Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
EMBO Rep ; 24(12): e57268, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37987220

RESUMO

Intermittent fasting (IF) is a promising strategy to counteract ageing shown to increase the number of adult-born neurons in the dentate gyrus of mice. However, it is unclear which steps of the adult neurogenesis process are regulated by IF. The number of adult neural stem cells (NSCs) decreases with age in an activation-dependent manner and, to counteract this loss, adult NSCs are found in a quiescent state which ensures their long-term maintenance. We aimed to determine if and how IF affects adult NSCs in the hippocampus. To identify the effects of every-other-day IF on NSCs and all following steps in the neurogenic lineage, we combined fasting with lineage tracing and label retention assays. We show here that IF does not affect NSC activation or maintenance and, that contrary to previous reports, IF does not increase neurogenesis. The same results are obtained regardless of strain, sex, diet length, tamoxifen administration or new-born neuron identification method. Our data suggest that NSCs maintain homeostasis upon IF and that this intervention is not a reliable strategy to increase adult neurogenesis.


Assuntos
Células-Tronco Adultas , Células-Tronco Neurais , Camundongos , Animais , Jejum Intermitente , Neurogênese , Neurônios , Hipocampo , Células-Tronco Adultas/fisiologia
2.
Development ; 148(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33558315

RESUMO

Quiescence is a cellular state in which a cell remains out of the cell cycle but retains the capacity to divide. The unique ability of adult stem cells to maintain quiescence is crucial for life-long tissue homeostasis and regenerative capacity. Quiescence has long been viewed as an inactive state but recent studies have shown that it is in fact an actively regulated process and that adult stem cells are highly reactive to extrinsic stimuli. This has fuelled hopes of boosting the reactivation potential of adult stem cells to improve tissue function during ageing. In this Review, we provide a perspective of the quiescent state and discuss how quiescent adult stem cells transition into the cell cycle. We also discuss current challenges in the field, highlighting recent technical advances that could help overcome some of these challenges.


Assuntos
Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Diferenciação Celular , Proliferação de Células , Epigenômica , Homeostase/fisiologia , Humanos
3.
Development ; 148(20)2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34557919

RESUMO

Adult mouse hippocampal neural stem cells (NSCs) generate new neurons that integrate into existing hippocampal networks and modulate mood and memory. These NSCs are largely quiescent and are stimulated by niche signals to activate and produce neurons. Wnt/ß-catenin signalling acts at different steps along the hippocampal neurogenic lineage, but whether it has a direct role in the regulation of NSCs remains unclear. Here, we used Wnt/ß-catenin reporters and transcriptomic data from in vivo and in vitro models to show that adult NSCs respond to Wnt/ß-catenin signalling. Wnt/ß-catenin stimulation instructed the neuronal differentiation of proliferating NSCs and promoted the activation or differentiation of quiescent NSCs in a dose-dependent manner. However, deletion of ß-catenin in NSCs did not affect either their activation or maintenance of their stem cell characteristics. Together, these results indicate that, although NSCs do respond to Wnt/ß-catenin stimulation in a dose-dependent and state-specific manner, Wnt/ß-catenin signalling is not cell-autonomously required to maintain NSC homeostasis, which reconciles some of the contradictions in the literature as to the role of Wnt/ß-catenin signalling in adult hippocampal NSCs.


Assuntos
Homeostase/fisiologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Neurogênese/fisiologia , Neurônios/metabolismo
4.
Genes Dev ; 27(16): 1769-86, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23964093

RESUMO

The majority of neural stem cells (NSCs) in the adult brain are quiescent, and this fraction increases with aging. Although signaling pathways that promote NSC quiescence have been identified, the transcriptional mechanisms involved are mostly unknown, largely due to lack of a cell culture model. In this study, we first demonstrate that NSC cultures (NS cells) exposed to BMP4 acquire cellular and transcriptional characteristics of quiescent cells. We then use epigenomic profiling to identify enhancers associated with the quiescent NS cell state. Motif enrichment analysis of these enhancers predicts a major role for the nuclear factor one (NFI) family in the gene regulatory network controlling NS cell quiescence. Interestingly, we found that the family member NFIX is robustly induced when NS cells enter quiescence. Using genome-wide location analysis and overexpression and silencing experiments, we demonstrate that NFIX has a major role in the induction of quiescence in cultured NSCs. Transcript profiling of NS cells overexpressing or silenced for Nfix and the phenotypic analysis of the hippocampus of Nfix mutant mice suggest that NFIX controls the quiescent state by regulating the interactions of NSCs with their microenvironment.


Assuntos
Epigênese Genética , Fatores de Transcrição NFI/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Animais , Proteína Morfogenética Óssea 4/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Elementos Facilitadores Genéticos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/efeitos dos fármacos , Ligação Proteica
5.
Cell Stem Cell ; 30(4): 333-334, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-37028395

RESUMO

Alzheimer's disease (AD) is associated with reduced adult hippocampal neurogenesis and impaired hippocampal-dependent behaviors. Li et al.1 report that stimulating adult neurogenesis combined with new-born neuron activation ameliorates behavioral symptoms and plaque deposition in AD mouse models. This supports boosting adult neurogenesis as a potential therapeutic approach for AD-related cognitive decline.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Doença de Alzheimer/terapia , Camundongos Transgênicos , Hipocampo , Neurônios , Neurogênese/fisiologia , Modelos Animais de Doenças
6.
Front Neurosci ; 16: 878875, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35431774

RESUMO

The majority of adult neural stem cells (aNSCs) are in a distinct metabolic state of reversible cell cycle exit also known as quiescence. The rate of aNSC activation determines the number of new neurons generated and directly influences the long-term maintenance of neurogenesis. Despite its relevance, it is still unclear how aNSC quiescence is regulated. Many factors contribute to this, like aNSC heterogeneity, the lack of reliable quiescence markers, the complexity of the neurogenic niches or the intricacy of the transcriptional and post-transcriptional mechanisms involved. In this perspective article I discuss possible solutions to these problems. But, first and foremost, I believe we require a model that goes beyond a simple transition toward activation. Instead, we must acknowledge the full complexity of aNSC states, which include not only activation but also differentiation and survival as behavioural outcomes. I propose a model where aNSCs dynamically transition through a cloud of highly interlinked cellular states driven by intrinsic and extrinsic cues. I also show how a new perspective enables us to integrate current results into a coherent framework leading to the formulation of new testable hypothesis. This model, like all others, is still far from perfect and will be reshaped by future findings. I believe that having a more complete view of aNSC transitions and embracing their complexity will bring us closer to understand how aNSC activity and neurogenesis are controlled throughout life.

7.
Cell Stem Cell ; 28(5): 863-876.e6, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33581058

RESUMO

Neural stem cell numbers fall rapidly in the hippocampus of juvenile mice but stabilize during adulthood, ensuring lifelong hippocampal neurogenesis. We show that this stabilization of stem cell numbers in young adults is the result of coordinated changes in stem cell behavior. Although proliferating neural stem cells in juveniles differentiate rapidly, they increasingly return to a resting state of shallow quiescence and progress through additional self-renewing divisions in adulthood. Single-cell transcriptomics, modeling, and label retention analyses indicate that resting cells have a higher activation rate and greater contribution to neurogenesis than dormant cells, which have not left quiescence. These changes in stem cell behavior result from a progressive reduction in expression of the pro-activation protein ASCL1 because of increased post-translational degradation. These cellular mechanisms help reconcile current contradictory models of hippocampal neural stem cell (NSC) dynamics and may contribute to the different rates of decline of hippocampal neurogenesis in mammalian species, including humans.


Assuntos
Células-Tronco Adultas , Células-Tronco Neurais , Animais , Proliferação de Células , Hipocampo , Camundongos , Neurogênese
8.
Cell Stem Cell ; 27(4): 508-510, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33007233

RESUMO

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.

9.
Neuron ; 104(5): 834-848, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31805262

RESUMO

Neural stem cells in the adult mammalian brain are the source of new neurons that contribute to complex sensory and cognitive functions. Most adult neural stem cells are maintained in a state of reversible cell cycle arrest, also called quiescence. Quiescent neural stem cells present a low rate of metabolic activity and a high sensitivity to their local signaling environment, and they can be activated by diverse physiological stimuli. The balance between stem cell quiescence and activity determines not only the rate of neurogenesis but also the long-term maintenance of the stem cell pool and the neurogenic capacity of the aging brain. In recent years, significant progress has been made in characterizing quiescent stem cells thanks to the introduction of new genomic and imaging techniques. We discuss in this review our current understanding of neural stem cell quiescence and its regulation by intrinsic and systemic factors.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Humanos
10.
Elife ; 82019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31552825

RESUMO

Quiescence is essential for the long-term maintenance of adult stem cells but how stem cells maintain quiescence is poorly understood. Here, we show that neural stem cells (NSCs) in the adult mouse hippocampus actively transcribe the pro-activation factor Ascl1 regardless of their activated or quiescent states. We found that the inhibitor of DNA binding protein Id4 is enriched in quiescent NSCs and that elimination of Id4 results in abnormal accumulation of Ascl1 protein and premature stem cell activation. Accordingly, Id4 and other Id proteins promote elimination of Ascl1 protein in NSC cultures. Id4 sequesters Ascl1 heterodimerization partner E47, promoting Ascl1 protein degradation and stem cell quiescence. Our results highlight the importance of non-transcriptional mechanisms for the maintenance of NSC quiescence and reveal a role for Id4 as a quiescence-inducing factor, in contrast with its role of promoting the proliferation of embryonic neural progenitors.


Assuntos
Células-Tronco Adultas/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Hipocampo/citologia , Proteínas Inibidoras de Diferenciação/metabolismo , Células-Tronco Neurais/fisiologia , Animais , Células Cultivadas , Camundongos , Ligação Proteica , Fator 3 de Transcrição/metabolismo
12.
Neuron ; 93(2): 348-361, 2017 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-28041881

RESUMO

Mutations in NIPBL are the most frequent cause of Cornelia de Lange syndrome (CdLS), a developmental disorder encompassing several neurological defects, including intellectual disability and seizures. How NIPBL mutations affect brain development is not understood. Here we identify Nipbl as a functional interaction partner of the neural transcription factor Zfp609 in brain development. Depletion of Zfp609 or Nipbl from cortical neural progenitors in vivo is detrimental to neuronal migration. Zfp609 and Nipbl overlap at genomic binding sites independently of cohesin and regulate genes that control cortical neuron migration. We find that Zfp609 and Nipbl interact with the Integrator complex, which functions in RNA polymerase 2 pause release. Indeed, Zfp609 and Nipbl co-localize at gene promoters containing paused RNA polymerase 2, and Integrator similarly regulates neuronal migration. Our data provide a rationale and mechanistic insights for the role of Nipbl in the neurological defects associated with CdLS.


Assuntos
Movimento Celular/genética , Córtex Cerebral/crescimento & desenvolvimento , Síndrome de Cornélia de Lange/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Neurais/citologia , Neurônios/citologia , Transativadores/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ciclo Celular/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Coesinas
13.
Cell Rep ; 17(5): 1383-1398, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27783951

RESUMO

Throughout life, adult neural stem cells (NSCs) produce new neurons and glia that contribute to crucial brain functions. Quiescence is an essential protective feature of adult NSCs; however, the establishment and maintenance of this state remain poorly understood. We demonstrate that in the adult zebrafish pallium, the brain-enriched miR-9 is expressed exclusively in a subset of quiescent NSCs, highlighting a heterogeneity within these cells, and is necessary to maintain NSC quiescence. Strikingly, miR-9, along with Argonaute proteins (Agos), is localized to the nucleus of quiescent NSCs, and manipulating their nuclear/cytoplasmic ratio impacts quiescence. Mechanistically, miR-9 permits efficient Notch signaling to promote quiescence, and we identify the RISC protein TNRC6 as a mediator of miR-9/Agos nuclear localization in vivo. We propose a conserved non-canonical role for nuclear miR-9/Agos in controlling the balance between NSC quiescence and activation, a key step in maintaining adult germinal pools.


Assuntos
Ciclo Celular , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Peixe-Zebra/genética , Envelhecimento/metabolismo , Animais , Proteínas Argonautas/metabolismo , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Camundongos , MicroRNAs/genética , Modelos Biológicos , Neuroglia/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Telencéfalo/metabolismo , Peixe-Zebra/metabolismo
14.
Science ; 353(6296): 292-5, 2016 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-27418510

RESUMO

Quiescence is essential for long-term maintenance of adult stem cells. Niche signals regulate the transit of stem cells from dormant to activated states. Here, we show that the E3-ubiquitin ligase Huwe1 (HECT, UBA, and WWE domain-containing 1) is required for proliferating stem cells of the adult mouse hippocampus to return to quiescence. Huwe1 destabilizes proactivation protein Ascl1 (achaete-scute family bHLH transcription factor 1) in proliferating hippocampal stem cells, which prevents accumulation of cyclin Ds and promotes the return to a resting state. When stem cells fail to return to quiescence, the proliferative stem cell pool becomes depleted. Thus, long-term maintenance of hippocampal neurogenesis depends on the return of stem cells to a transient quiescent state through the rapid degradation of a key proactivation factor.


Assuntos
Células-Tronco Adultas/fisiologia , Hipocampo/embriologia , Células-Tronco Neurais/fisiologia , Neurogênese , Ubiquitina-Proteína Ligases/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Hipocampo/citologia , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Estabilidade Proteica , Proteólise , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases/genética
15.
Front Cell Neurosci ; 8: 396, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25505873

RESUMO

Neurogenesis persists in adult mammals in specific brain areas, known as neurogenic niches. Adult neurogenesis is highly dynamic and is modulated by multiple physiological stimuli and pathological states. There is a strong interest in understanding how this process is regulated, particularly since active neuronal production has been demonstrated in both the hippocampus and the subventricular zone (SVZ) of adult humans. The molecular mechanisms that control neurogenesis have been extensively studied during embryonic development. Therefore, we have a broad knowledge of the intrinsic factors and extracellular signaling pathways driving proliferation and differentiation of embryonic neural precursors. Many of these factors also play important roles during adult neurogenesis, but essential differences exist in the biological responses of neural precursors in the embryonic and adult contexts. Because adult neural stem cells (NSCs) are normally found in a quiescent state, regulatory pathways can affect adult neurogenesis in ways that have no clear counterpart during embryogenesis. BMP signaling, for instance, regulates NSC behavior both during embryonic and adult neurogenesis. However, this pathway maintains stem cell proliferation in the embryo, while it promotes quiescence to prevent stem cell exhaustion in the adult brain. In this review, we will compare and contrast the functions of transcription factors (TFs) and other regulatory molecules in the embryonic brain and in adult neurogenic regions of the adult brain in the mouse, with a special focus on the hippocampal niche and on the regulation of the balance between quiescence and activation of adult NSCs in this region.

16.
Neuron ; 83(5): 1085-97, 2014 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-25189209

RESUMO

The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.


Assuntos
Células-Tronco Adultas/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/citologia , Neurogênese/genética , Células-Tronco Adultas/metabolismo , Fatores Etários , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Ventrículos Cerebrais/citologia , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Ácido Caínico/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas com Domínio T/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
17.
Cell Rep ; 4(3): 477-91, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23891001

RESUMO

FOXO transcription factors are central regulators of longevity from worms to humans. FOXO3, the FOXO isoform associated with exceptional human longevity, preserves adult neural stem cell pools. Here, we identify FOXO3 direct targets genome-wide in primary cultures of adult neural progenitor cells (NPCs). Interestingly, FOXO3-bound sites are enriched for motifs for bHLH transcription factors, and FOXO3 shares common targets with the proneuronal bHLH transcription factor ASCL1/MASH1 in NPCs. Analysis of the chromatin landscape reveals that FOXO3 and ASCL1 are particularly enriched at the enhancers of genes involved in neurogenic pathways. Intriguingly, FOXO3 inhibits ASCL1-dependent neurogenesis in NPCs and direct neuronal conversion in fibroblasts. FOXO3 also restrains neurogenesis in vivo. Our study identifies a genome-wide interaction between the prolongevity transcription factor FOXO3 and the cell-fate determinant ASCL1 and raises the possibility that FOXO3's ability to restrain ASCL1-dependent neurogenesis may help preserve the neural stem cell pool.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Genoma , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo
18.
Stem Cells Dev ; 21(12): 2239-51, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22142223

RESUMO

Development of the nervous system is finely regulated by consecutive expression of cell-specific transcription factors. Here we show that Helios, a member of the Ikaros transcription factor family, is expressed in ectodermal and neuroectodermal-derived tissues. During embryonic development, Helios is expressed by several brain structures including the lateral ganglionic eminence (LGE, the striatal anlage); the cingulated, insular and retrosplenial cortex; the hippocampus; and the accessory olfactory bulb. Moreover, Helios is also expressed by Purkinje neurons during postnatal cerebellar development. Within the LGE, Helios expression follows a dynamic spatio-temporal pattern starting at embryonic stages (E14.5), peaking at E18.5, and completely disappearing during postnatal development. Helios is expressed by a small population of nestin-positive neural progenitor cells located in the subventricular zone as well as by a larger population of immature neurons distributed throughout the mantle zone. In the later, Helios is preferentially expressed in the matrix compartment, where it colocalizes with Bcl11b and Foxp1, well-known markers of striatal projection neurons. In addition, we observed that Helios expression is not detected in Dlx1/2 and Gsx2 null mutants, while its expression is maintained in Ascl1 mutants. These findings allow us to introduce a new transcription factor in the cascade of events that take part of striatal development postulating the existence of at least 4 different neural progenitors in the LGE. An Ascl1-independent but Gsx2- & Dlx1/2-dependent precursor will express Helios defining a new lineage for a subset of matrix striatal neurons.


Assuntos
Corpo Estriado/citologia , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Neurônios/metabolismo , Fatores de Transcrição/fisiologia , Animais , Cerebelo/metabolismo , Corpo Estriado/embriologia , Corpo Estriado/crescimento & desenvolvimento , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Nestina , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
19.
Neural Dev ; 5: 21, 2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20735826

RESUMO

BACKGROUND: Nolz1 is a zinc finger transcription factor whose expression is enriched in the lateral ganglionic eminence (LGE), although its function is still unknown. RESULTS: Here we analyze the role of Nolz1 during LGE development. We show that Nolz1 expression is high in proliferating neural progenitor cells (NPCs) of the LGE subventricular zone. In addition, low levels of Nolz1 are detected in the mantle zone, as well as in the adult striatum. Similarly, Nolz1 is highly expressed in proliferating LGE-derived NPC cultures, but its levels rapidly decrease upon cell differentiation, pointing to a role of Nolz1 in the control of NPC proliferation and/or differentiation. In agreement with this hypothesis, we find that Nolz1 over-expression promotes cell cycle exit of NPCs in neurosphere cultures and negatively regulates proliferation in telencephalic organotypic cultures. Within LGE primary cultures, Nolz1 over-expression promotes the acquisition of a neuronal phenotype, since it increases the number of ß-III tubulin (Tuj1)- and microtubule-associated protein (MAP)2-positive neurons, and inhibits astrocyte generation and/or differentiation. Retinoic acid (RA) is one of the most important morphogens involved in striatal neurogenesis, and regulates Nolz1 expression in different systems. Here we show that Nolz1 also responds to this morphogen in E12.5 LGE-derived cell cultures. However, Nolz1 expression is not regulated by RA in E14.5 LGE-derived cell cultures, nor is it affected during LGE development in mouse models that present decreased RA levels. Interestingly, we find that Gsx2, which is necessary for normal RA signaling during LGE development, is also required for Nolz1 expression, which is lost in Gsx2 knockout mice. These findings suggest that Nolz1 might act downstream of Gsx2 to regulate RA-induced neurogenesis. Keeping with this hypothesis, we show that Nolz1 induces the selective expression of the RA receptor (RAR)ß without altering RARα or RARγ. In addition, Nozl1 over-expression increases RA signaling since it stimulates the RA response element. This RA signaling is essential for Nolz1-induced neurogenesis, which is impaired in a RA-free environment or in the presence of a RAR inverse agonist. It has been proposed that Drosophila Gsx2 and Nolz1 homologues could cooperate with the transcriptional co-repressors Groucho-TLE to regulate cell proliferation. In agreement with this view, we show that Nolz1 could act in collaboration with TLE-4, as they are expressed at the same time in NPC cultures and during mouse development. CONCLUSIONS: Nolz1 promotes RA signaling in the LGE, contributing to the striatal neurogenesis during development.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte/metabolismo , Corpo Estriado/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Transdução de Sinais/fisiologia , Tretinoína/farmacologia , Animais , Proteínas de Transporte/genética , Contagem de Células , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Corpos Geniculados/embriologia , Proteínas de Homeodomínio/genética , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos CBA , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neurogênese/fisiologia , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/fisiologia , Proteínas Nucleares/genética , Gravidez , Retinal Desidrogenase/deficiência , Transdução de Sinais/genética , Tubulina (Proteína)/metabolismo
20.
J Comp Neurol ; 518(3): 329-51, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19950118

RESUMO

During central nervous system development, several transcription factors regulate the differentiation of progenitor cells to postmitotic neurons. Here we describe a novel role for Ikaros-1 in the generation of late-born striatal neurons. Our results show that Ikaros-1 is expressed in the boundary of the striatal germinal zone (GZ)/mantle zone (MZ), where it induces cell cycle arrest of neural progenitors by up-regulation of the cyclin-dependent kinase inhibitor (CDKi) p21(Cip1/Waf1). This effect is coupled with the neuronal differentiation of late precursors, which in turn is critical for the second wave of striatal neurogenesis that gives rise to matrix neurons. Consistently, Ikaros(-/-) mice had fewer striatal projecting neurons and, in particular, enkephalin (ENK)-positive neurons. In addition, overexpression of Ikaros-1 in primary striatal cultures increases the number of calbindin- and ENK-positive neurons. Our results also show that Ikaros-1 acts downstream of the Dlx family of transcription factors, insofar as its expression is lost in Dlx1/2 double knockout mice. However, we demonstrate that Ikaros-1 and Ebf-1 independently regulate the final determination of the two populations of striatal projection neurons of the matrix compartment, ENK- and substance P-positive neurons. In conclusion, our findings identify Ikaros-1 as a modulator of cell cycle exit of neural progenitors that gives rise to the neurogenesis of ENK-positive striatal neurons.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Corpo Estriado/embriologia , Encefalinas/metabolismo , Fator de Transcrição Ikaros/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Animais , Calbindinas , Proteínas de Ciclo Celular/genética , Diferenciação Celular/fisiologia , Corpo Estriado/citologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Vias Eferentes/citologia , Vias Eferentes/embriologia , Genes cdc/fisiologia , Proteínas de Homeodomínio/genética , Fator de Transcrição Ikaros/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Proteína G de Ligação ao Cálcio S100/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Substância P/metabolismo , Transativadores/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA