Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 23(9): 623-640, 2022 09.
Article in English | MEDLINE | ID: mdl-35562425

ABSTRACT

Heterochromatin is characterized by dimethylated or trimethylated histone H3 Lys9 (H3K9me2 or H3K9me3, respectively) and is found at transposable elements, satellite repeats and genes, where it ensures their transcriptional silencing. The histone methyltransferases (HMTs) that methylate H3K9 - in mammals Suppressor of variegation 3-9 homologue 1 (SUV39H1), SUV39H2, SET domain bifurcated 1 (SETDB1), SETDB2, G9A and G9A-like protein (GLP) - and the 'readers' of H3K9me2 or H3K9me3 are highly conserved and show considerable redundancy. Despite their redundancy, genetic ablation or mistargeting of an individual H3K9 methyltransferase can correlate with impaired cell differentiation, loss of tissue identity, premature aging and/or cancer. In this Review, we discuss recent advances in understanding the roles of the known H3K9-specific HMTs in ensuring transcriptional homeostasis during tissue differentiation in mammals. We examine the effects of H3K9-methylation-dependent gene repression in haematopoiesis, muscle differentiation and neurogenesis in mammals, and compare them with mechanistic insights obtained from the study of model organisms, notably Caenorhabditis elegans and Drosophila melanogaster. In all these organisms, H3K9-specific HMTs have both unique and redundant roles that ensure the maintenance of tissue integrity by restricting the binding of transcription factors to lineage-specific promoters and enhancer elements.


Subject(s)
Heterochromatin , Histone-Lysine N-Methyltransferase , Animals , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Mammals/genetics , Methylation
3.
Cell ; 163(6): 1333-47, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26607792

ABSTRACT

Interphase chromatin is organized in distinct nuclear sub-compartments, reflecting its degree of compaction and transcriptional status. In Caenorhabditis elegans embryos, H3K9 methylation is necessary to silence and to anchor repeat-rich heterochromatin at the nuclear periphery. In a screen for perinuclear anchors of heterochromatin, we identified a previously uncharacterized C. elegans chromodomain protein, CEC-4. CEC-4 binds preferentially mono-, di-, or tri-methylated H3K9 and localizes at the nuclear envelope independently of H3K9 methylation and nuclear lamin. CEC-4 is necessary for endogenous heterochromatin anchoring, but not for transcriptional repression, in contrast to other known H3K9 methyl-binders in worms, which mediate gene repression but not perinuclear anchoring. When we ectopically induce a muscle differentiation program in embryos, cec-4 mutants fail to commit fully to muscle cell fate. This suggests that perinuclear sequestration of chromatin during development helps restrict cell differentiation programs by stabilizing commitment to a specific cell fate. PAPERCLIP.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Nonmammalian/cytology , Heterochromatin , Histone Code , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Embryo, Nonmammalian/metabolism , Molecular Sequence Data , Sequence Alignment
4.
Mol Cell ; 81(4): 811-829.e6, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33529595

ABSTRACT

Eukaryotic cells package their genomes around histone octamers. In response to DNA damage, checkpoint activation in yeast induces core histone degradation resulting in 20%-40% reduction in nucleosome occupancy. To gain insight into this process, we developed a new approach to analyze the chromatin-associated proteome comprehensively before and after damage. This revealed extensive changes in protein composition after Zeocin-induced damage. First, core histones and the H1 homolog Hho1 were partially lost from chromatin along with replication, transcription, and chromatin remodeling machineries, while ubiquitin ligases and the proteasome were recruited. We found that the checkpoint- and INO80C-dependent recruitment of five ubiquitin-conjugating factors (Rad6, Bre1, Pep5, Ufd4, and Rsp5) contributes to core and linker histone depletion, reducing chromatin compaction and enhancing DNA locus mobility. Importantly, loss of Rad6/Bre1, Ufd4/TRIP12, and Pep5/VPS11 compromise DNA strand invasion kinetics during homology-driven repair. Thus we provide a comprehensive overview of a functionally relevant genome-wide chromatin response to DNA damage.


Subject(s)
Chromatin Assembly and Disassembly , DNA Repair , DNA, Fungal/metabolism , Histones/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligases/metabolism , DNA, Fungal/genetics , Histones/genetics , Proteasome Endopeptidase Complex/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics
5.
Mol Cell ; 81(1): 183-197.e6, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33278361

ABSTRACT

Mre11-Rad50-Xrs2 (MRX) is a highly conserved complex with key roles in various aspects of DNA repair. Here, we report a new function for MRX in limiting transcription in budding yeast. We show that MRX interacts physically and colocalizes on chromatin with the transcriptional co-regulator Mediator. MRX restricts transcription of coding and noncoding DNA by a mechanism that does not require the nuclease activity of Mre11. MRX is required to tether transcriptionally active loci to the nuclear pore complex (NPC), and it also promotes large-scale gene-NPC interactions. Moreover, MRX-mediated chromatin anchoring to the NPC contributes to chromosome folding and helps to control gene expression. Together, these findings indicate that MRX has a role in transcription and chromosome organization that is distinct from its known function in DNA repair.


Subject(s)
Chromosomes, Fungal/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Chromosomes, Fungal/genetics , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/genetics , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
6.
Genes Dev ; 35(1-2): 82-101, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33303642

ABSTRACT

The establishment and maintenance of chromatin domains shape the epigenetic memory of a cell, with the methylation of histone H3 lysine 9 (H3K9me) defining transcriptionally silent heterochromatin. We show here that the C. elegans SET-25 (SUV39/G9a) histone methyltransferase (HMT), which catalyzes H3K9me1, me2 and me3, can establish repressed chromatin domains de novo, unlike the SETDB1 homolog MET-2. Thus, SET-25 is needed to silence novel insertions of RNA or DNA transposons, and repress tissue-specific genes de novo during development. We identify two partially redundant pathways that recruit SET-25 to its targets. One pathway requires LIN-61 (L3MBTL2), which uses its four MBT domains to bind the H3K9me2 deposited by MET-2. The second pathway functions independently of MET-2 and involves the somatic Argonaute NRDE-3 and small RNAs. This pathway targets primarily highly conserved RNA and DNA transposons. These redundant SET-25 targeting pathways (MET-2-LIN-61-SET-25 and NRDE-3-SET-25) ensure repression of intact transposons and de novo insertions, while MET-2 can act alone to repress simple and satellite repeats. Removal of both pathways in the met-2;nrde-3 double mutant leads to the loss of somatic H3K9me2 and me3 and the synergistic derepression of transposons in embryos, strongly elevating embryonic lethality.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Transposable Elements/genetics , Gene Expression Regulation/genetics , Heterochromatin/genetics , RNA-Binding Proteins/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Embryo, Nonmammalian , Gene Silencing , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methylation , Mutation , RNA-Binding Proteins/genetics
7.
Cell ; 152(6): 1355-64, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23498942

ABSTRACT

Mechanistic analyses based on improved imaging techniques have begun to explore the biological implications of chromatin movement within the nucleus. Studies in both prokaryotes and eukaryotes have shed light on what regulates the mobility of DNA over long distances. Interestingly, in eukaryotes, genomic loci increase their movement in response to double-strand break induction. Break mobility, in turn, correlates with the efficiency of repair by homologous recombination. We review here the source and regulation of DNA mobility and discuss how it can both contribute to and jeopardize genome stability.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Genomic Instability , Animals , DNA/metabolism , DNA Damage , DNA Repair , Humans , Movement , Transcription, Genetic
8.
Mol Cell ; 80(2): 311-326.e4, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32970994

ABSTRACT

To determine whether double-strand break (DSB) mobility enhances the physical search for an ectopic template during homology-directed repair (HDR), we tested the effects of factors that control chromatin dynamics, including cohesin loading and kinetochore anchoring. The former but not the latter is altered in response to DSBs. Loss of the nonhistone high-mobility group protein Nhp6 reduces histone occupancy and increases chromatin movement, decompaction, and ectopic HDR. The loss of nucleosome remodeler INO80-C did the opposite. To see whether enhanced HDR depends on DSB mobility or the global chromatin response, we tested the ubiquitin ligase mutant uls1Δ, which selectively impairs local but not global movement in response to a DSB. Strand invasion occurs in uls1Δ cells with wild-type kinetics, arguing that global histone depletion rather than DSB movement is rate limiting for HDR. Impaired break movement in uls1Δ correlates with elevated MRX and cohesin loading, despite normal resection and checkpoint activation.


Subject(s)
DNA Breaks, Double-Stranded , Nucleosomes/metabolism , Saccharomyces cerevisiae/metabolism , Bleomycin/pharmacology , Cell Cycle , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA, Fungal/metabolism , Histones/metabolism , Models, Biological , Phosphorylation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/metabolism , Spindle Pole Bodies/metabolism , Cohesins
9.
Genes Dev ; 34(7-8): 560-579, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32139421

ABSTRACT

Mutations in the nuclear structural protein lamin A produce rare, tissue-specific diseases called laminopathies. The introduction of a human Emery-Dreifuss muscular dystrophy (EDMD)-inducing mutation into the C. elegans lamin (LMN-Y59C), recapitulates many muscular dystrophy phenotypes, and correlates with hyper-sequestration of a heterochromatic array at the nuclear periphery in muscle cells. Using muscle-specific emerin Dam-ID in worms, we monitored the effects of the mutation on endogenous chromatin. An increased contact with the nuclear periphery along chromosome arms, and an enhanced release of chromosomal centers, coincided with the disease phenotypes of reduced locomotion and compromised sarcomere integrity. The coupling of the LMN-Y59C mutation with the ablation of CEC-4, a chromodomain protein that anchors H3K9-methylated chromatin at the nuclear envelope (NE), suppressed the muscle-associated disease phenotypes. Deletion of cec-4 also rescued LMN-Y59C-linked alterations in chromatin organization and some changes in transcription. Sequences that changed position in the LMN-Y59C mutant, are enriched for E2F (EFL-2)-binding sites, consistent with previous studies suggesting that altered Rb-E2F interaction with lamin A may contribute to muscle dysfunction. In summary, we were able to counteract the dominant muscle-specific defects provoked by LMNA mutation by the ablation of a lamin-associated H3K9me anchor, suggesting a novel therapeutic pathway for EDMD.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Cell Nucleus/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Deletion , Muscular Dystrophy, Emery-Dreifuss/genetics , Animals , Binding Sites/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Nucleus/pathology , Chromatin/genetics , Disease Models, Animal , Genome, Helminth/genetics , Laminin/genetics , Laminin/metabolism , Muscles/physiopathology , Muscular Dystrophy, Emery-Dreifuss/physiopathology , Mutation , Protein Structure, Tertiary/genetics , Sarcomeres/chemistry , Sarcomeres/genetics , Transcription, Genetic/genetics
10.
Cell ; 150(5): 934-47, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939621

ABSTRACT

The factors that sequester transcriptionally repressed heterochromatin at the nuclear periphery are currently unknown. In a genome-wide RNAi screen, we found that depletion of S-adenosylmethionine (SAM) synthetase reduces histone methylation globally and causes derepression and release of heterochromatin from the nuclear periphery in Caenorhabditis elegans embryos. Analysis of histone methyltransferases (HMTs) showed that elimination of two HMTs, MET-2 and SET-25, mimics the loss of SAM synthetase, abrogating the perinuclear attachment of heterochromatic transgenes and of native chromosomal arms rich in histone H3 lysine 9 methylation. The two HMTs target H3K9 in a consecutive fashion: MET-2, a SETDB1 homolog, mediates mono- and dimethylation, and SET-25, a previously uncharacterized HMT, deposits H3K9me3. SET-25 colocalizes with its own product in perinuclear foci, in a manner dependent on H3K9me3, but not on its catalytic domain. This colocalization suggests an autonomous, self-reinforcing mechanism for the establishment and propagation of repeat-rich heterochromatin.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Cell Nucleus/chemistry , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , Chromosomes/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Genome, Helminth , Histone-Lysine N-Methyltransferase/analysis , Histone-Lysine N-Methyltransferase/genetics , Lamins/metabolism , Methionine Adenosyltransferase/metabolism , Methylation , Mutation
11.
Genes Dev ; 33(7-8): 436-451, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30804228

ABSTRACT

Caenorhabditis elegans has two histone H3 Lys9 methyltransferases, MET-2 (SETDB1 homolog) and SET-25 (G9a/SUV39H1 related). In worms, we found simple repeat sequences primarily marked by H3K9me2, while transposable elements and silent tissue-specific genes bear H3K9me3. RNA sequencing (RNA-seq) in histone methyltransferase (HMT) mutants shows that MET-2-mediated H3K9me2 is necessary for satellite repeat repression, while SET-25 silences a subset of transposable elements and tissue-specific genes through H3K9me3. A genome-wide synthetic lethality screen showed that RNA processing, nuclear RNA degradation, the BRCA1/BARD1 complex, and factors mediating replication stress survival are necessary for germline viability in worms lacking MET-2 but not SET-25. Unlike set-25 mutants, met-2-null worms accumulated satellite repeat transcripts, which form RNA:DNA hybrids on repetitive sequences, additively with the loss of BRCA1 or BARD1. BRCA1/BARD1-mediated H2A ubiquitination and MET-2 deposited H3K9me2 on satellite repeats are partially interdependent, suggesting both that the loss of silencing generates BRCA-recruiting DNA damage and that BRCA1 recruitment by damage helps silence repeats. The artificial induction of MSAT1 transcripts can itself trigger damage-induced germline lethality in a wild-type background, arguing that the synthetic sterility upon BRCA1/BARD1 and H3K9me2 loss is directly linked to the DNA damage provoked by unscheduled satellite repeat transcription.


Subject(s)
BRCA1 Protein/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental/genetics , Histones/genetics , Animals , BRCA1 Protein/metabolism , Caenorhabditis elegans Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Transposable Elements/genetics , Embryo, Nonmammalian , Fertility/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Microsatellite Repeats/genetics , Mutation , RNA Processing, Post-Transcriptional/genetics , Temperature
12.
Annu Rev Genet ; 52: 295-319, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30208290

ABSTRACT

Recent advances in both the technologies used to measure chromatin movement and the biophysical analysis used to model them have yielded a fuller understanding of chromatin dynamics and the polymer structure that underlies it. Changes in nucleosome packing, checkpoint kinase activation, the cell cycle, chromosomal tethers, and external forces acting on nuclei in response to external and internal stimuli can alter the basal mobility of DNA in interphase nuclei of yeast or mammalian cells. Although chromatin movement is assumed to be necessary for many DNA-based processes, including gene activation by distal enhancer-promoter interaction or sequence-based homology searches during double-strand break repair, experimental evidence supporting an essential role in these activities is sparse. Nonetheless, high-resolution tracking of chromatin dynamics has led to instructive models of the higher-order folding and flexibility of the chromatin polymer. Key regulators of chromatin motion in physiological conditions or after damage induction are reviewed here.


Subject(s)
Cell Nucleus/genetics , Chromatin/genetics , Chromosomes/genetics , Nucleosomes/genetics , Animals , Cell Nucleus/chemistry , Chromatin/chemistry , Chromosomes/chemistry , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Repair/genetics , Mammals/genetics , Nucleosomes/chemistry , Saccharomyces cerevisiae/genetics
13.
Nature ; 587(7834): 377-386, 2020 11.
Article in English | MEDLINE | ID: mdl-32894860

ABSTRACT

Here we describe the LifeTime Initiative, which aims to track, understand and target human cells during the onset and progression of complex diseases, and to analyse their response to therapy at single-cell resolution. This mission will be implemented through the development, integration and application of single-cell multi-omics and imaging, artificial intelligence and patient-derived experimental disease models during the progression from health to disease. The analysis of large molecular and clinical datasets will identify molecular mechanisms, create predictive computational models of disease progression, and reveal new drug targets and therapies. The timely detection and interception of disease embedded in an ethical and patient-centred vision will be achieved through interactions across academia, hospitals, patient associations, health data management systems and industry. The application of this strategy to key medical challenges in cancer, neurological and neuropsychiatric disorders, and infectious, chronic inflammatory and cardiovascular diseases at the single-cell level will usher in cell-based interceptive medicine in Europe over the next decade.


Subject(s)
Cell- and Tissue-Based Therapy , Delivery of Health Care/methods , Delivery of Health Care/trends , Medicine/methods , Medicine/trends , Pathology , Single-Cell Analysis , Artificial Intelligence , Delivery of Health Care/ethics , Delivery of Health Care/standards , Early Diagnosis , Education, Medical , Europe , Female , Health , Humans , Legislation, Medical , Male , Medicine/standards
14.
Genes Dev ; 37(1-2): 25-26, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-37061994
15.
EMBO J ; 40(21): e108439, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34569643

ABSTRACT

Upon replication stress, budding yeast checkpoint kinase Mec1ATR triggers the downregulation of transcription, thereby reducing the level of RNA polymerase (RNAP) on chromatin to facilitate replication fork progression. Here, we identify a hydroxyurea-induced phosphorylation site on Mec1, Mec1-S1991, that contributes to the eviction of RNAPII and RNAPIII during replication stress. The expression of the non-phosphorylatable mec1-S1991A mutant reduces replication fork progression genome-wide and compromises survival on hydroxyurea. This defect can be suppressed by destabilizing chromatin-bound RNAPII through a TAP fusion to its Rpb3 subunit, suggesting that lethality in mec1-S1991A mutants arises from replication-transcription conflicts. Coincident with a failure to repress gene expression on hydroxyurea in mec1-S1991A cells, highly transcribed genes such as GAL1 remain bound at nuclear pores. Consistently, we find that nuclear pore proteins and factors controlling RNAPII and RNAPIII are phosphorylated in a Mec1-dependent manner on hydroxyurea. Moreover, we show that Mec1 kinase also contributes to reduced RNAPII occupancy on chromatin during an unperturbed S phase by promoting degradation of the Rpb1 subunit.


Subject(s)
DNA Replication , Intracellular Signaling Peptides and Proteins/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , RNA Polymerase III/genetics , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Chromatin/chemistry , Chromatin/drug effects , Chromatin/metabolism , Galactokinase/genetics , Galactokinase/metabolism , Gene Expression Regulation, Fungal , Hydroxyurea/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Phosphoproteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Polymerase II/metabolism , RNA Polymerase III/metabolism , S Phase/drug effects , S Phase/genetics , Saccharomyces cerevisiae/genetics , Stress, Physiological/drug effects , Stress, Physiological/genetics , Transcription, Genetic
16.
PLoS Biol ; 20(4): e3001623, 2022 04.
Article in English | MEDLINE | ID: mdl-35452449

ABSTRACT

Molecular biology holds a vast potential for tackling climate change and biodiversity loss. Yet, it is largely absent from the current strategies. We call for a community-wide action to bring molecular biology to the forefront of climate change solutions.


Subject(s)
Biodiversity , Climate Change , Ecosystem , Molecular Biology
17.
Nature ; 569(7758): 734-739, 2019 05.
Article in English | MEDLINE | ID: mdl-31118512

ABSTRACT

The execution of developmental programs of gene expression requires an accurate partitioning of the genome into subnuclear compartments, with active euchromatin enriched centrally and silent heterochromatin at the nuclear periphery1. The existence of degenerative diseases linked to lamin A mutations suggests that perinuclear binding of chromatin contributes to cell-type integrity2,3. The methylation of lysine 9 of histone H3 (H3K9me) characterizes heterochromatin and mediates both transcriptional repression and chromatin anchoring at the inner nuclear membrane4. In Caenorhabditis elegans embryos, chromodomain protein CEC-4 bound to the inner nuclear membrane tethers heterochromatin through H3K9me3,5, whereas in differentiated tissues, a second heterochromatin-sequestering pathway is induced. Here we use an RNA interference screen in the cec-4 background and identify MRG-1 as a broadly expressed factor that is necessary for this second chromatin anchor in intestinal cells. However, MRG-1 is exclusively bound to euchromatin, suggesting that it acts indirectly. Heterochromatin detachment in double mrg-1; cec-4 mutants is rescued by depleting the histone acetyltransferase CBP-1/p300 or the transcription factor ATF-8, a member of the bZIP family (which is known to recruit CBP/p300). Overexpression of CBP-1 in cec-4 mutants is sufficient to delocalize heterochromatin in an ATF-8-dependent manner. CBP-1 and H3K27ac levels increase in heterochromatin upon mrg-1 knockdown, coincident with delocalization. This suggests that the spatial organization of chromatin in C. elegans is regulated both by the direct perinuclear attachment of silent chromatin, and by an active retention of CBP-1/p300 in euchromatin. The two pathways contribute differentially in embryos and larval tissues, with CBP-1 sequestration by MRG-1 having a major role in differentiated cells.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Chromatin/genetics , Chromatin/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Animals , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Euchromatin/genetics , Euchromatin/metabolism , Gain of Function Mutation , Genes, Reporter/genetics , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/metabolism , Intestines/cytology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Mol Cell ; 68(2): 431-445.e5, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29033322

ABSTRACT

Mec1-Ddc2 (ATR-ATRIP) is a key DNA-damage-sensing kinase that is recruited through the single-stranded (ss) DNA-binding replication protein A (RPA) to initiate the DNA damage checkpoint response. Activation of ATR-ATRIP in the absence of DNA damage is lethal. Therefore, it is important that damage-specific recruitment precedes kinase activation, which is achieved at least in part by Mec1-Ddc2 homodimerization. Here, we report a structural, biochemical, and functional characterization of the yeast Mec1-Ddc2-RPA assembly. High-resolution co-crystal structures of Ddc2-Rfa1 and Ddc2-Rfa1-t11 (K45E mutant) N termini and of the Ddc2 coiled-coil domain (CCD) provide insight into Mec1-Ddc2 homodimerization and damage-site targeting. Based on our structural and functional findings, we present a Mec1-Ddc2-RPA-ssDNA composite structural model. By way of validation, we show that RPA-dependent recruitment of Mec1-Ddc2 is crucial for maintaining its homodimeric state at ssDNA and that Ddc2's recruitment domain and CCD are important for Mec1-dependent survival of UV-light-induced DNA damage.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Cell Cycle Proteins/chemistry , DNA, Fungal/chemistry , DNA, Single-Stranded/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Protein Serine-Threonine Kinases/chemistry , Replication Protein A/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation, Missense , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , Protein Structure, Secondary , Replication Protein A/genetics , Replication Protein A/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
19.
Genes Dev ; 31(22): 2204-2221, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29284710

ABSTRACT

Chromatin is organized into higher-order structures that form subcompartments in interphase nuclei. Different categories of specialized enzymes act on chromatin and regulate its compaction and biophysical characteristics in response to physiological conditions. We present an overview of the function of chromatin structure and its dynamic changes in response to genotoxic stress, focusing on both subnuclear organization and the physical mobility of DNA. We review the requirements and mechanisms that cause chromatin relocation, enhanced mobility, and chromatin unfolding as a consequence of genotoxic lesions. An intriguing link has been established recently between enhanced chromatin dynamics and histone loss.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , DNA Damage , DNA Repair , Nucleosomes/metabolism , Histones/metabolism , Nucleosomes/chemistry
20.
Trends Genet ; 37(5): 476-487, 2021 05.
Article in English | MEDLINE | ID: mdl-33608117

ABSTRACT

Recent structural analysis of Fe-S centers in replication proteins and insights into the structure and function of DNA polymerase δ (DNA Pol δ) subunits have shed light on the key role played by this polymerase at replication forks under stress. The sequencing of cancer genomes reveals multiple point mutations that compromise the activity of POLD1, the DNA Pol δ catalytic subunit, whereas the loci encoding the accessory subunits POLD2 and POLD3 are amplified in a very high proportion of human tumors. Consistently, DNA Pol δ is key for the survival of replication stress and is involved in multiple long-patch repair pathways. Synthetic lethality arises from compromising the function and availability of the noncatalytic subunits of DNA Pol δ under conditions of replication stress, opening the door to novel therapies.


Subject(s)
DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , DNA Replication/physiology , Neoplasms/genetics , Animals , DNA Polymerase III/genetics , DNA Repair , Humans , Hydroxyurea/chemistry , Neoplasms/pathology , Oncogenes
SELECTION OF CITATIONS
SEARCH DETAIL