Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30808650

ABSTRACT

FUS (fused in sarcoma) plays a key role in several steps of RNA metabolism, and dominant mutations in this protein are associated with neurodegenerative diseases. Here, we show that FUS is a component of the cellular response to topoisomerase I (TOP1)-induced DNA breakage; relocalising to the nucleolus in response to RNA polymerase II (Pol II) stalling at sites of TOP1-induced DNA breaks. This relocalisation is rapid and dynamic, reversing following the removal of TOP1-induced breaks and coinciding with the recovery of global transcription. Importantly, FUS relocalisation following TOP1-induced DNA breakage is associated with increased FUS binding at sites of RNA polymerase I transcription in ribosomal DNA and reduced FUS binding at sites of RNA Pol II transcription, suggesting that FUS relocates from sites of stalled RNA Pol II either to regulate pre-mRNA processing during transcriptional stress or to modulate ribosomal RNA biogenesis. Importantly, FUS-mutant patient fibroblasts are hypersensitive to TOP1-induced DNA breakage, highlighting the possible relevance of these findings to neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , RNA-Binding Protein FUS/genetics , Transcription, Genetic , A549 Cells , Amyotrophic Lateral Sclerosis/genetics , Animals , Binding Sites , Brain/cytology , Brain/embryology , Chromatin/metabolism , DNA Repair , Fibroblasts/metabolism , HeLa Cells , Humans , Mice , Mutant Proteins , Mutation/genetics , Neural Stem Cells/metabolism , Neurons/metabolism , RNA Polymerase I/metabolism , RNA Polymerase II/metabolism , RNA-Binding Protein FUS/metabolism
2.
Nat Commun ; 8(1): 233, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28794467

ABSTRACT

DNA double-strand breaks (DSBs) induced by abortive topoisomerase II (TOP2) activity are a potential source of genome instability and chromosome translocation. TOP2-induced DNA double-strand breaks are rejoined in part by tyrosyl-DNA phosphodiesterase 2 (TDP2)-dependent non-homologous end-joining (NHEJ), but whether this process suppresses or promotes TOP2-induced translocations is unclear. Here, we show that TDP2 rejoins DSBs induced during transcription-dependent TOP2 activity in breast cancer cells and at the translocation 'hotspot', MLL. Moreover, we find that TDP2 suppresses chromosome rearrangements induced by TOP2 and reduces TOP2-induced chromosome translocations that arise during gene transcription. Interestingly, however, we implicate TDP2-dependent NHEJ in the formation of a rare subclass of translocations associated previously with therapy-related leukemia and characterized by junction sequences with 4-bp of perfect homology. Collectively, these data highlight the threat posed by TOP2-induced DSBs during transcription and demonstrate the importance of TDP2-dependent non-homologous end-joining in protecting both gene transcription and genome stability.DNA double-strand breaks (DSBs) induced by topoisomerase II (TOP2) are rejoined by TDP2-dependent non-homologous end-joining (NHEJ) but whether this promotes or suppresses translocations is not clear. Here the authors show that TDP2 suppresses chromosome translocations from DSBs introduced during gene transcription.


Subject(s)
DNA Topoisomerases, Type II/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Translocation, Genetic , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins , Humans , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Transcription Factors/genetics
3.
Nat Commun ; 7: 12364, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27503537

ABSTRACT

There are two major and alternative pathways to repair DNA double-strand breaks: non-homologous end-joining and homologous recombination. Here we identify and characterize novel factors involved in choosing between these pathways; in this study we took advantage of the SeeSaw Reporter, in which the repair of double-strand breaks by homology-independent or -dependent mechanisms is distinguished by the accumulation of green or red fluorescence, respectively. Using a genome-wide human esiRNA (endoribonuclease-prepared siRNA) library, we isolate genes that control the recombination/end-joining ratio. Here we report that two distinct sets of genes are involved in the control of the balance between NHEJ and HR: those that are required to facilitate recombination and those that favour NHEJ. This last category includes CCAR2/DBC1, which we show inhibits recombination by limiting the initiation and the extent of DNA end resection, thereby acting as an antagonist of CtIP.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA End-Joining Repair , Genome, Human , Carrier Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , DNA Damage , Endodeoxyribonucleases , Gene Regulatory Networks , Humans , Models, Biological , Nuclear Proteins/metabolism , Protein Binding , Recombinational DNA Repair
4.
J Biol Chem ; 288(8): 5496-505, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23316050

ABSTRACT

DNA methylation patterns are the dynamic outcome of antagonist methylation and demethylation mechanisms, but the latter are still poorly understood. Active DNA demethylation in plants is mediated by a family of DNA glycosylases typified by Arabidopsis ROS1 (repressor of silencing 1). ROS1 and its homologs remove 5-methylcytosine and incise the sugar backbone at the abasic site, thus initiating a base excision repair pathway that finally inserts an unmethylated cytosine. The DNA 3'-phosphatase ZDP processes some of the incision products generated by ROS1, allowing subsequent DNA polymerization and ligation steps. In this work, we examined the possible role of plant XRCC1 (x-ray cross-complementing group protein 1) in DNA demethylation. We found that XRCC1 interacts in vitro with ROS1 and ZDP and stimulates the enzymatic activity of both proteins. Furthermore, extracts from xrcc1 mutant plants exhibit a reduced capacity to complete DNA demethylation initiated by ROS1. An anti-XRCC1 antibody inhibits removal of the blocking 3'-phosphate in the single-nucleotide gap generated during demethylation and reduces the capacity of Arabidopsis cell extracts to ligate a nicked DNA intermediate. Our results suggest that XRCC1 is a component of plant base excision repair and functions at several stages during active DNA demethylation in Arabidopsis.


Subject(s)
5-Methylcytosine/metabolism , Arabidopsis Proteins/metabolism , DNA-Binding Proteins/metabolism , Arabidopsis/metabolism , DNA/metabolism , DNA Glycosylases/metabolism , DNA Methylation , DNA Repair , Epigenesis, Genetic , Gene Silencing , Models, Biological , Models, Genetic , Mutation , Nuclear Proteins/metabolism , Protein Binding , Reactive Oxygen Species , X-ray Repair Cross Complementing Protein 1
5.
Mol Cell ; 45(3): 357-70, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22325353

ABSTRACT

DNA methylation is an important epigenetic mark established by the combined actions of methylation and demethylation reactions. Plants use a base excision repair pathway for active DNA demethylation. After 5-methylcytosine removal, the Arabidopsis DNA glycosylase/lyase ROS1 incises the DNA backbone and part of the product has a single-nucleotide gap flanked by 3'- and 5'-phosphate termini. Here we show that the DNA phosphatase ZDP removes the blocking 3' phosphate, allowing subsequent DNA polymerization and ligation steps needed to complete the repair reactions. ZDP and ROS1 interact in vitro and colocalize in vivo in nucleoplasmic foci. Extracts from zdp mutant plants are unable to complete DNA demethylation in vitro, and the mutations cause DNA hypermethylation and transcriptional silencing of a reporter gene. Genome-wide methylation analysis in zdp mutant plants identified hundreds of hypermethylated endogenous loci. Our results show that ZDP functions downstream of ROS1 in one branch of the active DNA demethylation pathway.


Subject(s)
Arabidopsis/enzymology , DNA Methylation , Nucleotidases/chemistry , 5-Methylcytosine/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Base Sequence , Cell Nucleus/metabolism , DNA Cleavage , Gene Expression Regulation, Plant , Gene Silencing , Genes, Reporter , Genetic Loci , Genome, Plant , Kinetics , Luciferases/biosynthesis , Luciferases/genetics , Molecular Sequence Data , Mutagenesis, Insertional , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleotidases/genetics , Nucleotidases/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Transcription, Genetic
6.
J Biol Chem ; 285(30): 23032-9, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20489198

ABSTRACT

DNA cytosine methylation is an epigenetic mark that promotes gene silencing and performs critical roles during reproduction and development in both plants and animals. The genomic distribution of DNA methylation is the dynamic outcome of opposing methylation and demethylation processes. In plants, active demethylation occurs through a base excision repair pathway initiated by 5-methycytosine (5-meC) DNA glycosylases of the REPRESSOR OF SILENCING 1 (ROS1)/DEMETER (DME) family. To gain insight into the mechanism by which Arabidopsis ROS1 recognizes and excises 5-meC, we have identified those protein regions that are required for efficient DNA binding and catalysis. We have found that a short N-terminal lysine-rich domain conserved in members of the ROS1/DME family mediates strong methylation-independent binding of ROS1 to DNA and is required for efficient activity on 5-meC.G, but not for T.G processing. Removal of this domain does not significantly affect 5-meC excision from short molecules, but strongly decreases ROS1 activity on long DNA substrates. This region is not required for product binding and is not involved in the distributive behavior of the enzyme on substrates containing multiple 5-meC residues. Altogether, our results suggest that methylation-independent DNA binding allows ROS1 to perform a highly redundant search for efficient excision of a nondamaged, correctly paired base such as 5-meC in long stretches of DNA. These findings may have implications for understanding the evolution of structure and target specificity in DNA glycosylases.


Subject(s)
Arabidopsis Proteins/metabolism , DNA Methylation , DNA/chemistry , DNA/metabolism , Nuclear Proteins/metabolism , 5-Methylcytosine/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Conserved Sequence , Lysine , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Structure, Tertiary , Sequence Deletion , Substrate Specificity
7.
Proc Natl Acad Sci U S A ; 103(18): 6853-8, 2006 May 02.
Article in English | MEDLINE | ID: mdl-16624880

ABSTRACT

Cytosine methylation is an epigenetic mark that promotes gene silencing and plays important roles in development and genome defense against transposons. Methylation patterns are established and maintained by DNA methyltransferases that catalyze transfer of a methyl group from S-adenosyl-L-methionine to cytosine bases in DNA. Erasure of cytosine methylation occurs during development, but the enzymatic basis of active demethylation remains controversial. In Arabidopsis thaliana, DEMETER (DME) activates the maternal expression of two imprinted genes silenced by methylation, and REPRESSOR OF SILENCING 1 (ROS1) is required for release of transcriptional silencing of a hypermethylated transgene. DME and ROS1 encode two closely related DNA glycosylase domain proteins, but it is unknown whether they participate directly in a DNA demethylation process or counteract silencing through an indirect effect on chromatin structure. Here we show that DME and ROS1 catalyze the release of 5-methylcytosine (5-meC) from DNA by a glycosylase/lyase mechanism. Both enzymes also remove thymine, but not uracil, mismatched to guanine. DME and ROS1 show a preference for 5-meC over thymine in the symmetric dinucleotide CpG context, where most plant DNA methylation occurs. Nevertheless, they also have significant activity on both substrates at CpApG and asymmetric sequences, which are additional methylation targets in plant genomes. These findings suggest that a function of ROS1 and DME is to initiate erasure of 5-meC through a base excision repair process and provide strong biochemical evidence for the existence of an active DNA demethylation pathway in plants.


Subject(s)
5-Methylcytosine/metabolism , Arabidopsis Proteins/metabolism , DNA Glycosylases/metabolism , N-Glycosyl Hydrolases/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Arabidopsis/enzymology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Cytosine/metabolism , DNA Glycosylases/genetics , DNA Methylation , DNA Repair , Epigenesis, Genetic , Gene Silencing , Humans , Molecular Sequence Data , N-Glycosyl Hydrolases/genetics , Nuclear Proteins/genetics , Sequence Alignment , Thymine/metabolism , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...