Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 616(7958): 814-821, 2023 04.
Article in English | MEDLINE | ID: mdl-37046086

ABSTRACT

Physiological homeostasis becomes compromised during ageing, as a result of impairment of cellular processes, including transcription and RNA splicing1-4. However, the molecular mechanisms leading to the loss of transcriptional fidelity are so far elusive, as are ways of preventing it. Here we profiled and analysed genome-wide, ageing-related changes in transcriptional processes across different organisms: nematodes, fruitflies, mice, rats and humans. The average transcriptional elongation speed (RNA polymerase II speed) increased with age in all five species. Along with these changes in elongation speed, we observed changes in splicing, including a reduction of unspliced transcripts and the formation of more circular RNAs. Two lifespan-extending interventions, dietary restriction and lowered insulin-IGF signalling, both reversed most of these ageing-related changes. Genetic variants in RNA polymerase II that reduced its speed in worms5 and flies6 increased their lifespan. Similarly, reducing the speed of RNA polymerase II by overexpressing histone components, to counter age-associated changes in nucleosome positioning, also extended lifespan in flies and the division potential of human cells. Our findings uncover fundamental molecular mechanisms underlying animal ageing and lifespan-extending interventions, and point to possible preventive measures.


Subject(s)
Aging , Longevity , Transcription Elongation, Genetic , Animals , Humans , Mice , Rats , Aging/genetics , Insulin/metabolism , Longevity/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , Drosophila melanogaster/genetics , Caenorhabditis elegans/genetics , RNA, Circular , Somatomedins , Nucleosomes , Histones , Cell Division , Caloric Restriction
2.
Mol Cell ; 70(4): 730-744.e6, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29706538

ABSTRACT

Processes like cellular senescence are characterized by complex events giving rise to heterogeneous cell populations. However, the early molecular events driving this cascade remain elusive. We hypothesized that senescence entry is triggered by an early disruption of the cells' three-dimensional (3D) genome organization. To test this, we combined Hi-C, single-cell and population transcriptomics, imaging, and in silico modeling of three distinct cells types entering senescence. Genes involved in DNA conformation maintenance are suppressed upon senescence entry across all cell types. We show that nuclear depletion of the abundant HMGB2 protein occurs early on the path to senescence and coincides with the dramatic spatial clustering of CTCF. Knocking down HMGB2 suffices for senescence-induced CTCF clustering and for loop reshuffling, while ectopically expressing HMGB2 rescues these effects. Our data suggest that HMGB2-mediated genomic reorganization constitutes a primer for the ensuing senescent program.


Subject(s)
CCCTC-Binding Factor/metabolism , Chromatin/metabolism , Genome, Human , HMGB2 Protein/metabolism , CCCTC-Binding Factor/genetics , Cell Proliferation , Cellular Senescence , Chromatin/genetics , HMGB2 Protein/genetics , Human Umbilical Vein Endothelial Cells , Humans
3.
RNA ; 28(11): 1481-1495, 2022 11.
Article in English | MEDLINE | ID: mdl-35973723

ABSTRACT

Circular RNAs are an endogenous long-lived and abundant noncoding species. Despite their prevalence, only a few circRNAs have been dissected mechanistically to date. Here, we cataloged nascent RNA-enriched circRNAs from primary human cells and functionally assigned a role to circRAB3IP in sustaining cellular homeostasis. We combined "omics" and functional experiments to show how circRAB3IP depletion deregulates hundreds of genes, suppresses cell cycle progression, and induces senescence-associated gene expression changes. Conversely, excess circRAB3IP delivered to endothelial cells via extracellular vesicles suffices for accelerating their division. We attribute these effects to an interplay between circRAB3IP and the general splicing factor SF3B1, which can affect transcript variant expression levels of cell cycle-related genes. Together, our findings link the maintenance of cell homeostasis to the presence of a single circRNA.


Subject(s)
MicroRNAs , RNA, Circular , Humans , RNA, Circular/genetics , Endothelial Cells/metabolism , Cell Proliferation/genetics , RNA, Messenger/genetics , Gene Expression , MicroRNAs/genetics
4.
Genome Res ; 30(4): 515-527, 2020 04.
Article in English | MEDLINE | ID: mdl-32253279

ABSTRACT

Cohesin is a ring-shaped multiprotein complex that is crucial for 3D genome organization and transcriptional regulation during differentiation and development. It also confers sister chromatid cohesion and facilitates DNA damage repair. Besides its core subunits SMC3, SMC1A, and RAD21, cohesin in somatic cells contains one of two orthologous STAG subunits, STAG1 or STAG2. How these variable subunits affect the function of the cohesin complex is still unclear. STAG1- and STAG2-cohesin were initially proposed to organize cohesion at telomeres and centromeres, respectively. Here, we uncover redundant and specific roles of STAG1 and STAG2 in gene regulation and chromatin looping using HCT116 cells with an auxin-inducible degron (AID) tag fused to either STAG1 or STAG2. Following rapid depletion of either subunit, we perform high-resolution Hi-C, gene expression, and sequential ChIP studies to show that STAG1 and STAG2 do not co-occupy individual binding sites and have distinct ways by which they affect looping and gene expression. These findings are further supported by single-molecule localizations via direct stochastic optical reconstruction microscopy (dSTORM) super-resolution imaging. Since somatic and congenital mutations of the STAG subunits are associated with cancer (STAG2) and intellectual disability syndromes with congenital abnormalities (STAG1 and STAG2), we verified STAG1-/STAG2-dependencies using human neural stem cells, hence highlighting their importance in particular disease contexts.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Diploidy , Humans , Nuclear Proteins/chemistry , Protein Binding , Protein Conformation , Proteolysis , Structure-Activity Relationship , Cohesins
5.
Mol Syst Biol ; 17(6): e9760, 2021 06.
Article in English | MEDLINE | ID: mdl-34166567

ABSTRACT

Spatial organization and gene expression of mammalian chromosomes are maintained and regulated in conjunction with cell cycle progression. This is perturbed once cells enter senescence and the highly abundant HMGB1 protein is depleted from nuclei to act as an extracellular proinflammatory stimulus. Despite its physiological importance, we know little about the positioning of HMGB1 on chromatin and its nuclear roles. To address this, we mapped HMGB1 binding genome-wide in two primary cell lines. We integrated ChIP-seq and Hi-C with graph theory to uncover clustering of HMGB1-marked topological domains that harbor genes involved in paracrine senescence. Using simplified Cross-Linking and Immuno-Precipitation and functional tests, we show that HMGB1 is also a bona fide RNA-binding protein (RBP) binding hundreds of mRNAs. It presents an interactome rich in RBPs implicated in senescence regulation. The mRNAs of many of these RBPs are directly bound by HMGB1 and regulate availability of SASP-relevant transcripts. Our findings reveal a broader than hitherto assumed role for HMGB1 in coordinating chromatin folding and RNA homeostasis as part of a regulatory loop controlling cell-autonomous and paracrine senescence.


Subject(s)
HMGB1 Protein , RNA , Animals , Cellular Senescence/genetics , Chromatin/genetics , HMGB1 Protein/genetics , Homeostasis/genetics
6.
Genome Res ; 26(11): 1478-1489, 2016 11.
Article in English | MEDLINE | ID: mdl-27633323

ABSTRACT

Mammalian cells have developed intricate mechanisms to interpret, integrate, and respond to extracellular stimuli. For example, tumor necrosis factor (TNF) rapidly activates proinflammatory genes, but our understanding of how this occurs against the ongoing transcriptional program of the cell is far from complete. Here, we monitor the early phase of this cascade at high spatiotemporal resolution in TNF-stimulated human endothelial cells. NF-κB, the transcription factor complex driving the response, interferes with the regulatory machinery by binding active enhancers already in interaction with gene promoters. Notably, >50% of these enhancers do not encode canonical NF-κB binding motifs. Using a combination of genomics tools, we find that binding site selection plays a key role in NF-κΒ-mediated transcriptional activation and repression. We demonstrate the latter by describing the synergy between NF-κΒ and the corepressor JDP2. Finally, detailed analysis of a 2.8-Mbp locus using sub-kbp-resolution targeted chromatin conformation capture and genome editing uncovers how NF-κΒ that has just entered the nucleus exploits pre-existing chromatin looping to exert its multimodal role. This work highlights the involvement of topology in cis-regulatory element function during acute transcriptional responses, where primary DNA sequence and its higher-order structure constitute a regulatory context leading to either gene activation or repression.


Subject(s)
Consensus Sequence , NF-kappa B/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Cells, Cultured , Chromatin/metabolism , Gene Editing , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , NF-kappa B/genetics , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology
7.
Nucleic Acids Res ; 43(9): 4721-32, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25897131

ABSTRACT

The conventional model for splicing involves excision of each intron in one piece; we demonstrate this inaccurately describes splicing in many human genes. First, after switching on transcription of SAMD4A, a gene with a 134 kb-long first intron, splicing joins the 3' end of exon 1 to successive points within intron 1 well before the acceptor site at exon 2 is made. Second, genome-wide analysis shows that >60% of active genes yield products generated by such intermediate intron splicing. These products are present at ∼15% the levels of primary transcripts, are encoded by conserved sequences similar to those found at canonical acceptors, and marked by distinctive structural and epigenetic features. Finally, using targeted genome editing, we demonstrate that inhibiting the formation of these splicing intermediates affects efficient exon-exon splicing. These findings greatly expand the functional and regulatory complexity of the human transcriptome.


Subject(s)
Introns , RNA Splicing , Cells, Cultured , Exons , Human Umbilical Vein Endothelial Cells/metabolism , Humans , RNA Splice Sites , Repressor Proteins/genetics , Transcription, Genetic
8.
Nucleic Acids Res ; 41(13): 6618-36, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23677615

ABSTRACT

The oncofetal IGF2 mRNA-binding protein 1 (IGF2BP1) controls the migration and invasiveness of primary as well as tumor-derived cells in vitro. Whether the protein also modulates epithelial-mesenchymal-transition (EMT), a hallmark of tumor progression involved in tumor cell dissemination, remained elusive. In this study, we reveal that IGF2BP1 enhances mesenchymal-like cell properties in tumor-derived cells by promoting the expression of the transcriptional regulators LEF1 and SLUG (SNAI2). IGF2BP1 associates with LEF1 transcripts and prevents their degradation in a 3'-UTR-dependent manner resulting in an upregulation of LEF1 expression. LEF1 promotes transcription of the mesenchymal marker fibronectin by associating with the fibronectin 1 promoter. Moreover, LEF1 enforces the synthesis of the 'EMT-driving' transcriptional regulator SNAI2. Accordingly, IGF2BP1 knockdown causes MET-like (mesenchymal-epithelial-transition) morphological changes, enhances the formation of cell-cell contacts and reduces cell migration in various mesenchymal-like tumor-derived cells. However, in epithelial-like tumor-derived cells characterized by a lack or low abundance of IGF2BP1, the protein fails to induce EMT. These findings identify IGF2BP1 as a pro-mesenchymal post-transcriptional determinant, which sustains the synthesis of 'EMT-driving' transcriptional regulators, mesenchymal markers and enhances tumor cell motility. This supports previous reports, suggesting a role of IGF2BP1 in tumor cell dissemination.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Lymphoid Enhancer-Binding Factor 1/metabolism , Mesoderm/cytology , Neoplasms/physiopathology , RNA-Binding Proteins/physiology , Transcription Factors/metabolism , Cell Line, Tumor , Fibronectins/biosynthesis , Fibronectins/genetics , HEK293 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Neoplasms/pathology , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic
9.
Biol Chem ; 395(11): 1301-5, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205722

ABSTRACT

Eukaryotic genomes - until recently dealt with as if they were a cohort of linear DNA molecules - are perplexed three-dimensional structures, the exact conformation of which profoundly affects genome function. Recent advances in molecular biology and DNA sequencing technologies have led to a new understanding of the folding of chromatin in the nucleus. Changes in chromatin structure underlie deployment of new gene expression programs during development, differentiation, or disease. In this review, we revisit data pointing to, arguably, the major force that shapes genomes: transcription of DNA into RNA.


Subject(s)
Genome , Transcription, Genetic , Animals , Chromosomes , DNA/genetics , Humans , RNA/genetics
10.
RNA ; 18(5): 958-72, 2012 May.
Article in English | MEDLINE | ID: mdl-22442037

ABSTRACT

Small nucleolar and small Cajal body RNAs (snoRNAs and scaRNAs) of the H/ACA box and C/D box type are generated by exonucleolytic shortening of longer precursors. Removal of the last few nucleotides at the 3' end is known to be a distinct step. We report that, in human cells, knock-down of the poly(A) specific ribonuclease (PARN), previously implicated only in mRNA metabolism, causes the accumulation of oligoadenylated processing intermediates of H/ACA box but not C/D box RNAs. In agreement with a role of PARN in snoRNA and scaRNA processing, the enzyme is concentrated in nucleoli and Cajal bodies. Oligo(A) tails are attached to a short stub of intron sequence remaining beyond the mature 3' end of the snoRNAs. The noncanonical poly(A) polymerase PAPD5 is responsible for addition of the oligo(A) tails. We suggest that deadenylation is coupled to clean 3' end trimming, which might serve to enhance snoRNA stability.


Subject(s)
Exoribonucleases/metabolism , RNA Nucleotidyltransferases/metabolism , RNA, Small Nucleolar/chemistry , RNA, Small Nucleolar/metabolism , Base Sequence , Catalysis , Cell Nucleolus/metabolism , Coiled Bodies/metabolism , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex , Humans , Nuclear Proteins/metabolism , Nucleotide Motifs , Polyadenylation , Protein Transport , RNA Editing
11.
Aging Cell ; 23(4): e14083, 2024 04.
Article in English | MEDLINE | ID: mdl-38196311

ABSTRACT

Cellular senescence is acknowledged as a key contributor to organismal ageing and late-life disease. Though popular, the study of senescence in vitro can be complicated by the prolonged and asynchronous timing of cells committing to it and by its paracrine effects. To address these issues, we repurposed a small molecule inhibitor, inflachromene (ICM), to induce senescence to human primary cells. Within 6 days of treatment with ICM, senescence hallmarks, including the nuclear eviction of HMGB1 and -B2, are uniformly induced across IMR90 cell populations. By generating and comparing various high throughput datasets from ICM-induced and replicative senescence, we uncovered a high similarity of the two states. Notably though, ICM suppresses the pro-inflammatory secretome associated with senescence, thus alleviating most paracrine effects. In summary, ICM rapidly and synchronously induces a senescent-like phenotype thereby allowing the study of its core regulatory program without confounding heterogeneity.


Subject(s)
Aging , Cellular Senescence , Humans , Aging/genetics , Cellular Senescence/genetics
12.
Nat Commun ; 12(1): 3014, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021162

ABSTRACT

Members of the chromodomain-helicase-DNA binding (CHD) protein family are chromatin remodelers implicated in human pathologies, with CHD6 being one of its least studied members. We discovered a de novo CHD6 missense mutation in a patient clinically presenting the rare Hallermann-Streiff syndrome (HSS). We used genome editing to generate isogenic iPSC lines and model HSS in relevant cell types. By combining genomics with functional in vivo and in vitro assays, we show that CHD6 binds a cohort of autophagy and stress response genes across cell types. The HSS mutation affects CHD6 protein folding and impairs its ability to recruit co-remodelers in response to DNA damage or autophagy stimulation. This leads to accumulation of DNA damage burden and senescence-like phenotypes. We therefore uncovered a molecular mechanism explaining HSS onset via chromatin control of autophagic flux and genotoxic stress surveillance.


Subject(s)
Autophagy/physiology , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Autophagy/genetics , Chromatin , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/metabolism , Epigenomics , Gene Editing , Gene Expression , Hallermann's Syndrome/genetics , Humans , Mutation , Phenotype
13.
Methods Mol Biol ; 1724: 69-75, 2018.
Article in English | MEDLINE | ID: mdl-29322441

ABSTRACT

Fluorescence in situ hybridization (FISH) coupled to high-resolution microscopy is a powerful method for analyzing the subcellular localization of RNA. However, the detection of circular RNAs (circRNAs) using microscopy is challenging because the only feature of a circRNA that can be used for the probe design is its junction. Circular RNAs are expressed at varying levels, and for their efficient monitoring by FISH, background fluorescence levels need to be kept low. Here, we describe a FISH protocol coupled to high-precision localizations using a single fluorescently labeled probe spanning the circRNA junction; this allows circRNA detection in mammalian cells with high signal-to-noise ratios.


Subject(s)
Gene Expression Regulation , In Situ Hybridization, Fluorescence/methods , RNA/analysis , RNA/genetics , Humans , RNA, Circular
14.
Methods Mol Biol ; 1724: 159-166, 2018.
Article in English | MEDLINE | ID: mdl-29322448

ABSTRACT

A substantial proportion of the currently annotated genes in eukaryotes are proposed to function as RNA molecules (>200 bp) with no significant protein coding potential, currently classified as long noncoding RNAs (lncRNA). A distinct subgroup of lncRNAs is circular RNAs (circRNAs), which can be easily identified by unique junction reads, resulting from their biogenesis. CircRNAs are largely cytosolic and thought to either code for micro-peptides or facilitate gene regulation by sequestering microRNAs (miRNAs) or RNA-binding proteins (RBPs) from their targets. Interrogation of the interaction of circRNAs with cellular macromolecular machineries could indicate their mode of action. Here, we detail a sucrose gradient-based method to pinpoint association of a given circRNA (or any transcript of interest) with distinct ribosomal fractions. This method can evaluate the coding potential of candidate circRNAs (or any transcript of interest) and its association with the translation machinery.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Humans , RNA/genetics , RNA, Circular , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
15.
Aging Cell ; 16(1): 183-191, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27785870

ABSTRACT

Replicative senescence has a major impact on function and integrity of cell preparations. This process is reflected by continuous DNA methylation (DNAm) changes at specific CpG dinucleotides in the course of in vitro culture, and such modifications can be used to estimate the state of cellular senescence for quality control of cell preparations. Still, it is unclear how senescence-associated DNAm changes are regulated and whether they occur simultaneously across a cell population. In this study, we analyzed global DNAm profiles of human mesenchymal stem cells (MSCs) and human umbilical vein endothelial cells (HUVECs) to demonstrate that senescence-associated DNAm changes are overall similar in these different cell types. Subsequently, an Epigenetic-Senescence-Signature, based on six CpGs, was either analyzed by pyrosequencing or by bar-coded bisulfite amplicon sequencing. There was a good correlation between predicted and real passage numbers in bulk populations of MSCs (R2  = 0.67) and HUVECs (R2  = 0.97). However, when we analyzed the Epigenetic-Senescence-Signature in subclones of MSCs, the predictions revealed high variation and they were not related to the adipogenic or osteogenic differentiation potential of the subclones. Notably, in clonally derived subpopulations, the DNAm levels of neighboring CpGs differed extensively, indicating that these genomic regions are not synchronously modified during senescence. Taken together, senescence-associated DNAm changes occur in a highly reproducible manner, but they are not synchronously co-regulated. They rather appear to be acquired stochastically-potentially evoked by other epigenetic modifications.


Subject(s)
Cellular Senescence/genetics , DNA Methylation/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Base Sequence , Cell Differentiation/genetics , Clone Cells , CpG Islands/genetics , Epigenesis, Genetic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Sequence Analysis, DNA , Stochastic Processes , Sulfites/metabolism
16.
Genome Biol ; 15(12): 536, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25608606

ABSTRACT

BACKGROUND: The rearrangement of nucleosomes along the DNA fiber profoundly affects gene expression, but little is known about how signalling reshapes the chromatin landscape, in three-dimensional space and over time, to allow establishment of new transcriptional programs. RESULTS: Using micrococcal nuclease treatment and high-throughput sequencing, we map genome-wide changes in nucleosome positioning in primary human endothelial cells stimulated with tumour necrosis factor alpha (TNFα) - a proinflammatory cytokine that signals through nuclear factor kappa-B (NF-κB). Within 10 min, nucleosomes reposition at regions both proximal and distal to NF-κB binding sites, before the transcription factor quantitatively binds thereon. Similarly, in long TNFα-responsive genes, repositioning precedes transcription by pioneering elongating polymerases and appears to nucleate from intragenic enhancer clusters resembling super-enhancers. By 30 min, widespread repositioning throughout megabase pair-long chromosomal segments, with consequential effects on three-dimensional structure (detected using chromosome conformation capture), is seen. CONCLUSIONS: Whilst nucleosome repositioning is viewed as a local phenomenon, our results point to effects occurring over multiple scales. Here, we present data in support of a TNFα-induced priming mechanism, mostly independent of NF-κB binding and/or elongating RNA polymerases, leading to a plastic network of interactions that affects DNA accessibility over large domains.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , NF-kappa B p50 Subunit/metabolism , Nucleosomes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Binding Sites , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , DNA/metabolism , High-Throughput Nucleotide Sequencing , Human Umbilical Vein Endothelial Cells , Humans , Molecular Sequence Data , NF-kappa B p50 Subunit/chemistry , Nucleosomes/genetics , Sequence Analysis, RNA , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL