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










Base de dados
Intervalo de ano de publicação
1.
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.

2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Nat Commun ; 9(1): 1623, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29691400

RESUMO

Non-pial neocortical astrocytes have historically been thought to comprise largely a nondiverse population of protoplasmic astrocytes. Here we show that astrocytes of the mouse somatosensory cortex manifest layer-specific morphological and molecular differences. Two- and three-dimensional observations revealed that astrocytes in the different layers possess distinct morphologies as reflected by differences in cell orientation, territorial volume, and arborization. The extent of ensheathment of synaptic clefts by astrocytes in layer II/III was greater than that by those in layer VI. Moreover, differences in gene expression were observed between upper-layer and deep-layer astrocytes. Importantly, layer-specific differences in astrocyte properties were abrogated in reeler and Dab1 conditional knockout mice, in which neuronal layers are disturbed, suggesting that neuronal layers are a prerequisite for the observed morphological and molecular differences of neocortical astrocytes. This study thus demonstrates the existence of layer-specific interactions between neurons and astrocytes, which may underlie their layer-specific functions.


Assuntos
Astrócitos/citologia , Neocórtex/crescimento & desenvolvimento , Animais , Astrócitos/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/citologia , Neocórtex/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Proteína Reelina , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Córtex Somatossensorial/citologia , Córtex Somatossensorial/crescimento & desenvolvimento , Córtex Somatossensorial/metabolismo
9.
Methods Mol Biol ; 1728: 263-277, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29405004

RESUMO

Deciphering neuronal networks governing specific brain functions is a longstanding mission in neuroscience, yet global manipulation of protein functions pharmacologically or genetically lacks sufficient specificity to reveal a neuronal protein's function in a particular neuron or a circuitry. Photostimulation presents a great venue for researchers to control neuronal proteins with high temporal and spatial resolution. Recently, an approach to optically control the function of a neuronal protein directly in neurons has been demonstrated using genetically encoded light-sensitive Unnatural amino acids (Uaas). Here, we describe procedures for genetically incorporating Uaas into target neuronal proteins in neurons in vitro and in embryonic mouse brain. As an example, a photocaged Uaa was incorporated into an inwardly rectifying potassium channel Kir2.1 to render Kir2.1 photo-activatable. This method has the potential to be generally applied to many neuronal proteins to achieve optical regulation of different processes in brains. Uaas with other properties can be similarly incorporated into neuronal proteins in neurons for various applications.


Assuntos
Aminoácidos/genética , Código Genético , Neurônios/metabolismo , Optogenética , Engenharia de Proteínas , Aminoácidos/química , Animais , Encéfalo/metabolismo , Células Cultivadas , Eletroporação , Ordem dos Genes , Vetores Genéticos/genética , Canais Iônicos , Camundongos , Microinjeções , Optogenética/métodos , Células Piramidais/metabolismo , Ratos , Transfecção
10.
J Neurosci ; 37(49): 11867-11880, 2017 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-29101245

RESUMO

In the adult mammalian brain, neural stem cells (NSCs) generate new neurons throughout the mammal's lifetime. The balance between quiescence and active cell division among NSCs is crucial in producing appropriate numbers of neurons while maintaining the stem cell pool for a long period. The Notch signaling pathway plays a central role in both maintaining quiescent NSCs (qNSCs) and promoting cell division of active NSCs (aNSCs), although no one knows how this pathway regulates these apparently opposite functions. Notch1 has been shown to promote proliferation of aNSCs without affecting qNSCs in the adult mouse subependymal zone (SEZ). In this study, we found that Notch3 is expressed to a higher extent in qNSCs than in aNSCs while Notch1 is preferentially expressed in aNSCs and transit-amplifying progenitors in the adult mouse SEZ. Furthermore, Notch3 is selectively expressed in the lateral and ventral walls of the SEZ. Knockdown of Notch3 in the lateral wall of the adult SEZ increased the division of NSCs. Moreover, deletion of the Notch3 gene resulted in significant reduction of qNSCs specifically in the lateral and ventral walls, compared with the medial and dorsal walls, of the lateral ventricles. Notch3 deletion also reduced the number of qNSCs activated after antimitotic cytosine ß-D-arabinofuranoside (Ara-C) treatment. Importantly, Notch3 deletion preferentially reduced specific subtypes of newborn neurons in the olfactory bulb derived from the lateral walls of the SEZ. These results indicate that Notch isoforms differentially control the quiescent and proliferative steps of adult SEZ NSCs in a domain-specific manner.SIGNIFICANCE STATEMENT In the adult mammalian brain, the subependymal zone (SEZ) of the lateral ventricles is the largest neurogenic niche, where neural stem cells (NSCs) generate neurons. In this study, we found that Notch3 plays an important role in the maintenance of quiescent NSCs (qNSCs), while Notch1 has been reported to act as a regulator of actively cycling NSCs. Furthermore, we found that Notch3 is specifically expressed in qNSCs located in the lateral and ventral walls of the lateral ventricles and regulates neuronal production of NSCs in a region-specific manner. Our results indicate that Notch3, by maintaining the quiescence of a subpopulation of NSCs, confers a region-specific heterogeneity among NSCs in the adult SEZ.


Assuntos
Células-Tronco Adultas/metabolismo , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Células-Tronco Neurais/metabolismo , Receptor Notch3/biossíntese , Fatores Etários , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Notch3/deficiência
11.
J Vis Exp ; (109): e53818, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-27078635

RESUMO

Photostimulation is a noninvasive way to control biological events with excellent spatial and temporal resolution. New methods are desired to photo-regulate endogenous proteins expressed in their native environment. Here, we present an approach to optically control the function of a neuronal protein directly in neurons using a genetically encoded unnatural amino acid (Uaa). By using an orthogonal tRNA/aminoacyl-tRNA synthetase pair to suppress the amber codon, a photo-reactive Uaa 4,5-dimethoxy-2-nitrobenzyl-cysteine (Cmn) is site-specifically incorporated in the pore of a neuronal protein Kir2.1, an inwardly rectifying potassium channel. The bulky Cmn physically blocks the channel pore, rendering Kir2.1 non-conducting. Light illumination instantaneously converts Cmn into a smaller natural amino acid Cys, activating Kir2.1 channel function. We express these photo-inducible inwardly rectifying potassium (PIRK) channels in rat hippocampal primary neurons, and demonstrate that light-activation of PIRK ceases the neuronal firing due to the outflux of K(+) current through the activated Kir2.1 channels. Using in utero electroporation, we also express PIRK in the embryonic mouse neocortex in vivo, showing the light-activation of PIRK in neocortical neurons. Genetically encoding Uaa imposes no restrictions on target protein type or cellular location, and a family of photoreactive Uaas is available for modulating different natural amino acid residues. This technique thus has the potential to be generally applied to many neuronal proteins to achieve optical regulation of different processes in brains. The current protocol presents an accessible procedure for intricate Uaa incorporation in neurons in vitro and in vivo to achieve photo control of neuronal protein activity on the molecular level.


Assuntos
Aminoácidos/metabolismo , Fármacos Fotossensibilizantes/farmacocinética , Aminoacil-tRNA Sintetases/química , Animais , Terapia Genética , Hipocampo/metabolismo , Luz , Camundongos , Neurônios/fisiologia , Fármacos Fotossensibilizantes/síntese química , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , RNA de Transferência , Ratos
12.
Dev Biol ; 412(1): 139-147, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26896590

RESUMO

Foxg1 expression is highly restricted to the telencephalon and other head structures in the early embryo. This expression pattern has been exploited to generate conditional knockout mice, based on a widely used Foxg1-Cre knock-in line (Foxg1(tm1(cre)Skm)), in which the Foxg1 coding region was replaced by the Cre gene. The utility of this line, however, is severely hampered for two reasons: (1) Foxg1-Cre mice display ectopic and unpredictable Cre activity, and (2) Foxg1 haploinsufficiency can produce neurodevelopmental phenotypes. To overcome these issues, we have generated a new Foxg1-IRES-Cre knock-in mouse line, in which an IRES-Cre cassette was inserted in the 3'UTR of Foxg1 locus, thus preserving the endogenous Foxg1 coding region and un-translated gene regulatory sequences in the 3'UTR, including recently discovered microRNA target sites. We further demonstrate that the new Foxg1-IRES-Cre line displays consistent Cre activity patterns that recapitulated the endogenous Foxg1 expression at embryonic and postnatal stages without causing defects in cortical development. We conclude that the new Foxg1-IRES-Cre mouse line is a unique and advanced tool for studying genes involved in the development of the telencephalon and other Foxg1-expressing regions starting from early embryonic stages.


Assuntos
Fatores de Transcrição Forkhead/genética , Integrases/genética , Proteínas do Tecido Nervoso/genética , Regiões 3' não Traduzidas , Animais , Camundongos
14.
Plant Cell Environ ; 38(12): 2508-18, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25630712

RESUMO

Development of xylem embolism during water stress in two diffuse-porous hardwoods, Katsura (Cercidiphyllum japonicum) and Japanese white birch (Betula platyphylla var. japonica), was observed non-destructively under a compact magnetic resonance imaging (MRI) system in addition to conventional quantitation of hydraulic vulnerability to cavitation from excised stem segments. Distribution of white and dark areas in MR images corresponded well to the distribution of water-filled/embolized vessels observed by cryo-scanning electron microscopy in both species. Water-filled vessels were observed in MR images as white areas in Katsura and as white dots in Japanese white birch, respectively, and embolisms could be detected as a change to dark areas. The increase in the relative embolized area (REA: %) in the cross-sectional area of total xylem during water stress, which was estimated from the binarized MR images, was consistent with the hydraulic vulnerability curves of these species. From the non-destructive MRI observations, cavitation induced by water stress was shown to develop earlier in 1- or 2-year-old xylem than in the current-year xylem in both species; that is, the vulnerability to cavitation differs between vessels in the current-year xylem and those in older annual rings.


Assuntos
Betula/fisiologia , Imageamento por Ressonância Magnética/métodos , Magnoliopsida/fisiologia , Xilema/fisiologia , Betula/ultraestrutura , Desidratação , Magnoliopsida/ultraestrutura , Caules de Planta/fisiologia , Caules de Planta/ultraestrutura , Porosidade , Água/fisiologia , Xilema/ultraestrutura
15.
Neuron ; 80(2): 358-70, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24139041

RESUMO

Optical control of protein function provides excellent spatial-temporal resolution for studying proteins in situ. Although light-sensitive exogenous proteins and ligands have been used to manipulate neuronal activity, a method for optical control of neuronal proteins using unnatural amino acids (Uaa) in vivo is lacking. Here, we describe the genetic incorporation of a photoreactive Uaa into the pore of an inwardly rectifying potassium channel Kir2.1. The Uaa occluded the pore, rendering the channel nonconducting, and, on brief light illumination, was released to permit outward K(+) current. Expression of this photoinducible inwardly rectifying potassium (PIRK) channel in rat hippocampal neurons created a light-activatable PIRK switch for suppressing neuronal firing. We also expanded the genetic code of mammals to express PIRK channels in embryonic mouse neocortex in vivo and demonstrated a light-activated PIRK current in cortical neurons. These principles could be generally expanded to other proteins expressed in the brain to enable optical regulation.


Assuntos
Agonistas de Aminoácidos Excitatórios/farmacologia , Expressão Gênica/fisiologia , Luz , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Canais de Potássio Corretores do Fluxo de Internalização/genética , Animais , Córtex Cerebral/metabolismo , Agonistas de Aminoácidos Excitatórios/síntese química , Células HEK293 , Hipocampo/fisiologia , Humanos , Potenciais da Membrana/fisiologia , Camundongos , Inibição Neural/fisiologia , Neurônios/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia
16.
Nat Commun ; 4: 1880, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23695674

RESUMO

Stem cells often divide asymmetrically to produce one stem cell and one differentiating cell, thus maintaining the stem cell pool. Although neural stem cells (NSCs) in the adult mouse subventricular zone have been suggested to divide asymmetrically, intrinsic cell fate determinants for asymmetric NSC division are largely unknown. Stem cell niches are important for stem cell maintenance, but the niche for the maintenance of adult quiescent NSCs has remained obscure. Here we show that the Notch ligand Delta-like 1 (Dll1) is required to maintain quiescent NSCs in the adult mouse subventricular zone. Dll1 protein is induced in activated NSCs and segregates to one daughter cell during mitosis. Dll1-expressing cells reside in close proximity to quiescent NSCs, suggesting a feedback signal for NSC maintenance by their sister cells and progeny. Our data suggest a model in which NSCs produce their own niche cells for their maintenance through asymmetric Dll1 inheritance at mitosis.


Assuntos
Células-Tronco Adultas/citologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mitose , Células-Tronco Neurais/citologia , Células-Tronco Adultas/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Células Cultivadas , Ventrículos Cerebrais/citologia , Deleção de Genes , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Receptores Notch/metabolismo
17.
Hum Gene Ther Methods ; 24(3): 141-50, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23470213

RESUMO

As the possibility of tumorigenesis and undesirable immune responses in patients cannot be completely excluded in gene and cell therapies, a conditional death switch to eliminate the therapeutic cells would be a valuable tool to enhance the safety of these therapies. A few ligand-receptor conditional death switches have already been developed; however, they cannot be used if patients exhibit side effects upon administration of the ligand. Here we demonstrate a death-inducing chimeric antibody named "death signalobody," in which the antigen-antibody system, having virtually infinite ligand-receptor combinations, is utilized for the activation of death signaling. We designed a death signalobody named "SFas," which has an antifluorescein single-chain variable fragment and the cytoplasmic domain of Fas. SFas efficiently induced conditional apoptosis in murine pro-B Ba/F3 cells in response to fluorescein-conjugated bovine serum albumin. Moreover, SFas was also able to induce antigen-dependent conditional apoptosis in human cancer cell lines. The death signalobody technique will be a valuable tool for the conditional elimination of cells of interest in multiple therapeutic applications.


Assuntos
Apoptose , Proteínas Recombinantes de Fusão/genética , Anticorpos de Cadeia Única/genética , Receptor fas/genética , Animais , Complexo Antígeno-Anticorpo , Antígenos/imunologia , Antígenos/metabolismo , Linhagem Celular , Sobrevivência Celular , Fluoresceína/química , Fluoresceína/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Camundongos , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Soroalbumina Bovina/química , Soroalbumina Bovina/metabolismo , Anticorpos de Cadeia Única/metabolismo , Transdução Genética , Receptor fas/metabolismo
18.
Curr Opin Neurobiol ; 20(1): 22-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20138502

RESUMO

Cortical development progresses through an early phase of progenitor expansion, a middle phase of neurogenesis, and a final phase of gliogenesis. During the middle phase, the neurogenic phase, the neocortical primordium balances the production of neurons against the maintenance of neural precursor cells (NPCs). The final number of neurons is determined by the duration of the neurogenic phase, the rate of NPC division, and the mode of NPC division, that is, whether a division gives rise to two NPCs, one NPC and one cell committed to the neuronal lineage, or two committed cells. We discuss here recent advances in understanding these key aspects that are fundamental for normal brain development.


Assuntos
Neocórtex/citologia , Neurogênese/fisiologia , Neurônios/citologia , Animais , Contagem de Células , Camundongos , Neocórtex/crescimento & desenvolvimento , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/fisiologia
19.
Development ; 135(23): 3849-58, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18997111

RESUMO

During the neurogenic phase of mammalian brain development, only a subpopulation of neural precursor cells (NPCs) differentiates into neurons. The mechanisms underlying this selection remain unclear. Here we provide evidence that the Notch-Delta pathway plays an important role in this selection in the developing mouse telencephalon. We found that the expression patterns of the Notch ligand delta-like 1 (Dll1) and of the active form of Notch1 were mutually exclusive and segregated into distinct NPC subpopulations in the ventricular zone of the telencephalon. When Dll1 was overexpressed in a small, but not a large, proportion of NPCs, these cells underwent neuronal differentiation in vitro and in vivo. This Dll1-induced neuronal differentiation did not occur when cells were plated at lower densities in an in vitro culture. Importantly, conditional deletion of the Dll1 gene in a small proportion of NPCs reduced neurogenesis in vivo, whereas deletion in a large proportion promoted premature neurogenesis. These results support the notion that different levels of Dll1 expression determine the fate of NPCs through cell-cell interactions, most likely through the Notch-Delta lateral inhibitory signaling pathway, thus contributing to the selection of differentiating cells.


Assuntos
Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurônios/citologia , Células-Tronco/citologia , Telencéfalo/citologia , Telencéfalo/embriologia , Animais , Padronização Corporal , Proteínas de Ligação ao Cálcio , Comunicação Celular , Linhagem da Célula , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos ICR , Neocórtex/citologia , Neocórtex/embriologia , Neocórtex/metabolismo , Neurogênese , Neurônios/metabolismo , Estrutura Terciária de Proteína , Receptor Notch1/genética , Receptor Notch1/metabolismo , Células-Tronco/metabolismo , Telencéfalo/metabolismo
20.
Development ; 133(13): 2553-63, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16728475

RESUMO

The transcription factor STAT3 promotes astrocytic differentiation of neural precursor cells (NPCs) during postnatal development of the mouse neocortex, but little has been known of the possible role of STAT3 in the embryonic neocortex. We now show that STAT3 is expressed in NPCs of the mouse embryonic neocortex and that the JAK-STAT3 signaling pathway plays an essential role in the maintenance of NPCs by fibroblast growth factor 2. Conditional deletion of the STAT3 gene in NPCs reduced their capacity to form neurospheres in vitro, as well as promoted neuronal differentiation both in vitro and in vivo. Furthermore, STAT3 was found to maintain NPCs in the undifferentiated state in a non-cell-autonomous manner. STAT3-dependent expression of the Notch ligand Delta-like1 (DLL1) appears to account for the non-cell-autonomous effect of STAT3 on NPC maintenance, as knockdown of DLL1 by RNA interference or inhibition of Notch activation with a gamma-secretase inhibitor abrogated the enhancement of neurosphere formation by STAT3. Our results reveal a previously unrecognized mechanism of interaction between the JAK-STAT3 and DLL1-Notch signaling pathways, as well as a pivotal role for this interaction in maintenance of NPCs during early neocortical development.


Assuntos
Neocórtex/citologia , Neocórtex/embriologia , Neurônios/fisiologia , Fator de Transcrição STAT3/genética , Animais , Sequência de Bases , Eletroporação , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Camundongos , RNA Interferente Pequeno/genética , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...