Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 175(1): 239-253.e17, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197081

ABSTRACT

Many disease-causing missense mutations affect intrinsically disordered regions (IDRs) of proteins, but the molecular mechanism of their pathogenicity is enigmatic. Here, we employ a peptide-based proteomic screen to investigate the impact of mutations in IDRs on protein-protein interactions. We find that mutations in disordered cytosolic regions of three transmembrane proteins (GLUT1, ITPR1, and CACNA1H) lead to an increased clathrin binding. All three mutations create dileucine motifs known to mediate clathrin-dependent trafficking. Follow-up experiments on GLUT1 (SLC2A1), the glucose transporter causative of GLUT1 deficiency syndrome, revealed that the mutated protein mislocalizes to intracellular compartments. Mutant GLUT1 interacts with adaptor proteins (APs) in vitro, and knocking down AP-2 reverts the cellular mislocalization and restores glucose transport. A systematic analysis of other known disease-causing variants revealed a significant and specific overrepresentation of gained dileucine motifs in structurally disordered cytosolic domains of transmembrane proteins. Thus, several mutations in disordered regions appear to cause "dileucineopathies."


Subject(s)
Glucose Transporter Type 1/physiology , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/physiology , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Carbohydrate Metabolism, Inborn Errors , Clathrin/metabolism , Cytoplasm/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/physiology , Intrinsically Disordered Proteins/metabolism , Leucine/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/deficiency , Mutation/genetics , Peptides , Protein Binding , Proteomics/methods
2.
Nature ; 599(7886): 684-691, 2021 11.
Article in English | MEDLINE | ID: mdl-34789882

ABSTRACT

The three-dimensional (3D) structure of chromatin is intrinsically associated with gene regulation and cell function1-3. Methods based on chromatin conformation capture have mapped chromatin structures in neuronal systems such as in vitro differentiated neurons, neurons isolated through fluorescence-activated cell sorting from cortical tissues pooled from different animals and from dissociated whole hippocampi4-6. However, changes in chromatin organization captured by imaging, such as the relocation of Bdnf away from the nuclear periphery after activation7, are invisible with such approaches8. Here we developed immunoGAM, an extension of genome architecture mapping (GAM)2,9, to map 3D chromatin topology genome-wide in specific brain cell types, without tissue disruption, from single animals. GAM is a ligation-free technology that maps genome topology by sequencing the DNA content from thin (about 220 nm) nuclear cryosections. Chromatin interactions are identified from the increased probability of co-segregation of contacting loci across a collection of nuclear slices. ImmunoGAM expands the scope of GAM to enable the selection of specific cell types using low cell numbers (approximately 1,000 cells) within a complex tissue and avoids tissue dissociation2,10. We report cell-type specialized 3D chromatin structures at multiple genomic scales that relate to patterns of gene expression. We discover extensive 'melting' of long genes when they are highly expressed and/or have high chromatin accessibility. The contacts most specific of neuron subtypes contain genes associated with specialized processes, such as addiction and synaptic plasticity, which harbour putative binding sites for neuronal transcription factors within accessible chromatin regions. Moreover, sensory receptor genes are preferentially found in heterochromatic compartments in brain cells, which establish strong contacts across tens of megabases. Our results demonstrate that highly specific chromatin conformations in brain cells are tightly related to gene regulation mechanisms and specialized functions.


Subject(s)
Brain/cytology , Cells/classification , Chromatin Assembly and Disassembly , Chromatin/chemistry , Chromatin/genetics , Genes , Molecular Conformation , Animals , Binding Sites , Cells/metabolism , Chromatin/metabolism , Gene Expression Regulation , Male , Mice , Multigene Family/genetics , Neurons/classification , Neurons/metabolism , Nucleic Acid Denaturation , Transcription Factors/metabolism
3.
EMBO J ; 40(6): e104296, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33459422

ABSTRACT

The IκB kinase (IKK)-NF-κB pathway is activated as part of the DNA damage response and controls both inflammation and resistance to apoptosis. How these distinct functions are achieved remained unknown. We demonstrate here that DNA double-strand breaks elicit two subsequent phases of NF-κB activation in vivo and in vitro, which are mechanistically and functionally distinct. RNA-sequencing reveals that the first-phase controls anti-apoptotic gene expression, while the second drives expression of senescence-associated secretory phenotype (SASP) genes. The rapidly activated first phase is driven by the ATM-PARP1-TRAF6-IKK cascade, which triggers proteasomal destruction of inhibitory IκBα, and is terminated through IκBα re-expression from the NFKBIA gene. The second phase, which is activated days later in senescent cells, is on the other hand independent of IKK and the proteasome. An altered phosphorylation status of NF-κB family member p65/RelA, in part mediated by GSK3ß, results in transcriptional silencing of NFKBIA and IKK-independent, constitutive activation of NF-κB in senescence. Collectively, our study reveals a novel physiological mechanism of NF-κB activation with important implications for genotoxic cancer treatment.


Subject(s)
Cellular Senescence/physiology , I-kappa B Kinase/metabolism , NF-KappaB Inhibitor alpha/biosynthesis , Transcription Factor RelA/metabolism , Transcription, Genetic/genetics , Animals , Apoptosis/genetics , Cell Line , Cell Proliferation/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Female , Gene Silencing/physiology , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/genetics , Phosphorylation , Proteasome Endopeptidase Complex/metabolism
4.
Cell ; 140(5): 744-52, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20211142

ABSTRACT

Combinatorial interactions among transcription factors are critical to directing tissue-specific gene expression. To build a global atlas of these combinations, we have screened for physical interactions among the majority of human and mouse DNA-binding transcription factors (TFs). The complete networks contain 762 human and 877 mouse interactions. Analysis of the networks reveals that highly connected TFs are broadly expressed across tissues, and that roughly half of the measured interactions are conserved between mouse and human. The data highlight the importance of TF combinations for determining cell fate, and they lead to the identification of a SMAD3/FLI1 complex expressed during development of immunity. The availability of large TF combinatorial networks in both human and mouse will provide many opportunities to study gene regulation, tissue differentiation, and mammalian evolution.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Transcription Factors/metabolism , Animals , Cell Differentiation , Evolution, Molecular , Humans , Mice , Monocytes/cytology , Organ Specificity , Smad3 Protein/metabolism , Trans-Activators/metabolism
5.
Genome Res ; 30(7): 1060-1072, 2020 07.
Article in English | MEDLINE | ID: mdl-32718982

ABSTRACT

Long noncoding RNAs (lncRNAs) constitute the majority of transcripts in the mammalian genomes, and yet, their functions remain largely unknown. As part of the FANTOM6 project, we systematically knocked down the expression of 285 lncRNAs in human dermal fibroblasts and quantified cellular growth, morphological changes, and transcriptomic responses using Capped Analysis of Gene Expression (CAGE). Antisense oligonucleotides targeting the same lncRNAs exhibited global concordance, and the molecular phenotype, measured by CAGE, recapitulated the observed cellular phenotypes while providing additional insights on the affected genes and pathways. Here, we disseminate the largest-to-date lncRNA knockdown data set with molecular phenotyping (over 1000 CAGE deep-sequencing libraries) for further exploration and highlight functional roles for ZNF213-AS1 and lnc-KHDC3L-2.


Subject(s)
RNA, Long Noncoding/physiology , Cell Growth Processes/genetics , Cell Movement/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , KCNQ Potassium Channels/metabolism , Molecular Sequence Annotation , Oligonucleotides, Antisense , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering
6.
Proc Natl Acad Sci U S A ; 117(25): 14421-14432, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32522871

ABSTRACT

Epstein-Barr virus (EBV) is a B cell transforming virus that causes B cell malignancies under conditions of immune suppression. EBV orchestrates B cell transformation through its latent membrane proteins (LMPs) and Epstein-Barr nuclear antigens (EBNAs). We here identify secondary mutations in mouse B cell lymphomas induced by LMP1, to predict and identify key functions of other EBV genes during transformation. We find aberrant activation of early B cell factor 1 (EBF1) to promote transformation of LMP1-expressing B cells by inhibiting their differentiation to plasma cells. EBV EBNA3A phenocopies EBF1 activities in LMP1-expressing B cells, promoting transformation while inhibiting differentiation. In cells expressing LMP1 together with LMP2A, EBNA3A only promotes lymphomagenesis when the EBNA2 target Myc is also overexpressed. Collectively, our data support a model where proproliferative activities of LMP1, LMP2A, and EBNA2 in combination with EBNA3A-mediated inhibition of terminal plasma cell differentiation critically control EBV-mediated B cell lymphomagenesis.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/pathogenicity , Lymphoma, B-Cell/pathology , Plasma Cells/pathology , Animals , Cell Differentiation , Cell Line, Tumor , DNA-Binding Proteins/genetics , Disease Models, Animal , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/metabolism , Fibroblasts , Herpesvirus 4, Human/metabolism , Humans , Lymphoma, B-Cell/virology , Mice , Mice, Knockout , Plasma Cells/virology , Primary Cell Culture , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism
7.
Int J Mol Sci ; 24(3)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36769095

ABSTRACT

Critical illness myopathy (CIM) is an acquired, devastating, multifactorial muscle-wasting disease with incomplete recovery. The impact on hospital costs and permanent loss of quality of life is enormous. Incomplete recovery might imply that the function of muscle stem cells (MuSC) is impaired. We tested whether epigenetic alterations could be in part responsible. We characterized human muscle stem cells (MuSC) isolated from early CIM and analyzed epigenetic alterations (CIM n = 15, controls n = 21) by RNA-Seq, immunofluorescence, analysis of DNA repair, and ATAC-Seq. CIM-MuSC were transplanted into immunodeficient NOG mice to assess their regenerative potential. CIM-MuSC exhibited significant growth deficits, reduced ability to differentiate into myotubes, and impaired DNA repair. The chromatin structure was damaged, as characterized by alterations in mRNA of histone 1, depletion or dislocation of core proteins of nucleosome remodeling and deacetylase complex, and loosening of multiple nucleosome-spanning sites. Functionally, CIM-MuSC had a defect in building new muscle fibers. Further, MuSC obtained from the electrically stimulated muscle of CIM patients was very similar to control MuSC, indicating the impact of muscle contraction in the onset of CIM. CIM not only affects working skeletal muscle but has a lasting and severe epigenetic impact on MuSC.


Subject(s)
Mi-2 Nucleosome Remodeling and Deacetylase Complex , Muscular Diseases , Humans , Animals , Mice , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Critical Illness , Quality of Life , Muscular Diseases/metabolism , Muscle, Skeletal/metabolism , Stem Cells
8.
EMBO J ; 37(24)2018 12 14.
Article in English | MEDLINE | ID: mdl-30467221

ABSTRACT

The IκB kinase (IKK) is considered to control gene expression primarily through activation of the transcription factor NF-κB. However, we show here that IKK additionally regulates gene expression on post-transcriptional level. IKK interacted with several mRNA-binding proteins, including a Processing (P) body scaffold protein, termed enhancer of decapping 4 (EDC4). IKK bound to and phosphorylated EDC4 in a stimulus-sensitive manner, leading to co-recruitment of P body components, mRNA decapping proteins 1a and 2 (DCP1a and DCP2) and to an increase in P body numbers. Using RNA sequencing, we identified scores of transcripts whose stability was regulated via the IKK-EDC4 axis. Strikingly, in the absence of stimulus, IKK-EDC4 promoted destabilization of pro-inflammatory cytokines and regulators of apoptosis. Our findings expand the reach of IKK beyond its canonical role as a regulator of transcription.


Subject(s)
I-kappa B Kinase/metabolism , Multiprotein Complexes/metabolism , Proteins/metabolism , RNA Stability , RNA, Messenger/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , HEK293 Cells , Hep G2 Cells , Humans , I-kappa B Kinase/genetics , Multiprotein Complexes/genetics , Proteins/genetics , RNA, Messenger/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
9.
Blood ; 133(13): 1489-1494, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30696620

ABSTRACT

Persistent NF-κB activation is a hallmark of the malignant Hodgkin/Reed-Sternberg (HRS) cells in classical Hodgkin lymphoma (cHL). Genomic lesions, Epstein-Barr virus infection, soluble factors, and tumor-microenvironment interactions contribute to this activation. Here, in an unbiased approach to identify the cHL cell-secreted key factors for NF-κB activation, we have dissected the secretome of cultured cHL cells by chromatography and subsequent mass spectrometry. We identified lymphotoxin-α (LTA) as the causative factor for autocrine and paracrine activation of canonical and noncanonical NF-κB in cHL cell lines. In addition to inducing NF-κB, LTA promotes JAK2/STAT6 signaling. LTA and its receptor TNFRSF14 are transcriptionally activated by noncanonical NF-κB, creating a continuous feedback loop. Furthermore, LTA shapes the expression of cytokines, receptors, immune checkpoint ligands and adhesion molecules, including CSF2, CD40, PD-L1/PD-L2, and VCAM1. Comparison with single-cell gene-activity profiles of human hematopoietic cells showed that LTA induces genes restricted to the lymphoid lineage, as well as those largely restricted to the myeloid lineage. Thus, LTA sustains autocrine NF-κB activation, impacts activation of several signaling pathways, and drives expression of genes essential for microenvironmental interactions and lineage ambiguity. These data provide a robust rationale for targeting LTA as a treatment strategy for cHL patients.


Subject(s)
Hodgkin Disease/immunology , Janus Kinase 2/immunology , Lymphotoxin-alpha/immunology , NF-kappa B/immunology , STAT6 Transcription Factor/immunology , Cell Line , Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Humans , Lymphotoxin-alpha/genetics , Reed-Sternberg Cells/immunology , Reed-Sternberg Cells/metabolism , Signal Transduction , Transcriptional Activation
10.
Nature ; 520(7546): 243-7, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25607372

ABSTRACT

DNA methylation is an epigenetic modification associated with transcriptional repression of promoters and is essential for mammalian development. Establishment of DNA methylation is mediated by the de novo DNA methyltransferases DNMT3A and DNMT3B, whereas DNMT1 ensures maintenance of methylation through replication. Absence of these enzymes is lethal, and somatic mutations in these genes have been associated with several human diseases. How genomic DNA methylation patterns are regulated remains poorly understood, as the mechanisms that guide recruitment and activity of DNMTs in vivo are largely unknown. To gain insights into this matter we determined genomic binding and site-specific activity of the mammalian de novo DNA methyltransferases DNMT3A and DNMT3B. We show that both enzymes localize to methylated, CpG-dense regions in mouse stem cells, yet are excluded from active promoters and enhancers. By specifically measuring sites of de novo methylation, we observe that enzymatic activity reflects binding. De novo methylation increases with CpG density, yet is excluded from nucleosomes. Notably, we observed selective binding of DNMT3B to the bodies of transcribed genes, which leads to their preferential methylation. This targeting to transcribed sequences requires SETD2-mediated methylation of lysine 36 on histone H3 and a functional PWWP domain of DNMT3B. Together these findings reveal how sequence and chromatin cues guide de novo methyltransferase activity to ensure methylome integrity.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Genome/genetics , Animals , Cell Line , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methyltransferase 3A , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Genomics , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Mice , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport , Transcription, Genetic/genetics , DNA Methyltransferase 3B
11.
Nucleic Acids Res ; 47(11): 5735-5745, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31114922

ABSTRACT

High-occupancy target (HOT) regions are segments of the genome with unusually high number of transcription factor binding sites. These regions are observed in multiple species and thought to have biological importance due to high transcription factor occupancy. Furthermore, they coincide with house-keeping gene promoters and consequently associated genes are stably expressed across multiple cell types. Despite these features, HOT regions are solely defined using ChIP-seq experiments and shown to lack canonical motifs for transcription factors that are thought to be bound there. Although, ChIP-seq experiments are the golden standard for finding genome-wide binding sites of a protein, they are not noise free. Here, we show that HOT regions are likely to be ChIP-seq artifacts and they are similar to previously proposed 'hyper-ChIPable' regions. Using ChIP-seq data sets for knocked-out transcription factors, we demonstrate presence of false positive signals on HOT regions. We observe sequence characteristics and genomic features that are discriminatory of HOT regions, such as GC/CpG-rich k-mers, enrichment of RNA-DNA hybrids (R-loops) and DNA tertiary structures (G-quadruplex DNA). The artificial ChIP-seq enrichment on HOT regions could be associated to these discriminatory features. Furthermore, we propose strategies to deal with such artifacts for the future ChIP-seq studies.


Subject(s)
Binding Sites , Chromatin Immunoprecipitation/methods , Promoter Regions, Genetic , Transcription Factors/chemistry , Amino Acid Motifs , Animals , Artifacts , Caenorhabditis elegans , DNA/chemistry , Drosophila melanogaster , False Positive Reactions , G-Quadruplexes , Genome , Genome, Human , Genomics , Humans , Mice , Protein Binding , Protein Domains , RNA/chemistry , Sequence Analysis, DNA
12.
Nucleic Acids Res ; 47(5): 2560-2573, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30590745

ABSTRACT

The proper subcellular localization of RNAs and local translational regulation is crucial in highly compartmentalized cells, such as neurons. RNA localization is mediated by specific cis-regulatory elements usually found in mRNA 3'UTRs. Therefore, processes that generate alternative 3'UTRs-alternative splicing and polyadenylation-have the potential to diversify mRNA localization patterns in neurons. Here, we performed mapping of alternative 3'UTRs in neurites and soma isolated from mESC-derived neurons. Our analysis identified 593 genes with differentially localized 3'UTR isoforms. In particular, we have shown that two isoforms of Cdc42 gene with distinct functions in neuronal polarity are differentially localized between neurites and soma of mESC-derived and mouse primary cortical neurons, at both mRNA and protein level. Using reporter assays and 3'UTR swapping experiments, we have identified the role of alternative 3'UTRs and mRNA transport in differential localization of alternative CDC42 protein isoforms. Moreover, we used SILAC to identify isoform-specific Cdc42 3'UTR-bound proteome with potential role in Cdc42 localization and translation. Our analysis points to usage of alternative 3'UTR isoforms as a novel mechanism to provide for differential localization of functionally diverse alternative protein isoforms.


Subject(s)
Alternative Splicing/genetics , Neurons/metabolism , Protein Isoforms/genetics , cdc42 GTP-Binding Protein/genetics , 3' Untranslated Regions , Animals , Mice , Mouse Embryonic Stem Cells/metabolism , Neurites/metabolism , Neurons/cytology , Polyadenylation/genetics , Protein Isoforms/metabolism , RNA Stability/genetics , RNA Transport/genetics , RNA, Messenger/genetics
13.
Nucleic Acids Res ; 47(2): 570-581, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30517751

ABSTRACT

RNA-binding proteins (RBPs) control and coordinate each stage in the life cycle of RNAs. Although in vivo binding sites of RBPs can now be determined genome-wide, most studies typically focused on individual RBPs. Here, we examined a large compendium of 114 high-quality transcriptome-wide in vivo RBP-RNA cross-linking interaction datasets generated by the same protocol in the same cell line and representing 64 distinct RBPs. Comparative analysis of categories of target RNA binding preference, sequence preference, and transcript region specificity was performed, and identified potential posttranscriptional regulatory modules, i.e. specific combinations of RBPs that bind to specific sets of RNAs and targeted regions. These regulatory modules represented functionally related proteins and exhibited distinct differences in RNA metabolism, expression variance, as well as subcellular localization. This integrative investigation of experimental RBP-RNA interaction evidence and RBP regulatory function in a human cell line will be a valuable resource for understanding the complexity of post-transcriptional regulation.


Subject(s)
Gene Expression Regulation , RNA/metabolism , Ribonucleoproteins/metabolism , Base Sequence , Binding Sites , HEK293 Cells , Humans , RNA/chemistry , Ribonucleoproteins/classification
14.
Genes Dev ; 27(18): 1986-98, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-24065766

ABSTRACT

More than 50% of patients with chondrosarcomas exhibit gain-of-function mutations in either isocitrate dehydrogenase 1 (IDH1) or IDH2. In this study, we performed genome-wide CpG methylation sequencing of chondrosarcoma biopsies and found that IDH mutations were associated with DNA hypermethylation at CpG islands but not other genomic regions. Regions of CpG island hypermethylation were enriched for genes implicated in stem cell maintenance/differentiation and lineage specification. In murine 10T1/2 mesenchymal progenitor cells, expression of mutant IDH2 led to DNA hypermethylation and an impairment in differentiation that could be reversed by treatment with DNA-hypomethylating agents. Introduction of mutant IDH2 also induced loss of contact inhibition and generated undifferentiated sarcomas in vivo. The oncogenic potential of mutant IDH2 correlated with the ability to produce 2-hydroxyglutarate. Together, these data demonstrate that neomorphic IDH2 mutations can be oncogenic in mesenchymal cells.


Subject(s)
Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Chondrosarcoma/enzymology , Chondrosarcoma/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Animals , Bone Neoplasms/physiopathology , Cell Differentiation , Cell Line , Chondrosarcoma/physiopathology , CpG Islands/genetics , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Genome , Glutarates/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Mice , Mice, Nude , Transplantation, Heterologous
15.
Nature ; 507(7492): 381-385, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24531765

ABSTRACT

A core promoter is a stretch of DNA surrounding the transcription start site (TSS) that integrates regulatory inputs and recruits general transcription factors to initiate transcription. The nature and causative relationship of the DNA sequence and chromatin signals that govern the selection of most TSSs by RNA polymerase II remain unresolved. Maternal to zygotic transition represents the most marked change of the transcriptome repertoire in the vertebrate life cycle. Early embryonic development in zebrafish is characterized by a series of transcriptionally silent cell cycles regulated by inherited maternal gene products: zygotic genome activation commences at the tenth cell cycle, marking the mid-blastula transition. This transition provides a unique opportunity to study the rules of TSS selection and the hierarchy of events linking transcription initiation with key chromatin modifications. We analysed TSS usage during zebrafish early embryonic development at high resolution using cap analysis of gene expression, and determined the positions of H3K4me3-marked promoter-associated nucleosomes. Here we show that the transition from the maternal to zygotic transcriptome is characterized by a switch between two fundamentally different modes of defining transcription initiation, which drive the dynamic change of TSS usage and promoter shape. A maternal-specific TSS selection, which requires an A/T-rich (W-box) motif, is replaced with a zygotic TSS selection grammar characterized by broader patterns of dinucleotide enrichments, precisely aligned with the first downstream (+1) nucleosome. The developmental dynamics of the H3K4me3-marked nucleosomes reveal their DNA-sequence-associated positioning at promoters before zygotic transcription and subsequent transcription-independent adjustment to the final position downstream of the zygotic TSS. The two TSS-defining grammars coexist, often physically overlapping, in core promoters of constitutively expressed genes to enable their expression in the two regulatory environments. The dissection of overlapping core promoter determinants represents a framework for future studies of promoter structure and function across different regulatory contexts.


Subject(s)
Promoter Regions, Genetic/genetics , Transcription Initiation Site , Zebrafish/genetics , Animals , Base Sequence , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Histones/metabolism , Methylation , Mothers , Nucleosomes/genetics , Transcription Initiation, Genetic , Transcriptome/genetics , Zebrafish/embryology , Zygote/metabolism
16.
Nucleic Acids Res ; 45(10): e91, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28334930

ABSTRACT

In the field of RNA, the technologies for studying the transcriptome have created a tremendous potential for deciphering the puzzles of the RNA biology. Along with the excitement, the unprecedented volume of RNA related omics data is creating great challenges in bioinformatics analyses. Here, we present the RNA Centric Annotation System (RCAS), an R package, which is designed to ease the process of creating gene-centric annotations and analysis for the genomic regions of interest obtained from various RNA-based omics technologies. The design of RCAS is modular, which enables flexible usage and convenient integration with other bioinformatics workflows. RCAS is an R/Bioconductor package but we also created graphical user interfaces including a Galaxy wrapper and a stand-alone web service. The application of RCAS on published datasets shows that RCAS is not only able to reproduce published findings but also helps generate novel knowledge and hypotheses. The meta-gene profiles, gene-centric annotation, motif analysis and gene-set analysis provided by RCAS provide contextual knowledge which is necessary for understanding the functional aspects of different biological events that involve RNAs. In addition, the array of different interfaces and deployment options adds the convenience of use for different levels of users. RCAS is available at http://bioconductor.org/packages/release/bioc/html/RCAS.html and http://rcas.mdc-berlin.de.


Subject(s)
Genome , Molecular Sequence Annotation/methods , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Transcriptome , User-Computer Interface , Animals , Base Sequence , Binding Sites , Chickens/genetics , Chickens/metabolism , Computational Biology/methods , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Protein Binding , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
17.
Nucleic Acids Res ; 45(W1): W560-W566, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28582575

ABSTRACT

RNA-based regulation has become a major research topic in molecular biology. The analysis of epigenetic and expression data is therefore incomplete if RNA-based regulation is not taken into account. Thus, it is increasingly important but not yet standard to combine RNA-centric data and analysis tools with other types of experimental data such as RNA-seq or ChIP-seq. Here, we present the RNA workbench, a comprehensive set of analysis tools and consolidated workflows that enable the researcher to combine these two worlds. Based on the Galaxy framework the workbench guarantees simple access, easy extension, flexible adaption to personal and security needs, and sophisticated analyses that are independent of command-line knowledge. Currently, it includes more than 50 bioinformatics tools that are dedicated to different research areas of RNA biology including RNA structure analysis, RNA alignment, RNA annotation, RNA-protein interaction, ribosome profiling, RNA-seq analysis and RNA target prediction. The workbench is developed and maintained by experts in RNA bioinformatics and the Galaxy framework. Together with the growing community evolving around this workbench, we are committed to keep the workbench up-to-date for future standards and needs, providing researchers with a reliable and robust framework for RNA data analysis. AVAILABILITY: The RNA workbench is available at https://github.com/bgruening/galaxy-rna-workbench.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA/chemistry , Sequence Analysis, RNA/methods , Software , Computational Biology , Internet , Nucleic Acid Conformation , RNA/metabolism , RNA, Untranslated/chemistry , Workflow
18.
Mol Syst Biol ; 13(10): 946, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038337

ABSTRACT

Polycomb repression in mouse embryonic stem cells (ESCs) is tightly associated with promoter co-occupancy of RNA polymerase II (RNAPII) which is thought to prime genes for activation during early development. However, it is unknown whether RNAPII poising is a general feature of Polycomb repression, or is lost during differentiation. Here, we map the genome-wide occupancy of RNAPII and Polycomb from pluripotent ESCs to non-dividing functional dopaminergic neurons. We find that poised RNAPII complexes are ubiquitously present at Polycomb-repressed genes at all stages of neuronal differentiation. We observe both loss and acquisition of RNAPII and Polycomb at specific groups of genes reflecting their silencing or activation. Strikingly, RNAPII remains poised at transcription factor genes which are silenced in neurons through Polycomb repression, and have major roles in specifying other, non-neuronal lineages. We conclude that RNAPII poising is intrinsically associated with Polycomb repression throughout differentiation. Our work suggests that the tight interplay between RNAPII poising and Polycomb repression not only instructs promoter state transitions, but also may enable promoter plasticity in differentiated cells.


Subject(s)
Dopaminergic Neurons/cytology , Genes, Developmental , Mouse Embryonic Stem Cells/cytology , Polycomb-Group Proteins/metabolism , RNA Polymerase II/metabolism , Animals , Cell Differentiation , Dopaminergic Neurons/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Promoter Regions, Genetic , Sequence Analysis, RNA , Transcription Factors/genetics
19.
Nucleic Acids Res ; 43(Database issue): D160-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25416797

ABSTRACT

The expression of almost all genes in animals is subject to post-transcriptional regulation by RNA binding proteins (RBPs) and microRNAs (miRNAs). The interactions between both RBPs and miRNAs with mRNA can be mapped on a whole-transcriptome level using experimental and computational techniques established in the past years. The combined action of RBPs and miRNAs is thought to form a post-transcriptional regulatory code. Here we present doRiNA 2.0, available at http://dorina.mdc-berlin.de. In this highly improved new version, we have completely reworked the user interface and expanded the database to improve the usability of the website. Taking into account user feedback over the past years, the input forms for both the simple and the combinatorial search function have been streamlined and combined into a single web page that will also display the search results. Especially, custom uploads is one of the key new features in doRiNA 2.0. To enable the inclusion of doRiNA into third-party analysis pipelines, all operations are accessible via a REST API. Alternatively, local installations can be queried using a Python API. Both the web application and the APIs are available under an OSI-approved Open Source license that allows research and commercial access and re-use.


Subject(s)
Databases, Genetic , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Animals , Argonaute Proteins/metabolism , Exons , HEK293 Cells , Humans , Internet , Mice , Nuclear Proteins/metabolism , RNA/metabolism , RNA, Circular , Serine-Arginine Splicing Factors , Software
20.
Genome Res ; 23(11): 1938-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002785

ABSTRACT

Spatiotemporal control of gene expression is central to animal development. Core promoters represent a previously unanticipated regulatory level by interacting with cis-regulatory elements and transcription initiation in different physiological and developmental contexts. Here, we provide a first and comprehensive description of the core promoter repertoire and its dynamic use during the development of a vertebrate embryo. By using cap analysis of gene expression (CAGE), we mapped transcription initiation events at single nucleotide resolution across 12 stages of zebrafish development. These CAGE-based transcriptome maps reveal genome-wide rules of core promoter usage, structure, and dynamics, key to understanding the control of gene regulation during vertebrate ontogeny. They revealed the existence of multiple classes of pervasive intra- and intergenic post-transcriptionally processed RNA products and their developmental dynamics. Among these RNAs, we report splice donor site-associated intronic RNA (sRNA) to be specific to genes of the splicing machinery. For the identification of conserved features, we compared the zebrafish data sets to the first CAGE promoter map of Tetraodon and the existing human CAGE data. We show that a number of features, such as promoter type, newly discovered promoter properties such as a specialized purine-rich initiator motif, as well as sRNAs and the genes in which they are detected, are conserved in mammalian and Tetraodon CAGE-defined promoter maps. The zebrafish developmental promoterome represents a powerful resource for studying developmental gene regulation and revealing promoter features shared across vertebrates.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Purines/metabolism , Transcription Initiation Site , Zebrafish/embryology , Zebrafish/genetics , Animals , Evolution, Molecular , Gene Expression Profiling , Genes , Genome , Phylogeny , Promoter Regions, Genetic , RNA/genetics , RNA/metabolism , RNA Caps/genetics , RNA Splicing , Transcriptome , Vertebrates/genetics
SELECTION OF CITATIONS
SEARCH DETAIL