Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 48(4): 1872-1885, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31802118

ABSTRACT

Efficient double-strand break repair in eukaryotes requires manipulation of chromatin structure. ATP-dependent chromatin remodelling enzymes facilitate different DNA repair pathways, during different stages of the cell cycle and in varied chromatin environments. The contribution of remodelling factors to double-strand break repair within heterochromatin during G2 is unclear. The human HELLS protein is a Snf2-like chromatin remodeller family member and is mutated or misregulated in several cancers and some cases of ICF syndrome. HELLS has been implicated in the DNA damage response, but its mechanistic function in repair is not well understood. We discover that HELLS facilitates homologous recombination at two-ended breaks and contributes to repair within heterochromatic regions during G2. HELLS promotes initiation of HR by facilitating end-resection and accumulation of CtIP at IR-induced foci. We identify an interaction between HELLS and CtIP and establish that the ATPase domain of HELLS is required to promote DSB repair. This function of HELLS in maintenance of genome stability is likely to contribute to its role in cancer biology and demonstrates that different chromatin remodelling activities are required for efficient repair in specific genomic contexts.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , DNA Helicases/genetics , Homologous Recombination/genetics , DNA Damage/genetics , DNA Repair/genetics , Genome, Human/genetics , Genomic Instability/genetics , Heterochromatin/genetics , Humans
2.
Elife ; 62017 11 07.
Article in English | MEDLINE | ID: mdl-29111974

ABSTRACT

General transcription factor TFIID is a key component of RNA polymerase II transcription initiation. Human TFIID is a megadalton-sized complex comprising TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs). TBP binds to core promoter DNA, recognizing the TATA-box. We identified a ternary complex formed by TBP and the histone fold (HF) domain-containing TFIID subunits TAF11 and TAF13. We demonstrate that TAF11/TAF13 competes for TBP binding with TATA-box DNA, and also with the N-terminal domain of TAF1 previously implicated in TATA-box mimicry. In an integrative approach combining crystal coordinates, biochemical analyses and data from cross-linking mass-spectrometry (CLMS), we determine the architecture of the TAF11/TAF13/TBP complex, revealing TAF11/TAF13 interaction with the DNA binding surface of TBP. We identify a highly conserved C-terminal TBP-interaction domain (CTID) in TAF13, which is essential for supporting cell growth. Our results thus have implications for cellular TFIID assembly and suggest a novel regulatory state for TFIID function.


Subject(s)
TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/metabolism , Crystallography, X-Ray , DNA/metabolism , Histone Acetyltransferases/metabolism , Humans , Mass Spectrometry , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Protein Interaction Mapping , Transcription Factor TFIID/chemistry
3.
Proc Natl Acad Sci U S A ; 112(33): E4565-70, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26240319

ABSTRACT

Faithful genome duplication and inheritance require the complete resolution of all intertwines within the parental DNA duplex. This is achieved by topoisomerase action ahead of the replication fork or by fork rotation and subsequent resolution of the DNA precatenation formed. Although fork rotation predominates at replication termination, in vitro studies have suggested that it also occurs frequently during elongation. However, the factors that influence fork rotation and how rotation and precatenation may influence other replication-associated processes are unknown. Here we analyze the causes and consequences of fork rotation in budding yeast. We find that fork rotation and precatenation preferentially occur in contexts that inhibit topoisomerase action ahead of the fork, including stable protein-DNA fragile sites and termination. However, generally, fork rotation and precatenation are actively inhibited by Timeless/Tof1 and Tipin/Csm3. In the absence of Tof1/Timeless, excessive fork rotation and precatenation cause extensive DNA damage following DNA replication. With Tof1, damage related to precatenation is focused on the fragile protein-DNA sites where fork rotation is induced. We conclude that although fork rotation and precatenation facilitate unwinding in hard-to-replicate contexts, they intrinsically disrupt normal chromosome duplication and are therefore restricted by Timeless/Tipin.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Instability , DNA Replication , DNA-Binding Proteins/physiology , DNA/chemistry , Saccharomyces cerevisiae Proteins/physiology , Cell Cycle , DNA Topoisomerases, Type II/metabolism , DNA, Fungal/chemistry , Gene Deletion , Genotype , Phosphorylation , Plasmids/metabolism , Saccharomycetales/genetics , Stochastic Processes
4.
Mol Cell ; 55(5): 723-32, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25066234

ABSTRACT

Actively transcribed regions of the genome are vulnerable to genomic instability. Recently, it was discovered that transcription is repressed in response to neighboring DNA double-strand breaks (DSBs). It is not known whether a failure to silence transcription flanking DSBs has any impact on DNA repair efficiency or whether chromatin remodelers contribute to the process. Here, we show that the PBAF remodeling complex is important for DSB-induced transcriptional silencing and promotes repair of a subset of DNA DSBs at early time points, which can be rescued by inhibiting transcription globally. An ATM phosphorylation site on BAF180, a PBAF subunit, is required for both processes. Furthermore, we find that subunits of the PRC1 and PRC2 polycomb group complexes are similarly required for DSB-induced silencing and promoting repair. Cancer-associated BAF180 mutants are unable to restore these functions, suggesting PBAF's role in repressing transcription near DSBs may contribute to its tumor suppressor activity.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , DNA Breaks , DNA Repair , Gene Expression Regulation , Transcription Factors/physiology , Binding Sites , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , DNA End-Joining Repair , DNA-Binding Proteins , HeLa Cells , Histones/metabolism , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phosphorylation , Transcription Factors/chemistry , Transcription Factors/metabolism , Ubiquitination
5.
Cell Rep ; 6(6): 973-981, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24613357

ABSTRACT

BAF180, a subunit of the PBAF chromatin remodeling complex, is frequently mutated in cancer. Although PBAF regulates transcription, it remains unclear whether this is what drives tumorigenesis in cells lacking BAF180. Based on data from yeast, we hypothesized that BAF180 may prevent tumorigenesis by promoting cohesion. Here, we show BAF180 is required for centromeric cohesion in mouse and human cells. Mutations identified in tumor samples are unable to support this activity, and also compromise cohesion-dependent functions in yeast. We provide evidence of genome instability in line with loss of cohesion, and importantly, we find dynamic chromosome instability following DNA damage in cells lacking BAF180. These data demonstrate a function for BAF180 in promoting genome stability that is distinct from its well-characterized role in transcriptional regulation, uncovering a potent mechanism for its tumor-suppressor activity.


Subject(s)
Aneuploidy , Genomic Instability , HMGB Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , DNA-Binding Proteins , Gene Expression Regulation , Humans , Mice
6.
Nucleic Acids Res ; 41(19): 9168-82, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23907388

ABSTRACT

Bromo-adjacent homology (BAH) domains are commonly found in chromatin-associated proteins and fall into two classes; Remodels the Structure of Chromatin (RSC)-like or Sir3-like. Although Sir3-like BAH domains bind nucleosomes, the binding partners of RSC-like BAH domains are currently unknown. The Rsc2 subunit of the RSC chromatin remodeling complex contains an RSC-like BAH domain and, like the Sir3-like BAH domains, we find Rsc2 BAH also interacts with nucleosomes. However, unlike Sir3-like BAH domains, we find that Rsc2 BAH can bind to recombinant purified H3 in vitro, suggesting that the mechanism of nucleosome binding is not conserved. To gain insight into the Rsc2 BAH domain, we determined its crystal structure at 2.4 Å resolution. We find that it differs substantially from Sir3-like BAH domains and lacks the motifs in these domains known to be critical for making contacts with histones. We then go on to identify a novel motif in Rsc2 BAH that is critical for efficient H3 binding in vitro and show that mutation of this motif results in defective Rsc2 function in vivo. Moreover, we find this interaction is conserved across Rsc2-related proteins. These data uncover a binding target of the Rsc2 family of BAH domains and identify a novel motif that mediates this interaction.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Histones/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Sequence , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA, Ribosomal , Gene Silencing , Models, Molecular , Molecular Sequence Data , Mutation , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
Genes Dev ; 26(23): 2590-603, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23207916

ABSTRACT

The INO80 chromatin remodeling complex functions in transcriptional regulation, DNA repair, and replication. Here we uncover a novel role for INO80 in regulating chromosome segregation. First, we show that the conserved Ies6 subunit is critical for INO80 function in vivo. Strikingly, we found that loss of either Ies6 or the Ino80 catalytic subunit results in rapid increase in ploidy. One route to polyploidy is through chromosome missegregation due to aberrant centromere structure, and we found that loss of either Ies6 or Ino80 leads to defective chromosome segregation. Importantly, we show that chromatin structure flanking centromeres is altered in cells lacking these subunits and that these alterations occur not in the Cse4-containing centromeric nucleosome, but in pericentric chromatin. We provide evidence that these effects are mediated through misincorporation of H2A.Z, and these findings indicate that H2A.Z-containing pericentric chromatin, as in higher eukaryotes with regional centromeres, is important for centromere function in budding yeast. These data reveal an important additional mechanism by which INO80 maintains genome stability.


Subject(s)
Centromere/metabolism , Chromatin Assembly and Disassembly , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Polyploidy , Saccharomyces cerevisiae Proteins/metabolism , Centromere/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , DNA Damage , Gene Expression Regulation, Fungal , Histones/genetics , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
8.
Prog Mol Biol Transl Sci ; 110: 229-61, 2012.
Article in English | MEDLINE | ID: mdl-22749148

ABSTRACT

In eukaryotes, DNA is packaged into chromatin and is therefore relatively inaccessible to DNA repair enzymes. In order to perform efficient DNA repair, ATP-dependent chromatin-remodeling enzymes are required to alter the chromatin structure near the site of damage to facilitate processing and allow access to repair enzymes. Two of the best-studied remodeling complexes involved in repair are RSC (Remodels the Structure of Chromatin) and INO80 from Saccharomyces cerevisiae, which are both conserved in higher eukaryotes. RSC is very rapidly recruited to breaks and mobilizes nucleosomes to promote phosphorylation of H2A S129 and resection. INO80 enrichment at a break occurs later and is dependent on phospho-S129 H2A. INO80 activity at the break site also facilitates resection. Consequently, both homologous recombination and nonhomologous end-joining are defective in rsc mutants, while subsets of these repair pathways are affected in ino80 mutants.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Multiprotein Complexes/metabolism , Animals , DNA Helicases/metabolism , Humans , Models, Biological
9.
Nucleic Acids Res ; 40(15): 7393-403, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22638582

ABSTRACT

In eukaryotic cells, replication past damaged sites in DNA is regulated by the ubiquitination of proliferating cell nuclear antigen (PCNA). Little is known about how this process is affected by chromatin structure. There are two isoforms of the Remodels the Structure of Chromatin (RSC) remodelling complex in yeast. We show that deletion of RSC2 results in a dramatic reduction in the level of PCNA ubiquitination after DNA-damaging treatments, whereas no such effect was observed after deletion of RSC1. Similarly, depletion of the BAF180 component of the corresponding PBAF (Polybromo BRG1 (Brahma-Related Gene 1) Associated Factor) complex in human cells led to a similar reduction in PCNA ubiquitination. Remarkably, we found that depletion of BAF180 resulted after UV-irradiation, in a reduction not only of ubiquitinated PCNA but also of chromatin-associated unmodified PCNA and Rad18 (the E3 ligase that ubiquitinates PCNA). This was accompanied by a modest decrease in fork progression. We propose a model to account for these findings that postulates an involvement of PBAF in repriming of replication downstream from replication forks blocked at sites of DNA damage. In support of this model, chromatin immunoprecipitation data show that the RSC complex in yeast is present in the vicinity of the replication forks, and by extrapolation, this is also likely to be the case for the PBAF complex in human cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle , Cell Line , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins/physiology , Gene Deletion , Humans , Nuclear Proteins/antagonists & inhibitors , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology , Ubiquitin-Protein Ligases , Ubiquitination
10.
Biochem Soc Trans ; 40(2): 364-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22435813

ABSTRACT

Chromatin remodelling complexes alter the structure of chromatin and have central roles in all DNA-templated activities, including regulation of gene expression and DNA repair. Mutations in subunits of the PBAF (polybromo/Brg1-associated factor) or SWI/SNF-B remodelling complex, including BAF180, are frequently associated with cancer. There are six potential acetyl-lysine-binding BDs (bromodomains) in BAF180, which may function to target the PBAF complex to promoters or sites of DNA repair. In the present review, we discuss what is currently known about the BDs of BAF180 and their potential significance in cancer.


Subject(s)
Neoplasms/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Animals , Humans , Mutation/genetics , Neoplasms/genetics , Protein Binding , Protein Structure, Tertiary , Protein Subunits/metabolism
11.
PLoS One ; 7(2): e32016, 2012.
Article in English | MEDLINE | ID: mdl-22359657

ABSTRACT

The RSC chromatin remodeling complex has been implicated in contributing to DNA double-strand break (DSB) repair in a number of studies. Both survival and levels of H2A phosphorylation in response to damage are reduced in the absence of RSC. Importantly, there is evidence for two isoforms of this complex, defined by the presence of either Rsc1 or Rsc2. Here, we investigated whether the two isoforms of RSC provide distinct contributions to DNA damage responses. First, we established that the two isoforms of RSC differ in the presence of Rsc1 or Rsc2 but otherwise have the same subunit composition. We found that both rsc1 and rsc2 mutant strains have intact DNA damage-induced checkpoint activity and transcriptional induction. In addition, both strains show reduced non-homologous end joining activity and have a similar spectrum of DSB repair junctions, suggesting perhaps that the two complexes provide the same functions. However, the hypersensitivity of a rsc1 strain cannot be complemented with an extra copy of RSC2, and likewise, the hypersensitivity of the rsc2 strain remains unchanged when an additional copy of RSC1 is present, indicating that the two proteins are unable to functionally compensate for one another in DNA damage responses. Rsc1, but not Rsc2, is required for nucleosome sliding flanking a DNA DSB. Interestingly, while swapping the domains from Rsc1 into the Rsc2 protein does not compromise hypersensitivity to DNA damage suggesting they are functionally interchangeable, the BAH domain from Rsc1 confers upon Rsc2 the ability to remodel chromatin at a DNA break. These data demonstrate that, despite the similarity between Rsc1 and Rsc2, the two different isoforms of RSC provide distinct functions in DNA damage responses, and that at least part of the functional specificity is dictated by the BAH domains.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/physiology , DNA Repair , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/physiology , DNA Damage , Fungal Proteins , Protein Isoforms , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics
12.
J Biol Chem ; 282(38): 27693-701, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17652077

ABSTRACT

DNA double strand breaks (DSBs) are potentially serious chromosomal lesions. However, cells sometimes deliberately cleave their own DNA to facilitate certain chromosomal processes, and there is much interest in how such self-inflicted breaks are effectively managed. Eukaryotic DSBs occur in the context of chromatin and the RSC chromatin-remodeling ATPase complex has been shown to promote DSB repair at the budding yeast MAT locus DSB, created by the HO endonuclease during mating type switching. We show that the role of RSC at MAT is highly specialized. The Rsc1p subunit of RSC directs nucleosome sliding immediately after DSB creation at both MAT and generally and is required for efficient DNA damage-induced histone H2A phosphorylation and strand resection during repair by homologous recombination. However, the Rsc2p and Rsc7p subunits are additionally required to set up a basal MAT locus structure. This RSC-dependent chromatin structure at MAT ensures accessibility to the HO endonuclease. The RSC complex therefore has chromatin remodeling roles both before and after DSB induction at MAT, promoting both DNA cleavage and subsequent repair.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Repair , Gene Expression Regulation, Fungal , Chromosomal Proteins, Non-Histone/metabolism , Fungal Proteins , Histones/chemistry , Models, Biological , Models, Genetic , Nucleosomes/metabolism , Phosphorylation , Protein Structure, Tertiary , Recombination, Genetic , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
13.
Cell ; 124(3): 507-20, 2006 Feb 10.
Article in English | MEDLINE | ID: mdl-16469698

ABSTRACT

Coupling of transcription and DNA repair in bacteria is mediated by transcription-repair coupling factor (TRCF, the product of the mfd gene), which removes transcription elongation complexes stalled at DNA lesions and recruits the nucleotide excision repair machinery to the site. Here we describe the 3.2 A-resolution X-ray crystal structure of Escherichia coli TRCF. The structure consists of a compact arrangement of eight domains, including a translocation module similar to the SF2 ATPase RecG, and a region of structural similarity to UvrB. Biochemical and genetic experiments establish that another domain with structural similarity to the Tudor-like domain of the transcription elongation factor NusG plays a critical role in TRCF/RNA polymerase interactions. Comparison with the translocation module of RecG as well as other structural features indicate that TRCF function involves large-scale conformational changes. These data, along with a structural model for the interaction of TRCF with the transcription elongation complex, provide mechanistic insights into TRCF function.


Subject(s)
Bacterial Proteins/chemistry , DNA Repair , Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Transcription Factors/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...