Sie sind auf Seite 1von 8

Mutation Research 750 (2013) 23–30

Contents lists available at ScienceDirect


Mutation Research/Fundamental and Molecular
Mechanisms of Mutagenesis
journal homepage: www.elsevier.com/locate/molmut
Community address: www.elsevier.com/locate/mutres

Review

Mammalian DNA repair: HATs and HDACs make their mark through
histone acetylation
Fade Gong, Kyle M. Miller ∗
Section of Molecular Genetics and Microbiology, Institute for Molecular and Cellular Biology, University of Texas at Austin, 2506 Speedway Stop A5000,
Austin, TX 78712, USA

a r t i c l e i n f o a b s t r a c t

Article history: Genetic information is recorded in specific DNA sequences that must be protected to preserve normal
Received 30 April 2013 cellular function. Genome maintenance pathways have evolved to sense and repair DNA damage. Impor-
Received in revised form 3 July 2013 tantly, deleterious mutations that occur from mis-repaired lesions can lead to diseases such as cancer. As
Accepted 9 July 2013
eukaryotic DNA is bound by histone proteins and organized into chromatin, the true in vivo substrate of
Available online 6 August 2013
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
Keywords:
transcription associated histone acetyltransferase (HAT) and histone deacetylase (HDAC) enzymes that
Chromatin
DNA repair write and erase the histone acetylation mark respectively, it was realized that this histone modification
DNA damage response could be dynamically regulated. Chromatin is subjected to numerous PTMs that regulate chromatin struc-
Genome stability ture and function, including DNA repair. As different organisms contain different histone modifications,
Histone acetylation 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 acety-
lation 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.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
1.1. DNA damage response (DDR) in mammalian cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
1.2. Chromatin regulation by histone modifications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
1.3. Regulation of histone acetylation by HATs and HDACs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2. Histone acetylation and DNA damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
3. Histone acetylation and DNA double-strand break repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
3.1. Histone variant H2AX and histone H2A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
3.2. Histone H4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
3.3. Histone H3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
4. Additional histone acetylations and DDR pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
5. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Conflict of interest statement. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29

∗ Corresponding author. Tel.: +1 5124715045.


E-mail address: kyle.miller@austin.utexas.edu (K.M. Miller).

0027-5107/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.mrfmmm.2013.07.002
24 F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30

1. Introduction transcription correlates with an “open” chromatin configuration


(i.e. euchromatin) while transcriptionally repressed regions results
1.1. DNA damage response (DDR) in mammalian cells in a more “closed” chromatin state (i.e. heterochromatin). Chro-
matin remodeling activities including nucleosome repositioning,
The nuclear milieu is a harsh environment for our genetic infor- histone eviction/deposition, and histone variant incorporation
mation since high incidences of DNA damage occur every day can regulate chromatin. Another mechanism that regulates chro-
within our cells [1,2]. Whether it be replication-dependent DNA matin states is the post-translational modifications (PTMs) that
mismatches, transcriptional damage, the production of metabolic decorate histones. Histones are modified on distinct amino acid
by-products, exposure to exogenous DNA damaging agents or residues with a vast array of different PTMs including phosphory-
therapeutic radiotherapy, these processes can all create damaged lation, methylation, ubiquitylation, sumoylation and acetylation,
DNA that must be faithfully repaired to maintain genome fidelity. including several that are involved in DSB repair [14–17].
Eukaryotic cells respond to these genetic assaults by activating a Histone modifications can affect chromatin structure in multi-
set of pathways termed the DNA damage response (DDR) [1,2]. ple ways. The basic charge of histones is due to the high proportion
The DDR represents a complex network of cellular pathways that of positively charged lysine and arginine residues that are found
function to detect the damaged lesion in DNA, signal the presence in all histone proteins. Negatively charged DNA interacts electro-
of the lesion to the cell to activate the appropriate response, and statically to regions of the histone that contain these amino acids.
ultimately to repair the lesion to maintain the correct sequence Modifications of either lysines or arginines have the potential to
of the DNA [1,2]. One particularly dangerous type of lesion is the disrupt DNA/protein and protein/protein interactions by changing
DNA double-strand break (DSB). A DSB makes a discontinuous DNA the charge of the modified amino acid [14,18]. Histone marks can
molecule creating a precarious situation as unprotected DNA ends also act as high affinity binding sites for proteins that contain bind-
can be acted upon by nucleases leading to degradation and/or inap- ing domains that recognize histone PTMs. Histone binding domains
propriately religated to other free ends resulting in a loss of genetic exists for many PTMs including phosphorylation, methylation and
information, chromosomal translocations or complete chromo- acetylation. For example, twin-BRCT domains can bind phospho-
some loss. Due to these particularly deleterious outcomes, DSBs rylated histones, chromodomains can exhibit binding preferences
constitute the most cytotoxic class of all DNA lesions. In mammalian for methylated lysines and bromodomains can specifically interact
cells, DSBs are repaired mainly by either homologous recombina- with acetylated lysine residues [14,18,19]. As an example, meth-
tion (HR) or non-homologous end-joining (NHEJ) [3]. HR requires ylation of lysine 9 of histone H3 provides an interaction motif for
the processing of DNA ends and a template for the accurate repair the chromodomains of HP1 proteins, which facilitate the formation
of a DSB while NHEJ ligates the free DNA ends with little to no of heterochromatin and gene repression [20,21]. Trimethylation
processing of the DNA ends prior to ligation. It is important to note of lysine 4 of histone H3 creates a binding site for transcriptional
that in mammalian cells, NHEJ is the main DSB repair pathway in complexes, leading to gene activation and euchromatin formation.
G1, and is also active in the G2 phase of the cell cycle [4,5]. For this Thus, different chromatin states and functions found throughout
reason, the choice between HR and NHEJ has been an important and the genome can be regulated and established by the modification
active area of research in the field [6]. As DSB repair occurs within of context specific amino acid residues on histones.
the context of chromatin and many chromatin modifying enzymes
have been linked with DSB repair, the understanding of the rela- 1.3. Regulation of histone acetylation by HATs and HDACs
tionship between DNA repair and chromatin is fundamental for
revealing the mechanistic details of how DNA repair pathways are Acetylation of histones was one of the first PTMs identified [22]
regulated to promote stability of the entire eukaryotic genome [7]. and represents the addition of an acetyl group ( COCH3 ) onto the
Yeast has provided important insights into the role of histone acety- ␧-amino group of lysines (Fig. 1). This is not the only modification
lation in genome stability and DNA repair [8–11]. Here, we focus that occurs on lysines as other PTMs including methylation,
on our current understanding of the function and regulation of his- ubiquitylation, sumoylation, biotinylation and succinylation have
tone acetylation in the DNA damage response (DDR) in mammalian been identified, making lysines one of the most diversely modified
cells. Specifically, we aim to review how HAT and HDAC enzymes, amino acids [14,23]. Histone acetylation is regulated by histone
and their histone acetylation targets, participate in DNA double- acetyltransferases (HATs) and histone deacetylases (HDACs) that
strand break (DSB) repair. Researchers are actively pursuing both catalyze the addition and removal, respectively, of the acetyl
chromatin factors and DDR proteins as potential therapeutic tar- moiety from lysines (Fig. 1). From the initial discovery of the first
gets for diseases, including cancer, so understanding the interplay HAT and HDAC involved in gene activation, it became apparent
between chromatin and the DDR could reveal novel therapeutic that histone acetylation was a regulatable phenomenon that par-
opportunities. ticipated in site-specific processes such as transcription [24,25].
Mammalian cells contain at least 17 HATs and 18 HDACs (Table 1).
1.2. Chromatin regulation by histone modifications HATs are diverse and fall into several families including GNAT (ex.
GCN5), MYST (ex. TIP60) and the “orphan” family (ex. p300) [26].
Duplexed DNA in the nucleus of eukaryotic cells is largely com- Both GCN5 and p300 contain bromodomains, and TIP60 exists in
pacted into chromatin. The fundamental unit of chromatin is the the p400-Tip60/NuA4 complex that comprises a bromodomain
nucleosome [12], which consists of positively charged histone pro- containing protein, which reinforces the notion of how histone
teins and negatively charged DNA [13]. Specifically, the nucleosome acetylation regulates the specificity, targeting and activity of these
consists of two copies each of the four core histones, H2A, H2B, enzymes [27,28]. Mammalian HDACs fall into 4 classes (Table 1)
H3 and H4, as well as ∼146 bp of DNA wrapped nearly two times [29]. Class I HDACs (HDAC 1-3, 8) are nuclear and ubiquitously
around the histone octamer. Eukaryotic cells also contain linker his- expressed in all cells. Class II HDACs (HDAC 4-7, 9 and 10) are
tones, histone 1 (H1), that bind linker DNA between nucleosomes cell-type specific and are expressed mostly in the cytoplasm but
as well as binding surfaces within the nucleosome themselves can become nuclear upon various stimuli. Class III HDACs are
to promote chromatin compaction. Eukaryotic genomes contain the NAD-dependent Sirtuins (SIRT1-7) where SIRT1, 6 and 7 are
multiple chromatin states that change depending on cell type and nuclear, SIRT2 is mainly cytoplasmic and SIRT 3-5 are mitochon-
cell-cycle phase as well as the processes that act upon a particular drial [30]. Class IV contains only one member, HDAC11, which is
region of the genome at any given time. For example, active mainly expressed in the nucleus in a cell-type specific manner.
F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30 25

Ac-Lys Readers
H O
Bromodomain
+
NH 3 εN C CH 3
Double Bromodomain
CH 2
CH CH 2
CH
Double PHD finger
CH 2
CH CH 2
CH
Double PH domain
CH 2
CH CH 2
CH
CH 2
CH CH 2
CH
CH CH

Lys
HAT Ac-Lys

HDAC
Fig. 1. Lysine acetylation. The addition of an acetyl (Ac) moiety on the ␧-amino group of a lysine residue is catalyzed by histone acetyltransferases (HATs) and this reaction
is reversed by histone deacetylases (HDACs). Acetyl-lysine readers are comprised mainly of proteins containing a bromodomain but other acetyl-lysine binding domains
(indicated) have been reported [98].

Collectively, many of these enzymes have been shown to function in 3.1. Histone variant H2AX and histone H2A
the DNA damage response by targeting specific histone acetylation
sites (Table 1 and Fig. 2A). These enzymes also target non-histone Upon formation of a DSB in mammalian cells, one of the very
substrates, for example ATM [31] and p53 [32], that are critical for first chromatin events that occurs is the phosphorylation of the
the DDR, although this review will focus mainly on their histone histone variant H2AX on S139 to form ␥H2AX [39]. ␥H2AX is crit-
targets. ical for many aspects of the DDR, including the maintenance of
genome integrity. This histone modification marks DSBs and is used
as a biomarker of DNA damage in cancer cells, making ␥H2AX the
2. Histone acetylation and DNA damage most well-studied histone modification involved in the DDR [40].
However, acetylation of several lysines in H2AX have also been
Histone acetylation was first linked with a DNA damage described to impact DSB repair [17]. Upon DNA damage, the HAT
response in human cells when it was observed that histones TIP60 acetylates H2AX on lysine 5 [28]. SIRT1 regulates TIP60 activ-
are rapidly acetylated after exposure to ultraviolet radiation (UV) ity and depletion of SIRT1 results in H2AX K5Ac hyperacetylation
[33]. Interestingly, hyperacetylation of histones is rapidly followed resulting in defective DDR activation at DSBs [41]. H2AX K5Ac also
by hypoacetylation and this response is sensitive to UV dose regulates H2AX ubiquitylation after DSB formation, which affects
as low doses promoted more hyperacetylation and high doses the dynamics of H2AX at break sites. H2AX lysine 36 acetylation
resulted in hypoacetylation. Repair of UV lesions occurs more has also been identified [42,43]. This site is hyperacetylated by
rapidly in hyperacetylated histones, which led to the idea that the overexpression of the HATs, p300 or CBP [42]. H2AX−/− mouse
chromatin accessibility occurring after histone acetylation would cells reconstituted with an unmodifiable H2AX at lysine 36 results
allow repair proteins to gain access to the damaged lesion [34]. in defective IR-resistance while an unmodifiable H2AX at lysine
These data led to the “access-repair-restore” model whereby chro- 5 does not [42,43]. For H2AX acetylation, several important ques-
matin would be altered to allow access and repair of lesions tions remain. The core histone H2A also contains K5 and K36 so it is
followed by a restoration of chromatin organization [35]. Sub- unclear whether the acetylation of these residues is H2AX-specific
sequent work has given more molecular details of the enzymes in regards to DSB repair. In addition, no HDAC has been reported
and histone acetylation sites involved in this process. GCN5- for H2AX K36Ac and whether the function of H2AX acetylations
dependent H3K9 acetylation, and H4K16Ac, are rapidly induced involves changes in chromatin structure and/or the involvement
following UV damage [36]. Global hypoacetylation of H3K9 and of a histone acetylation binding protein is yet to be determined.
H3K56 is observed after high doses of UV as well [37,38]. These H2A has several additional acetylation sites and both H2AZ and
histone acetylation changes potentially regulate chromatin orga- H2B have many acetylated lysines but the potential role of these
nization following UV damage. Defining how global changes in acetylation sites in DSB repair in mammalian cells awaits further
histone acetylation upon UV damage regulates chromatin struc- characterization [44].
ture, nuclear processes, such as transcription, as well as the repair
of the UV lesion will require further analysis in mammalian cells.
3.2. Histone H4
Regardless, the “access-repair-restore” model has been applied to
DSB repair in mammalian cells where it is believed that a con-
Histone H3 and H4 bind as a tetramer in the nucleosome and
certed effort from chromatin modifying enzymes, such as HATs
several acetylation sites on H4 are involved in the cellular response
and HDACs, as well as chromatin remodeling complexes, histone
to DNA damage. Early experiments identified the human HAT TIP60
variants and histone chaperones collectively regulate chromatin
as an important factor involved in the DDR [45]. TIP60 acetylates
organization to promote DSB repair by HR and NHEJ pathways
both H3 and H4, as well as H2A, in vitro and mutating the acetyl-
(Fig. 2A) [7].
transferase activity of TIP60 rendered cells defective in repairing
DSBs following IR suggesting that acetylation was critical for DSB
repair [45]. The first histone acetylation sites identified as targets
3. Histone acetylation and DNA double-strand break repair of the DDR in mammalian cells were on H4. Human MOF acetylates
H4K16Ac after IR exposure. Interestingly, this HAT also interacts
Core histones and histone variants are modified by acetylation with ATM and reduction of MOF results in defective ATM activation
and several sites have been shown to be involved in DSB repair and DSB repair [46]. TIP60 functions in the p400-Tip60/NuA4 com-
(Table 1). plex to acetylate histones to promote remodeling of chromatin at
26 F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30

DSB
A
HATs/
HDACs HATs
HDACs

DSB repair

Repaired
DNA break
HATs/HDACs, Chromatin Remodeling complexes
Histone chaperones, Proteases

Chromatin
Restoration

acetylated &
Histones: deacetylated acetylated
deacetylated

53BP1
B 53BP1 BRCA1
Me2
HAT Ac Me2
K16 BRCA1
K20 (TIP60) K16 K20
H4 H4

HDAC
(HDAC1/2)

NHEJ HR

Fig. 2. Histone acetylation and DSB repair. (A) DSBs occur within a pre-existing chromatin acetylation landscape. HATs and HDACs, individually or in concert, are recruited to
the DSB site where deacetyation, acetylation or both activities occur on specific histone residues to create a repair proficient chromatin state that will promote either NHEJ or
HR. Restoration of chromatin to its pre-damaged state can occur through modification of existing chromatin or de novo chromatin assembly by the action of several distinct
chromatin modifying enzymes, histone chaperones and/or chromatin remodeling complexes. These activities, with or without HDACs and HATs, can also act upstream of
chromatin restoration at the level of detection and repair of DSBs. (B) Regulation of DSB repair pathway choice by histone acetylation.
Adapted from model presented in Tang et al. [56]. See text for details.

DSBs [28,47–50]. TIP60 activity is stimulated by H3K9me3 resulting shown to increase near a DSB [52]. However, these DSB-induced
in acetylation of ATM to promote its activity [49]. The interaction acetylations were dependent on either p300 or CBP as depletion
between TIP60 and ATM is regulated by TIP60 phosphorylation by of either of these HATs resulted in a defect in H4 Ac and H3K18Ac
c-Abl, which is critical for the ability of ATM to sense chromatin inductions at a DSB. p300 and CBP, as well as TIP60, were shown to
alterations, perhaps caused by aberrant histone acetylation, even decrease efficient NHEJ using this reporter system. ChIP analysis of
without the presence of damaged DNA [51]. Using an I-SceI (site- these histone marks adjacent to the DSB was performed in a region
specific homing endonuclease) single DSB system in mouse ES cells, that was transcriptionally silent in undamaged conditions but
DSB induction induces the hyperacetylation of H4 0.5KB from the became transcriptionally active after repair [52]. However, histone
DSB as determined by an antibody raised against H4 acetylated acetylations are also associated with active transcription, which
at K5, K8, K12, K16 [48]. These acetylations were dependent on could also affect the levels of these marks in these types of assays. In
the Trrap-TIP60 complex. Trrap-TIP60 is required for efficient HR addition, these systems rely on persistent DSBs. How these results
and this defect could be suppressed by the HDAC inhibitor sodium from persistent single-locus DSB systems relate to endogenous and
butyrate, although the HDAC(s) were not identified. In another exogenous DSB formation that are occurring throughout the chro-
study, using a DSB system with two I-SceI sites that could monitor matin landscape from normal cellular processes or upon treatment
NHEJ specifically, H4 tetra acetylation on K5, K8, K12, and K16 was with DNA damaging agents, requires additional studies.
F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30 27

Table 1
Histone acetylation sites and enzymes involved in the DDR in mammalian cells.

Name Localization DDR histone sites DDR pathways Recruitment

Mammalian histone acetyltransferases (HATs)


KAT1 HAT1 N, C n.d. DDR [80] n.d.
KAT2A GCN5 N H3K9/K14/K18/K23/56 DDR [37,62]; NER [36] UV [36]
[36,37,62]; H4K16 [36]
KAT2B PCAF N H3K9, H3K14 [81] DDR [81] n.d.
KAT3A CBP N H3K56 [60]; H3K18, NHEJ [52]; NER [82] Laser damage [52]
H4K5/K8/K12/K16
[52]; H2AXK36 [42]
KAT3B p300 N H3K56 [37,60,83]; NHEJ [52]; DDR [84,85]; NER [82] Laser damage [52]; UV [86]
H3K18,
H4K5/K8/K12/K16
[52]; H2AXK36 [42]
KAT4 TAF1 N n.d. DDR [87]
KAT5 TIP60 N H2AXK5 [28]; H4K16 HR [48,56,57]; DDR [45,88] IR [88]; R.E. [48,56]
[56,57]
KAT8 MOF N H4K16[46,53,89] DDR[46,53]; NHEJ & HR [89] n.d.
KAT13D CLOCK N, C n.d. DDR [90] Laser damage [90]
Other HATs: MOZ (KAT6A), MORF (KAT6B), HBO1 (KAT7), ELP3 (KAT9), TFIIIC90 (KAT12), SRC1 (KAT13A), ACTR (KAT13B), p160 (KAT13C)

Mammalian histone deacetylases (HDACs)


Class I HDAC1 N H3K56, H4K16 [54] NHEJ [54] Laser damage [54]
HDAC2 N H3K56, H4K16 [54] NHEJ [54] Laser damage [54]
HDAC3 N H3K9/K14, H4K5/K12 DDR [65] n.d.
[65]

Class II HDAC4 N n.d. DDR [91,92] IR [91]


HDAC9 N n.d. HR [93] n.d.
HDAC10 N,C n.d. HR [93] n.d.

Class III SIRT1 N H1K26 [66], H2AXK5 DDR [41,60,94,95]; HR [66] R.E. [66,94]
[41], H3K56 [60],
H4K16 [94]
SIRT2 C H3K56 [60] DDR [60] n.d.
SIRT6 N H3K9 [61] NHEJ [61]; HR [96]; BER [97] Laser damage [96]; R.E. [61]
Other HDACs: HDAC8 (Class I); HDAC5, HDAC6, HDAC7 (Class II); SIRT3, SIRT4, SIRT5, SIRT7 (Class III); HADC11 (Class IV)

Histone acetylation sites and enzymes involved in the DDR in mammalian cells. All mammalian HATs and HDACs are indicated. DNA damage affects on specific lysine sites, DDR
pathways and localization are provided for each enzyme. HATs and HDACs where a role in the DDR has not been reported are listed in the last line. Abbreviations: N-nuclear,
C-cytoplasmic, M-mitochondrial, n.d.-not determined, DDR-DNA damage response, NER-Nucleotide excision repair, NHEJ-Non homologous end-joining, HR-Homologous
recombination, BER-Base excision repair, UV-ultraviolet radiation, IR-Ionizing radiation, R.E.-restriction enzyme.

Of the potential H4 acetylation sites, H4K16Ac levels increase creates a steric hindrance to regulate protein binding at DSBs but
after DNA damage and this site appears to play a critical role in could also regulate repair through both chromatin structure and
DSB repair [46,53,54]. After laser-induced DNA damage, H4K16Ac direct histone binding of an acetyl-lysine reader. Multiple HATs,
levels decrease rapidly followed by a phase of acetylation result- including MOF and TIP60, have been reported to induce H4 acetyla-
ing in increased levels at DNA damage sites [54]. The behavior of tions upon DNA damage (Table 1). Defective chromatin remodeling
H4K16Ac upon DNA damage mirrors those of DSB repair in human mediated by H4 acetylations in TIP60 or MOF deficient cells could
cells. Indeed, repair of DSB by NHEJ occurs rapidly while HR repair help explain their observed defects in DSB repair. How coordinated
is much slower. Interestingly, HDAC1 and HDAC2 localize to laser- HAT activities regulate these H4 sites to promote DSB repair by both
induced damage rapidly, at a time where H4K16 deacetylation is NHEJ and HR is unclear. H4 HATs could potentially target a subset
observed [54]. Depletion of both HDAC1 and HDAC2 results in of histones at both damaged and undamaged sites to promote DDR
hyper-acetylation of H4K16Ac and defects in NHEJ in cancer cell processes, including DNA repair and/or transcriptional regulation.
lines and genome instability in mice [54,55]. A molecular explana- Increases in histone acetylations can also result from the inhibition
tion for some of these results has recently been provided. Using a of HDACs, which could also be controlled by HAT activities on both
single locus DSB reporter system in human cells, TIP60 promoted histone and non-histone targets.
the accumulation of BRCA1 while inhibiting 53BP1 recruitment to
DSBs [56]. HDAC inhibitors, as well as depletion of both HDAC1 and 3.3. Histone H3
HDAC2, resulted in 53BP1 inhibition and accumulation of BRCA1 at
DSBs. Thus, the balance between BRCA1, which is involved in HR, Several acetylations on histone H3 are regulated by DNA
and 53BP1, which is involved in NHEJ, at DSBs is regulated by acety- damage. Upon DSB induction, global levels of H3K9Ac, H3K56Ac
lation and deacetylation [56,57]. Using peptides containing specific and H3K14Ac levels are regulated by the DDR [37,54,59–61].
histone marks as well as NMR structural studies, acetylation of The coordinated potential functions, for example transcription,
H4K16 was found to be inhibitory toward the binding of the tudor replication, chromatin structure and/or chromatin organization,
domains of 53BP1 to H4K20me2 [56]. Thus, regulation of H4K16Ac of these global changes in H3 acetylation are not yet fully under-
can contribute to DSB repair pathway choice at DSBs by modulat- stood. For H3K14Ac, the nucleosomal binding protein HMGN1
ing the association of both 53BP1 and BRCA1 to promote NHEJ and was found to be required for H3K14Ac upon IR treatment [59].
HR respectively (Fig. 2B). Of note, H4K16Ac also inhibits both intra- Loss of HMGN1 resulted in defective ATM signaling suggesting
and inter-nucleosomal folding to impede higher order chromatin an additional link between histone acetylation and chromatin for
structure formation [58] and H4K16Ac could also affect interactions ATM activation. Several studies analyzed H3 acetylations specif-
of acetyl binding proteins with chromatin at DSBs. Thus, H4K16Ac ically at DSBs and found similar, as well as additional, alterations
28 F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30

in histone acetylation levels. Purification of damaged ␥H2AX as well as specific histone acetylations, in nearly all aspects of the
containing chromatin revealed higher levels of H3K9, K14, K18 and DDR, as well as other cellular stresses such as aging, are sure to be
K23 acetylation levels compared to chromatin that contained an discovered.
unphosphorylatable form of H2AX (S139A) [62]. These N-terminal
acetylations on H3 are dependent on GCN5 and were shown to
recruit the SWI/SNF chromatin remodeling complex to ␥H2AX 5. Summary
nucleosomes. Interestingly, the SWI/SNF complex includes BRG1,
a bromodomain-containing protein, which is recruited specifi- Histone acetylations are determinant marks for both chromatin
cally to damaged chromatin through a bromodomain-dependent structure and chromatin binding proteins. Regulation of these chro-
interaction with H3K14Ac. Mutation of the bromodomain in BRG1 matin states by the concerted action of both HATs and HDACs
also resulted in sensitivity to IR suggesting that repair of DSBs are critical for orchestrating the DDR in mammalian cells and
requires the binding of SWI/SNF to acetylated histones [62]. This for promoting DSB repair. Several HATs and HDACs are recruited
study shows that histone acetylations can modulate the DDR by to sites of DNA damage and several histone acetylation sites are
directly affecting the binding of factors through lysine acetylation affected by DNA damage (Table 1). Thus, it is clear that HATs and
interactions at DSBs. Using site-specific restriction enzyme based HDACs modulate chromatin surrounding a break to promote its
systems, H3K18Ac levels are increased while H3K56Ac levels are repair (Fig. 2A). Upon DSB formation, we envision that the pre-
reduced at DSBs [52,54]. HDAC1 and HDAC2 are recruited to sites damaged chromatin acetylation state will be regulated by specific
of damage where they regulate H3K56Ac levels and deacetylate HDACs or HATs, as well as potentially a combination of both, to
H3K56Ac to promote NHEJ [54]. p300/CBP acetylates H3K18, create repair-proficient chromatin. This is reminiscent of transcrip-
which facilitates SWI/SNF chromatin remodeling activity at DSBs. tion where rapid acetylation followed by deacetylation by HATs
The activity of SWI/SNF is required for the efficient recruitment of and HDACs respectively occurs to regulate chromatin dynamics
NHEJ factors, including KU, and inhibition of this pathway results required for gene activation [69,70]. At a DSB, once repair is com-
in defective NHEJ [52]. Collectively, certain histone acetylations on plete, restoration of the damage-modified chromatin can occur
H3 are regulated upon DNA damage and function to promote DSB through the concerted activities of HDACs and/or HATs. De novo
repair. H3 acetylations appear to not only regulate the recruitment chromatin assembly can also restore chromatin using the activities
of DDR factors to sites of damage but also to promote nucleosome of chromatin remodeling complexes and histone chaperones. His-
and chromatin alterations at DSBs, as well as throughout the tone degradation following DNA damage has recently been shown
genome. These activities clearly contribute to the DDR and to DSB to occur in mammalian cells through a non-ubiquitin mechanism
repair although the mechanistic details are still emerging. that requires the proteosome activator PA200 [71]. Interestingly,
PA200 contains a bromodomain-like region that binds acetylated
histones to catalyze their degradation. These activities can also
4. Additional histone acetylations and DDR pathways act upstream of chromatin restoration for DSB repair, as has been
demonstrated for chromatin remodeling complexes. Thus, several
Although this review has focused on histone acetylations that potential pathways exist, either through modifying the existing
occur upon DSB formation, several other DNA damage pathways chromatin or through de novo chromatin assembly at the site of a
also result in changes to acetylations on histones. DNA damage DSB that have the potential to restore chromatin to its pre-damaged
occurring during replication is a major cause of endogenous DNA state.
damage and threatens both genetic and epigenetic integrity. His- A comprehensive view of how and why specific histone acety-
tone acetylations are involved in both genome maintenance during lations are regulated by DNA damage globally, as well as at DSBs,
replication as well as promoting the stability of the epigenome is lacking in mammalian cells. What the DNA damage-dependent
[63]. Analysis of histone acetylations and the enzymes that regulate functions of each individual histone acetylation are at each genome
them at active, stalled or collapsed replication forks has now been location, for a given cellular response and repair pathway, remains
made possible by a powerful technique called iPOND [64]. Using unclear. The variability of chromatin states within cell-cycle phases,
this method, HDAC1, HDAC2 and HDAC3 were enriched at active cell types and physiological conditions compounds the ability to
replication forks, and deacetylation of H4K5Ac and H4K12Ac, marks provide an all-inclusive model of DSB repair and histone acetyla-
required for chromatin maturation during S-phase, was observed. tions. Rather, like transcription, histone acetylations are likely to
Interestingly, deletion of HDAC3 in mouse cells results in the hyper- regulate DSB repair in a site-specific context that depends on multi-
acetylation of both of these histone marks, as well as H3K9Ac ple factors including, but not limited to, existing chromatin modifi-
and H3K14Ac, in S-phase [65]. HDAC3 null cells exhibit defects in cations, chromatin structure, cell cycle phase, genome location and
heterochromatin formation as well as increased genome instability DSB repair pathway choice. Utilization of genome-wide DSB sys-
and defects in DNA repair [65]. Thus, these examples highlight the tems that are capable of analyzing multiple site-specific DSBs in a
importance of regulated histone acetylations by HATs and HDACs variety of chromatin contexts can overcome the normal limitations
during DNA replication and further work is required to detail the of many DNA damaging agents that act in a non site-specific man-
precise regulation of these pathways in preventing epigenetic and ner. This type of system allows for the use of ChIP and genome-wide
genome instability during replication. Histone acetylations are also sequencing to map histone modifications, including acetylations,
affected by oxidative stress. Indeed, SIRT1 binding to chromatin at multiple defined DSBs [17,72]. Genetic systems using histone
is reduced upon oxidative stress resulting in increased histone variant knockouts, including H2AX, in mammalian cells can also
H1K26Ac [66]. SIRT1 is recruited to sites of DNA damage and is provide insights into the function of site-specific acetylations using
required for DSB repair and genome stability [66]. As oxidative complementation analysis of acetylation site mutants [43,73,74].
stress, DNA damage and SIRT1 are implicated in aging in mam- Development of these and additional approaches to analyze DNA
mals, histone acetylation is likely to also be involved in aging, as damage and histone acetylations across the genome, as well as
has been shown in yeast for H4K16Ac [67]. Replicative aging of pri- defining the acetyl-lysine interactome, will no doubt advance
mary human cells also results in changes in histone acetylations at our understanding of how DNA repair occurs within the context
both telomeres and throughout the genome [68]. Collectively, these of chromatin. HDAC, HAT and bromodomain containing proteins
studies highlight a variety of biological processes that rely on his- are potential therapeutic anti-cancer targets due to their roles in
tone acetylation regulation. Additional roles for HATs and HDACs, transcription, the radiosensitization effects observed upon their
F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30 29

inhibition and importantly, their druggability due to the develop- [22] V.G. Allfrey, R. Faulkner, A.E. Mirsky, Acetylation and methylation of histones
ment and availability of small molecule inhibitors targeting these and their possible role in the regulation of RNA synthesis, Proc. Natl. Acad. Sci.
U. S. A. 51 (1964) 786–794.
proteins [75–77]. The success of inhibitors of the BET family of [23] Z. Zhang, M. Tan, Z. Xie, L. Dai, Y. Chen, Y. Zhao, Identification of lysine suc-
bromodomain-containing proteins, that include BRD4, in suppress- cinylation as a new post-translational modification, Nat. Chem. Biol. 7 (2011)
ing growth in various cancer cell lines and murine cancer models 58–63.
[24] J.E. Brownell, J. Zhou, T. Ranalli, R. Kobayashi, D.G. Edmondson, S.Y. Roth, C.D.
has raised considerable interest in targeting epigenetic readers in Allis, Tetrahymena histone acetyltransferase A: a homolog to yeast Gcn5p link-
cancer [78]. Interestingly, an isoform of BRD4 has been described ing histone acetylation to gene activation, Cell 84 (1996) 843–851.
that acts as an inhibitor of the chromatin response to DNA damage [25] J. Taunton, C.A. Hassig, S.L. Schreiber, A mammalian histone deacetylase related
to the yeast transcriptional regulator Rpd3p, Science 272 (1996) 408–411.
through an association with the condensin II complex [79]. These
[26] K.K. Lee, J.L. Workman, Histone acetyltransferase complexes: one size doesn’t
functions of BRD4 required the lysine recognition bromodomain, fit all, Nat. Rev. Mol. Cell Biol. 8 (2007) 284–295.
which is yet another example that highlights the critical relation- [27] Y. Doyon, W. Selleck, W.S. Lane, S. Tan, J. Cote, Structural and functional conser-
vation of the NuA4 histone acetyltransferase complex from yeast to humans,
ship between chromatin, histone acetylation and the DDR. Thus,
Mol. Cell. Biol. 24 (2004) 1884–1896.
understanding the DDR and DNA repair within the context of chro- [28] T. Ikura, S. Tashiro, A. Kakino, H. Shima, N. Jacob, R. Amunugama, K. Yoder, S.
matin will be critical in determining the utility of targeting the Izumi, I. Kuraoka, K. Tanaka, H. Kimura, M. Ikura, S. Nishikubo, T. Ito, A. Muto, K.
DDR and histone acetylation pathways, including bromodomain Miyagawa, S. Takeda, R. Fishel, K. Igarashi, K. Kamiya, DNA damage-dependent
acetylation and ubiquitination of H2AX enhances chromatin dynamics, Mol.
epigenetic readers, in diseases such as cancer. Cell. Biol. 27 (2007) 7028–7040.
[29] X.J. Yang, E. Seto, The Rpd3/Hda1 family of lysine deacetylases: from bacteria
and yeast to mice and men, Nat. Rev. Mol. Cell Biol. 9 (2008) 206–218.
Conflict of interest statement [30] J.L. Feldman, K.E. Dittenhafer-Reed, J.M. Denu, Sirtuin catalysis and regulation,
J. Biol. Chem. 287 (2012) 42419–42427.
[31] Y. Sun, X. Jiang, B.D. Price, Tip60: connecting chromatin to DNA damage
The authors declare that there is no conflict of interest. signaling, Cell Cycle 9 (2010) 930–936.
[32] K.H. Vousden, C. Prives, Blinded by the light: the growing complexity of p53,
Cell 137 (2009) 413–431.
Acknowledgements [33] B. Ramanathan, M.J. Smerdon, Changes in nuclear protein acetylation in u.v.-
damaged human cells, Carcinogenesis 7 (1986) 1087–1094.
[34] B. Ramanathan, M.J. Smerdon, Enhanced DNA repair synthesis in hyperacety-
The Miller laboratory is in part supported by the Cancer Preven- lated nucleosomes, J. Biol. Chem. 264 (1989) 11026–11034.
tion Research Institute of Texas (CPRIT). Dr. Kyle Miller is a CPRIT [35] M.J. Smerdon, DNA repair and the role of chromatin structure, Curr. Opin. Cell
Biol. 3 (1991) 422–428.
scholar.
[36] R. Guo, J. Chen, D.L. Mitchell, D.G. Johnson, GCN5 and E2F1 stimulate nucleotide
excision repair by promoting H3K9 acetylation at sites of damage, Nucleic Acids
Res. 39 (2011) 1390–1397.
References [37] J.V. Tjeertes, K.M. Miller, S.P. Jackson, Screen for DNA-damage-responsive his-
tone modifications identifies H3K9Ac and H3K56Ac in human cells, EMBO J. 28
[1] S.P. Jackson, J. Bartek, The DNA-damage response in human biology and disease, (2009) 1878–1889.
Nature 461 (2009) 1071–1078. [38] A. Battu, A. Ray, A.A. Wani, ASF1A and ATM regulate H3K56-mediated cell-cycle
[2] A. Ciccia, S.J. Elledge, The DNA damage response: making it safe to play with checkpoint recovery in response to UV irradiation, Nucleic Acids Res. 39 (2011)
knives, Mol. Cell 40 (2010) 179–204. 7931–7945.
[3] P. Huertas, DNA resection in eukaryotes: deciding how to fix the break, Nat. [39] E.P. Rogakou, D.R. Pilch, A.H. Orr, V.S. Ivanova, W.M. Bonner, DNA double-
Struct. Mol. Biol. 17 (2010) 11–16. stranded breaks induce histone H2AX phosphorylation on serine 139, J. Biol.
[4] A. Beucher, J. Birraux, L. Tchouandong, O. Barton, A. Shibata, S. Conrad, A.A. Chem. 273 (1998) 5858–5868.
Goodarzi, A. Krempler, P.A. Jeggo, M. Lobrich, ATM and Artemis promote homol- [40] W.M. Bonner, C.E. Redon, J.S. Dickey, A.J. Nakamura, O.A. Sedelnikova, S. Solier,
ogous recombination of radiation-induced DNA double-strand breaks in G2, Y. Pommier, GammaH2AX and cancer, Nat. Rev. Cancer 8 (2008) 957–967.
EMBO J. 28 (2009) 3413–3427. [41] K. Yamagata, I. Kitabayashi, Sirt1 physically interacts with Tip60 and nega-
[5] M.R. Lieber, The mechanism of double-strand DNA break repair by the nonho- tively regulates Tip60-mediated acetylation of H2AX, Biochem. Biophys. Res.
mologous DNA end-joining pathway, Annu. Rev. Biochem. 79 (2010) 181–211. Commun. 390 (2009) 1355–1360.
[6] J.R. Chapman, M.R. Taylor, S.J. Boulton, Playing the end game: DNA double- [42] X. Jiang, Y. Xu, B.D. Price, Acetylation of H2AX on lysine 36 plays a key role in the
strand break repair pathway choice, Mol. Cell 47 (2012) 497–510. DNA double-strand break repair pathway, FEBS Lett. 584 (2010) 2926–2930.
[7] G. Soria, S.E. Polo, G. Almouzni, Prime, repair, restore: the active role of chro- [43] A. Xie, S. Odate, G. Chandramouly, R. Scully, H2AX post-translational modifica-
matin in the DNA damage response, Mol. Cell 46 (2012) 722–734. tions in the ionizing radiation response and homologous recombination, Cell
[8] A. Gospodinov, Z. Herceg, Shaping chromatin for repair, Mutat. Res. 752 (2013) Cycle 9 (2010) 3602–3610.
45–60. [44] J.J. Wyrick, M.A. Parra, The role of histone H2A and H2B post-translational mod-
[9] B.D. Price, A.D. D’Andrea, Chromatin remodeling at DNA double-strand breaks, ifications in transcription: a genomic perspective, Biochim. Biophys. Acta 1789
Cell 152 (2013) 1344–1354. (2009) 37–44.
[10] D. Rossetto, A.W. Truman, S.J. Kron, J. Cote, Epigenetic modifications in double- [45] T. Ikura, V.V. Ogryzko, M. Grigoriev, R. Groisman, J. Wang, M. Horikoshi, R. Scully,
strand break DNA damage signaling and repair, Clin. Cancer Res. 16 (2010) J. Qin, Y. Nakatani, Involvement of the TIP60 histone acetylase complex in DNA
4543–4552. repair and apoptosis, Cell 102 (2000) 463–473.
[11] Y. Xu, B.D. Price, Chromatin dynamics and the repair of DNA double strand [46] A. Gupta, G.G. Sharma, C.S. Young, M. Agarwal, E.R. Smith, T.T. Paull, J.C. Lucch-
breaks, Cell Cycle 10 (2011) 261–267. esi, K.K. Khanna, T. Ludwig, T.K. Pandita, Involvement of human MOF in ATM
[12] R.D. Kornberg, Chromatin structure: a repeating unit of histones and DNA, function, Mol. Cell. Biol. 25 (2005) 5292–5305.
Science 184 (1974) 868–871. [47] S. Jha, E. Shibata, A. Dutta, Human Rvb1/Tip49 is required for the histone
[13] R. Margueron, D. Reinberg, Chromatin structure and the inheritance of epige- acetyltransferase activity of Tip60/NuA4 and for the downregulation of phos-
netic information, Nat. Rev. Genet. 11 (2010) 285–296. phorylation on H2AX after DNA damage, Mol. Cell. Biol. 28 (2008) 2690–2700.
[14] T. Kouzarides, Chromatin modifications and their function, Cell 128 (2007) [48] R. Murr, J.I. Loizou, Y.G. Yang, C. Cuenin, H. Li, Z.Q. Wang, Z. Herceg, Histone
693–705. acetylation by Trrap-Tip60 modulates loading of repair proteins and repair of
[15] E.I. Campos, D. Reinberg, Histones: annotating chromatin, Annu. Rev. Genet. 43 DNA double-strand breaks, Nat. Cell Biol. 8 (2006) 91–99.
(2009) 559–599. [49] Y. Sun, X. Jiang, Y. Xu, M.K. Ayrapetov, L.A. Moreau, J.R. Whetstine, B.D. Price,
[16] T. Suganuma, J.L. Workman, Signals and combinatorial functions of histone Histone H3 methylation links DNA damage detection to activation of the
modifications, Annu. Rev. Biochem. 80 (2011) 473–499. tumour suppressor Tip60, Nat. Cell Biol. 11 (2009) 1376–1382.
[17] K.M. Miller, S.P. Jackson, Histone marks: repairing DNA breaks within the con- [50] Y. Xu, Y. Sun, X. Jiang, M.K. Ayrapetov, P. Moskwa, S. Yang, D.M. Weinstock, B.D.
text of chromatin, Biochem. Soc. Trans. 40 (2012) 370–376. Price, The p400 ATPase regulates nucleosome stability and chromatin ubiquit-
[18] M.D. Shahbazian, M. Grunstein, Functions of site-specific histone acetylation ination during DNA repair, J. Cell Biol. 191 (2010) 31–43.
and deacetylation, Annu. Rev. Biochem. 76 (2007) 75–100. [51] A. Kaidi, S.P. Jackson, KAT5 tyrosine phosphorylation couples chromatin
[19] A.J. Ruthenburg, H. Li, D.J. Patel, C.D. Allis, Multivalent engagement of chromatin sensing to ATM signalling, Nature 498 (2013) 70–74.
modifications by linked binding modules, Nat. Rev. Mol. Cell Biol. 8 (2007) [52] H. Ogiwara, A. Ui, A. Otsuka, H. Satoh, I. Yokomi, S. Nakajima, A. Yasui, J.
983–994. Yokota, T. Kohno, Histone acetylation by CBP and p300 at double-strand break
[20] A.J. Bannister, P. Zegerman, J.F. Partridge, E.A. Miska, J.O. Thomas, R.C. Allshire, sites facilitates SWI/SNF chromatin remodeling and the recruitment of non-
T. Kouzarides, Selective recognition of methylated lysine 9 on histone H3 by homologous end joining factors, Oncogene 30 (2011) 2135–2146.
the HP1 chromo domain, Nature 410 (2001) 120–124. [53] X. Li, C.A. Corsa, P.W. Pan, L. Wu, D. Ferguson, X. Yu, J. Min, Y. Dou, MOF and
[21] S.I. Grewal, S. Jia, Heterochromatin revisited, Nat. Rev. Genet. 8 (2007) 35–46. H4 K16 acetylation play important roles in DNA damage repair by modulating
30 F. Gong, K.M. Miller / Mutation Research 750 (2013) 23–30

recruitment of DNA damage repair protein Mdc1, Mol. Cell. Biol. 30 (2010) [76] V. Di Cerbo, R. Schneider, Cancers with wrong HATs: the impact of acetylation,
5335–5347. Brief. Funct. Genomics 12 (2013) 231–243.
[54] K.M. Miller, J.V. Tjeertes, J. Coates, G. Legube, S.E. Polo, S. Britton, S.P. Jackson, [77] B. Groselj, N.L. Sharma, F.C. Hamdy, M. Kerr, A.E. Kiltie, Histone deacetylase
Human HDAC1 and HDAC2 function in the DNA-damage response to promote inhibitors as radiosensitisers: effects on DNA damage signalling and repair, Br.
DNA nonhomologous end-joining, Nat. Struct. Mol. Biol. 17 (2010) 1144–1151. J. Cancer 108 (2013) 748–754.
[55] O.M. Dovey, C.T. Foster, N. Conte, S.A. Edwards, J.M. Edwards, R. Singh, G. Vas- [78] M.A. Dawson, T. Kouzarides, B.J. Huntly, Targeting epigenetic readers in cancer,
siliou, A. Bradley, S.M. Cowley, Histone deacetylase 1 and 2 are essential for New Engl. J. Med. 367 (2012) 647–657.
normal T-cell development and genomic stability in mice, Blood 121 (2013) [79] S.R. Floyd, M.E. Pacold, Q. Huang, S.M. Clarke, F.C. Lam, I.G. Cannell, B.D. Bryson,
1335–1344. J. Rameseder, M.J. Lee, E.J. Blake, A. Fydrych, R. Ho, B.A. Greenberger, G.C. Chen,
[56] J. Tang, N.W. Cho, G. Cui, E.M. Manion, N.M. Shanbhag, M.V. Botuyan, G. Mer, R.A. A. Maffa, A.M. Del Rosario, D.E. Root, A.E. Carpenter, W.C. Hahn, D.M. Sabatini,
Greenberg, Acetylation limits 53BP1 association with damaged chromatin to C.C. Chen, F.M. White, J.E. Bradner, M.B. Yaffe, The bromodomain protein Brd4
promote homologous recombination, Nat. Struct. Mol. Biol. 20 (2013) 317–325. insulates chromatin from DNA damage signalling, Nature 498 (2013) 246–250.
[57] K.Y. Hsiao, C.A. Mizzen, Histone H4 deacetylation facilitates 53BP1 DNA damage [80] E.A. Lebel, P. Boukamp, S.T. Tafrov, Irradiation with heavy-ion particles changes
signaling and double-strand break repair, J. Mol. Cell Biol. 5 (2013) 157–165. the cellular distribution of human histone acetyltransferase HAT1, Mol. Cell.
[58] M. Shogren-Knaak, H. Ishii, J.M. Sun, M.J. Pazin, J.R. Davie, C.L. Peterson, His- Biochem. 339 (2010) 271–284.
tone H4-K16 acetylation controls chromatin structure and protein interactions, [81] I.M. Love, P. Sekaric, D. Shi, S.R. Grossman, E.J. Androphy, The histone
Science 311 (2006) 844–847. acetyltransferase PCAF regulates p21 transcription through stress-induced
[59] Y.C. Kim, G. Gerlitz, T. Furusawa, F. Catez, A. Nussenzweig, K.S. Oh, K.H. Krae- acetylation of histone H3, Cell Cycle 11 (2012) 2458–2466.
mer, Y. Shiloh, M. Bustin, Activation of ATM depends on chromatin interactions [82] M. Tillhon, O. Cazzalini, T. Nardo, D. Necchi, S. Sommatis, L.A. Stivala, A.I. Sco-
occurring before induction of DNA damage, Nat. Cell Biol. 11 (2009) 92–96. vassi, E. Prosperi, p300/CBP acetyl transferases interact with and acetylate the
[60] C. Das, M.S. Lucia, K.C. Hansen, J.K. Tyler, CBP/p300-mediated acetylation of nucleotide excision repair factor XPG, DNA Repair 11 (2012) 844–852.
histone H3 on lysine 56, Nature 459 (2009) 113–117. [83] R.K. Vempati, R.S. Jayani, D. Notani, A. Sengupta, S. Galande, D. Haldar, p300-
[61] R.A. McCord, E. Michishita, T. Hong, E. Berber, L.D. Boxer, R. Kusumoto, S. Guan, mediated acetylation of histone H3 lysine 56 functions in DNA damage
X. Shi, O. Gozani, A.L. Burlingame, V.A. Bohr, K.F. Chua, SIRT6 stabilizes DNA- response in mammals, J. Biol. Chem. 285 (2010) 28553–28564.
dependent protein kinase at chromatin for DNA double-strand break repair, [84] M.K. Kim, J.M. Shin, H.C. Eun, J.H. Chung, The role of p300 histone acetyltrans-
Aging (Albany, N.Y.) (Milano) 1 (2009) 109–121. ferase in UV-induced histone modifications and MMP-1 gene transcription,
[62] H.S. Lee, J.H. Park, S.J. Kim, S.J. Kwon, J. Kwon, A cooperative activation loop PloS ONE 4 (2009) e4864.
among SWI/SNF, gamma-H2AX and H3 acetylation for DNA double-strand [85] E.R. Jang, J.D. Choi, J.S. Lee, Acetyltransferase p300 regulates NBS1-mediated
break repair, EMBO J. 29 (2010) 1434–1445. DNA damage response, FEBS Lett. 585 (2011) 47–52.
[63] C. Alabert, A. Groth, Chromatin replication and epigenome maintenance, Nat. [86] C.P. Rubbi, J. Milner, p53 is a chromatin accessibility factor for nucleotide exci-
Rev. Mol. Cell. Biol. 13 (2012) 153–167. sion repair of DNA damage, EMBO J. 22 (2003) 975–986.
[64] B.M. Sirbu, F.B. Couch, J.T. Feigerle, S. Bhaskara, S.W. Hiebert, D. Cortez, Analysis [87] A.M. Buchmann, J.R. Skaar, J.A. DeCaprio, Activation of a DNA damage check-
of protein dynamics at active, stalled, and collapsed replication forks, Genes point response in a TAF1-defective cell line, Mol. Cell. Biol. 24 (2004)
Dev. 25 (2011) 1320–1327. 5332–5339.
[65] S. Bhaskara, S.K. Knutson, G. Jiang, M.B. Chandrasekharan, A.J. Wilson, S. Zheng, [88] Y. Sun, X. Jiang, S. Chen, N. Fernandes, B.D. Price, A role for the Tip60 histone
A. Yenamandra, K. Locke, J.L. Yuan, A.R. Bonine-Summers, C.E. Wells, J.F. Kaiser, acetyltransferase in the acetylation and activation of ATM, Proc. Natl. Acad. Sci.
M.K. Washington, Z. Zhao, F.F. Wagner, Z.W. Sun, F. Xia, E.B. Holson, D. Khabele, U. S. A. 102 (2005) 13182–13187.
S.W. Hiebert, Hdac3 is essential for the maintenance of chromatin structure [89] G.G. Sharma, S. So, A. Gupta, R. Kumar, C. Cayrou, N. Avvakumov, U. Bhadra,
and genome stability, Cancer Cell 18 (2010) 436–447. R.K. Pandita, M.H. Porteus, D.J. Chen, J. Cote, T.K. Pandita, MOF and histone H4
[66] P. Oberdoerffer, S. Michan, M. McVay, R. Mostoslavsky, J. Vann, S.K. Park, A. acetylation at lysine 16 are critical for DNA damage response and double-strand
Hartlerode, J. Stegmuller, A. Hafner, P. Loerch, S.M. Wright, K.D. Mills, A. Bonni, break repair, Mol. Cell. Biol. 30 (2010) 3582–3595.
B.A. Yankner, R. Scully, T.A. Prolla, F.W. Alt, D.A. Sinclair, SIRT1 redistribution on [90] C. Cotta-Ramusino, E.R. McDonald 3rd, K. Hurov, M.E. Sowa, J.W. Harper, S.J.
chromatin promotes genomic stability but alters gene expression during aging, Elledge, A DNA damage response screen identifies RHINO, a 9-1-1 and TopBP1
Cell 135 (2008) 907–918. interacting protein required for ATR signaling, Science 332 (2011) 1313–1317.
[67] W. Dang, K.K. Steffen, R. Perry, J.A. Dorsey, F.B. Johnson, A. Shilatifard, M. Kae- [91] G.D. Kao, W.G. McKenna, M.G. Guenther, R.J. Muschel, M.A. Lazar, T.J. Yen, His-
berlein, B.K. Kennedy, S.L. Berger, Histone H4 lysine 16 acetylation regulates tone deacetylase 4 interacts with 53BP1 to mediate the DNA damage response,
cellular lifespan, Nature 459 (2009) 802–807. J. Cell Biol. 160 (2003) 1017–1027.
[68] R.J. O’Sullivan, S. Kubicek, S.L. Schreiber, S.L. Schreiber, KarlsederF J., Reduced [92] V. Basile, R. Mantovani, C. Imbriano, DNA damage promotes histone deacetylase
histone biosynthesis and chromatin changes arising from a damage signal at 4 nuclear localization and repression of G2/M promoters, via p53 C-terminal
telomeres, Nat. Struct. Mol. Biol. 17 (2010) 1218–1225. lysines, J. Biol. Chem. 281 (2006) 2347–2357.
[69] Z. Wang, C. Zang, K. Cui, D.E. Schones, A. Barski, W. Peng, K. Zhao, Genome-wide [93] S. Kotian, S. Liyanarachchi, A. Zelent, J.D. Parvin, Histone deacetylases 9 and
mapping of HATs and HDACs reveals distinct functions in active and inactive 10 are required for homologous recombination, J. Biol. Chem. 286 (2011)
genes, Cell 138 (2009) 1019–1031. 7722–7726.
[70] J.H. Waterborg, Dynamics of histone acetylation in vivo. A function for acety- [94] H.M. O’Hagan, H.P. Mohammad, S.B. Baylin, Double strand breaks can initiate
lation turnover? Biochem. Cell Biol. 80 (2002) 363–378. gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous
[71] M.X. Qian, Y. Pang, C.H. Liu, K. Haratake, B.Y. Du, D.Y. Ji, G.F. Wang, Q.Q. Zhu, W. promoter CpG island, PLoS Genet. 4 (2008) e1000155.
Song, Y. Yu, X.X. Zhang, H.T. Huang, S. Miao, L.B. Chen, Z.H. Zhang, Y.N. Liang, [95] R.H. Wang, K. Sengupta, C. Li, H.S. Kim, L. Cao, C. Xiao, S. Kim, X. Xu, Y. Zheng, B.
S. Liu, H. Cha, D. Yang, Y. Zhai, T. Komatsu, F. Tsuruta, H. Li, C. Cao, W. Li, G.H. Chilton, R. Jia, Z.M. Zheng, E. Appella, X.W. Wang, T. Ried, C.X. Deng, Impaired
Li, Y. Cheng, T. Chiba, L. Wang, A.L. Goldberg, Y. Shen, X.B. Qiu, Acetylation- DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant
mediated proteasomal degradation of core histones during DNA repair and mice, Cancer Cell 14 (2008) 312–323.
spermatogenesis, Cell 153 (2013) 1012–1024. [96] A. Kaidi, B.T. Weinert, C. Choudhary, S.P. Jackson, Human SIRT6 promotes DNA
[72] J.S. Iacovoni, P. Caron, I. Lassadi, E. Nicolas, L. Massip, D. Trouche, G. Legube, end resection through CtIP deacetylation, Science 329 (2010) 1348–1353.
High-resolution profiling of gammaH2AX around DNA double strand breaks in [97] R. Mostoslavsky, K.F. Chua, D.B. Lombard, W.W. Pang, M.R. Fischer, L. Gellon, P.
the mammalian genome, EMBO J. 29 (2010) 1446–1457. Liu, G. Mostoslavsky, S. Franco, M.M. Murphy, K.D. Mills, P. Patel, J.T. Hsu, A.L.
[73] Y. Xu, M.K. Ayrapetov, C. Xu, O. Gursoy-Yuzugullu, Y. Hu, B.D. Price, Histone Hong, E. Ford, H.L. Cheng, C. Kennedy, N. Nunez, R. Bronson, D. Frendewey, W.
H2A.Z. controls a critical chromatin remodeling step required for DNA double- Auerbach, D. Valenzuela, M. Karow, M.O. Hottiger, S. Hursting, J.C. Barrett, L.
strand break repair, Mol. Cell 48 (2012) 723–733. Guarente, R. Mulligan, B. Demple, G.D. Yancopoulos, F.W. Alt, Genomic insta-
[74] W.T. Chen, A. Alpert, C. Leiter, F. Gong, S.P. Jackson, K.M. Miller, Systematic bility and aging-like phenotype in the absence of mammalian SIRT6, Cell 124
identification of functional residues in mammalian histone H2AX, Mol. Cell. (2006) 315–329.
Biol. 33 (2013) 111–126. [98] C.A. Musselman, M.E. Lalonde, J. Cote, T.G. Kutateladze, Perceiving the epi-
[75] I. Barbieri, E. Cannizzaro, M.A. Dawson, Bromodomains as therapeutic targets genetic landscape through histone readers, Nat. Struct. Mol. Biol. 19 (2012)
in cancer, Brief. Funct. Genomics 12 (2013) 219–230. 1218–1227.

Das könnte Ihnen auch gefallen