Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuron ; 112(9): 1426-1443.e11, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38442714

RESUMO

Glucocorticoids are important for proper organ maturation, and their levels are tightly regulated during development. Here, we use human cerebral organoids and mice to study the cell-type-specific effects of glucocorticoids on neurogenesis. We show that glucocorticoids increase a specific type of basal progenitors (co-expressing PAX6 and EOMES) that has been shown to contribute to cortical expansion in gyrified species. This effect is mediated via the transcription factor ZBTB16 and leads to increased production of neurons. A phenome-wide Mendelian randomization analysis of an enhancer variant that moderates glucocorticoid-induced ZBTB16 levels reveals causal relationships with higher educational attainment and altered brain structure. The relationship with postnatal cognition is also supported by data from a prospective pregnancy cohort study. This work provides a cellular and molecular pathway for the effects of glucocorticoids on human neurogenesis that relates to lasting postnatal phenotypes.


Assuntos
Córtex Cerebral , Glucocorticoides , Neurogênese , Proteína com Dedos de Zinco da Leucemia Promielocítica , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Humanos , Animais , Camundongos , Glucocorticoides/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/citologia , Feminino , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Gravidez , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Masculino
2.
Sci Adv ; 9(20): eadd8164, 2023 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-37205765

RESUMO

Disruption in neurogenesis and neuronal migration can influence the assembly of cortical circuits, affecting the excitatory-inhibitory balance and resulting in neurodevelopmental and neuropsychiatric disorders. Using ventral cerebral organoids and dorsoventral cerebral assembloids with mutations in the extracellular matrix gene LGALS3BP, we show that extracellular vesicles released into the extracellular environment regulate the molecular differentiation of neurons, resulting in alterations in migratory dynamics. To investigate how extracellular vesicles affect neuronal specification and migration dynamics, we collected extracellular vesicles from ventral cerebral organoids carrying a mutation in LGALS3BP, previously identified in individuals with cortical malformations and neuropsychiatric disorders. These results revealed differences in protein composition and changes in dorsoventral patterning. Proteins associated with cell fate decision, neuronal migration, and extracellular matrix composition were altered in mutant extracellular vesicles. Moreover, we show that treatment with extracellular vesicles changes the transcriptomic profile in neural progenitor cells. Our results indicate that neuronal molecular differentiation can be influenced by extracellular vesicles.


Assuntos
Vesículas Extracelulares , Neurônios , Humanos , Neurônios/metabolismo , Interneurônios , Neurogênese , Diferenciação Celular/genética
3.
Front Neurosci ; 16: 1009125, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36340763

RESUMO

The neural stem cell niche is a key regulator participating in the maintenance, regeneration, and repair of the brain. Within the niche neural stem cells (NSC) generate new neurons throughout life, which is important for tissue homeostasis and brain function. NSCs are regulated by intrinsic and extrinsic factors with cellular metabolism being lately recognized as one of the most important ones, with evidence suggesting that it may serve as a common signal integrator to ensure mammalian brain homeostasis. The aim of this review is to summarize recent insights into how metabolism affects NSC fate decisions in adult neural stem cell niches, with occasional referencing of embryonic neural stem cells when it is deemed necessary. Specifically, we will highlight the implication of mitochondria as crucial regulators of NSC fate decisions and the relationship between metabolism and ependymal cells. The link between primary cilia dysfunction in the region of hypothalamus and metabolic diseases will be examined as well. Lastly, the involvement of metabolic pathways in ependymal cell ciliogenesis and physiology regulation will be discussed.

4.
Front Neurosci ; 16: 1031075, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36340790

RESUMO

Neurodevelopmental disorders (NDDs) are a heterogeneous group of impairments that affect the development of the central nervous system leading to abnormal brain function. NDDs affect a great percentage of the population worldwide, imposing a high societal and economic burden and thus, interest in this field has widely grown in recent years. Nevertheless, the complexity of human brain development and function as well as the limitations regarding human tissue usage make their modeling challenging. Animal models play a central role in the investigation of the implicated molecular and cellular mechanisms, however many of them display key differences regarding human phenotype and in many cases, they partially or completely fail to recapitulate them. Although in vitro two-dimensional (2D) human-specific models have been highly used to address some of these limitations, they lack crucial features such as complexity and heterogeneity. In this review, we will discuss the advantages, limitations and future applications of in vivo and in vitro models that are used today to model NDDs. Additionally, we will describe the recent development of 3-dimensional brain (3D) organoids which offer a promising approach as human-specific in vitro models to decipher these complex disorders.

5.
Am J Psychiatry ; 179(5): 375-387, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34698522

RESUMO

OBJECTIVE: A fine-tuned balance of glucocorticoid receptor (GR) activation is essential for organ formation, with disturbances influencing many health outcomes. In utero, glucocorticoids have been linked to brain-related negative outcomes, with unclear underlying mechanisms, especially regarding cell-type-specific effects. An in vitro model of fetal human brain development, induced human pluripotent stem cell (hiPSC)-derived cerebral organoids, was used to test whether cerebral organoids are suitable for studying the impact of prenatal glucocorticoid exposure on the developing brain. METHODS: The GR was activated with the synthetic glucocorticoid dexamethasone, and the effects were mapped using single-cell transcriptomics across development. RESULTS: The GR was expressed in all cell types, with increasing expression levels through development. Not only did its activation elicit translocation to the nucleus and the expected effects on known GR-regulated pathways, but also neurons and progenitor cells showed targeted regulation of differentiation- and maturation-related transcripts. Uniquely in neurons, differentially expressed transcripts were significantly enriched for genes associated with behavior-related phenotypes and disorders. This human neuronal glucocorticoid response profile was validated across organoids from three independent hiPSC lines reprogrammed from different source tissues from both male and female donors. CONCLUSIONS: These findings suggest that excessive glucocorticoid exposure could interfere with neuronal maturation in utero, leading to increased disease susceptibility through neurodevelopmental processes at the interface of genetic susceptibility and environmental exposure. Cerebral organoids are a valuable translational resource for exploring the effects of glucocorticoids on early human brain development.


Assuntos
Células-Tronco Pluripotentes Induzidas , Receptores de Glucocorticoides , Encéfalo/metabolismo , Dexametasona/metabolismo , Dexametasona/farmacologia , Feminino , Glucocorticoides/efeitos adversos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Organoides/metabolismo , Gravidez , Receptores de Glucocorticoides/genética
6.
Nat Commun ; 12(1): 6298, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728600

RESUMO

Basal progenitors (BPs), including intermediate progenitors and basal radial glia, are generated from apical radial glia and are enriched in gyrencephalic species like humans, contributing to neuronal expansion. Shortly after generation, BPs delaminate towards the subventricular zone, where they further proliferate before differentiation. Gene expression alterations involved in BP delamination and function in humans are poorly understood. Here, we study the role of LGALS3BP, so far known as a cancer biomarker, which is a secreted protein enriched in human neural progenitors (NPCs). We show that individuals with LGALS3BP de novo variants exhibit altered local gyrification, sulcal depth, surface area and thickness in their cortex. Additionally, using cerebral organoids, human fetal tissues and mice, we show that LGALS3BP regulates the position of NPCs. Single-cell RNA-sequencing and proteomics reveal that LGALS3BP-mediated mechanisms involve the extracellular matrix in NPCs' anchoring and migration within the human brain. We propose that its temporal expression influences NPCs' delamination, corticogenesis and gyrification extrinsically.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Córtex Cerebral/citologia , Vesículas Extracelulares/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Neocórtex/citologia , Células-Tronco Neurais/citologia , Neuroglia/metabolismo , Animais , Diferenciação Celular , Córtex Cerebral/metabolismo , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo
7.
Front Mol Biosci ; 7: 578137, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33330619

RESUMO

Cortical development is a very complex process in which any temporal or spatial alterations can give rise to a wide range of cortical malformations. Among those malformations, periventricular heterotopia (PH) is characterized by clusters of neurons that do not migrate to the correct place. Cerebral organoids derived from patients with mutations in DCHS1 and FAT4, which have been associated with PH, exhibit higher levels of GNG5 expression in a patient-specific cluster of neurons. Here we investigate the role of GNG5 during the development of the cerebral cortex in mice and human cerebral organoids. GNG5, highly expressed in progenitors and downregulated in neurons, is critical for controlling the number of apical and basal progenitors and neuronal migration. Moreover, forced expression of GNG5 recapitulates some of the alterations observed upon downregulation of Dchs1 and Fat4 in mice and human cerebral organoids derived from DCHS1 and FAT4 patients, suggesting a critical role of GNG5 in cortical development.

8.
Adv Exp Med Biol ; 1195: 35-41, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32468456

RESUMO

Human brain possesses a unique anatomy and physiology. For centuries, methodological barriers and ethical challenges in accessing human brain tissues have restricted researchers into using 2-D cell culture systems and model organisms as a tool for investigating the mechanisms underlying neurological disorders in humans. However, our understanding regarding the human brain development and diseases has been recently extended due to the generation of 3D brain organoids, grown from human stem cells or induced pluripotent stem cells (iPSCs). This system evolved into an attractive model of brain diseases as it recapitulates to a great extend the cellular organization and the microenvironment of a human brain. This chapter focuses on the application of brain organoids in modelling several neurodevelopmental and neurodegenerative diseases.


Assuntos
Encéfalo/patologia , Doenças Neurodegenerativas/patologia , Transtornos do Neurodesenvolvimento/patologia , Organoides/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia
9.
EMBO Mol Med ; 12(6): e11419, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32378798

RESUMO

Progressive myoclonus epilepsy (PME) of Unverricht-Lundborg type (EPM1) is an autosomal recessive neurodegenerative disorder with the highest incidence of PME worldwide. Mutations in the gene encoding cystatin B (CSTB) are the primary genetic cause of EPM1. Here, we investigate the role of CSTB during neurogenesis in vivo in the developing mouse brain and in vitro in human cerebral organoids (hCOs) derived from EPM1 patients. We find that CSTB (but not one of its pathological variants) is secreted into the mouse cerebral spinal fluid and the conditioned media from hCOs. In embryonic mouse brain, we find that functional CSTB influences progenitors' proliferation and modulates neuronal distribution by attracting interneurons to the site of secretion via cell-non-autonomous mechanisms. Similarly, in patient-derived hCOs, low levels of functional CSTB result in an alteration of progenitor's proliferation, premature differentiation, and changes in interneurons migration. Secretion and extracellular matrix organization are the biological processes particularly affected as suggested by a proteomic analysis in patients' hCOs. Overall, our study sheds new light on the cellular mechanisms underlying the development of EPM1.


Assuntos
Síndrome de Unverricht-Lundborg , Animais , Proliferação de Células , Cistatina B/genética , Humanos , Interneurônios , Camundongos , Neurogênese , Proteômica
10.
EMBO Rep ; 21(5): e48204, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32207244

RESUMO

During embryonic development, excitatory projection neurons migrate in the cerebral cortex giving rise to organised layers. Periventricular heterotopia (PH) is a group of aetiologically heterogeneous disorders in which a subpopulation of newborn projection neurons fails to initiate their radial migration to the cortex, ultimately resulting in bands or nodules of grey matter lining the lateral ventricles. Although a number of genes have been implicated in its cause, currently they only satisfactorily explain the pathogenesis of the condition for 50% of patients. Novel gene discovery is complicated by the extreme genetic heterogeneity recently described to underlie its cause. Here, we study the neurodevelopmental role of endothelin-converting enzyme-2 (ECE2) for which two biallelic variants have been identified in two separate patients with PH. Our results show that manipulation of ECE2 levels in human cerebral organoids and in the developing mouse cortex leads to ectopic localisation of neural progenitors and neurons. We uncover the role of ECE2 in neurogenesis, and mechanistically, we identify its involvement in the generation and secretion of extracellular matrix proteins in addition to cytoskeleton and adhesion.


Assuntos
Neurogênese , Heterotopia Nodular Periventricular , Movimento Celular/genética , Córtex Cerebral , Feminino , Humanos , Neurogênese/genética , Neurônios , Gravidez
11.
RSC Chem Biol ; 1(4): 209-213, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34458760

RESUMO

Monoamine oxidases MAOA and MAOB catalyze important cellular functions such as the deamination of neurotransmitters. Correspondingly, MAO inhibitors are used for the treatment of severe neuropsychiatric disorders such as depression. A commonly prescribed drug against refractory depression is tranylcypromine, however, the side effects are poorly understood. In order to decipher putative off-targets, we synthesized two tranylcypromine probes equipped with either an alkyne moiety or an alkyne-diazirine minimal photocrosslinker for in situ proteome profiling. Surprisingly, LC-MS/MS analysis revealed low enrichment of MAOA and relatively promiscuous labeling of proteins. Photoprobe labeling paired with fluorescent imaging studies revealed lysosomal trapping which could be largely reverted by the addition of lysosomotropic drugs.

12.
Wiley Interdiscip Rev Dev Biol ; 9(1): e347, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31071759

RESUMO

The brain is one of the most complex organs, responsible for the advanced intellectual and cognitive ability of humans. Although primates are to some extent capable of performing cognitive tasks, their abilities are less evolved. One of the reasons for this is the vast differences in the brain of humans compared to other mammals, in terms of shape, size and complexity. Such differences make the study of human brain development fascinating. Interestingly, the cerebral cortex is by far the most complex brain region resulting from its selective evolution within mammals over millions of years. Unraveling the molecular and cellular mechanisms regulating brain development, as well as the evolutionary differences seen across species and the need to understand human brain disorders, are some of the reasons why scientists are interested in improving their current knowledge on human corticogenesis. Toward this end, several animal models including primates have been used, however, these models are limited in their extent to recapitulate human-specific features. Recent technological achievements in the field of stem cell research, which have enabled the generation of human models of corticogenesis, called brain or cerebral organoids, are of great importance. This review focuses on the main cellular and molecular features of human corticogenesis and the use of brain organoids to study it. We will discuss the key differences between cortical development in human and nonhuman mammals, the technological applications of brain organoids and the different aspects of cortical development in normal and pathological conditions, which can be modeled using brain organoids. This article is categorized under: Comparative Development and Evolution > Regulation of Organ Diversity Nervous System Development > Vertebrates: General Principles.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Neurogênese/fisiologia , Organoides/crescimento & desenvolvimento , Animais , Humanos , Mamíferos/crescimento & desenvolvimento
13.
Cereb Cortex ; 30(6): 3467-3482, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-31867667

RESUMO

Neocortex development depends on neural stem cell proliferation, cell differentiation, neurogenesis, and neuronal migration. Cytoskeletal regulation is critical for all these processes, but the underlying mechanisms are only poorly understood. We previously implicated the cytoskeletal regulator profilin1 in cerebellar granule neuron migration. Since we found profilin1 expressed throughout mouse neocortex development, we here tested the hypothesis that profilin1 is crucial for neocortex development. We found no evidence for impaired neuron migration or layering in the neocortex of profilin1 mutant mice. However, proliferative activity at basal positions was doubled in the mutant neocortex during mid-neurogenesis, with a drastic and specific increase in basal Pax6+ cells indicative for elevated numbers of basal radial glia (bRG). This was accompanied by transiently increased neurogenesis and associated with mild invaginations resembling rudimentary neocortex folds. Our data are in line with a model in which profilin1-dependent actin assembly controls division of apical radial glia (aRG) and thereby the fate of their progenies. Via this mechanism, profilin1 restricts cell delamination from the ventricular surface and, hence, bRG production and thereby controls neocortex development in mice. Our data support the radial cone hypothesis" claiming that elevated bRG number causes neocortex folds.


Assuntos
Actinas/metabolismo , Proliferação de Células/genética , Células Ependimogliais/citologia , Neocórtex/embriologia , Neurogênese/genética , Profilinas/genética , Citoesqueleto de Actina , Animais , Divisão Celular/genética , Camundongos , Mutação , Células-Tronco Neurais
14.
Glia ; 67(12): 2360-2373, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31328313

RESUMO

The subventricular zone (SVZ) is one of two main niches where neurogenesis persists during adulthood, as it retains neural stem cells (NSCs) with self-renewal capacity and multi-lineage potency. Another critical cellular component of the niche is the population of postmitotic multiciliated ependymal cells. Both cell types are derived from radial glial cells that become specified to each lineage during embryogenesis. We show here that GemC1, encoding Geminin coiled-coil domain-containing protein 1, is associated with congenital hydrocephalus in humans and mice. Our results show that GemC1 deficiency drives cells toward a NSC phenotype, at the expense of multiciliated ependymal cell generation. The increased number of NSCs is accompanied by increased levels of proliferation and neurogenesis in the postnatal SVZ. Finally, GemC1-knockout cells display altered chromatin organization at multiple loci, further supporting a NSC identity. Together, these findings suggest that GemC1 regulates the balance between NSC generation and ependymal cell differentiation, with implications for the pathogenesis of human congenital hydrocephalus.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proteínas de Ciclo Celular/deficiência , Genes de Troca/fisiologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Animais , Encéfalo/citologia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Gravidez
15.
J Cell Sci ; 132(11)2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31028178

RESUMO

A distinct combination of transcription factors elicits the acquisition of a specific fate and the initiation of a differentiation program. Multiciliated cells (MCCs) are a specialized type of epithelial cells that possess dozens of motile cilia on their apical surface. Defects in cilia function have been associated with ciliopathies that affect many organs, including brain and airway epithelium. Here we show that the geminin coiled-coil domain-containing protein 1 GemC1 (also known as Lynkeas) regulates the transcriptional activation of p73, a transcription factor central to multiciliogenesis. Moreover, we show that GemC1 acts in a trimeric complex with transcription factor E2F5 and tumor protein p73 (officially known as TP73), and that this complex is important for the activation of the p73 promoter. We also provide in vivo evidence that GemC1 is necessary for p73 expression in different multiciliated epithelia. We further show that GemC1 regulates multiciliogenesis through the control of chromatin organization, and the epigenetic marks/tags of p73 and Foxj1. Our results highlight novel signaling cues involved in the commitment program of MCCs across species and tissues.This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cílios/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/genética , Proteína Tumoral p73/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Células Epiteliais/citologia , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Ativação Transcricional/genética , Proteína Tumoral p73/genética
16.
Nat Med ; 25(4): 561-568, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30858616

RESUMO

Malformations of the human cortex represent a major cause of disability1. Mouse models with mutations in known causal genes only partially recapitulate the phenotypes and are therefore not unlimitedly suited for understanding the molecular and cellular mechanisms responsible for these conditions2. Here we study periventricular heterotopia (PH) by analyzing cerebral organoids derived from induced pluripotent stem cells (iPSCs) of patients with mutations in the cadherin receptor-ligand pair DCHS1 and FAT4 or from isogenic knockout (KO) lines1,3. Our results show that human cerebral organoids reproduce the cortical heterotopia associated with PH. Mutations in DCHS1 and FAT4 or knockdown of their expression causes changes in the morphology of neural progenitor cells and result in defective neuronal migration dynamics only in a subset of neurons. Single-cell RNA-sequencing (scRNA-seq) data reveal a subpopulation of mutant neurons with dysregulated genes involved in axon guidance, neuronal migration and patterning. We suggest that defective neural progenitor cell (NPC) morphology and an altered navigation system in a subset of neurons underlie this form of PH.


Assuntos
Movimento Celular , Cérebro/patologia , Neurônios/patologia , Organoides/patologia , Heterotopia Nodular Periventricular/patologia , Proteínas Relacionadas a Caderinas , Caderinas/genética , Linhagem Celular , Humanos , Recém-Nascido , Mutação/genética , Análise de Sequência de RNA , Análise de Célula Única , Imagem com Lapso de Tempo , Proteínas Supressoras de Tumor/genética
17.
Cell Rep ; 25(10): 2729-2741.e6, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517861

RESUMO

The mammalian neocortex has undergone remarkable changes through evolution. A consequence of such rapid evolutionary events could be a trade-off that has rendered the brain susceptible to certain neurodevelopmental and neuropsychiatric conditions. We analyzed the exomes of 65 patients with the structural brain malformation periventricular nodular heterotopia (PH). De novo coding variants were observed in excess in genes defining a transcriptomic signature of basal radial glia, a cell type linked to brain evolution. In addition, we located two variants in human isoforms of two genes that have no ortholog in mice. Modulating the levels of one of these isoforms for the gene PLEKHG6 demonstrated its role in regulating neuroprogenitor differentiation and neuronal migration via RhoA, with phenotypic recapitulation of PH in human cerebral organoids. This suggests that this PLEKHG6 isoform is an example of a primate-specific genomic element supporting brain development.


Assuntos
Movimento Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Alelos , Animais , Sequência de Bases , Encéfalo/embriologia , Encéfalo/metabolismo , Exoma/genética , Regulação da Expressão Gênica , Genoma , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Recém-Nascido , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Organoides/embriologia , Primatas , Isoformas de Proteínas/metabolismo , Especificidade da Espécie , Proteína rhoA de Ligação ao GTP/metabolismo
18.
Cell ; 174(3): 590-606.e21, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29961574

RESUMO

Cerebral cortex size differs dramatically between reptiles, birds, and mammals, owing to developmental differences in neuron production. In mammals, signaling pathways regulating neurogenesis have been identified, but genetic differences behind their evolution across amniotes remain unknown. We show that direct neurogenesis from radial glia cells, with limited neuron production, dominates the avian, reptilian, and mammalian paleocortex, whereas in the evolutionarily recent mammalian neocortex, most neurogenesis is indirect via basal progenitors. Gain- and loss-of-function experiments in mouse, chick, and snake embryos and in human cerebral organoids demonstrate that high Slit/Robo and low Dll1 signaling, via Jag1 and Jag2, are necessary and sufficient to drive direct neurogenesis. Attenuating Robo signaling and enhancing Dll1 in snakes and birds recapitulates the formation of basal progenitors and promotes indirect neurogenesis. Our study identifies modulation in activity levels of conserved signaling pathways as a primary mechanism driving the expansion and increased complexity of the mammalian neocortex during amniote evolution.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Córtex Cerebral/metabolismo , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Proteína Jagged-2 , Mamíferos/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Neocórtex/fisiologia , Células-Tronco Neurais , Neurogênese/fisiologia , Neuroglia/fisiologia , Neurônios , Fator de Transcrição PAX6/metabolismo , Proteínas Repressoras , Transdução de Sinais , Serpentes/embriologia , Proteínas Roundabout
19.
Front Cell Neurosci ; 12: 57, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29593499

RESUMO

Disorders of neuronal mispositioning during brain development are phenotypically heterogeneous and their genetic causes remain largely unknown. Here, we report biallelic variants in a Hippo signaling factor-MOB2-in a patient with one such disorder, periventricular nodular heterotopia (PH). Genetic and cellular analysis of both variants confirmed them to be loss-of-function with enhanced sensitivity to transcript degradation via nonsense mediated decay (NMD) or increased protein turnover via the proteasome. Knockdown of Mob2 within the developing mouse cortex demonstrated its role in neuronal positioning. Cilia positioning and number within migrating neurons was also impaired with comparable defects detected following a reduction in levels of an upstream modulator of Mob2 function, Dchs1, a previously identified locus associated with PH. Moreover, reduced Mob2 expression increased phosphorylation of Filamin A, an actin cross-linking protein frequently mutated in cases of this disorder. These results reveal a key role for Mob2 in correct neuronal positioning within the developing cortex and outline a new candidate locus for PH development.

20.
Stem Cells Dev ; 26(16): 1214-1222, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28557659

RESUMO

Neural stem cells have been considered as a source of stem cells that can be used for cell replacement therapies in neurodegenerative diseases, as they can be isolated and expanded in vitro and can be used for autologous grafting. However, due to low percentages of survival and varying patterns of differentiation, strategies that will enhance the efficacy of transplantation are under scrutiny. In this article, we have examined whether alterations in Geminin's expression, a protein that coordinates the balance between self-renewal and differentiation, can improve the properties of stem cells transplanted in 6-OHDA hemiparkinsonian mouse model. Our results indicate that, in the absence of Geminin, grafted cells differentiating into dopaminergic neurons were decreased, while an increased number of oligodendrocytes were detected. The number of proliferating multipotent cells was not modified by the absence of Geminin. These findings encourage research related to the impact of Geminin on transplantations for neurodegenerative disorders, as an important molecule in influencing differentiation decisions of the cells composing the graft.


Assuntos
Células-Tronco Adultas/citologia , Geminina/genética , Células-Tronco Neurais/citologia , Neurogênese , Doença de Parkinson/terapia , Transplante de Células-Tronco , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/transplante , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Proliferação de Células , Células Cultivadas , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Geminina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Oxidopamina/toxicidade , Doença de Parkinson/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...