Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
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
2.
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
3.
J Cell Sci ; 132(11)2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-31076512

RESUMO

Peroxisomes cooperate with mitochondria in the performance of cellular metabolic functions, such as fatty acid oxidation and the maintenance of redox homeostasis. However, whether peroxisomes also regulate mitochondrial fission-fusion dynamics or mitochondrion-dependent apoptosis remained unclear. We now show that genetic ablation of the peroxins Pex3 or Pex5, which are essential for peroxisome biogenesis, results in mitochondrial fragmentation in mouse embryonic fibroblasts (MEFs) in a manner dependent on Drp1 (also known as DNM1L). Conversely, treatment with 4-PBA, which results in peroxisome proliferation, resulted in mitochondrial elongation in wild-type MEFs, but not in Pex3-knockout MEFs. We further found that peroxisome deficiency increased the levels of cytosolic cytochrome c and caspase activity under basal conditions without inducing apoptosis. It also greatly enhanced etoposide-induced caspase activation and apoptosis, which is indicative of an enhanced cellular sensitivity to death signals. Taken together, our data unveil a previously unrecognized role for peroxisomes in the regulation of mitochondrial dynamics and mitochondrion-dependent apoptosis. Effects of peroxin gene mutations on mitochondrion-dependent apoptosis may contribute to pathogenesis of peroxisome biogenesis disorders.This article has an associated First Person interview with the first author of the paper.


Assuntos
Apoptose/fisiologia , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , Peroxissomos/metabolismo , Animais , Butilaminas/farmacologia , Caspases/metabolismo , Linhagem Celular , Citocromos c/metabolismo , Dinaminas/metabolismo , Humanos , Lipoproteínas/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Peroxinas/genética , Transtornos Peroxissômicos/patologia , Receptor 1 de Sinal de Orientação para Peroxissomos/genética , Interferência de RNA , RNA Interferente Pequeno/genética
4.
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.

5.
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
6.
Proc Natl Acad Sci U S A ; 113(21): E2955-64, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27170189

RESUMO

Neurons migrate a long radial distance by a process known as locomotion in the developing mammalian neocortex. During locomotion, immature neurons undergo saltatory movement along radial glia fibers. The molecular mechanisms that regulate the speed of locomotion are largely unknown. We now show that the serine/threonine kinase Akt and its activator phosphoinositide-dependent protein kinase 1 (PDK1) regulate the speed of locomotion of mouse neocortical neurons through the cortical plate. Inactivation of the PDK1-Akt pathway impaired the coordinated movement of the nucleus and centrosome, a microtubule-dependent process, during neuronal migration. Moreover, the PDK1-Akt pathway was found to control microtubules, likely by regulating the binding of accessory proteins including the dynactin subunit p150(glued) Consistent with this notion, we found that PDK1 regulates the expression of cytoplasmic dynein intermediate chain and light intermediate chain at a posttranscriptional level in the developing neocortex. Our results thus reveal an essential role for the PDK1-Akt pathway in the regulation of a key step of neuronal migration.


Assuntos
Movimento Celular/fisiologia , Microtúbulos/metabolismo , Neocórtex/crescimento & desenvolvimento , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Animais , Complexo Dinactina/genética , Complexo Dinactina/metabolismo , Camundongos , Camundongos Transgênicos , Microtúbulos/genética , Neocórtex/citologia , Neurônios/citologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil
7.
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
8.
EMBO J ; 32(7): 970-81, 2013 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-23481253

RESUMO

Throughout life, neural stem cells (NSCs) in the adult hippocampus persistently generate new neurons that modify the neural circuitry. Adult NSCs constitute a relatively quiescent cell population but can be activated by extrinsic neurogenic stimuli. However, the molecular mechanism that controls such reversible quiescence and its physiological significance have remained unknown. Here, we show that the cyclin-dependent kinase inhibitor p57(kip2) (p57) is required for NSC quiescence. In addition, our results suggest that reduction of p57 protein in NSCs contributes to the abrogation of NSC quiescence triggered by extrinsic neurogenic stimuli such as running. Moreover, deletion of p57 in NSCs initially resulted in increased neurogenesis in young adult and aged mice. Long-term p57 deletion, on the contrary, led to NSC exhaustion and impaired neurogenesis in aged mice. The regulation of NSC quiescence by p57 might thus have important implications for the short-term (extrinsic stimuli-dependent) and long-term (age-related) modulation of neurogenesis.


Assuntos
Células-Tronco Adultas/metabolismo , Envelhecimento/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Células-Tronco Adultas/citologia , Envelhecimento/genética , Animais , Inibidor de Quinase Dependente de Ciclina p57/genética , Deleção de Genes , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/citologia
9.
Development ; 141(22): 4343-53, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25344075

RESUMO

In the developing neocortex, neural precursor cells (NPCs) sequentially generate various neuronal subtypes in a defined order. Although the precise timing of the NPC fate switches is essential for determining the number of neurons of each subtype and for precisely generating the cortical layer structure, the molecular mechanisms underlying these switches are largely unknown. Here, we show that epigenetic regulation through Ring1B, an essential component of polycomb group (PcG) complex proteins, plays a key role in terminating NPC-mediated production of subcerebral projection neurons (SCPNs). The level of histone H3 residue K27 trimethylation at and Ring1B binding to the promoter of Fezf2, a fate determinant of SCPNs, increased in NPCs as Fezf2 expression decreased. Moreover, deletion of Ring1B in NPCs, but not in postmitotic neurons, prolonged the expression of Fezf2 and the generation of SCPNs that were positive for CTIP2. These results indicate that Ring1B mediates the timed termination of Fezf2 expression and thereby regulates the number of SCPNs.


Assuntos
Epigênese Genética/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neocórtex/embriologia , Neurogênese/fisiologia , Neurônios/fisiologia , Complexo Repressor Polycomb 1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Bromodesoxiuridina , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Neurônios/citologia , RNA Interferente Pequeno/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor/metabolismo
10.
Clin Calcium ; 27(6): 815-822, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28536319

RESUMO

Recent advances in analysis technology have revealed that non-coding RNAs(ncRNA)that are not translated and function as RNA itself. It has been revealed that long ncRNA(lncRNA), which has more than and 200 nucleotides, is involved in the regulation of fate decision in neural stem cells. In this article, we introduce some functional lncRNAs regulating neural stem cells and discuss the future perspective about this field.


Assuntos
Regulação da Expressão Gênica , Células-Tronco Neurais/metabolismo , RNA Longo não Codificante/genética , Animais , Encéfalo/metabolismo , Linhagem da Célula , Humanos , Células-Tronco Neurais/citologia , Conformação de Ácido Nucleico , RNA Longo não Codificante/química
11.
Genes Cells ; 20(2): 108-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25441120

RESUMO

Immature neurons undergo morphological and physiological changes including axonal and dendritic development to establish neuronal networks. As the transcriptional status changes at a large number of genes during neuronal maturation, global changes in chromatin modifiers may take place in this process. We now show that the amount of heterochromatin protein 1γ (HP1γ) increases during neuronal maturation in the mouse neocortex. Knockdown of HP1γ suppressed axonal and dendritic development in mouse embryonic neocortical neurons in culture, and either knockdown or knockout of HP1γ impaired the projection of callosal axons of superficial layer neurons to the contralateral hemisphere in the developing neocortex. Conversely, forced expression of HP1γ facilitated axonal and dendritic development, suggesting that the increase of HP1γ is a rate limiting step in neuronal maturation. These results together show an important role for HP1γ in promoting axonal and dendritic development in maturing neurons.


Assuntos
Axônios/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Dendritos/metabolismo , Neocórtex/citologia , Neurogênese , Animais , Proteínas Cromossômicas não Histona/genética , Camundongos , Camundongos Endogâmicos ICR/embriologia , Células NIH 3T3 , Neocórtex/embriologia , Neocórtex/metabolismo , Cultura Primária de Células , Regulação para Cima
12.
J Cell Sci ; 126(Pt 3): 745-55, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264741

RESUMO

The crosstalk between spatial adhesion signals and temporal soluble signals is key in regulating cellular responses such as cell migration. Here we show that soluble growth factors enhance integrin signaling through Akt phosphorylation of FAK at Ser695 and Thr700. PDGF treatment or overexpression of active Akt1 in fibroblasts increased autophosphorylation of FAK at Tyr397, an essential event for integrin turnover and cell migration. Phosphorylation-defective mutants of FAK (S695A and T700A) underwent autophosphorylation at Tyr397 and promoted cell migration in response to the integrin ligand fibronectin, but importantly, not in response to PDGF. This study has unveiled a novel function of Akt as an 'ignition kinase' of FAK in growth factor signaling and may shed light on the mechanism by which growth factors regulate integrin signaling.


Assuntos
Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Células COS , Movimento Celular/genética , Chlorocebus aethiops , Fibronectinas/metabolismo , Adesões Focais/genética , Integrinas/metabolismo , Camundongos , Células NIH 3T3 , Fosforilação/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , RNA Interferente Pequeno/genética , Receptor Cross-Talk , Transdução de Sinais/genética , Transgenes/genética
13.
Nat Rev Neurosci ; 11(6): 377-88, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20485363

RESUMO

The temporally and spatially restricted nature of the differentiation capacity of cells in the neural lineage has been studied extensively in recent years. Epigenetic control of developmental genes, which is heritable through cell divisions, has emerged as a key mechanism defining the differentiation potential of cells. Short-term or reversible repression of developmental genes puts them in a 'poised state', ready to be activated in response to differentiation-inducing cues, whereas long-term or permanent repression of developmental genes restricts the cell fates they regulate. Here, we review the molecular mechanisms that underlie the establishment and regulation of differentiation potential along the neural lineage during development.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/crescimento & desenvolvimento , Epigênese Genética/fisiologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Cromatina/fisiologia , Metilação de DNA/fisiologia , Humanos , Modelos Biológicos , Neurônios/citologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
14.
Stem Cells ; 32(11): 2983-97, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25069414

RESUMO

Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Although previous studies have shown that the STAT signaling pathway or its regulators promote the generation of astrocytes from multipotent neural precursor cells (NPCs) in the developing mammalian brain, the molecular mechanisms that regulate the astrocytic fate decision have still remained largely unclear. Here, we show that the high mobility group nucleosome-binding (HMGN) family proteins, HMGN1, 2, and 3, promote astrocyte differentiation of NPCs during brain development. HMGN proteins were expressed in NPCs, Sox9(+) glial progenitors, and GFAP(+) astrocytes in perinatal and adult brains. Forced expression of either HMGN1, 2, or 3 in NPCs in cultures or in the late embryonic neocortex increased the generation of astrocytes at the expense of neurons. Conversely, knockdown of either HMGN1, 2, or 3 in NPCs suppressed astrocyte differentiation and promoted neuronal differentiation. Importantly, overexpression of HMGN proteins did not induce the phosphorylation of STAT3 or activate STAT reporter genes. In addition, HMGN family proteins did not enhance DNA demethylation and acetylation of histone H3 around the STAT-binding site of the gfap promoter. Moreover, knockdown of HMGN family proteins significantly reduced astrocyte differentiation induced by gliogenic signal ciliary neurotrophic factor, which activates the JAK-STAT pathway. Therefore, we propose that HMGN family proteins are novel chromatin regulatory factors that control astrocyte fate decision/differentiation in parallel with or downstream of the JAK-STAT pathway through modulation of the responsiveness to gliogenic signals.


Assuntos
Astrócitos/citologia , Diferenciação Celular/fisiologia , Proteínas HMGN/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Neurogênese/fisiologia , Neuroglia/citologia , Gravidez , Fator de Transcrição STAT3/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(42): 16939-44, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-23027973

RESUMO

The proneural basic helix-loop-helix (bHLH) transcription factor neurogenin1 (Neurog1) plays a pivotal role in neuronal differentiation during mammalian development. The spatiotemporal control of the Neurog1 gene expression is mediated by several specific enhancer elements, although how these elements regulate the Neurog1 locus has remained largely unclear. Recently it has been shown that a large number of enhancer elements are transcribed, but the regulation and function of the resulting transcripts have been investigated for only several such elements. We now show that an enhancer element located 5.8-7.0 kb upstream of the mouse Neurog1 locus is transcribed. The production of this transcript, designated utNgn1, is highly correlated with that of Neurog1 mRNA during neuronal differentiation. Moreover, knockdown of utNgn1 by a corresponding short interfering RNA inhibits the production of Neurog1 mRNA in response to induction of neuronal differentiation. We also found that production of utNgn1 is suppressed by polycomb group (PcG) proteins, which inhibit the expression of Neurog1. Our results thus suggest that a noncoding RNA transcribed from an enhancer element positively regulates transcription at the Neurog1 locus.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Neocórtex/embriologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , RNA não Traduzido/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Northern Blotting , Diferenciação Celular/genética , Imunoprecipitação da Cromatina , Primers do DNA/genética , Técnicas de Silenciamento de Genes , Hibridização In Situ , Camundongos , Neocórtex/citologia , Proteínas do Tecido Nervoso/genética , Interferência de RNA , RNA não Traduzido/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Genes Cells ; 18(2): 79-89, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23331702

RESUMO

Neural precursor cells (NPCs) in the mammalian neocortex generate various neuronal and glial cell types in a developmental stage-dependent manner. Most neocortical NPCs lose their neurogenic potential after birth. We have previously shown that high-mobility group A (HMGA) proteins confer the neurogenic potential on early-stage NPCs during the midgestation period, although the underlying mechanisms are not fully understood. In this study, we found that HMGA2 promotes the expression of insulin-like growth factor 2 mRNA-binding protein 2 (IMP2, Igf2bp2) in neocortical NPCs. The level of IMP2 was indeed high in early-stage NPCs compared with that in late-stage NPCs. Importantly, over-expression of IMP2 increased the neurogenic potential and suppressed astrocytic differentiation of late-stage NPCs, whereas knockdown of IMP2 promoted astrocytic differentiation and reduced the neurogenic potential of early-stage neocortical NPCs without overtly affecting cell proliferation. Our results thus identified IMP2 as a developmental stage-dependent regulator of the differentiation potentials of NPCs in the mouse neocortex.


Assuntos
Diferenciação Celular/fisiologia , Neocórtex/citologia , Neocórtex/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteína HMGA2/metabolismo , Camundongos , Neocórtex/embriologia , Proteínas de Ligação a RNA/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
17.
Genes Cells ; 18(6): 493-501, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23574001

RESUMO

The RIG-I-like receptor (RLR) family of intracellular receptors detects viral nucleic acids and transmits an antiviral signal through the adaptor IPS-1. IPS-1 activation triggers host defense mechanisms, including rapid production of type I interferon (IFN), such as IFN-ß, and induction of apoptosis. IPS-1 is mainly localized to mitochondria, and this localization has been proposed to be essential for inducing production of type I IFN and IFN-stimulated genes (ISGs). However, the importance of this mitochondrial localization of IPS-1 in executing apoptosis has remained unclear. Here, using IPS-1 mutants that were directed to specific subcellular locations such as cytoplasm, plasma membrane and mitochondria, we found that IPS-1's localization to mitochondria is important to activate caspase, but not to signal for IFN-ß gene induction. We also found that IPS-1 possesses a BH3-like motif, which is commonly found among members of the Bcl-2 family. Mutations within this motif promoted IPS-1-induced caspase activation, suggesting that this domain acts as an intrinsic inhibitor domain of apoptosis induction. These results establish that the mitochondrial location of IPS-1 is essential to its ability to induce apoptosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caspases/metabolismo , Mitocôndrias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Ativação Enzimática , Células HEK293 , Humanos
18.
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
19.
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.

20.
Development ; 137(7): 1035-44, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20215343

RESUMO

Basal progenitors (also called non-surface dividing or intermediate progenitors) have been proposed to regulate the number of neurons during neocortical development through expanding cells committed to a neuronal fate, although the signals that govern this population have remained largely unknown. Here, we show that N-myc mediates the functions of Wnt signaling in promoting neuronal fate commitment and proliferation of neural precursor cells in vitro. Wnt signaling and N-myc also contribute to the production of basal progenitors in vivo. Expression of a stabilized form of beta-catenin, a component of the Wnt signaling pathway, or of N-myc increased the numbers of neocortical basal progenitors, whereas conditional deletion of the N-myc gene reduced these and, as a likely consequence, the number of neocortical neurons. These results reveal that Wnt signaling via N-myc is crucial for the control of neuron number in the developing neocortex.


Assuntos
Células-Tronco Multipotentes/fisiologia , Neocórtex/citologia , Neocórtex/embriologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Deleção de Genes , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Neocórtex/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA