Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 41(11): 1582-1592, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36823354

ABSTRACT

Cell state changes in development and disease are controlled by gene regulatory networks, the dynamics of which are difficult to track in real time. In this study, we used an inducible DCM-RNA polymerase subunit b fusion protein which labels active genes and enhancers with a bacterial methylation mark that does not affect gene transcription and is propagated in S-phase. This DCM-RNA polymerase fusion protein enables transcribed genes and active enhancers to be tagged and then examined at later stages of development or differentiation. We apply this DCM-time machine (DCM-TM) technology to study intestinal homeostasis, revealing rapid and coordinated activation of enhancers and nearby genes during enterocyte differentiation. We provide new insights in absorptive-secretory lineage decision-making in intestinal stem cell (ISC) differentiation and show that ISCs retain a unique chromatin landscape required to maintain ISC identity and delineate future expression of differentiation-associated genes. DCM-TM has wide applicability in tracking cell states, providing new insights in the regulatory networks underlying cell state changes.


Subject(s)
Chromatin , Transcriptome , Cell Lineage/genetics , Transcriptome/genetics , Retrospective Studies , Cell Differentiation/genetics , Chromatin/genetics , DNA-Directed RNA Polymerases/metabolism , Enhancer Elements, Genetic/genetics
2.
PLoS One ; 17(2): e0263262, 2022.
Article in English | MEDLINE | ID: mdl-35176052

ABSTRACT

Genome-wide screens that have viability as a readout have been instrumental to identify essential genes. The development of gene knockout screens with the use of CRISPR-Cas has provided a more sensitive method to identify these genes. Here, we performed an exhaustive genome-wide CRISPR/Cas9 phenotypic rescue screen to identify modulators of cytotoxicity induced by the pioneer transcription factor, DUX4. Misexpression of DUX4 due to a failure in epigenetic repressive mechanisms underlies facioscapulohumeral muscular dystrophy (FHSD), a complex muscle disorder that thus far remains untreatable. As the name implies, FSHD generally starts in the muscles of the face and shoulder girdle. Our CRISPR/Cas9 screen revealed no key effectors other than DUX4 itself that could modulate DUX4 cytotoxicity, suggesting that treatment efforts in FSHD should be directed towards direct modulation of DUX4 itself. Our screen did however reveal some rare and unexpected genomic events, that had an important impact on the interpretation of our data. Our findings may provide important considerations for planning future CRISPR/Cas9 phenotypic survival screens.


Subject(s)
CRISPR-Cas Systems , Gene Expression Regulation , Homeodomain Proteins/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Muscle Cells/pathology , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/pathology , Cell Survival , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Muscle Cells/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts/metabolism
3.
Front Immunol ; 13: 1101999, 2022.
Article in English | MEDLINE | ID: mdl-36685500

ABSTRACT

Introduction: Dendritic cells (DC) are crucial for initiating and shaping immune responses. So far, little is known about the functional specialization of human DC subsets in (local) inflammatory conditions. We profiled conventional (c)DC1, cDC2 and monocytes based on phenotype, transcriptome and function from a local inflammatory site, namely synovial fluid (SF) from patients suffering from a chronic inflammatory condition, Juvenile Idiopathic Arthritis (JIA) as well as patients with rheumatoid arthritis (RA). Methods: Paired PB and SF samples from 32 JIA and 4 RA patients were collected for mononuclear cell isolation. Flow cytometry was done for definition of antigen presenting cell (APC) subsets. Cell sorting was done on the FACSAria II or III. RNA sequencing was done on SF APC subsets. Proliferation assays were done on co-cultures after CD3 magnetic activated cell sorting (MACS). APC Toll-like receptor (TLR) stimulation was done using Pam3CSK4, Poly(I:C), LPS, CpG-A and R848. Cytokine production was measured by Luminex. Results: cDC1, a relatively small DC subset in blood, are strongly enriched in SF, and showed a quiescent immune signature without a clear inflammatory profile, low expression of pathogen recognition receptors (PRRs), chemokine and cytokine receptors, and poor induction of T cell proliferation and cytokine production, but selective production of IFNλ upon polyinosinic:polycytidylic acid exposure. In stark contrast, cDC2 and monocytes from the same environment, showed a pro-inflammatory transcriptional profile, high levels of (spontaneous) pro-inflammatory cytokine production, and strong induction of T cell proliferation and cytokine production, including IL-17. Although the cDC2 and monocytes showed an overlapping transcriptional core profile, there were clear differences in the transcriptional landscape and functional features, indicating that these cell types retain their lineage identity in chronic inflammatory conditions. Discussion: Our findings suggest that at the site of inflammation, there is specific functional programming of human DCs, especially cDC2. In contrast, the enriched cDC1 remain relatively quiescent and seemingly unchanged under inflammatory conditions, pointing to a potentially more regulatory role.


Subject(s)
Arthritis, Juvenile , Arthritis, Rheumatoid , Humans , Synovial Fluid , Dendritic Cells , Cytokines/metabolism
4.
Brain Commun ; 3(4): fcab253, 2021.
Article in English | MEDLINE | ID: mdl-34746789

ABSTRACT

X-linked dystonia-parkinsonism (XDP) is a monogenic neurodegenerative disorder of the basal ganglia, which presents as a combination of hyperkinetic movements and parkinsonian features. The underlying genetic mechanism involves the insertion of a SINE-VNTR-Alu retrotransposon within the TAF1 gene. Interestingly, alterations of TAF1 have been involved in multiple neurological diseases. In XDP, the SINE-VNTR-Alu insertion in TAF1 has been proposed to result in alternative splicing defects, including the decreased incorporation of a neuron-specific microexon annotated as 34'. This mechanism has become controversial as recent studies failed to provide support. In order to resolve this conundrum, we examined the alternative splicing patterns of TAF1 mRNAs in XDP and control brains. The impact of the disease-associated SINE-VNTR-Alu on alternative splicing of microexon 34' was further investigated in cellular assays. Subsequently, microexon 34' incorporation was explored by RT-PCR and Nanopore long-read sequencing of TAF1 mRNAs from XDP and control brains tissues. Using cell-based splicing assays, we demonstrate that presence of the disease-associated SINE-VNTR-Alu does not affect the inclusion of microexon 34'. In addition, we show that (1) microexon 34'-containing TAF1 mRNAs are detected at similar levels in XDP as in controls and that (2) the architecture of TAF1 transcripts is remarkably similar between XDP and controls brains. These results indicate that microexon 34' incorporation into TAF1 mRNA is not affected in XDP brains. Our findings shift the current paradigm of XDP by discounting alternative splicing of TAF1 microexon 34' as the molecular basis for this disease.

5.
Proc Natl Acad Sci U S A ; 117(45): 28422-28432, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33109720

ABSTRACT

The human cerebral cortex contains many cell types that likely underwent independent functional changes during evolution. However, cell-type-specific regulatory landscapes in the cortex remain largely unexplored. Here we report epigenomic and transcriptomic analyses of the two main cortical neuronal subtypes, glutamatergic projection neurons and GABAergic interneurons, in human, chimpanzee, and rhesus macaque. Using genome-wide profiling of the H3K27ac histone modification, we identify neuron-subtype-specific regulatory elements that previously went undetected in bulk brain tissue samples. Human-specific regulatory changes are uncovered in multiple genes, including those associated with language, autism spectrum disorder, and drug addiction. We observe preferential evolutionary divergence in neuron subtype-specific regulatory elements and show that a substantial fraction of pan-neuronal regulatory elements undergoes subtype-specific evolutionary changes. This study sheds light on the interplay between regulatory evolution and cell-type-dependent gene-expression programs, and provides a resource for further exploration of human brain evolution and function.


Subject(s)
Cerebral Cortex/metabolism , Evolution, Molecular , Neurons/metabolism , Animals , Autism Spectrum Disorder/genetics , Brain/metabolism , Epigenesis, Genetic , Epigenomics , Gene Expression , Histone Code , Humans , Interneurons/metabolism , Macaca mulatta/genetics , Pan troglodytes/genetics , Primates/genetics , Regulatory Elements, Transcriptional , Regulatory Sequences, Nucleic Acid , Transcriptome
6.
Cell Rep ; 31(12): 107799, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579926

ABSTRACT

Mutations in non-coding regulatory DNA such as enhancers underlie a wide variety of diseases including developmental disorders and cancer. As enhancers rapidly evolve, understanding their function and configuration in non-human disease models can have important clinical applications. Here, we analyze enhancer configurations in tissues isolated from the common marmoset, a widely used primate model for human disease. Integrating these data with human and mouse data, we find that enhancers containing trait-associated variants are preferentially conserved. In contrast, most human-specific enhancers are highly variable between individuals, with a subset failing to contact promoters. These are located further away from genes and more often reside in inactive B-compartments. Our data show that enhancers typically emerge as instable elements with minimal biological impact prior to their integration in a transcriptional program. Furthermore, our data provide insight into which trait variations in enhancers can be faithfully modeled using the common marmoset.


Subject(s)
Disease/genetics , Enhancer Elements, Genetic , Evolution, Molecular , Genetic Predisposition to Disease , Animals , Callithrix/genetics , Conserved Sequence/genetics , Humans , Mice , Molecular Sequence Annotation , Phylogeny , Promoter Regions, Genetic , Quantitative Trait, Heritable
7.
Nat Commun ; 11(1): 301, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949148

ABSTRACT

Speciation is associated with substantial rewiring of the regulatory circuitry underlying the expression of genes. Determining which changes are relevant and underlie the emergence of the human brain or its unique susceptibility to neural disease has been challenging. Here we annotate changes to gene regulatory elements (GREs) at cell type resolution in the brains of multiple primate species spanning most of primate evolution. We identify a unique set of regulatory elements that emerged in hominins prior to the separation of humans and chimpanzees. We demonstrate that these hominin gains perferentially affect oligodendrocyte function postnatally and are preferentially affected in the brains of autism patients. This preference is also observed for human-specific GREs suggesting this system is under continued selective pressure. Our data provide a roadmap of regulatory rewiring across primate evolution providing insight into the genomic changes that underlie the emergence of the brain and its susceptibility to neural disease.


Subject(s)
Autistic Disorder/metabolism , Brain/metabolism , Hominidae/metabolism , Oligodendroglia/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Animals , Autistic Disorder/genetics , Callithrix , Chromatin , Chromatin Immunoprecipitation , Chromosomes/chemistry , Disease Susceptibility , Evolution, Molecular , Female , Gene Expression Regulation , Genomics , Hominidae/genetics , Humans , Macaca mulatta , Pan troglodytes
8.
RNA Biol ; 17(1): 62-74, 2020 01.
Article in English | MEDLINE | ID: mdl-31559909

ABSTRACT

Neuronal microexons represent the most highly conserved class of alternative splicing events and their timed expression shapes neuronal biology, including neuronal commitment and differentiation. The six-nt microexon 34' is included in the neuronal form of TAF1 mRNA, which encodes the largest subunit of the basal transcription factor TFIID. In this study, we investigate the tissue distribution of TAF1-34' mRNA and protein and the mechanism responsible for its neuronal-specific splicing. Using isoform-specific RNA probes and antibodies, we observe that canonical TAF1 and TAF1-34' have different distributions in the brain, which distinguish proliferating from post-mitotic neurons. Knockdown and ectopic expression experiments demonstrate that the neuronal-specific splicing factor SRRM4/nSR100 promotes the inclusion of microexon 34' into TAF1 mRNA, through the recognition of UGC sequences in the poly-pyrimidine tract upstream of the regulated microexon. These results show that SRRM4 regulates temporal and spatial expression of alternative TAF1 mRNAs to generate a neuronal-specific TFIID complex.


Subject(s)
Exons , Gene Expression Regulation , Histone Acetyltransferases/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA Splicing , RNA, Messenger/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Animals , Brain/metabolism , Cell Differentiation , Immunohistochemistry , Mice , Neurogenesis/genetics , Neurons/cytology
9.
Hemasphere ; 3(4): e270, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31723844

ABSTRACT

The clinical use of histone deacetylase inhibitors (HDACi) for the treatment of bone marrow failure and hematopoietic malignancies has increased dramatically over the last decades. Nonetheless, their effects on normal myelopoiesis remain poorly evaluated. Here, we treated cord blood derived CD34+ progenitor cells with two chemically distinct HDACi inhibitors MS-275 or SAHA and analyzed their effects on the transcriptome (RNA-seq), epigenome (H3K27ac ChIP-seq) and functional and morphological characteristics during neutrophil development. MS-275 (entinostat) selectively inhibits class I HDACs, with a preference for HDAC1, while SAHA (vorinostat) is a non-selective class I/II HDACi. Treatment with individual HDACi resulted in both overlapping and distinct effects on both transcriptome and epigenome, whereas functional effects were relatively similar. Both HDACi resulted in reduced expansion and increased apoptosis in neutrophil progenitor cells. Morphologically, HDACi disrupted normal neutrophil differentiation what was illustrated by decreased percentages of mature neutrophils. In addition, while SAHA treatment clearly showed a block at the promyelocytic stage, MS-275 treatment was characterized by dysplastic features and skewing towards the monocytic lineage. These effects could be mimicked using shRNA-mediated knockdown of HDAC1. Taken together, our data provide novel insights into the effects of HDAC inhibition on normal hematopoietic cells during neutrophil differentiation. These findings should be taken into account when considering the clinical use of MS-275 and SAHA, and can be potentially utilized to tailor more specific, hematopoietic-directed HDACi in the future.

10.
Cell Rep ; 28(11): 2866-2877.e5, 2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31509748

ABSTRACT

Relapse in acute myeloid leukemia (AML) may result from variable genetic origins or convergence on common biological processes. Exploiting the specificity and sensitivity of regulatory DNA, we analyze patient samples of multiple clinical outcomes covering various AML molecular subtypes. We uncover regulatory variation among patients translating into a transcriptional signature that predicts relapse risk. In addition, we find clusters of coexpressed genes within this signature selectively link to relapse risk in distinct patient subgroups defined by molecular subtype or AML maturation. Analyzing these gene clusters and the AML subtypes separately enhances their prognostic value substantially and provides insight in the mechanisms underlying relapse risk across the distinct patient subgroups. We propose that prognostic gene expression signatures in AML are valid only within patient subgroups and do not transcend these subgroups.


Subject(s)
Gene Expression Regulation, Leukemic/genetics , Histones/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Acetylation , Adolescent , Child , Child, Preschool , Chromatin Immunoprecipitation Sequencing , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Ontology , Histones/chemistry , Humans , Infant , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Multigene Family , Mutation , Prognosis , Recurrence , Regulatory Sequences, Nucleic Acid , Risk Factors , Transcriptome
11.
Cell Stem Cell ; 24(3): 462-476.e6, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30849367

ABSTRACT

The SOX2 transcription factor is critical for neural stem cell (NSC) maintenance and brain development. Through chromatin immunoprecipitation (ChIP) and chromatin interaction analysis (ChIA-PET), we determined genome-wide SOX2-bound regions and Pol II-mediated long-range chromatin interactions in brain-derived NSCs. SOX2-bound DNA was highly enriched in distal chromatin regions interacting with promoters and carrying epigenetic enhancer marks. Sox2 deletion caused widespread reduction of Pol II-mediated long-range interactions and decreased gene expression. Genes showing reduced expression in Sox2-deleted cells were significantly enriched in interactions between promoters and SOX2-bound distal enhancers. Expression of one such gene, Suppressor of Cytokine Signaling 3 (Socs3), rescued the self-renewal defect of Sox2-ablated NSCs. Our work identifies SOX2 as a major regulator of gene expression through connections to the enhancer network in NSCs. Through the definition of such a connectivity network, our study shows the way to the identification of genes and enhancers involved in NSC maintenance and neurodevelopmental disorders.


Subject(s)
Chromatin/metabolism , Neural Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cells, Cultured , Gene Regulatory Networks/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Zebrafish
12.
Stroke ; 49(2): 447-453, 2018 02.
Article in English | MEDLINE | ID: mdl-29301971

ABSTRACT

BACKGROUND AND PURPOSE: Genome-wide association studies significantly link intracranial aneurysm (IA) to single-nucleotide polymorphisms (SNPs) in 6 genomic loci. To gain insight into the relevance of these IA-associated SNPs, we aimed to identify regulatory regions and analyze overall gene expression in the human circle of Willis (CoW), on which these aneurysms develop. METHODS: We performed chromatin immunoprecipitation and sequencing for histone modifications H3K4me1 and H3K27ac to identify regulatory regions, including distal enhancers and active promoters, in postmortem specimens of the human CoW. These experiments were complemented with RNA sequencing on the same specimens. We determined whether these regulatory regions overlap with IA-associated SNPs, using computational methods. By combining our results with publicly available data, we investigated the effect of IA-associated SNPs on the newly identified regulatory regions and linked them to potential target genes. RESULTS: We find that IA-associated SNPs are significantly enriched in CoW regulatory regions. Some of the IA-associated SNPs that overlap with a regulatory region are likely to alter transcription factor binding, and in proximity to these regulatory regions are 102 genes that are expressed in the CoW. In addition, gene expression in the CoW is enriched for genes related to cell adhesion and the extracellular matrix. CONCLUSIONS: CoW regulatory regions link IA-associated SNPs to genes with a potential role in the development of IAs. Our data refine previous predictions on SNPs associated with IA and provide a substantial resource from which candidates for follow-up studies can be prioritized.


Subject(s)
Circle of Willis/diagnostic imaging , DNA/metabolism , Genetic Predisposition to Disease , Intracranial Aneurysm/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Female , Genetic Loci/genetics , Genome, Human , Genome-Wide Association Study , Humans , Male , Middle Aged , Risk Factors
13.
Circulation ; 136(15): 1396-1409, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28724751

ABSTRACT

BACKGROUND: Cardiac ischemic injury induces a pathological remodeling response, which can ultimately lead to heart failure. Detailed mechanistic insights into molecular signaling pathways relevant for different aspects of cardiac remodeling will support the identification of novel therapeutic targets. METHODS: Although genome-wide transcriptome analysis on diseased tissues has greatly advanced our understanding of the regulatory networks that drive pathological changes in the heart, this approach has been disadvantaged by the fact that the signals are derived from tissue homogenates. Here we used tomo-seq to obtain a genome-wide gene expression signature with high spatial resolution spanning from the infarcted area to the remote to identify new regulators of cardiac remodeling. Cardiac tissue samples from patients suffering from ischemic heart disease were used to validate our findings. RESULTS: Tracing transcriptional differences with a high spatial resolution across the infarcted heart enabled us to identify gene clusters that share a comparable expression profile. The spatial distribution patterns indicated a separation of expressional changes for genes involved in specific aspects of cardiac remodeling, such as fibrosis, cardiomyocyte hypertrophy, and calcium handling (Col1a2, Nppa, and Serca2). Subsequent correlation analysis allowed for the identification of novel factors that share a comparable transcriptional regulation pattern across the infarcted tissue. The strong correlation between the expression levels of these known marker genes and the expression of the coregulated genes could be confirmed in human ischemic cardiac tissue samples. Follow-up analysis identified SOX9 as common transcriptional regulator of a large portion of the fibrosis-related genes that become activated under conditions of ischemic injury. Lineage-tracing experiments indicated that the majority of COL1-positive fibroblasts stem from a pool of SOX9-expressing cells, and in vivo loss of Sox9 blunted the cardiac fibrotic response on ischemic injury. The colocalization between SOX9 and COL1 could also be confirmed in patients suffering from ischemic heart disease. CONCLUSIONS: Based on the exact local expression cues, tomo-seq can serve to reveal novel genes and key transcription factors involved in specific aspects of cardiac remodeling. Using tomo-seq, we were able to unveil the unknown relevance of SOX9 as a key regulator of cardiac fibrosis, pointing to SOX9 as a potential therapeutic target for cardiac fibrosis.


Subject(s)
Gene Expression Regulation , Muscle Proteins/biosynthesis , Myocardial Ischemia/metabolism , Myocardium/metabolism , SOX9 Transcription Factor/biosynthesis , Collagen Type I/biosynthesis , Collagen Type I/genetics , Female , Fibrosis , High-Throughput Nucleotide Sequencing , Humans , Male , Muscle Proteins/genetics , Myocardial Ischemia/genetics , SOX9 Transcription Factor/genetics
14.
EMBO Rep ; 18(7): 1199-1212, 2017 07.
Article in English | MEDLINE | ID: mdl-28536247

ABSTRACT

Cell-autonomous circadian oscillations strongly influence tissue physiology and pathophysiology of peripheral organs including the heart, in which the circadian clock is known to determine cardiac metabolism and the outcome of for instance ischemic stress. Human pluripotent stem cells represent a powerful tool to study developmental processes in vitro, but the extent to which human embryonic stem (ES) cell-derived cardiomyocytes establish circadian rhythmicity in the absence of a systemic context is unknown. Here we demonstrate that while undifferentiated human ES cells do not possess an intrinsic functional clock, oscillatory expression of known core clock genes emerges spontaneously during directed cardiac differentiation. We identify a set of clock-controlled output genes that contain an oscillatory network of stress-related transcripts. Furthermore, we demonstrate that this network results in a time-dependent functional response to doxorubicin, a frequently used anti-cancer drug with known cardiotoxic side effects. Taken together, our data provide a framework from which the effect of oscillatory gene expression on cardiomyocyte physiology can be modeled in vitro, and demonstrate the influence of a functional clock on experimental outcome.


Subject(s)
CLOCK Proteins/genetics , Circadian Clocks , Human Embryonic Stem Cells/physiology , Myocytes, Cardiac/physiology , Period Circadian Proteins/genetics , Cell Differentiation , Circadian Rhythm , Doxorubicin/pharmacology , Gene Expression , Humans , Myocytes, Cardiac/drug effects , Period Circadian Proteins/metabolism , Topoisomerase II Inhibitors/pharmacology
15.
Cell Rep ; 17(12): 3165-3177, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009287

ABSTRACT

In vertebrate embryos, anterior tissues are generated early, followed by the other axial structures that emerge sequentially from a posterior growth zone. The genetic network driving posterior axial elongation in mice, and its disturbance in mutants with posterior truncation, is not yet fully understood. Here, we show that the combined expression of Cdx2 and T Brachyury is essential to establish the core signature of posterior axial progenitors. Cdx2 and T Brachyury are required for extension of a similar trunk portion of the axis. Simultaneous loss of function of these two genes disrupts axial elongation to a much greater extent than each single mutation alone. We identify and validate common targets for Cdx2 and T Brachyury in vivo, including Wnt and Fgf pathway components active in the axial progenitor niche. Our data demonstrate that integration of the Cdx/Hox and T Brachyury transcriptional networks controls differential axial growth during vertebrate trunk elongation.


Subject(s)
Body Patterning/genetics , CDX2 Transcription Factor/genetics , Fetal Proteins/genetics , Morphogenesis/genetics , T-Box Domain Proteins/genetics , Animals , Embryo, Mammalian , Gene Expression Regulation, Developmental , Gene Regulatory Networks/genetics , Mice , Mutation
16.
Curr Neurol Neurosci Rep ; 16(10): 94, 2016 10.
Article in English | MEDLINE | ID: mdl-27628759

ABSTRACT

Over the last decade, the noncoding part of the genome has been shown to harbour thousands of cis-regulatory elements, such as enhancers, that activate well-defined gene expression programs. Driven by the development of numerous techniques, many of these elements are now identified in multiple tissues and cell types, and their characteristics as well as importance in development and disease are becoming increasingly clear. Here, we provide an overview of the insights that were gained from the analysis of noncoding gene regulatory elements in the brain and describe their potential contribution to cell type specialization, brain function and neurodegenerative disease.


Subject(s)
Brain/metabolism , Enhancer Elements, Genetic/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation/physiology , Neurodegenerative Diseases/genetics , Animals , Humans
17.
Genes Dev ; 30(17): 1937-42, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27633012

ABSTRACT

Sequential 3'-to-5' activation of the Hox gene clusters in early embryos is a most fascinating issue in developmental biology. Neither the trigger nor the regulatory elements involved in the transcriptional initiation of the 3'-most Hox genes have been unraveled in any organism. We demonstrate that a series of enhancers, some of which are Wnt-dependent, is located within a HoxA 3' subtopologically associated domain (subTAD). This subTAD forms the structural basis for multiple layers of 3'-polarized features, including DNA accessibility and enhancer activation. Deletion of the cassette of Wnt-dependent enhancers proves its crucial role in initial transcription of HoxA at the 3' side of the cluster.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcriptional Activation/genetics , Wnt Proteins/metabolism , Animals , Embryo, Mammalian , Enhancer Elements, Genetic/genetics , Mice , Mice, Inbred C57BL , Wnt Proteins/genetics
18.
Nat Commun ; 7: 10967, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26997371

ABSTRACT

DNA methylation likely plays a role in the regulation of human stress reactivity. Here we show that in a genome-wide analysis of blood DNA methylation in 85 healthy individuals, a locus in the Kit ligand gene (KITLG; cg27512205) showed the strongest association with cortisol stress reactivity (P=5.8 × 10(-6)). Replication was obtained in two independent samples using either blood (N=45, P=0.001) or buccal cells (N=255, P=0.004). KITLG methylation strongly mediates the relationship between childhood trauma and cortisol stress reactivity in the discovery sample (32% mediation). Its genomic location, a CpG island shore within an H3K27ac enhancer mark, and the correlation between methylation in the blood and prefrontal cortex provide further evidence that KITLG methylation is functionally relevant for the programming of stress reactivity in the human brain. Our results extend preclinical evidence for epigenetic regulation of stress reactivity to humans and provide leads to enhance our understanding of the neurobiological pathways underlying stress vulnerability.


Subject(s)
DNA Methylation/genetics , Genome, Human , Hydrocortisone/metabolism , Stress, Psychological/genetics , Wounds and Injuries/genetics , Adolescent , Adult , Age Factors , Aged , Child , Epigenesis, Genetic , Ethnicity/genetics , Female , Gene Regulatory Networks , Genetic Loci , Genome-Wide Association Study , Histones/metabolism , Humans , Male , Middle Aged , Models, Genetic , Stem Cell Factor/genetics , Stress, Psychological/blood , Wounds and Injuries/blood , Young Adult
19.
Nat Neurosci ; 19(3): 494-503, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26807951

ABSTRACT

Although genome sequencing has identified numerous noncoding alterations between primate species, which of those are regulatory and potentially relevant to the evolution of the human brain is unclear. Here we annotated cis-regulatory elements (CREs) in the human, rhesus macaque and chimpanzee genomes using chromatin immunoprecipitation followed by sequencing (ChIP-seq) in different anatomical regions of the adult brain. We found high similarity in the genomic positioning of rhesus macaque and human CREs, suggesting that the majority of these elements were already present in a common ancestor 25 million years ago. Most of the observed regulatory changes between humans and rhesus macaques occurred before the ancestral separation of humans and chimpanzees, leaving a modest set of regulatory elements with predicted human specificity. Our data refine previous predictions and hypotheses on the consequences of genomic changes between primate species and allow the identification of regulatory alterations relevant to the evolution of the brain.


Subject(s)
Brain/metabolism , Epigenesis, Genetic/genetics , Epigenomics , Evolution, Molecular , Macaca mulatta/genetics , Pan troglodytes/genetics , Regulatory Elements, Transcriptional/genetics , Animals , Chromatin Immunoprecipitation , Humans
20.
Genome Biol ; 16: 289, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26700097

ABSTRACT

BACKGROUND: Linker histone H1 is a core chromatin component that binds to nucleosome core particles and the linker DNA between nucleosomes. It has been implicated in chromatin compaction and gene regulation and is anticipated to play a role in higher-order genome structure. Here we have used a combination of genome-wide approaches including DNA methylation, histone modification and DNase I hypersensitivity profiling as well as Hi-C to investigate the impact of reduced cellular levels of histone H1 in embryonic stem cells on chromatin folding and function. RESULTS: We find that depletion of histone H1 changes the epigenetic signature of thousands of potential regulatory sites across the genome. Many of them show cooperative loss or gain of multiple chromatin marks. Epigenetic alterations cluster to gene-dense topologically associating domains (TADs) that already showed a high density of corresponding chromatin features. Genome organization at the three-dimensional level is largely intact, but we find changes in the structural segmentation of chromosomes specifically for the epigenetically most modified TADs. CONCLUSIONS: Our data show that cells require normal histone H1 levels to expose their proper regulatory landscape. Reducing the levels of histone H1 results in massive epigenetic changes and altered topological organization particularly at the most active chromosomal domains. Changes in TAD configuration coincide with epigenetic landscape changes but not with transcriptional output changes, supporting the emerging concept that transcriptional control and nuclear positioning of TADs are not causally related but independently controlled by the locally associated trans-acting factors.


Subject(s)
Chromatin Assembly and Disassembly , Epigenesis, Genetic , Histones/metabolism , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , Histones/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...