Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Nat Cell Biol ; 25(12): 1873-1883, 2023 Dec.
Article En | MEDLINE | ID: mdl-37996647

Gene expression is regulated by multiple epigenetic mechanisms, which are coordinated in development and disease. However, current multiomics methods are frequently limited to one or two modalities at a time, making it challenging to obtain a comprehensive gene regulatory signature. Here, we describe a method-3D genome, RNA, accessibility and methylation sequencing (3DRAM-seq)-that simultaneously interrogates spatial genome organization, chromatin accessibility and DNA methylation genome-wide and at high resolution. We combine 3DRAM-seq with immunoFACS and RNA sequencing in cortical organoids to map the cell-type-specific regulatory landscape of human neural development across multiple epigenetic layers. Finally, we apply a massively parallel reporter assay to profile cell-type-specific enhancer activity in organoids and to functionally assess the role of key transcription factors for human enhancer activation and function. More broadly, 3DRAM-seq can be used to profile the multimodal epigenetic landscape in rare cell types and different tissues.


Chromatin , Epigenome , Humans , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic , DNA Methylation/genetics , Organoids/metabolism
2.
Nat Neurosci ; 25(2): 154-167, 2022 02.
Article En | MEDLINE | ID: mdl-35132236

How multiple epigenetic layers and transcription factors (TFs) interact to facilitate brain development is largely unknown. Here, to systematically map the regulatory landscape of neural differentiation in the mouse neocortex, we profiled gene expression and chromatin accessibility in single cells and integrated these data with measurements of enhancer activity, DNA methylation and three-dimensional genome architecture in purified cell populations. This allowed us to identify thousands of new enhancers, their predicted target genes and the temporal relationships between enhancer activation, epigenome remodeling and gene expression. We characterize specific neuronal transcription factors associated with extensive and frequently coordinated changes across multiple epigenetic modalities. In addition, we functionally demonstrate a new role for Neurog2 in directly mediating enhancer activity, DNA demethylation, increasing chromatin accessibility and facilitating chromatin looping in vivo. Our work provides a global view of the gene regulatory logic of lineage specification in the cerebral cortex.


Chromatin , Epigenome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Mice , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Development ; 149(3)2022 02 01.
Article En | MEDLINE | ID: mdl-35147187

Corticogenesis consists of a series of synchronised events, including fate transition of cortical progenitors, neuronal migration, specification and connectivity. NeuroD1, a basic helix-loop-helix (bHLH) transcription factor (TF), contributes to all of these events, but how it coordinates these independently is still unknown. Here, we demonstrate that NeuroD1 expression is accompanied by a gain of active chromatin at a large number of genomic loci. Interestingly, transcriptional activation of these loci relied on a high local density of adjacent bHLH TFs motifs, including, predominantly, Tcf12. We found that activity and expression levels of Tcf12 were high in cells with induced levels of NeuroD1 that spanned the transition of cortical progenitors from proliferative to neurogenic divisions. Moreover, Tcf12 forms a complex with NeuroD1 and co-occupies a subset of NeuroD1 target loci. This Tcf12-NeuroD1 cooperativity is essential for gaining active chromatin and targeted expression of genes involved in cell migration. By functional manipulation in vivo, we further show that Tcf12 is essential during cortical development for the correct migration of newborn neurons and, hence, for proper cortical lamination.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Movement , Cerebral Cortex/metabolism , Chromatin/metabolism , Embryonic Development/genetics , Female , Histones/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neurogenesis , Neurons/cytology , Neurons/metabolism , RNA Interference , RNA, Small Interfering/metabolism
4.
Life Sci Alliance ; 2(2)2019 04.
Article En | MEDLINE | ID: mdl-30814272

Dynamic changes in DNA (hydroxy-)methylation are fundamental for stem cell differentiation. However, the signature of these epigenetic marks in specific cell types during corticogenesis is unknown. Moreover, site-specific manipulation of cytosine modifications is needed to reveal the significance and function of these changes. Here, we report the first assessment of (hydroxy-)methylation in neural stem cells, neurogenic progenitors, and newborn neurons during mammalian corticogenesis. We found that gain in hydroxymethylation and loss in methylation occur sequentially at specific cellular transitions during neurogenic commitment. We also found that these changes predominantly occur within enhancers of neurogenic genes up-regulated during neurogenesis and target of pioneer transcription factors. We further optimized the use of dCas9-Tet1 manipulation of (hydroxy-)methylation, locus-specifically, in vivo, showing the biological relevance of our observations for Dchs1, a regulator of corticogenesis involved in developmental malformations and cognitive impairment. Together, our data reveal the dynamics of cytosine modifications in lineage-related cell types, whereby methylation is reduced and hydroxymethylation gained during the neurogenic lineage concurrently with up-regulation of pioneer transcription factors and activation of enhancers for neurogenic genes.


5-Methylcytosine/analogs & derivatives , DNA Methylation/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics , 5-Methylcytosine/physiology , Animals , CRISPR-Associated Protein 9/metabolism , Cadherins/metabolism , Cell Differentiation , Cell Lineage/physiology , Cytosine/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Oncogene Proteins, Fusion/metabolism , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors , Transcriptome
5.
Front Neurosci ; 12: 85, 2018.
Article En | MEDLINE | ID: mdl-29515357

Epigenetic modifications of DNA and chromatin are long known to control stem cell differentiation and organ function but the role of similar modifications at the level or regulatory RNAs is just beginning to emerge. Over 160 RNA modifications have been identified but their abundance, distribution and functional significance are not known. The few available maps of RNA modifications indicated their dynamic regulation during somatic stem cell differentiation, brain development and function in adulthood suggesting a hitherto unsuspected layer of regulation both at the level of RNA metabolism and post-transcriptional control of gene expression. The advent of programmable, RNA-specific CRISPR-Cas editing platforms together with the identification of RNA modifying enzymes now offers the opportunity to investigate the functional role of these elusive epitranscriptome changes. Here, we discuss recent insights in studying the most abundant modifications in functional mRNAs and lncRNAs, N6-methyladenosine and 5-(hydroxy-)methylcytosine, and their role in regulating somatic stem cell differentiation with particular attention to neural stem cells during mammalian corticogenesis. An outlook on novel CRISPR-Cas based systems that allow stem cell reprogramming by epitranscriptome-editing will also be discussed.

6.
J Am Chem Soc ; 138(38): 12364-7, 2016 09 28.
Article En | MEDLINE | ID: mdl-27632643

Total syntheses of oridamycin A, triptoquinones B and C, and isoiresin are accomplished from a common intermediate prepared via iridium-catalyzed alcohol C-H tert-(hydroxy)prenylation - a byproduct-free process that forms an all-carbon quaternary stereocenter with excellent control of diastereo- and enantioselectivity.


Sesquiterpenes/chemical synthesis , Terpenes/chemistry , Catalysis , Molecular Structure , Sesquiterpenes/chemistry
7.
EMBO J ; 35(8): 803-19, 2016 Apr 15.
Article En | MEDLINE | ID: mdl-26929011

A mutation in the centrosomal-P4.1-associated protein (CPAP) causes Seckel syndrome with microcephaly, which is suggested to arise from a decline in neural progenitor cells (NPCs) during development. However, mechanisms ofNPCs maintenance remain unclear. Here, we report an unexpected role for the cilium inNPCs maintenance and identifyCPAPas a negative regulator of ciliary length independent of its role in centrosome biogenesis. At the onset of cilium disassembly,CPAPprovides a scaffold for the cilium disassembly complex (CDC), which includes Nde1, Aurora A, andOFD1, recruited to the ciliary base for timely cilium disassembly. In contrast, mutatedCPAPfails to localize at the ciliary base associated with inefficientCDCrecruitment, long cilia, retarded cilium disassembly, and delayed cell cycle re-entry leading to premature differentiation of patientiPS-derivedNPCs. AberrantCDCfunction also promotes premature differentiation ofNPCs in SeckeliPS-derived organoids. Thus, our results suggest a role for cilia in microcephaly and its involvement during neurogenesis and brain size control.


Cilia/metabolism , Microcephaly/pathology , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/pathology , Aurora Kinase A/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cilia/genetics , Cilia/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/physiology , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Mutation , Neural Stem Cells/metabolism , Proteins/metabolism , Syndrome
8.
EMBO J ; 35(1): 24-45, 2016 Jan 04.
Article En | MEDLINE | ID: mdl-26516211

Cell fate specification relies on the action of critical transcription factors that become available at distinct stages of embryonic development. One such factor is NeuroD1, which is essential for eliciting the neuronal development program and possesses the ability to reprogram other cell types into neurons. Given this capacity, it is important to understand its targets and the mechanism underlying neuronal specification. Here, we show that NeuroD1 directly binds regulatory elements of neuronal genes that are developmentally silenced by epigenetic mechanisms. This targeting is sufficient to initiate events that confer transcriptional competence, including reprogramming of transcription factor landscape, conversion of heterochromatin to euchromatin, and increased chromatin accessibility, indicating potential pioneer factor ability of NeuroD1. The transcriptional induction of neuronal fate genes is maintained via epigenetic memory despite a transient NeuroD1 induction during neurogenesis. NeuroD1 also induces genes involved in the epithelial-to-mesenchymal transition, thereby promoting neuronal migration. Our study not only reveals the NeuroD1-dependent gene regulatory program driving neurogenesis but also increases our understanding of how cell fate specification during development involves a concerted action of transcription factors and epigenetic mechanisms.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Chromatin/metabolism , Gene Expression Regulation, Developmental , Neurons/physiology , Transcription Factors/metabolism , Animals , Cell Line , Epigenesis, Genetic , Gene Regulatory Networks , Mice
9.
EMBO Rep ; 15(12): 1224-5, 2014 Dec.
Article En | MEDLINE | ID: mdl-25366322

Recent studies have highlighted the importance of regulatory non­coding RNAs and epigenetics in controlling the differentiation of somatic stem cells. Two major pathways characterize these fields: micro­RNAs (miRNAs) and DNA methylation. In this issue of EMBO Reports, Lv et al show that during mammalian corticogenesis, miR­15b inhibits cytosine demethylation by targeting Tet3, a key methylcytosine dioxygenase. This leads to the epigenetic downregulation of cyclin D1. As a result, cell cycle and differentiation of neural progenitors are altered, promoting their switch to neurogenesis. Hence, Lv et al elegantly bring together miRNAs and DNA methylation in the cell cycle control of neural progenitors and neurogenesis.


DNA-Binding Proteins/metabolism , MicroRNAs/physiology , Neocortex/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Proto-Oncogene Proteins/metabolism , Animals , Female , Pregnancy
...