Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(2): 113778, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38341854

ABSTRACT

During genome duplication, replication forks (RFs) can be stalled by different obstacles or by depletion of replication factors or nucleotides. A limited number of histone post-translational modifications at stalled RFs are involved in RF protection and restart. Provided the recent observation that the SIN3A histone deacetylase complex reduces transcription-replication conflicts, we explore the role of the SIN3A complex in protecting RFs under stressed conditions. We observe that Sin3A protein is enriched at replicating DNA in the presence of hydroxyurea. In this situation, Sin3A-depleted cells show increased RF stalling, H3 acetylation, and DNA breaks at stalled RFs. Under Sin3A depletion, RF recovery is impaired, and DNA damage accumulates. Importantly, these effects are partially dependent on the MUS81 endonuclease, which promotes DNA breaks and MRE11-dependent DNA degradation of such breaks. We propose that chromatin deacetylation triggered by the SIN3A complex limits MUS81 cleavage of stalled RFs, promoting genome stability when DNA replication is challenged.


Subject(s)
Cell Cycle Proteins , Chromatin , Acetylation , Protein Processing, Post-Translational , DNA
2.
Cell Rep ; 42(3): 112148, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36827184

ABSTRACT

Unscheduled R loops can be a source of genome instability, a hallmark of cancer cells. Although targeted proteomic approaches and cellular analysis of specific mutants have uncovered factors potentially involved in R-loop homeostasis, we report a more open screening of factors whose depletion causes R loops based on the ability of activation-induced cytidine deaminase (AID) to target R loops. Immunofluorescence analysis of γH2AX caused by small interfering RNAs (siRNAs) covering 3,205 protein-coding genes identifies 59 potential candidates, from which 13 are analyzed further and show a significant increase of R loops. Such candidates are enriched in factors involved in chromatin, transcription, and RNA biogenesis and other processes. A more focused study shows that the DDX47 helicase is an R-loop resolvase, whereas the MeCP2 methyl-CpG-binding protein uncovers a link between DNA methylation and R loops. Thus, our results suggest that a plethora of gene dysfunctions can alter cell physiology via affecting R-loop homeostasis by different mechanisms.


Subject(s)
Proteomics , R-Loop Structures , Humans , Chromatin , Chromosomes/metabolism , DNA Helicases/metabolism , Genomic Instability
3.
Nat Commun ; 13(1): 3743, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768435

ABSTRACT

Perturbation in the replication-stress response (RSR) and DNA-damage response (DDR) causes genomic instability. Genomic instability occurs in Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency disorder, yet the mechanism remains largely uncharacterized. Replication protein A (RPA), a single-strand DNA (ssDNA) binding protein, has key roles in the RSR and DDR. Here we show that human WAS-protein (WASp) modulates RPA functions at perturbed replication forks (RFs). Following genotoxic insult, WASp accumulates at RFs, associates with RPA, and promotes RPA:ssDNA complexation. WASp deficiency in human lymphocytes destabilizes RPA:ssDNA-complexes, impairs accumulation of RPA, ATR, ETAA1, and TOPBP1 at genotoxin-perturbed RFs, decreases CHK1 activation, and provokes global RF dysfunction. las17 (yeast WAS-homolog)-deficient S. cerevisiae also show decreased ScRPA accumulation at perturbed RFs, impaired DNA recombination, and increased frequency of DNA double-strand break (DSB)-induced single-strand annealing (SSA). Consequently, WASp (or Las17)-deficient cells show increased frequency of DSBs upon genotoxic insult. Our study reveals an evolutionarily conserved, essential role of WASp in the DNA stress-resolution pathway, such that WASp deficiency provokes RPA dysfunction-coupled genomic instability.


Subject(s)
DNA Breaks, Double-Stranded , DNA Replication , DNA, Single-Stranded , Replication Protein A , Saccharomyces cerevisiae Proteins , Wiskott-Aldrich Syndrome Protein , Animals , Antigens, Surface/metabolism , DNA Repair , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Protein Binding , Replication Protein A/genetics , Replication Protein A/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
4.
Nucleic Acids Res ; 49(22): 12785-12804, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34871443

ABSTRACT

Genome instability is a condition characterized by the accumulation of genetic alterations and is a hallmark of cancer cells. To uncover new genes and cellular pathways affecting endogenous DNA damage and genome integrity, we exploited a Synthetic Genetic Array (SGA)-based screen in yeast. Among the positive genes, we identified VID22, reported to be involved in DNA double-strand break repair. vid22Δ cells exhibit increased levels of endogenous DNA damage, chronic DNA damage response activation and accumulate DNA aberrations in sequences displaying high probabilities of forming G-quadruplexes (G4-DNA). If not resolved, these DNA secondary structures can block the progression of both DNA and RNA polymerases and correlate with chromosome fragile sites. Vid22 binds to and protects DNA at G4-containing regions both in vitro and in vivo. Loss of VID22 causes an increase in gross chromosomal rearrangement (GCR) events dependent on G-quadruplex forming sequences. Moreover, the absence of Vid22 causes defects in the correct maintenance of G4-DNA rich elements, such as telomeres and mtDNA, and hypersensitivity to the G4-stabilizing ligand TMPyP4. We thus propose that Vid22 is directly involved in genome integrity maintenance as a novel regulator of G4 metabolism.


Subject(s)
G-Quadruplexes , Genomic Instability , Membrane Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Chromosome Aberrations , DNA Damage , Genome, Fungal , Membrane Proteins/genetics , Membrane Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere Homeostasis
5.
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
6.
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
7.
PLoS Genet ; 12(4): e1005966, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27035147

ABSTRACT

Yra1 is an essential nuclear factor of the evolutionarily conserved family of hnRNP-like export factors that when overexpressed impairs mRNA export and cell growth. To investigate further the relevance of proper Yra1 stoichiometry in the cell, we overexpressed Yra1 by transforming yeast cells with YRA1 intron-less constructs and analyzed its effect on gene expression and genome integrity. We found that YRA1 overexpression induces DNA damage and leads to a transcription-associated hyperrecombination phenotype that is mediated by RNA:DNA hybrids. In addition, it confers a genome-wide replication retardation as seen by reduced BrdU incorporation and accumulation of the Rrm3 helicase. In addition, YRA1 overexpression causes a cell senescence-like phenotype and telomere shortening. ChIP-chip analysis shows that overexpressed Yra1 is loaded to transcribed chromatin along the genome and to Y' telomeric regions, where Rrm3 is also accumulated, suggesting an impairment of telomere replication. Our work not only demonstrates that a proper stoichiometry of the Yra1 mRNA binding and export factor is required to maintain genome integrity and telomere homeostasis, but suggests that the cellular imbalance between transcribed RNA and specific RNA-binding factors may become a major cause of genome instability mediated by co-transcriptional replication impairment.


Subject(s)
DNA Replication , Genomic Instability , Nuclear Proteins/physiology , RNA-Binding Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Telomere Shortening , Transcription, Genetic , Genes, Fungal , Nucleic Acid Hybridization , Recombination, Genetic
8.
PLoS Genet ; 11(11): e1005674, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26584049

ABSTRACT

Co-transcriptional RNA-DNA hybrids (R loops) cause genome instability. To prevent harmful R loop accumulation, cells have evolved specific eukaryotic factors, one being the BRCA2 double-strand break repair protein. As BRCA2 also protects stalled replication forks and is the FANCD1 member of the Fanconi Anemia (FA) pathway, we investigated the FA role in R loop-dependent genome instability. Using human and murine cells defective in FANCD2 or FANCA and primary bone marrow cells from FANCD2 deficient mice, we show that the FA pathway removes R loops, and that many DNA breaks accumulated in FA cells are R loop-dependent. Importantly, FANCD2 foci in untreated and MMC-treated cells are largely R loop dependent, suggesting that the FA functions at R loop-containing sites. We conclude that co-transcriptional R loops and R loop-mediated DNA damage greatly contribute to genome instability and that one major function of the FA pathway is to protect cells from R loops.


Subject(s)
BRCA2 Protein/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Genomic Instability/genetics , Animals , DNA/chemistry , DNA/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , HeLa Cells , Humans , Mice , RNA/chemistry , RNA/genetics
9.
EMBO J ; 34(2): 236-50, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25452497

ABSTRACT

Transcription is a major contributor to genome instability. A main cause of transcription-associated instability relies on the capacity of transcription to stall replication. However, we know little of the possible role, if any, of the RNA polymerase (RNAP) in this process. Here, we analyzed 4 specific yeast RNAPII mutants that show different phenotypes of genetic instability including hyper-recombination, DNA damage sensitivity and/or a strong dependency on double-strand break repair functions for viability. Three specific alleles of the RNAPII core, rpb1-1, rpb1-S751F and rpb9∆, cause a defect in replication fork progression, compensated for by additional origin firing, as the main action responsible for instability. The transcription elongation defects of rpb1-S751F and rpb9∆ plus our observation that rpb1-1 causes RNAPII retention on chromatin suggest that RNAPII could participate in facilitating fork progression upon a transcription-replication encounter. Our results imply that the RNAPII or ancillary factors actively help prevent transcription-associated genome instability.


Subject(s)
DNA Repair/genetics , DNA Replication/genetics , Genomic Instability , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Biomarkers/metabolism , Chromatin Immunoprecipitation , Gene Expression Profiling , Mutation/genetics , Oligonucleotide Array Sequence Analysis , RNA Polymerase II/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
10.
Mol Cell ; 56(6): 777-85, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25435140

ABSTRACT

R-loops, consisting of an RNA-DNA hybrid and displaced single-stranded DNA, are physiological structures that regulate various cellular processes occurring on chromatin. Intriguingly, changes in R-loop dynamics have also been associated with DNA damage accumulation and genome instability; however, the mechanisms underlying R-loop-induced DNA damage remain unknown. Here we demonstrate in human cells that R-loops induced by the absence of diverse RNA processing factors, including the RNA/DNA helicases Aquarius (AQR) and Senataxin (SETX), or by the inhibition of topoisomerase I, are actively processed into DNA double-strand breaks (DSBs) by the nucleotide excision repair endonucleases XPF and XPG. Surprisingly, DSB formation requires the transcription-coupled nucleotide excision repair (TC-NER) factor Cockayne syndrome group B (CSB), but not the global genome repair protein XPC. These findings reveal an unexpected and potentially deleterious role for TC-NER factors in driving R-loop-induced DNA damage and genome instability.


Subject(s)
DNA Repair , Genomic Instability , DNA Damage , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Genome, Human , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
11.
Nucleic Acids Res ; 42(19): 12000-14, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25294824

ABSTRACT

The THSC/TREX-2 complex of Saccharomyces cerevisiae mediates the anchoring of transcribed genes to the nuclear pore, linking transcription elongation with mRNA export and genome stability, as shown for specific reporters. However, it is still unknown whether the function of TREX-2 is global and the reason for its relevant role in genome integrity. Here, by studying two TREX-2 representative subunits, Thp1 and Sac3, we show that TREX-2 has a genome-wide role in gene expression. Both proteins show similar distributions along the genome, with a gradient disposition at active genes that increases towards the 3' end. Thp1 and Sac3 have a relevant impact on the expression of long, G+C-rich and highly transcribed genes. Interestingly, replication impairment detected by the genome-wide accumulation of the replicative Rrm3 helicase is increased preferentially at highly expressed genes in the thp1Δ and sac3Δ mutants analyzed. Therefore, our work provides evidence of a function of TREX-2 at the genome-wide level and suggests a role for TREX-2 in preventing transcription-replication conflicts, as a source of genome instability derived from a defective messenger ribonucleoprotein particle (mRNP) biogenesis.


Subject(s)
DNA Replication , Nucleocytoplasmic Transport Proteins/metabolism , Porins/metabolism , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic , 3' Flanking Region , DNA Helicases/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Genome, Fungal , Nucleocytoplasmic Transport Proteins/genetics , Porins/genetics , Ribonucleoproteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
12.
Nature ; 511(7509): 362-5, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24896180

ABSTRACT

Genome instability is central to ageing, cancer and other diseases. It is not only proteins involved in DNA replication or the DNA damage response (DDR) that are important for maintaining genome integrity: from yeast to higher eukaryotes, mutations in genes involved in pre-mRNA splicing and in the biogenesis and export of messenger ribonucleoprotein (mRNP) also induce DNA damage and genome instability. This instability is frequently mediated by R-loops formed by DNA-RNA hybrids and a displaced single-stranded DNA. Here we show that the human TREX-2 complex, which is involved in mRNP biogenesis and export, prevents genome instability as determined by the accumulation of γ-H2AX (Ser-139 phosphorylated histone H2AX) and 53BP1 foci and single-cell electrophoresis in cells depleted of the TREX-2 subunits PCID2, GANP and DSS1. We show that the BRCA2 repair factor, which binds to DSS1, also associates with PCID2 in the cell. The use of an enhanced green fluorescent protein-tagged hybrid-binding domain of RNase H1 and the S9.6 antibody did not detect R-loops in TREX-2-depleted cells, but did detect the accumulation of R-loops in BRCA2-depleted cells. The results indicate that R-loops are frequently formed in cells and that BRCA2 is required for their processing. This link between BRCA2 and RNA-mediated genome instability indicates that R-loops may be a chief source of replication stress and cancer-associated instability.


Subject(s)
BRCA2 Protein/metabolism , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/metabolism , Genomic Instability , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA Transport , RNA/metabolism , Acetyltransferases/metabolism , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , DNA Damage , DNA Replication , DNA, Single-Stranded/chemistry , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/deficiency , Histones/chemistry , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nucleic Acid Conformation , Phosphoproteins/chemistry , Phosphoproteins/deficiency , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA/chemistry , Ribonuclease H/chemistry , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/metabolism
13.
Genes Dev ; 28(7): 735-48, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24636987

ABSTRACT

FACT (facilitates chromatin transcription) is a chromatin-reorganizing complex that swaps nucleosomes around the RNA polymerase during transcription elongation and has a role in replication that is not fully understood yet. Here we show that recombination factors are required for the survival of yeast FACT mutants, consistent with an accumulation of DNA breaks that we detected by Rad52 foci and transcription-dependent hyperrecombination. Breaks also accumulate in FACT-depleted human cells, as shown by γH2AX foci and single-cell electrophoresis. Furthermore, FACT-deficient yeast and human cells show replication impairment, which in yeast we demonstrate by ChIP-chip (chromatin immunoprecipitation [ChIP] coupled with microarray analysis) of Rrm3 to occur genome-wide but preferentially at highly transcribed regions. Strikingly, in yeast FACT mutants, high levels of Rad52 foci are suppressed by RNH1 overexpression; R loops accumulate at high levels, and replication becomes normal when global RNA synthesis is inhibited in FACT-depleted human cells. The results demonstrate a key function of FACT in the resolution of R-loop-mediated transcription-replication conflicts, likely associated with a specific chromatin organization.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic/physiology , Transcriptional Elongation Factors/metabolism , Cell Survival/genetics , DNA Breaks , DNA Replication/genetics , DNA-Binding Proteins/genetics , Genomic Instability/genetics , High Mobility Group Proteins/genetics , Humans , Mutation , Ribonuclease H/genetics , Ribonuclease H/metabolism , S Phase , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic/genetics , Transcriptional Elongation Factors/genetics
14.
Genes Dev ; 27(22): 2445-58, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24240235

ABSTRACT

Transcription is a major obstacle for replication fork (RF) progression and a cause of genome instability. Part of this instability is mediated by cotranscriptional R loops, which are believed to increase by suboptimal assembly of the nascent messenger ribonucleoprotein particle (mRNP). However, no clear evidence exists that heterogeneous nuclear RNPs (hnRNPs), the basic mRNP components, prevent R-loop stabilization. Here we show that yeast Npl3, the most abundant RNA-binding hnRNP, prevents R-loop-mediated genome instability. npl3Δ cells show transcription-dependent and R-loop-dependent hyperrecombination and genome-wide replication obstacles as determined by accumulation of the Rrm3 helicase. Such obstacles preferentially occur at long and highly expressed genes, to which Npl3 is preferentially bound in wild-type cells, and are reduced by RNase H1 overexpression. The resulting replication stress confers hypersensitivity to double-strand break-inducing agents. Therefore, our work demonstrates that mRNP factors are critical for genome integrity and opens the option of using them as therapeutic targets in anti-cancer treatment.


Subject(s)
DNA Replication/genetics , Genomic Instability/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription, Genetic/genetics , 3' Flanking Region , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Deletion , Genome, Fungal , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Mutagens/pharmacology , Saccharomyces cerevisiae/drug effects
15.
Nucleic Acids Res ; 40(3): 1050-64, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21998294

ABSTRACT

Despite the theoretical bases for the association of topoisomerases and supercoiling changes with transcription and replication, our knowledge of the impact of topological constraints on transcription and replication is incomplete. Although mutation of topoisomerases affects expression and stability of the rDNA region it is not clear whether the same is the case for RNAPII transcription and genome integrity in other regions. We developed new assays in which two convergent RNAPII-driven genes are transcribed simultaneously. Plasmid-based systems were constructed with and without a transcription terminator between the two convergent transcription units, so that the impact of transcription interference could also be evaluated. Using these assays we show that Topos I and II play roles in RNAPII transcription in vivo and reduce the stability of RNAPII-transcribed genes in Saccharomyces cerevisiae. Supercoiling accumulation in convergent transcription units impairs RNAPII transcription in top1Δ strains, but Topo II is also required for efficient transcription independent of Topo I and of detectable supercoiling accumulation. Our work shows that topological constraints negatively affect RNAPII transcription and genetic integrity, and provides an assay to study gene regulation by transcription interference.


Subject(s)
DNA Topoisomerases, Type II/physiology , DNA Topoisomerases, Type I/physiology , Plasmids/genetics , RNA Polymerase II/metabolism , Saccharomyces cerevisiae Proteins/physiology , Transcription, Genetic , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type II/genetics , DNA, Superhelical/metabolism , Mutation , Phenotype , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Terminator Regions, Genetic
16.
Mol Cell Biol ; 31(4): 674-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21149575

ABSTRACT

Formation of a ribonucleoprotein particle (mRNP) competent for export requires the coupling of transcription with mRNA processing and RNA export. A key link between these processes is provided by the THO complex. To progress in our understanding of this coupling, we have performed a search for suppressors of the transcription defect caused by the hpr1Δ mutation. This has permitted us to identify mutations in the genes for the RNA polymerase II mediator component Med10, the Sch9 protein kinase, and the Ypr045c protein. We report a role in transcription elongation for Ypr045c (Thp3) and the Csn12 component of the COP9 signalosome. Thp3 and Csn12 form a complex that is recruited to transcribed genes. Their mutations suppress the gene expression defects of THO complex mutants involved in mRNP biogenesis and export and show defects in mRNA accumulation. Transcription elongation impairment of thp3Δ mutants is shown by in vivo transcript run-on analysis performed in G-less systems. Thp3-Csn12 establishes a novel link between transcription and mRNA processing that opens new perspectives on our understanding of gene expression and reveals novel functions for a component of the COP9 signalosome. Thp3-Csn12 also copurifies with ribosomal proteins, which opens the possibility that it has other functions in addition to transcription.


Subject(s)
Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Base Sequence , COP9 Signalosome Complex , DNA, Fungal/genetics , Genes, Fungal , Genome, Fungal , Mediator Complex/genetics , Mediator Complex/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Open Reading Frames , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Processing, Post-Transcriptional , Suppression, Genetic , Transcription, Genetic
17.
PLoS Genet ; 5(2): e1000364, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19197357

ABSTRACT

RNA polymerases frequently deal with a number of obstacles during transcription elongation that need to be removed for transcription resumption. One important type of hindrance consists of DNA lesions, which are removed by transcription-coupled repair (TC-NER), a specific sub-pathway of nucleotide excision repair. To improve our knowledge of transcription elongation and its coupling to TC-NER, we used the yeast library of non-essential knock-out mutations to screen for genes conferring resistance to the transcription-elongation inhibitor mycophenolic acid and the DNA-damaging agent 4-nitroquinoline-N-oxide. Our data provide evidence that subunits of the SAGA and Ccr4-Not complexes, Mediator, Bre1, Bur2, and Fun12 affect transcription elongation to different extents. Given the dependency of TC-NER on RNA Polymerase II transcription and the fact that the few proteins known to be involved in TC-NER are related to transcription, we performed an in-depth TC-NER analysis of a selection of mutants. We found that mutants of the PAF and Ccr4-Not complexes are impaired in TC-NER. This study provides evidence that PAF and Ccr4-Not are required for efficient TC-NER in yeast, unraveling a novel function for these transcription complexes and opening new perspectives for the understanding of TC-NER and its functional interconnection with transcription elongation.


Subject(s)
DNA Repair/physiology , Genome, Fungal , Ribonucleases/physiology , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/physiology , Transcription, Genetic , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Fungal/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Messenger/metabolism , Ribonucleases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
18.
Mol Cell Biol ; 26(20): 7451-65, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16908536

ABSTRACT

THO/TREX, a conserved eukaryotic protein complex, is a key player at the interface between transcription and mRNP metabolism. The lack of a functional THO complex impairs transcription, leads to transcription-dependent hyperrecombination, causes mRNA export defects and fast mRNA decay, and retards replication fork progression in a transcription-dependent manner. To get more insight into the interconnection between mRNP biogenesis and genomic instability, we searched for HPR1 mutations that differentially affect gene expression and recombination. We isolated mutants that were barely affected in gene expression but exhibited a hyperrecombination phenotype. In addition, we isolated a mutant, hpr1-101, with a strong defect in transcription, as observed for lacZ, and a general defect in mRNA export that did not display a relevant hyperrecombination phenotype. In THO single-null mutants, but not in the hpr1 point mutants studied, THO and its subunits were unstable. Interestingly, in contrast to hyperrecombinant null mutants, hpr1-101 did not cause retardation of replication fork progression. Transcription and mRNP biogenesis can therefore be impaired by THO/TREX dysfunction without increasing recombination, suggesting that it is possible to separate the mechanism(s) responsible for mRNA biogenesis defects from the further step of triggering transcription-dependent recombination.


Subject(s)
Protein Biosynthesis/genetics , Recombination, Genetic/genetics , Ribonucleoproteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription, Genetic/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Cell Nucleus/genetics , Chromatin/metabolism , DNA Replication/genetics , DNA, Fungal/genetics , Gene Expression , Nuclear Proteins , Point Mutation/genetics , Protein Binding , RNA Transport , RNA, Messenger/metabolism , Saccharomyces cerevisiae/isolation & purification , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...