Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.816
Filter
1.
Cell ; 186(11): 2438-2455.e22, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37178687

ABSTRACT

The generation of distinct messenger RNA isoforms through alternative RNA processing modulates the expression and function of genes, often in a cell-type-specific manner. Here, we assess the regulatory relationships between transcription initiation, alternative splicing, and 3' end site selection. Applying long-read sequencing to accurately represent even the longest transcripts from end to end, we quantify mRNA isoforms in Drosophila tissues, including the transcriptionally complex nervous system. We find that in Drosophila heads, as well as in human cerebral organoids, 3' end site choice is globally influenced by the site of transcription initiation (TSS). "Dominant promoters," characterized by specific epigenetic signatures including p300/CBP binding, impose a transcriptional constraint to define splice and polyadenylation variants. In vivo deletion or overexpression of dominant promoters as well as p300/CBP loss disrupted the 3' end expression landscape. Our study demonstrates the crucial impact of TSS choice on the regulation of transcript diversity and tissue identity.


Subject(s)
Alternative Splicing , RNA Isoforms , Transcription Initiation Site , Humans , Polyadenylation , Promoter Regions, Genetic , RNA Isoforms/metabolism , RNA, Messenger/metabolism
2.
Cell ; 179(7): 1551-1565.e17, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31787377

ABSTRACT

The processing of RNA transcripts from mammalian genes occurs in proximity to their transcription. Here, we describe a phenomenon affecting thousands of genes that we call exon-mediated activation of transcription starts (EMATS), in which the splicing of internal exons impacts promoter choice and the expression level of the gene. We observed that evolutionary gain of internal exons is associated with gain of new transcription start sites (TSSs) nearby and increased gene expression. Inhibiting exon splicing reduced transcription from nearby promoters, and creation of new spliced exons activated transcription from cryptic promoters. The strongest effects occurred for weak promoters located proximal and upstream of efficiently spliced exons. Together, our findings support a model in which splicing recruits transcription machinery locally to influence TSS choice and identify exon gain, loss, and regulatory change as major contributors to the evolution of alternative promoters and gene expression in mammals.


Subject(s)
Exons , Promoter Regions, Genetic , Transcriptional Activation/genetics , 3T3 Cells , Animals , Evolution, Molecular , HeLa Cells , Humans , Mice , RNA Splicing , Transcription Initiation Site
3.
Cell ; 176(3): 520-534.e25, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661750

ABSTRACT

Elucidating the global and local rules that govern genome-wide, hierarchical chromatin architecture remains a critical challenge. Current high-throughput chromosome conformation capture (Hi-C) technologies have identified large-scale chromatin structural motifs, such as topologically associating domains and looping. However, structural rules at the smallest or nucleosome scale remain poorly understood. Here, we coupled nucleosome-resolved Hi-C technology with simulated annealing-molecular dynamics (SA-MD) simulation to reveal 3D spatial distributions of nucleosomes and their genome-wide orientation in chromatin. Our method, called Hi-CO, revealed distinct nucleosome folding motifs across the yeast genome. Our results uncovered two types of basic secondary structural motifs in nucleosome folding: α-tetrahedron and ß-rhombus analogous to α helix and ß sheet motifs in protein folding. Using mutants and cell-cycle-synchronized cells, we further uncovered motifs with specific nucleosome positioning and orientation coupled to epigenetic features at individual loci. By illuminating molecular-level structure-function relationships in eukaryotic chromatin, our findings establish organizational principles of nucleosome folding.


Subject(s)
Chromatin/ultrastructure , Nucleosomes/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Chromosomes/metabolism , Chromosomes/ultrastructure , Nucleosomes/genetics , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Initiation Site
4.
Cell ; 174(5): 1127-1142.e19, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30078706

ABSTRACT

Replication origins, fragile sites, and rDNA have been implicated as sources of chromosomal instability. However, the defining genomic features of replication origins and fragile sites are among the least understood elements of eukaryote genomes. Here, we map sites of replication initiation and breakage in primary cells at high resolution. We find that replication initiates between transcribed genes within nucleosome-depleted structures established by long asymmetrical poly(dA:dT) tracts flanking the initiation site. Paradoxically, long (>20 bp) (dA:dT) tracts are also preferential sites of polar replication fork stalling and collapse within early-replicating fragile sites (ERFSs) and late-replicating common fragile sites (CFSs) and at the rDNA replication fork barrier. Poly(dA:dT) sequences are fragile because long single-strand poly(dA) stretches at the replication fork are unprotected by the replication protein A (RPA). We propose that the evolutionary expansion of poly(dA:dT) tracts in eukaryotic genomes promotes replication initiation, but at the cost of chromosome fragility.


Subject(s)
DNA Replication , DNA, Ribosomal/chemistry , Nucleosomes/metabolism , Poly dA-dT/chemistry , Replication Origin , Amino Acid Motifs , Animals , Cell Line , Chromatin Immunoprecipitation , Chromosomal Instability , Chromosome Fragile Sites , Chromosome Fragility , Female , Male , Mice , Mice, Inbred C57BL , Saccharomyces cerevisiae , Schizosaccharomyces , Transcription Initiation Site , Transcription, Genetic
5.
Cell ; 173(4): 1031-1044.e13, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727662

ABSTRACT

Full understanding of eukaryotic transcriptomes and how they respond to different conditions requires deep knowledge of all sites of intron excision. Although RNA sequencing (RNA-seq) provides much of this information, the low abundance of many spliced transcripts (often due to their rapid cytoplasmic decay) limits the ability of RNA-seq alone to reveal the full repertoire of spliced species. Here, we present "spliceosome profiling," a strategy based on deep sequencing of RNAs co-purifying with late-stage spliceosomes. Spliceosome profiling allows for unambiguous mapping of intron ends to single-nucleotide resolution and branchpoint identification at unprecedented depths. Our data reveal hundreds of new introns in S. pombe and numerous others that were previously misannotated. By providing a means to directly interrogate sites of spliceosome assembly and catalysis genome-wide, spliceosome profiling promises to transform our understanding of RNA processing in the nucleus, much as ribosome profiling has transformed our understanding mRNA translation in the cytoplasm.


Subject(s)
Schizosaccharomyces/genetics , Spliceosomes/metabolism , Transcriptome , Algorithms , Introns , RNA Splicing , RNA, Fungal/metabolism , Ribonucleoproteins/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Sequence Analysis, RNA , Transcription Initiation Site
6.
Mol Cell ; 84(16): 3080-3097.e9, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39043178

ABSTRACT

Alternative transcription start sites can affect transcript isoform diversity and translation levels. In a recently described form of gene regulation, coordinated transcriptional and translational interference results in transcript isoform-dependent changes in protein expression. Specifically, a long undecoded transcript isoform (LUTI) is transcribed from a gene-distal promoter, interfering with expression of the gene-proximal promoter. Although transcriptional and chromatin features associated with LUTI expression have been described, the mechanism underlying LUTI-based transcriptional interference is not well understood. Using an unbiased genetic approach followed by functional genomics, we uncovered that the Swi/Snf chromatin remodeling complex is required for co-transcriptional nucleosome remodeling that leads to LUTI-based repression. We identified genes with tandem promoters that rely on Swi/Snf function for transcriptional interference during protein folding stress, including LUTI-regulated genes. This study provides clear evidence for Swi/Snf playing a direct role in gene repression via a cis transcriptional interference mechanism.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Nucleosomes , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Nucleosomes/metabolism , Nucleosomes/genetics , Gene Expression Regulation, Fungal , Transcription Initiation Site , Chromatin/metabolism , Chromatin/genetics
7.
Cell ; 167(6): 1555-1570.e15, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27889238

ABSTRACT

Nucleosome organization influences gene activity by controlling DNA accessibility to transcription machinery. Here, we develop a chemical biology approach to determine mammalian nucleosome positions genome-wide. We uncovered surprising features of nucleosome organization in mouse embryonic stem cells. In contrast to the prevailing model, we observe that for nearly all mouse genes, a class of fragile nucleosomes occupies previously designated nucleosome-depleted regions around transcription start sites and transcription termination sites. We show that nucleosomes occupy DNA targets for a subset of DNA-binding proteins, including CCCTC-binding factor (CTCF) and pluripotency factors. Furthermore, we provide evidence that promoter-proximal nucleosomes, with the +1 nucleosome in particular, contribute to the pausing of RNA polymerase II. Lastly, we find a characteristic preference for nucleosomes at exon-intron junctions. Taken together, we establish an accurate method for defining the nucleosome landscape and provide a valuable resource for studying nucleosome-mediated gene regulation in mammalian cells.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Nucleosomes/genetics , Animals , CCCTC-Binding Factor , Genome-Wide Association Study , Mice , RNA Polymerase II/metabolism , RNA Splice Sites , RNA Splicing , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription Initiation Site , Transcription, Genetic
8.
Cell ; 165(2): 357-71, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058666

ABSTRACT

We report a mechanism through which the transcription machinery directly controls topoisomerase 1 (TOP1) activity to adjust DNA topology throughout the transcription cycle. By comparing TOP1 occupancy using chromatin immunoprecipitation sequencing (ChIP-seq) versus TOP1 activity using topoisomerase 1 sequencing (TOP1-seq), a method reported here to map catalytically engaged TOP1, TOP1 bound at promoters was discovered to become fully active only after pause-release. This transition coupled the phosphorylation of the carboxyl-terminal-domain (CTD) of RNA polymerase II (RNAPII) with stimulation of TOP1 above its basal rate, enhancing its processivity. TOP1 stimulation is strongly dependent on the kinase activity of BRD4, a protein that phosphorylates Ser2-CTD and regulates RNAPII pause-release. Thus the coordinated action of BRD4 and TOP1 overcame the torsional stress opposing transcription as RNAPII commenced elongation but preserved negative supercoiling that assists promoter melting at start sites. This nexus between transcription and DNA topology promises to elicit new strategies to intercept pathological gene expression.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , DNA/chemistry , DNA Topoisomerases, Type I/genetics , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , RNA Polymerase II/chemistry , RNA Polymerase II/isolation & purification , Transcription Elongation, Genetic , Transcription Factors/isolation & purification , Transcription Initiation Site
9.
Mol Cell ; 83(11): 1798-1809.e7, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37148879

ABSTRACT

At active human genes, the +1 nucleosome is located downstream of the RNA polymerase II (RNA Pol II) pre-initiation complex (PIC). However, at inactive genes, the +1 nucleosome is found further upstream, at a promoter-proximal location. Here, we establish a model system to show that a promoter-proximal +1 nucleosome can reduce RNA synthesis in vivo and in vitro, and we analyze its structural basis. We find that the PIC assembles normally when the edge of the +1 nucleosome is located 18 base pairs (bp) downstream of the transcription start site (TSS). However, when the nucleosome edge is located further upstream, only 10 bp downstream of the TSS, the PIC adopts an inhibited state. The transcription factor IIH (TFIIH) shows a closed conformation and its subunit XPB contacts DNA with only one of its two ATPase lobes, inconsistent with DNA opening. These results provide a mechanism for nucleosome-dependent regulation of transcription initiation.


Subject(s)
Nucleosomes , RNA Polymerase II , Humans , Nucleosomes/genetics , RNA Polymerase II/metabolism , Promoter Regions, Genetic , Transcription Factor TFIIH/metabolism , DNA/genetics , DNA/chemistry , Transcription, Genetic , Transcription Initiation Site
10.
Cell ; 161(4): 879-892, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25936837

ABSTRACT

N(6)-methyldeoxyadenosine (6mA or m(6)A) is a DNA modification preserved in prokaryotes to eukaryotes. It is widespread in bacteria and functions in DNA mismatch repair, chromosome segregation, and virulence regulation. In contrast, the distribution and function of 6mA in eukaryotes have been unclear. Here, we present a comprehensive analysis of the 6mA landscape in the genome of Chlamydomonas using new sequencing approaches. We identified the 6mA modification in 84% of genes in Chlamydomonas. We found that 6mA mainly locates at ApT dinucleotides around transcription start sites (TSS) with a bimodal distribution and appears to mark active genes. A periodic pattern of 6mA deposition was also observed at base resolution, which is associated with nucleosome distribution near the TSS, suggesting a possible role in nucleosome positioning. The new genome-wide mapping of 6mA and its unique distribution in the Chlamydomonas genome suggest potential regulatory roles of 6mA in gene expression in eukaryotic organisms.


Subject(s)
Adenine/analogs & derivatives , Chlamydomonas reinhardtii/genetics , Transcription Initiation Site , 5-Methylcytosine/metabolism , Adenine/metabolism , Chlamydomonas reinhardtii/metabolism , DNA, Algal/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Nucleosomes/metabolism , Transcription, Genetic
11.
Cell ; 163(2): 445-55, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26451488

ABSTRACT

RNA-directed DNA methylation in Arabidopsis thaliana is driven by the plant-specific RNA Polymerase IV (Pol IV). It has been assumed that a Pol IV transcript can give rise to multiple 24-nt small interfering RNAs (siRNAs) that target DNA methylation. Here, we demonstrate that Pol IV-dependent RNAs (P4RNAs) from wild-type Arabidopsis are surprisingly short in length (30 to 40 nt) and mirror 24-nt siRNAs in distribution, abundance, strand bias, and 5'-adenine preference. P4RNAs exhibit transcription start sites similar to Pol II products and are featured with 5'-monophosphates and 3'-misincorporated nucleotides. The 3'-misincorporation preferentially occurs at methylated cytosines on the template DNA strand, suggesting a co-transcriptional feedback to siRNA biogenesis by DNA methylation to reinforce silencing locally. These results highlight an unusual mechanism of Pol IV transcription and suggest a "one precursor, one siRNA" model for the biogenesis of 24-nt siRNAs in Arabidopsis.


Subject(s)
Arabidopsis/metabolism , RNA, Plant/genetics , RNA, Small Interfering/genetics , Adenine/metabolism , Arabidopsis/enzymology , Arabidopsis/genetics , DNA Methylation , DNA-Directed RNA Polymerases/metabolism , Models, Biological , Transcription Initiation Site
12.
Nature ; 631(8022): 891-898, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39020164

ABSTRACT

Patterns of transcriptional activity are encoded in our genome through regulatory elements such as promoters or enhancers that, paradoxically, contain similar assortments of sequence-specific transcription factor (TF) binding sites1-3. Knowledge of how these sequence motifs encode multiple, often overlapping, gene expression programs is central to understanding gene regulation and how mutations in non-coding DNA manifest in disease4,5. Here, by studying gene regulation from the perspective of individual transcription start sites (TSSs), using natural genetic variation, perturbation of endogenous TF protein levels and massively parallel analysis of natural and synthetic regulatory elements, we show that the effect of TF binding on transcription initiation is position dependent. Analysing TF-binding-site occurrences relative to the TSS, we identified several motifs with highly preferential positioning. We show that these patterns are a combination of a TF's distinct functional profiles-many TFs, including canonical activators such as NRF1, NFY and Sp1, activate or repress transcription initiation depending on their precise position relative to the TSS. As such, TFs and their spacing collectively guide the site and frequency of transcription initiation. More broadly, these findings reveal how similar assortments of TF binding sites can generate distinct gene regulatory outcomes depending on their spatial configuration and how DNA sequence polymorphisms may contribute to transcription variation and disease and underscore a critical role for TSS data in decoding the regulatory information of our genome.


Subject(s)
Gene Expression Regulation , Nucleotide Motifs , Promoter Regions, Genetic , Transcription Factors , Transcription Initiation Site , Transcription Initiation, Genetic , Humans , Binding Sites , Gene Expression Regulation/genetics , Genome, Human/genetics , Nucleotide Motifs/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Transcription Factors/metabolism , Genetic Variation
13.
Nature ; 627(8003): 424-430, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418874

ABSTRACT

Mycobacterium tuberculosis (Mtb) is a bacterial pathogen that causes tuberculosis (TB), an infectious disease that is responsible for major health and economic costs worldwide1. Mtb encounters diverse environments during its life cycle and responds to these changes largely by reprogramming its transcriptional output2. However, the mechanisms of Mtb transcription and how they are regulated remain poorly understood. Here we use a sequencing method that simultaneously determines both termini of individual RNA molecules in bacterial cells3 to profile the Mtb transcriptome at high resolution. Unexpectedly, we find that most Mtb transcripts are incomplete, with their 5' ends aligned at transcription start sites and 3' ends located 200-500 nucleotides downstream. We show that these short RNAs are mainly associated with paused RNA polymerases (RNAPs) rather than being products of premature termination. We further show that the high propensity of Mtb RNAP to pause early in transcription relies on the binding of the σ-factor. Finally, we show that a translating ribosome promotes transcription elongation, revealing a potential role for transcription-translation coupling in controlling Mtb gene expression. In sum, our findings depict a mycobacterial transcriptome that prominently features incomplete transcripts resulting from RNAP pausing. We propose that the pausing phase constitutes an important transcriptional checkpoint in Mtb that allows the bacterium to adapt to environmental changes and could be exploited for TB therapeutics.


Subject(s)
Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis , RNA, Bacterial , Transcriptome , DNA-Directed RNA Polymerases/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , RNA, Bacterial/analysis , RNA, Bacterial/biosynthesis , RNA, Bacterial/genetics , Transcriptome/genetics , Tuberculosis/microbiology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Initiation Site , Sigma Factor/metabolism , Ribosomes/metabolism , Protein Biosynthesis
14.
Nat Immunol ; 18(10): 1160-1172, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28783152

ABSTRACT

Regulatory T cells (Treg cells) perform two distinct functions: they maintain self-tolerance, and they support organ homeostasis by differentiating into specialized tissue Treg cells. We found that epigenetic modifications defined the molecular characteristics of tissue Treg cells. Tagmentation-based whole-genome bisulfite sequencing revealed more than 11,000 regions that were methylated differentially in pairwise comparisons of tissue Treg cell populations and lymphoid T cells. Similarities in the epigenetic landscape led to the identification of a common tissue Treg cell population that was present in many organs and was characterized by gain and loss of DNA methylation that included many gene sites associated with the TH2 subset of helper T cells, such as the gene encoding cytokine IL-33 receptor ST2, as well as the production of tissue-regenerative factors. Furthermore, the ST2-expressing population was dependent on the transcriptional regulator BATF and could be expanded by IL-33. Thus, tissue Treg cells integrate multiple waves of epigenetic reprogramming that define their tissue-restricted specialization.


Subject(s)
DNA Methylation , Genome-Wide Association Study , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cluster Analysis , Computational Biology/methods , CpG Islands , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Mice, Transgenic , Molecular Sequence Annotation , Organ Specificity/genetics , Organ Specificity/immunology , Promoter Regions, Genetic , Th2 Cells/metabolism , Transcription Initiation Site , Transcriptome
15.
Cell ; 159(7): 1538-48, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25483776

ABSTRACT

Activation-induced cytidine deaminase (AID) initiates both somatic hypermutation (SHM) for antibody affinity maturation and DNA breakage for antibody class switch recombination (CSR) via transcription-dependent cytidine deamination of single-stranded DNA targets. Though largely specific for immunoglobulin genes, AID also acts on a limited set of off-targets, generating oncogenic translocations and mutations that contribute to B cell lymphoma. How AID is recruited to off-targets has been a long-standing mystery. Based on deep GRO-seq studies of mouse and human B lineage cells activated for CSR or SHM, we report that most robust AID off-target translocations occur within highly focal regions of target genes in which sense and antisense transcription converge. Moreover, we found that such AID-targeting "convergent" transcription arises from antisense transcription that emanates from super-enhancers within sense transcribed gene bodies. Our findings provide an explanation for AID off-targeting to a small subset of mostly lineage-specific genes in activated B cells.


Subject(s)
Cytidine Deaminase/metabolism , Enhancer Elements, Genetic , Genomic Instability , Transcription, Genetic , Animals , B-Lymphocytes/metabolism , Humans , Immunoglobulin Class Switching , Mice , Transcription Initiation Site
16.
Cell ; 156(5): 920-34, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581493

ABSTRACT

Argonaute (Ago) proteins mediate posttranscriptional gene repression by binding guide miRNAs to regulate targeted RNAs. To confidently assess Ago-bound small RNAs, we adapted a mouse embryonic stem cell system to express a single epitope-tagged Ago protein family member in an inducible manner. Here, we report the small RNA profile of Ago-deficient cells and show that Ago-dependent stability is a common feature of mammalian miRNAs. Using this criteria and immunopurification, we identified an Ago-dependent class of noncanonical miRNAs derived from protein-coding gene promoters, which we name transcriptional start site miRNAs (TSS-miRNAs). A subset of promoter-proximal RNA polymerase II (RNAPII) complexes produces hairpin RNAs that are processed in a DiGeorge syndrome critical region gene 8 (Dgcr8)/Drosha-independent but Dicer-dependent manner. TSS-miRNA activity is detectable from endogenous levels and following overexpression of mRNA constructs. Finally, we present evidence of differential expression and conservation in humans, suggesting important roles in gene regulation.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA, Small Untranslated/metabolism , Transcription Elongation, Genetic , Animals , Argonaute Proteins , Base Sequence , Cleavage And Polyadenylation Specificity Factor/metabolism , Embryonic Stem Cells/metabolism , Genetic Techniques , Humans , Mice , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/genetics , Transcription Initiation Site
17.
Cell ; 159(5): 1153-1167, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416952

ABSTRACT

The endoribonuclease Dicer is known for its central role in the biogenesis of eukaryotic small RNAs/microRNAs. Despite its importance, Dicer target transcripts have not been directly mapped. Here, we apply biochemical methods to human cells and C. elegans and identify thousands of Dicer-binding sites. We find known and hundreds of additional miRNAs with high sensitivity and specificity. We also report structural RNAs, promoter RNAs, and mitochondrial transcripts as Dicer targets. Interestingly, most Dicer-binding sites reside on mRNAs/lncRNAs and are not significantly processed into small RNAs. These passive sites typically harbor small, Dicer-bound hairpins within intact transcripts and generally stabilize target expression. We show that passive sites can sequester Dicer and reduce microRNA expression. mRNAs with passive sites were in human and worm significantly associated with processing-body/granule function. Together, we provide the first transcriptome-wide map of Dicer targets and suggest conserved binding modes and functions outside of the miRNA pathway.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Ribonuclease III/metabolism , Animals , Caenorhabditis elegans/metabolism , Chromatin Immunoprecipitation , Humans , MicroRNAs/metabolism , Photochemistry , RNA/metabolism , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , RNA, Mitochondrial , RNA-Binding Proteins/metabolism , Transcription Initiation Site , Transcriptome
18.
Nature ; 622(7981): 173-179, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37731000

ABSTRACT

Lysine residues in histones and other proteins can be modified by post-translational modifications that encode regulatory information1. Lysine acetylation and methylation are especially important for regulating chromatin and gene expression2-4. Pathways involving these post-translational modifications are targets for clinically approved therapeutics to treat human diseases. Lysine methylation and acetylation are generally assumed to be mutually exclusive at the same residue. Here we report cellular lysine residues that are both methylated and acetylated on the same side chain to form Nε-acetyl-Nε-methyllysine (Kacme). We show that Kacme is found on histone H4 (H4Kacme) across a range of species and across mammalian tissues. Kacme is associated with marks of active chromatin, increased transcriptional initiation and is regulated in response to biological signals. H4Kacme can be installed by enzymatic acetylation of monomethyllysine peptides and is resistant to deacetylation by some HDACs in vitro. Kacme can be bound by chromatin proteins that recognize modified lysine residues, as we demonstrate with the crystal structure of acetyllysine-binding protein BRD2 bound to a histone H4Kacme peptide. These results establish Kacme as a cellular post-translational modification with the potential to encode information distinct from methylation and acetylation alone and demonstrate that Kacme has all the hallmarks of a post-translational modification with fundamental importance to chromatin biology.


Subject(s)
Acetylation , Chromatin , Lysine , Methylation , Protein Processing, Post-Translational , Transcription Initiation Site , Animals , Humans , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Histones/chemistry , Histones/metabolism , Lysine/analogs & derivatives , Lysine/chemistry , Lysine/metabolism , Peptides/chemistry , Peptides/metabolism , Histone Deacetylases/metabolism
19.
Mol Cell ; 81(14): 2975-2988.e6, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34157308

ABSTRACT

The heterogeneous nature of eukaryotic replication kinetics and the low efficiency of individual initiation sites make mapping the location and timing of replication initiation in human cells difficult. To address this challenge, we have developed optical replication mapping (ORM), a high-throughput single-molecule approach, and used it to map early-initiation events in human cells. The single-molecule nature of our data and a total of >2,500-fold coverage of the human genome on 27 million fibers averaging ∼300 kb in length allow us to identify initiation sites and their firing probability with high confidence. We find that the distribution of human replication initiation is consistent with inefficient, stochastic activation of heterogeneously distributed potential initiation complexes enriched in accessible chromatin. These observations are consistent with stochastic models of initiation-timing regulation and suggest that stochastic regulation of replication kinetics is a fundamental feature of eukaryotic replication, conserved from yeast to humans.


Subject(s)
DNA Replication/genetics , Eukaryotic Cells/physiology , Genome, Human/genetics , Cell Line, Tumor , Chromatin/genetics , DNA Replication Timing/genetics , Genome, Fungal/genetics , Genome-Wide Association Study/methods , HeLa Cells , Humans , Replication Origin/genetics , Saccharomyces cerevisiae/genetics , Transcription Initiation Site/physiology
20.
Nat Rev Genet ; 23(1): 55-68, 2022 01.
Article in English | MEDLINE | ID: mdl-34526697

ABSTRACT

Plant intra-individual and inter-individual variation can be determined by the epigenome, a set of covalent modifications of DNA and chromatin that can alter genome structure and activity without changes to the genome sequence. The epigenome of plant cells is plastic, that is, it can change in response to internal or external cues, such as during development or due to environmental changes, to create a memory of such events. Ongoing advances in technologies to read and write epigenomic patterns with increasing resolution, scale and precision are enabling the extent of plant epigenome variation to be more extensively characterized and functionally interrogated. In this Review, we discuss epigenome dynamics and variation within plants during development and in response to environmental changes, including stress, as well as between plants. We review known or potential functions of such plasticity and emphasize the importance of investigating the causality of epigenomic changes. Finally, we discuss emerging technologies that may underpin future research into plant epigenome plasticity.


Subject(s)
DNA Methylation , Epigenesis, Genetic/genetics , Epigenome/genetics , Epigenomics , Genetic Variation , Plants/genetics , Gene Expression Regulation, Plant , Genes, Plant/genetics , Models, Genetic , Mutation , Plant Proteins/genetics , Plants/classification , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL