Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
EMBO Rep ; 23(9): e54078, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35861333

RESUMO

According to the current consensus, murine neural stem cells (NSCs) apically contacting the lateral ventricle generate differentiated progenitors by rare asymmetric divisions or by relocating to the basal side of the ventricular-subventricular zone (V-SVZ). Both processes will ultimately lead to the generation of adult-born olfactory bulb (OB) interneurons. In contrast to this view, we here find that adult-born OB interneurons largely derive from an additional NSC-type resident in the basal V-SVZ. Despite being both capable of self-renewal and long-term quiescence, apical and basal NSCs differ in Nestin expression, primary cilia extension and frequency of cell division. The expression of Notch-related genes also differs between the two NSC groups, and Notch activation is greatest in apical NSCs. Apical downregulation of Notch-effector Hes1 decreases Notch activation while increasing proliferation across the niche and neurogenesis from apical NSCs. Underscoring their different roles in neurogenesis, lactation-dependent increase in neurogenesis is paralleled by extra activation of basal but not apical NSCs. Thus, basal NSCs support OB neurogenesis, whereas apical NSCs impart Notch-mediated lateral inhibition across the V-SVZ.


Assuntos
Ventrículos Laterais , Células-Tronco Neurais , Animais , Diferenciação Celular/genética , Feminino , Ventrículos Laterais/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Bulbo Olfatório/metabolismo
2.
Stem Cells ; 35(12): 2417-2429, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28869691

RESUMO

The glycoprotein Prominin-1 and the carbohydrate Lewis X stage-specific embryonic antigen 1 (LeX-SSEA1) both have been extensively used as cell surface markers to purify neural stem cells (NSCs). While Prominin-1 labels a specialized membrane region in NSCs and ependymal cells, the specificity of LeX-SSEA1 expression and its biological significance are still unknown. To address these issues, we have here monitored the expression of the carbohydrate in neonatal and adult NSCs and in their progeny. Our results show that the percentage of immunopositive cells and the levels of LeX-SSEA1 immunoreactivity both increase with postnatal age across all stages of the neural lineage. This is associated with decreased proliferation in precursors including NSCs, which accumulate the carbohydrate at the cell surface while remaining quiescent. Exposure of precursors to bone morphogenetic protein (BMP) increases LEX-SSEA1 expression, which promotes cell cycle withdrawal by a mechanism involving LeX-SSEA1-mediated interaction at the cell surface. Conversely, interference with either BMP signaling or with LeX-SSEA1 promotes proliferation to a similar degree. Thus, in the postnatal germinal niche, the expression of LeX-SSEA1 increases with age and exposure to BMP signaling, thereby downregulating the proliferation of subependymal zone precursors including NSCs. Stem Cells 2017;35:2417-2429.


Assuntos
Antígenos CD15/metabolismo , Células-Tronco Neurais/metabolismo , Antígeno AC133/genética , Antígeno AC133/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proliferação de Células/genética , Proliferação de Células/fisiologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Antígenos CD15/genética , Camundongos , Células-Tronco Neurais/citologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
3.
Development ; 141(4): 773-83, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24496615

RESUMO

The activation of epidermal growth factor receptor (EGFR) affects multiple aspects of neural precursor behaviour, including proliferation and migration. Telencephalic precursors acquire EGF responsiveness and upregulate EGFR expression at late stages of development. The events regulating this process and its significance are still unclear. We here show that in the developing and postnatal hippocampus (HP), growth/differentiation factor (GDF) 15 and EGFR are co-expressed in primitive precursors as well as in more differentiated cells. We also provide evidence that GDF15 promotes responsiveness to EGF and EGFR expression in hippocampal precursors through a mechanism that requires active CXC chemokine receptor (CXCR) 4. Besides EGFR expression, GDF15 ablation also leads to decreased proliferation and migration. In particular, lack of GDF15 impairs both processes in the cornu ammonis (CA) 1 and only proliferation in the dentate gyrus (DG). Importantly, migration and proliferation in the mutant HP were altered only perinatally, when EGFR expression was also affected. These data suggest that GDF15 regulates migration and proliferation by promoting EGFR signalling in the perinatal HP and represent a first description of a functional role for GDF15 in the developing telencephalon.


Assuntos
Animais Recém-Nascidos , Movimento Celular/fisiologia , Receptores ErbB/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Hipocampo/crescimento & desenvolvimento , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Bromodesoxiuridina , Carbocianinas , Proliferação de Células , Citometria de Fluxo , Fluorescência , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/metabolismo , Imuno-Histoquímica , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR4/metabolismo , beta-Galactosidase/metabolismo
4.
J Biol Chem ; 290(7): 4343-55, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25540202

RESUMO

Activation of γ-aminobutyric A receptors (GABA(A)Rs) in the subependymal zone (SEZ) induces hyperpolarization and osmotic swelling in precursors, thereby promoting surface expression of the epidermal growth factor receptor (EGFR) and cell cycle entry. However, the mechanisms underlying the GABAergic modulation of cell swelling are unclear. Here, we show that GABA(A)Rs colocalize with the water channel aquaporin (AQP) 4 in prominin-1 immunopositive (P(+)) precursors in the postnatal SEZ, which include neural stem cells. GABA(A)R signaling promotes AQP4 expression by decreasing serine phosphorylation associated with the water channel. The modulation of AQP4 expression by GABA(A)R signaling is key to its effect on cell swelling and EGFR expression. In addition, GABA(A)R function also affects the ability of neural precursors to swell in response to an osmotic challenge in vitro and in vivo. Thus, the regulation of AQP4 by GABA(A)Rs is involved in controlling activation of neural stem cells and water exchange dynamics in the SEZ.


Assuntos
Aquaporina 4/fisiologia , Epêndima/metabolismo , Regulação da Expressão Gênica , Ventrículos Laterais/metabolismo , Receptores de GABA-A/metabolismo , Água/metabolismo , Animais , Western Blotting , Tamanho Celular , Células Cultivadas , Epêndima/citologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Imunofluorescência , Imunoprecipitação , Ventrículos Laterais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pressão Osmótica , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de GABA-A/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
5.
Life Sci Alliance ; 7(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38719753

RESUMO

We recently reported that growth/differentiation factor 15 (GDF15) and its receptor GDNF family receptor alpha-like (GFRAL) are expressed in the periventricular germinal epithelium thereby regulating apical progenitor proliferation. However, the mechanisms are unknown. We now found GFRAL in primary cilia and altered cilia morphology upon GDF15 ablation. Mutant progenitors also displayed increased histone deacetylase 6 (Hdac6) and ciliary adenylate cyclase 3 (Adcy3) transcript levels. Consistently, microtubule acetylation, endogenous sonic hedgehog (SHH) activation and ciliary ADCY3 were all affected in this group. Application of exogenous GDF15 or pharmacological antagonists of either HDAC6 or ADCY3 similarly normalized ciliary morphology, proliferation and SHH signalling. Notably, Gdf15 ablation affected Hdac6 expression and cilia length only in the mutant periventricular niche, in concomitance with ciliary localization of GFRAL. In contrast, in the hippocampus, where GFRAL was not expressed in the cilium, progenitors displayed altered Adcy3 expression and SHH signalling, but Hdac6 expression, cilia morphology and ciliary ADCY3 levels remained unchanged. Thus, ciliary signalling underlies the effect of GDF15 on primary cilia elongation and proliferation in apical progenitors.


Assuntos
Adenilil Ciclases , Proliferação de Células , Cílios , Proteínas Hedgehog , Desacetilase 6 de Histona , Transdução de Sinais , Animais , Camundongos , Acetilação , Adenilil Ciclases/metabolismo , Adenilil Ciclases/genética , Proliferação de Células/genética , Cílios/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Desacetilase 6 de Histona/metabolismo , Desacetilase 6 de Histona/genética , Camundongos Knockout , Células-Tronco/metabolismo , Células-Tronco/citologia
6.
Stem Cell Reports ; 19(3): 351-365, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38366596

RESUMO

The expression of growth/differentiation factor (GDF) 15 increases in the ganglionic eminence (GE) late in neural development, especially in neural stem cells (NSCs). However, GDF15 function in this region remains unknown. We report that GDF15 receptor is expressed apically in the GE and that GDF15 ablation promotes proliferation and cell division in the embryonic GE and in the adult ventricular-subventricular zone (V-SVZ). This causes a transient generation of additional neuronal progenitors, compensated by cell death, and a lasting increase in the number of ependymal cells and apical NSCs. Finally, both GDF15 receptor and the epidermal growth factor receptor (EGFR) were expressed in progenitors and mutation of GDF15 affected EGFR signaling. However, only exposure to exogenous GDF15, but not to EGF, normalized proliferation and the number of apical progenitors. Thus, GDF15 regulates proliferation of apical progenitors in the GE, thereby affecting the number of ependymal cells and NSCs.


Assuntos
Ventrículos Laterais , Células-Tronco Neurais , Receptores ErbB/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Contagem de Células , Proliferação de Células , Diferenciação Celular/fisiologia
7.
Methods Cell Biol ; 175: 17-31, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36967140

RESUMO

Primary cilia provide a specialized subcellular environment favoring ordered and timely interaction and modification of signaling molecules, necessary for the sensing and transduction of extracellular signals and environmental conditions. Crucial to the understanding of ciliary function is the knowledge of the signaling molecules composing the ciliary compartment. While proteomes of primary cilia have been published recently, the selective isolation of primary cilia from specific cell types and whole tissue still proves difficult, and many laboratories instead resort to the analysis of cultured cells, which may introduce experimental artifacts. Here we present a flow cytometry-based method to isolate and characterize primary cilia from the murine ventricular-subventricular zone. After deciliation, primary cilia are immunolabeled with antibodies against ciliary markers. As an example, we here use a double-staining with acetylated tubulin, which stains the ciliary axoneme, and ciliary membrane protein ADP-ribosylation-like factor 13b (Arl13b); additionally, we triple-labeled primary cilia using the ciliary marker adenylate cyclase 3 (AC3). Besides analysis at the single particle level, fluorescence activated cell sorting (FACS) allows collection of pure preparations of primary cilia suited for subsequent proteomic analyses like mass spectrometry or western blot. As an example of analytical application, we performed triple immunostaining and FACS analysis to reveal cilia heterogeneity. Thus, our cilia isolation method, which can readily be applied to other tissues or cell culture, will facilitate the study of this key cellular organelle and shed light on its role in normal conditions and disease.


Assuntos
Cílios , Proteômica , Animais , Camundongos , Cílios/metabolismo , Citometria de Fluxo , Tubulina (Proteína)/metabolismo , Transporte Proteico
8.
Stem Cells ; 29(9): 1415-26, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21714038

RESUMO

Niche homeostasis in the postnatal subependymal zone of the lateral ventricle (lSEZ) requires coordinated proliferation and differentiation of neural progenitor cells. The mechanisms regulating this balance are scarcely known. Recent observations indicate that the orphan nuclear receptor Tlx is an intrinsic factor essential in maintaining this balance. However, the effect of Tlx on gene expression depends on age and cell-type cues. Therefore, it is essential to establish its expression pattern at different developmental ages. Here, we show for the first time that in the neonatal lSEZ activated neural stem cells (NSCs) and especially transit-amplifying progenitors (TAPs) express Tlx and that its expression may be regulated at the posttranscriptional level. We also provide evidence that in both cell types Tlx affects gene expression in a positive and negative manner. In activated NSCs, but not in TAPs, absence of Tlx leads to overexpression of negative cell cycle regulators and impairment of proliferation. Moreover, in both cell types, the homeobox transcription factor Dlx2 is downregulated in the absence of Tlx. This is paralleled by increased expression of Olig2 in activated NSCs and glial fibrillary acidic protein in TAPs, indicating that in both populations Tlx decreases gliogenesis. Consistent with this, we found a higher proportion of cells expressing glial makers in the neonatal lSEZ of mutant mice than in the wild type counterpart. Thus, Tlx playing a dual role affects the expression of distinct genes in these two lSEZ cell types.


Assuntos
Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Feminino , Genótipo , Ventrículos Laterais/crescimento & desenvolvimento , Camundongos , Neurogênese , Gravidez , Receptores Citoplasmáticos e Nucleares/genética
9.
Stem Cells ; 29(2): 307-19, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21732488

RESUMO

Signal-regulated changes in cell size affect cell division and survival and therefore are central to tissue morphogenesis and homeostasis. In this respect, GABA receptors (GABA(A)Rs) are of particular interest because allowing anions flow across the cell membrane modulates the osmolyte flux and the cell volume. Therefore, we have here investigated the hypothesis that GABA may regulate neural stem cell proliferation by inducing cell size changes. We found that, besides neuroblasts, also neural precursors in the neonatal murine subependymal zone sense GABA via GABA(A) Rs. However, unlike in neuroblasts, where it induced depolarization-mediated [Ca(2+)](i) increase, GABA(A) Rs activation in precursors caused hyperpolarization. This resulted in osmotic swelling and increased surface expression of epidermal growth factor receptors (EGFRs). Furthermore, activation of GABA(A) Rs signaling in vitro in the presence of EGF modified the expression of the cell cycle regulators, phosphatase and tensin homolog and cyclin D1, increasing the pool of cycling precursors without modifying cell cycle length. A similar effect was observed on treatment with diazepam. We also demonstrate that GABA and diazepam responsive precursors represent prominin(+) stem cells. Finally, we show that as in in vitro also in in vivo a short administration of diazepam promotes EGFR expression in prominin(+) stem cells causing activation and cell cycle entry. Thus, our data indicate that endogenous GABA is a part of a regulatory mechanism of size and cell cycle entry of neonatal stem cells. Our results also have potential implications for the therapeutic practices that involve exposure to GABA(A) Rs modulators during neurodevelopment.


Assuntos
Antígenos CD/metabolismo , Ciclo Celular/efeitos dos fármacos , Glicoproteínas/metabolismo , Células-Tronco Neurais/metabolismo , Peptídeos/metabolismo , Receptores de GABA-A/metabolismo , Transdução de Sinais , Antígeno AC133 , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , Diazepam/farmacologia , Receptores ErbB/biossíntese , Receptores ErbB/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Pressão Osmótica/fisiologia , Precursores de Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo
10.
Glia ; 59(4): 615-26, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21294160

RESUMO

In the adult mammalian brain, neurogenesis originates from astrocyte-like stem cells. We generated a transgenic mouse line in which the tetracycline dependent transactivator (tTA) is expressed under the control of the murine GFAP promoter. In this mouse line, inducible gene expression targets virtually all GFAP-expressing stem-like cells in the dentate gyrus and a subset of GFAP-expressing progenitors located primarily in the dorsal wall/dorsolateral corner of the subventricular zone. Outside the neurogenic zones, astrocytes are infrequently targeted. We introduce a panel of transgenic mice which allow both inducible expression of candidate genes under control of the murine GFAP promoter and, at the same time, lineage tracing of all cells descendant from the original GFAP-positive cell. This new mouse line represents a versatile tool for functional analysis of neurogenesis and lineage tracing.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Células-Tronco/metabolismo , Animais , Linhagem Celular , Expressão Gênica , Proteína Glial Fibrilar Ácida , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Regiões Promotoras Genéticas
11.
Mol Cell Neurosci ; 44(4): 318-29, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20417282

RESUMO

Precursors within the subventricular zone (SVZ) exhibit regional variations in the expression of transcription factors important for the regulation of their proliferation and differentiation. In the anterior SVZ (aSVZ) the homeobox transcription factor distalless (Dlx)2 modulates both processes by promoting neural stem cell (NSC) activation as well as neurogenesis. Activated NSCs and transit-amplifying precursors (TAPs) in the aSVZ both express high levels of epidermal growth factor receptor (EGFR(high)) and form clones in response to exogenous EGF. EGF-responsive cells are also present in the hippocampal subependyma (hSVZ). However, it is not clear whether they represent NSCs or TAPs and whether their proliferation and differentiation are regulated as in the aSVZ. Here we have purified EGFR(high) cells from both the aSVZ and hSVZ at different ages. When isolated from perinatal tissue both populations were enriched in multipotent clonogenic precursors, which generated GABAergic neurons. Although they differed in absolute expression levels, activated NSCs and TAPs in both regions displayed similar signatures of transcription factor expression. However, activated NSCs were less frequent in the hSVZ than in the aSVZ. Furthermore, increasing age had a greater inhibitory effect on NSC proliferation in the hSVZ than in the aSVZ. This suggests that NSC activation is differentially regulated in the two regions. Consistent with this hypothesis, we found that in hippocampal precursors Dlx2 promoted neurogenesis but not NSC activation. Thus, most clonogenic EGFR(high) precursors in the hSVZ represent TAPs and NSC proliferation in the aSVZ and hSVZ is regulated by different mechanisms.


Assuntos
Receptores ErbB/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Nicho de Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Receptores ErbB/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Neurônios/citologia , Especificidade de Órgãos , Gravidez , Nicho de Células-Tronco/citologia
12.
Stem Cells ; 27(6): 1443-54, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489104

RESUMO

In the adult subventricular zone (SVZ), astroglial stem cells generate transit-amplifying precursors (TAPs). Both stem cells and TAPs form clones in response to epidermal growth factor (EGF). However, in vivo, in the absence of sustained EGF receptor (EGFR) activation, TAPs divide a few times before differentiating into neuroblasts. The lack of suitable markers has hampered the analysis of stem cell lineage progression and associated functional changes in the neonatal germinal epithelium. Here we purified neuroblasts and clone-forming precursors from the neonatal SVZ using expression levels of EGFR and polysialylated neural cell adhesion molecule (PSANCAM). As in the adult SVZ, most neonatal clone-forming precursors did not express the neuroglia proteoglycan 2 (NG2) but displayed characteristics of TAPs, and only a subset exhibited antigenic characteristics of astroglial stem cells. Both precursors and neuroblasts were PSANCAM(+); however, neuroblasts also expressed doublecortin and functional voltage-dependent Ca(2+) channels. Neuroblasts and precursors had distinct outwardly rectifying K(+) current densities and passive membrane properties, particularly in precursors contacting each other, because of the contribution of gap junction coupling. Confirming the hypothesis that most are TAPs, cell tracing in brain slices revealed that within 2 days the majority of EGFR(+) cells had exited the cell cycle and differentiated into a progenitor displaying intermediate antigenic and functional properties between TAPs and neuroblasts. Thus, distinct functional and antigenic properties mark stem cell lineage progression in the neonatal SVZ.


Assuntos
Encéfalo/citologia , Linhagem da Célula , Neurogênese/fisiologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Encéfalo/metabolismo , Diferenciação Celular , Receptores ErbB/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Proteoglicanas/metabolismo
13.
Mol Cell Neurosci ; 42(4): 308-14, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19683576

RESUMO

In the postnatal subventricular zone (SVZ) neural stem cells (NSCs) give rise to transit-amplifying precursors (TAPs) expressing high levels of epidermal growth factor receptor (EGFR) that in turn generate neuroblasts. Both TAPs and neuroblasts express distal-less (DLX)2 homeobox transcription factor but the latter proliferate less. Modulation of its expression in vivo has revealed that DLX2 affects both neurogenesis and proliferation in the postnatal SVZ. However, the mechanisms underlying these effects are not clear. To investigate this issue we have here forced the expression of DLX2 in SVZ isolated NSCs growing in defined in vitro conditions. This analysis revealed that DLX2 affects the proliferation of SVZ precursors by regulating two distinct steps of neural lineage progression. Firstly, it promotes the lineage transition from NSCs to TAPs. Secondly it enhances the proliferative response of neuronal progenitors to EGF. Thus DLX2 and EGFR signalling interact at multiple levels to coordinate proliferation in the postnatal SVZ.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Proteínas de Homeodomínio/metabolismo , Neurogênese/fisiologia , Neurônios/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Proteínas de Homeodomínio/genética , Camundongos , Neurônios/citologia , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/citologia , Células-Tronco/citologia , Fatores de Transcrição/genética
14.
Stem Cell Reports ; 13(1): 132-146, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31178417

RESUMO

In the adult subependymal zone (SEZ), neural stem cells (NSCs) apically contacting the lateral ventricle on activation generate progenitors proliferating at the niche basal side. We here show that Tailless (TLX) coordinates NSC activation and basal progenitor proliferation by repressing the NOTCH effector Hes1. Consistent with this, besides quiescence Hes1 expression also increases on Tlx mutation. Since HES1 levels are higher at the apical SEZ, NOTCH activation is increased in Tlx-/- NSCs, but not in surrounding basal progenitors. Underscoring the causative relationship between higher HES1/NOTCH and increased quiescence, downregulation of Hes1 only in mutant NSCs normalizes NOTCH activation and resumes proliferation and neurogenesis not only in NSCs, but especially in basal progenitors. Since pharmacological blockade of NOTCH signaling also promotes proliferation of basal progenitors, we conclude that TLX, by repressing Hes1 expression, counteracts quiescence and NOTCH activation in NSCs, thereby relieving NOTCH-mediated lateral inhibition of proliferation in basal progenitors.


Assuntos
Regulação da Expressão Gênica , Ventrículos Laterais/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1/genética , Animais , Diferenciação Celular , Linhagem da Célula/genética , Proliferação de Células , Células Cultivadas , Imunofluorescência , Ventrículos Laterais/citologia , Camundongos , Camundongos Knockout , Mutação , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo
15.
Eur J Neurosci ; 27(2): 269-83, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18190521

RESUMO

Neuronal activity regulates neurogenesis and neuronal differentiation in the mammalian brain. The commencement of neurotransmitter expression establishes the neuronal phenotype and enables the formation of functional connectivity between neurons. In addition, release of neurotransmitters from differentiating neurons may modulate the behaviour of neural precursors. Here, we show that neuronal activity regulates gamma-aminobutyric acid (GABA) expression in neurons generated from stem cells of the striatum and adult subventricular zone (SVZ). Differentiating neurons display spontaneous Ca2+ events, which are voltage-gated calcium channel (VGCC) dependent. Depolarization increases both the frequency of Ca2+ transients and the amount of Ca2+ influx in differentiating neurons. We show that depolarization-dependent GABA expression is regulated by the amplitude and not by the frequency of Ca2+ influx. Brief activation of VGCCs leads to Ca2+ influx that in turn promotes a rapid expression of GABA. Depolarization-dependent GABA expression does not require changes in gene expression. Instead, it involves cAMP-dependent protein kinase (PKA) and Ca2+ and phospholipid-dependent protein kinase (PKC) signalling. Activity increases the number of glutamic acid decarboxylase (GAD) 65-immunoreactive neurons in a PKA-dependent manner, without altering the expression of GAD 65, suggesting that depolarization promotes recruitment of GAD 65 by a post-translational mechanism. In line with this, depolarization does not permanently increase the expression of GABA in neurons derived from neural stem cells of the embryonic striatum, cortex and adult SVZ. Thus, neuronal activity does not merely accelerate neuronal differentiation but it may alter the mechanism of GABA synthesis in newly generated neurons.


Assuntos
Glutamato Descarboxilase/biossíntese , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Células-Tronco/metabolismo , Ácido gama-Aminobutírico/biossíntese , Potenciais de Ação/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Diferenciação Celular/fisiologia , Feminino , Glutamato Descarboxilase/genética , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Gravidez , Células-Tronco/citologia , Ácido gama-Aminobutírico/genética
16.
Eur J Neurosci ; 28(5): 871-82, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18717733

RESUMO

It is known that activity modulates neuronal differentiation in the adult brain but the signalling mechanisms underlying this process remain to be identified. We show here that activity requires soluble amyloid precursor protein (sAPP) to enhance neurite outgrowth of young neurons differentiating from neural stem cells. Inhibition of sAPP secretion and anti-APP antibodies both abolished the effect of depolarization on neurite outgrowth, whereas exogenous sAPPalpha, similar to depolarization, induced neurite elongation. Depolarization and sAPPalpha both required active N-methyl-D-aspartic acid receptor (NMDAR) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) recruitment to induce neurite outgrowth. However, depolarization and sAPPalpha played different roles in modulating this signalling cascade. Depolarization induced ERK phosphorylation with fast kinetics via activation of NMDAR. By contrast, acute application of sAPPalpha did not lead to ERK activation. However, continuous generation of sAPPalpha was necessary for depolarization-induced ERK phosphorylation, indicating that sAPPalpha promotes MAPK/ERK recruitment by an indirect mechanism. In addition, we found that blockade of NMDAR down-regulated APP expression, whereas depolarization increased sAPPalpha, suggesting that activity may also act upstream of sAPP signalling by regulating the amount of cellular APP and extracellular sAPPalpha. Finally, we show that soluble amyloid precursor-like protein 2 (sAPLP2), but not sAPLP1, is functionally redundant to sAPP in promoting neurite outgrowth and that soluble members of the APP family require membrane-bound APP to enhance neurite outgrowth. In summary, these experiments indicate a novel role of APP family members in activity-dependent neuronal differentiation.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neuritos/enzimologia , Neurogênese/fisiologia , Células-Tronco/enzimologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Encéfalo/citologia , Encéfalo/embriologia , Células Cultivadas , Citoproteção/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuritos/ultraestrutura , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Esferoides Celulares , Células-Tronco/citologia
17.
Front Cell Neurosci ; 12: 519, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30692915

RESUMO

In the adult mammalian brain, the apical surface of the subependymal zone (SEZ) is covered by many motile ependymal cilia and a few primary cilia originating from rare intermingled neural stem cells (NSCs). In NSCs the primary cilia are key for the transduction of essential extracellular signals such as Sonic hedgehog (SHH) and platelet-derived growth factor (PDGF). Despite their importance, the analysis of NSC primary cilia is greatly hampered by the fact that they are overwhelmingly outnumbered by the motile cilia. We here take advantage of flow cytometry to purify the two cilia types and allow their molecular characterization. Primary cilia were identified based on immunoreactivity to the marker adenylate cyclase type III (AC3) and differential levels of prominin-1 whereas motile cilia displayed immunoreactivity only to the latter. Consistent with the morphological differences between the two classes of cilia, enrichment of motile cilia positively correlated with size. Moreover, we observed age-dependent variations in the abundance of the two groups of ciliary organelles reflecting the changes associated with their development. The two cilia groups also differed with respect to the expression of signaling molecules, since PDGF receptor (PDGFR)α, smoothened (Smo) and CXC chemokine receptor (CXCR)4 were only detected in isolated primary but not motile cilia. Thus, our novel method of cilia isolation and characterization by flow cytometry has the potential to be extended to the study of cilia from different tissues and organs, providing a powerful tool for the investigation of primary cilia in physiological and pathological conditions.

18.
J Neurosci ; 23(1): 103-11, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12514206

RESUMO

Neural stem cells can generate in vitro progenitors of the three main cell lineages found in the CNS. The signaling pathways underlying the acquisition of differentiated phenotypes in these cells are poorly understood. Here we tested the hypothesis that Ca(2+) signaling controls differentiation of neural precursors. We found low-frequency global and local Ca(2+) transients occurring predominantly during early stages of differentiation. Spontaneous Ca(2+) signals in individual precursors were not synchronized with Ca(2+) transients in surrounding cells. Experimentally induced changes in the frequency of local Ca(2+) signals and global Ca(2+) rises correlated positively with neurite outgrowth and the onset of GABAergic neurotransmitter phenotype, respectively. NMDA receptor activity was critical for alterations in neuronal morphology but not for the timing of the acquisition of the neurotransmitter phenotype. Thus, spontaneous Ca(2+) signals are an intrinsic property of differentiating neurosphere-derived precursors. Their frequency may specify neuronal morphology and acquisition of neurotransmitter phenotype.


Assuntos
Sinalização do Cálcio , Sistema Nervoso Central/embriologia , Neuritos/metabolismo , Neuritos/ultraestrutura , Células-Tronco/fisiologia , Ácido gama-Aminobutírico/biossíntese , Animais , Diferenciação Celular , Células Cultivadas , Sistema Nervoso Central/citologia , Cinética , Camundongos , Neurônios/citologia , Fenótipo , Cloreto de Potássio/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo
19.
Neurogenesis (Austin) ; 1(1): e970883, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27504469

RESUMO

The transcription factor CREB (cAMP-response element binding protein) regulates differentiation, migration, survival and activity-dependent gene expression in the developing and mature nervous system. However, its specific role in the proliferation of embryonic neural progenitors is still not completely understood. Here we investigated how CREB regulates proliferation of mouse embryonic neural progenitors by a conditional mutant lacking Creb gene in neural progenitors. In parallel, we explored possible compensatory effects by the genetic ablation of another member of the same gene family, the cAMP-responsive element modulator (Crem). We show that CREB loss differentially impaired the proliferation, clonogenic potential and self-renewal of precursors derived from the ganglionic eminence (GE), in comparison to those derived from the cortex. This phenotype was associated with a specific reduction of histone acetylation in the GE of CREB mutant mice, and this reduction was rescued in vivo by inhibition of histone deacetylation. These observations indicate that the impaired proliferation could be caused by a reduced acetyltransferase activity in Creb conditional knock-out mice. These findings support a crucial role of CREB in controlling embryonic neurogenesis and propose a novel mechanism by which CREB regulates embryonic neural development.

20.
Sci Rep ; 4: 3803, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24448162

RESUMO

Neural stem cells (NSCs) generate new neurons in vivo and in vitro throughout adulthood and therefore are physiologically and clinically relevant. Unveiling the mechanisms regulating the lineage progression from NSCs to newborn neurons is critical for the transition from basic research to clinical application. However, the direct analysis of NSCs and their progeny is still elusive due to the problematic identification of the cells. We here describe the isolation of highly purified genetically unaltered NSCs and transit-amplifying precursors (TAPs) from the adult subependymal zone (SEZ). Using this approach we show that a primary cilium and high levels of epidermal growth factor receptor (EGFR) at the cell membrane characterize quiescent and cycling NSCs, respectively. However, we also observed non-ciliated quiescent NSCs and NSCs progressing into the cell cycle without up-regulating EGFR expression. Thus, the existence of NSCs displaying distinct molecular and structural conformations provides more flexibility to the regulation of quiescence and cell cycle progression.


Assuntos
Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Cílios/fisiologia , Epêndima/citologia , Células-Tronco Neurais/citologia , Neurônios/citologia , Animais , Animais Recém-Nascidos , Apoptose , Western Blotting , Ciclo Celular , Linhagem da Célula , Membrana Celular/metabolismo , Células Cultivadas , Epêndima/metabolismo , Receptores ErbB/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA