Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38260523

ABSTRACT

Mammalian DNA replication employs several RecQ DNA helicases to orchestrate the faithful duplication of genetic information. Helicase function is often coupled to the activity of specific nucleases, but how helicase and nuclease activities are co-directed is unclear. Here we identify the inactive ubiquitin-specific protease, USP50, as a ubiquitin-binding and chromatin-associated protein required for ongoing replication, fork restart, telomere maintenance and cellular survival during replicative stress. USP50 supports WRN:FEN1 at stalled replication forks, suppresses MUS81-dependent fork collapse and restricts double-strand DNA breaks at GC-rich sequences. Surprisingly we find that cells depleted for USP50 and recovering from a replication block exhibit increased DNA2 and RECQL4 foci and that the defects in ongoing replication, poor fork restart and increased fork collapse seen in these cells are mediated by DNA2, RECQL4 and RECQL5. These data define a novel ubiquitin-dependent pathway that promotes the balance of helicase: nuclease use at ongoing and stalled replication forks.

2.
Nat Commun ; 14(1): 7834, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38030626

ABSTRACT

A synthetic lethal relationship exists between disruption of polymerase theta (Polθ), and loss of either 53BP1 or homologous recombination (HR) proteins, including BRCA1; however, the mechanistic basis of these observations are unclear. Here we reveal two distinct mechanisms of Polθ synthetic lethality, identifying dual influences of 1) whether Polθ is lost or inhibited, and 2) the underlying susceptible genotype. Firstly, we find that the sensitivity of BRCA1/2- and 53BP1-deficient cells to Polθ loss, and 53BP1-deficient cells to Polθ inhibition (ART558) requires RAD52, and appropriate reduction of RAD52 can ameliorate these phenotypes. We show that in the absence of Polθ, RAD52 accumulations suppress ssDNA gap-filling in G2/M and encourage MRE11 nuclease accumulation. In contrast, the survival of BRCA1-deficient cells treated with Polθ inhibitor are not restored by RAD52 suppression, and ssDNA gap-filling is prevented by the chemically inhibited polymerase itself. These data define an additional role for Polθ, reveal the mechanism underlying synthetic lethality between 53BP1, BRCA1/2 and Polθ loss, and indicate genotype-dependent Polθ inhibitor mechanisms.


Subject(s)
BRCA1 Protein , Synthetic Lethal Mutations , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Homologous Recombination , DNA Repair , Tumor Suppressor p53-Binding Protein 1/metabolism , DNA Polymerase theta
3.
Sci Rep ; 12(1): 21343, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494414

ABSTRACT

Monoclonal antibodies (MAb) to members of the Small Ubiquitin-like modifier (SUMO) family are essential tools in the study of cellular SUMOylation. However, many anti-SUMO MAbs are poorly validated, and antibody matching to detection format is without an evidence base. Here we test the specificity and sensitivity of twenty-four anti-SUMO MAbs towards monomeric and polymeric SUMO1-4 in dot-blots, immunoblots, immunofluorescence and immunoprecipitation. We find substantial variability between SUMO MAbs for different conjugation states, for detecting increased SUMOylation in response to thirteen different stress agents, and as enrichment reagents for SUMOylated RanGAP1 or KAP1. All four anti-SUMO4 monoclonal antibodies tested cross-reacted wit SUMO2/3, and several SUMO2/3 monoclonal antibodies cross-reacted with SUMO4. These data characterize the specificity of twenty-four anti-SUMO antibodies across commonly used assays, creating an enabling resource for the SUMO research community.


Subject(s)
Antibodies, Monoclonal , Small Ubiquitin-Related Modifier Proteins , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin , Sumoylation , Immunoprecipitation
4.
Metallomics ; 14(7)2022 07 25.
Article in English | MEDLINE | ID: mdl-35689667

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a disease that remains refractory to existing treatments including the nucleoside analogue gemcitabine. In the current study we demonstrate that an organometallic nucleoside analogue, the ferronucleoside 1-(S,Rp), is cytotoxic in a panel of PDAC cell lines including gemcitabine-resistant MIAPaCa2, with IC50 values comparable to cisplatin. Biochemical studies show that the mechanism of action is inhibition of DNA replication, S-phase cell cycle arrest and stalling of DNA-replication forks, which were directly observed at single molecule resolution by DNA-fibre fluorography. In agreement with this, transcriptional changes following treatment with 1-(S,Rp) include activation of three of the four genes (HUS1, RAD1, RAD17) of the 9-1-1 check point complex clamp and two of the three genes (MRE11, NBN) that form the MRN complex as well as activation of multiple downstream targets. Furthermore, there was evidence of phosphorylation of checkpoint kinases 1 and 2 as well as RPA1 and gamma H2AX, all of which are considered biochemical markers of replication stress. Studies in p53-deficient cell lines showed activation of CDKN1A (p21) and GADD45A by 1-(S,Rp) was at least partially independent of p53. In conclusion, because of its potency and activity in gemcitabine-resistant cells, 1-(S,Rp) is a promising candidate molecule for development of new treatments for PDAC.


Subject(s)
DNA Replication , Nucleosides , Pancreatic Neoplasms , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Metallocenes , Nucleosides/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , S Phase , Tumor Suppressor Protein p53/metabolism , Pancreatic Neoplasms
5.
J Cell Sci ; 135(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35748225

ABSTRACT

Commonly applied super-resolution light microscopies have provided insight into subcellular processes at the nanoscale. However, imaging depth, speed, throughput and cost remain significant challenges, limiting the numbers of three-dimensional (3D) nanoscale processes that can be investigated and the number of laboratories able to undertake such analysis. Expansion microscopy (ExM) solves many of these limitations, but its application to imaging nuclear processes has been constrained by concerns of unequal nuclear expansion. Here, we demonstrate the conditions for isotropic expansion of the nucleus at a resolution equal to or better than 120-130 nm (pre-expansion). Using the DNA damage response proteins BRCA1, 53BP1 (also known as TP53BP1) and RAD51 as exemplars, we quantitatively describe the 3D nanoscale organisation of over 50,000 DNA damage response structures. We demonstrate the ability to assess chromatin-regulated events and show the simultaneous assessment of four elements. This study thus demonstrates how ExM can contribute to the investigation of nanoscale nuclear processes.


Subject(s)
Chromatin , Microscopy , Cell Nucleus , Microscopy/methods
7.
Semin Cell Dev Biol ; 113: 14-26, 2021 05.
Article in English | MEDLINE | ID: mdl-32653304

ABSTRACT

In response to replication hindrances, DNA replication forks frequently stall and are remodelled into a four-way junction. In such a structure the annealed nascent strand is thought to resemble a DNA double-strand break and remodelled forks are vulnerable to nuclease attack by MRE11 and DNA2. Proteins that promote the recruitment, loading and stabilisation of RAD51 onto single-stranded DNA for homology search and strand exchange in homologous recombination (HR) repair and inter-strand cross-link repair also act to set up RAD51-mediated protection of nascent DNA at stalled replication forks. However, despite the similarities of these pathways, several lines of evidence indicate that fork protection is not simply analogous to the RAD51 loading step of HR. Protection of stalled forks not only requires separate functions of a number of recombination proteins, but also utilises nucleases important for the resection steps of HR in alternative ways. Here we discuss how fork protection arises and how its differences with HR give insights into the differing contexts of these two pathways.


Subject(s)
DNA Damage/genetics , DNA Replication/genetics , Genomic Instability/genetics , Homologous Recombination/genetics , Humans
8.
Nat Commun ; 11(1): 5863, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33203852

ABSTRACT

Stalled replication forks can be restarted and repaired by RAD51-mediated homologous recombination (HR), but HR can also perform post-replicative repair after bypass of the obstacle. Bulky DNA adducts are important replication-blocking lesions, but it is unknown whether they activate HR at stalled forks or behind ongoing forks. Using mainly BPDE-DNA adducts as model lesions, we show that HR induced by bulky adducts in mammalian cells predominantly occurs at post-replicative gaps formed by the DNA/RNA primase PrimPol. RAD51 recruitment under these conditions does not result from fork stalling, but rather occurs at gaps formed by PrimPol re-priming and resection by MRE11 and EXO1. In contrast, RAD51 loading at double-strand breaks does not require PrimPol. At bulky adducts, PrimPol promotes sister chromatid exchange and genetic recombination. Our data support that HR at bulky adducts in mammalian cells involves post-replicative gap repair and define a role for PrimPol in HR-mediated DNA damage tolerance.


Subject(s)
DNA Adducts/genetics , DNA Primase/metabolism , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination/physiology , Multifunctional Enzymes/metabolism , 4-Nitroquinoline-1-oxide/toxicity , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/metabolism , Benz(a)Anthracenes/administration & dosage , Benz(a)Anthracenes/toxicity , Cell Line , DNA Adducts/metabolism , DNA Primase/genetics , DNA, Single-Stranded , DNA-Directed DNA Polymerase/genetics , Humans , Multifunctional Enzymes/genetics , Quinolones/toxicity , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Single Molecule Imaging , Sister Chromatid Exchange
9.
Mol Cell Oncol ; 6(6): e1656500, 2019.
Article in English | MEDLINE | ID: mdl-31692900

ABSTRACT

The breast cancer type-1 susceptibility protein (BRCA1) contributes to genome integrity through homologous recombinational DNA repair and by protecting stalled replication forks from nucleolytic degradation. We recently discovered that fork protection requires a conformational change of BRCA1 unimportant to homologous recombination repair, indicating separate roles for BRCA1 in these pathways.

10.
Front Mol Biosci ; 6: 79, 2019.
Article in English | MEDLINE | ID: mdl-31552267

ABSTRACT

DNA double-strand breaks (DSBs) occur in our cells in the context of chromatin. This type of lesion is toxic, entirely preventing genome continuity and causing cell death or terminal arrest. Several repair mechanisms can act on DNA surrounding a DSB, only some of which carry a low risk of mutation, so that which repair process is utilized is critical to the stability of genetic material of cells. A key component of repair outcome is the degree of DNA resection directed to either side of the break site. This in turn determines the subsequent forms of repair in which DNA homology plays a part. Here we will focus on chromatin and chromatin-bound complexes which constitute the "mountains" that block resection, with a particular focus on how the breast and ovarian cancer predisposition protein-1 (BRCA1) contributes to repair outcomes through overcoming these blocks.

11.
Nature ; 571(7766): 521-527, 2019 07.
Article in English | MEDLINE | ID: mdl-31270457

ABSTRACT

The integrity of genomes is constantly threatened by problems encountered by the replication fork. BRCA1, BRCA2 and a subset of Fanconi anaemia proteins protect stalled replication forks from degradation by nucleases, through pathways that involve RAD51. The contribution and regulation of BRCA1 in replication fork protection, and how this role relates to its role in homologous recombination, is unclear. Here we show that BRCA1 in complex with BARD1, and not the canonical BRCA1-PALB2 interaction, is required for fork protection. BRCA1-BARD1 is regulated by a conformational change mediated by the phosphorylation-directed prolyl isomerase PIN1. PIN1 activity enhances BRCA1-BARD1 interaction with RAD51, thereby increasing the presence of RAD51 at stalled replication structures. We identify genetic variants of BRCA1-BARD1 in patients with cancer that exhibit poor protection of nascent strands but retain homologous recombination proficiency, thus defining domains of BRCA1-BARD1 that are required for fork protection and associated with cancer development. Together, these findings reveal a BRCA1-mediated pathway that governs replication fork protection.


Subject(s)
BRCA1 Protein/chemistry , BRCA1 Protein/metabolism , DNA Replication , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , BRCA1 Protein/genetics , Cell Line, Tumor , DNA Replication/genetics , Genomic Instability/genetics , Humans , Isomerism , Mutation , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Rad51 Recombinase/metabolism
12.
Nucleic Acids Res ; 47(9): 4476-4494, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30854564

ABSTRACT

IRF1 (Interferon Regulatory Factor-1) is the prototype of the IRF family of DNA binding transcription factors. IRF1 protein expression is regulated by transient up-regulation in response to external stimuli followed by rapid degradation via the ubiquitin-proteasome system. Here we report that DNA bound IRF1 turnover is promoted by GSK3ß (Glycogen Synthase Kinase 3ß) via phosphorylation of the T181 residue which generates a phosphodegron for the SCF (Skp-Cul-Fbox) ubiquitin E3-ligase receptor protein Fbxw7α (F-box/WD40 7). This regulated turnover is essential for IRF1 activity, as mutation of T181 results in an improperly stabilized protein that accumulates at target promoters but fails to induce RNA-Pol-II elongation and subsequent transcription of target genes. Consequently, the anti-proliferative activity of IRF1 is lost in cell lines expressing T181A mutant. Further, cell lines with dysfunctional Fbxw7 are less sensitive to IRF1 overexpression, suggesting an important co-activator function for this ligase complex. As T181 phosphorylation requires both DNA binding and RNA-Pol-II elongation, we propose that this event acts to clear 'spent' molecules of IRF1 from transcriptionally engaged target promoters.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/genetics , Glycogen Synthase Kinase 3 beta/genetics , Interferon Regulatory Factor-1/genetics , SKP Cullin F-Box Protein Ligases/genetics , Animals , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Mice , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Protein Binding/genetics , Transcription Factors/genetics , Ubiquitination/genetics
13.
Genes Dev ; 33(5-6): 333-347, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30796017

ABSTRACT

SUMOylation (small ubiquitin-like modifier) in the DNA double-strand break (DSB) response regulates recruitment, activity, and clearance of repair factors. However, our understanding of a role for deSUMOylation in this process is limited. Here we identify different mechanistic roles for deSUMOylation in homologous recombination (HR) and nonhomologous end joining (NHEJ) through the investigation of the deSUMOylase SENP2. We found that regulated deSUMOylation of MDC1 prevents excessive SUMOylation and its RNF4-VCP mediated clearance from DSBs, thereby promoting NHEJ. In contrast, we show that HR is differentially sensitive to SUMO availability and SENP2 activity is needed to provide SUMO. SENP2 is amplified as part of the chromosome 3q amplification in many cancers. Increased SENP2 expression prolongs MDC1 focus retention and increases NHEJ and radioresistance. Collectively, our data reveal that deSUMOylation differentially primes cells for responding to DSBs and demonstrates the ability of SENP2 to tune DSB repair responses.


Subject(s)
Cysteine Endopeptidases/metabolism , DNA End-Joining Repair/genetics , DNA Repair/genetics , Homologous Recombination/genetics , Sumoylation/genetics , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/radiation effects , Cysteine Endopeptidases/genetics , DNA Breaks, Double-Stranded , HEK293 Cells , HeLa Cells , Humans , Infrared Rays , Nuclear Proteins/metabolism , Radiation Tolerance/genetics , Signal Transduction/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Valosin Containing Protein/metabolism
14.
J Biol Chem ; 294(2): 424-436, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30373771

ABSTRACT

Ubiquitin-specific proteases (USPs) reverse ubiquitination and regulate virtually all cellular processes. Defined noncatalytic domains in USP4 and USP15 are known to interact with E3 ligases and substrate recruitment factors. No such interactions have been reported for these domains in the paralog USP11, a key regulator of DNA double-strand break repair by homologous recombination. We hypothesized that USP11 domains adjacent to its protease domain harbor unique peptide-binding sites. Here, using a next-generation phage display (NGPD) strategy, combining phage display library screening with next-generation sequencing, we discovered unique USP11-interacting peptide motifs. Isothermal titration calorimetry disclosed that the highest affinity peptides (KD of ∼10 µm) exhibit exclusive selectivity for USP11 over USP4 and USP15 in vitro Furthermore, a crystal structure of a USP11-peptide complex revealed a previously unknown binding site in USP11's noncatalytic ubiquitin-like (UBL) region. This site interacted with a helical motif and is absent in USP4 and USP15. Reporter assays using USP11-WT versus a binding pocket-deficient double mutant disclosed that this binding site modulates USP11's function in homologous recombination-mediated DNA repair. The highest affinity USP11 peptide binder fused to a cellular delivery sequence induced significant nuclear localization and cell cycle arrest in S phase, affecting the viability of different mammalian cell lines. The USP11 peptide ligands and the paralog-specific functional site in USP11 identified here provide a framework for the development of new biochemical tools and therapeutic agents. We propose that an NGPD-based strategy for identifying interacting peptides may be applied also to other cellular targets.


Subject(s)
Peptides/chemistry , Peptides/metabolism , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Crystallography, X-Ray , DNA Repair , Homologous Recombination , Humans , Kinetics , Ligands , Mice , Molecular Sequence Data , Peptides/genetics , Protein Domains , Thiolester Hydrolases/genetics , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitination
15.
Nat Commun ; 9(1): 229, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335415

ABSTRACT

BRCA1-BARD1-catalyzed ubiquitination of histone H2A is an important regulator of the DNA damage response, priming chromatin for repair by homologous recombination. However, no specific deubiquitinating enzymes (DUBs) are known to antagonize this function. Here we identify ubiquitin specific protease-48 (USP48) as a H2A DUB, specific for the C-terminal BRCA1 ubiquitination site. Detailed biochemical analysis shows that an auxiliary ubiquitin, an additional ubiquitin that itself does not get cleaved, modulates USP48 activity, which has possible implications for its regulation in vivo. In cells we reveal that USP48 antagonizes BRCA1 E3 ligase function and in BRCA1-proficient cells loss of USP48 results in positioning 53BP1 further from the break site and in extended resection lengths. USP48 repression confers a survival benefit to cells treated with camptothecin and its activity acts to restrain gene conversion and mutagenic single-strand annealing. We propose that USP48 promotes genome stability by antagonizing BRCA1 E3 ligase function.


Subject(s)
BRCA1 Protein/metabolism , Histones/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin/metabolism , Animals , BRCA1 Protein/genetics , Base Sequence , Cell Line, Tumor , Cells, Cultured , DNA Repair , HeLa Cells , Humans , Kinetics , Mice, Knockout , RNA Interference , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
16.
Philos Trans R Soc Lond B Biol Sci ; 372(1731)2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28847818

ABSTRACT

The response to a DNA double-stranded break in mammalian cells is a process of sensing and signalling the lesion. It results in halting the cell cycle and local transcription and in the mediation of the DNA repair process itself. The response is launched through a series of post-translational modification signalling events coordinated by phosphorylation and ubiquitination. More recently modifications of proteins by Small Ubiquitin-like MOdifier (SUMO) isoforms have also been found to be key to coordination of the response (Morris et al. 2009 Nature462, 886-890 (doi:10.1038/nature08593); Galanty et al. 2009 Nature462, 935-939 (doi:10.1038/nature08657)). However our understanding of the role of SUMOylation is slight compared with our growing knowledge of how ubiquitin drives signal amplification and key chromatin interactions. In this review we consider our current knowledge of how SUMO isoforms, SUMO conjugation machinery, SUMO proteases and SUMO-interacting proteins contribute to directing altered chromatin states and to repair-protein kinetics at a double-stranded DNA lesion in mammalian cells. We also consider the gaps in our understanding.This article is part of the themed issue 'Chromatin modifiers and remodellers in DNA repair and signalling'.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Mammals/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Animals , Humans
17.
J Mol Biol ; 429(22): 3376-3387, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28527786

ABSTRACT

In recent years, our knowledge of the varied role that ubiquitination plays in promoting signal amplification, novel protein interactions, and protein turnover has progressed rapidly. This is particularly remarkable in the examination of how DNA double-stranded breaks (DSBs) are repaired, with many components of the ubiquitin (Ub) conjugation, de-conjugation, and recognition machinery now identified as key factors in DSB repair. In addition, a member of the Ub-like family, small Ub-like modifier (SUMO), has also been recognised as integral for efficient repair. Here, we summarise our emerging understanding of SUMOylation both as a distinct modification and as a cooperative modification with Ub, using the cellular response to DNA DSBs as the primary setting to compare these modifications.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Eukaryotic Cells/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitin/metabolism , Eukaryotic Cells/enzymology
18.
Nucleus ; 8(2): 116-125, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28032817

ABSTRACT

The protein product of the breast and ovarian cancer gene, BRCA1, is part of an obligate heterodimer with BARD1. Together these RING bearing proteins act as an E3 ubiquitin ligase. Several functions have been attributed to BRCA1 that contribute to genome integrity but which of these, if any, require this enzymatic function was unclear. Here we review recent studies clarifying the role of BRCA1 E3 ubiquitin ligase in DNA repair. Perhaps the most surprising finding is the narrow range of BRCA1 functions this activity relates to. Remarkably ligase activity promotes chromatin remodelling and 53BP1 positioning through the remodeller SMARCAD1, but the activity is dispensable for the cellular survival in response to cisplatin or replication stressing agents. Implications for therapy response and tumor susceptibility are discussed.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Ubiquitin-Protein Ligases/metabolism , Animals , BRCA1 Protein/chemistry , Chromatin/metabolism , Gene Silencing , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
19.
Nat Struct Mol Biol ; 23(7): 647-55, 2016 07.
Article in English | MEDLINE | ID: mdl-27239795

ABSTRACT

The opposing activities of 53BP1 and BRCA1 influence pathway choice in DNA double-strand-break repair. How BRCA1 counteracts the inhibitory effect of 53BP1 on DNA resection and homologous recombination is unknown. Here we identify the site of BRCA1-BARD1 required for priming ubiquitin transfer from E2∼ubiquitin and demonstrate that BRCA1-BARD1's ubiquitin ligase activity is required for repositioning 53BP1 on damaged chromatin. We confirm H2A ubiquitination by BRCA1-BARD1 and show that an H2A-ubiquitin fusion protein promotes DNA resection and repair in BARD1-deficient cells. BRCA1-BARD1's function in homologous recombination requires the chromatin remodeler SMARCAD1. SMARCAD1 binding to H2A-ubiquitin and optimal localization to sites of damage and activity in DNA repair requires its ubiquitin-binding CUE domains. SMARCAD1 is required for 53BP1 repositioning, and the need for SMARCAD1 in olaparib or camptothecin resistance is alleviated by 53BP1 loss. Thus, BRCA1-BARD1 ligase activity and subsequent SMARCAD1-dependent chromatin remodeling are critical regulators of DNA repair.


Subject(s)
BRCA1 Protein/genetics , Chromatin/metabolism , DNA Helicases/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Recombinational DNA Repair , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , BRCA1 Protein/metabolism , Binding Sites , Camptothecin/pharmacology , Chromatin/chemistry , Chromatin/drug effects , Cloning, Molecular , DNA Breaks, Double-Stranded , DNA Cleavage/drug effects , DNA Helicases/metabolism , DNA, Neoplasm/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Phthalazines/pharmacology , Piperazines/pharmacology , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
20.
Epigenetics ; 10(12): 1121-32, 2015.
Article in English | MEDLINE | ID: mdl-26727311

ABSTRACT

Germline pathogenic mutations in BRCA1 increase risk of developing breast cancer. Screening for mutations in BRCA1 frequently identifies sequence variants of unknown pathogenicity and recent work has aimed to develop methods for determining pathogenicity. We previously observed that tumor DNA methylation can differentiate BRCA1-mutated from BRCA1-wild type tumors. We hypothesized that we could predict pathogenicity of variants based on DNA methylation profiles of tumors that had arisen in carriers of unclassified variants. We selected 150 FFPE breast tumor DNA samples [47 BRCA1 pathogenic mutation carriers, 65 BRCAx (BRCA1-wild type), 38 BRCA1 test variants] and analyzed a subset (n=54) using the Illumina 450K methylation platform, using the remaining samples for bisulphite pyrosequencing validation. Three validated markers (BACH2, C8orf31, and LOC654342) were combined with sequence bioinformatics in a model to predict pathogenicity of 27 variants (independent test set). Predictions were compared with standard multifactorial likelihood analysis. Prediction was consistent for c.5194-12G>A (IVS 19-12 G>A) (P>0.99); 13 variants were considered not pathogenic or likely not pathogenic using both approaches. We conclude that tumor DNA methylation data alone has potential to be used in prediction of BRCA1 variant pathogenicity but is not independent of estrogen receptor status and grade, which are used in current multifactorial models to predict pathogenicity.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Genes, BRCA1 , Genetic Predisposition to Disease , Breast Neoplasms/pathology , Computational Biology , DNA Mutational Analysis , Female , Genetic Markers , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...