Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell Stem Cell ; 31(1): 39-51.e6, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38181749

RESUMO

Research on human cerebellar development and disease has been hampered by the need for a human cell-based system that recapitulates the human cerebellum's cellular diversity and functional features. Here, we report a human organoid model (human cerebellar organoids [hCerOs]) capable of developing the complex cellular diversity of the fetal cerebellum, including a human-specific rhombic lip progenitor population that have never been generated in vitro prior to this study. 2-month-old hCerOs form distinct cytoarchitectural features, including laminar organized layering, and create functional connections between inhibitory and excitatory neurons that display coordinated network activity. Long-term culture of hCerOs allows healthy survival and maturation of Purkinje cells that display molecular and electrophysiological hallmarks of their in vivo counterparts, addressing a long-standing challenge in the field. This study therefore provides a physiologically relevant, all-human model system to elucidate the cell-type-specific mechanisms governing cerebellar development and disease.


Assuntos
Cerebelo , Células de Purkinje , Humanos , Lactente , Metencéfalo , Organoides
2.
Nat Neurosci ; 26(12): 2090-2103, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37946050

RESUMO

Genes involved in synaptic function are enriched among those with autism spectrum disorder (ASD)-associated rare genetic variants. Dysregulated cortical neurogenesis has been implicated as a convergent mechanism in ASD pathophysiology, yet it remains unknown how 'synaptic' ASD risk genes contribute to these phenotypes, which arise before synaptogenesis. Here, we show that the synaptic Ras GTPase-activating (RASGAP) protein 1 (SYNGAP1, a top ASD risk gene) is expressed within the apical domain of human radial glia cells (hRGCs). In a human cortical organoid model of SYNGAP1 haploinsufficiency, we find dysregulated cytoskeletal dynamics that impair the scaffolding and division plane of hRGCs, resulting in disrupted lamination and accelerated maturation of cortical projection neurons. Additionally, we confirmed an imbalance in the ratio of progenitors to neurons in a mouse model of Syngap1 haploinsufficiency. Thus, SYNGAP1-related brain disorders may arise through non-synaptic mechanisms, highlighting the need to study genes associated with neurodevelopmental disorders (NDDs) in diverse human cell types and developmental stages.


Assuntos
Transtorno do Espectro Autista , Transtornos do Neurodesenvolvimento , Animais , Camundongos , Humanos , Transtorno do Espectro Autista/genética , Proteínas Ativadoras de ras GTPase/genética , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Neurogênese/genética
3.
Biol Psychiatry ; 93(7): 606-615, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36759258

RESUMO

Brain organoids derived from human pluripotent stem cells are emerging as a powerful tool to model cellular aspects of neuropsychiatric disorders, including alterations in cell proliferation, differentiation, migration, and lineage trajectory. To date, most contributions in the field have focused on modeling cellular impairment of the cerebral cortex, with few studies probing dysfunction in local network connectivity. However, it is increasingly more apparent that these psychiatric disorders are connectopathies involving multiple brain structures and the connections between them. Therefore, the lack of reproducible anatomical features in these 3-dimensional cultures represents a major bottleneck for effectively modeling brain connectivity at the micro(cellular) level and at the macroscale level between brain regions. In this perspective, we review the use of current organoid protocols to model neuropsychiatric disorders with a specific emphasis on the potential and limitations of the current strategies to model impairments in functional connectivity. Finally, we discuss the importance of adopting interdisciplinary strategies to establish next-generation, multiregional organoids that can model, with higher fidelity, the dysfunction in the development and functionality of long-range connections within the brain of patients affected by psychiatric disorders.


Assuntos
Células-Tronco Pluripotentes Induzidas , Transtornos Mentais , Humanos , Encéfalo , Organoides , Diferenciação Celular
4.
Ann N Y Acad Sci ; 1518(1): 196-208, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36177906

RESUMO

Complex three-dimensional in vitro organ-like models, or organoids, offer a unique biological tool with distinct advantages over two-dimensional cell culture systems, which can be too simplistic, and animal models, which can be too complex and may fail to recapitulate human physiology and pathology. Significant progress has been made in driving stem cells to differentiate into different organoid types, though several challenges remain. For example, many organoid models suffer from high heterogeneity, and it can be difficult to fully incorporate the complexity of in vivo tissue and organ development to faithfully reproduce human biology. Successfully addressing such limitations would increase the viability of organoids as models for drug development and preclinical testing. On April 3-6, 2022, experts in organoid development and biology convened at the Keystone Symposium "Organoids as Tools for Fundamental Discovery and Translation" to discuss recent advances and insights from this relatively new model system into human development and disease.


Assuntos
Modelos Biológicos , Organoides , Animais , Humanos , Organoides/metabolismo , Células-Tronco , Modelos Animais
6.
Nature ; 602(7896): 268-273, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35110736

RESUMO

Genetic risk for autism spectrum disorder (ASD) is associated with hundreds of genes spanning a wide range of biological functions1-6. The alterations in the human brain resulting from mutations in these genes remain unclear. Furthermore, their phenotypic manifestation varies across individuals7,8. Here we used organoid models of the human cerebral cortex to identify cell-type-specific developmental abnormalities that result from haploinsufficiency in three ASD risk genes-SUV420H1 (also known as KMT5B), ARID1B and CHD8-in multiple cell lines from different donors, using single-cell RNA-sequencing (scRNA-seq) analysis of more than 745,000 cells and proteomic analysis of individual organoids, to identify phenotypic convergence. Each of the three mutations confers asynchronous development of two main cortical neuronal lineages-γ-aminobutyric-acid-releasing (GABAergic) neurons and deep-layer excitatory projection neurons-but acts through largely distinct molecular pathways. Although these phenotypes are consistent across cell lines, their expressivity is influenced by the individual genomic context, in a manner that is dependent on both the risk gene and the developmental defect. Calcium imaging in intact organoids shows that these early-stage developmental changes are followed by abnormal circuit activity. This research uncovers cell-type-specific neurodevelopmental abnormalities that are shared across ASD risk genes and are finely modulated by human genomic context, finding convergence in the neurobiological basis of how different risk genes contribute to ASD pathology.


Assuntos
Transtorno do Espectro Autista , Predisposição Genética para Doença , Neurônios , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Córtex Cerebral/citologia , Proteínas de Ligação a DNA/genética , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Histona-Lisina N-Metiltransferase/genética , Humanos , Neurônios/classificação , Neurônios/metabolismo , Neurônios/patologia , Organoides/citologia , Proteômica , RNA-Seq , Análise de Célula Única , Fatores de Transcrição/genética
7.
Semin Cell Dev Biol ; 111: 23-31, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32718852

RESUMO

Human brain organoids are self-organizing three-dimensional structures that emerge from human pluripotent stem cells and mimic aspects of the cellular composition and functionality of the developing human brain. Despite their impressive self-organizing capacity, organoids lack the stereotypic structural anatomy of their in vivo counterpart, making conventional analysis techniques underpowered to assess cellular composition and gene network regulation in organoids. Advances in single cell transcriptomics have recently allowed characterization and improvement of organoid protocols, as they continue to evolve, by enabling identification of cell types and states along with their developmental origins. In this review, we summarize recent approaches, progresses and challenges in resolving brain organoid's complexity through single-cell transcriptomics. We then discuss emerging technologies that may complement single-cell RNA sequencing by providing additional readouts of cellular states to generate an organ-level view of developmental processes. Altogether, these integrative technologies will allow monitoring of global gene regulation in thousands of individual cells and will offer an unprecedented opportunity to investigate features of human brain development and disease across multiple cellular modalities and with cell-type resolution.


Assuntos
Encéfalo/metabolismo , Proteínas do Tecido Nervoso/genética , Malformações do Sistema Nervoso/genética , Organoides/metabolismo , Análise de Célula Única/métodos , Transcriptoma , Encéfalo/patologia , Diferenciação Celular , Linhagem da Célula/genética , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Regulação da Expressão Gênica , Humanos , Modelos Biológicos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/patologia , Malformações do Sistema Nervoso/fisiopatologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Organoides/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Análise de Sequência de RNA
8.
Neuron ; 107(6): 1014-1028, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32970996

RESUMO

The recent advent of human pluripotent stem cell (PSC)-derived 3D brain organoids has opened a window into aspects of human brain development that were not accessible before, allowing tractable monitoring and assessment of early developmental processes. However, their broad and effective use for modeling later stages of human brain development and disease is hampered by the lack of a stereotypic anatomical organization, which limits maturation processes dependent upon formation of unique cellular interactions and short- and long-range network connectivity. Emerging methods and technologies aimed at tighter regulatory control through bioengineering approaches, along with newer unbiased organoid analysis readouts, should resolve several of the current limitations. Here, we review recent advances in brain organoid generation and characterization with a focus on highlighting future directions utilizing interdisciplinary strategies that will be important for improving the physiological relevance of this model system.


Assuntos
Encéfalo/citologia , Crescimento Neuronal , Organoides/citologia , Cultura Primária de Células/métodos , Encéfalo/metabolismo , Encéfalo/fisiologia , Genômica/métodos , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Organoides/metabolismo , Organoides/fisiologia
9.
Nature ; 570(7762): 523-527, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31168097

RESUMO

Experimental models of the human brain are needed for basic understanding of its development and disease1. Human brain organoids hold unprecedented promise for this purpose; however, they are plagued by high organoid-to-organoid variability2,3. This has raised doubts as to whether developmental processes of the human brain can occur outside the context of embryogenesis with a degree of reproducibility that is comparable to the endogenous tissue. Here we show that an organoid model of the dorsal forebrain can reliably generate a rich diversity of cell types appropriate for the human cerebral cortex. We performed single-cell RNA-sequencing analysis of 166,242 cells isolated from 21 individual organoids, finding that 95% of the organoids generate a virtually indistinguishable compendium of cell types, following similar developmental trajectories and with a degree of organoid-to-organoid variability comparable to that of individual endogenous brains. Furthermore, organoids derived from different stem cell lines show consistent reproducibility in the cell types produced. The data demonstrate that reproducible development of the complex cellular diversity of the central nervous system does not require the context of the embryo, and that establishment of terminal cell identity is a highly constrained process that can emerge from diverse stem cell origins and growth environments.


Assuntos
Córtex Cerebral/citologia , Organoides/citologia , Técnicas de Cultura de Tecidos , Linhagem Celular , Córtex Cerebral/metabolismo , Feminino , Feto/citologia , Feto/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Organoides/metabolismo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , RNA-Seq , Reprodutibilidade dos Testes , Análise de Célula Única , Fatores de Tempo , Técnicas de Cultura de Tecidos/normas , Transcriptoma/genética
10.
11.
Curr Top Dev Biol ; 129: 99-122, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29801532

RESUMO

The study of the cellular and molecular processes of the developing human brain has been hindered by access to suitable models of living human brain tissue. Recently developed 3D cell culture models offer the promise of studying fundamental brain processes in the context of human genetic background and species-specific developmental mechanisms. Here, we review the current state of 3D human brain organoid models and consider their potential to enable investigation of complex aspects of human brain development and the underpinning of human neurological disease.


Assuntos
Encefalopatias/patologia , Encéfalo/embriologia , Técnicas de Cultura de Células/métodos , Modelos Biológicos , Animais , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
12.
Curr Opin Cell Biol ; 49: 47-52, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29227864

RESUMO

Three-dimensional (3D) brain organoids derived from human pluripotent stem cells hold great potential to investigate complex human genetic states and to model aspects of human brain development and pathology. However, the field of brain organoids is still in its infancy, and their use has been limited by their variability and their inability to differentiate into 3D structures with reproducible anatomical organization. Here, starting from a review of basic principles of in vitro 'brain organogenesis', we discuss which aspects of human brain development and disease can be faithfully modeled with current brain organoid protocols, and discuss improvements that would allow them to become reliable tools to investigate complex features of human brain development and disease.


Assuntos
Encéfalo/patologia , Organogênese/genética , Organoides/crescimento & desenvolvimento , Células-Tronco Pluripotentes/metabolismo , Humanos
13.
Nature ; 545(7652): 48-53, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28445462

RESUMO

In vitro models of the developing brain such as three-dimensional brain organoids offer an unprecedented opportunity to study aspects of human brain development and disease. However, the cells generated within organoids and the extent to which they recapitulate the regional complexity, cellular diversity and circuit functionality of the brain remain undefined. Here we analyse gene expression in over 80,000 individual cells isolated from 31 human brain organoids. We find that organoids can generate a broad diversity of cells, which are related to endogenous classes, including cells from the cerebral cortex and the retina. Organoids could be developed over extended periods (more than 9 months), allowing for the establishment of relatively mature features, including the formation of dendritic spines and spontaneously active neuronal networks. Finally, neuronal activity within organoids could be controlled using light stimulation of photosensitive cells, which may offer a way to probe the functionality of human neuronal circuits using physiological sensory stimuli.


Assuntos
Encéfalo/citologia , Vias Neurais/fisiologia , Neurogênese , Organoides/citologia , Organoides/efeitos da radiação , Linhagem Celular , Separação Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Dendritos , Perfilação da Expressão Gênica , Humanos , Técnicas In Vitro , Luz , Rede Nervosa/citologia , Rede Nervosa/efeitos da radiação , Vias Neurais/citologia , Vias Neurais/efeitos da radiação , Especificidade de Órgãos , Organoides/crescimento & desenvolvimento , Células Fotorreceptoras de Vertebrados/citologia , Células-Tronco Pluripotentes/citologia , Retina/citologia , Retina/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Fatores de Tempo , Transcriptoma
14.
Nat Med ; 22(11): 1220-1228, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27783065

RESUMO

Neuropsychiatric disorders such as autism spectrum disorder (ASD), schizophrenia (SCZ) and bipolar disorder (BPD) are of great societal and medical importance, but the complexity of these diseases and the challenges of modeling the development and function of the human brain have made these disorders difficult to study experimentally. The recent development of 3D brain organoids derived from human pluripotent stem cells offers a promising approach for investigating the phenotypic underpinnings of these highly polygenic disorders and for understanding the contribution of individual risk variants and complex genetic background to human pathology. Here we discuss the advantages, limitations and future applications of human brain organoids as in vitro models of neuropsychiatric disease.


Assuntos
Encéfalo/fisiopatologia , Células-Tronco Pluripotentes Induzidas/citologia , Transtornos Mentais/fisiopatologia , Rede Nervosa/fisiopatologia , Organoides/fisiopatologia , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/fisiopatologia , Transtorno Bipolar/genética , Transtorno Bipolar/fisiopatologia , Humanos , Técnicas In Vitro , Transtornos Mentais/genética , Modelos Neurológicos , Modelos Psicológicos , Rede Nervosa/citologia , Neurogênese , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Esquizofrenia/genética , Esquizofrenia/fisiopatologia
15.
Cell Stem Cell ; 18(3): 297-9, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26942845

RESUMO

How do astrocyte-derived neurons deal with the sudden loss of their glial identity? Exciting new findings from Gascón et al. (2016) single out metabolic conversion as a critical checkpoint for direct neuronal reprogramming.


Assuntos
Neuroglia , Neurônios/metabolismo , Astrócitos
16.
Brain ; 138(Pt 7): 1843-62, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25981963

RESUMO

Regeneration of injured central nervous system axons is highly restricted, causing neurological impairment. To date, although the lack of intrinsic regenerative potential is well described, a key regulatory molecular mechanism for the enhancement of both axonal regrowth and functional recovery after central nervous system injury remains elusive. While ubiquitin ligases coordinate neuronal morphogenesis and connectivity during development as well as after axonal injury, their role specifically in axonal regeneration is unknown. Following a bioinformatics network analysis combining ubiquitin ligases with previously defined axonal regenerative proteins, we found a triad composed of the ubiquitin ligases MDM4, MDM2 and the transcription factor p53 (encoded by TP53) as a putative central signalling complex restricting the regeneration program. Indeed, conditional deletion of MDM4 or pharmacological inhibition of MDM2/p53 interaction in the eye and spinal cord promote axonal regeneration and sprouting of the optic nerve after crush and of supraspinal tracts after spinal cord injury. The double conditional deletion of MDM4-p53 as well as MDM2 inhibition in p53-deficient mice blocks this regenerative phenotype, showing its dependence upon p53. Genome-wide gene expression analysis from ex vivo fluorescence-activated cell sorting in MDM4-deficient retinal ganglion cells identifies the downstream target IGF1R, whose activity and expression was found to be required for the regeneration elicited by MDM4 deletion. Importantly, we demonstrate that pharmacological enhancement of the MDM2/p53-IGF1R axis enhances axonal sprouting as well as functional recovery after spinal cord injury. Thus, our results show MDM4-MDM2/p53-IGF1R as an original regulatory mechanism for CNS regeneration and offer novel targets to enhance neurological recovery.media-1vid110.1093/brain/awv125_video_abstractawv125_video_abstract.


Assuntos
Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/metabolismo , Recuperação de Função Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Traumatismos da Medula Espinal/metabolismo , Animais , Axônios/metabolismo , Axônios/patologia , Biologia Computacional , Modelos Animais de Doenças , Citometria de Fluxo , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Compressão Nervosa , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/fisiopatologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Transcriptoma , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
17.
Nat Cell Biol ; 16(11): 1045-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25283993

RESUMO

The vasculature is a prominent component of the subventricular zone neural stem cell niche. Although quiescent neural stem cells physically contact blood vessels at specialized endfeet, the significance of this interaction is not understood. In contrast, it is well established that vasculature-secreted soluble factors promote lineage progression of committed progenitors. Here we specifically investigated the role of cell-cell contact-dependent signalling in the vascular niche. Unexpectedly, we find that direct cell-cell interactions with endothelial cells enforce quiescence and promote stem cell identity. Mechanistically, endothelial ephrinB2 and Jagged1 mediate these effects by suppressing cell-cycle entry downstream of mitogens and inducing stemness genes to jointly inhibit differentiation. In vivo, endothelial-specific ablation of either of the genes which encode these proteins, Efnb2 and Jag1 respectively, aberrantly activates quiescent stem cells, resulting in depletion. Thus, we identify the vasculature as a critical niche compartment for stem cell maintenance, furthering our understanding of how anchorage to the niche maintains stem cells within a pro-differentiative microenvironment.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Efrina-B2/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Neurais/citologia , Nicho de Células-Tronco/fisiologia , Animais , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Células Endoteliais/citologia , Humanos , Proteína Jagged-1 , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Proteínas Serrate-Jagged , Nicho de Células-Tronco/genética
18.
J Neurosci ; 34(25): 8630-45, 2014 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-24948817

RESUMO

Correlative evidence suggests that GABAergic signaling plays an important role in the regulation of activity-dependent hippocampal neurogenesis and emotional behavior in adult mice. However, whether these are causally linked at the molecular level remains elusive. Nuclear factor of activated T cell (NFAT) proteins are activity-dependent transcription factors that respond to environmental stimuli in different cell types, including hippocampal newborn neurons. Here, we identify NFATc4 as a key activity-dependent transcriptional regulator of GABA signaling in hippocampal progenitor cells via an unbiased high-throughput genome-wide study. Next, we demonstrate that GABAA receptor (GABAAR) signaling modulates hippocampal neurogenesis through NFATc4 activity, which in turn regulates GABRA2 and GABRA4 subunit expression via binding to specific promoter responsive elements, as assessed by ChIP and luciferase assays. Furthermore, we show that selective pharmacological enhancement of GABAAR activity promotes hippocampal neurogenesis via the calcineurin/NFATc4 axis. Importantly, the NFATc4-dependent increase in hippocampal neurogenesis after GABAAR stimulation is required for the suppression of the anxiety response in mice. Together, these data provide a novel molecular insight into the regulation of the anxiety response in mice, suggesting that the GABAAR/NFATc4 axis is a druggable target for the therapy of emotional disorders.


Assuntos
Ansiedade/metabolismo , Ansiedade/prevenção & controle , Fatores de Transcrição NFATC/metabolismo , Neurogênese/fisiologia , Receptores de GABA-A/fisiologia , Transdução de Sinais/fisiologia , Animais , Ansiedade/patologia , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout
20.
J Neurosci ; 33(36): 14318-30, 2013 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-24005285

RESUMO

Mounting evidence points to a role for endogenous reactive oxygen species (ROS) in cell signaling, including in the control of cell proliferation, differentiation, and fate. However, the function of ROS and their molecular regulation in embryonic mouse neural progenitor cells (eNPCs) has not yet been clarified. Here, we describe that physiological ROS are required for appropriate timing of neurogenesis in the developing telencephalon in vivo and in cultured NPCs, and that the tumor suppressor p53 plays a key role in the regulation of ROS-dependent neurogenesis. p53 loss of function leads to elevated ROS and early neurogenesis, while restoration of p53 and antioxidant treatment partially reverse the phenotype associated with premature neurogenesis. Furthermore, we describe that the expression of a number of neurogenic and oxidative stress genes relies on p53 and that both p53 and ROS-dependent induction of neurogenesis depend on PI3 kinase/phospho-Akt signaling. Our results suggest that p53 fine-tunes endogenous ROS levels to ensure the appropriate timing of neurogenesis in eNPCs. This may also have implications for the generation of tumors of neurodevelopmental origin.


Assuntos
Células-Tronco Neurais/metabolismo , Neurogênese , Fosfatidilinositol 3-Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Células Cultivadas , Camundongos , Células-Tronco Neurais/citologia , Estresse Oxidativo/genética , Telencéfalo/citologia , Telencéfalo/embriologia , Telencéfalo/metabolismo , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA