Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Stem Cell ; 29(1): 52-69.e8, 2022 01 06.
Article de Anglais | MEDLINE | ID: mdl-34624206

RÉSUMÉ

The human forebrain has expanded in size and complexity compared to chimpanzees despite limited changes in protein-coding genes, suggesting that gene expression regulation is an important driver of brain evolution. Here, we identify a KRAB-ZFP transcription factor, ZNF558, that is expressed in human but not chimpanzee forebrain neural progenitor cells. ZNF558 evolved as a suppressor of LINE-1 transposons but has been co-opted to regulate a single target, the mitophagy gene SPATA18. ZNF558 plays a role in mitochondrial homeostasis, and loss-of-function experiments in cerebral organoids suggests that ZNF558 influences developmental timing during early human brain development. Expression of ZNF558 is controlled by the size of a variable number tandem repeat that is longer in chimpanzees compared to humans, and variable in the human population. Thus, this work provides mechanistic insight into how a cis-acting structural variation establishes a regulatory network that affects human brain evolution.


Sujet(s)
Réseaux de régulation génique , Organoïdes , Encéphale/métabolisme , Protéines de liaison à l'ADN , Régulation de l'expression des gènes , Humains , Organoïdes/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
2.
Autophagy ; 17(6): 1316-1329, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-32374203

RÉSUMÉ

Activation of macroautophagy/autophagy, a key mechanism involved in the degradation and removal of aggregated proteins, can successfully reverse Huntington disease phenotypes in various model systems. How neuronal autophagy impairments need to be considered in Huntington disease progression to achieve a therapeutic effect is currently not known. In this study, we used a mouse model of HTT (huntingtin) protein aggregation to investigate how different methods and timing of autophagy activation influence the efficacy of autophagy-activating treatment in vivo. We found that overexpression of human TFEB, a master regulator of autophagy, did not decrease mutant HTT aggregation. On the other hand, Becn1 overexpression, an autophagic regulator that plays a key role in autophagosome formation, partially cleared mutant HTT aggregates and restored neuronal pathology, but only when administered early in the disease progression. When Becn1 was administered at a later stage, when prominent mutant HTT accumulation and autophagy impairments have occurred, Becn1 overexpression did not rescue the mutant HTT-associated phenotypes. Together, these results demonstrate that the targets used to activate autophagy, as well as the timing of autophagy activation, are crucial for achieving efficient therapeutic effects.Abbreviations: AAV: adeno-associated viral vectors; ACTB: actin beta; BECN1: beclin 1, autophagy related; DAPI: 4',6-diamidino-2-phenylindole; GO: gene ontology; HD: Huntington disease; HTT: huntingtin; ICQ: Li's intensity correlation quotient; IHC: immunohistochemistry; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; mHTT: mutant huntingtin; PCA: principal component analysis; PPP1R1B/DARPP-32: protein phosphatase 1 regulatory inhibitor subunit 1B; SQSTM1: sequestosome 1; TFEB: transcription factor EB; WB: western blot; WT: wild-type.


Sujet(s)
Autophagosomes/métabolisme , Autophagie/physiologie , Maladie de Huntington/métabolisme , Maladie de Huntington/thérapie , Animaux , Bécline-1/métabolisme , Modèles animaux de maladie humaine , Femelle , Souris de lignée C57BL , Facteurs temps
3.
Heliyon ; 6(1): e03067, 2020 Jan.
Article de Anglais | MEDLINE | ID: mdl-31909251

RÉSUMÉ

Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) can be differentiated into many different cell types of the central nervous system. One challenge when using pluripotent stem cells is to develop robust and efficient differentiation protocols that result in homogenous cultures of the desired cell type. Here, we have utilized the SMAD-inhibitors SB431542 and Noggin in a fully defined monolayer culture model to differentiate human pluripotent cells into homogenous forebrain neural progenitors. Temporal fate analysis revealed that this protocol results in forebrain-patterned neural progenitor cells that start to express early neuronal markers after two weeks of differentiation, allowing for the analysis of gene expression changes during neurogenesis. Using this system, we were able to identify many previously uncharacterized long intergenic non-coding RNAs that display dynamic expression during human forebrain neurogenesis.

4.
PLoS Genet ; 15(3): e1008036, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30865625

RÉSUMÉ

Transposable elements (TEs) are dynamically expressed at high levels in multiple human tissues, but the function of TE-derived transcripts remains largely unknown. In this study, we identify numerous TE-derived microRNAs (miRNAs) by conducting Argonaute2 RNA immunoprecipitation followed by small RNA sequencing (AGO2 RIP-seq) on human brain tissue. Many of these miRNAs originated from LINE-2 (L2) elements, which entered the human genome around 100-300 million years ago. L2-miRNAs derived from the 3' end of the L2 consensus sequence and thus shared very similar sequences, indicating that L2-miRNAs could target transcripts with L2s in their 3'UTR. In line with this, many protein-coding genes carried fragments of L2-derived sequences in their 3'UTR: these sequences served as target sites for L2-miRNAs. L2-miRNAs and their targets were generally ubiquitously expressed at low levels in multiple human tissues, suggesting a role for this network in buffering transcriptional levels of housekeeping genes. In addition, we also found evidence that this network is perturbed in glioblastoma. In summary, our findings uncover a TE-based post-transcriptional network that shapes transcriptional regulation in human cells.


Sujet(s)
Éléments transposables d'ADN , Éléments LINE , microARN/génétique , Régions 3' non traduites , Animaux , Protéines Argonaute/génétique , Protéines Argonaute/métabolisme , Séquence nucléotidique , Encéphale/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Régulation de l'expression des gènes , Réseaux de régulation génique , Génome humain , Glioblastome/génétique , Glioblastome/métabolisme , Humains , Souris , microARN/métabolisme
5.
Brain Res ; 1705: 43-47, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-29522722

RÉSUMÉ

TRIM28 is an epigenetic co-repressor protein that mediates transcriptional silencing. TRIM28 participates, together with the large family of Kruppel-associated box domain zinc finger proteins (KRAB-ZFP) transcription factors, in the repression of transposable elements (TE). Recent advances indicate that TRIM28-based repression of TEs occurs in the mammalian brain and may provide beneficial effects through the regulation of transcriptional networks. Here, we provide an overview of TRIM28-related functions, highlighting the role of controlling TEs in neural progenitor cells and discuss how this mechanism may have contributed to the evolution of the complex human brain. Finally, we outline future considerations for the field.


Sujet(s)
Encéphale/métabolisme , Éléments transposables d'ADN/génétique , Protéine-28 à motif tripartite/génétique , Animaux , Épigenèse génétique/génétique , Régulation de l'expression des gènes/génétique , Réseaux de régulation génique , Humains , Protéines de répression/métabolisme , Facteurs de transcription/métabolisme , Protéine-28 à motif tripartite/métabolisme , Protéine-28 à motif tripartite/physiologie
6.
Cell Rep ; 24(6): 1397-1406, 2018 08 07.
Article de Anglais | MEDLINE | ID: mdl-30089251

RÉSUMÉ

Many neurodegenerative diseases are characterized by the presence of intracellular protein aggregates, resulting in alterations in autophagy. However, the consequences of impaired autophagy for neuronal function remain poorly understood. In this study, we used cell culture and mouse models of huntingtin protein aggregation as well as post-mortem material from patients with Huntington's disease to demonstrate that Argonaute-2 (AGO2) accumulates in the presence of neuronal protein aggregates and that this is due to impaired autophagy. Accumulation of AGO2, a key factor of the RNA-induced silencing complex that executes microRNA functions, results in global alterations of microRNA levels and activity. Together, these results demonstrate that impaired autophagy found in neurodegenerative diseases not only influences protein aggregation but also directly contributes to global alterations of intracellular post-transcriptional networks.


Sujet(s)
Protéines Argonaute/génétique , Autophagie/physiologie , Maladie de Huntington/génétique , microARN/métabolisme , Humains
7.
EMBO Mol Med ; 9(8): 1117-1131, 2017 08.
Article de Anglais | MEDLINE | ID: mdl-28646119

RÉSUMÉ

Direct conversion of human fibroblasts into mature and functional neurons, termed induced neurons (iNs), was achieved for the first time 6 years ago. This technology offers a promising shortcut for obtaining patient- and disease-specific neurons for disease modeling, drug screening, and other biomedical applications. However, fibroblasts from adult donors do not reprogram as easily as fetal donors, and no current reprogramming approach is sufficiently efficient to allow the use of this technology using patient-derived material for large-scale applications. Here, we investigate the difference in reprogramming requirements between fetal and adult human fibroblasts and identify REST as a major reprogramming barrier in adult fibroblasts. Via functional experiments where we overexpress and knockdown the REST-controlled neuron-specific microRNAs miR-9 and miR-124, we show that the effect of REST inhibition is only partially mediated via microRNA up-regulation. Transcriptional analysis confirmed that REST knockdown activates an overlapping subset of neuronal genes as microRNA overexpression and also a distinct set of neuronal genes that are not activated via microRNA overexpression. Based on this, we developed an optimized one-step method to efficiently reprogram dermal fibroblasts from elderly individuals using a single-vector system and demonstrate that it is possible to obtain iNs of high yield and purity from aged individuals with a range of familial and sporadic neurodegenerative disorders including Parkinson's, Huntington's, as well as Alzheimer's disease.


Sujet(s)
Transdifférenciation cellulaire , Fibroblastes/physiologie , Techniques de knock-down de gènes , Neurones/physiologie , Protéines de répression/biosynthèse , Adulte , Techniques cytologiques/méthodes , Analyse de profil d'expression de gènes , Humains , microARN/analyse , Protéines de répression/génétique
8.
EMBO J ; 36(10): 1379-1391, 2017 05 15.
Article de Anglais | MEDLINE | ID: mdl-28336683

RÉSUMÉ

During adult neurogenesis, newly formed olfactory bulb (OB) interneurons migrate radially to integrate into specific layers of the OB Despite the importance of this process, the intracellular mechanisms that regulate radial migration remain poorly understood. Here, we find that microRNA (miRNA) let-7 regulates radial migration by modulating autophagy in new-born neurons. Using Argonaute2 immunoprecipitation, we performed global profiling of miRNAs in adult-born OB neurons and identified let-7 as a highly abundant miRNA family. Knockdown of let-7 in migrating neuroblasts prevented radial migration and led to an immature morphology of newly formed interneurons. This phenotype was accompanied by a decrease in autophagic activity. Overexpression of Beclin-1 or TFEB in new-born neurons lacking let-7 resulted in re-activation of autophagy and restored radial migration. Thus, these results reveal a miRNA-dependent link between autophagy and adult neurogenesis with implications for neurodegenerative diseases where these processes are impaired.


Sujet(s)
Autophagie , Mouvement cellulaire , microARN/métabolisme , Neurones/physiologie , Animaux , Techniques de knock-down de gènes , Immunoprécipitation , Souris de lignée C57BL , Souris knockout , microARN/génétique
9.
Cell Rep ; 18(1): 1-11, 2017 01 03.
Article de Anglais | MEDLINE | ID: mdl-28052240

RÉSUMÉ

Endogenous retroviruses (ERVs), which make up 8% of the human genome, have been proposed to participate in the control of gene regulatory networks. In this study, we find a region- and developmental stage-specific expression pattern of ERVs in the developing human brain, which is linked to a transcriptional network based on ERVs. We demonstrate that almost 10,000, primarily primate-specific, ERVs act as docking platforms for the co-repressor protein TRIM28 in human neural progenitor cells, which results in the establishment of local heterochromatin. Thereby, TRIM28 represses ERVs and consequently regulates the expression of neighboring genes. These results uncover a gene regulatory network based on ERVs that participates in control of gene expression of protein-coding transcripts important for brain development.


Sujet(s)
Rétrovirus endogènes/génétique , Réseaux de régulation génique , Cellules souches neurales/métabolisme , Cellules souches neurales/virologie , Protéine-28 à motif tripartite/métabolisme , Encéphale/embryologie , Encéphale/virologie , Régulation de l'expression des gènes au cours du développement , Techniques de knock-down de gènes , Humains , Cadres ouverts de lecture/génétique , Liaison aux protéines , Activation de la transcription/génétique , Régulation positive/génétique
10.
Sci Rep ; 6: 31701, 2016 08 23.
Article de Anglais | MEDLINE | ID: mdl-27550741

RÉSUMÉ

Motor symptoms in Parkinson's disease are attributed to degeneration of midbrain dopaminergic neurons (DNs). Heterozygosity for Engrailed-1 (En1), one of the key factors for programming and maintenance of DNs, results in a parkinsonian phenotype featuring progressive degeneration of DNs in substantia nigra pars compacta (SNpc), decreased striatal dopamine levels and swellings of nigro-striatal axons in the SwissOF1-En1+/- mouse strain. In contrast, C57Bl/6-En1+/- mice do not display this neurodegenerative phenotype, suggesting that susceptibility to En1 heterozygosity is genetically regulated. Our goal was to identify quantitative trait loci (QTLs) that regulate the susceptibility to PD-like neurodegenerative changes in response to loss of one En1 allele. We intercrossed SwissOF1-En1+/- and C57Bl/6 mice to obtain F2 mice with mixed genomes and analyzed number of DNs in SNpc and striatal axonal swellings in 120 F2-En1+/- 17 week-old male mice. Linkage analyses revealed 8 QTLs linked to number of DNs (p = 2.4e-09, variance explained = 74%), 7 QTLs linked to load of axonal swellings (p = 1.7e-12, variance explained = 80%) and 8 QTLs linked to size of axonal swellings (p = 7.0e-11, variance explained = 74%). These loci should be of prime interest for studies of susceptibility to Parkinson's disease-like damage in rodent disease models and considered in clinical association studies in PD.


Sujet(s)
Modèles animaux de maladie humaine , Protéines à homéodomaine/génétique , Maladie de Parkinson/génétique , Locus de caractère quantitatif/génétique , Animaux , Neurones dopaminergiques/métabolisme , Femelle , Prédisposition génétique à une maladie/génétique , Hétérozygote , Mâle , Souris de lignée C57BL , Souris knockout , Maladies neurodégénératives/génétique , Neuropathologie , Substantia nigra/métabolisme , Substantia nigra/anatomopathologie
11.
Sci Rep ; 6: 19879, 2016 Jan 27.
Article de Anglais | MEDLINE | ID: mdl-26813637

RÉSUMÉ

MicroRNAs (miRNA) are small, non-coding RNAs mediating post-transcriptional regulation of gene expression. miRNAs have recently been implicated in hippocampus-dependent functions such as learning and memory, although the roles of individual miRNAs in these processes remain largely unknown. Here, we achieved stable inhibition using AAV-delivered miRNA sponges of individual, highly expressed and brain-enriched miRNAs; miR-124, miR-9 and miR-34, in hippocampal neurons. Molecular and cognitive studies revealed a role for miR-124 in learning and memory. Inhibition of miR-124 resulted in an enhanced spatial learning and working memory capacity, potentially through altered levels of genes linked to synaptic plasticity and neuronal transmission. In contrast, inhibition of miR-9 or miR-34 led to a decreased capacity of spatial learning and of reference memory, respectively. On a molecular level, miR-9 inhibition resulted in altered expression of genes related to cell adhesion, endocytosis and cell death, while miR-34 inhibition caused transcriptome changes linked to neuroactive ligand-receptor transduction and cell communication. In summary, this study establishes distinct roles for individual miRNAs in hippocampal function.


Sujet(s)
Cognition , microARN/génétique , Cellules pyramidales/métabolisme , Transcriptome , Animaux , Cellules cultivées , Dependovirus/génétique , Expression des gènes , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Ordre des gènes , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique , Hippocampe/cytologie , Hippocampe/métabolisme , Mâle , Souris , Souris transgéniques , Interférence par ARN , Transduction du signal
12.
Mol Cancer ; 13: 215, 2014 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-25216995

RÉSUMÉ

BACKGROUND: The DEK gene is highly expressed in a wide range of cancer cells, and a recurrent translocation partner in acute myeloid leukemia. While DEK has been identified as one of the most abundant proteins in human chromatin, its function and binding properties are not fully understood. METHODS: We performed ChIP-seq analysis in the myeloid cell line U937 and coupled it with epigenetic and gene expression analysis to explore the genome-wide binding pattern of DEK and its role in gene regulation. RESULTS: We show that DEK preferentially binds to open chromatin, with a low degree of DNA methylation and scarce in the heterochromatin marker H3K9me(3) but rich in the euchromatin marks H3K4me(2/3), H3K27ac and H3K9ac. More specifically, DEK binding is predominantly located at the transcription start sites of highly transcribed genes and a comparative analysis with previously established transcription factor binding patterns shows a similarity with that of RNA polymerase II. Further bioinformatic analysis demonstrates that DEK mainly binds to genes that are ubiquitously expressed across tissues. The functional significance of DEK binding was demonstrated by knockdown of DEK by shRNA, resulting in both significant upregulation and downregulation of DEK-bound genes. CONCLUSIONS: We find that DEK binds to transcription start sites with a dual role in activation and repression of highly and ubiquitously expressed genes.


Sujet(s)
Chromatine/génétique , Protéines chromosomiques nonhistones/génétique , Protéines chromosomiques nonhistones/métabolisme , Régulation de l'expression des gènes , Cellules myéloïdes/métabolisme , Protéines oncogènes/génétique , Protéines oncogènes/métabolisme , Transcription génétique , Sites de fixation , Cellules cultivées , Chromatine/métabolisme , Méthylation de l'ADN , Épigenèse génétique , Analyse de profil d'expression de gènes , Génome humain , Histone/métabolisme , Humains , Protéines liant le poly-adp-ribose , Régions promotrices (génétique) , RNA polymerase II/métabolisme , Analyse de séquence d'ADN
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...