fbpx
Wikipedia

Histone acetyltransferase

Histone acetyltransferases (HATs) are enzymes that acetylate conserved lysine amino acids on histone proteins by transferring an acetyl group from acetyl-CoA to form ε-N-acetyllysine. DNA is wrapped around histones, and, by transferring an acetyl group to the histones, genes can be turned on and off. In general, histone acetylation increases gene expression.

GCN5 Histone acetyltransferase
GCN5 histone acetyltransferase domain homo24-mer, Human. Based on PDB structure: 5trm[1]
Identifiers
EC no.2.3.1.48
CAS no.9054-51-7
Databases
IntEnzIntEnz view
BRENDABRENDA entry
ExPASyNiceZyme view
KEGGKEGG entry
MetaCycmetabolic pathway
PRIAMprofile
PDB structuresRCSB PDB PDBe PDBsum
Gene OntologyAmiGO / QuickGO
Search
PMCarticles
PubMedarticles
NCBIproteins

In general, histone acetylation is linked to transcriptional activation and associated with euchromatin. Euchromatin, which is less densely compact, allows transcription factors to bind more easily to regulatory sites on DNA, causing transcriptional activation. When it was first discovered, it was thought that acetylation of lysine neutralizes the positive charge normally present, thus reducing affinity between histone and (negatively charged) DNA, which renders DNA more accessible to transcription factors. Research has emerged, since, to show that lysine acetylation and other posttranslational modifications of histones generate binding sites for specific protein–protein interaction domains, such as the acetyllysine-binding bromodomain[citation needed]. Histone acetyltransferases can also acetylate non-histone proteins, such as nuclear receptors and other transcription factors to facilitate gene expression.

HAT families edit

HATs are traditionally divided into two different classes based on their subcellular localization.[2] Type A HATs are located in the nucleus and are involved in the regulation of gene expression through acetylation of nucleosomal histones in the context of chromatin.[3] They contain a bromodomain, which helps them recognize and bind to acetylated lysine residues on histone substrates. Gcn5, p300/CBP, and TAFII250 are some examples of type A HATs that cooperate with activators to enhance transcription. Type B HATs are located in the cytoplasm and are responsible for acetylating newly synthesized histones prior to their assembly into nucleosomes. These HATs lack a bromodomain, as their targets are unacetylated. The acetyl groups added by type B HATs to the histones are removed by HDACs once they enter the nucleus and are incorporated into chromatin. Hat1 is one of the few known examples of a type B HAT.[4] Despite this historical classification of HATs, some HAT proteins function in multiple complexes or locations and would thus not easily fit into a particular class.[5]

 
Relative sizes and locations of important domains for representative HATs (HAT = catalytic acetyltransferase domain; Bromo = bromodomain; Chromo = chromodomain; Zn = zinc finger domain). The number of amino acid residues in each HAT is indicated at the right in each example.

Gcn5-related N-acetyltransferases (GNATs) edit

HATs can be grouped into several different families based on sequence homology as well as shared structural features and functional roles. The Gcn5-related N-acetyltransferase (GNAT) family includes Gcn5, PCAF, Hat1, Elp3, Hpa2, Hpa3, ATF-2, and Nut1. These HATs are generally characterized by the presence of a bromodomain, and they are found to acetylate lysine residues on histones H2B, H3,[6] and H4.[2] All members of the GNAT family are characterized by up to four conserved motifs (A-D) found within the catalytic HAT domain. This includes the most highly conserved motif A, which contains an Arg/Gln-X-X-Gly-X-Gly/Ala sequence that is important for acetyl-CoA recognition and binding.[4] The C motif is found in most GNATs, but it is not present in the majority of other known HATs.[5] The yeast Gcn5 (general control nonderepressible-5) HAT is one of the best-characterized members of this family. It has four functional domains, including an N-terminal domain, a highly conserved catalytic (HAT) domain, an Ada2 interaction domain, and a C-terminal bromodomain. PCAF (p300/CBP-associated factor) and GCN5 are mammalian GNATs that share a high degree of homology throughout their sequences. These proteins have a 400-residue N-terminal region that is absent in yeast Gcn5, but their HAT functions are evolutionarily conserved with respect to the latter. Hat1 was the first HAT protein to be identified. It is responsible for most of the cytoplasmic HAT activity in yeast, and it binds strongly to histone H4 by virtue of its association with an additional subunit, Hat2. Elp3 is an example of a type A HAT found in yeast. It is part of the RNA polymerase II holoenzyme and plays a role in transcriptional elongation.

MYST HATs edit

The MYST family of HATs is named after its four founding members MOZ, Ybf2 (Sas3), Sas2, and Tip60.[2] Other important members include Esa1, MOF, MORF, and HBO1. These HATs are typically characterized by the presence of zinc fingers and chromodomains, and they are found to acetylate lysine residues on histones H2A, H3, and H4. Several MYST family proteins contain zinc fingers as well as the highly conserved motif A found among GNATs that facilitates acetyl-CoA binding.[4] A cysteine-rich region located in the N terminus of the HAT domain of MYST proteins is involved in zinc binding, which is essential for HAT activity.[7] Tip60 (Tat-interactive protein, 60 kDa) was the first human MYST family member to exhibit HAT activity. Sas3 found in yeast is a homolog of MOZ (monocytic leukemia zinc finger protein), which is an oncogene found in humans. Esa1 was the first essential HAT to be found in yeast, and MOF is its homolog in fruit flies. The HAT activity of the latter is required for the twofold increased transcription of the male X chromosome (dosage compensation) in flies. Human HBO1 (HAT bound to ORC1) was the first HAT shown to associate with components of the origin of replication complex. MORF (MOZ-related factor) exhibits very close homology to MOZ throughout its entire length.[5] It contains an N-terminal repression region that decreases its HAT activity in vitro as well as a C-terminal activation domain that is functional in the absence of the HAT domain.

Others edit

In addition to those that are members of the GNAT and MYST families, there are several other proteins found typically in higher eukaryotes that exhibit HAT activity. These include p300/CBP, nuclear receptor coactivators (e.g., ACTR/SRC-1), TAFII250, TFIIIC, Rtt109, and CLOCK. p300/CBP are metazoan-specific[8] and contain several zinc finger regions, a bromodomain, a catalytic (HAT) domain, and regions that interact with other transcription factors.[4] Importantly, the HAT domain shows no sequence homology to other known HATs,[9] and it is required for p300/CBP to function in transcriptional activation.[4] In addition, these proteins contain several HAT domain motifs (A, B, and D) that are similar to those of the GNATs. They also possess a novel motif E that is homologous to sequences in the HAT domains of GNATs. TFIIIC is one of the general transcription factors involved in RNA polymerase III-mediated transcription. Three components in the human protein have been shown to possess independent HAT activity (hTFIIIC220, hTFIIIC110, and hTFIIIC90).[10] Rtt109 is a fungal-specific HAT that requires association with histone chaperone proteins for activity.[8] The HAT activities of the human TAFII250 and CLOCK coactivators have not been studied as extensively. TAFII250 is one of the TBP-associated factor subunits of TFIID, and it shares a Gly-X-Gly pattern with Gcn5 that is important for HAT activity.[5] CLOCK is a circadian rhythm master regulator that functions with BMAL1 to carry out its HAT activity.[11]

Nuclear receptor coactivators edit

Three important nuclear receptor coactivators that display HAT activity are SRC-1, ACTR, and TIF-2. Human SRC-1 (steroid receptor coactivator-1) is known to interact with p300/CBP and PCAF, and its HAT domain is located in its C-terminal region. ACTR (also known as RAC3, AIB1, and TRAM-1 in humans) shares significant sequence homology with SRC-1, in particular in the N-terminal and C-terminal (HAT) regions as well as in the receptor and coactivator interaction domains.[5] ACTR also interacts with p300/CBP and PCAF. The former can prevent ACTR from binding to and activating its receptor by acetylating it in its receptor interaction domain. TIF-2 (transcriptional intermediary factor 2; also known as GRIP1) is another nuclear receptor coactivator with HAT activity, and it also interacts with p300/CBP.

A table summarizing the different families of HATs along with their associated members, parent organisms, multisubunit complexes, histone substrates, and structural features is presented below.[2][5][8][10][12][13][14][15][16][17]

Family Members Organism Associated complexes Substrate specificity Structural features
GNAT Gcn5 S. cerevisiae SAGA, SLIK (SALSA), ADA,

HAT-A2

H2B, H3, (H4) Bromodomain
GCN5 D. melanogaster SAGA, ATAC H3, H4 Bromodomain
GCN5 H. sapiens STAGA, TFTC H3, (H4, H2B) Bromodomain
PCAF H. sapiens PCAF H3, H4 Bromodomain
Hat1 S. cerevisiae
H. sapiens
HAT-B, NuB4, HAT-A3 H4, (H2A)
Elp3 S. cerevisiae Elongator H3, H4, (H2A, H2B)
Hpa2 S. cerevisiae HAT-B H3, H4
Hpa3 S. cerevisiae H3, H4
ATF-2 S. cerevisiae
H. sapiens
H2B, H4
Nut1 S. cerevisiae Mediator H3, H4
MYST Esa1 S. cerevisiae NuA4, piccolo NuA4 H2A, H4, (H2B, H3) Chromodomain
Sas2 S. cerevisiae SAS, NuA4 H4, (H2A, H3)
Sas3 (Ybf2) S. cerevisiae NuA3 H3, (H4, H2A)
Tip60 H. sapiens Tip60, NuA4 H2A, H4, (H3) Chromodomain
MOF D. melanogaster MSL H4, (H2A, H3) Chromodomain
MOZ H. sapiens MSL H3, H4
MORF H. sapiens MSL H3, H4
HBO1 H. sapiens ORC H3, H4
p300/CBP p300 H. sapiens H2A, H2B, H3, H4 Bromodomain
CBP H. sapiens H2A, H2B, H3, H4 Bromodomain
SRC

(nuclear receptor coactivators)

SRC-1 H. sapiens ACTR/SRC-1 H3, H4
ACTR (RAC3, AIB1, TRAM-1, SRC-3) H. sapiens ACTR/SRC-1 H3, H4
TIF-2 (GRIP1) H. sapiens H3, H4
Other TAFII250 (TAF1) S. cerevisiae
H. sapiens
TFIID H3, H4, (H2A) Bromodomain
TFIIIC (p220, p110, p90) H. sapiens TFIIIC H2A, H3, H4
Rtt109 S. cerevisiae Histone chaperones H3
CLOCK H. sapiens H3, H4

Overall structure edit

 
Crystal structure of Tetrahymena Gcn5 with bound coenzyme A and histone H3 peptide (PDB 1QSN). The central core (green), flanking N- and C-terminal segments (blue), coenzyme A (orange), and histone peptide (red) are shown.

In general, HATs are characterized by a structurally conserved core region made up of a three-stranded β-sheet followed by a long α-helix parallel to and spanning one side of it.[7][8] The core region, which corresponds to motifs A, B, and D of the GNAT proteins,[4] is flanked on opposite sides by N- and C-terminal α/β segments that are structurally unique for a given HAT family.[7][8] The central core and the flanking segments together form a cleft over the former, which is where histone substrates can bind prior to catalysis.[8] While the central core domain (motif A in GNATs) is involved in acetyl-CoA binding and catalysis, the N- and C-terminal segments assist in binding histone substrates.[7] Unique features related to the sequence and/or structure of the N- and C-terminal regions for different HAT families may help to explain some observed differences among HATs in histone substrate specificity. CoA binding has been observed to widen the histone binding groove in the central core by moving the C-terminal segment of Gcn5 outward. In addition, since contacts between CoA and protein facilitate the formation of favorable histone-protein contacts, it is likely that CoA binding precedes histone binding in vivo.

GNAT and MYST families edit

HATs in the GNAT family are most notably characterized by an approximately 160-residue HAT domain and a C-terminal bromodomain, which binds to acetylated lysine residues.[7] Those in the MYST family have HAT domains that are about 250 residues in length. Many MYST proteins also contain a cysteine-rich, zinc-binding domain within the HAT region in addition to an N-terminal chromodomain, which binds to methylated lysine residues.

On a broader scale, the structures of the catalytic domains of GNAT proteins (Gcn5, PCAF) exhibit a mixed α/β globular fold with a total of five α-helices and six β-strands.[4] The overall topology resembles a vise, with the central core of the protein at the base and the N- and C-terminal segments on the sides.

p300/CBP family edit

The p300/CBP HATs have larger HAT domains (about 500 residues) than those present in the GNAT and MYST families.[7] They also contain a bromodomain as well as three cysteine/histidine-rich domains that are thought to mediate interactions with other proteins. The structure of p300/CBP is characterized by an elongated globular domain, which contains a seven-stranded β-sheet in the center that is surrounded by nine α-helices and several loops.[9] The structure of the central core region associated with acetyl-CoA binding is conserved with respect to GNAT and MYST HATs, but there are many structural differences in the regions flanking this central core. Overall, the structural data is consistent with the fact that p300/CBP HATs are more promiscuous than GNAT and MYST HATs with respect to substrate binding.

Rtt109 edit

The structure of Rtt109 is very similar to that of p300, despite there only being 7% sequence identity between the two proteins.[9] There is a seven-stranded β-sheet that is surrounded by α-helices as well as a loop that is involved in acetyl-CoA substrate binding. Despite the conserved structure, Rtt109 and p300/CBP are functionally unique. For instance, the substrate binding site of the former is more similar to that of the GNAT and MYST HATs. In addition, the residues in the active site of each enzyme are distinct, which suggests that they employ different catalytic mechanisms for acetyl group transfer.

Catalytic mechanisms edit

The basic mechanism catalyzed by HATs involves the transfer of an acetyl group from acetyl-CoA to the ε-amino group of a target lysine side-chain within a histone.[8] Different families of HATs employ unique strategies in order to effect such a transformation.

 
Catalytic mechanisms of GNAT and MYST family HATs. (A) General mechanism of GNAT HATs. (B) General mechanism of MYST HATs.

GNAT family edit

Members of the GNAT family have a conserved glutamate residue that acts as a general base for catalyzing the nucleophilic attack of the lysine amine on the acetyl-CoA thioester bond.[8] These HATs use an ordered sequential bi-bi mechanism wherein both substrates (acetyl-CoA and histone) must bind to form a ternary complex with the enzyme before catalysis can occur. Acetyl-CoA binds first, followed by the histone substrate. A conserved glutamate residue (Glu173 in yeast Gcn5) activates a water molecule for removal of a proton from the amine group on lysine, which activates it for direct nucleophilic attack on the carbonyl carbon of enzyme-bound acetyl-CoA. After the reaction, the acetylated histone is released first followed by CoA.[4][8]

MYST family edit

Studies of yeast Esa1 from the MYST family of HATs have revealed a ping-pong mechanism involving conserved glutamate and cysteine residues.[18] The first part of the reaction involves the formation of a covalent intermediate in which a cysteine residue becomes acetylated following nucleophilic attack of this residue on the carbonyl carbon of acetyl-CoA. Then, a glutamate residue acts as a general base to facilitate transfer of the acetyl group from the cysteine to the histone substrate in a manner analogous to the mechanism used by GNATs. When Esa1 is assembled in the piccolo NuA4 complex, it loses its dependence on the cysteine residue for catalysis, which suggests that the reaction may proceed via a ternary bi-bi mechanism when the enzyme is part of a physiologically relevant multiprotein complex.

p300/CBP family edit

In human p300, Tyr1467 acts as a general acid and Trp1436 helps orient the target lysine residue of the histone substrate into the active site.[8] These two residues are highly conserved within the p300/CBP HAT family and, unlike enzymes in the GNAT and MYST families, p300 does not employ a general base for catalysis. Rather, it is likely that members of the p300/CBP family use a Theorell-Chance (i.e., “hit-and-run”) acetyl transfer mechanism.

Rtt109 edit

Rtt109 is likely to employ a mechanism that is different from that of the other HATs.[9] The yeast enzyme has very low catalytic activity in the absence of the histone chaperone proteins Asf1 and Vps75, which may be involved in delivering histone substrates to the enzyme for acetylation.[8] Moreover, a general acid or base have not yet been identified for this HAT.

Substrate binding and specificity edit

The structures of several HAT domains bound to acetyl-CoA and histone substrate peptides reveal that the latter bind across a groove on the protein that is formed by the central core region at the base and is flanked on opposite sides by the variable N- and C-terminal segments that mediate the majority of the interactions with the substrate peptide.[8] It is likely that these variable regions are at least in part responsible for the observed specificity of different HATs for various histone substrates.

Members of the GNAT and MYST families as well as Rtt109 exhibit greater substrate selectivity than p300/CBP, which is rather promiscuous with regard to substrate binding.[8] Whereas it appears that only three to five residues on either side of the lysine to be acetylated are necessary for effective substrate binding and catalysis by members of the GNAT and p300/CBP families, more distal regions of the substrate may be important for efficient acetylation by MYST family HATs.[19]

Lysine selectivity edit

Different HATs, usually in the context of multisubunit complexes, have been shown to acetylate specific lysine residues in histones.

GNAT family edit

Gcn5 cannot acetylate nucleosomal histones in the absence of other protein factors.[5] In the context of complexes like SAGA and ADA, however, Gcn5 is able to acetylate H3K14 among other sites within histones H2B, H3, and H4 (e.g., H3K9, H3K36, H4K8, H4K16).[3][4][7][19] Both Gcn5 and PCAF have the strongest site preference for H3K14, either as a free histone or within a nucleosome.[4][7] Hat1 acetylates H4K5 and H4K12, and Hpa2 acetylates H3K14 in vitro.[4][5]

MYST family edit

In flies, acetylation of H4K16 on the male X chromosome by MOF in the context of the MSL complex is correlated with transcriptional upregulation as a mechanism for dosage compensation in these organisms.[2] In humans, the MSL complex carries out the majority of genome-wide H4K16 acetylation. In the context of their cognate complexes, Sas2 (SAS) and Esa1 (NuA4) also carry out acetylation of H4K16, in particular in the telomere regions of chromosomes. Sas2 is also observed to acetylate H3K14 in vitro on free histones.[12] Esa1 can also acetylate H3K14 in vitro on free histones as well as H2AK5, H4K5, H4K8, and H4K12 either in vitro or in vivo on nucleosomal histones. H2AK7 and H2BK16 are also observed to be acetylated by Esa1 in vivo. Notably, neither Sas2 nor Esa1 can acetylate nucleosomal histones in vitro as a free enzyme. This happens to be the case as well for Sas3, which is observed to acetylate H3K9 and H3K14 in vivo as well as lysine residues on H2A and H4. MOZ can also acetylate H3K14.[19]

Others edit

p300/CBP acetylate all four nucleosomal core histones equally well.[4] In vitro, they have been observed to acetylate H2AK5, H2BK12, H2BK15, H3K14, H3K18, H4K5, and H4K8.[5] SRC-1 acetylates H3K9 and H3K14, TAFII230 (Drosophila homolog of human TAFII250) acetylates H3K14, and Rtt109 acetylates H3K9, H3K23,[19] and H3K56 in the presence of either Asf1 or Vps75.[9]

Non-histone substrates (in vitro) edit

In addition to the core histones, certain HATs acetylate a number of other cellular proteins including transcriptional activators, basal transcription factors, structural proteins, polyamines, and proteins involved in nuclear import.[4] Acetylation of these proteins can alter their ability to interact with their cognate DNA and/or protein substrates. The idea that acetylation can affect protein function in this manner has led to inquiry regarding the role of acetyltransferases in signal transduction pathways and whether an appropriate analogy to kinases and phosphorylation events can be made in this respect.

PCAF edit

PCAF and p300/CBP are the main HATs that have been observed to acetylate a number of non-histone proteins. For PCAF, these include the non-histone chromatin (high-mobility group (HMG)) proteins HMG-N2/HMG17 and HMG-I(Y), the transcriptional activators p53, MyoD, E2F(1-3), and HIV Tat, and the general transcription factors TFIIE and TFIIF.[5] Other proteins include CIITA, Brm (chromatin remodeler), NF-κB (p65), TAL1/SCL, Beta2/NeuroD, C/EBPβ, IRF2, IRF7, YY1, KLF13, EVI1, AME, ER81, and the androgen receptor (AR).[20] PCAF has also been observed to acetylate c-MYC, GATA-2, retinoblastoma (Rb), Ku70, and E1A adenovirus protein.[21] It can also autoacetylate, which facilitates intramolecular interactions with its bromodomain that may be involved in the regulation of its HAT activity.[4]

p300/CBP edit

p300/CBP have many non-histone substrates, including the non-histone chromatin proteins HMG1, HMG-N1/HMG14, and HMG-I(Y), the transcriptional activators p53, c-Myb, GATA-1, EKLF, TCF, and HIV Tat, the nuclear receptor coactivators ACTR, SRC-1, and TIF-2, and the general transcription factors TFIIE and TFIIF.[5] Other substrates include the transcription factors Sp1, KLF5, FOXO1, MEF2C, SRY, GATA-4, and HNF-6,[12] HMG-B2,[21] STAT3, the androgen and estrogen (α) receptors, GATA-2, GATA-3, MyoD, E2F(1-3), p73α, retinoblastoma (Rb), NF-κB (p50, p65), Smad7, importin-α, Ku70, YAP1,[22] E1A adenovirus protein, and S-HDAg (hepatitis delta virus small delta antigen).[21] p300/CBP have also been observed to acetylate β-catenin, RIP140, PCNA, the DNA metabolic enzymes flap endonuclease-1, thymine DNA glycosylase, and Werner syndrome DNA helicase, STAT6, Runx1 (AML1), UBF, Beta2/NeuroD, CREB, c-Jun, C/EBPβ, NF-E2, SREBP, IRF2, Sp3, YY1, KLF13, EVI1, BCL6, HNF-4, ER81 and FOXO4 (AFX).[20]

Multisubunit HAT complexes edit

The formation of multisubunit complexes has been observed to modulate the substrate specificity of HATs.[12] In general, while recombinant HATs are able to acetylate free histones, HATs can acetylate nucleosomal histones only when they are in their respective in vivo HAT complexes.[5] Some of the proteins that associate with HATs in these complexes function by targeting the HAT complex to nucleosomes at specific regions in the genome.[2][12] For instance, it has been observed that HAT complexes (e.g. SAGA, NuA3) often use methylated histones as docking sites so that the catalytic HAT subunit can carry out histone acetylation more effectively.[2]

In addition, the formation of multisubunit HAT complexes influences the lysine specificity of HATs.[12] The specific lysine residues that a given HAT acetylates may become either broader or more restricted in scope upon association with its respective complex. For example, the lysine specificity of MYST family HATs toward their histone substrates becomes more restricted when they associate with their complexes. In contrast, Gcn5 acquires the ability to acetylate multiple sites in both histones H2B and H3 when it joins other subunits to form the SAGA and ADA complexes.[4] Moreover, the acetylation site specificity of Rtt109 is dictated by its association with either Vps75 or Asf1.[19] When in complex with the former, Rtt109 acetylates H3K9 and H3K27, but, when in complex with the latter, it preferentially acetylates H3K56.[8]

Regulation of HAT activity edit

The catalytic activity of HATs is regulated by two types of mechanisms: (1) interaction with regulatory protein subunits and (2) autoacetylation.[8] A given HAT may be regulated in multiple ways, and the same effector may actually lead to different outcomes under different conditions.[4] Although it is clear that the association of HATs with multiprotein complexes provides a mechanism for the regulation of both HAT activity and substrate specificity in vivo, the molecular basis for how this actually occurs is still largely unknown.[8] However, data suggests that associated subunits may contribute to catalysis at least in part by facilitating productive binding of the HAT complex to its native histone substrates.

The MYST family of HATs, p300/CBP, and Rtt109 have all been shown to be regulated by autoacetylation.[8] Human MOF as well as yeast Esa1 and Sas2 are autoacetylated at a conserved active site lysine residue, and this modification is required for their function in vivo. Human p300 contains a highly basic loop embedded in the middle of its HAT domain that is hyperacetylated in the active form of the enzyme.[8][9] It has been proposed that, upon autoacetylation, this loop is released from the electronegative substrate binding site where it sits in the inactive HAT.[23] Acetylation of yeast Rtt109 at Lys290 is also required for it to exhibit full catalytic activity.[24] Some HATs are also inhibited by acetylation. For example, the HAT activity of the nuclear receptor coactivator ACTR is inhibited upon acetylation by p300/CBP.[4]

Interaction with HDACs edit

Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are recruited to their target promoters through physical interactions with sequence-specific transcription factors. They usually function within a multisubunit complex in which the other subunits are necessary for them to modify histone residues around the binding site. These enzymes can also modify non-histone proteins.

Biological role edit

Chromatin remodeling edit

 
Histone tails and their function in chromatin formation

Histone acetyltransferases serve many biological roles inside the cell. Chromatin is a combination of proteins and DNA found in the nucleus, and it undergoes many structural changes as different cellular events such as DNA replication, DNA repair, and transcription occur.[25] Chromatin in the cell can be found in two states: condensed and uncondensed. The latter, known as euchromatin, is transcriptionally active, whereas the former, known as heterochromatin, is transcriptionally inactive.[25][26] Histones comprise the protein portion of chromatin. There are five different histone proteins: H1, H2A, H2B, H3, and H4. A core histone is formed when two of each histone subtype, excluding H1, form a quaternary complex. This octameric complex, in association with the 147 base pairs of DNA coiled around it, forms the nucleosome.[4] Histone H1 locks the nucleosome complex together, and it is the last protein to bind in the complex.

Histones tend to be positively charged proteins with N-terminal tails that stem from the core. The phosphodiester backbone of DNA is negative, which allows for strong ionic interactions between histone proteins and DNA. Histone acetyltransferases transfer an acetyl group to specific lysine residues on histones, which neutralizes their positive charge and thus reduces the strong interactions between the histone and DNA.[25] Acetylation is also thought to perturb interactions between individual nucleosomes and act as interaction sites for other DNA-associated proteins.[4]

There can be different levels of histone acetylation as well as other types of modifications, allowing the cell to have control over the level of chromatin packing during different cellular events such as replication, transcription, recombination, and repair. Acetylation is not the only regulatory post-translational modification to histones that dictates chromatin structure; methylation, phosphorylation, ADP-ribosylation, and ubiquitination have also been reported.[4][25] These combinations of different covalent modifications on the N-terminal tails of histones have been referred to as the histone code, and it is thought that this code may be heritable and preserved in the next cell generation.[26]

H3 and H4 histone proteins are the primary targets of HATs, but H2A and H2B are also acetylated in vivo. Lysines 9, 14, 18, and 23 of H3 and lysines 5, 8, 12, and 16 of H4 are all targeted for acetylation.[4][25] Lysines 5, 12, 15, and 20 are acetylated on histone H2B, while only lysines 5 and 9 have been observed to be acetylated on histone H2A.[4][25][26] With so many different sites for acetylation, a high level of specificity can be achieved in triggering specific responses. An example of this specificity is when histone H4 is acetylated at lysines 5 and 12. This acetylation pattern has been seen during histone synthesis. Another example is acetylation of H4K16, which has been associated with dosage compensation of the male X chromosome in Drosophila melanogaster.[2][4]

Gene expression edit

 
Schematic showing the role of HATs in gene transcription.

Histone modifications modulate the packing of chromatin. The level of packing of the DNA is important for gene transcription, since the transcriptional machinery must have access to the promoter in order for transcription to occur.[4] Neutralization of charged lysine residues by HATs allows for the chromatin to decondense so that this machinery has access to the gene to be transcribed. However, acetylation is not always associated with enhanced transcriptional activity. For instance, acetylation of H4K12 has been associated with condensed and transcriptionally inactive chromatin.[27] In addition, some histone modifications are associated with both enhanced and repressed activity, in a context-dependent manner.[28]

HATs act as transcriptional co-activators or gene silencers and are most often found in large complexes made up of 10 to 20 subunits, some of which shared among different HAT complexes.[25] These complexes include SAGA (Spt/Ada/Gcn5L acetyltransferase), PCAF, ADA (transcriptional adaptor), TFIID (transcription factor II D), TFTC (TBP-free TAF-containing complex), and NuA3/NuA4 (nucleosomal acetyltransferases of H3 and H4).[2][25] These complexes modulate HAT specificity by bringing HATs to their target genes where they can then acetylate nucleosomal histones.[25] Some HAT transcriptional co-activators contain a bromodomain, a 110-amino acid module that recognizes acetylated lysine residues and is functionally linked to the co-activators in the regulation of transcription.[29]

Clinical significance edit

The ability of histone acetyltransferases to manipulate chromatin structure and lay an epigenetic framework makes them essential in cell maintenance and survival. The process of chromatin remodeling involves several enzymes, including HATs, that assist in the reformation of nucleosomes and are required for DNA damage repair systems to function.[30] HATs have been implicated as accessories to disease progression, specifically in neurodegenerative disorders. For instance, Huntington's disease is a disease that affects motor skills and mental abilities. The only known mutation that has been implicated in the disease is in the N-terminal region of the protein huntingtin (htt).[31] It has been reported that htt directly interacts with HATs and represses the catalytic activity of p300/CBP and PCAF in vitro.

The human premature aging syndrome Hutchinson Gilford progeria is caused by a mutational defect in the processing of lamin A, a nuclear matrix protein. In a mouse model of this condition, recruitment of repair proteins to sites of DNA damage is delayed. The molecular mechanism underlying this delayed repair response involves a histone acetylation defect.[32] Specifically, histone H4 is hypoacetylated at a lysine 16 residue (H4K16) and this defect is due to reduced association of histone acetyltransferase, Mof, to the nuclear matrix[32]

Spinocerebellar ataxia type 1 is a neurodegenerative disease that arises as a result of a defective mutant Ataxin-1 protein. Mutant Ataxin-1 reduces histone acetylation resulting in repressed histone acetyltransferase-mediated transcription.[33]

HATs have also been associated with control of learning and memory functions. Studies have shown that mice without PCAF or CBP display evidence of neurodegeneration.[31] Mice with PCAF deletion are incompetent with respect to learning, and those with CBP deletion seem to suffer from long-term memory loss.[34]

The misregulation of the equilibrium between acetylation and deacetylation has also been associated with the manifestation of certain cancers. If histone acetyltransferases are inhibited, then damaged DNA may not be repaired, eventually leading to cell death. Controlling the chromatin remodeling process within cancer cells may provide a novel drug target for cancer research.[35] Attacking these enzymes within cancer cells could lead to increased apoptosis due to high accumulation of DNA damage. One such inhibitor of histone acetyltransferases is called garcinol. This compound is found within the rinds of the garcinia indica fruit, otherwise known as mangosteen. To explore the effects of garcinol on histone acetyltransferases, researchers used HeLa cells. The cells underwent irradiation, creating double-strand breaks within the DNA, and garcinol was introduced into the cells to see if it influenced the DNA damage response. If garcinol is successful at inhibiting the process of non-homologous end joining, a DNA repair mechanism that shows preference in fixing double-strand breaks,[36] then it may serve as a radiosensitizer, a molecule that increases the sensitivity of cells to radiation damage. Increases in radiosensitivity may increase the effectiveness of radiotherapy.[35]

See also edit

References edit

  1. ^ Wang Y, Guo Y, Liu K, Yin Z, Liu R, Xia Y, Tan L, Yang P, Lee J, et al. (December 6, 2017). "KAT2A coupled with the α-KGDH complex acts as a histone H3 succinyltransferase". Nature. 552 (7684): 273–277. Bibcode:2017Natur.552..273W. doi:10.1038/nature25003. PMC 5841452. PMID 29211711.
  2. ^ a b c d e f g h i Lee KK, Workman JL (April 2007). "Histone acetyltransferase complexes: one size doesn't fit all". Nature Reviews. Molecular Cell Biology. 8 (4): 284–95. doi:10.1038/nrm2145. PMID 17380162. S2CID 1091590.
  3. ^ a b Weaver R (2007). Molecular Biology. McGraw-Hill. ISBN 978-0073319940.
  4. ^ a b c d e f g h i j k l m n o p q r s t u v w x Roth SY, Denu JM, Allis CD (2001). "Histone acetyltransferases". Annual Review of Biochemistry. 70: 81–120. doi:10.1146/annurev.biochem.70.1.81. PMID 11395403.
  5. ^ a b c d e f g h i j k l Sterner DE, Berger SL (June 2000). "Acetylation of histones and transcription-related factors". Microbiology and Molecular Biology Reviews. 64 (2): 435–59. doi:10.1128/MMBR.64.2.435-459.2000. PMC 98999. PMID 10839822.
  6. ^ Müller, Manuel M.; Muir, Tom W. (2014-10-20). "Histones: At the Crossroads of Peptide and Protein Chemistry". Chemical Reviews. 115 (6). American Chemical Society: 2296–2349. doi:10.1021/cr5003529. ISSN 0009-2665. PMC 4378460. PMID 25330018. S2CID 33098192. This review cites this research. Mittal, C; Blacketer, M.J; Shogren-Knaak, M.A (2014). "Nucleosome acetylation sequencing to study the establishment of chromatin acetylation". Analytical Biochemistry. 457 (457): 51–8. doi:10.1016/j.ab.2014.04.024. PMID 24769374.
  7. ^ a b c d e f g h Marmorstein R (August 2001). "Structure of histone acetyltransferases". Journal of Molecular Biology. 311 (3): 433–44. doi:10.1006/jmbi.2001.4859. PMID 11492997.
  8. ^ a b c d e f g h i j k l m n o p q r Yuan H, Marmorstein R (February 2013). "Histone acetyltransferases: Rising ancient counterparts to protein kinases". Biopolymers. 99 (2): 98–111. doi:10.1002/bip.22128. PMC 4017165. PMID 23175385.
  9. ^ a b c d e f Marmorstein R, Trievel RC (January 2009). "Histone modifying enzymes: structures, mechanisms, and specificities". Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 1789 (1): 58–68. doi:10.1016/j.bbagrm.2008.07.009. PMC 4059211. PMID 18722564.
  10. ^ a b Ogryzko VV (May 2001). "Mammalian histone acetyltransferases and their complexes". Cellular and Molecular Life Sciences. 58 (5–6): 683–92. doi:10.1007/PL00000892. PMID 11437230. S2CID 20905209.
  11. ^ Doi M, Hirayama J, Sassone-Corsi P (May 2006). "Circadian regulator CLOCK is a histone acetyltransferase". Cell. 125 (3): 497–508. doi:10.1016/j.cell.2006.03.033. PMID 16678094. S2CID 5968161.
  12. ^ a b c d e f Kimura A, Matsubara K, Horikoshi M (December 2005). "A decade of histone acetylation: marking eukaryotic chromosomes with specific codes". Journal of Biochemistry. 138 (6): 647–62. doi:10.1093/jb/mvi184. PMID 16428293.
  13. ^ Marmorstein R, Roth SY (April 2001). "Histone acetyltransferases: function, structure, and catalysis". Current Opinion in Genetics & Development. 11 (2): 155–61. doi:10.1016/S0959-437X(00)00173-8. PMID 11250138.
  14. ^ Anamika K, Krebs AR, Thompson J, Poch O, Devys D, Tora L (October 2010). "Lessons from genome-wide studies: an integrated definition of the coactivator function of histone acetyl transferases". Epigenetics & Chromatin. 3 (1): 18. doi:10.1186/1756-8935-3-18. PMC 2972259. PMID 20961410.
  15. ^ Carrozza MJ, Utley RT, Workman JL, Côté J (June 2003). "The diverse functions of histone acetyltransferase complexes". Trends in Genetics. 19 (6): 321–9. doi:10.1016/S0168-9525(03)00115-X. PMID 12801725.
  16. ^ Torok MS, Grant PA (2004). "Histone acetyltransferase proteins contribute to transcriptional processes at multiple levels". Proteins in Eukaryotic Transcription. Advances in Protein Chemistry. Vol. 67. Academic Press. pp. 181–99. doi:10.1016/S0065-3233(04)67007-0. ISBN 9780120342679. PMID 14969728.
  17. ^ Vernarecci S, Tosi F, Filetici P (February 2010). "Tuning acetylated chromatin with HAT inhibitors: a novel tool for therapy". Epigenetics. 5 (2): 105–11. doi:10.4161/epi.5.2.10942. PMID 20160510.
  18. ^ Berndsen CE, Albaugh BN, Tan S, Denu JM (January 2007). "Catalytic mechanism of a MYST family histone acetyltransferase". Biochemistry. 46 (3): 623–9. doi:10.1021/bi602513x. PMC 2752042. PMID 17223684.
  19. ^ a b c d e Berndsen CE, Denu JM (December 2008). "Catalysis and substrate selection by histone/protein lysine acetyltransferases". Current Opinion in Structural Biology. 18 (6): 682–9. doi:10.1016/j.sbi.2008.11.004. PMC 2723715. PMID 19056256.
  20. ^ a b Yang XJ (October 2004). "Lysine acetylation and the bromodomain: a new partnership for signaling". BioEssays. 26 (10): 1076–87. doi:10.1002/bies.20104. PMID 15382140. S2CID 36755688.
  21. ^ a b c Glozak MA, Sengupta N, Zhang X, Seto E (December 2005). "Acetylation and deacetylation of non-histone proteins". Gene. 363: 15–23. doi:10.1016/j.gene.2005.09.010. PMID 16289629.
  22. ^ Hata S, Hirayama J, Kajiho H, Nakagawa K, Hata Y, Katada T, Furutani-Seiki M, Nishina H (June 2012). "A novel acetylation cycle of transcription co-activator Yes-associated protein that is downstream of Hippo pathway is triggered in response to SN2 alkylating agents". The Journal of Biological Chemistry. 287 (26): 22089–98. doi:10.1074/jbc.M111.334714. PMC 3381167. PMID 22544757.
  23. ^ Liu X, Wang L, Zhao K, Thompson PR, Hwang Y, Marmorstein R, Cole PA (February 2008). "The structural basis of protein acetylation by the p300/CBP transcriptional coactivator". Nature. 451 (7180): 846–50. Bibcode:2008Natur.451..846L. doi:10.1038/nature06546. PMID 18273021. S2CID 4426988.
  24. ^ Albaugh BN, Arnold KM, Lee S, Denu JM (July 2011). "Autoacetylation of the histone acetyltransferase Rtt109". The Journal of Biological Chemistry. 286 (28): 24694–701. doi:10.1074/jbc.M111.251579. PMC 3137045. PMID 21606491.
  25. ^ a b c d e f g h i Voet D, Voet JG (2004). Biochemistry (3rd ed.). Hoboken, N.J.: John Wiley & Sons. ISBN 978-0-471-19350-0.
  26. ^ a b c Tropp BE (2008). Molecular biology : genes to proteins (3rd ed.). Sudbury, Mass.: Jones and Bartlett Publishers. ISBN 9780763709167.
  27. ^ Grunstein M (September 1997). "Histone acetylation in chromatin structure and transcription". Nature. 389 (6649): 349–52. Bibcode:1997Natur.389..349G. doi:10.1038/38664. PMID 9311776. S2CID 4419816.
  28. ^ Voichek Y, Bar-Ziv R, Barkai N (March 2016). "Expression homeostasis during DNA replication". Science. 351 (6277): 1087–90. Bibcode:2016Sci...351.1087V. doi:10.1126/science.aad1162. PMID 26941319. S2CID 32751800.
  29. ^ Dhalluin C, Carlson JE, Zeng L, He C, Aggarwal AK, Zhou MM (June 1999). "Structure and ligand of a histone acetyltransferase bromodomain". Nature. 399 (6735): 491–6. Bibcode:1999Natur.399..491D. doi:10.1038/20974. PMID 10365964. S2CID 1210925.
  30. ^ Rossetto D, Truman AW, Kron SJ, Côté J (September 2010). "Epigenetic modifications in double-strand break DNA damage signaling and repair". Clinical Cancer Research. 16 (18): 4543–52. doi:10.1158/1078-0432.CCR-10-0513. PMC 2940951. PMID 20823147.
  31. ^ a b Klein G, Vande Woude GF (2002). Advances in Cancer Research, Volume 86. Boston: Academic Press. ISBN 978-0-12-006686-5.
  32. ^ a b Krishnan V, Chow MZ, Wang Z, Zhang L, Liu B, Liu X, Zhou Z (July 2011). "Histone H4 lysine 16 hypoacetylation is associated with defective DNA repair and premature senescence in Zmpste24-deficient mice". Proc. Natl. Acad. Sci. U.S.A. 108 (30): 12325–30. Bibcode:2011PNAS..10812325K. doi:10.1073/pnas.1102789108. PMC 3145730. PMID 21746928.
  33. ^ Cvetanovic M, Kular RK, Opal P (December 2012). "LANP mediates neuritic pathology in Spinocerebellar ataxia type 1". Neurobiol. Dis. 48 (3): 526–32. doi:10.1016/j.nbd.2012.07.024. PMC 3987943. PMID 22884877.
  34. ^ Furdas SD, Kannan S, Sippl W, Jung M (January 2012). "Small molecule inhibitors of histone acetyltransferases as epigenetic tools and drug candidates". Archiv der Pharmazie. 345 (1): 7–21. doi:10.1002/ardp.201100209. PMID 22234972. S2CID 3125138.
  35. ^ a b Oike T, Ogiwara H, Torikai K, Nakano T, Yokota J, Kohno T (November 2012). "Garcinol, a histone acetyltransferase inhibitor, radiosensitizes cancer cells by inhibiting non-homologous end joining". International Journal of Radiation Oncology, Biology, Physics. 84 (3): 815–21. doi:10.1016/j.ijrobp.2012.01.017. PMID 22417805.
  36. ^ Burma S, Chen BP, Chen DJ (September 2006). "Role of non-homologous end joining (NHEJ) in maintaining genomic integrity". DNA Repair. 5 (9–10): 1042–8. doi:10.1016/j.dnarep.2006.05.026. PMID 16822724.

External links edit

histone, acetyltransferase, other, uses, disambiguation, hats, enzymes, that, acetylate, conserved, lysine, amino, acids, histone, proteins, transferring, acetyl, group, from, acetyl, form, acetyllysine, wrapped, around, histones, transferring, acetyl, group, . For other uses see Hat disambiguation Histone acetyltransferases HATs are enzymes that acetylate conserved lysine amino acids on histone proteins by transferring an acetyl group from acetyl CoA to form e N acetyllysine DNA is wrapped around histones and by transferring an acetyl group to the histones genes can be turned on and off In general histone acetylation increases gene expression GCN5 Histone acetyltransferaseGCN5 histone acetyltransferase domain homo24 mer Human Based on PDB structure 5trm 1 IdentifiersEC no 2 3 1 48CAS no 9054 51 7DatabasesIntEnzIntEnz viewBRENDABRENDA entryExPASyNiceZyme viewKEGGKEGG entryMetaCycmetabolic pathwayPRIAMprofilePDB structuresRCSB PDB PDBe PDBsumGene OntologyAmiGO QuickGOSearchPMCarticlesPubMedarticlesNCBIproteins In general histone acetylation is linked to transcriptional activation and associated with euchromatin Euchromatin which is less densely compact allows transcription factors to bind more easily to regulatory sites on DNA causing transcriptional activation When it was first discovered it was thought that acetylation of lysine neutralizes the positive charge normally present thus reducing affinity between histone and negatively charged DNA which renders DNA more accessible to transcription factors Research has emerged since to show that lysine acetylation and other posttranslational modifications of histones generate binding sites for specific protein protein interaction domains such as the acetyllysine binding bromodomain citation needed Histone acetyltransferases can also acetylate non histone proteins such as nuclear receptors and other transcription factors to facilitate gene expression Contents 1 HAT families 1 1 Gcn5 related N acetyltransferases GNATs 1 2 MYST HATs 1 3 Others 1 3 1 Nuclear receptor coactivators 2 Overall structure 2 1 GNAT and MYST families 2 2 p300 CBP family 2 3 Rtt109 3 Catalytic mechanisms 3 1 GNAT family 3 2 MYST family 3 3 p300 CBP family 3 4 Rtt109 4 Substrate binding and specificity 4 1 Lysine selectivity 4 1 1 GNAT family 4 1 2 MYST family 4 1 3 Others 4 2 Non histone substrates in vitro 4 2 1 PCAF 4 2 2 p300 CBP 4 3 Multisubunit HAT complexes 5 Regulation of HAT activity 6 Interaction with HDACs 7 Biological role 7 1 Chromatin remodeling 7 2 Gene expression 8 Clinical significance 9 See also 10 References 11 External linksHAT families editHATs are traditionally divided into two different classes based on their subcellular localization 2 Type A HATs are located in the nucleus and are involved in the regulation of gene expression through acetylation of nucleosomal histones in the context of chromatin 3 They contain a bromodomain which helps them recognize and bind to acetylated lysine residues on histone substrates Gcn5 p300 CBP and TAFII250 are some examples of type A HATs that cooperate with activators to enhance transcription Type B HATs are located in the cytoplasm and are responsible for acetylating newly synthesized histones prior to their assembly into nucleosomes These HATs lack a bromodomain as their targets are unacetylated The acetyl groups added by type B HATs to the histones are removed by HDACs once they enter the nucleus and are incorporated into chromatin Hat1 is one of the few known examples of a type B HAT 4 Despite this historical classification of HATs some HAT proteins function in multiple complexes or locations and would thus not easily fit into a particular class 5 nbsp Relative sizes and locations of important domains for representative HATs HAT catalytic acetyltransferase domain Bromo bromodomain Chromo chromodomain Zn zinc finger domain The number of amino acid residues in each HAT is indicated at the right in each example Gcn5 related N acetyltransferases GNATs edit HATs can be grouped into several different families based on sequence homology as well as shared structural features and functional roles The Gcn5 related N acetyltransferase GNAT family includes Gcn5 PCAF Hat1 Elp3 Hpa2 Hpa3 ATF 2 and Nut1 These HATs are generally characterized by the presence of a bromodomain and they are found to acetylate lysine residues on histones H2B H3 6 and H4 2 All members of the GNAT family are characterized by up to four conserved motifs A D found within the catalytic HAT domain This includes the most highly conserved motif A which contains an Arg Gln X X Gly X Gly Ala sequence that is important for acetyl CoA recognition and binding 4 The C motif is found in most GNATs but it is not present in the majority of other known HATs 5 The yeast Gcn5 general control nonderepressible 5 HAT is one of the best characterized members of this family It has four functional domains including an N terminal domain a highly conserved catalytic HAT domain an Ada2 interaction domain and a C terminal bromodomain PCAF p300 CBP associated factor and GCN5 are mammalian GNATs that share a high degree of homology throughout their sequences These proteins have a 400 residue N terminal region that is absent in yeast Gcn5 but their HAT functions are evolutionarily conserved with respect to the latter Hat1 was the first HAT protein to be identified It is responsible for most of the cytoplasmic HAT activity in yeast and it binds strongly to histone H4 by virtue of its association with an additional subunit Hat2 Elp3 is an example of a type A HAT found in yeast It is part of the RNA polymerase II holoenzyme and plays a role in transcriptional elongation MYST HATs edit The MYST family of HATs is named after its four founding members MOZ Ybf2 Sas3 Sas2 and Tip60 2 Other important members include Esa1 MOF MORF and HBO1 These HATs are typically characterized by the presence of zinc fingers and chromodomains and they are found to acetylate lysine residues on histones H2A H3 and H4 Several MYST family proteins contain zinc fingers as well as the highly conserved motif A found among GNATs that facilitates acetyl CoA binding 4 A cysteine rich region located in the N terminus of the HAT domain of MYST proteins is involved in zinc binding which is essential for HAT activity 7 Tip60 Tat interactive protein 60 kDa was the first human MYST family member to exhibit HAT activity Sas3 found in yeast is a homolog of MOZ monocytic leukemia zinc finger protein which is an oncogene found in humans Esa1 was the first essential HAT to be found in yeast and MOF is its homolog in fruit flies The HAT activity of the latter is required for the twofold increased transcription of the male X chromosome dosage compensation in flies Human HBO1 HAT bound to ORC1 was the first HAT shown to associate with components of the origin of replication complex MORF MOZ related factor exhibits very close homology to MOZ throughout its entire length 5 It contains an N terminal repression region that decreases its HAT activity in vitro as well as a C terminal activation domain that is functional in the absence of the HAT domain Others edit In addition to those that are members of the GNAT and MYST families there are several other proteins found typically in higher eukaryotes that exhibit HAT activity These include p300 CBP nuclear receptor coactivators e g ACTR SRC 1 TAFII250 TFIIIC Rtt109 and CLOCK p300 CBP are metazoan specific 8 and contain several zinc finger regions a bromodomain a catalytic HAT domain and regions that interact with other transcription factors 4 Importantly the HAT domain shows no sequence homology to other known HATs 9 and it is required for p300 CBP to function in transcriptional activation 4 In addition these proteins contain several HAT domain motifs A B and D that are similar to those of the GNATs They also possess a novel motif E that is homologous to sequences in the HAT domains of GNATs TFIIIC is one of the general transcription factors involved in RNA polymerase III mediated transcription Three components in the human protein have been shown to possess independent HAT activity hTFIIIC220 hTFIIIC110 and hTFIIIC90 10 Rtt109 is a fungal specific HAT that requires association with histone chaperone proteins for activity 8 The HAT activities of the human TAFII250 and CLOCK coactivators have not been studied as extensively TAFII250 is one of the TBP associated factor subunits of TFIID and it shares a Gly X Gly pattern with Gcn5 that is important for HAT activity 5 CLOCK is a circadian rhythm master regulator that functions with BMAL1 to carry out its HAT activity 11 Nuclear receptor coactivators edit Three important nuclear receptor coactivators that display HAT activity are SRC 1 ACTR and TIF 2 Human SRC 1 steroid receptor coactivator 1 is known to interact with p300 CBP and PCAF and its HAT domain is located in its C terminal region ACTR also known as RAC3 AIB1 and TRAM 1 in humans shares significant sequence homology with SRC 1 in particular in the N terminal and C terminal HAT regions as well as in the receptor and coactivator interaction domains 5 ACTR also interacts with p300 CBP and PCAF The former can prevent ACTR from binding to and activating its receptor by acetylating it in its receptor interaction domain TIF 2 transcriptional intermediary factor 2 also known as GRIP1 is another nuclear receptor coactivator with HAT activity and it also interacts with p300 CBP A table summarizing the different families of HATs along with their associated members parent organisms multisubunit complexes histone substrates and structural features is presented below 2 5 8 10 12 13 14 15 16 17 Family Members Organism Associated complexes Substrate specificity Structural features GNAT Gcn5 S cerevisiae SAGA SLIK SALSA ADA HAT A2 H2B H3 H4 Bromodomain GCN5 D melanogaster SAGA ATAC H3 H4 Bromodomain GCN5 H sapiens STAGA TFTC H3 H4 H2B Bromodomain PCAF H sapiens PCAF H3 H4 Bromodomain Hat1 S cerevisiaeH sapiens HAT B NuB4 HAT A3 H4 H2A Elp3 S cerevisiae Elongator H3 H4 H2A H2B Hpa2 S cerevisiae HAT B H3 H4 Hpa3 S cerevisiae H3 H4 ATF 2 S cerevisiaeH sapiens H2B H4 Nut1 S cerevisiae Mediator H3 H4 MYST Esa1 S cerevisiae NuA4 piccolo NuA4 H2A H4 H2B H3 Chromodomain Sas2 S cerevisiae SAS NuA4 H4 H2A H3 Sas3 Ybf2 S cerevisiae NuA3 H3 H4 H2A Tip60 H sapiens Tip60 NuA4 H2A H4 H3 Chromodomain MOF D melanogaster MSL H4 H2A H3 Chromodomain MOZ H sapiens MSL H3 H4 MORF H sapiens MSL H3 H4 HBO1 H sapiens ORC H3 H4 p300 CBP p300 H sapiens H2A H2B H3 H4 Bromodomain CBP H sapiens H2A H2B H3 H4 Bromodomain SRC nuclear receptor coactivators SRC 1 H sapiens ACTR SRC 1 H3 H4 ACTR RAC3 AIB1 TRAM 1 SRC 3 H sapiens ACTR SRC 1 H3 H4 TIF 2 GRIP1 H sapiens H3 H4 Other TAFII250 TAF1 S cerevisiaeH sapiens TFIID H3 H4 H2A Bromodomain TFIIIC p220 p110 p90 H sapiens TFIIIC H2A H3 H4 Rtt109 S cerevisiae Histone chaperones H3 CLOCK H sapiens H3 H4Overall structure edit nbsp Crystal structure of Tetrahymena Gcn5 with bound coenzyme A and histone H3 peptide PDB 1QSN The central core green flanking N and C terminal segments blue coenzyme A orange and histone peptide red are shown In general HATs are characterized by a structurally conserved core region made up of a three stranded b sheet followed by a long a helix parallel to and spanning one side of it 7 8 The core region which corresponds to motifs A B and D of the GNAT proteins 4 is flanked on opposite sides by N and C terminal a b segments that are structurally unique for a given HAT family 7 8 The central core and the flanking segments together form a cleft over the former which is where histone substrates can bind prior to catalysis 8 While the central core domain motif A in GNATs is involved in acetyl CoA binding and catalysis the N and C terminal segments assist in binding histone substrates 7 Unique features related to the sequence and or structure of the N and C terminal regions for different HAT families may help to explain some observed differences among HATs in histone substrate specificity CoA binding has been observed to widen the histone binding groove in the central core by moving the C terminal segment of Gcn5 outward In addition since contacts between CoA and protein facilitate the formation of favorable histone protein contacts it is likely that CoA binding precedes histone binding in vivo GNAT and MYST families edit HATs in the GNAT family are most notably characterized by an approximately 160 residue HAT domain and a C terminal bromodomain which binds to acetylated lysine residues 7 Those in the MYST family have HAT domains that are about 250 residues in length Many MYST proteins also contain a cysteine rich zinc binding domain within the HAT region in addition to an N terminal chromodomain which binds to methylated lysine residues On a broader scale the structures of the catalytic domains of GNAT proteins Gcn5 PCAF exhibit a mixed a b globular fold with a total of five a helices and six b strands 4 The overall topology resembles a vise with the central core of the protein at the base and the N and C terminal segments on the sides p300 CBP family edit The p300 CBP HATs have larger HAT domains about 500 residues than those present in the GNAT and MYST families 7 They also contain a bromodomain as well as three cysteine histidine rich domains that are thought to mediate interactions with other proteins The structure of p300 CBP is characterized by an elongated globular domain which contains a seven stranded b sheet in the center that is surrounded by nine a helices and several loops 9 The structure of the central core region associated with acetyl CoA binding is conserved with respect to GNAT and MYST HATs but there are many structural differences in the regions flanking this central core Overall the structural data is consistent with the fact that p300 CBP HATs are more promiscuous than GNAT and MYST HATs with respect to substrate binding Rtt109 edit The structure of Rtt109 is very similar to that of p300 despite there only being 7 sequence identity between the two proteins 9 There is a seven stranded b sheet that is surrounded by a helices as well as a loop that is involved in acetyl CoA substrate binding Despite the conserved structure Rtt109 and p300 CBP are functionally unique For instance the substrate binding site of the former is more similar to that of the GNAT and MYST HATs In addition the residues in the active site of each enzyme are distinct which suggests that they employ different catalytic mechanisms for acetyl group transfer Catalytic mechanisms editThe basic mechanism catalyzed by HATs involves the transfer of an acetyl group from acetyl CoA to the e amino group of a target lysine side chain within a histone 8 Different families of HATs employ unique strategies in order to effect such a transformation nbsp Catalytic mechanisms of GNAT and MYST family HATs A General mechanism of GNAT HATs B General mechanism of MYST HATs GNAT family edit Members of the GNAT family have a conserved glutamate residue that acts as a general base for catalyzing the nucleophilic attack of the lysine amine on the acetyl CoA thioester bond 8 These HATs use an ordered sequential bi bi mechanism wherein both substrates acetyl CoA and histone must bind to form a ternary complex with the enzyme before catalysis can occur Acetyl CoA binds first followed by the histone substrate A conserved glutamate residue Glu173 in yeast Gcn5 activates a water molecule for removal of a proton from the amine group on lysine which activates it for direct nucleophilic attack on the carbonyl carbon of enzyme bound acetyl CoA After the reaction the acetylated histone is released first followed by CoA 4 8 MYST family edit Studies of yeast Esa1 from the MYST family of HATs have revealed a ping pong mechanism involving conserved glutamate and cysteine residues 18 The first part of the reaction involves the formation of a covalent intermediate in which a cysteine residue becomes acetylated following nucleophilic attack of this residue on the carbonyl carbon of acetyl CoA Then a glutamate residue acts as a general base to facilitate transfer of the acetyl group from the cysteine to the histone substrate in a manner analogous to the mechanism used by GNATs When Esa1 is assembled in the piccolo NuA4 complex it loses its dependence on the cysteine residue for catalysis which suggests that the reaction may proceed via a ternary bi bi mechanism when the enzyme is part of a physiologically relevant multiprotein complex p300 CBP family edit In human p300 Tyr1467 acts as a general acid and Trp1436 helps orient the target lysine residue of the histone substrate into the active site 8 These two residues are highly conserved within the p300 CBP HAT family and unlike enzymes in the GNAT and MYST families p300 does not employ a general base for catalysis Rather it is likely that members of the p300 CBP family use a Theorell Chance i e hit and run acetyl transfer mechanism Rtt109 edit Rtt109 is likely to employ a mechanism that is different from that of the other HATs 9 The yeast enzyme has very low catalytic activity in the absence of the histone chaperone proteins Asf1 and Vps75 which may be involved in delivering histone substrates to the enzyme for acetylation 8 Moreover a general acid or base have not yet been identified for this HAT Substrate binding and specificity editThe structures of several HAT domains bound to acetyl CoA and histone substrate peptides reveal that the latter bind across a groove on the protein that is formed by the central core region at the base and is flanked on opposite sides by the variable N and C terminal segments that mediate the majority of the interactions with the substrate peptide 8 It is likely that these variable regions are at least in part responsible for the observed specificity of different HATs for various histone substrates Members of the GNAT and MYST families as well as Rtt109 exhibit greater substrate selectivity than p300 CBP which is rather promiscuous with regard to substrate binding 8 Whereas it appears that only three to five residues on either side of the lysine to be acetylated are necessary for effective substrate binding and catalysis by members of the GNAT and p300 CBP families more distal regions of the substrate may be important for efficient acetylation by MYST family HATs 19 Lysine selectivity edit Different HATs usually in the context of multisubunit complexes have been shown to acetylate specific lysine residues in histones GNAT family edit Gcn5 cannot acetylate nucleosomal histones in the absence of other protein factors 5 In the context of complexes like SAGA and ADA however Gcn5 is able to acetylate H3K14 among other sites within histones H2B H3 and H4 e g H3K9 H3K36 H4K8 H4K16 3 4 7 19 Both Gcn5 and PCAF have the strongest site preference for H3K14 either as a free histone or within a nucleosome 4 7 Hat1 acetylates H4K5 and H4K12 and Hpa2 acetylates H3K14 in vitro 4 5 MYST family edit In flies acetylation of H4K16 on the male X chromosome by MOF in the context of the MSL complex is correlated with transcriptional upregulation as a mechanism for dosage compensation in these organisms 2 In humans the MSL complex carries out the majority of genome wide H4K16 acetylation In the context of their cognate complexes Sas2 SAS and Esa1 NuA4 also carry out acetylation of H4K16 in particular in the telomere regions of chromosomes Sas2 is also observed to acetylate H3K14 in vitro on free histones 12 Esa1 can also acetylate H3K14 in vitro on free histones as well as H2AK5 H4K5 H4K8 and H4K12 either in vitro or in vivo on nucleosomal histones H2AK7 and H2BK16 are also observed to be acetylated by Esa1 in vivo Notably neither Sas2 nor Esa1 can acetylate nucleosomal histones in vitro as a free enzyme This happens to be the case as well for Sas3 which is observed to acetylate H3K9 and H3K14 in vivo as well as lysine residues on H2A and H4 MOZ can also acetylate H3K14 19 Others edit p300 CBP acetylate all four nucleosomal core histones equally well 4 In vitro they have been observed to acetylate H2AK5 H2BK12 H2BK15 H3K14 H3K18 H4K5 and H4K8 5 SRC 1 acetylates H3K9 and H3K14 TAFII230 Drosophila homolog of human TAFII250 acetylates H3K14 and Rtt109 acetylates H3K9 H3K23 19 and H3K56 in the presence of either Asf1 or Vps75 9 Non histone substrates in vitro edit In addition to the core histones certain HATs acetylate a number of other cellular proteins including transcriptional activators basal transcription factors structural proteins polyamines and proteins involved in nuclear import 4 Acetylation of these proteins can alter their ability to interact with their cognate DNA and or protein substrates The idea that acetylation can affect protein function in this manner has led to inquiry regarding the role of acetyltransferases in signal transduction pathways and whether an appropriate analogy to kinases and phosphorylation events can be made in this respect PCAF edit PCAF and p300 CBP are the main HATs that have been observed to acetylate a number of non histone proteins For PCAF these include the non histone chromatin high mobility group HMG proteins HMG N2 HMG17 and HMG I Y the transcriptional activators p53 MyoD E2F 1 3 and HIV Tat and the general transcription factors TFIIE and TFIIF 5 Other proteins include CIITA Brm chromatin remodeler NF kB p65 TAL1 SCL Beta2 NeuroD C EBPb IRF2 IRF7 YY1 KLF13 EVI1 AME ER81 and the androgen receptor AR 20 PCAF has also been observed to acetylate c MYC GATA 2 retinoblastoma Rb Ku70 and E1A adenovirus protein 21 It can also autoacetylate which facilitates intramolecular interactions with its bromodomain that may be involved in the regulation of its HAT activity 4 p300 CBP edit p300 CBP have many non histone substrates including the non histone chromatin proteins HMG1 HMG N1 HMG14 and HMG I Y the transcriptional activators p53 c Myb GATA 1 EKLF TCF and HIV Tat the nuclear receptor coactivators ACTR SRC 1 and TIF 2 and the general transcription factors TFIIE and TFIIF 5 Other substrates include the transcription factors Sp1 KLF5 FOXO1 MEF2C SRY GATA 4 and HNF 6 12 HMG B2 21 STAT3 the androgen and estrogen a receptors GATA 2 GATA 3 MyoD E2F 1 3 p73a retinoblastoma Rb NF kB p50 p65 Smad7 importin a Ku70 YAP1 22 E1A adenovirus protein and S HDAg hepatitis delta virus small delta antigen 21 p300 CBP have also been observed to acetylate b catenin RIP140 PCNA the DNA metabolic enzymes flap endonuclease 1 thymine DNA glycosylase and Werner syndrome DNA helicase STAT6 Runx1 AML1 UBF Beta2 NeuroD CREB c Jun C EBPb NF E2 SREBP IRF2 Sp3 YY1 KLF13 EVI1 BCL6 HNF 4 ER81 and FOXO4 AFX 20 Multisubunit HAT complexes edit The formation of multisubunit complexes has been observed to modulate the substrate specificity of HATs 12 In general while recombinant HATs are able to acetylate free histones HATs can acetylate nucleosomal histones only when they are in their respective in vivo HAT complexes 5 Some of the proteins that associate with HATs in these complexes function by targeting the HAT complex to nucleosomes at specific regions in the genome 2 12 For instance it has been observed that HAT complexes e g SAGA NuA3 often use methylated histones as docking sites so that the catalytic HAT subunit can carry out histone acetylation more effectively 2 In addition the formation of multisubunit HAT complexes influences the lysine specificity of HATs 12 The specific lysine residues that a given HAT acetylates may become either broader or more restricted in scope upon association with its respective complex For example the lysine specificity of MYST family HATs toward their histone substrates becomes more restricted when they associate with their complexes In contrast Gcn5 acquires the ability to acetylate multiple sites in both histones H2B and H3 when it joins other subunits to form the SAGA and ADA complexes 4 Moreover the acetylation site specificity of Rtt109 is dictated by its association with either Vps75 or Asf1 19 When in complex with the former Rtt109 acetylates H3K9 and H3K27 but when in complex with the latter it preferentially acetylates H3K56 8 Regulation of HAT activity editThe catalytic activity of HATs is regulated by two types of mechanisms 1 interaction with regulatory protein subunits and 2 autoacetylation 8 A given HAT may be regulated in multiple ways and the same effector may actually lead to different outcomes under different conditions 4 Although it is clear that the association of HATs with multiprotein complexes provides a mechanism for the regulation of both HAT activity and substrate specificity in vivo the molecular basis for how this actually occurs is still largely unknown 8 However data suggests that associated subunits may contribute to catalysis at least in part by facilitating productive binding of the HAT complex to its native histone substrates The MYST family of HATs p300 CBP and Rtt109 have all been shown to be regulated by autoacetylation 8 Human MOF as well as yeast Esa1 and Sas2 are autoacetylated at a conserved active site lysine residue and this modification is required for their function in vivo Human p300 contains a highly basic loop embedded in the middle of its HAT domain that is hyperacetylated in the active form of the enzyme 8 9 It has been proposed that upon autoacetylation this loop is released from the electronegative substrate binding site where it sits in the inactive HAT 23 Acetylation of yeast Rtt109 at Lys290 is also required for it to exhibit full catalytic activity 24 Some HATs are also inhibited by acetylation For example the HAT activity of the nuclear receptor coactivator ACTR is inhibited upon acetylation by p300 CBP 4 Interaction with HDACs editHistone acetyltransferases HATs and histone deacetylases HDACs are recruited to their target promoters through physical interactions with sequence specific transcription factors They usually function within a multisubunit complex in which the other subunits are necessary for them to modify histone residues around the binding site These enzymes can also modify non histone proteins Biological role editChromatin remodeling edit nbsp Histone tails and their function in chromatin formation Histone acetyltransferases serve many biological roles inside the cell Chromatin is a combination of proteins and DNA found in the nucleus and it undergoes many structural changes as different cellular events such as DNA replication DNA repair and transcription occur 25 Chromatin in the cell can be found in two states condensed and uncondensed The latter known as euchromatin is transcriptionally active whereas the former known as heterochromatin is transcriptionally inactive 25 26 Histones comprise the protein portion of chromatin There are five different histone proteins H1 H2A H2B H3 and H4 A core histone is formed when two of each histone subtype excluding H1 form a quaternary complex This octameric complex in association with the 147 base pairs of DNA coiled around it forms the nucleosome 4 Histone H1 locks the nucleosome complex together and it is the last protein to bind in the complex Histones tend to be positively charged proteins with N terminal tails that stem from the core The phosphodiester backbone of DNA is negative which allows for strong ionic interactions between histone proteins and DNA Histone acetyltransferases transfer an acetyl group to specific lysine residues on histones which neutralizes their positive charge and thus reduces the strong interactions between the histone and DNA 25 Acetylation is also thought to perturb interactions between individual nucleosomes and act as interaction sites for other DNA associated proteins 4 There can be different levels of histone acetylation as well as other types of modifications allowing the cell to have control over the level of chromatin packing during different cellular events such as replication transcription recombination and repair Acetylation is not the only regulatory post translational modification to histones that dictates chromatin structure methylation phosphorylation ADP ribosylation and ubiquitination have also been reported 4 25 These combinations of different covalent modifications on the N terminal tails of histones have been referred to as the histone code and it is thought that this code may be heritable and preserved in the next cell generation 26 H3 and H4 histone proteins are the primary targets of HATs but H2A and H2B are also acetylated in vivo Lysines 9 14 18 and 23 of H3 and lysines 5 8 12 and 16 of H4 are all targeted for acetylation 4 25 Lysines 5 12 15 and 20 are acetylated on histone H2B while only lysines 5 and 9 have been observed to be acetylated on histone H2A 4 25 26 With so many different sites for acetylation a high level of specificity can be achieved in triggering specific responses An example of this specificity is when histone H4 is acetylated at lysines 5 and 12 This acetylation pattern has been seen during histone synthesis Another example is acetylation of H4K16 which has been associated with dosage compensation of the male X chromosome in Drosophila melanogaster 2 4 Gene expression edit nbsp Schematic showing the role of HATs in gene transcription Histone modifications modulate the packing of chromatin The level of packing of the DNA is important for gene transcription since the transcriptional machinery must have access to the promoter in order for transcription to occur 4 Neutralization of charged lysine residues by HATs allows for the chromatin to decondense so that this machinery has access to the gene to be transcribed However acetylation is not always associated with enhanced transcriptional activity For instance acetylation of H4K12 has been associated with condensed and transcriptionally inactive chromatin 27 In addition some histone modifications are associated with both enhanced and repressed activity in a context dependent manner 28 HATs act as transcriptional co activators or gene silencers and are most often found in large complexes made up of 10 to 20 subunits some of which shared among different HAT complexes 25 These complexes include SAGA Spt Ada Gcn5L acetyltransferase PCAF ADA transcriptional adaptor TFIID transcription factor II D TFTC TBP free TAF containing complex and NuA3 NuA4 nucleosomal acetyltransferases of H3 and H4 2 25 These complexes modulate HAT specificity by bringing HATs to their target genes where they can then acetylate nucleosomal histones 25 Some HAT transcriptional co activators contain a bromodomain a 110 amino acid module that recognizes acetylated lysine residues and is functionally linked to the co activators in the regulation of transcription 29 Clinical significance editThe ability of histone acetyltransferases to manipulate chromatin structure and lay an epigenetic framework makes them essential in cell maintenance and survival The process of chromatin remodeling involves several enzymes including HATs that assist in the reformation of nucleosomes and are required for DNA damage repair systems to function 30 HATs have been implicated as accessories to disease progression specifically in neurodegenerative disorders For instance Huntington s disease is a disease that affects motor skills and mental abilities The only known mutation that has been implicated in the disease is in the N terminal region of the protein huntingtin htt 31 It has been reported that htt directly interacts with HATs and represses the catalytic activity of p300 CBP and PCAF in vitro The human premature aging syndrome Hutchinson Gilford progeria is caused by a mutational defect in the processing of lamin A a nuclear matrix protein In a mouse model of this condition recruitment of repair proteins to sites of DNA damage is delayed The molecular mechanism underlying this delayed repair response involves a histone acetylation defect 32 Specifically histone H4 is hypoacetylated at a lysine 16 residue H4K16 and this defect is due to reduced association of histone acetyltransferase Mof to the nuclear matrix 32 Spinocerebellar ataxia type 1 is a neurodegenerative disease that arises as a result of a defective mutant Ataxin 1 protein Mutant Ataxin 1 reduces histone acetylation resulting in repressed histone acetyltransferase mediated transcription 33 HATs have also been associated with control of learning and memory functions Studies have shown that mice without PCAF or CBP display evidence of neurodegeneration 31 Mice with PCAF deletion are incompetent with respect to learning and those with CBP deletion seem to suffer from long term memory loss 34 The misregulation of the equilibrium between acetylation and deacetylation has also been associated with the manifestation of certain cancers If histone acetyltransferases are inhibited then damaged DNA may not be repaired eventually leading to cell death Controlling the chromatin remodeling process within cancer cells may provide a novel drug target for cancer research 35 Attacking these enzymes within cancer cells could lead to increased apoptosis due to high accumulation of DNA damage One such inhibitor of histone acetyltransferases is called garcinol This compound is found within the rinds of the garcinia indica fruit otherwise known as mangosteen To explore the effects of garcinol on histone acetyltransferases researchers used HeLa cells The cells underwent irradiation creating double strand breaks within the DNA and garcinol was introduced into the cells to see if it influenced the DNA damage response If garcinol is successful at inhibiting the process of non homologous end joining a DNA repair mechanism that shows preference in fixing double strand breaks 36 then it may serve as a radiosensitizer a molecule that increases the sensitivity of cells to radiation damage Increases in radiosensitivity may increase the effectiveness of radiotherapy 35 See also editHistone modifying enzymes Histone deacetylase HDAC Histone methyltransferase HMT RNA polymerase control by chromatin structure AcetyltransferaseReferences edit Wang Y Guo Y Liu K Yin Z Liu R Xia Y Tan L Yang P Lee J et al December 6 2017 KAT2A coupled with the a KGDH complex acts as a histone H3 succinyltransferase Nature 552 7684 273 277 Bibcode 2017Natur 552 273W doi 10 1038 nature25003 PMC 5841452 PMID 29211711 a b c d e f g h i Lee KK Workman JL April 2007 Histone acetyltransferase complexes one size doesn t fit all Nature Reviews Molecular Cell Biology 8 4 284 95 doi 10 1038 nrm2145 PMID 17380162 S2CID 1091590 a b Weaver R 2007 Molecular Biology McGraw Hill ISBN 978 0073319940 a b c d e f g h i j k l m n o p q r s t u v w x Roth SY Denu JM Allis CD 2001 Histone acetyltransferases Annual Review of Biochemistry 70 81 120 doi 10 1146 annurev biochem 70 1 81 PMID 11395403 a b c d e f g h i j k l Sterner DE Berger SL June 2000 Acetylation of histones and transcription related factors Microbiology and Molecular Biology Reviews 64 2 435 59 doi 10 1128 MMBR 64 2 435 459 2000 PMC 98999 PMID 10839822 Muller Manuel M Muir Tom W 2014 10 20 Histones At the Crossroads of Peptide and Protein Chemistry Chemical Reviews 115 6 American Chemical Society 2296 2349 doi 10 1021 cr5003529 ISSN 0009 2665 PMC 4378460 PMID 25330018 S2CID 33098192 This review cites this research Mittal C Blacketer M J Shogren Knaak M A 2014 Nucleosome acetylation sequencing to study the establishment of chromatin acetylation Analytical Biochemistry 457 457 51 8 doi 10 1016 j ab 2014 04 024 PMID 24769374 a b c d e f g h Marmorstein R August 2001 Structure of histone acetyltransferases Journal of Molecular Biology 311 3 433 44 doi 10 1006 jmbi 2001 4859 PMID 11492997 a b c d e f g h i j k l m n o p q r Yuan H Marmorstein R February 2013 Histone acetyltransferases Rising ancient counterparts to protein kinases Biopolymers 99 2 98 111 doi 10 1002 bip 22128 PMC 4017165 PMID 23175385 a b c d e f Marmorstein R Trievel RC January 2009 Histone modifying enzymes structures mechanisms and specificities Biochimica et Biophysica Acta BBA Gene Regulatory Mechanisms 1789 1 58 68 doi 10 1016 j bbagrm 2008 07 009 PMC 4059211 PMID 18722564 a b Ogryzko VV May 2001 Mammalian histone acetyltransferases and their complexes Cellular and Molecular Life Sciences 58 5 6 683 92 doi 10 1007 PL00000892 PMID 11437230 S2CID 20905209 Doi M Hirayama J Sassone Corsi P May 2006 Circadian regulator CLOCK is a histone acetyltransferase Cell 125 3 497 508 doi 10 1016 j cell 2006 03 033 PMID 16678094 S2CID 5968161 a b c d e f Kimura A Matsubara K Horikoshi M December 2005 A decade of histone acetylation marking eukaryotic chromosomes with specific codes Journal of Biochemistry 138 6 647 62 doi 10 1093 jb mvi184 PMID 16428293 Marmorstein R Roth SY April 2001 Histone acetyltransferases function structure and catalysis Current Opinion in Genetics amp Development 11 2 155 61 doi 10 1016 S0959 437X 00 00173 8 PMID 11250138 Anamika K Krebs AR Thompson J Poch O Devys D Tora L October 2010 Lessons from genome wide studies an integrated definition of the coactivator function of histone acetyl transferases Epigenetics amp Chromatin 3 1 18 doi 10 1186 1756 8935 3 18 PMC 2972259 PMID 20961410 Carrozza MJ Utley RT Workman JL Cote J June 2003 The diverse functions of histone acetyltransferase complexes Trends in Genetics 19 6 321 9 doi 10 1016 S0168 9525 03 00115 X PMID 12801725 Torok MS Grant PA 2004 Histone acetyltransferase proteins contribute to transcriptional processes at multiple levels Proteins in Eukaryotic Transcription Advances in Protein Chemistry Vol 67 Academic Press pp 181 99 doi 10 1016 S0065 3233 04 67007 0 ISBN 9780120342679 PMID 14969728 Vernarecci S Tosi F Filetici P February 2010 Tuning acetylated chromatin with HAT inhibitors a novel tool for therapy Epigenetics 5 2 105 11 doi 10 4161 epi 5 2 10942 PMID 20160510 Berndsen CE Albaugh BN Tan S Denu JM January 2007 Catalytic mechanism of a MYST family histone acetyltransferase Biochemistry 46 3 623 9 doi 10 1021 bi602513x PMC 2752042 PMID 17223684 a b c d e Berndsen CE Denu JM December 2008 Catalysis and substrate selection by histone protein lysine acetyltransferases Current Opinion in Structural Biology 18 6 682 9 doi 10 1016 j sbi 2008 11 004 PMC 2723715 PMID 19056256 a b Yang XJ October 2004 Lysine acetylation and the bromodomain a new partnership for signaling BioEssays 26 10 1076 87 doi 10 1002 bies 20104 PMID 15382140 S2CID 36755688 a b c Glozak MA Sengupta N Zhang X Seto E December 2005 Acetylation and deacetylation of non histone proteins Gene 363 15 23 doi 10 1016 j gene 2005 09 010 PMID 16289629 Hata S Hirayama J Kajiho H Nakagawa K Hata Y Katada T Furutani Seiki M Nishina H June 2012 A novel acetylation cycle of transcription co activator Yes associated protein that is downstream of Hippo pathway is triggered in response to SN2 alkylating agents The Journal of Biological Chemistry 287 26 22089 98 doi 10 1074 jbc M111 334714 PMC 3381167 PMID 22544757 Liu X Wang L Zhao K Thompson PR Hwang Y Marmorstein R Cole PA February 2008 The structural basis of protein acetylation by the p300 CBP transcriptional coactivator Nature 451 7180 846 50 Bibcode 2008Natur 451 846L doi 10 1038 nature06546 PMID 18273021 S2CID 4426988 Albaugh BN Arnold KM Lee S Denu JM July 2011 Autoacetylation of the histone acetyltransferase Rtt109 The Journal of Biological Chemistry 286 28 24694 701 doi 10 1074 jbc M111 251579 PMC 3137045 PMID 21606491 a b c d e f g h i Voet D Voet JG 2004 Biochemistry 3rd ed Hoboken N J John Wiley amp Sons ISBN 978 0 471 19350 0 a b c Tropp BE 2008 Molecular biology genes to proteins 3rd ed Sudbury Mass Jones and Bartlett Publishers ISBN 9780763709167 Grunstein M September 1997 Histone acetylation in chromatin structure and transcription Nature 389 6649 349 52 Bibcode 1997Natur 389 349G doi 10 1038 38664 PMID 9311776 S2CID 4419816 Voichek Y Bar Ziv R Barkai N March 2016 Expression homeostasis during DNA replication Science 351 6277 1087 90 Bibcode 2016Sci 351 1087V doi 10 1126 science aad1162 PMID 26941319 S2CID 32751800 Dhalluin C Carlson JE Zeng L He C Aggarwal AK Zhou MM June 1999 Structure and ligand of a histone acetyltransferase bromodomain Nature 399 6735 491 6 Bibcode 1999Natur 399 491D doi 10 1038 20974 PMID 10365964 S2CID 1210925 Rossetto D Truman AW Kron SJ Cote J September 2010 Epigenetic modifications in double strand break DNA damage signaling and repair Clinical Cancer Research 16 18 4543 52 doi 10 1158 1078 0432 CCR 10 0513 PMC 2940951 PMID 20823147 a b Klein G Vande Woude GF 2002 Advances in Cancer Research Volume 86 Boston Academic Press ISBN 978 0 12 006686 5 a b Krishnan V Chow MZ Wang Z Zhang L Liu B Liu X Zhou Z July 2011 Histone H4 lysine 16 hypoacetylation is associated with defective DNA repair and premature senescence in Zmpste24 deficient mice Proc Natl Acad Sci U S A 108 30 12325 30 Bibcode 2011PNAS 10812325K doi 10 1073 pnas 1102789108 PMC 3145730 PMID 21746928 Cvetanovic M Kular RK Opal P December 2012 LANP mediates neuritic pathology in Spinocerebellar ataxia type 1 Neurobiol Dis 48 3 526 32 doi 10 1016 j nbd 2012 07 024 PMC 3987943 PMID 22884877 Furdas SD Kannan S Sippl W Jung M January 2012 Small molecule inhibitors of histone acetyltransferases as epigenetic tools and drug candidates Archiv der Pharmazie 345 1 7 21 doi 10 1002 ardp 201100209 PMID 22234972 S2CID 3125138 a b Oike T Ogiwara H Torikai K Nakano T Yokota J Kohno T November 2012 Garcinol a histone acetyltransferase inhibitor radiosensitizes cancer cells by inhibiting non homologous end joining International Journal of Radiation Oncology Biology Physics 84 3 815 21 doi 10 1016 j ijrobp 2012 01 017 PMID 22417805 Burma S Chen BP Chen DJ September 2006 Role of non homologous end joining NHEJ in maintaining genomic integrity DNA Repair 5 9 10 1042 8 doi 10 1016 j dnarep 2006 05 026 PMID 16822724 External links editHistone Acetyltransferases at the U S National Library of Medicine Medical Subject Headings MeSH Portal nbsp Biology Retrieved from https en wikipedia org w index php title Histone acetyltransferase amp oldid 1188072393, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.