Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Physiol ; 238(2): 434-446, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36585955

RESUMO

Cyclo-glycylproline (cGP), a cyclic dipeptide containing a condensation bond between glycine and proline, is produced by the cyclization of the N-terminal tripeptide of insulin-like growth factor-1. Previous studies have shown that cGP administration exerts a neuroprotective effect and enhances the regenerative ability in rats with ischemic brain injury. The efficacy of cGP is medicated by regulating the bioavailability of insulin-like growth factor-1 (IGF-1), however, the molecular mechanisms underlying the neuroprotective effects of cGP on brain damage remains to be elucidated. In the current study, we investigated the cGP-mediated molecular mechanism in human fetal neural stem cells (hfNSCs) exposed to oxidative stress, which is a key factor affecting the development of several brain diseases, including traumatic brain injury and Parkinson's disease. We found that cGP treatment attenuated oxidative stress-induced cell death in cultured hfNSCs in a dose-dependent manner. Transcriptome analysis revealed that under oxidative stress conditions, p53-mediated signaling was activated, accompanied by upregulation of mouse double minute 2 homolog (MDM2), a p53-specific E3 ubiquitin ligase, in cGP-treated hfNSCs. By using a comprehensive protein phosphorylation array, we found that cGP induced the activation of Akt signaling pathway, which enhanced the expression of MDM2, in hfNSCs exposed to oxidative stress. Moreover, the MDM2 inhibitor nutlin-3 inhibited the protective effect of cGP on oxidative stress-induced cell death and apoptosis. Therefore, cGP attenuates oxidative stress-induced cell death mediated by the interplay between IGF-1 signaling and the MDM2-p53 pathway in human NSCs. We revealed the molecular mechanism underlying cGP-induced neuroprotective properties in a model of brain damage.


Assuntos
Fator de Crescimento Insulin-Like I , Células-Tronco Neurais , Camundongos , Humanos , Ratos , Animais , Fator de Crescimento Insulin-Like I/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Peróxido de Hidrogênio/metabolismo , Dipeptídeos , Células-Tronco Neurais/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo
2.
Sci Rep ; 12(1): 5593, 2022 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379840

RESUMO

Tissue aging is a major cause of aging-related disabilities and a shortened life span. Understanding how tissue aging progresses and identifying the factors underlying tissue aging are crucial; however, the mechanism of tissue aging is not fully understood. Here we show that the biosynthesis of S-adenosyl-methionine (SAM), the major cellular donor of methyl group for methylation modifications, potently accelerates the aging-related defects during Drosophila oogenesis. An aging-related increase in the SAM-synthetase (Sam-S) levels in the germline leads to an increase in ovarian SAM levels. Sam-S-dependent biosynthesis of SAM controls aging-related defects in oogenesis through two mechanisms, decreasing the ability to maintain germline stem cells and accelerating the improper formation of egg chambers. Aging-related increases in SAM commonly occur in mouse reproductive tissue and the brain. Therefore, our results raise the possibility suggesting that SAM is the factor related to tissue aging beyond the species and tissues.


Assuntos
Drosophila , S-Adenosilmetionina , Envelhecimento , Animais , Metionina Adenosiltransferase , Camundongos , Oogênese
3.
Behav Brain Res ; 417: 113561, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-34509530

RESUMO

Cyclic glycyl-proline (cGP) exerts neuroprotective effects against ischemic stroke and may promote neural plasticity or network remodeling. We sought to determine to what extent oral administration of cGP could facilitate task learning in rats with ischemic lesions. We trained rats to perform a choice reaction time task using their forepaws. One week after changing the food to pellets containing cGP (no cGP: 0 mg/kg; low cGP: 25 mg/kg; and high cGP: 75 mg/kg), we made a focal ischemic lesion on the left or right forepaw area of the sensorimotor cortex. After recovery of task performance, we altered the correct-response side of the task, and then analyzed the number of training days required for the rat to reach a learning criterion (error rate < 15%) and the regulation of adult neurogenesis in the subventricular zones (SVZs), taking lesion size into account. The low-cGP group required fewer training days for task learning than the no-cGP group. Unexpectedly, rats with larger lesions required fewer training days in the no-cGP and low-cGP groups, but more training days in the high-cGP group. The number of Ki67-immunopositive cells (indicating proliferative cells) in ipsilesional SVZ increased more rapidly in the low-cGP and high-cGP groups than in the no-cGP group. However, lesion size had only a small effect on required training days and the number of Ki67-immunopositive cells. We conclude that oral administration of cGP can facilitate task learning in rats with focal ischemic infarction through neural plasticity and network remodeling, even with minimal neuroprotective effects.


Assuntos
Relação Dose-Resposta a Droga , Aprendizagem/efeitos dos fármacos , Plasticidade Neuronal , Fármacos Neuroprotetores , Peptídeos Cíclicos/farmacologia , Acidente Vascular Cerebral/fisiopatologia , Administração Oral , Animais , Modelos Animais de Doenças , Masculino , Neurogênese , Ratos
4.
Cell Rep ; 37(2): 109820, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34644564

RESUMO

Lactate has diverse roles in the brain at the molecular and behavioral levels under physiological and pathophysiological conditions. This study investigates whether lysine lactylation (Kla), a lactate-derived post-translational modification in macrophages, occurs in brain cells and if it does, whether Kla is induced by the stimuli that accompany changes in lactate levels. Here, we show that Kla in brain cells is regulated by neural excitation and social stress, with parallel changes in lactate levels. These stimuli increase Kla, which is associated with the expression of the neuronal activity marker c-Fos, as well as with decreased social behavior and increased anxiety-like behavior in the stress model. In addition, we identify 63 candidate lysine-lactylated proteins and find that stress preferentially increases histone H1 Kla. This study may open an avenue for the exploration of a role of neuronal activity-induced lactate mediated by protein lactylation in the brain.


Assuntos
Comportamento Animal , Encéfalo/metabolismo , Histonas/metabolismo , Ácido Láctico/metabolismo , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional , Potenciais de Ação , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Ansiedade/psicologia , Encéfalo/fisiopatologia , Células Cultivadas , Lisina , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Proteoma , Proteínas Proto-Oncogênicas c-fos/metabolismo , Derrota Social
5.
Biosens Bioelectron ; 167: 112472, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32763827

RESUMO

DNA methylation at the 5-position of cytosine bases (5-methylcytosine, 5mC) in genomic DNA is representative epigenetic modification and is involved in many cellular processes, including gene expression and embryonic development. The hydroxylation of 5mC provide 5-hydroxymethylcytosine (5hmC), the so-called sixth base rediscovered recently in mammalian cells, is also considered to act as an epigenetic regulator. We report herein the immunochemical assessment of 5hmC achieved by an enzyme-linked immunosorbent assay (ELISA) using our linker technology. The keys to this assay are 1) the immobilization of genomic DNA with the bifunctional linker molecule, and 2) quantitative analysis by using guaranteed standard samples containing defined amounts of 5hmC. We succeeded in the sensitive and quantitative detection of 5hmC as well as 5mC in HEK293T cells transfected with TET1, and also monitored the effect of ascorbate on the TET1 catalyzed conversion of 5mC to 5hmC. Our linker technology enables the rapid and stable immobilization of genomic samples and thus contributes to the realization of a reproducible 5hmC evaluation method.


Assuntos
5-Metilcitosina , Técnicas Biossensoriais , 5-Metilcitosina/análogos & derivados , Animais , Citosina , Metilação de DNA , Células HEK293 , Humanos , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
6.
Int J Mol Sci ; 21(13)2020 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-32635647

RESUMO

Indonesian ginger (Zingiber purpureum Rosc.), also known as Bangle, exhibits neurotrophic effects on cultured murine cortical neurons and in the adult mouse brain, but the underlying mechanisms remain unknown. Here, using human fetal neural stem cells (hfNSCs) as a model system for in vitro human neurogenesis, we show that Bangle extracts activate canonical WNT/ß-catenin signaling. Bangle extract-treatment of hfNSCs not only promoted neuronal differentiation, but also accelerated neurite outgrowth from immature neurons. Furthermore, Bangle extracts induced expression of neurogenic genes and WNT signaling-target genes, and facilitated the accumulation of ß-catenin in nuclei of hfNSC. Interestingly, altered histone modifications were also observed in Bangle-treated hfNSCs. Together, these findings demonstrate that Bangle contributes to hfNSC neurogenesis by WNT pathway and epigenetic regulation.


Assuntos
Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Extratos Vegetais/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Zingiber officinale , Células Cultivadas , Código das Histonas/efeitos dos fármacos , Humanos , Doenças do Sistema Nervoso/tratamento farmacológico , Fitoterapia , Extratos Vegetais/uso terapêutico
7.
Neurosci Res ; 143: 31-43, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29859850

RESUMO

Adult neurogenesis is a process of generating new neurons from neural stem/precursor cells (NS/PCs) in restricted adult brain regions throughout life. It is now generally known that adult neurogenesis in the hippocampal dentate gyrus (DG) and subventricular zone participates in various higher brain functions, such as learning and memory formation, olfactory discrimination and repair after brain injury. However, the mechanisms underlying adult neurogenesis remain to be fully understood. Here, we show that Nuclear protein 95 KDa (Np95, also known as UHRF1 or ICBP90), which is an essential protein for maintaining DNA methylation during cell division, is involved in multiple processes of adult neurogenesis. Specific ablation of Np95 in adult NS/PCs (aNS/PCs) led to a decrease in their proliferation and an impairment of neuronal differentiation and to suppression of neuronal maturation associated with the impairment of dendritic formation in the hippocampal DG. We also found that deficiency of Np95 in NS/PCs increased the expression of tumor suppressor genes p16 and p53, and confirmed that expression of these genes in NS/PCs recapitulates the phenotype of Np95-deficient NS/PCs. Taken together, our findings suggest that Np95 plays an essential role in proliferation and differentiation of aNS/PCs through the regulation of tumor suppressor gene expression in adult neurogenesis.


Assuntos
Células-Tronco Adultas/fisiologia , Regulação da Expressão Gênica , Genes Supressores de Tumor , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Giro Denteado/metabolismo , Hipocampo/metabolismo , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Proteínas Nucleares/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases
8.
FEBS Open Bio ; 7(12): 1932-1942, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29226080

RESUMO

Flavin adenine dinucleotide (FAD), synthesized from riboflavin, is redox cofactor in energy production and plays an important role in cell survival. More recently, riboflavin deficiency has been linked to developmental disorders, but its role in stem cell differentiation remains unclear. Here, we show that FAD treatment, using DMSO as a solvent, enabled an increase in the amount of intracellular FAD and promoted neuronal differentiation of human neural stem cells (NSCs) derived not only from fetal brain, but also from induced pluripotent stem cells. Depression of FAD-dependent histone demethylase, lysine-specific demethylase-1 (LSD1), prevented FAD-induced neuronal differentiation. Furthermore, FAD influx facilitated nuclear localization of LSD1 and its enzymatic activity. Together, these findings led us to propose that FAD contributes to proper neuronal production from NSCs in the human fetal brain during development.

9.
Neurogenesis (Austin) ; 3(1): e1249195, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27900345

RESUMO

The cerebral cortex of primates has evolved massively and intricately in comparison to that of other species. Accumulating evidence indicates that this is caused by changes in cell biological features of neural stem cells (NSCs), which differentiate into neurons and glial cells during development. The fate of NSCs during rodent cortical development is stringently regulated by epigenetic factors, such as histone modification enzymes, but the role of these factors in human corticogenesis is largely unknown. We have recently discovered that a lysine-specific demethylase 1 (LSD1), which catalyzes the demethylation of methyl groups in the histone tail, plays a unique role in human fetal NSCs (hfNSCs). We show that, unlike the role previously reported in mice, LSD1 in hfNSCs is necessary for neuronal differentiation and controls the expression of HEYL, one of the NOTCH target genes, by modulating the methylation level of histones on its promoter region. Interestingly, LSD1-regulation of Heyl expression is not observed in mouse NSCs. Furthermore, we first demonstrated that HEYL is able to maintain the undifferentiated state of hfNSCs. Our findings provide a new insight indicating that LSD1 may be a key player in the development and characterization of the evolved cerebral cortex.

10.
Neurogenesis (Austin) ; 3(1): e1232679, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27844025

RESUMO

Despite recent advances in our understanding of epigenetic regulation of central nervous system development, little is known regarding the effects of epigenetic dysregulation on neurogenesis and brain function in adulthood. In the present study, we show that prenatal deletion of DNA methyltransferase 1 (Dnmt1) in neural stem cells results in impaired neurogenesis as well as increases in inflammatory features (e.g., elevated glial fibrillary acidic protein [GFAP] expression in astrocytes and increased numbers of microglia) in the adult mouse brain. Moreover, these mice exhibited anxiety-like behavior during an open-field test. These findings suggest that Dnmt1 plays a critical role in regulating neurogenesis and behavior in the developing brain and into adulthood.

11.
J Neurosci ; 36(22): 6050-68, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27251626

RESUMO

UNLABELLED: Development of the hippocampal dentate gyrus (DG) in the mammalian brain is achieved through multiple processes during late embryonic and postnatal stages, with each developmental step being strictly governed by extracellular cues and intracellular mechanisms. Here, we show that the maintenance DNA methyltransferase 1 (Dnmt1) is critical for development of the DG in the mouse. Deletion of Dnmt1 in neural stem cells (NSCs) at the beginning of DG development led to a smaller size of the granule cell layer in the DG. NSCs lacking Dnmt1 failed to establish proper radial processes or to migrate into the subgranular zone, resulting in aberrant neuronal production in the molecular layer of the DG and a reduction of integrated neurons in the granule cell layer. Interestingly, prenatal deletion of Dnmt1 in NSCs affected not only the developmental progression of the DG but also the properties of NSCs maintained into adulthood: Dnmt1-deficient NSCs displayed impaired neurogenic ability and proliferation. We also found that Dnmt1 deficiency in NSCs decreased the expression of Reelin signaling components in the developing DG and increased that of the cell cycle inhibitors p21 and p57 in the adult DG. Together, these findings led us to propose that Dnmt1 functions as a key regulator to ensure the proper development of the DG, as well as the proper status of NSCs maintained into adulthood, by modulating extracellular signaling and intracellular mechanisms. SIGNIFICANCE STATEMENT: Here, we provide evidence that Dnmt1 is required for the proper development of the hippocampal dentate gyrus (DG). Deletion of Dnmt1 in neural stem cells (NSCs) at an early stage of DG development impaired the ability of NSCs to establish secondary radial glial scaffolds and to migrate into the subgranular zone of the DG, leading to aberrant neuronal production in the molecular layer, increased cell death, and decreased granule neuron production. Prenatal deletion of Dnmt1 in NSCs also induced defects in the proliferation and neurogenic ability of adult NSCs. Furthermore, we found that Dnmt1 regulates the expression of key extracellular signaling components during developmental stages while modulating intracellular mechanisms for proliferation and neuronal production of NSCs in the adult.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Giro Denteado , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Células Cultivadas , DNA (Citosina-5-)-Metiltransferase 1 , Giro Denteado/citologia , Giro Denteado/embriologia , Giro Denteado/crescimento & desenvolvimento , Proteínas do Domínio Duplacortina , Embrião de Mamíferos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina/genética , Nestina/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Proteína Reelina
12.
Stem Cells ; 34(7): 1872-82, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27018646

RESUMO

Histone-modifying enzymes dynamically regulate the chromatin status and have been implicated in the fate specification of stem cells, including neural stem cells (NSCs), which differentiate into three major cell types: neurons, astrocytes, and oligodendrocytes. Lysine-specific demethylase 1 (LSD1, also known as KDM1A) catalyzes the demethylation of H3K4me1/2 and H3K9me1/2, and it was recently suggested that functional disruption of LSD1 links to various human diseases. However, the mechanism by which LSD1 regulates human neural development remains unclear. Here, we present evidence that specific inhibition of LSD1 suppresses the neurogenesis of cultured human fetal NSCs (hfNSCs) isolated from the human fetal neocortex. Notably, we found that LSD1 directly associates with the promoter of the HEYL gene, and controls the demethylation of H3K4me2, which is accompanied by repression of HEYL expression during hfNSC neuronal differentiation. Furthermore, we also showed that HEYL expression is sufficient to inhibit the neuronal differentiation of hfNSCs. This mechanism seems to be primate-specific because mouse NSCs do not exhibit the LSD1 inhibitor-induced upregulation of Heyl. Our findings suggest that LSD1 plays an important role in primate neurogenesis and may contribute to the characterization of an evolved primate brain. Stem Cells 2016;34:1872-1882.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Feto/citologia , Regulação da Expressão Gênica , Histona Desmetilases/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Proteínas Repressoras/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Cromatina/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos Endogâmicos ICR , Neocórtex/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/metabolismo
13.
Neurosci Res ; 95: 1-11, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25659757

RESUMO

Adult neurogenesis persists throughout life in the dentate gyrus (DG) of the hippocampus, and its importance has been highlighted in hippocampus-dependent learning and memory. Adult neurogenesis consists of multiple processes: maintenance and neuronal differentiation of neural stem/precursor cells (NS/PCs), followed by survival and maturation of newborn neurons and their integration into existing neuronal circuitry. However, the mechanisms that govern these processes remain largely unclear. Here we show that DNA methyltransferase 1 (DNMT1), an enzyme responsible for the maintenance of DNA methylation, is highly expressed in proliferative cells in the adult DG and plays an important role in the survival of newly generated neurons. Deletion of Dnmt1 in adult NS/PCs (aNS/PCs) did not affect the proliferation and differentiation of aNS/PCs per se. However, it resulted in a decrease of newly generated mature neurons, probably due to gradual cell death after aNS/PCs differentiated into neurons in the hippocampus. Interestingly, loss of DNMT1 in post-mitotic neurons did not influence their survival. Taken together, these findings suggest that the presence of DNMT1 in aNS/PCs is crucial for the survival of newly generated neurons, but is dispensable once they accomplish neuronal differentiation in the adult hippocampus.


Assuntos
Células-Tronco Adultas/enzimologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Giro Denteado/enzimologia , Células-Tronco Neurais/enzimologia , Neurônios/enzimologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Giro Denteado/citologia , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/citologia
14.
Epigenomics ; 6(6): 637-49, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25531257

RESUMO

Differentiation of neural stem/precursor cells (NS/PCs) into neurons, astrocytes and oligodendrocytes during mammalian brain development is a carefully controlled and timed event. Increasing evidences suggest that epigenetic regulation is necessary to drive this. Here, we provide an overview of the epigenetic mechanisms involved in the developing mammalian embryonic forebrain. Histone methylation is a key factor but other epigenetic factors such as DNA methylation and noncoding RNAs also partake during fate determination. As numerous epigenetic modifications have been identified, future studies on timing and regional specificity of these modifications will further deepen our understanding of how intrinsic and extrinsic mechanisms participate together to precisely control brain development.


Assuntos
Epigênese Genética , Mamíferos/embriologia , Células-Tronco Neurais/citologia , Animais , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular , Metilação de DNA , Histonas/metabolismo , Humanos , Mamíferos/genética , Células-Tronco Neurais/fisiologia , RNA não Traduzido/metabolismo
15.
Neurogenesis (Austin) ; 1(1): e976026, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27504471

RESUMO

Nuclear protein 95 KDa (Np95, also known as UHRF1 or ICBP90) plays an important role in maintaining DNA methylation of newly synthesized DNA strands by recruiting DNA methyltransferase 1 (DNMT1) during cell division. In addition, Np95 participates in chromatin remodeling by interacting with histone modification enzymes such as histone deacetylases. However, its expression pattern and function in the brain have not been analyzed extensively. We here investigated the expression pattern of Np95 in the mouse brain, from developmental to adult stages. In the fetal brain, Np95 is abundantly expressed at the midgestational stage, when a large number of neural stem/precursor cells (NS/PCs) exist. Interestingly, Np95 is expressed specifically in NS/PCs but not in differentiated cells such as neurons or glial cells. Furthermore, we demonstrate that Np95 is preferentially expressed in type 2a cells, which are highly proliferative NS/PCs in the dentate gyrus of the adult hippocampus. Moreover, the number of Np95-expressing cells increases in response to kainic acid administration or to voluntary running, which are known to enhance the proliferation of adult NS/PCs. These results suggest that Np95 participates in the process of proliferation and differentiation of NS/PCs, and that it should be a useful novel marker for proliferating NS/PCs, facilitating the analysis of the complex behavior of NS/PCs in the brain.

16.
Curr Opin Neurobiol ; 23(6): 921-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23827784

RESUMO

In the mammalian central nervous system, astrocytes are the most abundant cell type and play crucial roles in brain development and function. Astrocytes are known to be produced from multipotent neural stem cells (NSCs) at the late gestational stage during brain development, and accumulating evidence indicates that this stage-dependent generation of astrocytes from NSCs is achieved by systematic cooperation between environmental cues and cell-intrinsic programs. Exemplifying the former is cytokine signaling through the gp130-Janus kinase/signal transducer and activator of transcription 3 pathway, and exemplifying the latter is epigenetic modification of astrocyte-specific genes. Here, we introduce recent advances in our understanding of the mechanisms that coordinate astrocytogenesis from NSCs by modulating signaling pathways and epigenetic programs, with a particular focus on the developing mammalian forebrain.


Assuntos
Astrócitos/citologia , Encéfalo/citologia , Diferenciação Celular/fisiologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Animais , Astrócitos/metabolismo , Encéfalo/embriologia , Humanos , Mamíferos , Células-Tronco Neurais/metabolismo , Neurônios/fisiologia
17.
Cell Struct Funct ; 38(1): 55-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23439558

RESUMO

DNA demethylation of astrocyte-specific gene promoters and STAT3 activation in neural precursor cells (NPCs) are essential for astrogliogenesis in the developing brain. To date, it remains unclear whether DNA methylation is the sole epigenetic determinant responsible for suppressing astrocyte-specific genes. Here, we used mouse embryonic stem cells (TKO ESCs) that lacked all 3 DNA methyltransferase genes, Dnmt1, Dnmt3a, and Dnmt3b, and thereby exhibit complete demethylation of the astrocyte-specific glial fibrillary acidic protein (Gfap) gene promoter. We found that although the Gfap promoter was demethylated, STAT3 failed to bind to its cognate element to induce Gfap transcription, whereas it induced transcription of a different target gene, Socs3. Moreover, although the Gfap promoter region containing the STAT3-binding site (GSBS) is enriched with transcription-repressive histone modifications, such as methylation of H3 at lysine 9 (H3K9me3) and H3K27me3, the reduction of these modifications in TKO ESCs was not sufficient for binding of STAT3 at GSBS. Furthermore, GSBS was digested by micrococcal nuclease in late-gestational NPCs that express GFAP upon LIF stimulation, but not in cells that show no expression of GFAP even in the presence of LIF, indicating that STAT3 can access GSBS in the former cells. We further showed that expression of NF-1A, which is known to potentiate differentiation of mid-gestational NPCs into astrocytes, increased its accessibility. Taken together, our results suggest that chromatin accessibility of GSBS plays a critical role in the regulation of Gfap expression.


Assuntos
Astrócitos , Diferenciação Celular/genética , Metilação de DNA/genética , Proteínas do Tecido Nervoso , Fator de Transcrição STAT3 , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Sítios de Ligação , Cromatina/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Proteína Glial Fibrilar Ácida , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica
18.
Stem Cells ; 30(6): 1163-73, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22419556

RESUMO

Because of their ability to self-renew, to differentiate into multiple lineages, and to migrate toward a damaged site, neural stem cells (NSCs), which can be derived from various sources such as fetal tissues and embryonic stem cells, are currently considered to be promising components of cell replacement strategies aimed at treating injuries of the central nervous system, including the spinal cord. Despite their efficiency in promoting functional recovery, these NSCs are not homogeneous and possess variable characteristics depending on their derivation protocols. The advent of induced pluripotent stem (iPS) cells has provided new prospects for regenerative medicine. We used a recently developed robust and stable protocol for the generation of long-term, self-renewing, neuroepithelial-like stem cells from human iPS cells (hiPS-lt-NES cells), which can provide a homogeneous and well-defined population of NSCs for standardized analysis. Here, we show that transplanted hiPS-lt-NES cells differentiate into neural lineages in the mouse model of spinal cord injury (SCI) and promote functional recovery of hind limb motor function. Furthermore, using two different neuronal tracers and ablation of the transplanted cells, we revealed that transplanted hiPS-lt-NES cell-derived neurons, together with the surviving endogenous neurons, contributed to restored motor function. Both types of neurons reconstructed the corticospinal tract by forming synaptic connections and integrating neuronal circuits. Our findings indicate that hiPS-lt-NES transplantation represents a promising avenue for effective cell-based treatment of SCI.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/cirurgia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/metabolismo , Traumatismos da Medula Espinal/patologia , Transplante de Células-Tronco/métodos
19.
J Clin Invest ; 120(9): 3255-66, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20714104

RESUMO

The body's capacity to restore damaged neural networks in the injured CNS is severely limited. Although various treatment regimens can partially alleviate spinal cord injury (SCI), the mechanisms responsible for symptomatic improvement remain elusive. Here, using a mouse model of SCI, we have shown that transplantation of neural stem cells (NSCs) together with administration of valproic acid (VPA), a known antiepileptic and histone deacetylase inhibitor, dramatically enhanced the restoration of hind limb function. VPA treatment promoted the differentiation of transplanted NSCs into neurons rather than glial cells. Transsynaptic anterograde corticospinal tract tracing revealed that transplant-derived neurons reconstructed broken neuronal circuits, and electron microscopic analysis revealed that the transplant-derived neurons both received and sent synaptic connections to endogenous neurons. Ablation of the transplanted cells abolished the recovery of hind limb motor function, confirming that NSC transplantation directly contributed to restored motor function. These findings raise the possibility that epigenetic status in transplanted NSCs can be manipulated to provide effective treatment for SCI.


Assuntos
Vias Neurais/fisiopatologia , Células-Tronco Neurais/transplante , Neurônios/citologia , Neurônios/transplante , Traumatismos da Medula Espinal/terapia , Animais , Diferenciação Celular/fisiologia , Inibidores de Histona Desacetilases , Masculino , Camundongos , Camundongos Endogâmicos ICR , Células-Tronco Multipotentes/transplante , Vias Neurais/fisiologia , Neuroglia/transplante , Tratos Piramidais/fisiopatologia , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/cirurgia
20.
No To Hattatsu ; 41(6): 411-4, 2009 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-19928537

RESUMO

Neural stem/progenitor cells (NSCs/NPCs) give rise to neurons, astrocytes, and oligodendrocytes. It has become apparent that intracellular epigenetic modification including DNA methylation, in concert with extracellular cues such as cytokine signaling, is deeply involved in specifiying the fate of NSCs/NPCs by defining cell-type specific gene expression. However, it is still unclear how differentiated neural cells retain their specific attributes by repressing cellular properties characteristic of other lineages. In previous work, we have shown that methyl-CpG binding protein transcriptional repressors (MBDs), which were expressed predominantly in neurons in the central nervous system, inhibited astrocyte-specific gene expression by binding to highly methylated regions of their target genes. Here we report that oligodendrocytes, which do not express MBDs, can transdifferentiate into astrocytes both in vitro (cytokine stimulation) and in vive (ischemic injury) through the activation of the JAK/STAT signaling pathway. These findings suggest that differentiation plasticity in neural cells is regulated by cell-intrinsic epigenetic mechanisms in collaboration with ambient cell-extrinsic cues.


Assuntos
Astrócitos/citologia , Epigênese Genética/fisiologia , Neurônios/fisiologia , Animais , Diferenciação Celular/fisiologia , Metilação de DNA , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Oligodendroglia/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...