Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
Add more filters

Country/Region as subject
Publication year range
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.
Annu Rev Biochem ; 85: 291-317, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27023844

ABSTRACT

Genomes undergo different types of sporadic alterations, including DNA damage, point mutations, and genome rearrangements, that constitute the basis for evolution. However, these changes may occur at high levels as a result of cell pathology and trigger genome instability, a hallmark of cancer and a number of genetic diseases. In the last two decades, evidence has accumulated that transcription constitutes an important natural source of DNA metabolic errors that can compromise the integrity of the genome. Transcription can create the conditions for high levels of mutations and recombination by its ability to open the DNA structure and remodel chromatin, making it more accessible to DNA insulting agents, and by its ability to become a barrier to DNA replication. Here we review the molecular basis of such events from a mechanistic perspective with particular emphasis on the role of transcription as a genome instability determinant.


Subject(s)
DNA Repair , Genomic Instability , Mutagenesis , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Transcription, Genetic , Chromatin Assembly and Disassembly , DNA/genetics , DNA/metabolism , DNA Breaks, Single-Stranded , DNA Replication , Genome, Human , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Nucleic Acid Conformation , Recombination, Genetic
3.
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
4.
Nat Rev Mol Cell Biol ; 17(9): 553-63, 2016 09.
Article in English | MEDLINE | ID: mdl-27435505

ABSTRACT

The frequent occurrence of transcription and DNA replication in cells results in many encounters, and thus conflicts, between the transcription and replication machineries. These conflicts constitute a major intrinsic source of genome instability, which is a hallmark of cancer cells. How the replication machinery progresses along a DNA molecule occupied by an RNA polymerase is an old question. Here we review recent data on the biological relevance of transcription-replication conflicts, and the factors and mechanisms that are involved in either preventing or resolving them, mainly in eukaryotes. On the basis of these data, we provide our current view of how transcription can generate obstacles to replication, including torsional stress and non-B DNA structures, and of the different cellular processes that have evolved to solve them.


Subject(s)
DNA Replication , Transcription, Genetic , Animals , Chromatin Assembly and Disassembly , DNA/chemistry , DNA Repair , DNA, Superhelical , Genomic Instability , Humans , RNA/chemistry , Regulatory Elements, Transcriptional
5.
Mol Cell ; 79(3): 361-362, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32763222

ABSTRACT

In this issue of Molecular Cell, Zhang et al. (2020) reveal that ATM triggers RNA methylation of DNA-RNA hybrids formed at double-strand breaks (DSBs) to modulate repair, adding a new layer of complexity to RNA's role in the DNA damage response.


Subject(s)
DNA Breaks, Double-Stranded , RNA , Adenosine/analogs & derivatives , Ataxia Telangiectasia Mutated Proteins , DNA , DNA Repair , Methylation
6.
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
7.
Mol Cell ; 76(4): 529-530, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31756322

ABSTRACT

The study by Tan-Wong et al. (2019) in this issue of Molecular Cell reveals a capacity of R-loops to promote antisense transcription expanding our view of the features that a DNA region may have to act as a promoter.


Subject(s)
Mammals , R-Loop Structures , Animals , Promoter Regions, Genetic
8.
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
9.
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
10.
Cell ; 146(2): 233-46, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21784245

ABSTRACT

Transcription hinders replication fork progression and stability, and the Mec1/ATR checkpoint protects fork integrity. Examining checkpoint-dependent mechanisms controlling fork stability, we find that fork reversal and dormant origin firing due to checkpoint defects are rescued in checkpoint mutants lacking THO, TREX-2, or inner-basket nucleoporins. Gene gating tethers transcribed genes to the nuclear periphery and is counteracted by checkpoint kinases through phosphorylation of nucleoporins such as Mlp1. Checkpoint mutants fail to detach transcribed genes from nuclear pores, thus generating topological impediments for incoming forks. Releasing this topological complexity by introducing a double-strand break between a fork and a transcribed unit prevents fork collapse. Mlp1 mutants mimicking constitutive checkpoint-dependent phosphorylation also alleviate checkpoint defects. We propose that the checkpoint assists fork progression and stability at transcribed genes by phosphorylating key nucleoporins and counteracting gene gating, thus neutralizing the topological tension generated at nuclear pore gated genes.


Subject(s)
DNA Replication , Nuclear Pore/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Checkpoint Kinase 2 , DNA Breaks, Double-Stranded , Hydroxyurea/pharmacology , Mutation , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism
11.
Mol Cell ; 70(1): 34-47.e4, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29551515

ABSTRACT

UV-induced photoproducts are responsible for the pathological effects of sunlight. Mutations in nucleotide excision repair (NER) cause severe pathologies characterized by sunlight sensitivity, coupled to elevated predisposition to cancer and/or neurological dysfunctions. We have previously shown that in UV-irradiated non-cycling cells, only a particular subset of lesions activates the DNA damage response (DDR), and this requires NER and EXO1 activities. To define the molecular mechanism acting at these lesions, we demonstrate that Y family TLS polymerases are recruited at NER- and EXO1-positive lesion sites in non-S phase cells. The coordinated action of EXO1 and Y family TLS polymerases promotes checkpoint activation, leads to lesion repair, and is crucial to prevent cytotoxic double-strand break (DSB) formation.


Subject(s)
Cell Cycle Checkpoints/radiation effects , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , DNA Repair/radiation effects , DNA-Directed DNA Polymerase/metabolism , Exodeoxyribonucleases/metabolism , Ultraviolet Rays/adverse effects , Cell Death/radiation effects , Cell Line , DNA Repair Enzymes/genetics , DNA-Directed DNA Polymerase/genetics , Exodeoxyribonucleases/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Transport , DNA Polymerase iota
12.
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
13.
Genes Dev ; 32(13-14): 965-977, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29954833

ABSTRACT

R loops are an important source of genome instability, largely due to their negative impact on replication progression. Yra1/ALY is an abundant RNA-binding factor conserved from yeast to humans and required for mRNA export, but its excess causes lethality and genome instability. Here, we show that, in addition to ssDNA and ssRNA, Yra1 binds RNA-DNA hybrids in vitro and, when artificially overexpressed, can be recruited to chromatin in an RNA-DNA hybrid-dependent manner, stabilizing R loops and converting them into replication obstacles in vivo. Importantly, an excess of Yra1 increases R-loop-mediated genome instability caused by transcription-replication collisions regardless of whether they are codirectional or head-on. It also induces telomere shortening in telomerase-negative cells and accelerates senescence, consistent with a defect in telomere replication. Our results indicate that RNA-DNA hybrids form transiently in cells regardless of replication and, after stabilization by excess Yra1, compromise genome integrity, in agreement with a two-step model of R-loop-mediated genome instability. This work opens new perspectives to understand transcription-associated genome instability in repair-deficient cells, including tumoral cells.


Subject(s)
Chromosomal Instability/genetics , DNA Replication , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere/genetics , Transcription, Genetic , Chromatin/metabolism , Nucleic Acid Hybridization , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/metabolism
14.
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
15.
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
16.
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
17.
Nucleic Acids Res ; 51(12): 6337-6354, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37224534

ABSTRACT

Accurate genome replication is essential for all life and a key mechanism of disease prevention, underpinned by the ability of cells to respond to replicative stress (RS) and protect replication forks. These responses rely on the formation of Replication Protein A (RPA)-single stranded (ss) DNA complexes, yet this process remains largely uncharacterized. Here, we establish that actin nucleation-promoting factors (NPFs) associate with replication forks, promote efficient DNA replication and facilitate association of RPA with ssDNA at sites of RS. Accordingly, their loss leads to deprotection of ssDNA at perturbed forks, impaired ATR activation, global replication defects and fork collapse. Supplying an excess of RPA restores RPA foci formation and fork protection, suggesting a chaperoning role for actin nucleators (ANs) (i.e. Arp2/3, DIAPH1) and NPFs (i.e, WASp, N-WASp) in regulating RPA availability upon RS. We also discover that ß-actin interacts with RPA directly in vitro, and in vivo a hyper-depolymerizing ß-actin mutant displays a heightened association with RPA and the same dysfunctional replication phenotypes as loss of ANs/NPFs, which contrasts with the phenotype of a hyper-polymerizing ß-actin mutant. Thus, we identify components of actin polymerization pathways that are essential for preventing ectopic nucleolytic degradation of perturbed forks by modulating RPA activity.


Subject(s)
Actins , DNA Replication , Actins/genetics , Replication Protein A/genetics , Replication Protein A/metabolism , DNA, Single-Stranded/genetics , Molecular Chaperones/genetics
18.
Mol Genet Genomics ; 299(1): 59, 2024 May 26.
Article in English | MEDLINE | ID: mdl-38796829

ABSTRACT

RECQL5 is a member of the conserved RecQ family of DNA helicases involved in the maintenance of genome stability that is specifically found in higher eukaryotes and associates with the elongating RNA polymerase II. To expand our understanding of its function we expressed human RECQL5 in the yeast Saccharomyces cerevisiae, which does not have a RECQL5 ortholog. We found that RECQL5 expression leads to cell growth inhibition, increased genotoxic sensitivity and transcription-associated hyperrecombination. Chromatin immunoprecipitation and transcriptomic analysis of yeast cells expressing human RECQL5 shows that this is recruited to transcribed genes and although it causes only a weak impact on gene expression, in particular at G + C-rich genes, it leads to a transcription termination defect detected as readthrough transcription. The data indicate that the interaction between RNAPII and RECQL5 is conserved from yeast to humans. Unexpectedly, however, the RECQL5-ID mutant, previously shown to have reduced the association with RNAPII in vitro, associates with the transcribing polymerase in cells. As a result, expression of RECQL5-ID leads to similar although weaker phenotypes than wild-type RECQL5 that could be transcription-mediated. Altogether, the data suggests that RECQL5 has the intrinsic ability to function in transcription-dependent and independent genome dynamics in S. cerevisiae.


Subject(s)
Genomic Instability , RecQ Helicases , Saccharomyces cerevisiae , Transcription, Genetic , Saccharomyces cerevisiae/genetics , Genomic Instability/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , Humans , Transcription, Genetic/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
19.
Mol Cell ; 64(2): 388-404, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27768874

ABSTRACT

Common fragile sites (CFSs) are genomic regions that are unstable under conditions of replicative stress. Although the characteristics of CFSs that render them vulnerable to stress are associated mainly with replication, the cellular pathways that protect CFSs during replication remain unclear. Here, we identify and describe a role for FANCD2 as a trans-acting facilitator of CFS replication, in the absence of exogenous replicative stress. In the absence of FANCD2, replication forks stall within the AT-rich fragility core of CFS, leading to dormant origin activation. Furthermore, FANCD2 deficiency is associated with DNA:RNA hybrid formation at CFS-FRA16D, and inhibition of DNA:RNA hybrid formation suppresses replication perturbation. In addition, we also found that FANCD2 reduces the number of potential sites of replication initiation. Our data demonstrate that FANCD2 protein is required to ensure efficient CFS replication and provide mechanistic insight into how FANCD2 regulates CFS stability.


Subject(s)
Chromosome Fragile Sites , DNA Replication , DNA/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , RNA/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line, Transformed , DNA/metabolism , Fanconi Anemia , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Genomic Instability , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Humans , Lymphocytes/cytology , Lymphocytes/metabolism , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL