Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
EMBO J ; 40(9): e106423, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33644903

ABSTRACT

Endogenous retroviruses (ERVs) make up a large fraction of mammalian genomes and are thought to contribute to human disease, including brain disorders. In the brain, aberrant activation of ERVs is a potential trigger for an inflammatory response, but mechanistic insight into this phenomenon remains lacking. Using CRISPR/Cas9-based gene disruption of the epigenetic co-repressor protein Trim28, we found a dynamic H3K9me3-dependent regulation of ERVs in proliferating neural progenitor cells (NPCs), but not in adult neurons. In vivo deletion of Trim28 in cortical NPCs during mouse brain development resulted in viable offspring expressing high levels of ERVs in excitatory neurons in the adult brain. Neuronal ERV expression was linked to activated microglia and the presence of ERV-derived proteins in aggregate-like structures. This study demonstrates that brain development is a critical period for the silencing of ERVs and provides causal in vivo evidence demonstrating that transcriptional activation of ERV in neurons results in an inflammatory response.


Subject(s)
Brain/growth & development , Encephalitis/genetics , Endogenous Retroviruses/genetics , Gene Deletion , Tripartite Motif-Containing Protein 28/genetics , Animals , Brain/immunology , Brain/virology , CRISPR-Cas Systems , Cells, Cultured , Encephalitis/immunology , Encephalitis/virology , Endogenous Retroviruses/immunology , Epigenesis, Genetic , Gene Expression Regulation , Histones/metabolism , Mice , Transcriptional Activation
2.
Nat Methods ; 19(1): 90-99, 2022 01.
Article in English | MEDLINE | ID: mdl-34969984

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived organoids provide models to study human organ development. Single-cell transcriptomics enable highly resolved descriptions of cell states within these systems; however, approaches are needed to directly measure lineage relationships. Here we establish iTracer, a lineage recorder that combines reporter barcodes with inducible CRISPR-Cas9 scarring and is compatible with single-cell and spatial transcriptomics. We apply iTracer to explore clonality and lineage dynamics during cerebral organoid development and identify a time window of fate restriction as well as variation in neurogenic dynamics between progenitor neuron families. We also establish long-term four-dimensional light-sheet microscopy for spatial lineage recording in cerebral organoids and confirm regional clonality in the developing neuroepithelium. We incorporate gene perturbation (iTracer-perturb) and assess the effect of mosaic TSC2 mutations on cerebral organoid development. Our data shed light on how lineages and fates are established during cerebral organoid formation. More broadly, our techniques can be adapted in any iPSC-derived culture system to dissect lineage alterations during normal or perturbed development.


Subject(s)
Cerebral Cortex/cytology , Genes, Reporter , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Single-Cell Analysis/methods , CRISPR-Cas Systems , Cell Lineage , Humans , Microscopy/methods , Mutation , Neurons/cytology , Neurons/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, RNA , Tuberous Sclerosis Complex 2 Protein/genetics
3.
PLoS Genet ; 15(3): e1008036, 2019 03.
Article in English | MEDLINE | ID: mdl-30865625

ABSTRACT

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.


Subject(s)
DNA Transposable Elements , Long Interspersed Nucleotide Elements , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Sequence , Brain/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Genome, Human , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , MicroRNAs/metabolism
4.
EMBO J ; 36(10): 1379-1391, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28336683

ABSTRACT

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.


Subject(s)
Autophagy , Cell Movement , MicroRNAs/metabolism , Neurons/physiology , Animals , Gene Knockdown Techniques , Immunoprecipitation , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics
5.
Development ; 141(7): 1580-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24598163

ABSTRACT

New neurons, originating from the subventricular zone, are continuously integrating into neuronal circuitry in the olfactory bulb (OB). Using a transgenic sensor mouse, we found that adult-born OB interneurons express microRNA-125 (miR-125), whereas the pre-existing developmentally generated OB interneurons represent a unique population of cells in the adult brain, without miR-125 activity. Stable inhibition of miR-125 in newborn OB neurons resulted in enhanced dendritic morphogenesis, as well as in increased synaptic activation in response to odour sensory stimuli. These data demonstrate that miR-125 controls functional synaptic integration of adult-born OB interneurons. Our results also suggest that absence of an otherwise broadly expressed miRNA is a novel mechanism with which to achieve neuronal subtype specification.


Subject(s)
Adult Stem Cells/physiology , Embryonic Stem Cells/physiology , Interneurons/physiology , MicroRNAs/physiology , Olfactory Bulb/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation/genetics , Female , Interneurons/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/physiology , Neurogenesis/genetics , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Synapses/genetics
6.
Exp Cell Res ; 321(1): 84-9, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24099990

ABSTRACT

MicroRNAs (miRNAs) are small, non-coding RNAs that negatively regulate gene expression at the post-transcriptional level. In the brain, a large number of miRNAs are expressed and there is a growing body of evidence demonstrating that miRNAs are essential for brain development and neuronal function. Conditional knockout studies of the core components in the miRNA biogenesis pathway, such as Dicer and DGCR8, have demonstrated a crucial role for miRNAs during the development of the central nervous system. Furthermore, mice deleted for specific miRNAs and miRNA-clusters demonstrate diverse functional roles for different miRNAs during the development of different brain structures. miRNAs have been proposed to regulate cellular functions such as differentiation, proliferation and fate-determination of neural progenitors. In this review we summarise the findings from recent studies that highlight the importance of miRNAs in brain development with a focus on the mouse model. We also discuss the technical limitations of current miRNA studies that still limit our understanding of this family of non-coding RNAs and propose the use of novel and refined technologies that are needed in order to fully determine the impact of specific miRNAs in brain development.


Subject(s)
Brain/growth & development , Brain/metabolism , Cell Differentiation , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Animals , Mice
7.
Autophagy ; 17(6): 1316-1329, 2021 06.
Article in English | MEDLINE | ID: mdl-32374203

ABSTRACT

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.


Subject(s)
Autophagosomes/metabolism , Autophagy/physiology , Huntington Disease/metabolism , Huntington Disease/therapy , Animals , Beclin-1/metabolism , Disease Models, Animal , Female , Mice, Inbred C57BL , Time Factors
8.
Nat Commun ; 10(1): 3182, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320637

ABSTRACT

DNA methylation contributes to the maintenance of genomic integrity in somatic cells, in part through the silencing of transposable elements. In this study, we use CRISPR-Cas9 technology to delete DNMT1, the DNA methyltransferase key for DNA methylation maintenance, in human neural progenitor cells (hNPCs). We observe that inactivation of DNMT1 in hNPCs results in viable, proliferating cells despite a global loss of DNA CpG-methylation. DNA demethylation leads to specific transcriptional activation and chromatin remodeling of evolutionarily young, hominoid-specific LINE-1 elements (L1s), while older L1s and other classes of transposable elements remain silent. The activated L1s act as alternative promoters for many protein-coding genes involved in neuronal functions, revealing a hominoid-specific L1-based transcriptional network controlled by DNA methylation that influences neuronal protein-coding genes. Our results provide mechanistic insight into the role of DNA methylation in silencing transposable elements in somatic human cells, as well as further implicating L1s in human brain development and disease.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Demethylation , DNA Methylation/genetics , Long Interspersed Nucleotide Elements/genetics , Neural Stem Cells/cytology , Brain/embryology , CRISPR-Cas Systems/genetics , Chromatin Assembly and Disassembly/genetics , CpG Islands/genetics , Gene Silencing/physiology , Humans , Neural Stem Cells/metabolism , Transcriptional Activation/genetics
9.
Cell Rep ; 28(8): 2064-2079.e11, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433983

ABSTRACT

Identifying cellular programs that drive cancers to be stem-like and treatment resistant is critical to improving outcomes in patients. Here, we demonstrate that constitutive extracellular signal-regulated kinase 1/2 (ERK1/2) activation sustains a stem-like state in glioblastoma (GBM), the most common primary malignant brain tumor. Pharmacological inhibition of ERK1/2 activation restores neurogenesis during murine astrocytoma formation, inducing neuronal differentiation in tumorspheres. Constitutive ERK1/2 activation globally regulates miRNA expression in murine and human GBMs, while neuronal differentiation of GBM tumorspheres following the inhibition of ERK1/2 activation requires the functional expression of miR-124 and the depletion of its target gene SOX9. Overexpression of miR124 depletes SOX9 in vivo and promotes a stem-like-to-neuronal transition, with reduced tumorigenicity and increased radiation sensitivity. Providing a rationale for reports demonstrating miR-124-induced abrogation of GBM aggressiveness, we conclude that reversal of an ERK1/2-miR-124-SOX9 axis induces a neuronal phenotype and that enforcing neuronal differentiation represents a therapeutic strategy to improve outcomes in GBM.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Glioblastoma/pathology , MAP Kinase Signaling System , MicroRNAs/metabolism , Neurons/pathology , SOX9 Transcription Factor/metabolism , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Benzamides/pharmacology , Brain Neoplasms/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Disease Progression , Female , Glioblastoma/genetics , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects
10.
Methods Mol Biol ; 1720: 131-140, 2018.
Article in English | MEDLINE | ID: mdl-29236255

ABSTRACT

microRNAs (miRNA) are small, noncoding RNAs that bind to messenger RNAs (mRNAs) and regulate their activity. They are, therefore, important posttranscriptional regulators. In recent years it has become clear that miRNAs regulate large genetic networks, rather than single genes, and that one gene can be targeted by several miRNAs. To understand the role of miRNAs in cells or tissues, it is therefore important to analyze the targetome of miRNAs. Here, we present a technique called Argonaute-RNA Immunoprecipitation (AGO-RIP) which takes advantages of the fact that miRNAs and their targets are directly bound by the Argonaute protein family. With this approach quantitative, genome-wide analysis of miRNA targets is possible. In this chapter we describe the RIP-methodology and provide advice for RNA sequencing and bioinformatic analyses.


Subject(s)
Argonaute Proteins/metabolism , Immunoprecipitation/methods , MicroRNAs/metabolism , RNA, Messenger/metabolism , Sequence Analysis, RNA/methods , Computational Biology/methods , Gene Library , MicroRNAs/genetics , MicroRNAs/isolation & purification , Protein Binding , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Software
11.
Brain Plast ; 3(2): 195-203, 2018 Aug 10.
Article in English | MEDLINE | ID: mdl-30151343

ABSTRACT

Adult neurogenesis in the mammalian brain, including in humans, occurs throughout life in distinct brain regions. Alterations in adult neurogenesis is a common phenomenon in several different neurodegenerative disorders, which is likely to contribute to the pathophysiology of these disorders. This review summarizes novel concepts related to the interplay between autophagy and microRNAs in control of adult neurogenesis, with a specific focus on its relevance to neurodegenerative diseases.

12.
Cell Syst ; 7(4): 438-452.e8, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30292704

ABSTRACT

Non-coding RNAs regulate many biological processes including neurogenesis. The brain-enriched miR-124 has been assigned as a key player of neuronal differentiation via its complex but little understood regulation of thousands of annotated targets. To systematically chart its regulatory functions, we used CRISPR/Cas9 gene editing to disrupt all six miR-124 alleles in human induced pluripotent stem cells. Upon neuronal induction, miR-124-deleted cells underwent neurogenesis and became functional neurons, albeit with altered morphology and neurotransmitter specification. Using RNA-induced-silencing-complex precipitation, we identified 98 high-confidence miR-124 targets, of which some directly led to decreased viability. By performing advanced transcription-factor-network analysis, we identified indirect miR-124 effects on apoptosis, neuronal subtype differentiation, and the regulation of previously uncharacterized zinc finger transcription factors. Our data emphasize the need for combined experimental- and system-level analyses to comprehensively disentangle and reveal miRNA functions, including their involvement in the neurogenesis of diverse neuronal cell types found in the human brain.


Subject(s)
Gene Regulatory Networks , MicroRNAs/genetics , Neurogenesis/genetics , Cells, Cultured , HEK293 Cells , Humans , MicroRNAs/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cell Rep ; 24(6): 1397-1406, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30089251

ABSTRACT

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.


Subject(s)
Argonaute Proteins/genetics , Autophagy/physiology , Huntington Disease/genetics , MicroRNAs/metabolism , Humans
14.
Sci Rep ; 6: 19879, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26813637

ABSTRACT

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.


Subject(s)
Cognition , MicroRNAs/genetics , Pyramidal Cells/metabolism , Transcriptome , Animals , Cells, Cultured , Dependovirus/genetics , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Gene Order , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Mice, Transgenic , RNA Interference , Signal Transduction
15.
Sci Rep ; 5: 12609, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26219083

ABSTRACT

MicroRNAs (miRNAs) are key players in the regulation of neuronal processes by targeting a large network of target messenger RNAs (mRNAs). However, the identity and function of mRNAs targeted by miRNAs in specific cells of the brain are largely unknown. Here, we established an adeno-associated viral vector (AAV)-based neuron-specific Argonaute2:GFP-RNA immunoprecipitation followed by high-throughput sequencing to analyse the regulatory role of miRNAs in mouse hippocampal neurons. Using this approach, we identified more than two thousand miRNA targets in hippocampal neurons, regulating essential neuronal features such as cell signalling, transcription and axon guidance. Furthermore, we found that stable inhibition of the highly expressed miR-124 and miR-125 in hippocampal neurons led to significant but distinct changes in the AGO2 binding of target mRNAs, resulting in subsequent upregulation of numerous miRNA target genes. These findings greatly enhance our understanding of the miRNA targetome in hippocampal neurons.


Subject(s)
Hippocampus/metabolism , MicroRNAs/genetics , Neurons/metabolism , Animals , Argonaute Proteins/metabolism , Axons/metabolism , Dependovirus/metabolism , Gene Expression/genetics , Immunoprecipitation/methods , Mice , RNA, Messenger/genetics , Transcription, Genetic/genetics
16.
Cell Rep ; 10(1): 20-8, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25543143

ABSTRACT

TRIM28 is a corepressor that mediates transcriptional silencing by establishing local heterochromatin. Here, we show that deletion of TRIM28 in neural progenitor cells (NPCs) results in high-level expression of two groups of endogenous retroviruses (ERVs): IAP1 and MMERVK10C. We find that NPCs use TRIM28-mediated histone modifications to dynamically regulate transcription and silencing of ERVs, which is in contrast to other somatic cell types using DNA methylation. We also show that derepression of ERVs influences transcriptional dynamics in NPCs through the activation of nearby genes and the expression of long noncoding RNAs. These findings demonstrate a unique dynamic transcriptional regulation of ERVs in NPCs. Our results warrant future studies on the role of ERVs in the healthy and diseased brain.


Subject(s)
Endogenous Retroviruses/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic , Animals , DNA Methylation/genetics , Embryonic Stem Cells/virology , Endogenous Retroviruses/pathogenicity , Gene Expression Regulation, Developmental , Heterochromatin/genetics , Histones/metabolism , Humans , Mice , Neurons/virology , Nuclear Proteins/biosynthesis , Repressor Proteins/biosynthesis , Stem Cells/metabolism , Stem Cells/virology , Tripartite Motif-Containing Protein 28
SELECTION OF CITATIONS
SEARCH DETAIL