Abstract

Post-translational modification of histones, a major protein component of eukaryotic chromosomes, contributes to the epigenetic regulation of gene expression. Distinct patterns of histone modification are observed at specific chromosomal regions and affect various reactions on chromosomes (transcription, replication, repair, and recombination). Histone modification has long been proposed to have a profound effect on eukaryotic gene expression since its discovery in 1964. Verification of this idea, however, was difficult until the identification of enzymes responsible for histone modifications. Ten years ago (1995), histone acetyltransferases (HATs), which acetylate lysine residues in histone amino-terminal tail regions, were isolated. HATs are involved in the regulation of both promoter-specific transcription and long-range/chromosome-wide transcription. Analyses of HATs and other modification enzymes have revealed mechanisms of epigenetic regulation that are mediated by post-translational modifications of histones. Here we review some major advances in the field, with emphasis on the lysine specificity of the acetylation reaction and on the regulation of gene expression over broad regions.

1. Introduction: Before 1995

The nucleosome, a fundamental unit of eukaryotic chromosomes, is composed of DNA and histone proteins (Fig. 1) (1, 2). Histones have a mass roughly equal to that of the DNA which they are associated with. Each of the core histones (H2A, H2B, H3, and H4) exhibits a similar structural feature called the “histone fold,” which consists of a long central α-helix flanked by shorter helices and loops that interact with DNA (3). All the core histones have 15–30 unstructured residues at their amino termini, which are commonly referred to as “tails”; their carboxy termini consist of similarly unstructured tails.

Fig. 1.

Chromosomes, chromatin, nucleosomes and histone modifications. (A) Schematic representation of a chromosome (upper), chromatin fiber (middle) and nucleosomes (lower). Histones are shown as yellow circles, and DNA is shown as a black line. The nucleosome, the fundamental unit of chromatin, consists of a histone octamer wrapped with 146 bp of DNA. Post-translational modifications of histones are shown in small circles: acetylation (Ac, blue), methylation (Me, green), phosphorylation (P, orange), ubiquitination (Ub, purple), sumoylation (SUMO, gray), and poly-ADP-ribosylation (ADP, pink). HATs (histone acetyltransferases) transfer acetyl groups to histones, and HDACs (histone deacetylases) remove them. (B) Specific acetylation of histone lysine residues in vivo. A schematic summary of the results of Turner et al. (8). K5, K8, K12, and K16 represent histone H4 lysine residues 5, 8, 12, and 16, respectively. Distinct patterns of acetylation are found in different chromosomal regions (see text for details).

The tail regions of core histones are subject to various post-translational modifications that are considered to be key reactions in the modulation of chromatin structure and function (Fig. 1A). Post-translational modifications of histones include the acetylation of lysine residues, lysine and arginine methylation, serine and threonine phosphorylation, lysine ubiquitination, lysine sumoylation, and the poly-ADP-ribosylation of glutamic acid (4, 5). Among these, acetylation has been the primary subject of research.

Histone acetylation was first discovered by Allfrey et al. in 1964 and proposed to regulate gene expression (6). This idea was supported by the observation that hyper-acetylation of histones correlates with transcriptional activation (7). Because acetylation of lysine residues neutralizes their positive charges, it was assumed that a decrease in the electrostatic interaction between DNA and histones is the major acetylation-dependent mechanism that regulates gene expression.

Turner et al. observed acetylation of distinct lysine residues in specific chromosomal regions in Drosophila melanogaster polytene nuclei (Fig. 1B) (8). Lysine 5 of histone H4 (H4-K5) or H4-K8 is frequently acetylated in euchromatic regions, where transcription is potentially active. In contrast, acetylation of H4-K12 is increased in heterochromatic regions, where transcription is potentially inactive. Acetylation of H4-K16 is found along the transcriptionally hyperactive male X chromosome (8). These observations suggested that residue-specific acetylation, rather than bulk neutralization of electrostatic charge, is important in regulation of gene expression through histone modification. They further suggested that histone modification mediates not only promoter-specific gene expression but also longer-range (and even chromosome-wide) gene expression.

Although assumed for more than 30 years, the cause-and-effect relationship between histone acetylation and transcriptional activity was not confirmed, mainly because the enzymes responsible for histone acetylation were unknown. Therefore the identification of such enzymes was a major objective in the field of eukaryotic gene expression.

2. Identification of HATs

In 1995, Brownell and Allis developed a histone acetyltransferase (HAT) assay and detected a single polypeptide of 55 kDa (p55) in macronuclear extracts of Tetrahymena thermophilia (9). Cloning of the cDNA encoding p55 revealed striking sequence similarity of the protein with a yeast transcriptional coactivator, Gcn5 (10), and this latter protein was then found to have HAT activity. The identification of a HAT in the transcriptional coactivator was a breakthrough in understanding the causal relationship between histone acetylation and gene expression.

Another gene encoding a HAT was identified in 1995. Kleff et al. screened for mutants defective in HAT activity among a collection of yeast temperature-sensitive mutants with an enzymatic assay that used fractionated cell extracts (11), which led to the discovery of the Hat1 protein. Because the hat1 mutant conferred no obvious phenotypes other than the enzymatic defect, the identification of Hat1 did not directly link histone acetylation and gene expression. However, Hat1 was found to share structural similarity with Gcn5, and both proteins were later assigned to the GNAT (GCN5-related N-acetyltransferases) superfamily (12). Hat1 was the first HAT whose tertiary structure was solved (13), thus contributing to an understanding of the relationship between HAT structure and function.

HAT activities of mammalian proteins were also reported. In 1996, HAT activity of PCAF (p300/CBP-associated factor), a protein that competes with the adenoviral oncoprotein E1A to bind to the coactivator p300/CBP, was reported (14). p300/CBP itself also turned out to be a HAT (15, 16), as did TAFII250 (CCG1/TAF1), the largest subunit of the general transcription initiation factor TFIID, which is conserved among eukaryotes (17). To date, various types of promoter-associated transcriptional coactivators and proteins in transcription machineries have been tested for HAT activity and some of these were shown to be HATs (Table 1) (18, 19). These findings strengthened the idea that localized acetylation of histones by transcription factors contributes to the activation of promoter-specific gene expression.

Table 1.

A list of identified histone acetyltransferases (HATs).

Group
HAT
Organism
Complex
Possible function
Ref.
Gcn5 familyGcn5yeastSAGA, SLIK, SALSA, ADA, HAT-A2transcriptional activation(10)
Gcn5Lmammal/flySTAGA, TFTCtranscriptional activation(95, 96)
PCAFmammalPCAF complextranscriptional activation(14)
MYST familyaTip60mammalTIP60 complextranscriptional activation/DNA repair(20)
HBO1mammalHBO1 complexgene expression?/DNA replication?(97)
MORFmammaltranscriptional activation(98)
MOZmammaltranscriptional activation(99)
MOFmammal/flyMSLdosage compensation(25, 100102)
Esa1yeastNuA4transcriptional activation(36)
Sas3yeastNuA3transcriptional activation?(103)
Sas2yeastSAS-Ianti-silencing(41, 42, 45)
OthersHat1yeastHat1/2 complexhistone deposition(11)
p300/CBPmammaltranscriptional activation(15, 16)
TAFII250 (TAF1)mammal/fly/yeastTFIIDRNA pol II transcription(17)
ACTR/SRC-1mammaltranscriptional activation(104, 105)
Elp3yeastelongatortranscriptional elongation(106)
hTFIIIC110mammalTFIIICRNA pol III transcription(107)
hTFIIIC90mammalTFIIICRNA pol III transcription(108)
Hpa2yeast?(109)
Nut1yeastmediatorRNA pol II transcription(110)
ATF-2mammaltranscriptional activation(111)
Group
HAT
Organism
Complex
Possible function
Ref.
Gcn5 familyGcn5yeastSAGA, SLIK, SALSA, ADA, HAT-A2transcriptional activation(10)
Gcn5Lmammal/flySTAGA, TFTCtranscriptional activation(95, 96)
PCAFmammalPCAF complextranscriptional activation(14)
MYST familyaTip60mammalTIP60 complextranscriptional activation/DNA repair(20)
HBO1mammalHBO1 complexgene expression?/DNA replication?(97)
MORFmammaltranscriptional activation(98)
MOZmammaltranscriptional activation(99)
MOFmammal/flyMSLdosage compensation(25, 100102)
Esa1yeastNuA4transcriptional activation(36)
Sas3yeastNuA3transcriptional activation?(103)
Sas2yeastSAS-Ianti-silencing(41, 42, 45)
OthersHat1yeastHat1/2 complexhistone deposition(11)
p300/CBPmammaltranscriptional activation(15, 16)
TAFII250 (TAF1)mammal/fly/yeastTFIIDRNA pol II transcription(17)
ACTR/SRC-1mammaltranscriptional activation(104, 105)
Elp3yeastelongatortranscriptional elongation(106)
hTFIIIC110mammalTFIIICRNA pol III transcription(107)
hTFIIIC90mammalTFIIICRNA pol III transcription(108)
Hpa2yeast?(109)
Nut1yeastmediatorRNA pol II transcription(110)
ATF-2mammaltranscriptional activation(111)
a

enok and Chameau (Chm) are members of the MYST family in fly; their in vitro HAT activity has yet to be confirmed (112, 113).

Table 1.

A list of identified histone acetyltransferases (HATs).

Group
HAT
Organism
Complex
Possible function
Ref.
Gcn5 familyGcn5yeastSAGA, SLIK, SALSA, ADA, HAT-A2transcriptional activation(10)
Gcn5Lmammal/flySTAGA, TFTCtranscriptional activation(95, 96)
PCAFmammalPCAF complextranscriptional activation(14)
MYST familyaTip60mammalTIP60 complextranscriptional activation/DNA repair(20)
HBO1mammalHBO1 complexgene expression?/DNA replication?(97)
MORFmammaltranscriptional activation(98)
MOZmammaltranscriptional activation(99)
MOFmammal/flyMSLdosage compensation(25, 100102)
Esa1yeastNuA4transcriptional activation(36)
Sas3yeastNuA3transcriptional activation?(103)
Sas2yeastSAS-Ianti-silencing(41, 42, 45)
OthersHat1yeastHat1/2 complexhistone deposition(11)
p300/CBPmammaltranscriptional activation(15, 16)
TAFII250 (TAF1)mammal/fly/yeastTFIIDRNA pol II transcription(17)
ACTR/SRC-1mammaltranscriptional activation(104, 105)
Elp3yeastelongatortranscriptional elongation(106)
hTFIIIC110mammalTFIIICRNA pol III transcription(107)
hTFIIIC90mammalTFIIICRNA pol III transcription(108)
Hpa2yeast?(109)
Nut1yeastmediatorRNA pol II transcription(110)
ATF-2mammaltranscriptional activation(111)
Group
HAT
Organism
Complex
Possible function
Ref.
Gcn5 familyGcn5yeastSAGA, SLIK, SALSA, ADA, HAT-A2transcriptional activation(10)
Gcn5Lmammal/flySTAGA, TFTCtranscriptional activation(95, 96)
PCAFmammalPCAF complextranscriptional activation(14)
MYST familyaTip60mammalTIP60 complextranscriptional activation/DNA repair(20)
HBO1mammalHBO1 complexgene expression?/DNA replication?(97)
MORFmammaltranscriptional activation(98)
MOZmammaltranscriptional activation(99)
MOFmammal/flyMSLdosage compensation(25, 100102)
Esa1yeastNuA4transcriptional activation(36)
Sas3yeastNuA3transcriptional activation?(103)
Sas2yeastSAS-Ianti-silencing(41, 42, 45)
OthersHat1yeastHat1/2 complexhistone deposition(11)
p300/CBPmammaltranscriptional activation(15, 16)
TAFII250 (TAF1)mammal/fly/yeastTFIIDRNA pol II transcription(17)
ACTR/SRC-1mammaltranscriptional activation(104, 105)
Elp3yeastelongatortranscriptional elongation(106)
hTFIIIC110mammalTFIIICRNA pol III transcription(107)
hTFIIIC90mammalTFIIICRNA pol III transcription(108)
Hpa2yeast?(109)
Nut1yeastmediatorRNA pol II transcription(110)
ATF-2mammaltranscriptional activation(111)
a

enok and Chameau (Chm) are members of the MYST family in fly; their in vitro HAT activity has yet to be confirmed (112, 113).

Meanwhile, our group identified a novel class of HATs that regulates long-range/chromosome-wide gene expression (20). A member of the MYST (MOZ, YBF2/SAS3, SAS2, Tip60) family of proteins, Tip60, was shown to possess HAT activity with novel substrate specificity. The HAT activities of other members of the MYST family, including Esa1, a yeast counterpart of Tip60, have been confirmed (Table 1). HAT activities for MYST family proteins were anticipated since they contain sequences similar to the acetyl-CoA binding motif (21) in an evolutionarily conserved region, the MYST domain (20). Interestingly, Esa1 has a sequence motif (the “ER motif ”) also found in a histone deacetylase (HDAC), yeast Rpd3 (22). Other MYST family members, yeast Sas2 and Sas3, are involved in long-range gene repression dependent on chromosomal location (23, 24), and Drosophila MOF is involved in hyperactivation of the male X chromosome (25). The identification of MYST proteins as HATs linked histone acetylation and long-range/chromosome-wide gene expression.

3. The residue-specificity of histone acetylation: A two-step classification and the allocation strategy

Many, but not all, lysines in the amino-terminal tail (N-tail) regions of core histones are acetylated in vivo (Fig. 2A) (26, 27). The site specificity of identified HATs is also divergent (18, 28). Analysis of primary sequences in the vicinity of lysines of core histone N-tails revealed that lysines acetylated in vivo can be classified into three classes and six groups (Fig. 2B) (28). This classification distinguishes lysines acetylated in vivo from others, and fits well with the in vitro site specificity of the catalytic domains of HATs. Therefore, it has been hypothesized that the catalytic domains of HATs recognize classes and groups according to this “two-step classification” hypothesis (28, 29).

Fig. 2.

Acetylation sites in histone amino-terminal tails and the “two-step classification” hypothesis. (A) Primary structures of histone tails in human cells. Acetylation sites, shown in red, were determined through primary structural analyses of histones in cellular extracts (26, 27). Acetylation of most of these lysines has also been detected using specific antibodies (e.g., Ref. 40). These sites coincide with those acetylated by identified HATs (e.g., Ref. 163). Recently, acetylation of four additional lysines in the amino-terminal tail regions of calf thymus histones has been detected (H2A-K9, H2A-K13, H2A-K15 and H4-K20) (164). Further analysis is required before concluding that these lysines are acetylated in vivo. (B) Two-step classification hypothesis. Lysines acetylated in vivo (shown in red in A) are subject to a “two-step classification.” Classes I to III are defined by residues located amino-terminal to acetylated lysines: glycine or alanine (G/A) for class I, serine or threonine (S/T) for class II, and lysine or arginine (K/R) for class III. Each class is subdivided into two groups according to additional flanking residues (groups A to F). For details, see (28). Of the four additional lysines that may be acetylated in vivo (164), the flanking sequences of three (H2A-K9, H2A-K13 and H2A-K15) are similar to those of classified lysines. H2A-K9 can be considered as a group B residue (28), and it is acetylated by a HAT in vitro (29); and H2A-K13 and H2A-K15 can be classified as group A lysines. Further analyses of the acetylation status of these lysine residues and comparison of obtained experimental results with the provisional classification will be of interest.

The hypothesis provides several insights into how HATs select specific lysines. First, the flanking sequences alone are unlikely to determine whether the lysine will be acetylated by a HAT. Some lysines that are not acetylated by HATs in the non-N-tail regions of histones or other proteins meet the classification criteria. HATs also acetylate non-histone substrates (Table 2). Some lysines in these substrates do not fall into any group in the classification. Because the higher order structure of the N-tail regions is considered flexible (3), this structure might be important for the N-tails of histones and non-histone substrates to be acetylated in addition to the flanking sequences of the target lysines. Second, the hypothesis provides a possible explanation for the broad but non-random specificity of Tip60 or p300 (Fig. 2B). For example, there is no apparent and strict consensus sequence for substrate recognition by a class I-specific HAT like Tip60. Because the ‘class I’ lysines consist solely of ‘group A’ and ‘group B’ lysines, the substrate recognition surfaces of the class I-specific HATs may consist of a surface that can recognize both sequences or two distinct surfaces of which one recognizes the consensus sequence of group A and the other that of group B. This speculation, based on the proposed classification, can explain specific lysine selection by HATs without an apparent target consensus sequence.

Table 2.

A list of nonhistone substrates for HATs.a

Function
Substrate
Enzyme
Effect of acetylation
Ref.
transcription factorSp1p300enhancement of DNA binding through interaction with p300, but not acetylation by p300(114)
KLF5p300enhancement of transcription activity(115)
FOXO1CBPinhibition of transcription activity(116)
MEF2Cp300enhancement of DNA binding, transcription and myogenic differentiation(117)
SRYp300increase in importin β binding and participate nuclear localization(118)
GATA-4p300enhancement of DNA binding, participates in transcription and involved in differentiation of embryonic stem cells into cardiac myocytes(119, 120)
HNF-6CBPincrease in stability and stimulate transcription(121)
signaling regulatorStat3p300stimulation of DNA binding and transcription(122)
Function
Substrate
Enzyme
Effect of acetylation
Ref.
transcription factorSp1p300enhancement of DNA binding through interaction with p300, but not acetylation by p300(114)
KLF5p300enhancement of transcription activity(115)
FOXO1CBPinhibition of transcription activity(116)
MEF2Cp300enhancement of DNA binding, transcription and myogenic differentiation(117)
SRYp300increase in importin β binding and participate nuclear localization(118)
GATA-4p300enhancement of DNA binding, participates in transcription and involved in differentiation of embryonic stem cells into cardiac myocytes(119, 120)
HNF-6CBPincrease in stability and stimulate transcription(121)
signaling regulatorStat3p300stimulation of DNA binding and transcription(122)
a

A comprehensive list can be obtained from Yang (2004) (123). Non-histone substrates that do not appear in the list of Yang (2004), are listed. The other non-histone substrates include chromosomal proteins, HAT autoacetylations, chromatin remodelers, transcriptional coregulators, general transcription factors, DNA metabolic enzymes, apoptosis regulator, nuclear import receptor, and viral proteins (123).

Table 2.

A list of nonhistone substrates for HATs.a

Function
Substrate
Enzyme
Effect of acetylation
Ref.
transcription factorSp1p300enhancement of DNA binding through interaction with p300, but not acetylation by p300(114)
KLF5p300enhancement of transcription activity(115)
FOXO1CBPinhibition of transcription activity(116)
MEF2Cp300enhancement of DNA binding, transcription and myogenic differentiation(117)
SRYp300increase in importin β binding and participate nuclear localization(118)
GATA-4p300enhancement of DNA binding, participates in transcription and involved in differentiation of embryonic stem cells into cardiac myocytes(119, 120)
HNF-6CBPincrease in stability and stimulate transcription(121)
signaling regulatorStat3p300stimulation of DNA binding and transcription(122)
Function
Substrate
Enzyme
Effect of acetylation
Ref.
transcription factorSp1p300enhancement of DNA binding through interaction with p300, but not acetylation by p300(114)
KLF5p300enhancement of transcription activity(115)
FOXO1CBPinhibition of transcription activity(116)
MEF2Cp300enhancement of DNA binding, transcription and myogenic differentiation(117)
SRYp300increase in importin β binding and participate nuclear localization(118)
GATA-4p300enhancement of DNA binding, participates in transcription and involved in differentiation of embryonic stem cells into cardiac myocytes(119, 120)
HNF-6CBPincrease in stability and stimulate transcription(121)
signaling regulatorStat3p300stimulation of DNA binding and transcription(122)
a

A comprehensive list can be obtained from Yang (2004) (123). Non-histone substrates that do not appear in the list of Yang (2004), are listed. The other non-histone substrates include chromosomal proteins, HAT autoacetylations, chromatin remodelers, transcriptional coregulators, general transcription factors, DNA metabolic enzymes, apoptosis regulator, nuclear import receptor, and viral proteins (123).

Most HATs form multisubunit complexes. In 1998, subunit compositions of human PCAF complex and yeast Gcn5 complexes (SAGA and ADA) were reported (30, 31). To date, subunits of various HAT complexes have been identified and revealed to play critical roles in regulating HAT activity, for example by targeting activity to specific chromosomal regions (Table 3) (32). The formation of multisubunit complexes also modulates the substrate specificity of HATs. The catalytic domains of most HATs alone are unable to acetylate the histones in the nucleosomal context. HAT complexes, however, can acetylate nucleosomal histones, and specific subunits of these complexes are required for targeting them to nucleosomes. For example, Gcn5 alone is unable to acetylate nucleosomal histones efficiently, whereas the Gcn5 complexes ADA and SAGA can (33). Ada2 and Ada3, subunits common to these two complexes, are required for their association with and acetylation of nucleosomal histones (34). Similarly, the Epl1 and Yng2 subunits of the Esa1 complex NuA4 are sufficient for Esa1 to acetylate nucleosomal histones (35).

Table 3.

A list of HAT complexes.

complex
Gcn5 family
MYST family
Others
SAGA
SLIK
ADA
HAT-A2
STAGA
TFTC
PCAF
NuA4
NuA3
SAS-I
TIP60
MSLd
Hat1/2
Elongator
TFIIDe
TFIIICf
ACTR/SRC-1
Mediator
organism
yeast
yeast
yeast
yeast
mammal
mammal
mammal
yeast
yeast
yeast
mammal
fly
yeast
yeast
mammal/fly/yeast
mammal
mammal
yeast
HAT vs. free histoneaH2AH2AN.D.cH2AH2AH2AH2AN.A.c
H2B
H3H3H3H3H3H3H3H3H3H3H3H3H3H3gH3

H4
H4
H4
H4
H4
H4
H4
H4
H4

H4
H4
H4
H4
H4

H4

HAT complex vs. nucleosomal histonea
H2BH2BH2BH2BH2AH2AN.D.cN.D.cH2AN.A.c
H3H3H3H3H3H3H3H3H3H3H3






H4
H4

H4
H4
H4

H4

H4

H4
catalytic subunit(s)
Gcn5
Gcn5
Gcn5
Gcn5
Gcn5L
Gcn5L
PCAF
Esa1
Sas3
Sas2
Tip60
MOF
Hat1
Elp3
TAFII250 (TAF1)
TFIIIC220 TFIIIC110 TFIIIC90
ACTR/SRC-1
MED5 (Nut1)
otherTralTralTRRAPTRRAPPAF400Yaf9TAF14Sas5GAS41Hat2Elp1TAF2TFIIIC102CBPMED1
subunitsbAda1Ada1STAF42Yng2Yng1ING3Hif1Elp2TAF3TFIIIC63SRC-2MED2
Ada2Ada2Ada2Ada2Ada2Arp4BAF53Elp4TAF4SS-AMED3
Ada3Ada3Ada3Ada3STAF54Ada3Ada3Epl1EPCElp5TAF5IKKβMED4
Ada5Ada5EPC-likeElp6TAF6IKKγMED6
Spt3Spt3Spt3Spt3Spt3Eaf3MRG15MSL3TAF7MED7
Spt7Spt7STAF65γMRGXTAF8MED8
Spt8Eaf7MRGBPTAF9MED9
TAF2Eaf6FLJ11730TAF10MED10
TAF4Eaf2DMAP1TAF11MED11
TAF5TAF5TAF5Tra1TRRAPTAF12MED12
TAF5LTAF5LTAF5LAct1actinTAF13MED13
TAF6TAF6TAF6Eaf1P400TAF14MED14
TAF6LTAF6LTAF6LEaf5TBPMED15
TAF9TAF9TAF9TAF9TAF9Sas4MED16
TAF10TAF10TAF10TAF10TAF10YL1MED17
TAF12TAF12TAF12TAF12TAF12TIP49aMED18
Sgf29TIP49bMED19
Sgf73SCA7SCA7TRCp120MED20
Ubp8Ubp8MLEMED21
Sin4MSL1MED22
Rtg2MSL2CDK8
Ahc1Jil1CycC
STAF36roX RNA
STAF46
STAF55
STAF60
SAP130SAP130
Ref.h(31, 33)(124, 125)(31, 33)(126)(127, 128)(129)(30)(37)(43)(45, 46, 130, 131)(132134)(101)(135, 136)(137, 138)(139)(107)(140)(110)
complex
Gcn5 family
MYST family
Others
SAGA
SLIK
ADA
HAT-A2
STAGA
TFTC
PCAF
NuA4
NuA3
SAS-I
TIP60
MSLd
Hat1/2
Elongator
TFIIDe
TFIIICf
ACTR/SRC-1
Mediator
organism
yeast
yeast
yeast
yeast
mammal
mammal
mammal
yeast
yeast
yeast
mammal
fly
yeast
yeast
mammal/fly/yeast
mammal
mammal
yeast
HAT vs. free histoneaH2AH2AN.D.cH2AH2AH2AH2AN.A.c
H2B
H3H3H3H3H3H3H3H3H3H3H3H3H3H3gH3

H4
H4
H4
H4
H4
H4
H4
H4
H4

H4
H4
H4
H4
H4

H4

HAT complex vs. nucleosomal histonea
H2BH2BH2BH2BH2AH2AN.D.cN.D.cH2AN.A.c
H3H3H3H3H3H3H3H3H3H3H3






H4
H4

H4
H4
H4

H4

H4

H4
catalytic subunit(s)
Gcn5
Gcn5
Gcn5
Gcn5
Gcn5L
Gcn5L
PCAF
Esa1
Sas3
Sas2
Tip60
MOF
Hat1
Elp3
TAFII250 (TAF1)
TFIIIC220 TFIIIC110 TFIIIC90
ACTR/SRC-1
MED5 (Nut1)
otherTralTralTRRAPTRRAPPAF400Yaf9TAF14Sas5GAS41Hat2Elp1TAF2TFIIIC102CBPMED1
subunitsbAda1Ada1STAF42Yng2Yng1ING3Hif1Elp2TAF3TFIIIC63SRC-2MED2
Ada2Ada2Ada2Ada2Ada2Arp4BAF53Elp4TAF4SS-AMED3
Ada3Ada3Ada3Ada3STAF54Ada3Ada3Epl1EPCElp5TAF5IKKβMED4
Ada5Ada5EPC-likeElp6TAF6IKKγMED6
Spt3Spt3Spt3Spt3Spt3Eaf3MRG15MSL3TAF7MED7
Spt7Spt7STAF65γMRGXTAF8MED8
Spt8Eaf7MRGBPTAF9MED9
TAF2Eaf6FLJ11730TAF10MED10
TAF4Eaf2DMAP1TAF11MED11
TAF5TAF5TAF5Tra1TRRAPTAF12MED12
TAF5LTAF5LTAF5LAct1actinTAF13MED13
TAF6TAF6TAF6Eaf1P400TAF14MED14
TAF6LTAF6LTAF6LEaf5TBPMED15
TAF9TAF9TAF9TAF9TAF9Sas4MED16
TAF10TAF10TAF10TAF10TAF10YL1MED17
TAF12TAF12TAF12TAF12TAF12TIP49aMED18
Sgf29TIP49bMED19
Sgf73SCA7SCA7TRCp120MED20
Ubp8Ubp8MLEMED21
Sin4MSL1MED22
Rtg2MSL2CDK8
Ahc1Jil1CycC
STAF36roX RNA
STAF46
STAF55
STAF60
SAP130SAP130
Ref.h(31, 33)(124, 125)(31, 33)(126)(127, 128)(129)(30)(37)(43)(45, 46, 130, 131)(132134)(101)(135, 136)(137, 138)(139)(107)(140)(110)
a

“HAT” indicates that a monomer of the catalytic subunit was examined as an enzyme, while “HAT complex” indicates that the complex forms were used. For the substrates, “free histones” and “nucleosomal histones” indicate that the corresponding forms of histones were used. The references for the specificity of the catalytic subunits alone against free histones are shown in Table 1.

b

Subunits conserved among complexes of the Gcn5 family and MYST family are represented on horizontal lines as in (32). Unified nomenclature for TFIID and mediator subunits is used (141, 142).

c

N.D. = not detected, N.A. = not analyzed.

d

A catalytic domain of MOF (aa. 518–827) acetylates H2A/H3/H4 (101), whereas the full-length MOF shows a strong preference for histone H4 (100). An N-terminal domain is involved in this gain of specificity (100). The full-length MOF is also capable of acetylating of nucleosomal histones with a strong preference for lysine 16 of histone H4 (100).

e

HAT activity toward nucleosomal histones of a TAFII250-containing complex (TFIIDβ) has not been detected (129). Subunit composition in yeast is shown.

f

Recombinant TFIIIC90 alone possesses a HAT activity toward nucleosomal histones with a strong preference for histone H3 (108).

g

Specificity of TFIIIC90. Specificity of TFIIIC220 or TFIIIC110 alone toward free histones has yet to be analyzed.

h

Two review articles (19, 32) were consulted to construct the table.

Table 3.

A list of HAT complexes.

complex
Gcn5 family
MYST family
Others
SAGA
SLIK
ADA
HAT-A2
STAGA
TFTC
PCAF
NuA4
NuA3
SAS-I
TIP60
MSLd
Hat1/2
Elongator
TFIIDe
TFIIICf
ACTR/SRC-1
Mediator
organism
yeast
yeast
yeast
yeast
mammal
mammal
mammal
yeast
yeast
yeast
mammal
fly
yeast
yeast
mammal/fly/yeast
mammal
mammal
yeast
HAT vs. free histoneaH2AH2AN.D.cH2AH2AH2AH2AN.A.c
H2B
H3H3H3H3H3H3H3H3H3H3H3H3H3H3gH3

H4
H4
H4
H4
H4
H4
H4
H4
H4

H4
H4
H4
H4
H4

H4

HAT complex vs. nucleosomal histonea
H2BH2BH2BH2BH2AH2AN.D.cN.D.cH2AN.A.c
H3H3H3H3H3H3H3H3H3H3H3






H4
H4

H4
H4
H4

H4

H4

H4
catalytic subunit(s)
Gcn5
Gcn5
Gcn5
Gcn5
Gcn5L
Gcn5L
PCAF
Esa1
Sas3
Sas2
Tip60
MOF
Hat1
Elp3
TAFII250 (TAF1)
TFIIIC220 TFIIIC110 TFIIIC90
ACTR/SRC-1
MED5 (Nut1)
otherTralTralTRRAPTRRAPPAF400Yaf9TAF14Sas5GAS41Hat2Elp1TAF2TFIIIC102CBPMED1
subunitsbAda1Ada1STAF42Yng2Yng1ING3Hif1Elp2TAF3TFIIIC63SRC-2MED2
Ada2Ada2Ada2Ada2Ada2Arp4BAF53Elp4TAF4SS-AMED3
Ada3Ada3Ada3Ada3STAF54Ada3Ada3Epl1EPCElp5TAF5IKKβMED4
Ada5Ada5EPC-likeElp6TAF6IKKγMED6
Spt3Spt3Spt3Spt3Spt3Eaf3MRG15MSL3TAF7MED7
Spt7Spt7STAF65γMRGXTAF8MED8
Spt8Eaf7MRGBPTAF9MED9
TAF2Eaf6FLJ11730TAF10MED10
TAF4Eaf2DMAP1TAF11MED11
TAF5TAF5TAF5Tra1TRRAPTAF12MED12
TAF5LTAF5LTAF5LAct1actinTAF13MED13
TAF6TAF6TAF6Eaf1P400TAF14MED14
TAF6LTAF6LTAF6LEaf5TBPMED15
TAF9TAF9TAF9TAF9TAF9Sas4MED16
TAF10TAF10TAF10TAF10TAF10YL1MED17
TAF12TAF12TAF12TAF12TAF12TIP49aMED18
Sgf29TIP49bMED19
Sgf73SCA7SCA7TRCp120MED20
Ubp8Ubp8MLEMED21
Sin4MSL1MED22
Rtg2MSL2CDK8
Ahc1Jil1CycC
STAF36roX RNA
STAF46
STAF55
STAF60
SAP130SAP130
Ref.h(31, 33)(124, 125)(31, 33)(126)(127, 128)(129)(30)(37)(43)(45, 46, 130, 131)(132134)(101)(135, 136)(137, 138)(139)(107)(140)(110)
complex
Gcn5 family
MYST family
Others
SAGA
SLIK
ADA
HAT-A2
STAGA
TFTC
PCAF
NuA4
NuA3
SAS-I
TIP60
MSLd
Hat1/2
Elongator
TFIIDe
TFIIICf
ACTR/SRC-1
Mediator
organism
yeast
yeast
yeast
yeast
mammal
mammal
mammal
yeast
yeast
yeast
mammal
fly
yeast
yeast
mammal/fly/yeast
mammal
mammal
yeast
HAT vs. free histoneaH2AH2AN.D.cH2AH2AH2AH2AN.A.c
H2B
H3H3H3H3H3H3H3H3H3H3H3H3H3H3gH3

H4
H4
H4
H4
H4
H4
H4
H4
H4

H4
H4
H4
H4
H4

H4

HAT complex vs. nucleosomal histonea
H2BH2BH2BH2BH2AH2AN.D.cN.D.cH2AN.A.c
H3H3H3H3H3H3H3H3H3H3H3






H4
H4

H4
H4
H4

H4

H4

H4
catalytic subunit(s)
Gcn5
Gcn5
Gcn5
Gcn5
Gcn5L
Gcn5L
PCAF
Esa1
Sas3
Sas2
Tip60
MOF
Hat1
Elp3
TAFII250 (TAF1)
TFIIIC220 TFIIIC110 TFIIIC90
ACTR/SRC-1
MED5 (Nut1)
otherTralTralTRRAPTRRAPPAF400Yaf9TAF14Sas5GAS41Hat2Elp1TAF2TFIIIC102CBPMED1
subunitsbAda1Ada1STAF42Yng2Yng1ING3Hif1Elp2TAF3TFIIIC63SRC-2MED2
Ada2Ada2Ada2Ada2Ada2Arp4BAF53Elp4TAF4SS-AMED3
Ada3Ada3Ada3Ada3STAF54Ada3Ada3Epl1EPCElp5TAF5IKKβMED4
Ada5Ada5EPC-likeElp6TAF6IKKγMED6
Spt3Spt3Spt3Spt3Spt3Eaf3MRG15MSL3TAF7MED7
Spt7Spt7STAF65γMRGXTAF8MED8
Spt8Eaf7MRGBPTAF9MED9
TAF2Eaf6FLJ11730TAF10MED10
TAF4Eaf2DMAP1TAF11MED11
TAF5TAF5TAF5Tra1TRRAPTAF12MED12
TAF5LTAF5LTAF5LAct1actinTAF13MED13
TAF6TAF6TAF6Eaf1P400TAF14MED14
TAF6LTAF6LTAF6LEaf5TBPMED15
TAF9TAF9TAF9TAF9TAF9Sas4MED16
TAF10TAF10TAF10TAF10TAF10YL1MED17
TAF12TAF12TAF12TAF12TAF12TIP49aMED18
Sgf29TIP49bMED19
Sgf73SCA7SCA7TRCp120MED20
Ubp8Ubp8MLEMED21
Sin4MSL1MED22
Rtg2MSL2CDK8
Ahc1Jil1CycC
STAF36roX RNA
STAF46
STAF55
STAF60
SAP130SAP130
Ref.h(31, 33)(124, 125)(31, 33)(126)(127, 128)(129)(30)(37)(43)(45, 46, 130, 131)(132134)(101)(135, 136)(137, 138)(139)(107)(140)(110)
a

“HAT” indicates that a monomer of the catalytic subunit was examined as an enzyme, while “HAT complex” indicates that the complex forms were used. For the substrates, “free histones” and “nucleosomal histones” indicate that the corresponding forms of histones were used. The references for the specificity of the catalytic subunits alone against free histones are shown in Table 1.

b

Subunits conserved among complexes of the Gcn5 family and MYST family are represented on horizontal lines as in (32). Unified nomenclature for TFIID and mediator subunits is used (141, 142).

c

N.D. = not detected, N.A. = not analyzed.

d

A catalytic domain of MOF (aa. 518–827) acetylates H2A/H3/H4 (101), whereas the full-length MOF shows a strong preference for histone H4 (100). An N-terminal domain is involved in this gain of specificity (100). The full-length MOF is also capable of acetylating of nucleosomal histones with a strong preference for lysine 16 of histone H4 (100).

e

HAT activity toward nucleosomal histones of a TAFII250-containing complex (TFIIDβ) has not been detected (129). Subunit composition in yeast is shown.

f

Recombinant TFIIIC90 alone possesses a HAT activity toward nucleosomal histones with a strong preference for histone H3 (108).

g

Specificity of TFIIIC90. Specificity of TFIIIC220 or TFIIIC110 alone toward free histones has yet to be analyzed.

h

Two review articles (19, 32) were consulted to construct the table.

The lysine specificity of HATs is also modulated through multisubunit complex formation. In some cases, the lysine specificity of HAT complexes toward nucleosomal histones is more restricted than that of the catalytic subunit alone toward free histones. This is the case for HATs in the MYST family. Interestingly, there appears to be a systematic pattern underlying the in vivo lysine specificity of MYST family HATs, which we now call “allocation” (Fig. 3A lower). Six lysine residues in histone N-tails (K5 of H2A, K14 of H3, and K5/8/12/16 of H4) are potential targets for acetylation by MYST family HATs (Table 4, Fig. 3A upper) (29, 36). The MYST family has three members (Esa1, Sas2, and Sas3) in Saccharomyces cerevisiae. Esa1 has major effects on H2A-K5 and H4-K5/8/12 acetylation, both in vivo and in the form of multisubunit complexes (3742), whereas Sas3 affects H3-K14 (43, 44) and Sas2 affects H4-K16 (Table 4 and Fig. 3A, lower) (41, 42, 45, 46). Based on these observations, we propose that there is an “allocation” strategy, such that HATs cover all potential acetylation sites while narrowing down the specificity of each member with little overlap (Fig. 3B, lower). Potential acetylation sites are defined by local structures of catalytic domains of HATs and histone N-tails, which may follow the “two-step classification” hypothesis (Fig. 3B, upper). This strategy does not decrease the number of lysines that are acetylated by all family members, while narrowing the specificity of an individual family member. This may explain why HATs form a family. Since the number of the MYST family members is increased in mammals (Table 1), it is intriguing to examine whether further allocation is observed and how it relates to complexity of gene regulation in mammals.

Fig. 3.

Lysine specificity of HAT complexes and the “allocation” hypothesis. (A) Allocation of lysine specificities among members of the MYST-HAT family. (Upper) Potential lysine specificity of the MYST-HAT domain toward free histones as substrates, based on experimental results found for Esa1, Sas3, and human Tip60 (Table 4). Positions of lysines are numbered according to the primary structures of vertebrate histones as in Fig. 2. (Lower) Lysine-specific acetylation of MYST-HAT family members in vivo (Table 4). The six lysines potentially acetylated by the MYST domain are allocated by the MYST-HAT family members for acetylation in vivo. H4-K16 may not be a preferred Esa1 acetylation site compared to other histone H4 lysines (dotted arrow; Table 4). (B) Two-step classification hypothesis and allocation strategy to select specific lysines by HAT. (Upper) The potential specificity of each enzyme is defined by catalytic domain structures, which correspond to the two-step classification of lysines (Fig. 2B). (Lower) Lysine specificity is allocated by family members, and each member regulates specific lysine(s) in vivo.

Table 4.

Lysine specificity of MYST-HATs in Saccharomyces cerevisiae.a

In vitrob
In vivoc
Ref.
MYST vs. free histone
MYST vs. nuc. histone
MYSTc vs. free histone
MYSTc vs. nuc. histone
Western blot
Chromatin IP
Sas2not detectednot detected(41, 42, 45, 46)
H3-K14
H4-K16H4H4-K16H4-K16
Sas3H2A (c)not detected(43, 44, 103)
H3H3H3H3-K9/14
H4H4
Esa1H2Ad-K5(c)not detectedH2A (h/y)H2Ad-K5(c)H2A-K7(y)(3642, 143)
H3-K14H3H2B-K16(y)
H4-K5, K8, K12, K16H4H4-K5, K8, K12eH4-K5, K8, K12H4-K5, K8, K12
In vitrob
In vivoc
Ref.
MYST vs. free histone
MYST vs. nuc. histone
MYSTc vs. free histone
MYSTc vs. nuc. histone
Western blot
Chromatin IP
Sas2not detectednot detected(41, 42, 45, 46)
H3-K14
H4-K16H4H4-K16H4-K16
Sas3H2A (c)not detected(43, 44, 103)
H3H3H3H3-K9/14
H4H4
Esa1H2Ad-K5(c)not detectedH2A (h/y)H2Ad-K5(c)H2A-K7(y)(3642, 143)
H3-K14H3H2B-K16(y)
H4-K5, K8, K12, K16H4H4-K5, K8, K12eH4-K5, K8, K12H4-K5, K8, K12
a

Experimental data shown are from the references indicated. Because the primary structures of amino-terminal tails of histones H2A and H2B vary from species to species (19), the species from which the histones were derived are shown in parentheses for histones H2A and H2B (i.e., c: chicken, h: human, y: yeast).

b

“MYST” indicates that the monomer of the corresponding member (Sas2, Sas3, or Esa1) was examined as an enzyme in the in vitro assays, while “MYSTc” indicates that the complex form of the member was used. For the substrates, “free histones” and “nuc. histones” indicate that free histones and nucleosomal histones were used in the assay, respectively.

c

The sites whose acetylation is reduced in the strains with mutations in the corresponding member (Sas2, Sas3, or Esa1) are detected either by “Western blot” or “Chromatin IP” assay.

d

Lysine 9 of chicken H2A is weakly acetylated by Esa1 compared to lysine 5 of chicken H2A. Because the site is not reported to be acetylated in vivo, the lysine 9 is omitted from further discussion (see Refs. 28 and 29).

e

The level of acetylation of H4-K16 by the Esa1 complex is slightly above the background level in vitro (38).

Table 4.

Lysine specificity of MYST-HATs in Saccharomyces cerevisiae.a

In vitrob
In vivoc
Ref.
MYST vs. free histone
MYST vs. nuc. histone
MYSTc vs. free histone
MYSTc vs. nuc. histone
Western blot
Chromatin IP
Sas2not detectednot detected(41, 42, 45, 46)
H3-K14
H4-K16H4H4-K16H4-K16
Sas3H2A (c)not detected(43, 44, 103)
H3H3H3H3-K9/14
H4H4
Esa1H2Ad-K5(c)not detectedH2A (h/y)H2Ad-K5(c)H2A-K7(y)(3642, 143)
H3-K14H3H2B-K16(y)
H4-K5, K8, K12, K16H4H4-K5, K8, K12eH4-K5, K8, K12H4-K5, K8, K12
In vitrob
In vivoc
Ref.
MYST vs. free histone
MYST vs. nuc. histone
MYSTc vs. free histone
MYSTc vs. nuc. histone
Western blot
Chromatin IP
Sas2not detectednot detected(41, 42, 45, 46)
H3-K14
H4-K16H4H4-K16H4-K16
Sas3H2A (c)not detected(43, 44, 103)
H3H3H3H3-K9/14
H4H4
Esa1H2Ad-K5(c)not detectedH2A (h/y)H2Ad-K5(c)H2A-K7(y)(3642, 143)
H3-K14H3H2B-K16(y)
H4-K5, K8, K12, K16H4H4-K5, K8, K12eH4-K5, K8, K12H4-K5, K8, K12
a

Experimental data shown are from the references indicated. Because the primary structures of amino-terminal tails of histones H2A and H2B vary from species to species (19), the species from which the histones were derived are shown in parentheses for histones H2A and H2B (i.e., c: chicken, h: human, y: yeast).

b

“MYST” indicates that the monomer of the corresponding member (Sas2, Sas3, or Esa1) was examined as an enzyme in the in vitro assays, while “MYSTc” indicates that the complex form of the member was used. For the substrates, “free histones” and “nuc. histones” indicate that free histones and nucleosomal histones were used in the assay, respectively.

c

The sites whose acetylation is reduced in the strains with mutations in the corresponding member (Sas2, Sas3, or Esa1) are detected either by “Western blot” or “Chromatin IP” assay.

d

Lysine 9 of chicken H2A is weakly acetylated by Esa1 compared to lysine 5 of chicken H2A. Because the site is not reported to be acetylated in vivo, the lysine 9 is omitted from further discussion (see Refs. 28 and 29).

e

The level of acetylation of H4-K16 by the Esa1 complex is slightly above the background level in vitro (38).

The formation of multisubunit complexes is also reported to broaden the lysine specificity of HATs. For example, the Gcn5 complexes ADA and SAGA can acetylate histone H2B in vitro, whereas Gcn5 alone cannot (33). Mechanisms underlying this gain of lysine specificity are unknown, although the Ada2 and Ada3 subunits of the complex are necessary and sufficient for broadening the lysine specificity of Gcn5 (34). Tertiary structure analyses of these complexes may provide insight into the underlying mechanisms.

4. The histone code hypothesis

The residue specificity of identified HATs and HAT complexes was a strong evidence against the original proposal that the neutralization of the positive charge of lysine residues and a resultant decrease in electrostatic interactions between DNA and histones is the major consequence of histone acetylation. Instead, the idea that residue-specific modification of histones has unique and specific effects on chromatin function has become widely accepted (8). Post-translational modifications of histones, including acetylation, constitute a code that allows specific interactions or reactions with chromatin-associated components to take place in a chromosomal context. This idea has gained currency as the “histone code hypothesis”, which holds that the code is generated by histone-modifying enzymes of defined specificity and read by nonhistone proteins that bind in a modification-sensitive manner (Table 5 and Fig. 4) (5, 47). The mechanism is likely to be functioning in reactions other than transcription which are regulated by post-translational modification of histones, such as DNA replication, repair and recombination (Fig. 4) (4850).

Fig. 4.

The histone code hypothesis. Schematic of the histone code hypothesis. Histones are labeled with “codes” by histone modification enzymes (“marking of histone” in the figure). These post-translational modifications are recognized by proteins that interact with histones in modification-dependent manners (“reading of the code”). Recruitment of these histone-interacting proteins triggers subsequent reactions on chromatin (“chromatin states”), which cause various changes (“cellular events”).

Table 5.

Examples of residue-specific histone modifications and induced downstream events.

Histone
Residue
Modification
Organism
Modification enzyme
Recognition
Chromatin states (next reaction)
Cellular events
Ref.
H4N.D.aacetylationmammalCDYBRDTchromatin reorganizationspermatogenesis(144, 145)
H3K4methylationyeastSet1Chd1acetylation of histone H3 by SLIK complexresponse to transcriptional stress(146148)
K9methylationyeastClr4Swi6silencingmaintenance of heterochromatin(69, 70)
K9methylationmammalSuv39h1HP1interfere with phosphorylation of H3-S10maintenance of heterochromatin(68, 69, 149)
K9 K27methylationplantKryptonite (for K9)CMT3DNA methylationflowering(150)
Unknown (for K27)
K27methylationflyE(Z)Polycomb (PC)silencinghomeotic gene repression(151153)
K79methylationmammalDOT1L53BP1change higher-order chromatin structurecell cycle(154, 155)
S10bphosphorylationtetrahymenayeastSnf1GCN5acetylation of K14 of H3 by Gcn5transcription(72, 156)
H2AS129phosphorylationyeastMec1Arp4acetylation by NuA4DNA repair(157, 158)
Histone
Residue
Modification
Organism
Modification enzyme
Recognition
Chromatin states (next reaction)
Cellular events
Ref.
H4N.D.aacetylationmammalCDYBRDTchromatin reorganizationspermatogenesis(144, 145)
H3K4methylationyeastSet1Chd1acetylation of histone H3 by SLIK complexresponse to transcriptional stress(146148)
K9methylationyeastClr4Swi6silencingmaintenance of heterochromatin(69, 70)
K9methylationmammalSuv39h1HP1interfere with phosphorylation of H3-S10maintenance of heterochromatin(68, 69, 149)
K9 K27methylationplantKryptonite (for K9)CMT3DNA methylationflowering(150)
Unknown (for K27)
K27methylationflyE(Z)Polycomb (PC)silencinghomeotic gene repression(151153)
K79methylationmammalDOT1L53BP1change higher-order chromatin structurecell cycle(154, 155)
S10bphosphorylationtetrahymenayeastSnf1GCN5acetylation of K14 of H3 by Gcn5transcription(72, 156)
H2AS129phosphorylationyeastMec1Arp4acetylation by NuA4DNA repair(157, 158)
a

Lysines acetylated by CDY are yet to be identified. Interaction between acetylated histones and BRDT was examined using a H4 amino-terminal peptide with acetylation at K5, K8, K12 and K16.

b

Snf1 and GCN5 derive from S. cerevisiae and Tetrahymena, respectively.

Table 5.

Examples of residue-specific histone modifications and induced downstream events.

Histone
Residue
Modification
Organism
Modification enzyme
Recognition
Chromatin states (next reaction)
Cellular events
Ref.
H4N.D.aacetylationmammalCDYBRDTchromatin reorganizationspermatogenesis(144, 145)
H3K4methylationyeastSet1Chd1acetylation of histone H3 by SLIK complexresponse to transcriptional stress(146148)
K9methylationyeastClr4Swi6silencingmaintenance of heterochromatin(69, 70)
K9methylationmammalSuv39h1HP1interfere with phosphorylation of H3-S10maintenance of heterochromatin(68, 69, 149)
K9 K27methylationplantKryptonite (for K9)CMT3DNA methylationflowering(150)
Unknown (for K27)
K27methylationflyE(Z)Polycomb (PC)silencinghomeotic gene repression(151153)
K79methylationmammalDOT1L53BP1change higher-order chromatin structurecell cycle(154, 155)
S10bphosphorylationtetrahymenayeastSnf1GCN5acetylation of K14 of H3 by Gcn5transcription(72, 156)
H2AS129phosphorylationyeastMec1Arp4acetylation by NuA4DNA repair(157, 158)
Histone
Residue
Modification
Organism
Modification enzyme
Recognition
Chromatin states (next reaction)
Cellular events
Ref.
H4N.D.aacetylationmammalCDYBRDTchromatin reorganizationspermatogenesis(144, 145)
H3K4methylationyeastSet1Chd1acetylation of histone H3 by SLIK complexresponse to transcriptional stress(146148)
K9methylationyeastClr4Swi6silencingmaintenance of heterochromatin(69, 70)
K9methylationmammalSuv39h1HP1interfere with phosphorylation of H3-S10maintenance of heterochromatin(68, 69, 149)
K9 K27methylationplantKryptonite (for K9)CMT3DNA methylationflowering(150)
Unknown (for K27)
K27methylationflyE(Z)Polycomb (PC)silencinghomeotic gene repression(151153)
K79methylationmammalDOT1L53BP1change higher-order chromatin structurecell cycle(154, 155)
S10bphosphorylationtetrahymenayeastSnf1GCN5acetylation of K14 of H3 by Gcn5transcription(72, 156)
H2AS129phosphorylationyeastMec1Arp4acetylation by NuA4DNA repair(157, 158)
a

Lysines acetylated by CDY are yet to be identified. Interaction between acetylated histones and BRDT was examined using a H4 amino-terminal peptide with acetylation at K5, K8, K12 and K16.

b

Snf1 and GCN5 derive from S. cerevisiae and Tetrahymena, respectively.

The histone codes are read by proteins that interact with histones in modification-dependent manners (Fig. 5A). One group, bromodomain proteins, is considered to bear acetylated histone interaction domains (51). To date, the bromodomains of various proteins have been reported to interact with acetylated histones in a lysine-specific manner in vitro (Table 6). Bromodomain-dependent binding of proteins to acetylated chromatin supports the binding of bromodomains to acetylated histone in vivo (5256). These bromodomain-containing proteins are components of nucleosome-modulating complexes that also include ATPases and HATs. Acetylation at specific lysines is thought to stabilize these complexes through bromodomain interaction and to stimulate nucleosome remodeling, further acetylation, or the recruitment of TFIID (52, 57). Initial recruitment of a HAT to chromatin may require nucleosome remodeling. In vivo observations indicate that a HAT is recruited to a promoter after a nucleosome remodeling ATPase complex (58, 59). HAT is proposed to be required for the subsequent stable binding of the ATPase complex in these cases (60). HAT may be recruited to chromatin before the ATPase complex and recruit ATPases in some instances (52, 57, 60).

Fig. 5.

Reading histone codes. (A) Depending on specific patterns established by various histone-modification enzymes, distinct proteins are recruited to chromatin, with specific results (e.g. transcriptional activation/inactivation). (B) Schematic of “chromatin crosstalk.” The efficiency of modification at particular residues depends on pre-existing histone modification patterns. Such interdependency might involve residues in the same histone-tail (‘cis tail’ regulation) or those in different histone tails (‘trans tail’ regulation). Abbreviations: HAT, histone acetyltransferase; HMT, histone methyltransferase.

Table 6.

Summary of bromodomains with known acetyl-histone-binding ability.

Protein
Organism
Acetyl-histone-binding
Detection
Ref.
Gcn5humanK5 acetylated-H2ANMR(159)
K8 acetylated-H4
K16 acetylated-H4
yeastK16 acetylated-H4NMR(160)
PCAFhumanacetylated-H3NMR(51)
acetylated-H4
TAFII250humanK16 acetylated-H4NMR(161)
K8/K16 acetylated-H4
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
K14 acetylated-H3Western blot(56)
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd2mouseK5 acetylated-H2BWestern blot(56)
K12 acetylated-H2B
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd4mouseK14 acetylated-H3Western blot(55)
K9/K14 acetylated-H3
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
BRDTmouseacetylated-H4Western blot(145)
Bdf1yeastacetylated-H3 acetylated-H4Coomassie brilliant blue staining(53, 54)
Rsc4yeastK14 acetylated-H3Western blot(162)
Protein
Organism
Acetyl-histone-binding
Detection
Ref.
Gcn5humanK5 acetylated-H2ANMR(159)
K8 acetylated-H4
K16 acetylated-H4
yeastK16 acetylated-H4NMR(160)
PCAFhumanacetylated-H3NMR(51)
acetylated-H4
TAFII250humanK16 acetylated-H4NMR(161)
K8/K16 acetylated-H4
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
K14 acetylated-H3Western blot(56)
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd2mouseK5 acetylated-H2BWestern blot(56)
K12 acetylated-H2B
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd4mouseK14 acetylated-H3Western blot(55)
K9/K14 acetylated-H3
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
BRDTmouseacetylated-H4Western blot(145)
Bdf1yeastacetylated-H3 acetylated-H4Coomassie brilliant blue staining(53, 54)
Rsc4yeastK14 acetylated-H3Western blot(162)
Table 6.

Summary of bromodomains with known acetyl-histone-binding ability.

Protein
Organism
Acetyl-histone-binding
Detection
Ref.
Gcn5humanK5 acetylated-H2ANMR(159)
K8 acetylated-H4
K16 acetylated-H4
yeastK16 acetylated-H4NMR(160)
PCAFhumanacetylated-H3NMR(51)
acetylated-H4
TAFII250humanK16 acetylated-H4NMR(161)
K8/K16 acetylated-H4
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
K14 acetylated-H3Western blot(56)
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd2mouseK5 acetylated-H2BWestern blot(56)
K12 acetylated-H2B
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd4mouseK14 acetylated-H3Western blot(55)
K9/K14 acetylated-H3
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
BRDTmouseacetylated-H4Western blot(145)
Bdf1yeastacetylated-H3 acetylated-H4Coomassie brilliant blue staining(53, 54)
Rsc4yeastK14 acetylated-H3Western blot(162)
Protein
Organism
Acetyl-histone-binding
Detection
Ref.
Gcn5humanK5 acetylated-H2ANMR(159)
K8 acetylated-H4
K16 acetylated-H4
yeastK16 acetylated-H4NMR(160)
PCAFhumanacetylated-H3NMR(51)
acetylated-H4
TAFII250humanK16 acetylated-H4NMR(161)
K8/K16 acetylated-H4
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
K14 acetylated-H3Western blot(56)
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd2mouseK5 acetylated-H2BWestern blot(56)
K12 acetylated-H2B
K8 acetylated-H4
K12 acetylated-H4
K16 acetylated-H4
Brd4mouseK14 acetylated-H3Western blot(55)
K9/K14 acetylated-H3
K5/K12 acetylated-H4
K5/K8/K12/K16 acetylated-H4
BRDTmouseacetylated-H4Western blot(145)
Bdf1yeastacetylated-H3 acetylated-H4Coomassie brilliant blue staining(53, 54)
Rsc4yeastK14 acetylated-H3Western blot(162)

Sir3 (61, 62) and Tup1 (63) are proposed to interact with hypoacetylated histones, and both repress gene expression in S. cerevisiae. Sir3 spreads along chromatin and contributes to gene repression over a range of several kilobases (64). Deacetylation of H4-K16 by an HDAC, Sir2 (65), stimulates binding of Sir3 to chromatin and thus gene silencing, whereas acetylation of this lysine by a MYST-HAT, Sas2, prevents Sir3 from spreading on chromatin and contributes to anti-silencing (41, 42) (see next section). In contrast, Tup1 represses gene expression in a promoter-specific manner. The local recruitment of Tup1 is accomplished by sequence-specific DNA binding proteins such as α2/Mcm1, Mig1 and Sko1 (66). Tup1 acts in concert with a histone H2B/H3-specific HDAC, Hda1, to repress gene expression, possibly by binding to hypoacetylated histones (67).

Other modifications of histones also regulate their interaction with proteins, and these alterations also function as codes (Table 5). For example, methylation of H3-K9 is known to stimulate the binding of chromodomain-containing proteins such as HP1 and Swi6 to chromatin, leading to gene repression (6870).

Modification of histones also influences other histone modifications, a phenomenon called cross talk (71) (Fig. 5B). In cis-tail crosstalk, a given modification affects modification of neighboring residues by physically stabilizing or inhibiting interaction between enzymes and substrates. For example, phosphorylation of serine 10 of histone H3 (H3-S10) enhances acetylation of H3-K14 by Gcn5 (72). In trans-tail crosstalk, a given modification affects modifications on other histone tails. For example, ubiquitination of H2B-K123 by Rad6/Ubc2 is required for methylation of H3-K4 and H3-K79 (73, 74). The interdependency of histone modifications led Fischle et al. to propose that histone modifications function as binary switches (75).

5. Long-range/chromosome-wide gene expression regulated by histone acetylation

Most HATs identified to date are involved mainly in promoter-specific gene expression. Analyses of these HATs have advanced our understanding of the molecular mechanisms by which histone acetylation controls gene expression at specific promoters. In contrast, the regulation of long-range/chromosome-wide gene expression by histone acetylation is poorly understood (Fig. 6). The MYST-type HAT family is a group of HATs involved mainly in long-range/chromosome-wide gene expression.

Fig. 6.

Long-range/chromosome-wide gene expression. (A) Examples of long-range gene regulation in yeast. At telomere-proximal regions (upper) and cryptic mating type loci (middle), Sir2 deacetylates H4-K16 and enhances the assembly of Sir2/Sir3/Sir4 proteins on chromatin in these regions. Sir1 is required for silencing at the mating type loci. The Sir proteins are recruited to the chromosomal regions through DNA-binding proteins that bind to telomere repeat or silencer elements. The RENT (Sir2, Net1, Cdc14) complex regulates silencing at the rDNA locus (lower). (B) Examples of long-range/chromosome-wide gene regulation in higher eukaryotes. At the HOX gene cluster (upper) and on the mammalian X chromosome (middle), H3-K27 is methylated by the EED-EZH2 complex (151, 165). At the HOX gene cluster, methylation enhances the binding of Polycomb (PC) in the PRC1 complex, which leads to silencing of the region. (Lower) Insulators at the β-globin locus are hyperacetylated, and H3-K9 in condensed chromatin region is methylated (80). A silencing protein, HP1, binds to histones methylated at H3-K9 by a histone methyltransferase, Suv39h.

Transcriptional silencing/anti-silencing of genes located near telomeres and the cryptic mating loci of yeast S. cerevisiae is a model system of long-range gene regulation in eukaryotes (64, 7678). Deacetylation of H4-K16 by the Sir2 deacetylase is thought to enhance the binding of Sir3 to chromatin and to cause gene silencing at these regions. However, it was not clear whether deacetylation of this lysine residue was the primary determinant for Sir3 localization and gene silencing in vivo because the HAT acetylating this lysine in vivo was unknown. A MYST protein, Sas2, has been identified as the HAT that acetylates H4-K16 at telomere-distal regions (41, 42). The role of H4-K16 acetylation was evaluated by manipulating the acetylation level in vivo. Mutation of Sas2 and Sir2 caused the boundary between the hyper- and hypo-acetylated regions to advance and recede from its original position. Importantly, the position of this boundary coincided with that of Sir3 localization and of overall transcriptional activity. These results indicate that H4-K16 plays a decisive role in establishing transcriptionally active and inactive regions (Fig. 7A).

Fig. 7.

Negotiable and fixed borders. (A) Chromosomal gradient of histone acetylation near telomeres. Regions near the telomere end are hypoacetylated at H4-K16 through the function of Sir2, whereas telomere-distal regions are hyperacetylated through the function of Sas2. The hypoacetylated regions are enriched in Sir3 and gene expression is silenced. For details, see Ref. 41. (B) (Left) Establishment of fixed borders. An “insulator” DNA element is shown as a green box, and components recruited to this element are indicated as green circles. These factors function as a barrier, an enhancer blocker or both. (Right) Establishment of negotiable borders. The localization of each of a pair of modification enzymes can be defined by specific DNA elements on chromosomes. These enzymes are recruited by proteins bound to the DNA elements and can modify surrounding chromosomal regions. The border of the modified state is established somewhere between the two DNA elements. Distinct chromosome states are established according to specific modification states. Fixed borders (left) are tethered to an “insulator”, whereas negotiable borders (right) can shift according to the position of a “chromosomal gradient” based on the balance of two chromatin modification activities. For details, see (79). (C) Non-biological examples of a fixed border (left: Great Wall of China) and a negotiable border (right: American Civil War). The Great Wall of China was built by the first emperor of China as a first line of defense against the invading tribes north of China. The position of this border has been fixed for over 2,000 years. The American Civil War was fought in the United States in the 1860s between the northern states of the Union and the southern states of the Confederacy. The territories of each army changed during the war, but finally the Union prevailed and unified the states.

More importantly, these results inspired a novel view of chromosomal border establishment (79). A prevailing view of chromosome borders, which we term the “fixed border” model, is that they are ‘walls’ that actively inhibit the function of transcriptional enhancers or silencers between distinct regions on chromosomes (Fig. 7B; reviewed in Ref. 80). Positions of the borders will be defined by specific DNA elements and fixed in chromosomes (Fig. 7C). Analysis of Sas2, however, implied another mechanism that does not require ‘walls’ at chromosomal borders. Instead, it provided evidence for a mechanism (the “negotiable border” model) that defines a border through the balance of opposing enzymatic activities (Fig. 7B) (79). Positions of the borders depend on the strength of each activity and are thus movable (Fig. 7C).

Deacetylation of H4-K16 by Sir2 at the telomere-proximal regions stabilizes binding of Sir3 and Sir4 on the chromosome and inactivates gene expression (76). In contrast, acetylation of H4-K16 by Sas2 at the telomere-distal regions stabilizes binding of a bromodomain containing protein Bdf1 on the chromosome (53). Bdf1 is a component of a chromatin remodeling SWR1 complex, which exchanges conventional histone H2A with a histone variant H2A.Z (Htz1) in nucleosomes (8183). This recruitment of H2A.Z on chromatin antagonizes telomeric silencing (84). Methylation of H3-K4 by Set1 and H3-K79 by Dot1 at telomere-distal regions may also play a role in anti-silencing by preventing Sir proteins to associate with chromatin of these regions (8587). Ubiquitination of H2B-K123 by Rad6/Ubc2 should regulate methylation of these lysines (73, 74).

To establish a negotiable border, histone (or DNA) modification enzymes should be recruited to specific regions on chromosomes. A straightforward scenario for enzyme recruitment is to assume specific DNA elements that interact directly or indirectly with these enzymes. We previously predicted that boundary DNA elements (or insulators) originally assumed to define fixed borders may establish negotiable borders instead (79). Consistent with this prediction, a boundary DNA element in vertebrates recruits HATs to prevent the spread of silenced chromatin (88), and DNA elements that recruit histone modification enzymes function as boundary DNA elements in yeast (89). A temporal analysis of silent chromatin assembly in mammalian cells detected the bidirectional spread of histone and DNA modifications nucleated at a specific chromosome region (90). Other explanations of the recruitment of histone/DNA modification enzymes involve repetitive DNA elements and noncoding RNAs (91). Such mechanisms are important in dosage compensation in mammals and Drosophila and in gene silencing mediated by RNA interference (RNAi) (92). Establishment of eukaryotic chromosomal boundaries to regulate long-range/chromosome-wide gene expression involves many mechanisms (93, 94). How these mechanisms act in concert to regulate long-range/chromosome-wide transcriptional activity is one of the major challenges in the field of eukaryotic gene expression.

6. Perspectives

Here, we have reviewed major advances in histone acetylation research with emphases on lysine specificity in the acetylation reaction and on the regulation of long-range/chromosome-wide gene expression. Post-translational modification of histones also plays critical roles in other reactions involving chromatin, such as DNA replication, repair and recombination (4850). Research in the past ten years has greatly advanced our knowledge not only of histone acetylation but also of acetylation of non-histone proteins, histone modification in general and epigenetic regulation in eukaryotes. Considering its highly packed structure and dynamic modulation throughout the cell cycle, and the tight control over efficient and specific gene expression, the eukaryotic chromosome is still full of mystery. Understanding the post-translational modifications of histones is a key for unlocking its secrets.

*

Present address: Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522.

We are grateful to H. Fukuda and N. Sano for helpful discussions. We thank Drs. Wendy Zhow (http://www.meet-greatwall.org) and Tim Harrison (http://www.swcivilwar.com) for permission to use the images presented in Figure 7C. During this study, A. K. was a Research Fellow of the Japan Society for the Promotion of Science. This work was supported in part by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan and by Exploratory Research for Advanced Technology (ERATO) of the Japan Science and Technology Agency (JST).

REFERENCES

1.

Kornberg, R.D. (

1974
) Chromatin structure: a repeating unit of histones and DNA.
Science
184
,
868
–871

2.

Kornberg, R.D. and Lorch, Y. (

1999
) Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome.
Cell
98
,
285
–294

3.

Luger, K., Mader, A.W., Richmond, R.K., Sargent, D.F., and Richmond, T.J. (

1997
) Crystal structure of the nucleosome core particle at 2.8 Å resolution.
Nature
389
,
251
–260

4.

Isenberg, I. (

1979
) Histones.
Annu. Rev. Biochem.
48
,
159
–191

5.

Strahl, B.D. and Allis, C.D. (

2000
) The language of covalent histone modifications.
Nature
403
,
41
–45

6.

Allfrey, V.G., Faulkner, R., and Mirsky, A.E. (

1964
) Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis.
Proc. Natl. Acad. Sci. USA
51
,
786
–794

7.

Hebbes, T.R., Thorne, A.W., and Crane-Robinson, C. (

1988
) A direct link between core histone acetylation and transcriptionally active chromatin.
EMBO J.
7
,
1395
–1402

8.

Turner, B.M., Birley, A.J., and Lavender, J. (

1992
) Histone H4 isoforms acetylated at specific lysine residues define individual chromosomes and chromatin domains in Drosophila polytene nuclei.
Cell
69
,
375
–384

9.

Brownell, J. and Allis, C.D. (

1995
) An activity gel assay detects a single, catalytically active histone acetyltransferase subunit in Tetrahymena macronuclei.
Proc. Natl. Acad. Sci. USA
92
,
6364
–6368

10.

Brownell, J., Zhou, J., Ranalli, T., Kobayashi, R., Edmondson, D., Roth, S., and Allis, C.D. (

1996
) Tetrahymena histone acetyltransferase A: a homolog to yeast Gcn5p linking histone acetylation to gene activation.
Cell
84
,
843
–851

11.

Kleff, S., Andrulis, E.D., Anderson, C.W., and Sternglanz, R. (

1995
) Identification of a gene encoding a yeast histone H4 acetyltransferase.
J. Biol. Chem.
270
,
24674
–24677

12.

Dyda, F., Klein, D.C., and Hickman, A.B. (

2000
) GCN5-related N-acetyltransferases: A structural overview.
Annu. Rev. Biophy. Biomol. Struc.
29
,
81
–103

13.

Dutnall, R.N., Tafrov, S.T., Sternglanz, R., and Ramakrishnan, V. (

1998
) Structure of the histone acetyltransferase Hat1: a paradigm for the GCN5-related N-acetyltransferase superfamily.
Cell
94
,
427
–438

14.

Yang, X.-J., Ogryzko, V.V., Nishikawa, J., Howard, B.H., and Nakatani, Y. (

1996
) A p300/CBP-associated factor that competes with the adenoviral oncoprotein E1A.
Nature
382
,
319
–324

15.

Ogryzko, V.V., Schiltz, R.L., Russanova, V., Howard, B.H., and Nakatani, Y. (

1996
) The transcriptional coactivators p300 and CBP are histone acetylases.
Cell
87
,
953
–959

16.

Bannister, A.J. and Kouzarides, T. (

1996
) The CBP co-activator is a histone acetyltransferase.
Nature
384
,
641
–643

17.

Mizzen, C.A., Yang, X.J., Kokubo, T., Brownell, J.E., Bannister, A.J., Owen-Hughes, T., Workman, J., Wang, L., Berger, S.L., Kouzarides, T., Nakatani, Y., and Allis, C.D. (

1996
) The TAFII250 subunit of TFIID has histone acetyltransferase activity.
Cell
87
,
1261
–1270

18.

Sterner, D.E. and Berger S.L. (

2000
) Acetylation of histones and transcription-related factors.
Microbiol. Mol. Biol. Rev.
64
,
435
–459

19.

Roth, S.Y., Denu, J.M., and Allis, C.D. (

2001
) Histone acetyltransferases.
Annu. Rev. Biochem.
70
,
81
–120

20.

Yamamoto, T. and Horikoshi, M. (

1997
) Novel substrate specificity of the histone acetyltransferase activity of HIV-1-Tat interactive protein Tip60.
J. Biol. Chem.
272
,
30595
–30598

21.

Lu, L., Berkey, K.A., and Casero, Jr., R.A. (

1996
) RGFGIGS is an amino acid sequence required for acetyl coenzyme A binding and activity of human spermidine/spermine N1 acetyltransferase.
J. Biol. Chem.
271
,
18920
–18924

22.

Adachi, N., Kimura, A., and Horikoshi, M. (

2002
) A conserved motif common to the histone acetyltransferase Esa1 and the histone deacetylase Rpd3.
J. Biol. Chem.
277
,
35688
–35695

23.

Reifsnyder, C., Lowell, J., Clarke, A., and Pillus, L. (

1996
) Yeast SAS silencing genes and human genes associated with AML and HIV-1 Tat interactions are homologous with acetyltransferases.
Nature Genet.
14
,
42
–49

24.

Ehrenhofer-Murray, A.E., Rivier, D.H., and Rine, J. (

1997
) The role of Sas2, an acetyltransferase homologue of Saccharomyces cerevisiae, in silencing and ORC function.
Genetics
145
,
923
–934

25.

Hilfiker, A., Hilfiker-Kleiner, D., Pannuti, A., and Lucchesi, J.C. (

1997
) mof, a putative acetyltransferase gene related to the Tip60 and MOZ human genes and to the SAS genes of yeast, is required for dosage compensation in Drosophila.
EMBO J.
16
,
2054
–2060

26.

Marvin, K.W., Yau, P., and Bradbury, E.M. (

1990
) Isolation and characterization of acetylated histones H3 and H4 and their assembly into nucleosomes.
J. Biol. Chem.
265
,
19839
–19847

27.

Thorne, A.W., Kmiciek, D., Mitchelsom, K., Sautiere, P., and Crane-Robinson, C. (

1990
) Patterns of histone acetylation.
Eur. J. Biochem.
193
,
701
–713

28.

Kimura, A. and Horikoshi, M. (

1998
) How do histone acetyltransferases select lysine residues in core histones?
FEBS Lett.
431
,
131
–133

29.

Kimura, A. and Horikoshi, M. (

1998
) Tip60 acetylates six lysines of a specific class in core histones in vitro.
Genes Cells
3
,
789
–800

30.

Ogryzko, V.V., Kotani, T., Zhang, X., Schiltz, R.L., Howard, T., Yang, X.J., Howard, B.H., Qin, J., and Nakatani, Y. (

1998
) Histone-like TAFs within the PCAF histone acetylase complex.
Cell
94
,
35
–44

31.

Grant, P.A., Schieltz, D., Pray-Grant, M.G., Steger, D.J., Reese, J.C., Yates, J.R. 3rd, and Workman, J.L. (

1998
) A subset of TAF(II)s are integral components of the SAGA complex required for nucleosome acetylation and transcriptional stimulation.
Cell
94
,
45
–53

32.

Carrozza, M.J., Utley, R.T., Workman, J.L., and Côté, J. (

2003
) The diverse functions of histone acetyltransferase complexes.
Trends Genet.
19
,
321
–329

33.

Grant, P.A., Duggan, L., Côté, J., Roberts, S.M., Brownell, J.E., Candau, R., Ohba, R., Owen-Hughes, T., Allis, C.D., Winston, F., Berger, S.L., and Workman, J.L. (

1997
) Yeast Gcn5 functions in two multisubunit complexes to acetylate nucleosomal histones: characterization of an Ada complex and the SAGA (Spt/Ada) complex.
Genes Dev.
11
,
1640
–1650

34.

Balasubramanian, R., Pray-Grant, M.G., Selleck, W., Grant, P.A., and Tan, S. (

2002
) Role of the Ada2 and Ada3 transcriptional coactivators in histone acetylation.
J. Biol. Chem.
277
,
7989
–7995

35.

Boudreault, A.A., Cronier, D., Selleck, W., Lacoste, N., Utley, R.T., Allard, S., Savard, J., Lane, W.S., Tan, S., and Côté, J. (

2003
) Yeast enhancer of polycomb defines global Esa1-dependent acetylation of chromatin.
Genes Dev.
17
,
1415
–1428

36.

Smith, E.R., Eisen, A., Gu, W., Sattah, M., Pannuti, A., Zhou, J., Cook, R.G., Lucchesi, J.C., and Allis, C.D. (

1998
) ESA1 is a histone acetyltransferase that is essential for growth in yeast.
Proc. Natl. Acad. Sci. USA
95
,
3561
–3565

37.

Allard, S., Utley, R.T., Savard, J., Clarke, A., Grant, P., Brandl, C.J., Pillus, L., Workman, J.L., and Côté, J. (

1999
) NuA4, an essential transcription adaptor/histone H4 acetyltransferase complex containing Esa1p and the ATM-related cofactor Tra1p.
EMBO J.
18
,
5108
–5119

38.

Ohba, R., Steger, D.J., Brownell, J.E., Mizzen, C.A., Cook, R.G., Côté, J., Workman, J.L., and Allis, C.D. (

1999
) A novel H2A/H4 nucleosomal histone acetyltransferase in Tetrahymena thermophila.
Mol. Cell. Biol.
19
,
2061
–2068

39.

Vogelauer, M., Wu, J., Suka, N., and Grunstein, M. (

2000
) Global histone acetylation and deacetylation in yeast.
Nature
408
,
495
–498

40.

Suka, N., Suka, Y., Carmen, A.A., Wu, J., and Grunstein, M. (

2001
) Highly specific antibodies determine histone acetylation site usage in yeast heterochromatin and euchromatin.
Mol. Cell
8
,
473
–479

41.

Kimura, A., Umehara, T., and Horikoshi, M. (

2002
) Chromosomal gradient of histone acetylation established by Sas2p and Sir2p functions as a shield against gene silencing.
Nat. Genet.
32
,
370
–377

42.

Suka, N., Luo, K., and Grunstein, M. (

2002
) Sir2p and Sas2p opposingly regulate acetylation of yeast histone H4 lysine16 and spreading of heterochromatin.
Nat. Genet.
32
,
378
–383

43.

John, S., Howe, L., Tafrov, S.T., Grant, P.A., Sternglanz, R., and Workman, J.L. (

2000
) The something about silencing protein, Sas3, is the catalytic subunit of NuA3, a yTAF(II)30-containing HAT complex that interacts with the Spt16 subunit of the yeast CP (Cdc68/Pob3)-FACT complex.
Genes Dev.
14
,
1196
–1208

44.

Howe, L., Auston, D., Grant, P., John, S., Cook, R.G., Workman, J.L., and Pillus, L. (

2001
) Histone H3 specific acetyltransferases are essential for cell cycle progression.
Genes Dev.
15
,
3144
–3154

45.

Sutton, A., Shia, W.J., Band, D., Kaufman, P.D., Osada, S., Workman, J.L., and Sternglanz, R. (

2003
) Sas4 and Sas5 are required for the histone acetyltransferase activity of Sas2 in the SAS complex.
J. Biol. Chem.
278
,
16887
–16892

46.

Shia, W.J., Osada, S., Florens, L., Swanson, S.K., Washburn, M.P., and Workman, J.L. (

2005
) Characterization of the yeast trimeric-SAS acetyltransferase complex.
J. Biol. Chem.
280
,
11987
–11994

47.

Turner, B.M. (

2000
) Histone acetylation and an epigenetic code.
Bioessays
22
,
836
–845

48.

Donaldson, A.D. (

2005
) Shaping time: chromatin structure and the DNA replication programme.
Trends Genet.
21
,
444
–449

49.

Peterson, C.L. and Côté, J. (

2004
) Cellular machineries for chromosomal DNA repair.
Genes Dev.
18
,
602
–616

50.

Thiriet, C. and Hayes, J.J. (

2005
) Chromatin in need of a fix: phosphorylation of H2AX connects chromatin to DNA repair.
Mol. Cell
18
,
617
–622

51.

Dhalluin, C., Carlson, J.E., Zeng, L., He, C., Aggarwal, A.K., and Zhou, M.M. (

1999
) Structure and ligand of a histone acetyltransferase bromodomain.
Nature
399
,
491
–496

52.

Hassan, A.H., Prochasson, P., Neely, K.E., Galasinski, S.C., Chandy, M., Carrozza, M.J., and Workman, J.L. (

2002
) Function and selectivity of bromodomains in anchoring chromatin-modifying complexes to promoter nucleosomes.
Cell
111
,
369
–379

53.

Ladurner, A.G., Inouye, C., Jain, R., and Tjian, R. (

2003
) Bromodomains mediate an acetyl-histone encoded antisilencing function at heterochromatin boundaries.
Mol. Cell
11
,
365
–376

54.

Matangkasombut, O. and Buratowski, S. (

2003
) Different sensitivities of bromodomain factors 1 and 2 to histone H4 acetylation.
Mol. Cell
11
,
353
–363

55.

Dey, A., Chitsaz, F., Abbasi, A., Misteli, T., and Ozato, K. (

2003
) The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis.
Proc. Natl. Acad. Sci. USA
100
,
8758
–8763

56.

Kanno, T., Kanno, Y., Siegel, R.M., Jang, M.K., Lenardo, M.J., and Ozato, K. (

2004
) Selective recognition of acetylated histones by bromodomain proteins visualized in living cells.
Mol. Cell
13
,
33
–43

57.

Agalioti, T., Chen, G., and Thanos, D. (

2002
) Deciphering the transcriptional histone acetylation code for a human gene.
Cell
111
,
381
–392

58.

Cosma, M.P., Tanaka, T., and Nasmyth, K. (

1999
) Ordered recruitment of transcription and chromatin remodeling factors to a cell cycle- and developmentally regulated promoter.
Cell
97
,
299
–311

59.

Krebs, J.E., Kuo, M.H., Allis, C.D., and Peterson, C.L. (

1999
) Cell cycle-regulated histone acetylation required for expression of the yeast HO gene.
Genes Dev.
13
,
1412
–1421

60.

Hassan, A.H., Neely, K.E., and Workman, J.L. (

2001
) Histone acetyltransferase complexes stabilize SWI/SNF binding to promoter nucleosomes.
Cell
104
,
817
–827

61.

Hecht, A., Laroche, T., Strahl-Bolsinger, S., Gasser, S.M., and Grunstein, M. (

1995
) Histone H3 and H4 N-termini interact with SIR3 and SIR4 proteins: a molecular model for the formation of heterochromatin in yeast.
Cell
80
,
583
–592

62.

Carmen, A.A., Milne, L., and Grunstein, M. (

2002
) Acetylation of the yeast histone H4 N terminus regulates its binding to heterochromatin protein SIR3.
J. Biol. Chem.
277
,
4778
–4781

63.

Edmondson, D.G., Smith, M.M., and Roth, S.Y. (

1996
) Repression domain of the yeast global repressor Tup1 interacts directly with histones H3 and H4.
Genes Dev.
10
,
1247
–1259

64.

Grunstein, M. (

1997
) Histone acetylation in chromatin structure and transcription.
Nature
389
,
349
–352

65.

Imai, S., Armstrong, C.M., Kaeberlein, M., and Guarente, L. (

2000
) Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase.
Nature
403
,
795
–800

66.

Smith, R.L. and Johnson, A.D. (

2000
) Turning genes off by Ssn6-Tup1: a conserved system of transcriptional repression in eukaryotes.
Trends Biochem. Sci.
25
,
325
–330

67.

Wu, J., Suka, N., Carlson, M., and Grunstein, M. (

2001
) TUP1 utilizes histone H3/H2B-specific HDA1 deacetylase to repress gene activity in yeast.
Mol. Cell
7
,
117
–126

68.

Lachner, M., O'Carroll, D., Rea, S., Mechtler, K., and Jenuwein, T. (

2001
) Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins.
Nature
410
,
116
–120

69.

Bannister, A.J., Zegerman, P., Partridge, J.F., Miska, E.A., Thomas, J.O., Allshire, R.C., and Kouzarides, T. (

2001
) Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain.
Nature
410
,
120
–124

70.

Nakayama, J., Rice, J.C., Strahl, B.D., Allis, C.D., and Grewal, S.I. (

2001
) Role of histone H3 lysine 9 methylation in epigenetic control of heterochromatin assembly.
Science
292
,
110
–113

71.

Fischle, W., Wang, Y., and Allis, C.D. (

2003
) Histone and chromatin cross-talk.
Curr. Opin. Cell Biol.
15
,
172
–183

72.

Clements, A., Poux, A.N., Lo, W.S., Pillus, L., Berger, S.L., and Marmorstein, R. (

2003
) Structural basis for histone and phosphohistone binding by the GCN5 histone acetyltransferase.
Mol. Cell
12
,
461
–473

73.

Sun, Z.W. and Allis, C.D. (

2002
) Ubiquitination of histone H2B regulates H3 methylation and gene silencing in yeast.
Nature
418
,
104
–108

74.

Briggs, S.D., Xiao, T., Sun, Z.W., Caldwell, J.A., Shabanowitz, J., Hunt, D.F., Allis, C.D., and Strahl, B.D. (

2002
) Gene silencing: trans-histone regulatory pathway in chromatin.
Nature
418
,
498

75.

Fischle, W., Wang, Y., and Allis, C.D. (

2003
) Binary switches and modification cassettes in histone biology and beyond.
Nature
425
,
475
–479

76.

Kurdistani, S.K. and Grunstein, M. (

2003
) Histone acetylation and deacetylation in yeast.
Nat. Rev. Mol. Cell Biol.
4
,
276
–284

77.

Rusche, L.N., Kirchmaier, A.L., and Rine, J. (

2003
) The establishment, inheritance, and function of silenced chromatin in Saccharomyces cerevisiae.
Annu. Rev. Biochem.
72
,
481
–516

78.

Pirrotta, V. and Gross, D.S. (

2005
) Epigenetic silencing mechanisms in budding yeast and fruit fly: different paths, same destinations.
Mol. Cell
18
,
395
–398

79.

Kimura, A. and Horikoshi, M. (

2004
) Partition of distinct chromosomal regions: negotiable border and fixed border.
Genes Cells
9
,
499
–508

80.

West, A.G., Gaszner, M., and Felsenfeld, G. (

2002
) Insulators: many functions, many mechanisms.
Genes Dev.
16
,
271
–288

81.

Mizuguchi, G., Shen, X., Landry, J., Wu, W.H., Sen, S., and Wu, C. (

2004
) ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex.
Science
303
,
343
–348

82.

Krogan, N.J., Keogh, M.C., Datta, N., Sawa, C., Ryan, O.W., Ding, H., Haw, R.A., Pootoolal, J., Tong, A., Canadien, V., Richards, D.P., Wu, X., Emili, A., Hughes, T.R., Buratowski, S., and Greenblatt, J.F. (

2004
) A Snf2 family ATPase complex required for recruitment of the histone H2A variant Htz1.
Mol. Cell
12
,
1565
–1576

83.

Kobor, M.S., Venkatasubrahmanyam, S., Meneghini, M.D., Gin, J.W., Jennings, J.L., Link, A.J., Madhani, H.D., and Rine, J. (

2004
) A protein complex containing the conserved Swi2/Snf2-related ATPase Swr1p deposits histone variant H2A.Z into euchromatin.
PLoS Biol.
2
,
E131

84.

Meneghini, M.D., Wu, M., and Madhani, H.D. (

2003
) Conserved histone variant H2A.Z protects euchromatin from the ectopic spread of silent heterochromatin.
Cell
112
,
725
–736

85.

van Leeuwen, F., Gafken, P.R., and Gottschling, D.E. (

2002
) Dot1p modulates silencing in yeast by methylation of the nucleosome core.
Cell
109
,
745
–756

86.

Ng, H.H., Feng, Q., Wang, H., Erdjument-Bromage, H., Tempst, P., Zhang, Y., and Struhl, K. (

2002
) Lysine methylation within the globular domain of histone H3 by Dot1 is important for telomeric silencing and Sir protein association.
Genes Dev.
16
,
1518
–1527

87.

Santos-Rosa, H., Bannister, A.J., Dehe, P.M., Geli, V., and Kouzarides, T. (

2004
) Methylation of H3 lysine 4 at euchromatin promotes Sir3p association with heterochromatin.
J. Biol. Chem.
279
,
47506
–47512

88.

West, A.G., Huang, S., Gaszner, M., Litt, M.D., and Felsenfeld, G. (

2004
) Recruitment of histone modifications by USF proteins at a vertebrate barrier element.
Mol. Cell
16
,
453
–463

89.

Oki, M., Valenzuela, L., Chiba, T., Ito, T., and Kamakaka, R.T. (

2004
) Barrier proteins remodel and modify chromatin to restrict silenced domains.
Mol. Cell. Biol.
24
,
1956
–1967

90.

Su, R.C., Brown, K.E., Saaber, S., Fisher, A.G., Merkenschlager, M., and Smale, S.T. (

2004
) Dynamic assembly of silent chromatin during thymocyte maturation.
Nat. Genet.
36
,
502
–506

91.

Grewal, S.I. and Moazed, D. (

2003
) Heterochromatin and epigenetic control of gene expression.
Science
301
,
798
–802

92.

Verdel, A., Jia, S., Gerber, S., Sugiyama, T., Gygi, S., Grewal, S.I., and Moazed, D. (

2004
) RNAi-mediated targeting of heterochromatin by the RITS complex.
Science
303
,
672
–676

93.

Henikoff, S., Furuyama, T., and Ahmad, K. (

2004
) Histone variants, nucleosome assembly and epigenetic inheritance.
Trends Genet.
20
,
320
–326

94.

Swaminathan, J., Baxter, E.M., and Corces, V.G. (

2005
) The role of histone H2Av variant replacement and histone H4 acetylation in the establishment of Drosophila heterochromatin.
Genes Dev.
19
,
65
–76

95.

Smith, E.R., Belote, J.M., Schiltz, R.L., Yang, X.J., Moore, P.A., Berger, S.L., Nakatani, Y., and Allis, C.D. (

1998
) Cloning of Drosophila GCN5: conserved features among metazoan GCN5 family members.
Nucleic Acids Res.
26
,
2948
–2954

96.

Xu, W., Edmondson, D.G., and Roth, S.Y. (

1998
) Mammalian GCN5 and P/CAF acetyltransferases have homologous amino-terminal domains important for recognition of nucleosomal substrates.
Mol. Cell. Biol.
18
,
5659
–5669

97.

Iizuka, M. and Stillman, B. (

1999
) Histone acetyltransferase HBO1 interacts with the ORC1 subunit of the human initiator protein.
J. Biol. Chem.
274
,
23027
–23034

98.

Champagne, N., Bertos, N.R., Pelletier, N., Wang, A.H., Vezmar, M., Yang, Y., Heng, H.H., and Yang, X.J. (

1999
) Identification of a human histone acetyltransferase related to monocytic leukemia zinc finger protein.
J. Biol. Chem.
274
,
28528
–28536

99.

Champagne, N., Pelletier, N., and Yang, X.J. (

2001
) The monocytic leukemia zinc finger protein MOZ is a histone acetyltransferase.
Oncogene
20
,
404
–409

100.

Akhtar, A. and Becker, P.B. (

2000
) Activation of transcription through histone H4 acetylation by MOF, an acetyltransferase essential for dosage compensation in Drosophila.
Mol. Cell
5
,
367
–375

101.

Smith, E.R., Pannuti, A., Gu, W., Steurnagel, A., Cook, R.G., Allis, C.D., and Lucchesi, J.C. (

2000
) The Drosophila MSL complex acetylates histone H4 at lysine 16, a chromatin modification linked to dosage compensation.
Mol. Cell. Biol.
20
,
312
–318

102.

Neal, K.C., Pannuti, A., Smith, E.R., and Lucchesi, J.C. (

2000
) A new human member of the MYST family of histone acetyl transferases with high sequence similarity to Drosophila MOF.
Biochim. Biophys. Acta
1490
,
170
–174

103.

Takechi, S. and Nakayama, T. (

1999
) Sas3 is a histone acetyltransferase and requires a zinc finger motif.
Biochem. Biophys. Res. Commun.
266
,
405
–410

104.

Chen, H., Lin, R.J., Schiltz, R.L., Chakravarti, D., Nash, A., Nagy, L., Privalsky, M.L., Nakatani, Y., and Evans, R.M. (

1997
) Nuclear receptor coactivator ACTR is a novel histone acetyltransferase and forms a multimeric activation complex with P/CAF and CBP/p300.
Cell
90
,
569
–580

105.

Spencer, T.E., Jenster, G., Burcin, M.M., Allis, C.D., Zhou, J., Mizzen, C.A., McKenna, N.J., Onate, S.A., Tsai, S.Y., Tsai, M.J., and O'Malley, B.W. (

1997
) Steroid receptor coactivator-1 is a histone acetyltransferase.
Nature
389
,
194
–198

106.

Wittschieben, B.O., Otero, G., de Bizemont, T., Fellows, J., Erdjument-Bromage, H., Ohba, R., Li, Y., Allis, C.D., Tempst, P., and Svejstrup, J.Q. (

1999
) A novel histone acetyltransferase is an integral subunit of elongating RNA polymerase II holoenzyme.
Mol. Cell
4
,
123
–128

107.

Kundu, T.K., Wang, Z., and Roeder, R.G. (

1999
) Human TFIIIC relieves chromatin-mediated repression of RNA polymerase III transcription and contains an intrinsic histone acetyltransferase activity.
Mol. Cell. Biol.
19
,
1605
–1615

108.

Hsieh, Y.J., Kundu, T.K., Wang, Z., Kovelman, R., and Roeder, R.G. (

1999
) The TFIIIC90 subunit of TFIIIC interacts with multiple components of the RNA polymerase III machinery and contains a histone-specific acetyltransferase activity.
Mol. Cell. Biol.
19
,
7697
–7704

109.

Angus-Hill, M.L., Dutnall, R.N., Tafrov, S.T., Sternglanz, R., and Ramakrishnan, V. (

1999
) Crystal structure of the histone acetyltransferase Hpa2: A tetrameric member of the Gcn5-related N-acetyltransferase superfamily.
J. Mol. Biol.
294
,
1311
–1325

110.

Lorch, Y., Beve, J., Gustafsson, C.M., Myers, L.C., and Kornberg, R.D. (

2000
) Mediator-nucleosome interaction.
Mol. Cell
6
,
197
–201

111.

Kawasaki, H., Schiltz, L., Chiu, R., Itakura, K., Taira, K., Nakatani, Y., and Yokoyama, K.K. (

2000
) ATF-2 has intrinsic histone acetyltransferase activity which is modulated by phosphorylation.
Nature
405
,
195
–200

112.

Scott, E.K., Lee, T., and Luo, L. (

2001
) enok encodes a Drosophila putative histone acetyltransferase required for mushroom body neuroblast proliferation.
Curr. Biol.
11
,
99
–104

113.

Grienenberger, A., Miotto, B., Sagnier, T., Cavalli, G., Schramke, V., Geli, V., Mariol, M., Berenger, H., Graba, Y., and Pradel, J. (

2002
) The MYST domain acetyltransferase Chameau functions in epigenetic mechanisms of transcriptional repression.
Curr. Biol.
12
,
762
–766

114.

Suzuki, T., Kimura, A., Nagai, R., Horikoshi, M. (

2000
) Regulation of interaction of the acetyltransferase region of p300 and the DNA-binding domain of Sp1 on and through DNA binding.
Genes Cells
5
,
29
–41

115.

Miyamoto, S., Suzuki, T., Muto, S., Aizawa, K., Kimura, A., Mizuno, Y., Nagino, T., Imai, Y., Adachi, N., Horikoshi, M., and Nagai, R. (

2003
) Positive and negative regulation of the cardiovascular transcription factor KLF5 by p300 and the oncogenic regulator SET through interaction and acetylation on the DNA-binding domain.
Mol. Cell. Biol.
23
,
8528
–8541

116.

Daitoku, H., Hatta, M., Matsuzaki, H., Aratani, S., Ohshima, T., Miyagishi, M., Nakajima, T., and Fukamizu, A. (

2004
) Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity.
Proc. Natl. Acad. Sci. USA
101
,
10042
–10047

117.

Ma, K., Chan, J.K., Zhu, G., and Wu, Z. (

2005
) Myocyte enhancer factor 2 acetylation by p300 enhances its DNA binding activity, transcriptional activity, and myogenic differentiation.
Mol. Cell. Biol.
25
,
3575
–3582

118.

Thevenet, L., Mejean, C., Moniot, B., Bonneaud, N., Galeotti, N., Aldrian-Herrada, G., Poulat, F., Berta, P., Benkirane, M., and Boizet-Bonhoure, B. (

2004
) Regulation of human SRY subcellular distribution by its acetylation/deacetylation.
EMBO J.
23
,
3336
–3345

119.

Yanazume, T., Hasegawa, K., Morimoto, T., Kawamura, T., Wada, H., Matsumori, A., Kawase, Y., Hirai, M., and Kita, T. (

2003
) Cardiac p300 is involved in myocyte growth with decompensated heart failure.
Mol. Cell. Biol.
23
,
3593
–3606

120.

Kawamura, T., Ono, K., Morimoto, T., Wada, H., Hirai, M., Hidaka, K., Morisaki, T., Heike, T., Nakahata, T., Kita, T., and Hasegawa, K. (

2005
) Acetylation of GATA-4 is involved in the differentiation of embryonic stem cells into cardiac myocytes.
J. Biol. Chem.
280
,
19682
–19688

121.

Rausa, F.M., 3rd, Hughes, D.E., and Costa, R.H. (

2004
) Stability of the hepatocyte nuclear factor 6 transcription factor requires acetylation by the CREB-binding protein coactivator.
J. Biol. Chem.
279
,
43070
–43076

122.

Wang, R., Cherukuri, P., and Luo, J. (

2005
) Activation of Stat3 sequence-specific DNA binding and transcription by p300/CREB-binding protein-mediated acetylation.
J. Biol. Chem.
280
,
11528
–11534

123.

Yang, X.J. (

2004
) Lysine acetylation and the bromodomain: a new partnership for signaling.
Bioessays
26
,
1076
–1087

124.

Pray-Grant, M.G., Schieltz, D., McMahon, S.J., Wood, J.M., Kennedy, E.L., Cook, R.G., Workman, J.L., Yates, J.R. 3rd, and Grant, P.A. (

2002
) The novel SLIK histone acetyltransferase complex functions in the yeast retrograde response pathway.
Mol. Cell. Biol.
22
,
8774
–8786

125.

Daniel, J.A., Torok, M.S., Sun, Z.W., Schieltz, D., Allis, C.D., Yates, J.R., 3rd, and Grant, P.A. (

2004
) Deubiquitination of histone H2B by a yeast acetyltransferase complex regulates transcription.
J. Biol. Chem.
279
,
1867
–1871

126.

Sendra, R., Tse, C., and Hansen, J.C. (

2000
) The yeast histone acetyltransferase A2 complex, but not free Gcn5p, binds stably to nucleosomal arrays.
J. Biol. Chem.
275
,
24928
–24934

127.

Martinez, E., Palhan, V.B., Tjernberg, A., Lymar, E.S., Gamper, A.M., Kundu, T.K., Chait, B.T., and Roeder, R.G. (

2001
) Human STAGA complex is a chromatin-acetylating transcription coactivator that interacts with pre-mRNA splicing and DNA damage-binding factors in vivo.
Mol. Cell. Biol.
21
,
6782
–6795

128.

Helmlinger, D., Hardy. S., Sasorith, S., Klein, F., Robert, F., Weber, C., Miguet, L., Potier, N., Van-Dorsselaer, A., Wurtz, J.M., Mandel, J.L., Tora, L., and Devys, D. (

2004
) Ataxin-7 is a subunit of GCN5 histone acetyltransferase-containing complexes.
Hum. Mol. Genet.
13
,
11257
–1265

129.

Brand, M., Yamamoto, K., Staub, A., and Tora, L. (

1999
) Identification of TATA-binding protein-free TAFII-containing complex subunits suggests a role in nucleosome acetylation and signal transduction.
J. Biol. Chem.
274
,
18285
–18289

130.

Osada, S., Sutton, A., Muster, N., Brown, C.E., Yates, J.R. 3rd, Sternglanz, R., and Workman, J.L. (

2001
) The yeast SAS (something about silencing) protein complex contains a MYST-type putative acetyltransferase and functions with chromatin assembly factor ASF1.
Genes Dev.
15
,
3155
–3168

131.

Meijsing, S.H. and Ehrenhofer-Murray, A.E. (

2001
) The silencing complex SAS-I links histone acetylation to the assembly of repressed chromatin by CAF-I and Asf1 in Saccharomyces cerevisiae.
Genes Dev.
15
,
3169
–3182

132.

Ikura, T., Ogryzko, V.V., Grigoriev, M., Groisman, R., Wang, J., Horikoshi, M., Scully, R., Qin, J., and Nakatani, Y. (

2000
) Involvement of the TIP60 histone acetylase complex in DNA repair and apoptosis.
Cell
102
,
463
–473

133.

Cai, Y., Jin, J., Tomomori-Sato, C., Sato, S., Sorokina, I., Parmely, T.J., Conaway, R.C., and Conaway, J.W. (

2003
) Identification of new subunits of the multiprotein mammalian TRRAP/TIP60-containing histone acetyltransferase complex.
J. Biol. Chem.
278
,
42733
–42736

134.

Cai, Y., Jin, J., Florens, L., Swanson, S.K., Kusch, T., Li, B., Workman, J.L., Washburn, M.P., Conaway, R.C., and Conaway, J.W. (

2005
) The mammalian YL1 protein is a shared subunit of the TRRAP/TIP60 histone acetyltransferase and SRCAP complexes.
J. Biol. Chem.
280
,
13665
–13670

135.

Parthun, M.R., Widom, J., and Gottschling, D.E. (

1996
) The major cytoplasmic histone acetyltransferase in yeast: links to chromatin replication and histone metabolism.
Cell
87
,
85
–94

136.

Poveda, A., Pamblanco, M., Tafrov, S., Tordera, V., Sternglanz, R., and Sendra, R. (

2004
) Hif1 is a component of yeast histone acetyltransferase B, a complex mainly localized in the nucleus.
J. Biol. Chem.
279
,
16033
–16043

137.

Winkler, G.S., Petrakis, T.G., Ethelberg, S., Tokunaga, M., Erdjument-Bromage, H., Tempst, P., and Svejstrup, J.Q. (

2001
) RNA polymerase II elongator holoenzyme is composed of two discrete subcomplexes.
J. Biol. Chem.
276
,
32743
–32749

138.

Winkler, G.S., Kristjuhan, A., Erdjument-Bromage, H., Tempst, P., and Svejstrup, J.Q. (

2002
) Elongator is a histone H3 and H4 acetyltransferase important for normal histone acetylation levels in vivo.
Proc. Natl. Acad. Sci. USA
99
,
3517
–3522

139.

Burley, S.K. and Roeder, R.G. (

1996
) Biochemistry and structural biology of transcription factor IID (TFIID).
Annu. Rev. Biochem.
65
,
769
–799

140.

Wu, R.C., Qin, J., Hashimoto, Y., Wong, J., Xu, J., Tsai, S.Y., Tsai, M.J., and O'Malley, B.W. (

2002
) Regulation of SRC-3 (pCIP/ACTR/AIB-1/RAC-3/TRAM-1) Coactivator activity by I kappa B kinase.
Mol. Cell. Biol.
22
,
3549
–3561

141.

Tora, L. (

2002
) A unified nomenclature for TATA box binding protein (TBP)-associated factors (TAFs) involved in RNA polymerase II transcription.
Genes Dev.
16
,
673
–675

142.

Bourbon, H.M., Aguilera, A., Ansari, A.Z., Asturias, F.J., Berk, A.J., Bjorklund, S., Blackwell, T.K., Borggrefe, T., Carey, M., Carlson, M., Conaway, J.W., Conaway, R.C., Emmons, S.W., Fondell, J.D., Freedman, L.P., Fukasawa, T., Gustafsson, C.M., Han, M., He, X., Herman, P.K., Hinnebusch, A.G., Holmberg, S., Holstege, F.C., Jaehning, J.A., Kim, Y.J., Kuras, L., Leutz, A., Lis, J.T., Meisterernest, M., Naar, A.M., Nasmyth, K., Parvin, J.D., Ptashne, M., Reinberg, D., Ronne, H., Sadowski, I., Sakurai, H., Sipiczki, M., Sternberg, P.W., Stillman, D.J., Strich, R., Struhl, K., Svejstrup, J.Q., Tuck, S., Winston, F., Roeder, R.G, and Kornberg, R.D. (

2004
) A unified nomenclature for protein subunits of mediator complexes linking transcriptional regulators to RNA polymerase II.
Mol. Cell
14
,
553
–557

143.

Clarke, A.S., Lowell, J.E., Jacobson, S.J., and Pillus, L. (

1999
) Esa1p is an essential histone acetyltransferase required for cell cycle progression.
Mol. Cell. Biol.
19
,
2515
–2526

144.

Lahn, B.T., Tang, Z.L., Zhou, J., Barndt, R.J., Parvinen, M., Allis, C.D., and Page, D.C. (

2002
) Previously uncharacterized histone acetyltransferases implicated in mammalian spermatogenesis.
Proc. Natl. Acad. Sci. USA
99
,
8707
–8712

145.

Pivot-Pajot, C., Caron, C., Govin, J., Vion, A., Rousseaux, S., and Khochbin, S. (

2003
) Acetylation-dependent chromatin reorganization by BRDT, a testis-specific bromodomain-containing protein.
Mol. Cell. Biol.
23
,
5354
–5365

146.

Santos-Rosa, H., Schneider, R., Bannister, A.J., Sherriff, J., Bernstein, B.E., Emre, N.C., Schreiber, S.L., Mellor, J., and Kouzarides, T. (

2002
) Active genes are tri-methylated at K4 of histone H3.
Nature
419
,
407
–411

147.

Pray-Grant, M.G., Daniel, J.A., Schieltz, D., Yates, JR, 3rd, and Grant, P.A. (

2005
) Chd1 chromodomain links histone H3 methylation with SAGA- and SLIK-dependent acetylation.
Nature
433
,
434
–438

148.

Zhang, L., Schroeder, S., Fong, N., and Bentley, D.L. (

2005
) Altered nucleosome occupancy and histone H3K4 methylation in response to ‘transcriptional stress’.
EMBO J.
24
,
2379
–2390

149.

Rea, S., Eisenhaber, F., O'Carroll, D., Strahl, B.D., Sun, Z.W., Schmid, M., Opravil, S., Mechtler, K., Ponting, C.P., Allis, C.D., and Jenuwein, T. (

2000
) Regulation of chromatin structure by site-specific histone H3 methyltransferases.
Nature
406
,
593
–599

150.

Lindroth, A.M., Shultis, D., Jasencakova, Z., Fuchs, J., Johnson, L., Schubert, D., Patnaik, D., Pradhan, S., Goodrich, J., Schubert, I., Jenuwein, T., Khorasanizadeh, S., and Jacobsen, S.E. (

2004
) Dual histone H3 methylation marks at lysines 9 and 27 required for interaction with CHROMOMETHYLASE3.
EMBO J.
23
,
4286
–4296

151.

Cao, R., Wang, L., Wang, H., Xia, L., Erdjument-Bromage, H., Tempst, P., Jones, R.S., and Zhang, Y. (

2002
) Role of histone H3 lysine 27 methylation in Polycomb-group silencing.
Science
298
,
1039
–1043

152.

Czermin, B., Melfi, R., McCabe, D., Seitz, V., Imhof, A., and Pirrotta, V. (

2002
) Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites.
Cell
111
,
185
–196

153.

Muller, J., Hart, C.M., Francis, N.J., Vargas, M.L., Sengupta, A., Wild, B., Miller, E.L., O'Connor, M.B., Kingston, R.E., and Simon, J.A. (

2002
) Histone methyltransferase activity of a Drosophila Polycomb group repressor complex.
Cell
111
,
197
–208

154.

Huyen, Y., Zgheib, O., Ditullio, R.A., Jr., Gorgoulis, V.G., Zacharatos, P., Petty, T.J., Sheston, E.A., Mellert, H.S., Stavridi, E.S., and Halazonetis, T.D. (

2004
) Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks.
Nature
432
,
406
–411

155.

Feng, Q., Wang, H., Ng, H.H., Erdjument-Bromage, H., Tempst, P., Struhl, K., and Zhang, Y. (

2002
) Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain.
Curr. Biol.
12
,
1052
–1058

156.

Lo, W.S., Duggan, L., Emre, N.C., Belotserkovskya, R., Lane, W.S., Shiekhattar, R., and Berger, S.L. (

2001
) Snf1—a histone kinase that works in concert with the histone acetyltransferase Gcn5 to regulate transcription.
Science
293
,
1142
–1146

157.

Downs, J.A., Lowndes, N.F., and Jackson, S.P. (

2000
) A role for Saccharomyces cerevisiae histone H2A in DNA repair.
Nature
408
,
1001
–1004

158.

Downs, J.A., Allard, S., Jobin-Robitaille, O., Javaheri, A., Auger, A., Bouchard, N., Kron, S.J., Jackson, S.P., and Côté, J (

2004
) Binding of chromatin-modifying activities to phosphorylated histone H2A at DNA damage sites.
Mol. Cell
16
,
979
–990

159.

Hudson, B.P., Martinez-Yamout, M.A., Dyson, H.J., and Wright, P.E. (

2000
) Solution structure and acetyl-lysine binding activity of the GCN5 bromodomain.
J. Mol. Biol.
304
,
355
–370

160.

Owen, D.J., Ornaghi, P., Yang, J.C., Lowe, N., Evans, P.R., Ballario, P., Neuhaus, D., Filetici, P., and Travers, A.A. (

2000
) The structural basis for the recognition of acetylated histone H4 by the bromodomain of histone acetyltransferase gcn5p.
EMBO J.
19
,
6141
–6149

161.

Jacobson, R.H., Ladurner, A.G., King, D.S., and Tjian, R. (

2000
) Structure and function of a human TAFII250 double bromodomain module.
Science
288
,
1422
–1425

162.

Kasten, M., Szerlong, H., Erdjument-Bromage, H., Tempst, P., Werner, M., and Cairns, B.R. (

2004
) Tandem bromodomains in the chromatin remodeler RSC recognize acetylated histone H3 Lys14.
EMBO J.
23
,
1348
–1359

163.

Schiltz, R.L., Mizzen, C.A., Vassilev, A., Cook, R.G., Allis, C.D., and Nakatani, Y. (

1999
) Overlapping but distinct patterns of histone acetylation by the human coactivators p300 and PCAF within nucleosomal substrates.
J. Biol. Chem.
274
,
1189
–1192

164.

Zhang, L., Eugeni, E.E., Parthun, M.K., and Freitas, M.A. (

2003
) Identification of novel histone post-translational modifications by peptide mass fingerprinting.
Chromosoma
112
,
77
–86

165.

Cao, R. and Zhang, Y. (

2004
) The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3.
Curr. Opin. Genet. Dev.
14
,
155
–164

Author notes

1Laboratory of Developmental Biology, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032; and 2Horikoshi Gene Selector Project, Exploratory Research for Advanced Technology (ERATO), Japan Science and Technology Agency (JST), 5-9-6 Tokodai, Tsukuba, Ibaraki 300-2635.