Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 161(7): 1644-55, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26091041

RESUMO

Adult neural stem/progenitor (B1) cells within the walls of the lateral ventricles generate different types of neurons for the olfactory bulb (OB). The location of B1 cells determines the types of OB neurons they generate. Here we show that the majority of mouse B1 cell precursors are produced between embryonic days (E) 13.5 and 15.5 and remain largely quiescent until they become reactivated postnatally. Using a retroviral library carrying over 100,000 genetic tags, we found that B1 cells share a common progenitor with embryonic cells of the cortex, striatum, and septum, but this lineage relationship is lost before E15.5. The regional specification of B1 cells is evident as early as E11.5 and is spatially linked to the production of neurons that populate different areas of the forebrain. This study reveals an early embryonic regional specification of postnatal neural stem cells and the lineage relationship between them and embryonic progenitor cells.


Assuntos
Células-Tronco Adultas/citologia , Linhagem da Célula , Embrião de Mamíferos/citologia , Células-Tronco Neurais/citologia , Bulbo Olfatório/citologia , Células-Tronco Adultas/classificação , Animais , Camundongos , Células-Tronco Neurais/classificação , Prosencéfalo/citologia
2.
Annu Rev Cell Dev Biol ; 30: 465-502, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000993

RESUMO

Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.


Assuntos
Neocórtex/crescimento & desenvolvimento , Neurogênese/fisiologia , Animais , Divisão Celular Assimétrica , Compartimento Celular , Linhagem da Célula , Membrana Celular/fisiologia , Núcleo Celular/fisiologia , Polaridade Celular , Líquido Cefalorraquidiano/fisiologia , Humanos , Junções Intercelulares/fisiologia , Ventrículos Laterais/embriologia , Lipídeos de Membrana/metabolismo , Microglia/fisiologia , Mitose , Neocórtex/citologia , Neocórtex/embriologia , Células-Tronco Neurais/classificação , Células-Tronco Neurais/fisiologia , Células Neuroepiteliais/citologia , Células Neuroepiteliais/fisiologia , Neurônios/fisiologia , Organelas/fisiologia , Especificidade da Espécie
3.
Med Sci Monit ; 28: e933830, 2022 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-35250022

RESUMO

BACKGROUND Ischemic cerebrovascular disease leads to the activation and differentiation of neural stem cells (NSCs) into mature neurons and glia cells to repair nerve damage. Astragalus flavone (ASF) has shown its potential role in proliferation and differentiation into dopamine neurons of NSCs. MATERIAL AND METHODS Cerebral infarction models were constructed to determine the effects of ASF on NSCs in vivo and in vitro. RESULTS ASF therapy had the ability to reduce the neurologic function scores and the cerebral infarction volume of the cerebral infarction model. Moreover, ASF was able to increase BrdU-positive cells and promote the expression of Nestin, ß-Tubulin III, and O4, while decreasing the expression of GFAP. qRT-PCR and western blot assays showed ASF promoted the expression of Mash1, Math1, and Ngn2 mRNA and protein in cerebral infarction rats. Meanwhile, ASF (20 µg/ml) was able to increase EdU-positive cells and promote the expression of Nestin, ß-Tubulin III, and O4 of NSCs at day14 in vitro. In normoxia, ASF obviously promoted the expression of Mash1, Ngn1, and Ngn2 mRNA and proteins, but in hypoxia, ASF promoted the expression of Notch1 and Math1 mRNA and proteins and inhibited the expression of Ngn1 and Ngn2 mRNA and proteins. CONCLUSIONS ASF therapy can improve the neurologic functions and reduce the cerebral infarction volume in a cerebral infarction model. Moreover, ASF promoted the proliferation of NSCs and induced differentiation into neurons and oligodendrocytes, which might be involved in regulating factors in Notch signaling.


Assuntos
Infarto Cerebral/patologia , Flavonas/farmacologia , Células-Tronco Neurais/classificação , Neurogênese/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Infarto Cerebral/tratamento farmacológico , Modelos Animais de Doenças , Masculino , Células-Tronco Neurais/efeitos dos fármacos , Ratos , Ratos Wistar , Transdução de Sinais
4.
Biochem Soc Trans ; 49(5): 1997-2006, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34397081

RESUMO

The mammalian neocortex is the seat of higher cognitive functions, such as thinking and language in human. A hallmark of the neocortex are the cortical neurons, which are generated from divisions of neural progenitor cells (NPCs) during development, and which constitute a key feature of the well-organized layered structure of the neocortex. Proper formation of neocortex structure requires an orchestrated cellular behavior of different cortical NPCs during development, especially during the process of cortical neurogenesis. Here, we review the great diversity of NPCs and their contribution to the development of the neocortex. First, we review the categorization of NPCs into different classes and types based on their cell biological features, and discuss recent advances in characterizing marker expression and cell polarity features in the different types of NPCs. Second, we review the different modes of cell divisions that NPCs undergo and discuss the importance of the balance between proliferation and differentiation of NPCs in neocortical development. Third, we review the different proliferative capacities among different NPC types and among the same type of NPC in different mammalian species. Dissecting the differences between NPC types and differences among mammalian species is beneficial to further understand the development and the evolutionary expansion of the neocortex and may open up new therapeutic avenues for neurodevelopmental and psychiatric disorders.


Assuntos
Neocórtex/citologia , Neocórtex/crescimento & desenvolvimento , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neurônios/citologia , Animais , Evolução Biológica , Divisão Celular/fisiologia , Polaridade Celular/fisiologia , Humanos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia
5.
Genes Dev ; 27(11): 1272-87, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23723414

RESUMO

Neural stem cells (NSCs) reside in widespread regions along the lateral ventricle and generate diverse olfactory bulb (OB) interneuron subtypes in the adult mouse brain. Molecular mechanisms underlying their regional diversity, however, are not well understood. Here we show that the homeodomain transcription factor Gsx2 plays a crucial role in the region-specific control of adult NSCs in both persistent and injury-induced neurogenesis. In the intact brain, Gsx2 is expressed in a regionally restricted subset of NSCs and promotes the activation and lineage progression of stem cells, thereby controlling the production of selective OB neuron subtypes. Moreover, Gsx2 is ectopically induced in damaged brains outside its normal expression domains and is required for injury-induced neurogenesis in the subventricular zone (SVZ). These results demonstrate that mobilization of adult NSCs is controlled in a region-specific manner and that distinct mechanisms operate in continuous and injury-induced neurogenesis in the adult brain.


Assuntos
Proteínas de Homeodomínio/metabolismo , Ventrículos Laterais/citologia , Ventrículos Laterais/lesões , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Células-Tronco Adultas/metabolismo , Animais , Linhagem da Célula , Ventrículos Laterais/metabolismo , Camundongos , Células-Tronco Neurais/classificação , Bulbo Olfatório/citologia , Especificidade de Órgãos , Nicho de Células-Tronco , Fatores de Transcrição/metabolismo
6.
Development ; 144(23): 4313-4321, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29183942

RESUMO

Cellular reprogramming is a dedifferentiation process during which cells continuously undergo phenotypical remodeling. Although the genetic and biochemical details of this remodeling are fairly well understood, little is known about the change in cell mechanical properties during the process. In this study, we investigated changes in the mechanical phenotype of murine fetal neural progenitor cells (fNPCs) during reprogramming to induced pluripotent stem cells (iPSCs). We find that fNPCs become progressively stiffer en route to pluripotency, and that this stiffening is mirrored by iPSCs becoming more compliant during differentiation towards the neural lineage. Furthermore, we show that the mechanical phenotype of iPSCs is comparable with that of embryonic stem cells. These results suggest that mechanical properties of cells are inherent to their developmental stage. They also reveal that pluripotent cells can differentiate towards a more compliant phenotype, which challenges the view that pluripotent stem cells are less stiff than any cells more advanced developmentally. Finally, our study indicates that the cell mechanical phenotype might be utilized as an inherent biophysical marker of pluripotent stem cells.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Animais , Biomarcadores/metabolismo , Fenômenos Biomecânicos , Antígeno CD24/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/classificação , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Antígenos CD15/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/classificação , Fenótipo , Análise de Célula Única
7.
Anal Bioanal Chem ; 411(21): 5423-5436, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31161326

RESUMO

It is necessary to characterize and classify neural stem cells (NSCs) and differentiated cells (DCs) for potential use of NSC to treat neurodegenerative diseases. We therefore performed an analysis of NSCs and DCs using gas chromatography mass spectrometry (GC-MS) and direct infusion mass spectrometry (DI-MS) with elaborate multivariate statistical analysis for the characterization and classification of rat NSCs and DCs. GC-MS and DI-MS detected a total of 92 metabolites and lipids in NSCs and DCs, and the levels of 72 of them differed significantly between NSCs and DCs. The optimal model for partial least squares (PLS) discriminant analysis was constructed by applying 3 and 2 PLS components with a unit-variance scaling method for classifying NSCs and DCs based on the data obtained in the GC-MS and DI-MS analyses, respectively. The obtained results from PCA and PLS-DA suggest that creatinine, lactic acid, lysine, glutamine, glycine, pyroglutamic acid, PG 18:1/20:2, PS 18:0/20:2, PI 18:0/20:3, PC 16:0/20:4, PI 16:0/20:4, and PI 18:1/20:4 were the main contributors that provided distinct characteristics of NSCs and DCs. The results of this study suggest objective and complementary criteria for the characterization and classification of NSCs and DCs for potential clinical applications. Graphical abstract.


Assuntos
Diferenciação Celular , Metabolismo dos Lipídeos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/citologia , Animais , Células Cultivadas , Análise Discriminante , Cromatografia Gasosa-Espectrometria de Massas/métodos , Análise dos Mínimos Quadrados , Espectrometria de Massas/métodos , Análise de Componente Principal , Ratos , Ratos Sprague-Dawley
8.
J Cell Biochem ; 119(4): 3394-3403, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29130544

RESUMO

Adult neural stem cells (NSCs) are a group of multi-potent, self-renewing progenitor cells that contribute to the generation of new neurons and oligodendrocytes. Three subtypes of NSCs can be isolated based on the stages of the NSC lineage, including quiescent neural stem cells (qNSCs), activated neural stem cells (aNSCs) and neural progenitor cells (NPCs). Although it is widely accepted that these three groups of NSCs play different roles in the development of the nervous system, their molecular signatures are poorly understood. In this study, we applied the Monte-Carlo Feature Selection (MCFS) method to identify the gene expression signatures, which can yield a Matthews correlation coefficient (MCC) value of 0.918 with a support vector machine evaluated by ten-fold cross-validation. In addition, some classification rules yielded by the MCFS program for distinguishing above three subtypes were reported. Our results not only demonstrate a high classification capacity and subtype-specific gene expression patterns but also quantitatively reflect the pattern of the gene expression levels across the NSC lineage, providing insight into deciphering the molecular basis of NSC differentiation.


Assuntos
Astrócitos/citologia , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Células-Tronco Neurais/classificação , Algoritmos , Linhagem da Célula , Células Cultivadas , Humanos , Método de Monte Carlo , Máquina de Vetores de Suporte
9.
Development ; 141(2): 253-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24306106

RESUMO

Drosophila type II neuroblasts (NBs), like mammalian neural stem cells, deposit neurons through intermediate neural progenitors (INPs) that can each produce a series of neurons. Both type II NBs and INPs exhibit age-dependent expression of various transcription factors, potentially specifying an array of diverse neurons by combinatorial temporal patterning. Not knowing which mature neurons are made by specific INPs, however, conceals the actual variety of neuron types and limits further molecular studies. Here we mapped neurons derived from specific type II NB lineages and found that sibling INPs produced a morphologically similar but temporally regulated series of distinct neuron types. This suggests a common fate diversification program operating within each INP that is modulated by NB age to generate slightly different sets of diverse neurons based on the INP birth order. Analogous mechanisms might underlie the expansion of neuron diversity via INPs in mammalian brain.


Assuntos
Drosophila/citologia , Drosophila/crescimento & desenvolvimento , Células-Tronco Neurais/citologia , Animais , Animais Geneticamente Modificados , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Linhagem da Célula , Drosophila/genética , Modelos Neurológicos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/metabolismo , Neurogênese
10.
Development ; 141(6): 1260-71, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24553291

RESUMO

Axon guidance by commissural neurons has been well documented, providing us with a molecular logic of how midline crossing is achieved during development. Despite these advances, knowledge of the intrinsic genetic programs is still limited and it remains obscure whether the expression of a single transcription factor is sufficient to activate transcriptional programs that ultimately enable midline crossing. Here, we show in the mouse that the homeodomain transcription factor Dbx1 is expressed by a subset of progenitor cells that give rise to commissural neurons in the dorsal midbrain. Gain- and loss-of-function analyses indicate that the expression of Dbx1 alone is sufficient and necessary to trigger midline crossing in vivo. We also show that Robo3 controls midline crossing as a crucial downstream effector of the Dbx1-activated molecular programs. Furthermore, Dbx1 suppresses the expression of the transcriptional program for ipsilateral neuron differentiation in parallel. These results suggest that a single transcription factor, Dbx1, has an essential function in assigning midline-crossing identity, thereby contributing crucially to the establishment of the wiring laterality in the developing nervous system.


Assuntos
Proteínas de Homeodomínio/metabolismo , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Animais , Axônios/metabolismo , Padronização Corporal/genética , Padronização Corporal/fisiologia , Elementos Facilitadores Genéticos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Células-Tronco Neurais/classificação , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Gravidez , Receptores de Superfície Celular , Transdução de Sinais
11.
Development ; 141(6): 1392-403, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24595292

RESUMO

In the ventral spinal cord, generation of neuronal and glial cell subtypes is controlled by Sonic hedgehog (Shh). This morphogen contributes to cell diversity by regulating spatial and temporal sequences of gene expression during development. Here, we report that establishing Shh source cells is not sufficient to induce the high-threshold response required to specify sequential generation of ventral interneurons and oligodendroglial cells at the right time and place in zebrafish. Instead, we show that Shh-producing cells must repeatedly upregulate the secreted enzyme Sulfatase1 (Sulf1) at two critical time points of development to reach their full inductive capacity. We provide evidence that Sulf1 triggers Shh signaling activity to establish and, later on, modify the spatial arrangement of gene expression in ventral neural progenitors. We further present arguments in favor of Sulf1 controlling Shh temporal activity by stimulating production of active forms of Shh from its source. Our work, by pointing out the key role of Sulf1 in regulating Shh-dependent neural cell diversity, highlights a novel level of regulation, which involves temporal evolution of Shh source properties.


Assuntos
Proteínas Hedgehog/metabolismo , Medula Espinal/embriologia , Medula Espinal/metabolismo , Sulfatases/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Camundongos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Transdução de Sinais , Medula Espinal/citologia , Sulfatases/genética , Sulfotransferases/genética , Sulfotransferases/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
12.
Proc Natl Acad Sci U S A ; 111(34): 12438-43, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114218

RESUMO

The apical domain of embryonic (radial glia) and adult (B1 cells) neural stem cells (NSCs) contains a primary cilium. This organelle has been suggested to function as an antenna for the detection of morphogens or growth factors. In particular, primary cilia are essential for Hedgehog (Hh) signaling, which plays key roles in brain development. Their unique location facing the ventricular lumen suggests that primary cilia in NSCs could play an important role in reception of signals within the cerebrospinal fluid. Surprisingly, ablation of primary cilia using conditional alleles for genes essential for intraflagellar transport [kinesin family member 3A (Kif3a) and intraflagellar transport 88 (Ift88)] and Cre drivers that are activated at early [Nestin; embryonic day 10.5 (E10.5)] and late [human glial fibrillary acidic protein (hGFAP); E13.5] stages of mouse neural development resulted in no apparent developmental defects. Neurogenesis in the ventricular-subventricular zone (V-SVZ) shortly after birth was also largely unaffected, except for a restricted ventral domain previously known to be regulated by Hh signaling. However, Kif3a and Ift88 genetic ablation also disrupts ependymal cilia, resulting in hydrocephalus by postnatal day 4. To directly study the role of B1 cells' primary cilia without the confounding effects of hydrocephalus, we stereotaxically targeted elimination of Kif3a from a subpopulation of radial glia, which resulted in ablation of primary cilia in a subset of B1 cells. Again, this experiment resulted in decreased neurogenesis only in the ventral V-SVZ. Primary cilia ablation led to disruption of Hh signaling in this subdomain. We conclude that primary cilia are required in a specific Hh-regulated subregion of the postnatal V-SVZ.


Assuntos
Cílios/fisiologia , Células-Tronco Neurais/classificação , Células-Tronco Neurais/ultraestrutura , Animais , Animais Recém-Nascidos , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proliferação de Células , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Feminino , Técnicas de Silenciamento de Genes , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas Hedgehog/fisiologia , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Cinesinas/metabolismo , Camundongos , Camundongos Transgênicos , Nestina/genética , Nestina/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Gravidez , Transdução de Sinais , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
13.
J Neurosci ; 35(15): 6142-52, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25878286

RESUMO

Several neural precursor populations contemporaneously generate neurons in the developing neocortex. Specifically, radial glial stem cells of the dorsal telencephalon divide asymmetrically to produce excitatory neurons, but also indirectly to produce neurons via three types of intermediate progenitor cells. Why so many precursor types are needed to produce neurons has not been established; whether different intermediate progenitor cells merely expand the output of radial glia or instead generate distinct types of neurons is unknown. Here we use a novel genetic fate mapping technique to simultaneously track multiple precursor streams in the developing mouse brain and show that layer 2 and 3 pyramidal neurons exhibit distinctive electrophysiological and structural properties depending upon their precursor cell type of origin. These data indicate that individual precursor subclasses synchronously produce functionally different neurons, even within the same lamina, and identify a primary mechanism leading to cortical neuronal diversity.


Assuntos
Linhagem da Célula/fisiologia , Neocórtex/citologia , Rede Nervosa/fisiologia , Células-Tronco Neurais/classificação , Células-Tronco Neurais/fisiologia , Células Piramidais/fisiologia , Animais , Eletroporação , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas Luminescentes/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Neocórtex/embriologia , Técnicas de Patch-Clamp , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
14.
Cell Tissue Res ; 349(1): 349-62, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22388657

RESUMO

Spinal cord injury (SCI) causes the irreversible loss of spinal cord parenchyma including astroglia, oligodendroglia and neurons. In particular, severe injuries can lead to an almost complete neural cell loss at the lesion site and structural and functional recovery might only be accomplished by appropriate cell and tissue replacement. Stem cells have the capacity to differentiate into all relevant neural cell types necessary to replace degenerated spinal cord tissue and can now be obtained from virtually any stage of development. Within the last two decades, many in vivo studies in small animal models of SCI have demonstrated that stem cell transplantation can promote morphological and, in some cases, functional recovery via various mechanisms including remyelination, axon growth and regeneration, or neuronal replacement. However, only two well-documented neural-stem-cell-based transplantation strategies have moved to phase I clinical trials to date. This review aims to provide an overview about the current status of preclinical and clinical neural stem cell transplantation and discusses future perspectives in the field.


Assuntos
Células-Tronco Neurais/transplante , Regeneração da Medula Espinal/fisiologia , Transplante de Células-Tronco , Animais , Ensaios Clínicos como Assunto , Humanos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/citologia
15.
J Neurosci ; 30(44): 14635-48, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21048121

RESUMO

Sox2 is expressed by neural stem and progenitor cells, and a sox2 enhancer identifies these cells in the forebrains of both fetal and adult transgenic mouse reporters. We found that an adenovirus encoding EGFP placed under the regulatory control of a 0.4 kb sox2 core enhancer selectively identified multipotential and self-renewing neural progenitor cells in dissociates of human fetal forebrain. Upon EGFP-based fluorescence-activated cell sorting (FACS), the E/sox2:EGFP(+) isolates were propagable for up to 1 year in vitro, and remained multilineage competent throughout. E/sox2:EGFP(+) cells expressed more telomerase enzymatic activity than matched E/sox2:EGFP-depleted populations, and maintained their telomeric lengths with successive passage. Gene expression analysis of E/sox2:EGFP-sorted neural progenitor cells, normalized to the unsorted forebrain dissociates from which they derived, revealed marked overexpression of genes within the notch and wnt pathways, and identified multiple elements of each pathway that appear selective to human neural progenitors. Sox2 enhancer-based FACS thus permits the prospective identification and direct isolation of a telomerase-active population of neural stem cells from the human fetal forebrain, and the elucidation of both the transcriptome and dominant signaling pathways of these critically important cells.


Assuntos
Células-Tronco Embrionárias/citologia , Elementos Facilitadores Genéticos/genética , Citometria de Fluxo/métodos , Células-Tronco Neurais/citologia , Fatores de Transcrição SOXB1/genética , Telomerase/biossíntese , Linhagem da Célula/genética , Separação Celular/métodos , Células Cultivadas , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/enzimologia , Feto , Humanos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/enzimologia , Estudos Prospectivos , Telomerase/genética
16.
Cell Physiol Biochem ; 26(6): 935-46, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21220924

RESUMO

Glutamate and its receptors are ascribed a pivotal role during acitivity-dependent neurogenesis. Nevertheless, their precise expression patterns during embryonic and adult differentiation remain elusive. An in vitro-approach that includes cells representing embryonic as well as adult neural stem cells that are both amenable to retinoic acid treatment is well-suited for assessing the developmental regulation of ionotropic glutamate receptors (iGluRs). The chosen system provides a continuous time line from embryonic to adult neurogenesis via two distinguishable cell populations, namely neuroepithelial precursors (NEPs) and radial glia-like neural stem cells (NSCs). We investigated the expression of cell type-specific differentiation markers and iGluR subunits before and after neuronal induction. A quantitative PCR assay was established for the determination of a hypothetical correlation of neuronal differentiation and iGluR expression. The NMDAR subunits NR1 and NR2B as well as the AMPAR subunit GluR2 present in Ca(2+)-impermeable AMPARs were found to be upregulated at the mRNA level in differentiated neuroepithelial precursors, indicating their likely contribution to neurotransmission after the first establishment of neuronal networks. Furthermore, with this approach, discrimination between NEPs and NSCs regarding their iGluR subunit expression patterns before and after the induction of neuronal differentiation was possible and pointed to diverse functions in these two cell types carried out by differentially assembled iGluRs.


Assuntos
Cálcio/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Diferenciação Celular , Camundongos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Receptores de AMPA/genética , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/fisiologia , Tretinoína/farmacologia
17.
Elife ; 92020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32762844

RESUMO

Different subtypes of interneurons, destined for the olfactory bulb, are continuously generated by neural stem cells located in the ventricular and subventricular zones along the lateral forebrain ventricles of mice. Neuronal identity in the olfactory bulb depends on the existence of defined microdomains of pre-determined neural stem cells along the ventricle walls. The molecular mechanisms underlying positional identity of these neural stem cells are poorly understood. Here, we show that the transcription factor Vax1 controls the production of two specific neuronal subtypes. First, it is directly necessary to generate Calbindin expressing interneurons from ventro-lateral progenitors. Second, it represses the generation of dopaminergic neurons by dorsolateral progenitors through inhibition of Pax6 expression. We present data indicating that this repression occurs, at least in part, via activation of microRNA miR-7.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese , Neuropeptídeos/metabolismo , Bulbo Olfatório/fisiologia , Fator de Transcrição PAX6/metabolismo , Animais , Calbindinas/genética , Diferenciação Celular , Feminino , Proteínas de Homeodomínio/genética , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neurais/classificação , Neuropeptídeos/genética , Fator de Transcrição PAX6/genética
18.
Epilepsy Res ; 150: 78-94, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30735971

RESUMO

Neurogenesis persists throughout life in the hippocampi of all mammals, including humans. In the healthy hippocampus, relatively quiescent Type-1 neural stem cells (NSCs) can give rise to more proliferative Type-2a neural progenitor cells (NPCs), which generate neuronal-committed Type-2b NPCs that mature into Type-3 neuroblasts. Many Type-3 neuroblasts survive and mature into functionally integrated granule neurons over several weeks. In kindling models of epilepsy, neurogenesis is drastically upregulated and many new neurons form aberrant connections that could support epileptogenesis and/or seizures. We have shown that sustained vector-mediated hippocampal somatostatin (SST) expression can both block epileptogenesis and reverse seizure susceptibility in fully kindled rats. Here we test whether adeno-associated virus (AAV) vector-mediated sustained SST expression modulates hippocampal neurogenesis and microglial activation in fully kindled rats. We found significantly more dividing Type-1 NSCs and a corresponding increased number of surviving new neurons in the hippocampi of kindled versus sham-kindled rats. Increased numbers of activated microglia were found in the granule cell layer and hilus of kindled rats at both time points. After intrahippocampal injection with either eGFP or SST-eGFP vector, we found similar numbers of dividing Type-1 NSCs and -2 NPCs and surviving BrdU+ neurons and glia in the hippocampi of kindled rats. Upon observed variability in responses to SST-eGFP (2/4 rats exhibited Grade 0 seizures in the test session), we conducted an additional experiment. We found significantly fewer dividing Type-1 NSCs in the hippocampi of SST-eGFP vector-treated responder rats (5/13 rats) relative to SST-eGFP vector-treated non-responders and eGFP vector-treated controls that exhibited high-grade seizures on the test session. The number of activated microglia was upregulated in the GCL and hilus of kindled rats, regardless of vector treatment. These data support the hypothesis that sustained SST expression exerts antiepileptic effects potentially through normalization of neurogenesis and suggests that abnormally high proliferating Type-1 NSC numbers may be a cellular mechanism of epilepsy.


Assuntos
Hipocampo/metabolismo , Hipocampo/patologia , Excitação Neurológica/patologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Somatostatina/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Antígenos/metabolismo , Bromodesoxiuridina/metabolismo , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Contagem de Células , Modelos Animais de Doenças , Estimulação Elétrica/efeitos adversos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Microglia/patologia , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/classificação , Neurônios/metabolismo , Proteoglicanas/metabolismo , Ratos , Ratos Sprague-Dawley , Somatostatina/genética , Transdução Genética , Ureia/análogos & derivados , Ureia/metabolismo
19.
Cell Transplant ; 25(4): 645-64, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26720923

RESUMO

Intracerebral cell transplantation is increasingly finding a clinical translation. However, the number of cells surviving after implantation is low (5-10%) compared to the number of cells injected. Although significant efforts have been made with regard to the investigation of apoptosis of cells after implantation, very little optimization of cell preparation and administration has been undertaken. Moreover, there is a general neglect of the biophysical aspects of cell injection. Cell transplantation can only be an efficient therapeutic approach if an optimal transfer of cells from the dish to the brain can be ensured. We therefore focused on the in vitro aspects of cell preparation of a clinical-grade human neural stem cell (NSC) line for intracerebral cell implantation. NSCs were suspended in five different vehicles: phosphate-buffered saline (PBS), Dulbecco's modified Eagle medium (DMEM), artificial cerebral spinal fluid (aCSF), HypoThermosol, and Pluronic. Suspension accuracy, consistency, and cell settling were determined for different cell volume fractions in addition to cell viability, cell membrane damage, and clumping. Maintenance of cells in suspension was evaluated while being stored for 8 h on ice, at room temperature, or physiological normothermia. Significant differences between suspension vehicles and cellular volume fractions were evident. HypoThermosol and Pluronic performed best, with PBS, aCSF, and DMEM exhibiting less consistency, especially in maintaining a suspension and preserving viability under different storage conditions. These results provide the basis to further investigate these preparation parameters during the intracerebral delivery of NSCs to provide an optimized delivery process that can ensure an efficient clinical translation.


Assuntos
Encéfalo , Técnicas de Cultura de Células/métodos , Células-Tronco Neurais/classificação , Transplante de Células-Tronco , Linhagem Celular , Humanos , Células-Tronco Neurais/metabolismo
20.
Sci Rep ; 6: 37540, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27857203

RESUMO

The CRISPR/Cas9 system is a rapid and customizable tool for gene editing in mammalian cells. In particular, this approach has widely opened new opportunities for genetic studies in neurological disease. Human neurons can be differentiated in vitro from hPSC (human Pluripotent Stem Cells), hNPCs (human Neural Precursor Cells) or even directly reprogrammed from fibroblasts. Here, we described a new platform which enables, rapid and efficient CRISPR/Cas9-mediated genome targeting simultaneously with three different paradigms for in vitro generation of neurons. This system was employed to inactivate two genes associated with neurological disorder (TSC2 and KCNQ2) and achieved up to 85% efficiency of gene targeting in the differentiated cells. In particular, we devised a protocol that, combining the expression of the CRISPR components with neurogenic factors, generated functional human neurons highly enriched for the desired genome modification in only 5 weeks. This new approach is easy, fast and that does not require the generation of stable isogenic clones, practice that is time consuming and for some genes not feasible.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/classificação , Sistemas CRISPR-Cas/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Inativação Gênica , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA