Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 220(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34003252

ABSTRACT

The histone demethylase KDM5A erases histone H3 lysine 4 methylation, which is involved in transcription and DNA damage responses (DDRs). While DDR functions of KDM5A have been identified, how KDM5A recognizes DNA lesion sites within chromatin is unknown. Here, we identify two factors that act upstream of KDM5A to promote its association with DNA damage sites. We have identified a noncanonical poly(ADP-ribose) (PAR)-binding region unique to KDM5A. Loss of the PAR-binding region or treatment with PAR polymerase (PARP) inhibitors (PARPi's) blocks KDM5A-PAR interactions and DNA repair functions of KDM5A. The histone variant macroH2A1.2 is also specifically required for KDM5A recruitment and function at DNA damage sites, including homology-directed repair of DNA double-strand breaks and repression of transcription at DNA breaks. Overall, this work reveals the importance of PAR binding and macroH2A1.2 in KDM5A recognition of DNA lesion sites that drive transcriptional and repair activities at DNA breaks within chromatin that are essential for maintaining genome integrity.


Subject(s)
DNA/genetics , Histones/genetics , Recombinational DNA Repair/genetics , Retinoblastoma-Binding Protein 2/genetics , Chromatin/genetics , DNA Breaks, Double-Stranded , DNA Damage , Humans , Poly Adenosine Diphosphate Ribose/genetics , Poly(ADP-ribose) Polymerases/genetics
2.
Cell ; 184(2): 384-403.e21, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450205

ABSTRACT

Many oncogenic insults deregulate RNA splicing, often leading to hypersensitivity of tumors to spliceosome-targeted therapies (STTs). However, the mechanisms by which STTs selectively kill cancers remain largely unknown. Herein, we discover that mis-spliced RNA itself is a molecular trigger for tumor killing through viral mimicry. In MYC-driven triple-negative breast cancer, STTs cause widespread cytoplasmic accumulation of mis-spliced mRNAs, many of which form double-stranded structures. Double-stranded RNA (dsRNA)-binding proteins recognize these endogenous dsRNAs, triggering antiviral signaling and extrinsic apoptosis. In immune-competent models of breast cancer, STTs cause tumor cell-intrinsic antiviral signaling, downstream adaptive immune signaling, and tumor cell death. Furthermore, RNA mis-splicing in human breast cancers correlates with innate and adaptive immune signatures, especially in MYC-amplified tumors that are typically immune cold. These findings indicate that dsRNA-sensing pathways respond to global aberrations of RNA splicing in cancer and provoke the hypothesis that STTs may provide unexplored strategies to activate anti-tumor immune pathways.


Subject(s)
Antiviral Agents/pharmacology , Immunity/drug effects , Spliceosomes/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Adaptive Immunity/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytoplasm/drug effects , Cytoplasm/metabolism , Female , Gene Amplification/drug effects , Humans , Introns/genetics , Mice , Molecular Targeted Therapy , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Double-Stranded/metabolism , Signal Transduction/drug effects , Spliceosomes/drug effects , Triple Negative Breast Neoplasms/genetics
3.
Biochem Cell Biol ; 98(1): 42-49, 2020 02.
Article in English | MEDLINE | ID: mdl-30620620

ABSTRACT

FK506-binding proteins (FKBPs) alter the conformation of proteins via cis-trans isomerization of prolyl-peptide bonds. While this activity can be demonstrated in vitro, the intractability of detecting prolyl isomerization events in cells has limited our understanding of the biological processes regulated by FKBPs. Here we report that FKBP25 is an active participant in the repair of DNA double-strand breaks (DSBs). FKBP25 influences DSB repair pathway choice by promoting homologous recombination (HR) and suppressing single-strand annealing (SSA). Consistent with this observation, cells depleted of FKBP25 form fewer Rad51 repair foci in response to etoposide and ionizing radiation, and they are reliant on the SSA repair factor Rad52 for viability. We find that FKBP25's catalytic activity is required for promoting DNA repair, which is the first description of a biological function for this enzyme activity. Consistent with the importance of the FKBP catalytic site in HR, rapamycin treatment also impairs homologous recombination, and this effect is at least in part independent of mTor. Taken together these results identify FKBP25 as a component of the DNA DSB repair pathway.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Tacrolimus Binding Proteins/metabolism , Cell Proliferation , Fluorescent Antibody Technique , Humans , Tumor Cells, Cultured
4.
Genes Dev ; 33(23-24): 1751-1774, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31753913

ABSTRACT

Bromodomain proteins (BRD) are key chromatin regulators of genome function and stability as well as therapeutic targets in cancer. Here, we systematically delineate the contribution of human BRD proteins for genome stability and DNA double-strand break (DSB) repair using several cell-based assays and proteomic interaction network analysis. Applying these approaches, we identify 24 of the 42 BRD proteins as promoters of DNA repair and/or genome integrity. We identified a BRD-reader function of PCAF that bound TIP60-mediated histone acetylations at DSBs to recruit a DUB complex to deubiquitylate histone H2BK120, to allowing direct acetylation by PCAF, and repair of DSBs by homologous recombination. We also discovered the bromo-and-extra-terminal (BET) BRD proteins, BRD2 and BRD4, as negative regulators of transcription-associated RNA-DNA hybrids (R-loops) as inhibition of BRD2 or BRD4 increased R-loop formation, which generated DSBs. These breaks were reliant on topoisomerase II, and BRD2 directly bound and activated topoisomerase I, a known restrainer of R-loops. Thus, comprehensive interactome and functional profiling of BRD proteins revealed new homologous recombination and genome stability pathways, providing a framework to understand genome maintenance by BRD proteins and the effects of their pharmacological inhibition.


Subject(s)
Genomic Instability , R-Loop Structures , Recombinational DNA Repair/genetics , Transcription Factors/genetics , Acetylation , Cell Line , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , HEK293 Cells , HeLa Cells , Humans , Trans-Activators/metabolism , Transcription Factors/analysis , Ubiquitination , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
5.
Mutat Res Rev Mutat Res ; 780: 37-47, 2019.
Article in English | MEDLINE | ID: mdl-31395347

ABSTRACT

Preserving genome function and stability are paramount for ensuring cellular homeostasis, an imbalance in which can promote diseases including cancer. In the presence of DNA lesions, cells activate pathways referred to as the DNA damage response (DDR). As nuclear DNA is bound by histone proteins and organized into chromatin in eukaryotes, DDR pathways have evolved to sense, signal and repair DNA damage within the chromatin environment. Histone proteins, which constitute the building blocks of chromatin, are highly modified by post-translational modifications (PTMs) that regulate chromatin structure and function. An essential histone PTM involved in the DDR is histone methylation, which is regulated by histone methyltransferase (HMT) and histone demethylase (HDM) enzymes that add and remove methyl groups on lysine and arginine residues within proteins respectively. Methylated histones can alter how proteins interact with chromatin, including their ability to be bound by reader proteins that recognize these PTMs. Here, we review histone methylation in the context of the DDR, focusing on DNA double-strand breaks (DSBs), a particularly toxic lesion that can trigger genome instability and cell death. We provide a comprehensive overview of histone methylation changes that occur in response to DNA damage and how the enzymes and reader proteins of these marks orchestrate the DDR. Finally, as many epigenetic pathways including histone methylation are altered in cancer, we discuss the potential involvement of these pathways in the etiology and treatment of this disease.


Subject(s)
DNA Damage/genetics , Histones/metabolism , Animals , Chromatin/genetics , Humans , Methylation , Signal Transduction/genetics
6.
Methods Mol Biol ; 1999: 61-74, 2019.
Article in English | MEDLINE | ID: mdl-31127569

ABSTRACT

Maintenance of genomic integrity depends on the spatiotemporal recruitment and regulation of DNA damage response and repair proteins at DNA damage sites. These highly dynamic processes have been widely studied using laser microirradiation coupled with fluorescence microscopy. Laser microirradiation has provided a powerful methodology to identify and determine mechanisms of DNA damage response pathways. Here we describe methods used to analyze protein recruitment dynamics of fluorescently tagged or endogenous proteins to laser-induced DNA damage sites using laser scanning and fluorescence microscopy. We further describe multiple applications employing these techniques to study additional processes at DNA damage sites including transcription. Together, we aim to provide robust visualization methods employing laser-microirradiation to detect and determine protein behavior, functions and dynamics in response to DNA damage in mammalian cells.


Subject(s)
DNA Repair/radiation effects , Intravital Microscopy/methods , Lasers , Cell Line, Tumor , DNA Damage/radiation effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Epigenome/radiation effects , Fluorescent Dyes/chemistry , Genomic Instability/radiation effects , Humans , Intravital Microscopy/instrumentation , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Spatio-Temporal Analysis , Transcription, Genetic/radiation effects
7.
Cell Cycle ; 17(4): 414-420, 2018.
Article in English | MEDLINE | ID: mdl-29393731

ABSTRACT

Our genetic information is organized into chromatin, which consists of histones and proteins involved in regulating DNA compaction, accessibility and function. Chromatin is decorated by histone modifications, which provide signals that coordinate DNA-based processes including transcription and DNA damage response (DDR) pathways. A major signal involved in these processes is acetylation, which when attached to lysines within proteins, including histones, can be recognized and read by bromodomain-containing proteins. We recently identified the bromodomain protein ZMYND8 (also known as RACK7 and PRKCBP1) as a critical DNA damage response factor involved in regulating transcriptional responses and DNA repair activities at DNA double-strand breaks. Other studies have further defined the molecular details for how ZMYND8 interacts with chromatin and other chromatin modifying proteins to exert its DNA damage response functions. ZMYND8 also plays essential roles in regulating transcription during normal cellular growth, perturbation of which promotes cellular processes involved in cancer initiation and progression. In addition to acetylation, histone methylation and demethylase enzymes have emerged as important regulators of ZMYND8. Here we discuss our current understanding of the molecular mechanisms that govern ZMYND8 function within chromatin, highlighting the importance of this protein for genome maintenance both during the DDR and in cancer.


Subject(s)
DNA Repair , Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Chromatin/metabolism , DNA Breaks, Double-Stranded , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Protein Processing, Post-Translational , Retinoblastoma-Binding Protein 2/genetics , Retinoblastoma-Binding Protein 2/metabolism , Signal Transduction , Transcription, Genetic , Tumor Suppressor Proteins/genetics
8.
Philos Trans R Soc Lond B Biol Sci ; 372(1731)2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28847823

ABSTRACT

Genome surveillance and repair, termed the DNA damage response (DDR), functions within chromatin. Chromatin-based DDR mechanisms sustain genome and epigenome integrity, defects that can disrupt cellular homeostasis and contribute to human diseases. An important chromatin DDR pathway is acetylation signalling which is controlled by histone acetyltransferase (HAT) and histone deacetylase (HDAC) enzymes, which regulate acetylated lysines within proteins. Acetylated proteins, including histones, can modulate chromatin structure and provide molecular signals that are bound by acetyl-lysine binders, including bromodomain (BRD) proteins. Acetylation signalling regulates several DDR pathways, as exemplified by the preponderance of HATs, HDACs and BRD proteins that localize at DNA breaks to modify chromatin for lesion repair. Here, we explore the involvement of acetylation signalling in the DDR, focusing on the involvement of BRD proteins in promoting chromatin remodelling to repair DNA double-strand breaks. BRD proteins have widespread DDR functions including chromatin remodelling, chromatin modification and transcriptional regulation. We discuss mechanistically how BRD proteins read acetylation signals within chromatin to trigger DDR and chromatin activities to facilitate genome-epigenome maintenance. Thus, DDR pathways involving BRD proteins represent key participants in pathways that preserve genome-epigenome integrity to safeguard normal genome and cellular functions.This article is part of the themed issue 'Chromatin modifiers and remodellers in DNA repair and signalling'.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/genetics , DNA Repair , Histone Acetyltransferases/genetics , Histone Deacetylases/genetics , Acetylation , Animals , DNA Damage , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Humans , Mice
9.
J Cell Biol ; 216(7): 1959-1974, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28572115

ABSTRACT

Upon DNA damage, histone modifications are dynamically reshaped to accommodate DNA damage signaling and repair within chromatin. In this study, we report the identification of the histone demethylase KDM5A as a key regulator of the bromodomain protein ZMYND8 and NuRD (nucleosome remodeling and histone deacetylation) complex in the DNA damage response. We observe KDM5A-dependent H3K4me3 demethylation within chromatin near DNA double-strand break (DSB) sites. Mechanistically, demethylation of H3K4me3 is required for ZMYND8-NuRD binding to chromatin and recruitment to DNA damage. Functionally, KDM5A deficiency results in impaired transcriptional silencing and repair of DSBs by homologous recombination. Thus, this study identifies a crucial function for KDM5A in demethylating H3K4 to allow ZMYND8-NuRD to operate within damaged chromatin to repair DSBs.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/enzymology , DNA Breaks, Double-Stranded , Histones/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Receptors, Cell Surface/metabolism , Recombinational DNA Repair , Retinoblastoma-Binding Protein 2/metabolism , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Chromatin/genetics , Dealkylation , Down-Regulation , HEK293 Cells , Humans , Methylation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , RNA Interference , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Retinoblastoma-Binding Protein 2/genetics , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Tumor Suppressor Proteins
10.
PLoS Genet ; 12(9): e1006272, 2016 09.
Article in English | MEDLINE | ID: mdl-27631103

ABSTRACT

Chromatin-based DNA damage response (DDR) pathways are fundamental for preventing genome and epigenome instability, which are prevalent in cancer. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) catalyze the addition and removal of acetyl groups on lysine residues, a post-translational modification important for the DDR. Acetylation can alter chromatin structure as well as function by providing binding signals for reader proteins containing acetyl-lysine recognition domains, including the bromodomain (BRD). Acetylation dynamics occur upon DNA damage in part to regulate chromatin and BRD protein interactions that mediate key DDR activities. In cancer, DDR and acetylation pathways are often mutated or abnormally expressed. DNA damaging agents and drugs targeting epigenetic regulators, including HATs, HDACs, and BRD proteins, are used or are being developed to treat cancer. Here, we discuss how histone acetylation pathways, with a focus on acetylation reader proteins, promote genome stability and the DDR. We analyze how acetylation signaling impacts the DDR in the context of cancer and its treatments. Understanding the relationship between epigenetic regulators, the DDR, and chromatin is integral for obtaining a mechanistic understanding of genome and epigenome maintenance pathways, information that can be leveraged for targeting acetylation signaling, and/or the DDR to treat diseases, including cancer.


Subject(s)
DNA Damage/genetics , Epigenesis, Genetic , Histone Deacetylases/genetics , Neoplasms/genetics , Acetylation , Chromatin/genetics , Genome, Human , Genomic Instability , Histone Acetyltransferases/genetics , Humans
11.
Proc Natl Acad Sci U S A ; 113(9): E1170-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26884156

ABSTRACT

Exonuclease 1 (Exo1) is a 5'→3' exonuclease and 5'-flap endonuclease that plays a critical role in multiple eukaryotic DNA repair pathways. Exo1 processing at DNA nicks and double-strand breaks creates long stretches of single-stranded DNA, which are rapidly bound by replication protein A (RPA) and other single-stranded DNA binding proteins (SSBs). Here, we use single-molecule fluorescence imaging and quantitative cell biology approaches to reveal the interplay between Exo1 and SSBs. Both human and yeast Exo1 are processive nucleases on their own. RPA rapidly strips Exo1 from DNA, and this activity is dependent on at least three RPA-encoded single-stranded DNA binding domains. Furthermore, we show that ablation of RPA in human cells increases Exo1 recruitment to damage sites. In contrast, the sensor of single-stranded DNA complex 1-a recently identified human SSB that promotes DNA resection during homologous recombination-supports processive resection by Exo1. Although RPA rapidly turns over Exo1, multiple cycles of nuclease rebinding at the same DNA site can still support limited DNA processing. These results reveal the role of single-stranded DNA binding proteins in controlling Exo1-catalyzed resection with implications for how Exo1 is regulated during DNA repair in eukaryotic cells.


Subject(s)
DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , Exodeoxyribonucleases/physiology , Biocatalysis , DNA Damage , Humans , Saccharomyces cerevisiae/metabolism
12.
Nature ; 518(7538): 254-7, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25642960

ABSTRACT

The alternative non-homologous end-joining (NHEJ) machinery facilitates several genomic rearrangements, some of which can lead to cellular transformation. This error-prone repair pathway is triggered upon telomere de-protection to promote the formation of deleterious chromosome end-to-end fusions. Using next-generation sequencing technology, here we show that repair by alternative NHEJ yields non-TTAGGG nucleotide insertions at fusion breakpoints of dysfunctional telomeres. Investigating the enzymatic activity responsible for the random insertions enabled us to identify polymerase theta (Polθ; encoded by Polq in mice) as a crucial alternative NHEJ factor in mammalian cells. Polq inhibition suppresses alternative NHEJ at dysfunctional telomeres, and hinders chromosomal translocations at non-telomeric loci. In addition, we found that loss of Polq in mice results in increased rates of homology-directed repair, evident by recombination of dysfunctional telomeres and accumulation of RAD51 at double-stranded breaks. Lastly, we show that depletion of Polθ has a synergistic effect on cell survival in the absence of BRCA genes, suggesting that the inhibition of this mutagenic polymerase represents a valid therapeutic avenue for tumours carrying mutations in homology-directed repair genes.


Subject(s)
Chromosomes, Mammalian/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Directed DNA Polymerase/metabolism , Recombination, Genetic , Telomere/genetics , Telomere/metabolism , Animals , Base Sequence , Cell Death/genetics , Cell Line , Chromosome Aberrations , Chromosomes, Mammalian/genetics , DNA-Directed DNA Polymerase/deficiency , Genes, BRCA1 , Genes, BRCA2 , HeLa Cells , Humans , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , Recombination, Genetic/genetics , Recombinational DNA Repair/genetics , Translocation, Genetic/genetics , DNA Polymerase theta
13.
Genes Dev ; 29(2): 197-211, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25593309

ABSTRACT

How chromatin shapes pathways that promote genome-epigenome integrity in response to DNA damage is an issue of crucial importance. We report that human bromodomain (BRD)-containing proteins, the primary "readers" of acetylated chromatin, are vital for the DNA damage response (DDR). We discovered that more than one-third of all human BRD proteins change localization in response to DNA damage. We identified ZMYND8 (zinc finger and MYND [myeloid, Nervy, and DEAF-1] domain containing 8) as a novel DDR factor that recruits the nucleosome remodeling and histone deacetylation (NuRD) complex to damaged chromatin. Our data define a transcription-associated DDR pathway mediated by ZMYND8 and the NuRD complex that targets DNA damage, including when it occurs within transcriptionally active chromatin, to repress transcription and promote repair by homologous recombination. Thus, our data identify human BRD proteins as key chromatin modulators of the DDR and provide novel insights into how DNA damage within actively transcribed regions requires chromatin-binding proteins to orchestrate the appropriate response in concordance with the damage-associated chromatin context.


Subject(s)
Chromatin/metabolism , DNA Damage , Homologous Recombination/genetics , Receptors, Cell Surface/metabolism , Autoantigens/metabolism , Cell Line, Tumor , Gene Expression Regulation , Gene Silencing , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Protein Binding , Protein Transport/genetics , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Tumor Suppressor Proteins
14.
PLoS Genet ; 10(3): e1004178, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603765

ABSTRACT

Histone ubiquitinations are critical for the activation of the DNA damage response (DDR). In particular, RNF168 and RING1B/BMI1 function in the DDR by ubiquitinating H2A/H2AX on Lys-13/15 and Lys-118/119, respectively. However, it remains to be defined how the ubiquitin pathway engages chromatin to provide regulation of ubiquitin targeting of specific histone residues. Here we identify the nucleosome acid patch as a critical chromatin mediator of H2A/H2AX ubiquitination (ub). The acidic patch is required for RNF168- and RING1B/BMI1-dependent H2A/H2AXub in vivo. The acidic patch functions within the nucleosome as nucleosomes containing a mutated acidic patch exhibit defective H2A/H2AXub by RNF168 and RING1B/BMI1 in vitro. Furthermore, direct perturbation of the nucleosome acidic patch in vivo by the expression of an engineered acidic patch interacting viral peptide, LANA, results in defective H2AXub and RNF168-dependent DNA damage responses including 53BP1 and BRCA1 recruitment to DNA damage. The acidic patch therefore is a critical nucleosome feature that may serve as a scaffold to integrate multiple ubiquitin signals on chromatin to compose selective ubiquitinations on histones for DNA damage signaling.


Subject(s)
Histones/genetics , Polycomb Repressive Complex 1/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA Damage/genetics , DNA Repair , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Nucleosomes/genetics , Protein Binding/genetics , Signal Transduction/genetics , Ubiquitin
15.
Mutat Res ; 750(1-2): 23-30, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23927873

ABSTRACT

Genetic information is recorded in specific DNA sequences that must be protected to preserve normal cellular function. Genome maintenance pathways have evolved to sense and repair DNA damage. Importantly, deleterious mutations that occur from mis-repaired lesions can lead to diseases such as cancer. As eukaryotic DNA is bound by histone proteins and organized into chromatin, the true in vivo substrate of transcription, replication and DNA repair is chromatin. Almost 50 years ago, it was found that histones contained the post-translational modification (PTM), acetylation. With the cloning and identification of transcription associated histone acetyltransferase (HAT) and histone deacetylase (HDAC) enzymes that write and erase the histone acetylation mark respectively, it was realized that this histone modification could be dynamically regulated. Chromatin is subjected to numerous PTMs that regulate chromatin structure and function, including DNA repair. As different organisms contain different histone modifications, chromatin-associated proteins and chromatin states, it is likely that chromatin-templated processes such as DNA repair will exhibit organismal differences. This article focuses on the DNA damage response (DDR) in mammalian cells and how the concerted activities of HAT and HDAC enzymes, and their histone acetylation targets, specifically participate in DNA double-strand break (DSB) repair. Defects in DNA repair and chromatin pathways are observed in cancer, and these pathways represent cancer therapeutic targets. Therefore, understanding the relationship between DNA repair and histone acetylations is important for providing mechanistic details of DSB repair within chromatin that has the potential to be exploited in the clinic.


Subject(s)
DNA Damage , DNA Repair/physiology , Histone Acetyltransferases/physiology , Histone Deacetylases/physiology , Histones/metabolism , Acetylation , Animals , DNA/genetics , Humans
16.
Mol Cell Biol ; 33(1): 111-26, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23109425

ABSTRACT

The histone variant H2AX is a principal component of chromatin involved in the detection, signaling, and repair of DNA double-strand breaks (DSBs). H2AX is thought to operate primarily through its C-terminal S139 phosphorylation, which mediates the recruitment of DNA damage response (DDR) factors to chromatin at DSB sites. Here, we describe a comprehensive screen of 67 residues in H2AX to determine their contributions to H2AX functions. Our analysis revealed that H2AX is both sumoylated and ubiquitylated. Individual residues defective for sumoylation, ubiquitylation, and S139 phosphorylation in untreated and damaged cells were identified. Specifically, we identified an acidic triad region in both H2A and H2AX that is required in cis for their ubiquitylation. We also report the characterization of a human H2AX knockout cell line, which exhibits DDR defects, including p53 activation, following DNA damage. Collectively, this work constitutes the first genetic complementation system for a histone in human cells. Finally, our data reveal new roles for several residues in H2AX and define distinct functions for H2AX in human cells.


Subject(s)
Histones/metabolism , Protein Processing, Post-Translational , Alanine , Amino Acid Motifs , Amino Acid Substitution , Cell Line , Cell Proliferation , Cell Survival , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair/genetics , Gene Knockout Techniques , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Serine/metabolism , Sumoylation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...