Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38527856

RESUMO

APP gene dosage is strongly associated with Alzheimer's disease (AD) pathogenesis. Genomic duplication of the APP locus leads to autosomal dominant early-onset AD. Individuals with Down syndrome (trisomy of chromosome 21) harbor 3 copies of the APP gene and invariably develop progressive AD with highly characteristic neuropathological features. Restoring expression of APP to the equivalent of that of two gene copies, or lower, is a rational therapeutic strategy, as it would restore physiological levels of neuronal APP protein without the potentially deleterious consequences of inadvertently inducing loss of APP function. Here we find that antisense oligonucleotides (ASOs) targeting APP are an effective approach to reduce APP protein levels and rescue endolysosome and autophagy dysfunction in APP duplication human induced pluripotent stem cell (hiPSC)-derived cortical neurons. Importantly, using ultrasensitive single-aggregate imaging techniques, we show that APP targeting ASOs significantly reduce both intracellular and extracellular Aß-containing aggregates. Our results highlight the potential of APP ASOs as a therapeutic approach for forms of AD caused by duplication of the APP gene, including monogenic AD and AD related to Down syndrome.

2.
Dev Biol ; 494: 60-70, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36509125

RESUMO

Neuroepithelial cells balance tissue growth requirement with the morphogenetic imperative of closing the neural tube. They apically constrict to generate mechanical forces which elevate the neural folds, but are thought to apically dilate during mitosis. However, we previously reported that mitotic neuroepithelial cells in the mouse posterior neuropore have smaller apical surfaces than non-mitotic cells. Here, we document progressive apical enrichment of non-muscle myosin-II in mitotic, but not non-mitotic, neuroepithelial cells with smaller apical areas. Live-imaging of the chick posterior neuropore confirms apical constriction synchronised with mitosis, reaching maximal constriction by anaphase, before division and re-dilation. Mitotic apical constriction amplitude is significantly greater than interphase constrictions. To investigate conservation in humans, we characterised early stages of iPSC differentiation through dual SMAD-inhibition to robustly produce pseudostratified neuroepithelia with apically enriched actomyosin. These cultured neuroepithelial cells achieve an equivalent apical area to those in mouse embryos. iPSC-derived neuroepithelial cells have large apical areas in G2 which constrict in M phase and retain this constriction in G1/S. Given that this differentiation method produces anterior neural identities, we studied the anterior neuroepithelium of the elevating mouse mid-brain neural tube. Instead of constricting, mid-brain mitotic neuroepithelial cells have larger apical areas than interphase cells. Tissue geometry differs between the apically convex early midbrain and flat posterior neuropore. Culturing human neuroepithelia on equivalently convex surfaces prevents mitotic apical constriction. Thus, neuroepithelial cells undergo high-amplitude apical constriction synchronised with cell cycle progression but the timing of their constriction if influenced by tissue geometry.


Assuntos
Mitose , Sistema Nervoso , Humanos , Animais , Camundongos , Constrição , Ciclo Celular , Diferenciação Celular/fisiologia
3.
Proc Natl Acad Sci U S A ; 116(45): 22754-22763, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31628250

RESUMO

Mutations in the thyroid hormone receptor α 1 gene (THRA) have recently been identified as a cause of intellectual deficit in humans. Patients present with structural abnormalities including microencephaly, reduced cerebellar volume and decreased axonal density. Here, we show that directed differentiation of THRA mutant patient-derived induced pluripotent stem cells to forebrain neural progenitors is markedly reduced, but mutant progenitor cells can generate deep and upper cortical layer neurons and form functional neuronal networks. Quantitative lineage tracing shows that THRA mutation-containing progenitor cells exit the cell cycle prematurely, resulting in reduced clonal output. Using a micropatterned chip assay, we find that spatial self-organization of mutation-containing progenitor cells in vitro is impaired, consistent with down-regulated expression of cell-cell adhesion genes. These results reveal that thyroid hormone receptor α1 is required for normal neural progenitor cell proliferation in human cerebral cortical development. They also exemplify quantitative approaches for studying neurodevelopmental disorders using patient-derived cells in vitro.


Assuntos
Mutação , Células-Tronco Neurais/citologia , Neurogênese/genética , Receptores alfa dos Hormônios Tireóideos/genética , Adolescente , Adesão Celular/genética , Diferenciação Celular , Proliferação de Células , Criança , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Pessoa de Meia-Idade
4.
Nature ; 526(7573): 443-7, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26322584

RESUMO

Alzheimer disease (AD) is characterized by the accumulation of amyloid plaques, which are predominantly composed of amyloid-ß peptide. Two principal physiological pathways either prevent or promote amyloid-ß generation from its precursor, ß-amyloid precursor protein (APP), in a competitive manner. Although APP processing has been studied in great detail, unknown proteolytic events seem to hinder stoichiometric analyses of APP metabolism in vivo. Here we describe a new physiological APP processing pathway, which generates proteolytic fragments capable of inhibiting neuronal activity within the hippocampus. We identify higher molecular mass carboxy-terminal fragments (CTFs) of APP, termed CTF-η, in addition to the long-known CTF-α and CTF-ß fragments generated by the α- and ß-secretases ADAM10 (a disintegrin and metalloproteinase 10) and BACE1 (ß-site APP cleaving enzyme 1), respectively. CTF-η generation is mediated in part by membrane-bound matrix metalloproteinases such as MT5-MMP, referred to as η-secretase activity. η-Secretase cleavage occurs primarily at amino acids 504-505 of APP695, releasing a truncated ectodomain. After shedding of this ectodomain, CTF-η is further processed by ADAM10 and BACE1 to release long and short Aη peptides (termed Aη-α and Aη-ß). CTFs produced by η-secretase are enriched in dystrophic neurites in an AD mouse model and in human AD brains. Genetic and pharmacological inhibition of BACE1 activity results in robust accumulation of CTF-η and Aη-α. In mice treated with a potent BACE1 inhibitor, hippocampal long-term potentiation was reduced. Notably, when recombinant or synthetic Aη-α was applied on hippocampal slices ex vivo, long-term potentiation was lowered. Furthermore, in vivo single-cell two-photon calcium imaging showed that hippocampal neuronal activity was attenuated by Aη-α. These findings not only demonstrate a major functionally relevant APP processing pathway, but may also indicate potential translational relevance for therapeutic strategies targeting APP processing.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/citologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Neurônios/fisiologia , Proteólise , Proteínas ADAM/metabolismo , Proteína ADAM10 , Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/líquido cefalorraquidiano , Secretases da Proteína Precursora do Amiloide/deficiência , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/deficiência , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Sinalização do Cálcio , Modelos Animais de Doenças , Feminino , Hipocampo/enzimologia , Hipocampo/fisiologia , Humanos , Técnicas In Vitro , Potenciação de Longa Duração , Masculino , Metaloproteinases da Matriz Associadas à Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Peso Molecular , Neuritos/enzimologia , Neuritos/metabolismo , Neurônios/enzimologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Placa Amiloide , Processamento de Proteína Pós-Traducional , Análise de Célula Única
5.
Development ; 142(1): 17-20, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516966

RESUMO

In September 2014, over 100 scientists from around the globe gathered at Wotton House near London for the Company of Biologists' workshop 'From Stem Cells to Human Development'. The workshop covered diverse aspects of human development, from the earliest stages of embryogenesis to differentiation of mature cell types of all three germ layers from pluripotent cells. In this Meeting Review, we summarise some of the exciting data presented at the workshop and draw together the main themes that emerged.


Assuntos
Desenvolvimento Embrionário , Células-Tronco/citologia , Animais , Doença , Implantação do Embrião , Saúde , Hematopoese , Humanos , Camundongos , Organogênese , Células-Tronco Pluripotentes/citologia
6.
Development ; 142(18): 3178-87, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26395144

RESUMO

A key aspect of nervous system development, including that of the cerebral cortex, is the formation of higher-order neural networks. Developing neural networks undergo several phases with distinct activity patterns in vivo, which are thought to prune and fine-tune network connectivity. We report here that human pluripotent stem cell (hPSC)-derived cerebral cortex neurons form large-scale networks that reflect those found in the developing cerebral cortex in vivo. Synchronised oscillatory networks develop in a highly stereotyped pattern over several weeks in culture. An initial phase of increasing frequency of oscillations is followed by a phase of decreasing frequency, before giving rise to non-synchronous, ordered activity patterns. hPSC-derived cortical neural networks are excitatory, driven by activation of AMPA- and NMDA-type glutamate receptors, and can undergo NMDA-receptor-mediated plasticity. Investigating single neuron connectivity within PSC-derived cultures, using rabies-based trans-synaptic tracing, we found two broad classes of neuronal connectivity: most neurons have small numbers (<10) of presynaptic inputs, whereas a small set of hub-like neurons have large numbers of synaptic connections (>40). These data demonstrate that the formation of hPSC-derived cortical networks mimics in vivo cortical network development and function, demonstrating the utility of in vitro systems for mechanistic studies of human forebrain neural network biology.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Técnicas In Vitro/métodos , Rede Nervosa/crescimento & desenvolvimento , Células-Tronco Pluripotentes/fisiologia , Córtex Cerebral/citologia , Espinhas Dendríticas/ultraestrutura , Humanos , Processamento de Imagem Assistida por Computador , Microscopia de Fluorescência , Rede Nervosa/citologia , Plasticidade Neuronal/fisiologia , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Célula Única , Gravação em Vídeo
7.
Nat Methods ; 12(6): 519-22, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25915121

RESUMO

The simultaneous sequencing of a single cell's genome and transcriptome offers a powerful means to dissect genetic variation and its effect on gene expression. Here we describe G&T-seq, a method for separating and sequencing genomic DNA and full-length mRNA from single cells. By applying G&T-seq to over 220 single cells from mice and humans, we discovered cellular properties that could not be inferred from DNA or RNA sequencing alone.


Assuntos
DNA/genética , Genômica/métodos , Técnicas de Amplificação de Ácido Nucleico/métodos , RNA Mensageiro/genética , Animais , Linhagem Celular Tumoral , Humanos , Camundongos
8.
Hum Mol Genet ; 24(18): 5260-9, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26136155

RESUMO

The alternative splicing of the tau gene, MAPT, generates six protein isoforms in the adult human central nervous system (CNS). Tau splicing is developmentally regulated and dysregulated in disease. Mutations in MAPT that alter tau splicing cause frontotemporal dementia (FTD) with tau pathology, providing evidence for a causal link between altered tau splicing and disease. The use of induced pluripotent stem cell (iPSC)-derived neurons has revolutionized the way we model neurological disease in vitro. However, as most tau mutations are located within or around the alternatively spliced exon 10, it is important that iPSC-neurons splice tau appropriately in order to be used as disease models. To address this issue, we analyzed the expression and splicing of tau in iPSC-derived cortical neurons from control patients and FTD patients with the 10 + 16 intronic mutation in MAPT. We show that control neurons only express the fetal tau isoform (0N3R), even at extended time points of 100 days in vitro. Neurons from FTD patients with the 10 + 16 mutation in MAPT express both 0N3R and 0N4R tau isoforms, demonstrating that this mutation overrides the developmental regulation of exon 10 inclusion in our in vitro model. Further, at extended time points of 365 days in vitro, we observe a switch in tau splicing to include six tau isoforms as seen in the adult human CNS. Our results demonstrate the importance of neuronal maturity for use in in vitro modeling and provide a system that will be important for understanding the functional consequences of altered tau splicing.


Assuntos
Processamento Alternativo , Demência Frontotemporal/genética , Mutação , Neurônios/metabolismo , Células-Tronco/metabolismo , Proteínas tau/genética , Biomarcadores , Diferenciação Celular , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Demência Frontotemporal/metabolismo , Haplótipos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactente , Recém-Nascido , Íntrons , Neurônios/citologia , Fosforilação , Sítios de Splice de RNA , Células-Tronco/citologia
9.
Development ; 141(11): 2216-24, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24821983

RESUMO

Generation of neurons from patient fibroblasts using a combination of developmentally defined transcription factors has great potential in disease modelling, as well as ultimately for use in regeneration and repair. However, generation of physiologically mature neurons in vitro remains problematic. Here we demonstrate the cell-cycle-dependent phosphorylation of a key reprogramming transcription factor, Ascl1, on multiple serine-proline sites. This multisite phosphorylation is a crucial regulator of the ability of Ascl1 to drive neuronal differentiation and maturation in vivo in the developing embryo; a phosphomutant form of Ascl1 shows substantially enhanced neuronal induction activity in Xenopus embryos. Mechanistically, we see that this un(der)phosphorylated Ascl1 is resistant to inhibition by both cyclin-dependent kinase activity and Notch signalling, both of which normally limit its neurogenic potential. Ascl1 is a central component of reprogramming transcription factor cocktails to generate neurons from human fibroblasts; the use of phosphomutant Ascl1 in place of the wild-type protein significantly promotes neuronal maturity after human fibroblast reprogramming in vitro. These results demonstrate that cell-cycle-dependent post-translational modification of proneural proteins directly regulates neuronal differentiation in vivo during development, and that this regulatory mechanism can be harnessed to promote maturation of neurons obtained by transdifferentiation of human cells in vitro.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas de Xenopus/metabolismo , Animais , Técnicas de Cultura de Células , Ciclo Celular , Linhagem Celular , Transdiferenciação Celular/fisiologia , Fibroblastos/metabolismo , Células HEK293 , Humanos , Neurogênese , Neurônios/metabolismo , Fosforilação , Prolina/metabolismo , Processamento de Proteína Pós-Traducional , Receptores Notch/metabolismo , Serina/metabolismo , Transdução de Sinais , Xenopus laevis
10.
Hum Mol Genet ; 23(R1): R35-9, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24939911

RESUMO

Positive predictions were made in the aftermath of the development of induced pluripotent stem cell technology for the use of patient-specific iPSCs to model neurological diseases, including dementia. Here, we review the current state of the field and explore how close we are to the goal of in vitro models that capture all aspects of the cell and molecular biology of dementia.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Humanos , Modelos Genéticos , Modelos Neurológicos , Neurônios/citologia , Prosencéfalo/citologia , Transplante de Células-Tronco
11.
Proc Natl Acad Sci U S A ; 110(8): E716-25, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382203

RESUMO

During cerebral cortex development, a series of projection neuron types is generated in a fixed temporal order. In Drosophila neuroblasts, the transcription factor hunchback encodes first-born identity within neural lineages. One of its mammalian homologs, Ikaros, was recently reported to play an equivalent role in retinal progenitor cells, raising the question as to whether Ikaros/Hunchback proteins could be general factors regulating the development of early-born fates throughout the nervous system. Ikaros is also expressed in progenitor cells of the mouse cerebral cortex, and this expression is highest during the early stages of neurogenesis and thereafter decreases over time. Transgenic mice with sustained Ikaros expression in cortical progenitor cells and neurons have developmental defects, including displaced progenitor cells within the cortical plate, increased early neural differentiation, and disrupted cortical lamination. Sustained Ikaros expression results in a prolonged period of generation of deep layer neurons into the stages when, normally, only late-born upper layer neurons are generated, as well as a delayed production of late-born neurons. Consequently, more early-born and fewer late-born neurons are present in the cortex of these mice at birth. This phenotype was observed in all parts of the cortex, including those with minimal structural defects, demonstrating that it is not secondary to abnormalities in cortical morphogenesis. These data suggest that Ikaros plays a similar role in regulating early temporal fates in the mammalian cerebral cortex as Ikaros/Hunchback proteins do in the Drosophila nerve cord.


Assuntos
Córtex Cerebral/citologia , Fator de Transcrição Ikaros/fisiologia , Neurônios/citologia , Animais , Animais Geneticamente Modificados , Linhagem da Célula , Eletroporação , Hibridização In Situ , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley
12.
Alzheimers Dement ; 10(5 Suppl): S374-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24331439

RESUMO

BACKGROUND: The first-line treatment in chronic myeloid leukemia (CML), imatinib, has been shown to decrease the production of amyloid-ß (Aß) in vitro and in animal studies. However, whether imatinib has this effect in humans is not known. METHODS: Plasma levels of Aß42 were analyzed in sequential samples from CML patients treated with imatinib (n=51). The effect of imatinib on Aß production was also investigated in human embryonic kidney 293 (HEK293) cells overexpressing the amyloid precursor protein (APP) with the Swedish mutation, in mouse primary cortical neurons and in human Down syndrome embryonic stem-cell-derived cortical neurons. RESULTS: Twelve months of imatinib treatment did not lower plasma Aß42 levels in CML patients, and imatinib treatment did not lead to less Aß42 production in any of the in vitro models whereas ß- and γ-secretase inhibitors did. CONCLUSION: These data question the previously described role of imatinib in inhibiting amyloidogenic APP processing and as a drug candidate for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Benzamidas/farmacologia , Fragmentos de Peptídeos/metabolismo , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Idoso , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Benzamidas/uso terapêutico , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Síndrome de Down/tratamento farmacológico , Síndrome de Down/fisiopatologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Feminino , Células HEK293 , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/sangue , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico
14.
Proc Natl Acad Sci U S A ; 107(36): 15957-62, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20798045

RESUMO

Multipotent progenitor cells of the cerebral cortex balance self-renewal and differentiation to produce complex neural lineages in a fixed temporal order in a cell-autonomous manner. We studied the role of the polycomb epigenetic system, a chromatin-based repressive mechanism, in controlling cortical progenitor cell self-renewal and differentiation. We found that the histone methyltransferase of polycomb repressive complex 2 (PCR2), enhancer of Zeste homolog 2 (Ezh2), is essential for controlling the rate at which development progresses within cortical progenitor cell lineages. Loss of function of Ezh2 removes the repressive mark of trimethylated histone H3 at lysine 27 (H3K27me3) in cortical progenitor cells and also prevents its establishment in postmitotic neurons. Removal of this repressive chromatin modification results in marked up-regulation in gene expression, the consequence of which is a shift in the balance between self-renewal and differentiation toward differentiation, both directly to neurons and indirectly via basal progenitor cell genesis. Although the temporal order of neurogenesis and gliogenesis are broadly conserved under these conditions, the timing of neurogenesis, the relative numbers of different cell types, and the switch to gliogenesis are all altered, narrowing the neurogenic period for progenitor cells and reducing their neuronal output. As a consequence, the timing of cortical development is altered significantly after loss of PRC2 function.


Assuntos
Diferenciação Celular/fisiologia , Córtex Cerebral/citologia , Histona-Lisina N-Metiltransferase/fisiologia , Concentração de Íons de Hidrogênio , Neurogênese , Animais , Regulação para Baixo , Proteína Potenciadora do Homólogo 2 de Zeste , Histonas/química , Histonas/metabolismo , Camundongos , Complexo Repressor Polycomb 2 , Regulação para Cima
15.
Nat Genet ; 34(1): 53-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692551

RESUMO

Retinal progenitor cells regulate their proliferation during development so that the correct number of each cell type is made at the appropriate time. We found that the homeodomain protein Prox1 regulates the exit of progenitor cells from the cell cycle in the embryonic mouse retina. Cells lacking Prox1 are less likely to stop dividing, and ectopic expression of Prox1 forces progenitor cells to exit the cell cycle. During retinogenesis, Prox1 can be detected in differentiating horizontal, bipolar and AII amacrine cells. Horizontal cells are absent in retinae of Prox1-/- mice and misexpression of Prox1 in postnatal progenitor cells promotes horizontal-cell formation. Thus, Prox1 activity is both necessary and sufficient for progenitor-cell proliferation and cell-fate determination in the vertebrate retina.


Assuntos
Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Retina/citologia , Retina/crescimento & desenvolvimento , Células-Tronco/citologia , Animais , Ciclo Celular , Divisão Celular/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Retina/embriologia , Proteínas Supressoras de Tumor
16.
Biol Psychiatry ; 94(10): 780-791, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37001843

RESUMO

BACKGROUND: Loss-of-function mutations in the contactin-associated protein-like 2 (CNTNAP2) gene are causal for neurodevelopmental disorders, including autism, schizophrenia, epilepsy, and intellectual disability. CNTNAP2 encodes CASPR2, a single-pass transmembrane protein that belongs to the neurexin family of cell adhesion molecules. These proteins have a variety of functions in developing neurons, including connecting presynaptic and postsynaptic neurons, and mediating signaling across the synapse. METHODS: To study the effect of loss of CNTNAP2 function on human cerebral cortex development, and how this contributes to the pathogenesis of neurodevelopmental disorders, we generated human induced pluripotent stem cells from one neurotypical control donor null for full-length CNTNAP2, modeling cortical development from neurogenesis through to neural network formation in vitro. RESULTS: CNTNAP2 is particularly highly expressed in the first two populations of early-born excitatory cortical neurons, and loss of CNTNAP2 shifted the relative proportions of these two neuronal types. Live imaging of excitatory neuronal growth showed that loss of CNTNAP2 reduced neurite branching and overall neuronal complexity. At the network level, developing cortical excitatory networks null for CNTNAP2 had complex changes in activity compared with isogenic controls: an initial period of relatively reduced activity compared with isogenic controls, followed by a lengthy period of hyperexcitability, and then a further switch to reduced activity. CONCLUSIONS: Complete loss of CNTNAP2 contributes to the pathogenesis of neurodevelopmental disorders through complex changes in several aspects of human cerebral cortex excitatory neuron development that culminate in aberrant neural network formation and function.


Assuntos
Córtex Cerebral , Proteínas de Membrana , Rede Nervosa , Proteínas do Tecido Nervoso , Transtornos do Neurodesenvolvimento , Neurônios , Humanos , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Córtex Cerebral/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação com Perda de Função/genética , Mutação com Perda de Função/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Rede Nervosa/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Esquizofrenia/genética , Esquizofrenia/metabolismo
17.
J Exp Med ; 203(11): 2519-27, 2006 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-17060477

RESUMO

Micro RNAs (miRNAs) regulate gene expression at the posttranscriptional level. Here we show that regulatory T (T reg) cells have a miRNA profile distinct from conventional CD4 T cells. A partial T reg cell-like miRNA profile is conferred by the enforced expression of Foxp3 and, surprisingly, by the activation of conventional CD4 T cells. Depleting miRNAs by eliminating Dicer, the RNAse III enzyme that generates functional miRNAs, reduces T reg cell numbers and results in immune pathology. Dicer facilitates, in a cell-autonomous fashion, the development of T reg cells in the thymus and the efficient induction of Foxp3 by transforming growth factor beta. These results suggest that T reg cell development involves Dicer-generated RNAs.


Assuntos
Ribonuclease III/fisiologia , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Fatores de Transcrição Forkhead/biossíntese , Camundongos , MicroRNAs/biossíntese , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Timo/citologia , Timo/imunologia , Fator de Crescimento Transformador beta/fisiologia
18.
PLoS Genet ; 5(6): e1000511, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19521500

RESUMO

Neural stem cell self-renewal, neurogenesis, and cell fate determination are processes that control the generation of specific classes of neurons at the correct place and time. The transcription factor Pax6 is essential for neural stem cell proliferation, multipotency, and neurogenesis in many regions of the central nervous system, including the cerebral cortex. We used Pax6 as an entry point to define the cellular networks controlling neural stem cell self-renewal and neurogenesis in stem cells of the developing mouse cerebral cortex. We identified the genomic binding locations of Pax6 in neocortical stem cells during normal development and ascertained the functional significance of genes that we found to be regulated by Pax6, finding that Pax6 positively and directly regulates cohorts of genes that promote neural stem cell self-renewal, basal progenitor cell genesis, and neurogenesis. Notably, we defined a core network regulating neocortical stem cell decision-making in which Pax6 interacts with three other regulators of neurogenesis, Neurog2, Ascl1, and Hes1. Analyses of the biological function of Pax6 in neural stem cells through phenotypic analyses of Pax6 gain- and loss-of-function mutant cortices demonstrated that the Pax6-regulated networks operating in neural stem cells are highly dosage sensitive. Increasing Pax6 levels drives the system towards neurogenesis and basal progenitor cell genesis by increasing expression of a cohort of basal progenitor cell determinants, including the key transcription factor Eomes/Tbr2, and thus towards neurogenesis at the expense of self-renewal. Removing Pax6 reduces cortical stem cell self-renewal by decreasing expression of key cell cycle regulators, resulting in excess early neurogenesis. We find that the relative levels of Pax6, Hes1, and Neurog2 are key determinants of a dynamic network that controls whether neural stem cells self-renew, generate cortical neurons, or generate basal progenitor cells, a mechanism that has marked parallels with the transcriptional control of embryonic stem cell self-renewal.


Assuntos
Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Neocórtex/metabolismo , Neurogênese , Neurônios/citologia , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Animais , Células Cultivadas , Proteínas do Olho/genética , Feminino , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/citologia , Neocórtex/embriologia , Neurônios/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/genética , Células-Tronco/metabolismo
19.
Front Mol Neurosci ; 15: 1017144, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36340692

RESUMO

The contactin-associated protein-like 2 (CNTNAP2) gene is associated with multiple neurodevelopmental disorders, including autism spectrum disorder (ASD), intellectual disability (ID), and specific language impairment (SLI). Experimental work has shown that CNTNAP2 is important for neuronal development and synapse formation. There is also accumulating evidence for the differential use of CNTNAP2 in the human cerebral cortex compared with other primates. Here, we review the current literature on CNTNAP2, including what is known about its expression, disease associations, and molecular/cellular functions. We also review the evidence for its role in human brain evolution, such as the presence of eight human accelerated regions (HARs) within the introns of the gene. While progress has been made in understanding the function(s) of CNTNAP2, more work is needed to clarify the precise mechanisms through which CNTNAP2 acts. Such information will be crucial for developing effective treatments for CNTNAP2 patients. It may also shed light on the longstanding question of what makes us human.

20.
Autophagy ; 17(11): 3882-3883, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34429033

RESUMO

Abnormalities of the neuronal endolysosome and macroautophagy/autophagy system are an early and prominent feature of Alzheimer disease (AD). SORL1 is notable as a gene in which mutations are causal for a rare, autosomal dominant form of AD, and also variants that increase the risk of developing the common form of late-onset AD. In our recent study, we used patient-derived stem cells and CRISPR engineering to study the effects of SORL1 mutations on the endolysosome and autophagy system in human forebrain neurons. SORL1 mutations causal for monogenic AD are typically truncating mutations, and we found, using stem cells generated from an individual with dementia due to a heterozygous SORL1 truncation mutation, that this class of mutation results in SORL1 haploinsufficiency. Reducing SORL1 protein by half results in disrupted endosomal trafficking in patient-derived neurons, which we confirmed by studying the endolysosomal system in isogenic CRISPR-engineered SORL1 heterozygous null neurons. We also found that SORL1 homozygous null neurons develop more severe phenotypes, with endosome abnormalities, lysosome dysfunction and defects in the degradative phase of autophagy. Endolysosome and autophagy defects in SORL1 mutant neurons are dependent on APP, a key AD gene, as they are rescued by extracellular antisense oligonucleotides that reduce APP protein.


Assuntos
Doença de Alzheimer/patologia , Autofagia , Lisossomos/patologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Edição de Genes , Humanos , Proteínas Relacionadas a Receptor de LDL/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Neurônios/metabolismo , Neurônios/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA