Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain ; 146(8): 3528-3541, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36732302

RESUMO

Biallelic loss-of-function variants in SMPD4 cause a rare and severe neurodevelopmental disorder with progressive congenital microcephaly and early death. SMPD4 encodes a sphingomyelinase that hydrolyses sphingomyelin into ceramide at neutral pH and can thereby affect membrane lipid homeostasis. SMPD4 localizes to the membranes of the endoplasmic reticulum and nuclear envelope and interacts with nuclear pore complexes (NPC). We refine the clinical phenotype of loss-of-function SMPD4 variants by describing five individuals from three unrelated families with longitudinal data due to prolonged survival. All individuals surviving beyond infancy developed insulin-dependent diabetes, besides presenting with a severe neurodevelopmental disorder and microcephaly, making diabetes one of the most frequent age-dependent non-cerebral abnormalities. We studied the function of SMPD4 at the cellular and organ levels. Knock-down of SMPD4 in human neural stem cells causes reduced proliferation rates and prolonged mitosis. Moreover, SMPD4 depletion results in abnormal nuclear envelope breakdown and reassembly during mitosis and decreased post-mitotic NPC insertion. Fibroblasts from affected individuals show deficient SMPD4-specific neutral sphingomyelinase activity, without changing (sub)cellular lipidome fractions, which suggests a local function of SMPD4 on the nuclear envelope. In embryonic mouse brain, knockdown of Smpd4 impairs cortical progenitor proliferation and induces premature differentiation by altering the balance between neurogenic and proliferative progenitor cell divisions. We hypothesize that, in individuals with SMPD4-related disease, nuclear envelope bending, which is needed to insert NPCs in the nuclear envelope, is impaired in the absence of SMPD4 and interferes with cerebral corticogenesis and survival of pancreatic beta cells.


Assuntos
Diabetes Mellitus , Microcefalia , Humanos , Animais , Camundongos , Membrana Nuclear/química , Membrana Nuclear/metabolismo , Microcefalia/genética , Microcefalia/metabolismo , Esfingomielina Fosfodiesterase/análise , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Poro Nuclear/metabolismo , Mitose , Diabetes Mellitus/metabolismo
3.
Nat Commun ; 12(1): 5878, 2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34620845

RESUMO

Microtubule (MT)-based transport is an evolutionary conserved process finely tuned by posttranslational modifications. Among them, α-tubulin acetylation, primarily catalyzed by a vesicular pool of α-tubulin N-acetyltransferase 1 (Atat1), promotes the recruitment and processivity of molecular motors along MT tracks. However, the mechanism that controls Atat1 activity remains poorly understood. Here, we show that ATP-citrate lyase (Acly) is enriched in vesicles and provide Acetyl-Coenzyme-A (Acetyl-CoA) to Atat1. In addition, we showed that Acly expression is reduced upon loss of Elongator activity, further connecting Elongator to Atat1 in a pathway regulating α-tubulin acetylation and MT-dependent transport in projection neurons, across species. Remarkably, comparable defects occur in fibroblasts from Familial Dysautonomia (FD) patients bearing an autosomal recessive mutation in the gene coding for the Elongator subunit ELP1. Our data may thus shine light on the pathophysiological mechanisms underlying FD.


Assuntos
ATP Citrato (pro-S)-Liase/metabolismo , Transporte Axonal/fisiologia , ATP Citrato (pro-S)-Liase/genética , Acetilcoenzima A/metabolismo , Acetilação , Acetiltransferases/genética , Animais , Transporte Axonal/genética , Drosophila melanogaster , Disautonomia Familiar/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Larva , Masculino , Camundongos , Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional , Tubulina (Proteína)/metabolismo
4.
Sci Adv ; 5(12): eaax2705, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31897425

RESUMO

Microtubules are polymerized dimers of α- and ß-tubulin that underlie a broad range of cellular activities. Acetylation of α-tubulin by the acetyltransferase ATAT1 modulates microtubule dynamics and functions in neurons. However, it remains unclear how this enzyme acetylates microtubules over long distances in axons. Here, we show that loss of ATAT1 impairs axonal transport in neurons in vivo, and cell-free motility assays confirm a requirement of α-tubulin acetylation for proper bidirectional vesicular transport. Moreover, we demonstrate that the main cellular pool of ATAT1 is transported at the cytosolic side of neuronal vesicles that are moving along axons. Together, our data suggest that axonal transport of ATAT1-enriched vesicles is the predominant driver of α-tubulin acetylation in axons.


Assuntos
Acetiltransferases/metabolismo , Transporte Axonal/fisiologia , Proteínas dos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Acetilação , Acetiltransferases/genética , Animais , Drosophila melanogaster/metabolismo , Feminino , Células HEK293 , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Larva/fisiologia , Locomoção , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microtúbulos/genética , Neurônios/metabolismo , Tubulina (Proteína)/metabolismo
5.
Cell ; 172(5): 1063-1078.e19, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29474907

RESUMO

Interneurons navigate along multiple tangential paths to settle into appropriate cortical layers. They undergo a saltatory migration paced by intermittent nuclear jumps whose regulation relies on interplay between extracellular cues and genetic-encoded information. It remains unclear how cycles of pause and movement are coordinated at the molecular level. Post-translational modification of proteins contributes to cell migration regulation. The present study uncovers that carboxypeptidase 1, which promotes post-translational protein deglutamylation, controls the pausing of migrating cortical interneurons. Moreover, we demonstrate that pausing during migration attenuates movement simultaneity at the population level, thereby controlling the flow of interneurons invading the cortex. Interfering with the regulation of pausing not only affects the size of the cortical interneuron cohort but also impairs the generation of age-matched projection neurons of the upper layers.


Assuntos
Movimento Celular , Córtex Cerebral/citologia , Interneurônios/citologia , Morfogênese , Actomiosina/metabolismo , Animais , Carboxipeptidases/metabolismo , Ciclo Celular , Fatores Quimiotáticos/metabolismo , Embrião de Mamíferos/citologia , Feminino , Deleção de Genes , Interneurônios/metabolismo , Camundongos , Camundongos Knockout , Quinase de Cadeia Leve de Miosina/metabolismo , Neurogênese , Fenótipo
6.
Sci Rep ; 6: 33377, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27640816

RESUMO

Some mutations of the LRRK2 gene underlie autosomal dominant form of Parkinson's disease (PD). The G2019S is a common mutation that accounts for about 2% of PD cases. To understand the pathophysiology of this mutation and its possible developmental implications, we developed an in vitro assay to model PD with human induced pluripotent stem cells (hiPSCs) reprogrammed from skin fibroblasts of PD patients suffering from the LRKK2 G2019S mutation. We differentiated the hiPSCs into neural stem cells (NSCs) and further into dopaminergic neurons. Here we show that NSCs bearing the mutation tend to differentiate less efficiently into dopaminergic neurons and that the latter exhibit significant branching defects as compared to their controls.


Assuntos
Neurônios Dopaminérgicos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/enzimologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Mutação/genética , Neuritos/metabolismo , Animais , Células Cultivadas , Humanos , Mesencéfalo/citologia , Camundongos , Células-Tronco Neurais/citologia , Doença de Parkinson/genética , Fenótipo
7.
Dev Cell ; 35(5): 553-567, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26651292

RESUMO

The cerebral cortex contains layers of neurons sequentially generated by distinct lineage-related progenitors. At the onset of corticogenesis, the first-born progenitors are apical progenitors (APs), whose asymmetric division gives birth directly to neurons. Later, they switch to indirect neurogenesis by generating intermediate progenitors (IPs), which give rise to projection neurons of all cortical layers. While a direct lineage relationship between APs and IPs has been established, the molecular mechanism that controls their transition remains elusive. Here we show that interfering with codon translation speed triggers ER stress and the unfolded protein response (UPR), further impairing the generation of IPs and leading to microcephaly. Moreover, we demonstrate that a progressive downregulation of UPR in cortical progenitors acts as a physiological signal to amplify IPs and promotes indirect neurogenesis. Thus, our findings reveal a contribution of UPR to cell fate acquisition during mammalian brain development.


Assuntos
Córtex Cerebral/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Histona Acetiltransferases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Resposta a Proteínas não Dobradas , Animais , Linhagem da Célula , Separação Celular , Córtex Cerebral/metabolismo , Códon , Drosophila melanogaster , Células-Tronco Embrionárias/citologia , Deleção de Genes , Genótipo , Histona Acetiltransferases/genética , Humanos , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Fosforilação , Biossíntese de Proteínas , Desnaturação Proteica , Dobramento de Proteína , Transdução de Sinais , Células-Tronco/citologia , Regulação para Cima
8.
PLoS One ; 7(8): e42704, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22880087

RESUMO

Promoter methylation profiles are proposed as potential prognosis and/or diagnosis biomarkers in cervical cancer. Up to now, little is known about the promoter methylation profile and expression pattern of stem cell (SC) markers during tumor development. In this study, we were interested to identify SC genes methylation profiles during cervical carcinogenesis. A genome-wide promoter methylation screening revealed a strong hypermethylation of Undifferentiated cell Transcription Factor 1 (UTF1) promoter in cervical cancer in comparison with normal ectocervix. By direct bisulfite pyrosequencing of DNA isolated from liquid-based cytological samples, we showed that UTF1 promoter methylation increases with lesion severity, the highest level of methylation being found in carcinoma. This hypermethylation was associated with increased UTF1 mRNA and protein expression. By using quantitative RT-PCR and Western Blot, we showed that both UTF1 mRNA and protein are present in epithelial cancer cell lines, even in the absence of its two main described regulators Oct4A and Sox2. Moreover, by immunofluorescence, we confirmed the nuclear localisation of UTF1 in cell lines. Surprisingly, direct bisulfite pyrosequencing revealed that the inhibition of DNA methyltransferase by 5-aza-2'-deoxycytidine was associated with decreased UTF1 gene methylation and expression in two cervical cancer cell lines of the four tested. These findings strongly suggest that UTF1 promoter methylation profile might be a useful biomarker for cervical cancer diagnosis and raise the questions of its role during epithelial carcinogenesis and of the mechanisms regulating its expression.


Assuntos
Transformação Celular Neoplásica/genética , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Transativadores/genética , Neoplasias do Colo do Útero/genética , Azacitidina/farmacologia , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Colo do Útero/efeitos dos fármacos , Colo do Útero/metabolismo , Colo do Útero/patologia , Análise por Conglomerados , Técnicas Citológicas , Metilação de DNA/efeitos dos fármacos , DNA de Neoplasias/isolamento & purificação , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Neoplásicos/genética , Humanos , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Análise de Sequência de DNA , Transativadores/metabolismo , Neoplasias do Colo do Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...