Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.078
Filter
Add more filters

Publication year range
1.
Cell ; 169(4): 679-692.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475896

ABSTRACT

The nuclear RNA exosome is an essential multi-subunit complex that controls RNA homeostasis. Congenital mutations in RNA exosome genes are associated with neurodegenerative diseases. Little is known about the role of the RNA exosome in the cellular response to pathogens. Here, using NGS and human and mouse genetics, we show that influenza A virus (IAV) ribogenesis and growth are suppressed by impaired RNA exosome activity. Mechanistically, the nuclear RNA exosome coordinates the initial steps of viral transcription with RNAPII at host promoters. The viral polymerase complex co-opts the nuclear RNA exosome complex and cellular RNAs en route to 3' end degradation. Exosome deficiency uncouples chromatin targeting of the viral polymerase complex and the formation of cellular:viral RNA hybrids, which are essential RNA intermediates that license transcription of antisense genomic viral RNAs. Our results suggest that evolutionary arms races have shaped the cellular RNA quality control machinery.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/virology , RNA Polymerase II/metabolism , A549 Cells , Animals , Chromatin Immunoprecipitation , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/metabolism , Humans , Mass Spectrometry , Mice , Mutation , Neurodegenerative Diseases/virology , RNA-Binding Proteins/genetics , Ribosomes/genetics , Transcription, Genetic
2.
Cell ; 167(4): 1001-1013.e7, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27881299

ABSTRACT

RNA-DNA hybrids are a major internal cause of DNA damage within cells, and their degradation by RNase H enzymes is important for maintaining genomic stability. Here, we identified an unexpected role for RNA-DNA hybrids and RNase H enzymes in DNA repair. Using a site-specific DNA double-strand break (DSB) system in Schizosaccharomyces pombe, we showed that RNA-DNA hybrids form as part of the homologous-recombination (HR)-mediated DSB repair process and that RNase H enzymes are essential for their degradation and efficient completion of DNA repair. Deleting RNase H stabilizes RNA-DNA hybrids around DSB sites and strongly impairs recruitment of the ssDNA-binding RPA complex. In contrast, overexpressing RNase H1 destabilizes these hybrids, leading to excessive strand resection and RPA recruitment and to severe loss of repeat regions around DSBs. Our study challenges the existing model of HR-mediated DSB repair and reveals a surprising role for RNA-DNA hybrids in maintaining genomic stability.


Subject(s)
Genomic Instability , Recombinational DNA Repair , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , DNA/metabolism , DNA Damage , Gene Expression , RNA/metabolism , RNA Polymerase II/metabolism , Ribonuclease H/genetics , Ribonuclease H/metabolism , Schizosaccharomyces/enzymology
3.
Mol Cell ; 82(21): 3985-4000.e4, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36265486

ABSTRACT

Alternative lengthening of telomeres (ALT), a telomerase-independent process maintaining telomeres, is mediated by break-induced replication (BIR). RAD52 promotes ALT by facilitating D-loop formation, but ALT also occurs through a RAD52-independent BIR pathway. Here, we show that the telomere non-coding RNA TERRA forms dynamic telomeric R-loops and contributes to ALT activity in RAD52 knockout cells. TERRA forms R-loops in vitro and at telomeres in a RAD51AP1-dependent manner. The formation of R-loops by TERRA increases G-quadruplexes (G4s) at telomeres. G4 stabilization enhances ALT even when TERRA is depleted, suggesting that G4s act downstream of R-loops to promote BIR. In vitro, the telomeric R-loops assembled by TERRA and RAD51AP1 generate G4s, which persist after R-loop resolution and allow formation of telomeric D-loops without RAD52. Thus, the dynamic telomeric R-loops formed by TERRA and RAD51AP1 enable the RAD52-independent ALT pathway, and G4s orchestrate an R- to D-loop switch at telomeres to stimulate BIR.


Subject(s)
RNA, Long Noncoding , Telomerase , Telomere Homeostasis , Telomere/genetics , Telomere/metabolism , Telomerase/genetics , Telomerase/metabolism , R-Loop Structures/genetics , DNA Repair
4.
EMBO J ; 43(12): 2453-2485, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719994

ABSTRACT

Double-strand breaks (DSBs) are the most lethal form of DNA damage. Transcriptional activity at DSBs, as well as transcriptional repression around DSBs, are both required for efficient DNA repair. The chromatin landscape defines and coordinates these two opposing events. However, how the open and condensed chromatin architecture is regulated remains unclear. Here, we show that the GATAD2B-NuRD complex associates with DSBs in a transcription- and DNA:RNA hybrid-dependent manner, to promote histone deacetylation and chromatin condensation. This activity establishes a spatio-temporal boundary between open and closed chromatin, which is necessary for the correct termination of DNA end resection. The lack of the GATAD2B-NuRD complex leads to chromatin hyperrelaxation and extended DNA end resection, resulting in homologous recombination (HR) repair failure. Our results suggest that the GATAD2B-NuRD complex is a key coordinator of the dynamic interplay between transcription and the chromatin landscape, underscoring its biological significance in the RNA-dependent DNA damage response.


Subject(s)
Chromatin , DNA Breaks, Double-Stranded , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Chromatin/metabolism , Chromatin/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , RNA/metabolism , RNA/genetics , DNA Damage , DNA/metabolism , DNA/genetics , Animals , Humans , Transcription, Genetic , DNA Repair , Mice
5.
Mol Cell ; 79(3): 425-442.e7, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32615088

ABSTRACT

Double-strand breaks (DSBs) are the most deleterious DNA lesions, which, if left unrepaired, may lead to genome instability or cell death. Here, we report that, in response to DSBs, the RNA methyltransferase METTL3 is activated by ATM-mediated phosphorylation at S43. Phosphorylated METTL3 is then localized to DNA damage sites, where it methylates the N6 position of adenosine (m6A) in DNA damage-associated RNAs, which recruits the m6A reader protein YTHDC1 for protection. In this way, the METTL3-m6A-YTHDC1 axis modulates accumulation of DNA-RNA hybrids at DSBs sites, which then recruit RAD51 and BRCA1 for homologous recombination (HR)-mediated repair. METTL3-deficient cells display defective HR, accumulation of unrepaired DSBs, and genome instability. Accordingly, depletion of METTL3 significantly enhances the sensitivity of cancer cells and murine xenografts to DNA damage-based therapy. These findings uncover the function of METTL3 and YTHDC1 in HR-mediated DSB repair, which may have implications for cancer therapy.


Subject(s)
Adenosine/analogs & derivatives , Head and Neck Neoplasms/genetics , Methyltransferases/genetics , Nerve Tissue Proteins/genetics , RNA Splicing Factors/genetics , Recombinational DNA Repair/drug effects , Squamous Cell Carcinoma of Head and Neck/genetics , Adenosine/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Bleomycin/pharmacology , Cell Line, Tumor , DNA/genetics , DNA/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , HEK293 Cells , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Methyltransferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/metabolism , Nucleic Acid Hybridization , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing Factors/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Ribonuclease H/genetics , Ribonuclease H/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Survival Analysis , Xenograft Model Antitumor Assays
6.
Genes Dev ; 34(13-14): 863-864, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32611612

ABSTRACT

R loops arise from hybridization of RNA transcripts with template DNA during transcription. Unrepaired R loops lead to transcription-replication collisions, causing DNA damage and genomic instability. In this issue of Genes & Development, Pérez-Calero and colleagues (pp. 898-912) identify UAP56 as a cotranscriptional RNA-DNA helicase that unwinds R loops. They found that UAP56 helicase activity is required to remove R loops formed from different sources and prevent R-loop accumulation genome-wide at actively transcribed genes.


Subject(s)
Genome/genetics , R-Loop Structures/genetics , Transcription, Genetic/genetics , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Genomic Instability/genetics , Humans , K562 Cells
7.
Genes Dev ; 34(13-14): 898-912, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32439635

ABSTRACT

Nonscheduled R loops represent a major source of DNA damage and replication stress. Cells have different ways to prevent R-loop accumulation. One mechanism relies on the conserved THO complex in association with cotranscriptional RNA processing factors including the RNA-dependent ATPase UAP56/DDX39B and histone modifiers such as the SIN3 deacetylase in humans. We investigated the function of UAP56/DDX39B in R-loop removal. We show that UAP56 depletion causes R-loop accumulation, R-loop-mediated genome instability, and replication fork stalling. We demonstrate an RNA-DNA helicase activity in UAP56 and show that its overexpression suppresses R loops and genome instability induced by depleting five different unrelated factors. UAP56/DDX39B localizes to active chromatin and prevents the accumulation of RNA-DNA hybrids over the entire genome. We propose that, in addition to its RNA processing role, UAP56/DDX39B is a key helicase required to eliminate harmful cotranscriptional RNA structures that otherwise would block transcription and replication.


Subject(s)
DEAD-box RNA Helicases/metabolism , Genome/genetics , R-Loop Structures/genetics , Transcription, Genetic/genetics , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , Gene Expression/genetics , Genomic Instability/genetics , Humans , K562 Cells
8.
EMBO J ; 42(1): e111703, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36326837

ABSTRACT

EXD2 is a recently identified exonuclease that cleaves RNA and DNA in double-stranded (ds) forms. It thus serves as a model system for investigating the similarities and discrepancies between exoribonuclease and exodeoxyribonuclease activities and for understanding the nucleic acid (NA) unwinding-degradation coordination of an exonuclease. Here, using a single-molecule fluorescence resonance energy transfer (smFRET) approach, we show that despite stable binding to both substrates, EXD2 barely cleaves dsDNA and yet displays both exoribonuclease and exodeoxyribonuclease activities toward RNA-DNA hybrids with a cleavage preference for RNA. Unexpectedly, EXD2-mediated hybrid cleavage proceeds in a discrete stepwise pattern, wherein a sudden 4-bp duplex unwinding increment and the subsequent dwell constitute a complete hydrolysis cycle. The relatively weak exodeoxyribonuclease activity of EXD2 partially originates from frequent hybrid rewinding. Importantly, kinetic analysis and comparison of the dwell times under varied conditions reveal two rate-limiting steps of hybrid unwinding and nucleotide excision. Overall, our findings help better understand the cellular functions of EXD2, and the cyclic coupling between duplex unwinding and exonucleolytic degradation may be generalizable to other exonucleases.


Subject(s)
Exoribonucleases , RNA , RNA/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Kinetics , DNA/metabolism , Exodeoxyribonucleases/metabolism
9.
Annu Rev Microbiol ; 76: 461-480, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35655343

ABSTRACT

During the essential processes of DNA replication and transcription, RNA-DNA hybrid intermediates are formed that pose significant risks to genome integrity when left unresolved. To manage RNA-DNA hybrids, all cells rely on RNase H family enzymes that specifically cleave the RNA portion of the many different types of hybrids that form in vivo. Recent experimental advances have provided new insight into how RNA-DNA hybrids form and the consequences to genome integrity that ensue when persistent hybrids remain unresolved. Here we review the types of RNA-DNA hybrids, including R-loops, RNA primers, and ribonucleotide misincorporations, that form during DNA replication and transcription and discuss how each type of hybrid can contribute to genome instability in bacteria. Further, we discuss how bacterial RNase HI, HII, and HIII and bacterial FEN enzymes contribute to genome maintenance through the resolution of hybrids.


Subject(s)
Bacterial Proteins , Ribonucleases , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , DNA , DNA Replication , RNA/genetics , Ribonucleases/genetics , Ribonucleases/metabolism
10.
Mol Cell ; 76(1): 57-69.e9, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31519522

ABSTRACT

Although correlations between RNA polymerase II (RNAPII) transcription stress, R-loops, and genome instability have been established, the mechanisms underlying these connections remain poorly understood. Here, we used a mutant version of the transcription elongation factor TFIIS (TFIISmut), aiming to specifically induce increased levels of RNAPII pausing, arrest, and/or backtracking in human cells. Indeed, TFIISmut expression results in slower elongation rates, relative depletion of polymerases from the end of genes, and increased levels of stopped RNAPII; it affects mRNA splicing and termination as well. Remarkably, TFIISmut expression also dramatically increases R-loops, which may form at the anterior end of backtracked RNAPII and trigger genome instability, including DNA strand breaks. These results shed light on the relationship between transcription stress and R-loops and suggest that different classes of R-loops may exist, potentially with distinct consequences for genome stability.


Subject(s)
Genomic Instability , R-Loop Structures , RNA, Messenger/genetics , Stress, Physiological , Transcription, Genetic , Transcriptional Elongation Factors/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Mutation , RNA Polymerase II/metabolism , RNA Splicing , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Structure-Activity Relationship , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/genetics
11.
Mol Cell ; 73(4): 670-683.e12, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30639241

ABSTRACT

Cellular mechanisms that safeguard genome integrity are often subverted in cancer. To identify cancer-related genome caretakers, we employed a convergent multi-screening strategy coupled to quantitative image-based cytometry and ranked candidate genes according to multivariate readouts reflecting viability, proliferative capacity, replisome integrity, and DNA damage signaling. This unveiled regulators of replication stress resilience, including components of the pre-mRNA cleavage and polyadenylation complex. We show that deregulation of pre-mRNA cleavage impairs replication fork speed and leads to excessive origin activity, rendering cells highly dependent on ATR function. While excessive formation of RNA:DNA hybrids under these conditions was tightly associated with replication-stress-induced DNA damage, inhibition of transcription rescued fork speed, origin activation, and alleviated replication catastrophe. Uncoupling of pre-mRNA cleavage from co-transcriptional processing and export also protected cells from replication-stress-associated DNA damage, suggesting that pre-mRNA cleavage provides a mechanism to efficiently release nascent transcripts and thereby prevent gene gating-associated genomic instability.


Subject(s)
DNA Damage , DNA Replication , Genomic Instability , Neoplasms/genetics , RNA Cleavage , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Active Transport, Cell Nucleus , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Neoplasms/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Heteroduplexes/genetics , Nucleic Acid Heteroduplexes/metabolism , Polyadenylation , RNA Precursors/biosynthesis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA-Binding Proteins
12.
Genes Dev ; 33(15-16): 1008-1026, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31123061

ABSTRACT

Genome replication involves dealing with obstacles that can result from DNA damage but also from chromatin alterations, topological stress, tightly bound proteins or non-B DNA structures such as R loops. Experimental evidence reveals that an engaged transcription machinery at the DNA can either enhance such obstacles or be an obstacle itself. Thus, transcription can become a potentially hazardous process promoting localized replication fork hindrance and stress, which would ultimately cause genome instability, a hallmark of cancer cells. Understanding the causes behind transcription-replication conflicts as well as how the cell resolves them to sustain genome integrity is the aim of this review.


Subject(s)
DNA Replication/physiology , Genomic Instability/genetics , Transcription, Genetic/physiology , Genome/genetics , Humans , Neoplasms/physiopathology , Transcription Elongation, Genetic/physiology
13.
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
14.
EMBO J ; 41(22): e108040, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36215697

ABSTRACT

The ribonuclease DIS3 is one of the most frequently mutated genes in the hematological cancer multiple myeloma, yet the basis of its tumor suppressor function in this disease remains unclear. Herein, exploiting the TCGA dataset, we found that DIS3 plays a prominent role in the DNA damage response. DIS3 inactivation causes genomic instability by increasing mutational load, and a pervasive accumulation of DNA:RNA hybrids that induces genomic DNA double-strand breaks (DSBs). DNA:RNA hybrid accumulation also prevents binding of the homologous recombination (HR) machinery to double-strand breaks, hampering DSB repair. DIS3-inactivated cells become sensitive to PARP inhibitors, suggestive of a defect in homologous recombination repair. Accordingly, multiple myeloma patient cells mutated for DIS3 harbor an increased mutational burden and a pervasive overexpression of pro-inflammatory interferon, correlating with the accumulation of DNA:RNA hybrids. We propose DIS3 loss in myeloma to be a driving force for tumorigenesis via DNA:RNA hybrid-dependent enhanced genome instability and increased mutational rate. At the same time, DIS3 loss represents a liability that might be therapeutically exploited in patients whose cancer cells harbor DIS3 mutations.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Ribonucleases/metabolism , Recombinational DNA Repair , Homologous Recombination , Genomic Instability , DNA Repair , DNA/metabolism , RNA , Exosome Multienzyme Ribonuclease Complex/metabolism
15.
Mol Cell ; 71(4): 487-497.e3, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30078723

ABSTRACT

DNA-RNA hybrids associated with R-loops promote DNA damage and genomic instability. The capacity of hybrids at different genomic sites to cause DNA damage was not known, and the mechanisms leading from hybrid to damage were poorly understood. Here, we adopt a new strategy to map and characterize the sites of hybrid-induced damage genome-wide in budding yeast. We show that hybrid removal is essential for life because persistent hybrids cause irreparable DNA damage and cell death. We identify that a subset of hybrids is prone to cause damage, and the chromosomal context of hybrids dramatically impacts their ability to induce damage. Furthermore, persistent hybrids affect the repair pathway, generating large regions of single-stranded DNA (ssDNA) by two distinct mechanisms, likely resection and re-replication. These damaged regions may act as potential precursors to gross chromosomal rearrangements like deletions and duplications that are associated with R-loops and cancers.


Subject(s)
DNA, Single-Stranded/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Genomic Instability , RNA/genetics , Saccharomyces cerevisiae/genetics , DNA Cleavage , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Hydroxyurea/pharmacology , Indoleacetic Acids/pharmacology , Nucleic Acid Conformation , Nucleic Acid Hybridization , RNA/chemistry , RNA/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
16.
EMBO J ; 40(7): e106018, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33634895

ABSTRACT

The BRCA2 tumor suppressor is a DNA double-strand break (DSB) repair factor essential for maintaining genome integrity. BRCA2-deficient cells spontaneously accumulate DNA-RNA hybrids, a known source of genome instability. However, the specific role of BRCA2 on these structures remains poorly understood. Here we identified the DEAD-box RNA helicase DDX5 as a BRCA2-interacting protein. DDX5 associates with DNA-RNA hybrids that form in the vicinity of DSBs, and this association is enhanced by BRCA2. Notably, BRCA2 stimulates the DNA-RNA hybrid-unwinding activity of DDX5 helicase. An impaired BRCA2-DDX5 interaction, as observed in cells expressing the breast cancer variant BRCA2-T207A, reduces the association of DDX5 with DNA-RNA hybrids, decreases the number of RPA foci, and alters the kinetics of appearance of RAD51 foci upon irradiation. Our findings are consistent with DNA-RNA hybrids constituting an impediment for the repair of DSBs by homologous recombination and reveal BRCA2 and DDX5 as active players in their removal.


Subject(s)
BRCA2 Protein/metabolism , DEAD-box RNA Helicases/metabolism , Recombinational DNA Repair , BRCA2 Protein/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DNA Breaks, Double-Stranded , HEK293 Cells , Humans , Nucleic Acid Heteroduplexes , Protein Binding
17.
EMBO Rep ; 24(12): e57801, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37818834

ABSTRACT

Double-strand breaks (DSBs) are the most harmful DNA lesions, with a strong impact on cell proliferation and genome integrity. Depending on cell cycle stage, DSBs are preferentially repaired by non-homologous end joining or homologous recombination (HR). In recent years, numerous reports have revealed that DSBs enhance DNA-RNA hybrid formation around the break site. We call these hybrids "break-induced RNA-DNA hybrids" (BIRDHs) to differentiate them from sporadic R-loops consisting of DNA-RNA hybrids and a displaced single-strand DNA occurring co-transcriptionally in intact DNA. Here, we review and discuss the most relevant data about BIRDHs, with a focus on two main questions raised: (i) whether BIRDHs form by de novo transcription after a DSB or by a pre-existing nascent RNA in DNA regions undergoing transcription and (ii) whether they have a positive role in HR or are just obstacles to HR accidentally generated as an intrinsic risk of transcription. We aim to provide a comprehensive view of the exciting and yet unresolved questions about the source and impact of BIRDHs in the cell.


Subject(s)
DNA Breaks, Double-Stranded , RNA , RNA/genetics , Homologous Recombination , DNA Repair , DNA/genetics , DNA End-Joining Repair
18.
Mol Cell ; 66(5): 597-609.e5, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575656

ABSTRACT

R loops have positive physiological roles, but they can also be deleterious by causing genome instability, and the mechanisms for this are unknown. Here we identified yeast histone H3 and H4 mutations that facilitate R loops but do not cause instability. R loops containing single-stranded DNA (ssDNA), versus RNA-DNA hybrids alone, were demonstrated using ssDNA-specific human AID and bisulfite. Notably, they are similar size regardless of whether or not they induce genome instability. Contrary to mutants causing R loop-mediated instability, these histone mutants do not accumulate H3 serine-10 phosphate (H3S10-P). We propose a two-step mechanism in which, first, an altered chromatin facilitates R loops, and second, chromatin is modified, including H3S10-P, as a requisite for compromising genome integrity. Consistently, these histone mutations suppress the high H3S10 phosphorylation and genomic instability of hpr1 and sen1 mutants. Therefore, contrary to what was previously believed, R loops do not cause genome instability by themselves.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/genetics , DNA, Fungal/genetics , Genome, Fungal , Genomic Instability , Histones/genetics , Point Mutation , RNA, Fungal/genetics , Saccharomyces cerevisiae/genetics , Chromatin/chemistry , Chromatin/metabolism , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA, Fungal/chemistry , DNA, Fungal/metabolism , Histones/chemistry , Histones/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Fungal/chemistry , RNA, Fungal/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
19.
Proc Natl Acad Sci U S A ; 119(18): e2115638119, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35476521

ABSTRACT

A key property of adult stem cells is their ability to persist in a quiescent state for prolonged periods of time. The quiescent state is thought to contribute to stem cell resilience by limiting accumulation of DNA replication­associated mutations. Moreover, cellular stress response factors are thought to play a role in maintaining quiescence and stem cell integrity. We utilized muscle stem cells (MuSCs) as a model of quiescent stem cells and find that the replication stress response protein, ATR (Ataxia Telangiectasia and Rad3-Related), is abundant and active in quiescent but not activated MuSCs. Concurrently, MuSCs display punctate RPA (replication protein A) and R-loop foci, both key triggers for ATR activation. To discern the role of ATR in MuSCs, we generated MuSC-specific ATR conditional knockout (ATRcKO) mice. Surprisingly, ATR ablation results in increased MuSC quiescence exit. Phosphoproteomic analysis of ATRcKO MuSCs reveals enrichment of phosphorylated cyclin F, a key component of the Skp1­Cul1­F-box protein (SCF) ubiquitin ligase complex and regulator of key cell-cycle transition factors, such as the E2F family of transcription factors. Knocking down cyclin F or inhibiting the SCF complex results in E2F1 accumulation and in MuSCs exiting quiescence, similar to ATR-deficient MuSCs. The loss of ATR could be counteracted by inhibiting casein kinase 2 (CK2), the kinase responsible for phosphorylating cyclin F. We propose a model in which MuSCs express cell-cycle progression factors but ATR, in coordination with the cyclin F­SCF complex, represses premature stem cell quiescence exit via ubiquitin­proteasome degradation of these factors.


Subject(s)
Cell Cycle Proteins , Cyclins , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Division , Cyclins/genetics , Cyclins/metabolism , Stem Cells/metabolism
20.
Genes Dev ; 31(4): 347-352, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28289143

ABSTRACT

Wilms' tumor 1 (WT1) is essential for the development and homeostasis of multiple mesodermal tissues. Despite evidence for post-transcriptional roles, no endogenous WT1 target RNAs exist. Using RNA immunoprecipitation and UV cross-linking, we show that WT1 binds preferentially to 3' untranslated regions (UTRs) of developmental targets. These target mRNAs are down-regulated upon WT1 depletion in cell culture and developing kidney mesenchyme. Wt1 deletion leads to rapid turnover of specific mRNAs. WT1 regulates reporter gene expression through interaction with 3' UTR-binding sites. Combining experimental and computational analyses, we propose that WT1 influences key developmental and disease processes in part through regulating mRNA turnover.


Subject(s)
3' Untranslated Regions/physiology , Gene Expression Regulation, Developmental/genetics , RNA, Messenger/genetics , Wilms Tumor/genetics , Wilms Tumor/metabolism , Animals , Cell Line , Down-Regulation , Gene Deletion , Kidney/cytology , Mesoderm/metabolism , Mice , Mouse Embryonic Stem Cells , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL