Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuron ; 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38582081

RESUMO

Integration of new neurons into adult hippocampal circuits is a process coordinated by local and long-range synaptic inputs. To achieve stable integration and uniquely contribute to hippocampal function, immature neurons are endowed with a critical period of heightened synaptic plasticity, yet it remains unclear which mechanisms sustain this form of plasticity during neuronal maturation. We found that as new neurons enter their critical period, a transient surge in fusion dynamics stabilizes elongated mitochondrial morphologies in dendrites to fuel synaptic plasticity. Conditional ablation of fusion dynamics to prevent mitochondrial elongation selectively impaired spine plasticity and synaptic potentiation, disrupting neuronal competition for stable circuit integration, ultimately leading to decreased survival. Despite profuse mitochondrial fragmentation, manipulation of competition dynamics was sufficient to restore neuronal survival but left neurons poorly responsive to experience at the circuit level. Thus, by enabling synaptic plasticity during the critical period, mitochondrial fusion facilitates circuit remodeling by adult-born neurons.

2.
bioRxiv ; 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38260428

RESUMO

The adult hippocampus generates new granule cells (aGCs) that exhibit distinct functional capabilities along development, conveying a unique form of plasticity to the preexisting circuits. While early differentiation of adult radial glia-like neural stem cells (RGL) has been studied extensively, the molecular mechanisms guiding the maturation of postmitotic neurons remain unknown. Here, we used a precise birthdating strategy to follow newborn aGCs along differentiation using single-nuclei RNA sequencing (snRNA-seq). Transcriptional profiling revealed a continuous trajectory from RGLs to mature aGCs, with multiple sequential immature stages bearing increasing levels of effector genes supporting growth, excitability and synaptogenesis. Remarkably, four discrete cellular states were defined by the expression of distinct sets of transcription factors (TFs): quiescent neural stem cells, proliferative progenitors, postmitotic immature aGCs, and mature aGCs. The transition from immature to mature aCGs involved a transcriptional switch that shutdown molecular cascades promoting cell growth, such as the SoxC family of TFs, to activate programs controlling neuronal homeostasis. Indeed, aGCs overexpressing Sox4 or Sox11 remained stalled at the immature state. Our results unveil precise molecular mechanisms driving adult neural stem cells through the pathway of neuronal differentiation.

3.
Dis Model Mech ; 16(6)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37260288

RESUMO

The nuclear receptor NR2F1 acts as a strong transcriptional regulator in embryonic and postnatal neural cells. In humans, mutations in the NR2F1 gene cause Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS), a rare neurodevelopmental disorder characterized by multiple clinical features including vision impairment, intellectual disability and autistic traits. In this study, we identified, by genome-wide and in silico analyses, a set of nuclear-encoded mitochondrial genes as potential genomic targets under direct NR2F1 transcriptional control in neurons. By combining mouse genetic, neuroanatomical and imaging approaches, we demonstrated that conditional NR2F1 loss of function within the adult mouse hippocampal neurogenic niche results in a reduced mitochondrial mass associated with mitochondrial fragmentation and downregulation of key mitochondrial proteins in newborn neurons, the genesis, survival and functional integration of which are impaired. Importantly, we also found dysregulation of several nuclear-encoded mitochondrial genes and downregulation of key mitochondrial proteins in the brain of Nr2f1-heterozygous mice, a validated BBSOAS model. Our data point to an active role for NR2F1 in the mitochondrial gene expression regulatory network in neurons and support the involvement of mitochondrial dysfunction in BBSOAS pathogenesis.


Assuntos
Fator I de Transcrição COUP , Anormalidades do Olho , Deficiência Intelectual , Atrofia Óptica , Animais , Humanos , Camundongos , Encéfalo/metabolismo , Fator I de Transcrição COUP/genética , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Deficiência Intelectual/genética , Mitocôndrias , Mutação/genética , Atrofia Óptica/genética , Atrofia Óptica/metabolismo
4.
Cells ; 10(9)2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34571902

RESUMO

Inflammatory bowel diseases (IBD) are characterized by chronic dysregulation of immune homeostasis, epithelial demise, immune cell activation, and microbial translocation. Each of these processes leads to proinflammatory changes via the release of cytokines, damage-associated molecular patterns (DAMPs), and pathogen-associated molecular patterns (PAMPs), respectively. The impact of these noxious agents on the survival and function of the enteric nervous system (ENS) is poorly understood. Here, we show that in contrast to an expected decrease, experimental as well as clinical colitis causes an increase in the transcript levels of enteric neuronal and glial genes. Immunostaining revealed an elevated neuronal innervation of the inflamed regions of the gut mucosa. The increase was seen in models with overt damage to epithelial cells and models of T cell-induced colitis. Transcriptomic data from treatment naïve pediatric IBD patients also confirmed the increase in the neuroglial genes and were replicated on an independent adult IBD dataset. This induction in the neuroglial genes was transient as levels returned to normal upon the induction of remission in both mouse models as well as colitis patients. Our data highlight the dynamic and robust nature of the enteric nervous system in colitis and open novel questions on its regulation.


Assuntos
Colite/patologia , Sistema Nervoso Entérico/patologia , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/inervação , Neurônios/patologia , Transcriptoma , Animais , Colite/etiologia , Colite/metabolismo , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/metabolismo , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Camundongos , Neurônios/imunologia , Neurônios/metabolismo
5.
Development ; 148(14)2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34184026

RESUMO

Transcription factor 4 (TCF4) is a crucial regulator of neurodevelopment and has been linked to the pathogenesis of autism, intellectual disability and schizophrenia. As a class I bHLH transcription factor (TF), it is assumed that TCF4 exerts its neurodevelopmental functions through dimerization with proneural class II bHLH TFs. Here, we aim to identify TF partners of TCF4 in the control of interhemispheric connectivity formation. Using a new bioinformatic strategy integrating TF expression levels and regulon activities from single cell RNA-sequencing data, we find evidence that TCF4 interacts with non-bHLH TFs and modulates their transcriptional activity in Satb2+ intercortical projection neurons. Notably, this network comprises regulators linked to the pathogenesis of neurodevelopmental disorders, e.g. FOXG1, SOX11 and BRG1. In support of the functional interaction of TCF4 with non-bHLH TFs, we find that TCF4 and SOX11 biochemically interact and cooperatively control commissure formation in vivo, and regulate the transcription of genes implicated in this process. In addition to identifying new candidate interactors of TCF4 in neurodevelopment, this study illustrates how scRNA-Seq data can be leveraged to predict TF networks in neurodevelopmental processes.


Assuntos
RNA Citoplasmático Pequeno/metabolismo , Análise de Célula Única , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , DNA Helicases , Embrião de Mamíferos , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Deficiência Intelectual , Proteínas de Ligação à Região de Interação com a Matriz , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso , Neurônios/fisiologia , Proteínas Nucleares , Domínios e Motivos de Interação entre Proteínas , RNA Citoplasmático Pequeno/genética , Fatores de Transcrição SOXC , Esquizofrenia/genética , Esquizofrenia/metabolismo
6.
EMBO J ; 39(21): e104472, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-32929771

RESUMO

In adult hippocampal neurogenesis, stem/progenitor cells generate dentate granule neurons that contribute to hippocampal plasticity. The establishment of a morphologically defined dendritic arbor is central to the functional integration of adult-born neurons. We investigated the role of canonical Wnt/ß-catenin signaling in dendritogenesis of adult-born neurons. We show that canonical Wnt signaling follows a biphasic pattern, with high activity in stem/progenitor cells, attenuation in immature neurons, and reactivation during maturation, and demonstrate that this activity pattern is required for proper dendrite development. Increasing ß-catenin signaling in maturing neurons of young adult mice transiently accelerated dendritic growth, but eventually produced dendritic defects and excessive spine numbers. In middle-aged mice, in which protracted dendrite and spine development were paralleled by lower canonical Wnt signaling activity, enhancement of ß-catenin signaling restored dendritic growth and spine formation to levels observed in young adult animals. Our data indicate that precise timing and strength of ß-catenin signaling are essential for the correct functional integration of adult-born neurons and suggest Wnt/ß-catenin signaling as a pathway to ameliorate deficits in adult neurogenesis during aging.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , beta Catenina/metabolismo , Envelhecimento/metabolismo , Animais , Proteína Axina/genética , Feminino , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Via de Sinalização Wnt , beta Catenina/genética
7.
Stem Cell Res ; 47: 101889, 2020 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-32682288

RESUMO

ARID1B haploinsufficiency induced by missense or nonsense mutations of ARID1B is a cause of Coffin-Siris syndrome (CSS), a neurodevelopmental disorder associated with intellectual disability. At present, no appropriate human stem cell model for ARID1B-associated CSS has been reported. Here, we describe the generation and validation of ARID1B+/- hESCs by introducing out of frame deletions into exon 5 or 6 of ARID1B with CRISPR/Cas9 genome editing. These ARID1B+/- hESC lines allow to study the pathophysiology of ARID1B-associated CSS in 2D and 3D models of human neurodevelopment.

8.
Hum Mol Genet ; 28(15): 2589-2599, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31035284

RESUMO

The SOXC transcription factors Sox4, Sox11 and Sox12, are critical neurodevelopmental regulators that are thought to function in a highly redundant fashion. Surprisingly, heterozygous missense mutations or deletions of SOX11 were recently detected in patients with Coffin-Siris syndrome-like syndrome (CSSLS), a neurodevelopmental disorder associated with intellectual disability, demonstrating that in humans SOX11 haploinsufficiency cannot be compensated and raising the question of the function of SOX11 in human neurodevelopment. Here, we describe the generation of SOX11+/- heterozygous human embryonic stem cell (hESC) lines by CRISPR/Cas9 genome engineering. SOX11 haploinsufficiency impaired the generation of neurons and resulted in a proliferation/differentiation imbalance of neural precursor cells and enhanced neuronal cell death. Using the SOX11+/- hESC model we provide for the first time experimental evidence that SOX11 haploinsufficiency is sufficient to impair key processes of human neurodevelopment, giving a first insight into the pathophysiology of CSSLS and SOX11 function in human neurodevelopment.


Assuntos
Linhagem Celular , Dosagem de Genes , Células-Tronco Embrionárias Humanas/fisiologia , Modelos Biológicos , Transtornos do Neurodesenvolvimento/metabolismo , Fatores de Transcrição SOXC/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Sistemas CRISPR-Cas , Diferenciação Celular , Proliferação de Células , Face/anormalidades , Edição de Genes , Regulação da Expressão Gênica , Deformidades Congênitas da Mão/genética , Deformidades Congênitas da Mão/metabolismo , Haploinsuficiência , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Micrognatismo/genética , Micrognatismo/metabolismo , Pescoço/anormalidades , Células-Tronco Neurais , Transtornos do Neurodesenvolvimento/genética
9.
Front Neurosci ; 12: 914, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30574063

RESUMO

Mutations in SPG11 cause a complicated autosomal recessive form of hereditary spastic paraplegia (HSP). Mechanistically, there are indications for the dysregulation of the GSK3ß/ßCat signaling pathway in SPG11. In this study, we tested the therapeutic potential of the GSK3ß inhibitor, tideglusib, to rescue neurodegeneration associated characteristics in an induced pluripotent stem cells (iPSCs) derived neuronal model from SPG11 patients and matched healthy controls as well as a CRISPR-Cas9 mediated SPG11 knock-out line and respective control. SPG11-iPSC derived cortical neurons, as well as the genome edited neurons exhibited shorter and less complex neurites than controls. Administration of tideglusib to these lines led to the rescue of neuritic impairments. Moreover, the treatment restored increased cell death and ameliorated the membranous inclusions in iPSC derived SPG11 neurons. Our results provide a first evidence for the rescue of neurite pathology in SPG11-HSP by tideglusib. The current lack of disease-modifying treatments for SPG11 and related types of complicated HSP renders tideglusib a candidate compound for future clinical application.

10.
Sci Rep ; 8(1): 16196, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30385877

RESUMO

The intellectual disability gene, Sox11, encodes for a critical neurodevelopmental transcription factor with functions in precursor survival, neuronal fate determination, migration and morphogenesis. The mechanisms regulating SOX11's activity remain largely unknown. Mass spectrometric analysis uncovered that SOX11 can be post-translationally modified by phosphorylation. Here, we report that phosphorylatable serines surrounding the high-mobility group box modulate SOX11's transcriptional activity. Through Mass Spectrometry (MS), co-immunoprecipitation assays and in vitro phosphorylation assays followed by MS we verified that protein kinase A (PKA) interacts with SOX11 and phosphorylates it on S133. In vivo replacement of SoxC factors in developing adult-generated hippocampal neurons with SOX11 S133 phospho-mutants indicated that phosphorylation on S133 modulates dendrite development of adult-born dentate granule neurons, while reporter assays suggested that S133 phosphorylation fine-tunes the activation of select target genes. These data provide novel insight into the control of the critical neurodevelopmental regulator SOX11 and imply SOX11 as a mediator of PKA-regulated neuronal development.


Assuntos
Morfogênese/genética , Neurogênese/genética , Neurônios/metabolismo , Fatores de Transcrição SOXC/genética , Animais , Núcleos Cerebelares/crescimento & desenvolvimento , Núcleos Cerebelares/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/genética , Dendritos/genética , Dendritos/metabolismo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Espectrometria de Massas , Camundongos , Fosforilação/genética , Serina/genética
11.
Front Mol Neurosci ; 11: 211, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29973868

RESUMO

SOX11 is a key Transcription Factor (TF) in the regulation of embryonic and adult neurogenesis, whose mutation has recently been linked to an intellectual disability syndrome in humans. SOX11's transient activity during neurogenesis is critical to ensure the precise execution of the neurogenic program. Here, we report that SOX11 displays differential subcellular localizations during the course of neurogenesis. Western-Blot analysis of embryonic mouse brain lysates indicated that SOX11 is post-translationally modified by phosphorylation. Using Mass Spectrometry, we found 10 serine residues in the SOX11 protein that are putatively phosphorylated. Systematic analysis of phospho-mutant SOX11 resulted in the identification of the S30 residue, whose phosphorylation promotes nuclear over cytoplasmic localization of SOX11. Collectively, these findings uncover phosphorylation as a novel layer of regulation of the intellectual disability gene Sox11.

12.
Stem Cell Reports ; 10(2): 347-355, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29337116

RESUMO

Swiprosin-1/Efhd2 (Efhd2) is highly expressed in the CNS during development and in the adult. EFHD2 is regulated by Ca2+ binding, stabilizes F-actin, and promotes neurite extension. Previous studies indicated a dysregulation of EFHD2 in human Alzheimer's disease brains. We hypothesized a detrimental effect of genetic ablation of Efhd2 on hippocampal integrity and specifically investigated adult hippocampal neurogenesis. Efhd2 was expressed throughout adult neuronal development and in mature neurons. We observed a severe reduction of the survival of adult newborn neurons in Efhd2 knockouts, starting at the early neuroblast stage. Spine formation and dendrite growth of newborn neurons were compromised in full Efhd2 knockouts, but not upon cell-autonomous Efhd2 deletion. Together with our finding of severe hippocampal tauopathy in Efhd2 knockout mice, these data connect Efhd2 to impaired synaptic plasticity as present in Alzheimer's disease and identify a role of Efhd2 in neuronal survival and synaptic integration in the adult hippocampus.


Assuntos
Doença de Alzheimer/genética , Proteínas de Ligação ao Cálcio/genética , Hipocampo/crescimento & desenvolvimento , Neurogênese/genética , Coluna Vertebral/crescimento & desenvolvimento , Actinas/genética , Doença de Alzheimer/patologia , Animais , Cálcio/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Knockout , Neuritos/metabolismo , Plasticidade Neuronal/genética , Neurônios/citologia , Neurônios/metabolismo , Coluna Vertebral/metabolismo
13.
Autophagy ; 14(1): 98-119, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29198173

RESUMO

The autophagy-lysosome pathway (ALP) regulates intracellular homeostasis of the cytosolic protein SNCA/alpha-synuclein and is impaired in synucleinopathies, including Parkinson disease and dementia with Lewy bodies (DLB). Emerging evidence suggests that ALP influences SNCA release, but the underlying cellular mechanisms are not well understood. Several studies identified SNCA in exosome/extracellular vesicle (EV) fractions. EVs are generated in the multivesicular body compartment and either released upon its fusion with the plasma membrane, or cleared via the ALP. We therefore hypothesized that inhibiting ALP clearance 1) enhances SNCA release via EVs by increasing extracellular shuttling of multivesicular body contents, 2) alters EV biochemical profile, and 3) promotes SNCA cell-to-cell transfer. Indeed, ALP inhibition increased the ratio of extra- to intracellular SNCA and upregulated SNCA association with EVs in neuronal cells. Ultrastructural analysis revealed a widespread, fused multivesicular body-autophagosome compartment. Biochemical characterization revealed the presence of autophagosome-related proteins, such as LC3-II and SQSTM1. This distinct "autophagosome-exosome-like" profile was also identified in human cerebrospinal fluid (CSF) EVs. After a single intracortical injection of SNCA-containing EVs derived from CSF into mice, human SNCA colocalized with endosome and neuronal markers. Prominent SNCA immunoreactivity and a higher number of neuronal SNCA inclusions were observed after DLB patient CSF EV injections. In summary, this study provides compelling evidence that a) ALP inhibition increases SNCA in neuronal EVs, b) distinct ALP components are present in EVs, and c) CSF EVs transfer SNCA from cell to cell in vivo. Thus, macroautophagy/autophagy may regulate EV protein composition and consequently progression in synucleinopathies.


Assuntos
Autofagossomos/metabolismo , Autofagia/fisiologia , Exossomos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , alfa-Sinucleína/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Biomarcadores/metabolismo , Células Cultivadas , Cloroquina/farmacologia , Exossomos/efeitos dos fármacos , Exossomos/ultraestrutura , Humanos , Doença por Corpos de Lewy/metabolismo , Lisossomos/efeitos dos fármacos , Macrolídeos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Doença de Parkinson/metabolismo , Transporte Proteico , Ratos Sprague-Dawley
14.
Stem Cells ; 35(3): 787-799, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27790794

RESUMO

We asked whether cell-cycle associated protein p27kip1 might be involved in the transition of precursor cells to postmitotic maturation in adult hippocampal neurogenesis. p27kip1 was expressed throughout the dentate gyrus with a strong nuclear expression in early postmitotic, calretinin-positive neurons and neuronally determined progenitor cells (type-3 and some type-2b), lower or absent expression in radial glia-like precursor cells (type-1) and type-2a cells and essentially no expression in granule cells. This suggested a transitory role in late proliferative and early postmitotic phases of neurogenesis. Inconsistent with a role limited to cell cycle arrest the acute stimuli, voluntary wheel running (RUN), environmental enrichment (ENR) and kainate-induced seizures increased p27kip1 expressing cells. Sequential short-term combination of RUN and ENR yielded more p27kip1 cells than either stimulus alone, indicating an additive effect. In vitro, p27kip1 was lowly expressed by proliferating precursor cells but increased upon differentiation. In p27kip1-/- mice neurogenesis was reduced in vivo, whereas the number of proliferating cells was increased. Accordingly, the microdissected dentate gyrus of p27kip1-/- mice generated more colonies in the neurosphere assay and an increased number of larger spheres with the differentiation potential unchanged. In p27kip1-/- monolayer cultures, proliferation was increased and cell cycle genes were upregulated. In the Morris water maze p27kip1-/- mice learned the task but were specifically impaired in the reversal phase explainable by the decrease in adult neurogenesis. We conclude that p27kip1 is involved in the decisive step around cell-cycle exit and plays an important role in activity-regulated and functionally relevant adult hippocampal neurogenesis. Stem Cells 2017;35:787-799.


Assuntos
Envelhecimento/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Hipocampo/metabolismo , Neurogênese , Animais , Comportamento Animal , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Feminino , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose , Neurônios/citologia , Neurônios/metabolismo , Fenótipo , Aprendizagem Espacial
15.
Stem Cells ; 34(4): 997-1010, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26729510

RESUMO

Adult neurogenesis is tightly regulated by the neurogenic niche. Cellular contacts between niche cells and neural stem cells are hypothesized to regulate stem cell proliferation or lineage choice. However, the structure of adult neural stem cells and the contact they form with niche cells are poorly described. Here, we characterized the morphology of radial glia-like (RGL) cells, their molecular identity, proliferative activity, and fate determination in the adult mouse hippocampus. We found the coexistence of two morphotypes of cells with prototypical morphological characteristics of RGL stem cells: Type α cells, which represented 76% of all RGL cells, displayed a long primary process modestly branching into the molecular layer and type ß cells, which represented 24% of all RGL cells, with a shorter radial process highly branching into the outer granule cell layer-inner molecular layer border. Stem cell markers were expressed in type α cells and coexpressed with astrocytic markers in type ß cells. Consistently, in vivo lineage tracing indicated that type α cells can give rise to neurons, astrocytes, and type ß cells, whereas type ß cells do not proliferate. Our results reveal that the adult subgranular zone of the dentate gyrus harbors two functionally different RGL cells, which can be distinguished by simple morphological criteria, supporting a morphofunctional role of their thin cellular processes. Type ß cells may represent an intermediate state in the transformation of type α, RGL stem cells, into astrocytes.


Assuntos
Células Ependimogliais/citologia , Hipocampo/citologia , Células-Tronco Neurais/citologia , Neurogênese , Animais , Biomarcadores/metabolismo , Linhagem da Célula/genética , Proliferação de Células , Células Ependimogliais/metabolismo , Células Ependimogliais/transplante , Hipocampo/patologia , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante
16.
Cold Spring Harb Perspect Biol ; 7(10): a018879, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26430216

RESUMO

Adult-generated dentate granule neurons have emerged as major contributors to hippocampal plasticity. New neurons are generated from neural stem cells through a complex sequence of proliferation, differentiation, and maturation steps. Development of the new neuron is dependent on the precise temporal activity of transcription factors, which coordinate the expression of stage-specific genetic programs. Here, we review current knowledge in transcription factor-mediated regulation of mammalian neural stem cells and neurogenesis and will discuss potential mechanisms of how transcription factor networks, on one hand, allow for precise execution of the developmental sequence and, on the other hand, allow for adaptation of the rate and timing of adult neurogenesis in response to complex stimuli. Understanding transcription factor-mediated control of neuronal development will provide new insights into the mechanisms underlying neurogenesis-dependent plasticity in health and disease.


Assuntos
Hipocampo/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Fatores de Transcrição/metabolismo , Animais , Encéfalo/embriologia , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células , Hipocampo/embriologia , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Células-Tronco/citologia , Sinapses/fisiologia
17.
Stem Cells ; 33(1): 219-29, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25185890

RESUMO

Members of the cyclin-dependent kinase (CDK)-inhibitory protein (CIP)/kinase-inhibitory protein (KIP) family of cyclin-dependent kinase inhibitors regulate proliferation and cell cycle exit of mammalian cells. In the adult brain, the CIP/KIP protein p27(kip1) has been related to the regulation of intermediate progenitor cells located in neurogenic niches. Here, we uncover a novel function of p27(kip1) in the adult hippocampus as a dual regulator of stem cell quiescence and of cell-cycle exit of immature neurons. In vivo, p27(kip1) is detected in radial stem cells expressing SOX2 and in newborn neurons of the dentate gyrus. In vitro, the Cdkn1b gene encoding p27(kip1) is transcriptionally upregulated by quiescence signals such as BMP4. The nuclear accumulation of p27(kip1) protein in adult hippocampal stem cells encompasses the BMP4-induced quiescent state and its overexpression is able to block proliferation. p27(kip1) is also expressed in immature neurons upon differentiation of adult hippocampal stem cell cultures. Loss of p27(kip1) leads to an increase in proliferation and neurogenesis in the adult dentate gyrus, which results from both a decrease in the percentage of radial stem cells that are quiescent and a delay in cell cycle exit of immature neurons. Analysis of animals carrying a disruption in the cyclin-CDK interaction domain of p27(kip1) indicates that the CDK inhibitory function of the protein is necessary to control the activity of radial stem cells. Thus, we report that p27(kip1) acts as a central player of the molecular program that keeps adult hippocampal stem cells out of the cell cycle.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Hipocampo/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/genética , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Knockout , Células-Tronco Neurais/metabolismo
18.
Stem Cell Reports ; 2(2): 153-62, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24527389

RESUMO

We describe the labeling of adult neural stem cells (aNSCs) in the mouse and human dentate gyrus (DG) by the combinatorial expression of glial fibrillary acidic protein (GFAP) and Prominin1, as revealed by immunohistochemistry. Split-Cre-based genetic fate mapping of these double-positive cells in the adult murine DG reveals their NSC identity, as they are self-renewing and contribute to neurogenesis over several months. Their progeny reacts to stimuli such as voluntary exercise with increased neurogenesis. Prominin1+/GFAP+ cells also exist in the adult human DG, the only region in the human brain for which adult neurogenesis has been consistently reported. Our data, together with previous evidence of such double-positive NSCs in the developing murine brain and in neurogenic regions of vertebrates with widespread neurogenesis, suggest that Prominin1- and GFAP-expressing cells are NSCs in a wide range of species in development and adulthood.


Assuntos
Células-Tronco Adultas/metabolismo , Giro Denteado/citologia , Giro Denteado/metabolismo , Marcação de Genes , Recombinação Homóloga , Células-Tronco Neurais/metabolismo , Antígeno AC133 , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Expressão Gênica , Genes Reporter , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Hipocampo/metabolismo , Humanos , Camundongos , Neuroglia/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Transporte Proteico
19.
Neuron ; 79(6): 1123-35, 2013 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-24050402

RESUMO

Neurons of the cerebral cortex are organized in layers and columns. Unlike laminar patterning, the mechanisms underlying columnar organization remain largely unexplored. Here, we show that ephrin-B1 plays a key role in this process through the control of nonradial steps of migration of pyramidal neurons. In vivo gain of function of ephrin-B1 resulted in a reduction of tangential motility of pyramidal neurons, leading to abnormal neuronal clustering. Conversely, following genetic disruption of ephrin-B1, cortical neurons displayed a wider lateral dispersion, resulting in enlarged ontogenic columns. Dynamic analyses revealed that ephrin-B1 controls the lateral spread of pyramidal neurons by limiting neurite extension and tangential migration during the multipolar phase. Furthermore, we identified P-Rex1, a guanine-exchange factor for Rac3, as a downstream ephrin-B1 effector required to control migration during the multipolar phase. Our results demonstrate that ephrin-B1 inhibits nonradial migration of pyramidal neurons, thereby controlling the pattern of cortical columns.


Assuntos
Movimento Celular/genética , Córtex Cerebral/citologia , Efrina-B1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Piramidais/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Proteínas de Transporte/metabolismo , Adesão Celular/genética , Proteínas de Ciclo Celular/metabolismo , Eletroporação , Embrião de Mamíferos , Efrina-B1/deficiência , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Imunoprecipitação , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso , Proteínas Nucleares/metabolismo , Gravidez , Proteínas Repressoras/metabolismo
20.
Stroke ; 43(9): 2468-75, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22738919

RESUMO

BACKGROUND AND PURPOSE: Adult neurogenesis in the dentate gyrus is a unique form of brain plasticity that is strongly stimulated after stroke. We investigate the morphological properties of new granule cells, which are born and develop after the ischemic insult, and query whether these adult-born neurons properly integrate into the pre-existing hippocampal circuitries. METHODS: Two well-established models were used to induce either small cortical infarcts (photothrombosis model) or large territorial infarcts (transient middle cerebral artery occlusion model). New granule cells were labeled 4 days after the initial insult by intrahippocampal injection of a retroviral vector encoding green fluorescent protein and newborn neurons were morphologically analyzed using a semiautomatic Neurolucida system and confocal laser scanning microscopy at 6 weeks. RESULTS: Approximately 5% to 10% of newborn granule cells displayed significant morphological abnormalities comprising additional basal dendrites and, after middle cerebral artery occlusion, also ectopic cell position. The extent of morphological abnormalities was higher after large territorial infarcts and seems to depend on the severity of ischemic damage. An increased portion of mushroom spines in aberrant neurons suggests stable synaptic integration. However, poststroke generated granule cells with regular appearance also demonstrate alterations in dendritic complexity and spine morphology. CONCLUSIONS: The remarkable stimulation of dentate neurogenesis after stroke coincides with an increased rate of aberrantly integrated neurons, which may contribute to functional impairments and, hypothetically, favor pathogenesis of adjustment disorders, cognitive deficits, or epilepsy often seen in stroke patients.


Assuntos
Neurogênese/fisiologia , Retroviridae/genética , Acidente Vascular Cerebral/patologia , Animais , Isquemia Encefálica/patologia , Grânulos Citoplasmáticos/patologia , Grânulos Citoplasmáticos/ultraestrutura , Dendritos/patologia , Dendritos/ultraestrutura , Espinhas Dendríticas/patologia , Espinhas Dendríticas/ultraestrutura , Giro Denteado/patologia , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/patologia , Trombose Intracraniana/patologia , Proteínas Luminescentes/genética , Masculino , Plasticidade Neuronal/fisiologia , Neurônios/patologia , Neurônios/ultraestrutura , Ratos , Ratos Wistar , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA