicate BMP signaling in suppression of ependymal differentiation from NPCs through regulation of Gemc1 and Mcidas expression during embryonic and early-postnatal stages of mouse telencephalic development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Epêndima/citologia , Células-Tronco Neurais/citologia , Telencéfalo/citologia , Animais , Proteínas de Ciclo Celular/metabolismo , Células-Tronco Embrionárias/metabolismo , Epêndima/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células-Tronco Neurais/metabolismo , Neurogênese , Telencéfalo/metabolismo
12.
J Immunol ; 206(1): 154-163, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33219146

RESUMO

Viral RNA in the cytoplasm of mammalian host cells is recognized by retinoic acid-inducible protein-I-like receptors (RLRs), which localize to cytoplasmic stress granules (SGs). Activated RLRs associate with the mitochondrial adaptor protein IPS-1, which activates antiviral host defense mechanisms, including type I IFN induction. It has remained unclear, however, how RLRs in SGs and IPS-1 in the mitochondrial outer membrane associate physically and engage in information transfer. In this study, we show that NUDT21, an RNA-binding protein that regulates alternative transcript polyadenylation, physically associates with IPS-1 and mediates its localization to SGs in response to transfection with polyinosinic-polycytidylic acid [poly(I:C)], a mimic of viral dsRNA. We found that despite its well-established function in the nucleus, a fraction of NUDT21 localizes to mitochondria in resting cells and becomes localized to SGs in response to poly(I:C) transfection. NUDT21 was also found to be required for efficient type I IFN induction in response to viral infection in both human HeLa cells and mouse macrophage cell line RAW264.7 cells. Our results together indicate that NUDT21 links RLRs in SGs to mitochondrial IPS-1 and thereby activates host defense responses to viral infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por Cardiovirus/metabolismo , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Proteína DEAD-box 58/metabolismo , Vírus da Encefalomiocardite/fisiologia , Mitocôndrias/metabolismo , Doença de Newcastle/metabolismo , Vírus da Doença de Newcastle/fisiologia , Receptores Imunológicos/metabolismo , Vesículas Secretórias/metabolismo , Animais , Fator de Especificidade de Clivagem e Poliadenilação/genética , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Poli I-C/imunologia , Transporte Proteico , Células RAW 264.7 , RNA Interferente Pequeno/genética , RNA Viral/imunologia , Estresse Fisiológico
13.
Chaos ; 31(12): 121101, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34972333

RESUMO

We propose an algorithm to refine the reconstruction of an original time series given a recurrence plot, which is also referred to as a contact map. The refinement process calculates the local distances based on the Jaccard coefficients with the neighbors in the previous resolution for each point and takes their weighted average using local distances. We demonstrate the utility of our method using two examples.

14.
Nat Commun ; 11(1): 5709, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33177537

RESUMO

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


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Complexo Repressor Polycomb 1/metabolismo , Telencéfalo/embriologia , Animais , Padronização Corporal , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Histonas/genética , Histonas/metabolismo , Lisina/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Complexo Repressor Polycomb 1/genética , Telencéfalo/anormalidades , Fatores de Transcrição/genética , Via de Sinalização Wnt/genética
15.
Cell Rep ; 32(11): 108126, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32937141

RESUMO

SETD1A encodes a histone methyltransferase whose de novo mutations are identified in schizophrenia (SCZ) patients and confer a large increase in disease risk. Here, we generate Setd1a mutant mice carrying the frameshift mutation that closely mimics a loss-of-function variant of SCZ. Our Setd1a (+/-) mice display various behavioral abnormalities relevant to features of SCZ, impaired excitatory synaptic transmission in layer 2/3 (L2/3) pyramidal neurons of the medial prefrontal cortex (mPFC), and altered expression of diverse genes related to neurodevelopmental disorders and synaptic functions in the mPFC. RNAi-mediated Setd1a knockdown (KD) specifically in L2/3 pyramidal neurons of the mPFC only recapitulates impaired sociality among multiple behavioral abnormalities of Setd1a (+/-) mice. Optogenetics-assisted selective stimulation of presynaptic neurons combined with Setd1a KD reveals that Setd1a at postsynaptic site is essential for excitatory synaptic transmission. Our findings suggest that reduced SETD1A may attenuate excitatory synaptic function and contribute to the pathophysiology of SCZ.


Assuntos
Comportamento Animal , Histona-Lisina N-Metiltransferase/deficiência , Esquizofrenia/fisiopatologia , Sinapses/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sistemas CRISPR-Cas/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Ácido Glutâmico/metabolismo , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Masculino , Camundongos Endogâmicos ICR , Mutação/genética , Transtornos do Neurodesenvolvimento/genética , Córtex Pré-Frontal/metabolismo , Terminações Pré-Sinápticas/fisiologia , Células Piramidais/metabolismo , Esquizofrenia/genética , Comportamento Social
16.
Sci Rep ; 10(1): 1884, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32024956

RESUMO

Imprinted genes are expressed from only one allele in a parent of origin-specific manner. The cyclin-dependent kinase inhibitor p57kip2 is encoded by an imprinted gene Cdkn1c, with the paternal allele being silenced. The possible expression and function of the paternal allele of Cdkn1c have remained little studied, however. We now show that the paternal allele of the Cdkn1c gene is expressed at a low level in the developing mouse neocortex. Surprisingly, the central nervous system-specific conditional deletion of the paternal allele (pat cKO) at the Cdkn1c locus resulted in a marked reduction in brain size. Furthermore, pat cKO gradually reduced the number of neural stem-progenitor cells (NPCs) during neocortical development, and thus reduced the number of upper-layer neurons, which were derived from late-stage NPCs. Our results thus show that the paternal allele of the Cdkn1c locus plays a key role in maintenance of NPCs during neocortical development.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/genética , Desenvolvimento Embrionário/genética , Impressão Genômica , Neocórtex/embriologia , Células-Tronco Neurais/fisiologia , Alelos , Animais , Diferenciação Celular/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Embrião de Mamíferos , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Neocórtex/citologia , Neurônios/fisiologia
18.
Genes Cells ; 24(10): 650-666, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31442350

RESUMO

Neural progenitor cells (NPCs, also known as radial glial progenitors) produce neurons and then glial cells such as astrocytes during development of the mouse neocortex. Given that this sequential generation of neural cells is critical for proper brain formation, the neurogenic potential of NPCs must be precisely controlled. Here, we show that the transcription factor Plag1 plays an important role in the regulation of neurogenic potential in mouse neocortical NPCs. We found that Hmga2, a key neurogenic factor in neocortical NPCs, induces expression of the Plag1 gene. Analysis of the effects of over-expression or knockdown of Plag1 indicated that Plag1 promotes the production of neurons at the expense of astrocyte production in embryonic neocortical cultures. Furthermore, over-expression of Plag1 promoted and knockdown of Plag1 suppressed neuronal differentiation of neocortical NPCs in vivo. Transcriptomic analysis showed that Plag1 increases the expression of a set of neuronal genes in NPCs. Our results thus identify Plag1 as a regulator of neuronal gene expression and neuronal differentiation in NPCs of the developing mouse neocortex.


Assuntos
Proteínas de Ligação a DNA/genética , Neocórtex/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Regulação da Expressão Gênica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Camundongos , Neocórtex/citologia , Neocórtex/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia
19.
Curr Opin Neurobiol ; 59: 164-173, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31398486

RESUMO

While all the developmental genes are temporarily repressed for future activation in the pluripotent stem cells, non-neural genes become persistently repressed in the course of commitment to the neuronal lineage. Although Polycomb group proteins (PcG) are key factors for both temporary and persistent repression of the developmental genes, how the same group of proteins can differentially repress target genes remains unclarified. The identification of a variety of PcG complexes and activities sheds light on these issues. In this review, based on the recent findings including those with the use of interactome and Chromosome Conformation Capture (3C)-type analyses, we summarize the molecular mechanisms of PcG-mediated gene regulation and discuss how PcG regulates cell fate specification during neural development.


Assuntos
Regulação da Expressão Gênica , Neurogênese , Diferenciação Celular , Proteínas do Grupo Polycomb
20.
J Cell Biol ; 218(8): 2442-2443, 2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31332023

RESUMO

The trans-synaptic cell adhesion molecule neurexin regulates synaptic functions but its high-resolution subcellular localization and dynamics were unknown. Trotter et al. (2019. J. Cell Biol. https://doi.org/10.1083/jcb.201812076) describe previously unrecognized nanoscale clusters of neurexin-1 in presynaptic terminals and their regulation by ADAM10-mediated proteolysis.


Assuntos
Terminações Pré-Sinápticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...