Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Cell ; 179(3): 604-618, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31607512

ABSTRACT

DNA-RNA hybrids play a physiological role in cellular processes, but often, they represent non-scheduled co-transcriptional structures with a negative impact on transcription, replication and DNA repair. Accumulating evidence suggests that they constitute a source of replication stress, DNA breaks and genome instability. Reciprocally, DNA breaks facilitate DNA-RNA hybrid formation by releasing the double helix torsional conformation. Cells avoid DNA-RNA accumulation by either preventing or removing hybrids directly or by DNA repair-coupled mechanisms. Given the R-loop impact on chromatin and genome organization and its potential relation with genetic diseases, we review R-loop homeostasis as well as their physiological and pathological roles.


Subject(s)
DNA/genetics , Nucleic Acid Conformation , R-Loop Structures/genetics , RNA/genetics , Chromatin/chemistry , Chromatin/genetics , DNA/chemistry , DNA Breaks , DNA Repair/genetics , DNA Replication/genetics , Genomic Instability/genetics , Homeostasis/genetics , Humans , RNA/chemistry , Transcription, Genetic
2.
Nat Rev Mol Cell Biol ; 21(3): 167-178, 2020 03.
Article in English | MEDLINE | ID: mdl-32005969

ABSTRACT

R-loops are three-stranded structures that harbour an RNA-DNA hybrid and frequently form during transcription. R-loop misregulation is associated with DNA damage, transcription elongation defects, hyper-recombination and genome instability. In contrast to such 'unscheduled' R-loops, evidence is mounting that cells harness the presence of RNA-DNA hybrids in scheduled, 'regulatory' R-loops to promote DNA transactions, including transcription termination and other steps of gene regulation, telomere stability and DNA repair. R-loops formed by cellular RNAs can regulate histone post-translational modification and may be recognized by dedicated reader proteins. The two-faced nature of R-loops implies that their formation, location and timely removal must be tightly regulated. In this Perspective, we discuss the cellular processes that regulatory R-loops modulate, the regulation of R-loops and the potential differences that may exist between regulatory R-loops and unscheduled R-loops.


Subject(s)
DNA/chemistry , Genomic Instability/genetics , R-Loop Structures/genetics , Animals , DNA/genetics , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Replication/genetics , DNA Replication/physiology , Gene Expression Regulation/genetics , Histone Code/genetics , Humans , Nucleic Acid Conformation , R-Loop Structures/physiology , RNA/chemistry , RNA/genetics , Telomere/genetics , Transcription, Genetic/genetics
3.
Mol Cell ; 84(16): 3141-3153.e5, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39047725

ABSTRACT

The metagenome-derived type I-E and type I-F variant CRISPR-associated complex for antiviral defense (Cascade) complexes, fused with HNH domains, precisely cleave target DNA, representing recently identified genome editing tools. However, the underlying working mechanisms remain unknown. Here, structures of type I-FHNH and I-EHNH Cascade complexes at different states are reported. In type I-FHNH Cascade, Cas8fHNH loosely attaches to Cascade head and is adjacent to the 5' end of the target single-stranded DNA (ssDNA). Formation of the full R-loop drives the Cascade head to move outward, allowing Cas8fHNH to detach and rotate ∼150° to accommodate target ssDNA for cleavage. In type I-EHNH Cascade, Cas5eHNH domain is adjacent to the 5' end of the target ssDNA. Full crRNA-target pairing drives the lift of the Cascade head, widening the substrate channel for target ssDNA entrance. Altogether, these analyses into both complexes revealed that crRNA-guided positioning of target DNA and target DNA-induced HNH unlocking are two key factors for their site-specific cleavage of target DNA.


Subject(s)
CRISPR-Associated Proteins , CRISPR-Cas Systems , DNA Cleavage , DNA, Single-Stranded , Gene Editing , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics , Gene Editing/methods , R-Loop Structures/genetics , Cryoelectron Microscopy
4.
Mol Cell ; 84(14): 2717-2731.e6, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38955179

ABSTRACT

The specific nature of CRISPR-Cas12a makes it a desirable RNA-guided endonuclease for biotechnology and therapeutic applications. To understand how R-loop formation within the compact Cas12a enables target recognition and nuclease activation, we used cryo-electron microscopy to capture wild-type Acidaminococcus sp. Cas12a R-loop intermediates and DNA delivery into the RuvC active site. Stages of Cas12a R-loop formation-starting from a 5-bp seed-are marked by distinct REC domain arrangements. Dramatic domain flexibility limits contacts until nearly complete R-loop formation, when the non-target strand is pulled across the RuvC nuclease and coordinated domain docking promotes efficient cleavage. Next, substantial domain movements enable target strand repositioning into the RuvC active site. Between cleavage events, the RuvC lid conformationally resets to occlude the active site, requiring re-activation. These snapshots build a structural model depicting Cas12a DNA targeting that rationalizes observed specificity and highlights mechanistic comparisons to other class 2 effectors.


Subject(s)
Acidaminococcus , Bacterial Proteins , CRISPR-Associated Proteins , CRISPR-Cas Systems , Catalytic Domain , Cryoelectron Microscopy , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/genetics , Acidaminococcus/enzymology , Acidaminococcus/genetics , Acidaminococcus/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , R-Loop Structures/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/chemistry , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , Models, Molecular , Protein Domains , Structure-Activity Relationship , Protein Binding
5.
Mol Cell ; 83(20): 3707-3719.e5, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37827159

ABSTRACT

R-loops, which consist of a DNA-RNA hybrid and a displaced DNA strand, are known to threaten genome integrity. To counteract this, different mechanisms suppress R-loop accumulation by either preventing the hybridization of RNA with the DNA template (RNA biogenesis factors), unwinding the hybrid (DNA-RNA helicases), or degrading the RNA moiety of the R-loop (type H ribonucleases [RNases H]). Thus far, RNases H are the only nucleases known to cleave DNA-RNA hybrids. Now, we show that the RNase DICER also resolves R-loops. Biochemical analysis reveals that DICER acts by specifically cleaving the RNA within R-loops. Importantly, a DICER RNase mutant impaired in R-loop processing causes a strong accumulation of R-loops in cells. Our results thus not only reveal a function of DICER as an R-loop resolvase independent of DROSHA but also provide evidence for the role of multi-functional RNA processing factors in the maintenance of genome integrity in higher eukaryotes.


Subject(s)
R-Loop Structures , Ribonucleases , Humans , R-Loop Structures/genetics , Ribonucleases/genetics , RNA/genetics , DNA , DNA Replication , DNA Helicases/genetics , Ribonuclease H/genetics , Ribonuclease H/metabolism , Genomic Instability
6.
Mol Cell ; 82(12): 2267-2297, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35508167

ABSTRACT

Although transcription is an essential cellular process, it is paradoxically also a well-recognized cause of genomic instability. R-loops, non-B DNA structures formed when nascent RNA hybridizes to DNA to displace the non-template strand as single-stranded DNA (ssDNA), are partially responsible for this instability. Yet, recent work has begun to elucidate regulatory roles for R-loops in maintaining the genome. In this review, we discuss the cellular contexts in which R-loops contribute to genomic instability, particularly during DNA replication and double-strand break (DSB) repair. We also summarize the evidence that R-loops participate as an intermediate during repair and may influence pathway choice to preserve genomic integrity. Finally, we discuss the immunogenic potential of R-loops and highlight their links to disease should they become pathogenic.


Subject(s)
R-Loop Structures , Transcription, Genetic , DNA/metabolism , DNA Repair , DNA Replication , DNA, Single-Stranded/genetics , Genomic Instability , Humans , R-Loop Structures/genetics
7.
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
8.
Nature ; 621(7979): 610-619, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37557913

ABSTRACT

The proper regulation of transcription is essential for maintaining genome integrity and executing other downstream cellular functions1,2. Here we identify a stable association between the genome-stability regulator sensor of single-stranded DNA (SOSS)3 and the transcription regulator Integrator-PP2A (INTAC)4-6. Through SSB1-mediated recognition of single-stranded DNA, SOSS-INTAC stimulates promoter-proximal termination of transcription and attenuates R-loops associated with paused RNA polymerase II to prevent R-loop-induced genome instability. SOSS-INTAC-dependent attenuation of R-loops is enhanced by the ability of SSB1 to form liquid-like condensates. Deletion of NABP2 (encoding SSB1) or introduction of cancer-associated mutations into its intrinsically disordered region leads to a pervasive accumulation of R-loops, highlighting a genome surveillance function of SOSS-INTAC that enables timely termination of transcription at promoters to constrain R-loop accumulation and ensure genome stability.


Subject(s)
Genomic Instability , Promoter Regions, Genetic , R-Loop Structures , Transcription Termination, Genetic , Humans , DNA, Single-Stranded/metabolism , Genomic Instability/genetics , Mutation , R-Loop Structures/genetics , RNA Polymerase II/metabolism , Promoter Regions, Genetic/genetics , Genome, Human , DNA-Binding Proteins/metabolism
9.
EMBO J ; 43(14): 3044-3071, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38858601

ABSTRACT

MCM8 has emerged as a core gene in reproductive aging and is crucial for meiotic homologous recombination repair. It also safeguards genome stability by coordinating the replication stress response during mitosis, but its function in mitotic germ cells remains elusive. Here we found that disabling MCM8 in mice resulted in proliferation defects of primordial germ cells (PGCs) and ultimately impaired fertility. We further demonstrated that MCM8 interacted with two known helicases DDX5 and DHX9, and loss of MCM8 led to R-loop accumulation by reducing the retention of these helicases at R-loops, thus inducing genome instability. Cells expressing premature ovarian insufficiency-causative mutants of MCM8 with decreased interaction with DDX5 displayed increased R-loop levels. These results show MCM8 interacts with R-loop-resolving factors to prevent R-loop-induced DNA damage, which may contribute to the maintenance of genome integrity of PGCs and reproductive reserve establishment. Our findings thus reveal an essential role for MCM8 in PGC development and improve our understanding of reproductive aging caused by genome instability in mitotic germ cells.


Subject(s)
DEAD-box RNA Helicases , Genomic Instability , Minichromosome Maintenance Proteins , R-Loop Structures , Animals , Female , Humans , Male , Mice , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , DNA Damage , Germ Cells/metabolism , Minichromosome Maintenance Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , R-Loop Structures/genetics
10.
Mol Cell ; 77(3): 514-527.e4, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31708417

ABSTRACT

R loops arising during transcription induce genomic instability, but how cells respond to the R loop-associated genomic stress is still poorly understood. Here, we show that cells harboring high levels of R loops rely on the ATR kinase for survival. In response to aberrant R loop accumulation, the ataxia telangiectasia and Rad3-related (ATR)-Chk1 pathway is activated by R loop-induced reversed replication forks. In contrast to the activation of ATR by replication inhibitors, R loop-induced ATR activation requires the MUS81 endonuclease. ATR protects the genome from R loops by suppressing transcription-replication collisions, promoting replication fork recovery, and enforcing a G2/M cell-cycle arrest. Furthermore, ATR prevents excessive cleavage of reversed forks by MUS81, revealing a MUS81-triggered and ATR-mediated feedback loop that fine-tunes MUS81 activity at replication forks. These results suggest that ATR is a key sensor and suppressor of R loop-induced genomic instability, uncovering a signaling circuitry that safeguards the genome against R loops.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , R-Loop Structures/genetics , Ataxia Telangiectasia Mutated Proteins/physiology , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/genetics , DNA Damage , DNA Repair , DNA Replication/genetics , DNA Replication/physiology , DNA-Binding Proteins/genetics , Endonucleases/genetics , Genomic Instability/physiology , HeLa Cells , Humans , Phosphorylation , Protein Kinases/metabolism , Signal Transduction
11.
Mol Cell ; 77(3): 528-541.e8, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31759821

ABSTRACT

Formation of co-transcriptional R-loops underlies replication fork stalling upon head-on transcription-replication encounters. Here, we demonstrate that RAD51-dependent replication fork reversal induced by R-loops is followed by the restart of semiconservative DNA replication mediated by RECQ1 and RECQ5 helicases, MUS81/EME1 endonuclease, RAD52 strand-annealing factor, the DNA ligase IV (LIG4)/XRCC4 complex, and the non-catalytic subunit of DNA polymerase δ, POLD3. RECQ5 disrupts RAD51 filaments assembled on stalled forks after RECQ1-mediated reverse branch migration, preventing a new round of fork reversal and facilitating fork cleavage by MUS81/EME1. MUS81-dependent DNA breaks accumulate in cells lacking RAD52 or LIG4 upon induction of R-loop formation, suggesting that RAD52 acts in concert with LIG4/XRCC4 to catalyze fork religation, thereby mediating replication restart. The resumption of DNA synthesis after R-loop-associated fork stalling also requires active transcription, the restoration of which depends on MUS81, RAD52, LIG4, and the transcription elongation factor ELL. These findings provide mechanistic insights into transcription-replication conflict resolution.


Subject(s)
DNA Replication/physiology , R-Loop Structures/genetics , Rad51 Recombinase/metabolism , Cell Line, Tumor , DNA Ligases/metabolism , DNA Polymerase III/metabolism , DNA Replication/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Endonucleases/genetics , Endonucleases/metabolism , HeLa Cells , Humans , R-Loop Structures/physiology , Rad51 Recombinase/genetics , Rad51 Recombinase/physiology , Rad52 DNA Repair and Recombination Protein/metabolism , RecQ Helicases/metabolism , RecQ Helicases/physiology , Transcription, Genetic/genetics
12.
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
13.
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
14.
Nature ; 594(7862): 283-288, 2021 06.
Article in English | MEDLINE | ID: mdl-33981036

ABSTRACT

Homologous recombination (HR) repairs DNA double-strand breaks (DSBs) in the S and G2 phases of the cell cycle1-3. Several HR proteins are preferentially recruited to DSBs at transcriptionally active loci4-10, but how transcription promotes HR is poorly understood. Here we develop an assay to assess the effect of local transcription on HR. Using this assay, we find that transcription stimulates HR to a substantial extent. Tethering RNA transcripts to the vicinity of DSBs recapitulates the effects of local transcription, which suggests that transcription enhances HR through RNA transcripts. Tethered RNA transcripts stimulate HR in a sequence- and orientation-dependent manner, indicating that they function by forming DNA-RNA hybrids. In contrast to most HR proteins, RAD51-associated protein 1 (RAD51AP1) only promotes HR when local transcription is active. RAD51AP1 drives the formation of R-loops in vitro and is required for tethered RNAs to stimulate HR in cells. Notably, RAD51AP1 is necessary for the DSB-induced formation of DNA-RNA hybrids in donor DNA, linking R-loops to D-loops. In vitro, RAD51AP1-generated R-loops enhance the RAD51-mediated formation of D-loops locally and give rise to intermediates that we term 'DR-loops', which contain both DNA-DNA and DNA-RNA hybrids and favour RAD51 function. Thus, at DSBs in transcribed regions, RAD51AP1 promotes the invasion of RNA transcripts into donor DNA, and stimulates HR through the formation of DR-loops.


Subject(s)
DNA/genetics , DNA/metabolism , Homologous Recombination/genetics , R-Loop Structures/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , DNA/chemistry , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Genes/genetics , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , RNA, Messenger/chemistry , RNA-Binding Proteins/metabolism , Rad51 Recombinase/metabolism
15.
Nucleic Acids Res ; 52(7): 3623-3635, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38281203

ABSTRACT

Certain DNA sequences can adopt a non-B form in the genome that interfere with DNA-templated processes, including transcription. Among the sequences that are intrinsically difficult to transcribe are those that tend to form R-loops, three-stranded nucleic acid structures formed by a DNA-RNA hybrid and the displaced ssDNA. Here we compared the transcription of an endogenous gene with and without an R-loop-forming sequence inserted. We show that, in agreement with previous in vivo and in vitro analyses, transcription elongation is delayed by R-loops in yeast. Importantly, we demonstrate that the Rat1 transcription terminator factor facilitates transcription throughout such structures by inducing premature termination of arrested RNAPIIs. We propose that RNase H degrades the RNA moiety of the hybrid, providing an entry site for Rat1. Thus, we have uncovered an unanticipated function of Rat1 as a transcription restoring factor opening up the possibility that it may also promote transcription through other genomic DNA structures intrinsically difficult to transcribe. If R-loop-mediated transcriptional stress is not relieved by Rat1, it will cause genomic instability, probably through the increase of transcription-replication conflicts, a deleterious situation that could lead to cancer.


Subject(s)
Exoribonucleases , R-Loop Structures , Ribonuclease H , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Termination, Genetic , R-Loop Structures/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ribonuclease H/metabolism , Ribonuclease H/genetics , Saccharomyces cerevisiae/genetics , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic
16.
PLoS Genet ; 19(5): e1010754, 2023 05.
Article in English | MEDLINE | ID: mdl-37141391

ABSTRACT

The prototype enzymes of the ubiquitous type IA topoisomerases (topos) family are Escherichia coli topo I (topA) and topo III (topB). Topo I shows preference for relaxation of negative supercoiling and topo III for decatenation. However, as they could act as backups for each other or even share functions, strains lacking both enzymes must be used to reveal the roles of type IA enzymes in genome maintenance. Recently, marker frequency analysis (MFA) of genomic DNA from topA topB null mutants revealed a major RNase HI-sensitive DNA peak bordered by Ter/Tus barriers, sites of replication fork fusion and termination in the chromosome terminus region (Ter). Here, flow cytometry for R-loop-dependent replication (RLDR), MFA, R-loop detection with S9.6 antibodies, and microscopy were used to further characterize the mechanism and consequences of over-replication in Ter. It is shown that the Ter peak is not due to the presence of a strong origin for RLDR in Ter region; instead RLDR, which is partly inhibited by the backtracking-resistant rpoB*35 mutation, appears to contribute indirectly to Ter over-replication. The data suggest that RLDR from multiple sites on the chromosome increases the number of replication forks trapped at Ter/Tus barriers which leads to RecA-dependent DNA amplification in Ter and to a chromosome segregation defect. Overproducing topo IV, the main cellular decatenase, does not inhibit RLDR or Ter over-replication but corrects the chromosome segregation defect. Furthermore, our data suggest that the inhibition of RLDR by topo I does not require its C-terminal-mediated interaction with RNA polymerase. Overall, our data reveal a pathway of genomic instability triggered by R-loops and its regulation by various topos activities at different steps.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Humans , Escherichia coli/metabolism , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA Replication/genetics , R-Loop Structures/genetics , Genomic Instability/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism
17.
J Virol ; 98(8): e0100324, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39046232

ABSTRACT

Three-stranded DNA-RNA structures known as R-loops that form during papillomavirus transcription can cause transcription-replication conflicts and lead to DNA damage. We found that R-loops accumulated at the viral early promoter in human papillomavirus (HPV) episomal cells but were greatly reduced in cells with integrated HPV genomes. RNA-DNA helicases unwind R-loops and allow for transcription and replication to proceed. Depletion of the RNA-DNA helicase senataxin (SETX) using siRNAs increased the presence of R-loops at the viral early promoter in HPV-31 (CIN612) and HPV-16 (W12) episomal HPV cell lines. Depletion of SETX reduced viral transcripts in episomal HPV cell lines. The viral E2 protein, which binds with high affinity to specific palindromes near the promoter and origin, complexes with SETX, and both SETX and E2 are present at the viral p97 promoter in CIN612 and W12 cells. SETX overexpression increased E2 transcription activity on the p97 promoter. SETX depletion also significantly increased integration of viral genomes in CIN612 cells. Our results demonstrate that SETX resolves viral R-loops to proceed with HPV transcription and prevent genome integration.IMPORTANCEPapillomaviruses contain small circular genomes of approximately 8 kilobase pairs and undergo unidirectional transcription from the sense strand of the viral genome. Co-transcriptional R-loops were recently reported to be present at high levels in cells that maintain episomal HPV and were also detected at the early viral promoter. R-loops can inhibit transcription and DNA replication. The process that removes R-loops from the PV genome and the requisite enzymes are unknown. We propose a model in which the host RNA-DNA helicase senataxin assembles on the HPV genome to resolve R-loops in order to maintain the episomal status of the viral genome.


Subject(s)
DNA Helicases , Multifunctional Enzymes , Promoter Regions, Genetic , R-Loop Structures , RNA Helicases , Humans , RNA Helicases/genetics , RNA Helicases/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , R-Loop Structures/genetics , Plasmids/genetics , Virus Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Papillomaviridae/genetics , Genome, Viral , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Transcription, Genetic , Cell Line , DNA, Viral/genetics
18.
J Biol Chem ; 299(10): 105237, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37690693

ABSTRACT

The protein FUS (FUSed in sarcoma) is a metazoan RNA-binding protein that influences RNA production by all three nuclear polymerases. FUS also binds nascent transcripts, RNA processing factors, RNA polymerases, and transcription machinery. Here, we explored the role of FUS binding interactions for activity during transcription. In vitro run-off transcription assays revealed FUS-enhanced RNA produced by a non-eukaryote polymerase. The activity also reduced the formation of R-loops between RNA products and their DNA template. Analysis by domain mutation and deletion indicated RNA-binding was required for activity. We interpret that FUS binds and sequesters nascent transcripts to prevent R-loops from forming with nearby DNA. DRIP-seq analysis showed that a knockdown of FUS increased R-loop enrichment near expressed genes. Prevention of R-loops by FUS binding to nascent transcripts has the potential to affect transcription by any RNA polymerase, highlighting the broad impact FUS can have on RNA metabolism in cells and disease.


Subject(s)
DNA , R-Loop Structures , RNA-Binding Protein FUS , RNA , DNA/metabolism , R-Loop Structures/genetics , RNA/metabolism , RNA-Binding Protein FUS/metabolism , Protein Binding , Humans , DNA-Directed RNA Polymerases/metabolism , HEK293 Cells
19.
Cell Mol Biol Lett ; 29(1): 89, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877420

ABSTRACT

CircR-loop, a recently unearthed regulatory mechanism situated at the crossroads of circular RNA and DNA interactions, constitute a subset of R-loop. This circR-loop have emerged as a crucial player in pivotal regulatory functions within both animal and plant systems. The journey into the realm of circR-loop commenced with their discovery within the human mitochondrial genome, where they serve as critical directors of mitochondrial DNA replication. In the plant kingdom, circR-loop wield influence over processes such as alternative splicing and centromere organization, impacting the intricacies of floral development and genome stability, respectively. Their significance extends to the animal domain, where circR-loop has captured attention for their roles in cancer-related phenomena, exerting control over transcription, chromatin architecture, and orchestrating responses to DNA damage. Moreover, their involvement in nuclear export anomalies further underscores their prominence in cellular regulation. This article summarizes the important regulatory mechanisms and physiological roles of circR-loop in plants and animals, and offers a comprehensive exploration of the methodologies employed for the identification, characterization, and functional analysis of circR-loop, underscoring the pressing need for innovative approaches that can effectively distinguish them from their linear RNA counterparts while elucidating their precise functions. Lastly, the article sheds light on the challenges and opportunities that lie ahead in the field of circR-loop research, emphasizing the vital importance of continued investigations to uncover their regulatory roles and potential applications in the realm of biology. In summary, circR-loop represents a captivating and novel regulatory mechanism with broad-reaching implications spanning the realms of genetics, epigenetics, and disease biology. Their exploration opens new avenues for comprehending gene regulation and holds significant promise for future therapeutic interventions.


Subject(s)
Genomic Instability , RNA, Circular , Genomic Instability/genetics , Humans , Animals , RNA, Circular/genetics , RNA, Circular/metabolism , DNA/metabolism , DNA/genetics , R-Loop Structures/genetics , RNA/metabolism , RNA/genetics , DNA Replication/genetics
20.
PLoS Genet ; 17(8): e1009784, 2021 08.
Article in English | MEDLINE | ID: mdl-34464389

ABSTRACT

Aberrant repair of DNA double-strand breaks can recombine distant chromosomal breakpoints. Chromosomal rearrangements compromise genome function and are a hallmark of ageing. Rearrangements are challenging to detect in non-dividing cell populations, because they reflect individually rare, heterogeneous events. The genomic distribution of de novo rearrangements in non-dividing cells, and their dynamics during ageing, remain therefore poorly characterized. Studies of genomic instability during ageing have focussed on mitochondrial DNA, small genetic variants, or proliferating cells. To characterize genome rearrangements during cellular ageing in non-dividing cells, we interrogated a single diagnostic measure, DNA breakpoint junctions, using Schizosaccharomyces pombe as a model system. Aberrant DNA junctions that accumulated with age were associated with microhomology sequences and R-loops. Global hotspots for age-associated breakpoint formation were evident near telomeric genes and linked to remote breakpoints elsewhere in the genome, including the mitochondrial chromosome. Formation of breakpoint junctions at global hotspots was inhibited by the Sir2 histone deacetylase and might be triggered by an age-dependent de-repression of chromatin silencing. An unexpected mechanism of genomic instability may cause more local hotspots: age-associated reduction in an RNA-binding protein triggering R-loops at target loci. This result suggests that biological processes other than transcription or replication can drive genome rearrangements. Notably, we detected similar signatures of genome rearrangements that accumulated in old brain cells of humans. These findings provide insights into the unique patterns and possible mechanisms of genome rearrangements in non-dividing cells, which can be promoted by ageing-related changes in gene-regulatory proteins.


Subject(s)
Gene Rearrangement/genetics , Genomic Instability/genetics , R-Loop Structures/genetics , Aging/genetics , Chromosome Aberrations , Chromosome Breakpoints , DNA Breaks, Double-Stranded , Genomics/methods , Models, Genetic , Mutation/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Telomere/genetics
SELECTION OF CITATIONS
SEARCH DETAIL