histone modification and regulation of chromatin function

15
ISSN 1022-7954, Russian Journal of Genetics, 2006, Vol. 42, No. 9, pp. 970–984. © Pleiades Publishing, Inc., 2006. Original Russian Text © D.E. Koryakov, 2006, published in Genetika, 2006, Vol. 42, No. 9, pp. 1170–1185. 970 NUCLEOSOME STRUCTURE Almost no free DNA occurs in eukaryotic cell nuclei. DNA is associated with many proteins to form a complex known as chromatin. The association of DNA with proteins is not random, but rather shows a strongly hierarchic multilevel organization wherein nucleosomes correspond to the first level. Nucleosomes are repetitive units that each represent a protein globule with DNA wrapping round it by 1.75 turns [1–3] (Fig. 1). A globule includes histones of five types: H1, H2A, H2B, H3, and H4, which are all enriched in basic amino acid residues, lysine and arginine. H2A, H2B, H3, and H4 molecules form a wedge-shaped core octamer: its narrow part is formed by an (H3–H4) 2 tetramer, and the wide part consists of two H2A–H2B dimers [4–6]. The C-termi- nal regions of the octamer histones are tightly folded, while the flexible N-terminal regions, also known as N tails, are unfolded and freely radiate outwards [7, 8]. One molecule of histone H1 binds to the outer surface of the nucleosome in the region of the (H3–H4) 2 tet- ramer, thereby fixing DNA on it [9–13]. Histone H1 has a globular central region of approximately 80 amino acid residues and free C and N ends [14]. In bird and reptilian erythrocytes, histone H1 is replaced by its variant known as H5 in inactive chromatin [15]. The DNA region corresponding to one nucleosome varies in size, reaching 200 bp [16, 17]. Of these, 146 bp are directly associated with the histone octamer, and the other several tens of base pairs link two neighbor nucleosomes [5, 13, 18, 19]. The histone sequences are highly conserved. The earliest sequencing data showed that histone H4 differs only in two residues between calf and pea [20, 21]. The histone sequences are now known for many organisms, from protozoa to humans [22]. Sequence comparisons demonstrated that histones H3 and H4 are conserved to the greatest extent, while histones H2A and H2B are least conserved [23]. A detailed study of the sequences showed that his- tones of every type but H4 are heterogeneous in cells of one organism, forming a family. Each family includes a major type and minor fractions known as “histone vari- ants.” Different organisms possess up to four variants of histone H3, approximately eight variants of histone H2A, up to seven variants of histone H1, and at least three variants of histone H2B. Most, if not all, variants are associated with a certain process, e.g., transcription or X-chromosome inactivation [24–27]. MOLECULAR MARKS Every process occurring in chromatin is a chain of enzymatic reactions and essentially concerns transmis- sion, protection, or realization of genetic information. Such processes can be grouped by stability and period- Histone Modification and Regulation of Chromatin Function D. E. Koryakov Institute of Cytology and Genetics, Russian Academy of Sciences, Novosibirsk, 630090 Russia; fax: (383) 330-16-65; e-mail: [email protected] Department of Cytology and Genetics, Novosibirsk State University, Novosibirsk, 630090 Russia Received March 2, 2006 Abstract—Nucleosomes play two main roles, acting as basal units in DNA compaction and coordinating most processes in chromosomes. The coordination is due to modification of histones, proteins forming nucleosomes. The review briefly describes the nucleosome structure and major modifications of histones and considers the role of such modifications in transcriptional suppression and activation. DOI: 10.1134/S1022795406090043 THEORETICAL PAPERS AND REVIEWS H1 DNA N tails Histone octamer 2 × (H2A–H2B–H3–H4) Fig. 1. Organization of an individual nucleosome. Different histones are differently shadowed. See text for comments.

Upload: d-e-koryakov

Post on 29-Jul-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Histone modification and regulation of chromatin function

ISSN 1022-7954, Russian Journal of Genetics, 2006, Vol. 42, No. 9, pp. 970–984. © Pleiades Publishing, Inc., 2006.Original Russian Text © D.E. Koryakov, 2006, published in Genetika, 2006, Vol. 42, No. 9, pp. 1170–1185.

970

NUCLEOSOME STRUCTURE

Almost no free DNA occurs in eukaryotic cellnuclei. DNA is associated with many proteins to form acomplex known as chromatin. The association of DNAwith proteins is not random, but rather shows a stronglyhierarchic multilevel organization wherein nucleosomescorrespond to the first level. Nucleosomes are repetitiveunits that each represent a protein globule with DNAwrapping round it by 1.75 turns [1–3] (Fig. 1). A globuleincludes histones of five types: H1, H2A, H2B, H3, andH4, which are all enriched in basic amino acid residues,lysine and arginine. H2A, H2B, H3, and H4 moleculesform a wedge-shaped core octamer: its narrow part isformed by an (H3–H4)

2

tetramer, and the wide partconsists of two H2A–H2B dimers [4–6]. The C-termi-nal regions of the octamer histones are tightly folded,while the flexible N-terminal regions, also known asN tails, are unfolded and freely radiate outwards [7, 8].One molecule of histone H1 binds to the outer surfaceof the nucleosome in the region of the (H3–H4)

2

tet-ramer, thereby fixing DNA on it [9–13]. Histone H1 hasa globular central region of approximately 80 aminoacid residues and free C and N ends [14]. In bird andreptilian erythrocytes, histone H1 is replaced by itsvariant known as H5 in inactive chromatin [15]. TheDNA region corresponding to one nucleosome varies insize, reaching 200 bp [16, 17]. Of these, 146 bp aredirectly associated with the histone octamer, and theother several tens of base pairs link two neighbornucleosomes [5, 13, 18, 19].

The histone sequences are highly conserved. Theearliest sequencing data showed that histone H4 differsonly in two residues between calf and pea [20, 21]. Thehistone sequences are now known for many organisms,from protozoa to humans [22]. Sequence comparisonsdemonstrated that histones H3 and H4 are conserved to

the greatest extent, while histones H2A and H2B areleast conserved [23].

A detailed study of the sequences showed that his-tones of every type but H4 are heterogeneous in cells ofone organism, forming a family. Each family includes amajor type and minor fractions known as “histone vari-ants.” Different organisms possess up to four variants ofhistone H3, approximately eight variants of histoneH2A, up to seven variants of histone H1, and at leastthree variants of histone H2B. Most, if not all, variantsare associated with a certain process, e.g., transcriptionor X-chromosome inactivation [24–27].

MOLECULAR MARKS

Every process occurring in chromatin is a chain ofenzymatic reactions and essentially concerns transmis-sion, protection, or realization of genetic information.Such processes can be grouped by stability and period-

Histone Modification and Regulation of Chromatin Function

D. E. Koryakov

Institute of Cytology and Genetics, Russian Academy of Sciences, Novosibirsk, 630090 Russia; fax: (383) 330-16-65; e-mail: [email protected]

Department of Cytology and Genetics, Novosibirsk State University, Novosibirsk, 630090 Russia

Received March 2, 2006

Abstract

—Nucleosomes play two main roles, acting as basal units in DNA compaction and coordinating mostprocesses in chromosomes. The coordination is due to modification of histones, proteins forming nucleosomes.The review briefly describes the nucleosome structure and major modifications of histones and considers therole of such modifications in transcriptional suppression and activation.

DOI:

10.1134/S1022795406090043

THEORETICAL PAPERS AND REVIEWS

H1

DNA

N tails

Histoneoctamer

2

×

(H2A–H2B–H3–H4)

Fig. 1.

Organization of an individual nucleosome. Differenthistones are differently shadowed. See text for comments.

Page 2: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 971

icity. For instance, the heterochromatin state can bemaintained stable through many cell generations, whileDNA replication in the cell cycle is strongly periodical.The timing of various processes and the temporal andspatial coordination of different processes take advan-tage of molecular marks. In other words, to trigger reac-tion B, a system must detect a mark indicating that reac-tion A is complete. Another problem is that free DNAis necessary for most processes taking place in chroma-tin, while nucleosomal folding seems at first glance tohinder such processes. Yet it is actually nucleosomesthat ensure coordinate functions of chromatin, becausehistones carry the above molecular marks. Acetylationand methylation of some residues in histones wereobserved long before the spatial structure of the nucleo-some was resolved [28–30]. Modification of these andother types provides molecular marks, which can beclassified as genetic and not epigenetic. The formergroup includes stable marks that are not associated withDNA nucleotide sequence, are inherited in successivegenerations, and convey information about a process.Other marks serve to temporally maintain a particularstate of chromatin.

Modification involves mostly free N tails, while theglobular and C-terminal regions of histones are modi-fied to a far lower extent. The best-studied posttransla-tional modifications of amino acid residues are acetyla-tion of lysine, methylation of lysine and arginine, phos-phorylation of serine and threonine, and ubiquitylationof lysine (Fig. 2). Note that such modifications are post-translational indeed; i.e., certain groups are added aftersynthesis of a histone molecule. This effect was demon-strated in experiments with a protein synthesis inhibi-tor: added into a system in vitro, the inhibitor sup-pressed translation but not acetylation and methylation

of amino acid residues in histone molecules synthe-sized before [29].

How do histone modifications affect the organiza-tion and function of chromatin? Two variants are possi-ble. For instance, addition of acetyl groups neutralizesthe positive charge of a histone molecule, while addi-tion of phosphate groups increases the negative charge.Such modifications change the conformation of nucleo-somes and the accessibility of DNA for various proteins[31–33]. According to another hypothesis, the key roleis played by a particular combination of modificationsin a nucleosome rather than by the net charge of a his-tone molecule. Occurring in a given nucleosome, sucha combination “codes” for a particular process. Thecoding mechanism is based on the assumption thatmodified residues selectively bind with certain pro-teins, which trigger a particular chain of reactions,thereby deciphering the histone code. In 2000 and2001, Strahl (United States), Allis (United States), andJenuwein (Austria) [34, 35] formulated this histonecode theory, which is rapidly developing now. Interestin this theory can be illustrated by the fact that the paper[35] was cited more than one thousand times fromAugust 2001 to December 2005. Finally, it is possiblethat the two mechanisms do not exclude each other,functioning in parallel; i.e., both the combination ofmodifications and the net charge of a chromatin regionare important.

METHYLATION

It is clear that stable histone marks are necessary forlong-term processes in chromatin, while short-term andperiodical processes require marks that are easy to setand remove. It was believed for a long time that meth-

Me Me MeMe MeAcPP Ac Ac PMeMe Me Me

Ac MeAcAcAcMe

Ac Ac AcP Ub

UbAcAcAcAc P

120201514125

1 5 9 13 119

3 5 8 12 16 20

2 4 8 9 10 14 17 18 23 26 27 28 36 79

Ac

H3

H4

H2A

H2B

Fig. 2.

Major modifications of amino acid residues in the human core histones. Residues are numbered starting from the N terminus.Modifications: Me, methylation; Ac, acetylation; P, phosphorylation; and Ub, ubiquitylation.

Page 3: Histone modification and regulation of chromatin function

972

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

KORYAKOV

ylation is stable and that such marks can be removedonly together with the whole histone molecule,although the possibility of enzymatic demethylationwas already known [36]. The concept of methylationstability arose because the attempts to isolate histonedemethylases failed; yet the situation has changedrecently [37].

Today, methylation is known in various organismsfor 17 lysine residues, which each can bear one to threemethyl groups, and for 7 arginine residues, each bear-ing one or two groups [34, 38–45] (Fig. 2). Modifica-tion of these residues plays a role in the formation ofheterochromatin, DNA repair, and transcription regula-tion; the same residue with different number of methylgroups may have different biological functions. Methy-lation is driven by numerous lysine and arginine histonemethyltransferases (HMTs). Most lysine HMTs pos-sess a catalytical SET domain and are divided into fourfamilies (SUV39, SET1, SET2, and RIZ), which differin the position of the SET domain, the substrate speci-ficity, and the presence of other domains [46–48]. Thename of the SET domain originates from the names of

Drosophila

proteins wherein it has been identified(Su(var)3-9, E(z), and Trx). Only a few enzymes meth-ylating H3K79 lack the SET domain [49].

The terminal nitrogen atoms of the side chain ofarginine differ, one belonging to the amino group andthe other, to the imino group. Thus, a dimethylated argi-nine residue can have both methyl groups at the amino-group nitrogen (asymmetrical methylation) or onemethyl group at each of the two different nitrogens(symmetrical methylation). Arginine HMTs are dividedinto two classes. Class I enzymes catalyze asymmetri-cal methylation, whereas class II enzymes are responsi-ble for symmetrical methylation [40, 50]. In both cases,S-adenosyl-L-methionine is utilized as a donor of themethyl group [51]. This compound is a universal donorfor both DNA- and protein-specific methyltransferases.Accordingly, all these enzymes contain a conserved S-adenosyl-L-methionine-binding motif regardless oftheir substrate specificity [52].

The spatial structure of the SET domain was studiedin detail for human SET7/9,

Drosophila

SET8,

Neuro-spora crassa

DIM-5,

Schizosaccharomyces pombe

Clr4, and pea LSMT (for review, see [53, 54]). The SETdomain contains two conserved regions, SET-N andSET-C, and intermediate region SET-I, which is con-served to a lesser extent [54]. The N-flanking sequenceof the domain is important for its stability, while theC-flanking region acts as a binding site for S-adenosyl-L-methionine [53].

Methyl groups are removed by histone demethy-lases (HDMs). The first HDM was recently identified inyeast

S. pombe

and designated as LSD1. This enzymedemethylates H3K4 via oxidation to yield formalde-hyde [55, 56]. Yeast

Saccharomyces cerevisiae

lacksLSD1 homologs, but has JHMD1, another HDM thatspecifically demethylates H3K36. This process is cata-

lyzed by the JmjC domain and utilizes another chemicalreaction, yielding formaldehyde and succinate [57, 58].Homologs of these demethylases were found in variousorganisms, including humans. Arginine demethylationis still poorly understood. It is known only that PAD4converts nonmodified or monomethylated arginine ofhistones H3 and H4 into citrulline via deimination[59, 60].

It is noteworthy that the enzymes known to add orremove the methyl group considerably differ in num-ber. There are 73 genes coding for putative HMTs withthe SET domain in the human genome, 41 in the

Droso-phila

genome, and up to 37 in the nematode

Cae-norhabditis elegans

genome [48]. At the same time, nomore than ten HDMs homologous to LSD1 are encodedin mammalian genomes [37]. Note, however, that pro-teins presumably possessing the JmjC domain are farmore numerous: their number reaches 52 in humans.Yet reliable data are available for only a few demethy-lases, which poses several questions. For instance, aremethylated lysine residues are all capable of demethy-lation? If so, how their demethylation is achieved sinceHDMs are few? Which factors determine the site spec-ificity of demethylation? At least the last question canbe answered. There is evidence that the specificity ofLSD1 is affected by cofactors: LSD1 demethylatesH3K4 when bound with CoREST and H3K9 whenbound with the androgen receptor [61–63].

Another feature of demethylation is also of particu-lar interest. The lysine-specific enzymes are active onlywith di- and monomethylated residues. Trimethylatedresidues are not affected by known HDMs [56, 58]. It ispossible that one or two methyl groups modifying oneresidue mark relatively stable but still reversible pro-cesses, while three methyl groups are marks of irrevers-ible processes.

It is essential for binding with methylated lysine thata protein deciphering the histone code has specificdomains, e.g., chromodomain or Tudor [64–67]. Thefirst identified domain was the chromodomain, whichconsists of approximately 50 residues forming three

β

-sheets opposite to an

α

-helix [68]. A key role in theresulting structure is played by approximately tenhydrophobic residues, which bind the side chain oflysine. To recognize the methyl group, the chromo-domain utilizes three perpendicular aromatic rings,which form a pocket cell [65].

ACETYLATION

The total number of acetylation sites in histones is13–16 according to different estimates [44, 69–72](Fig. 2). Unlike in the case of methylation, only oneacetyl group can be added to a lysine residue. Suchmolecular marks are highly labile, which is due tonumerous histone acetyltransferases (HATs) and his-tone deacetylases (HDACs). Ample evidence accumu-lated to date implicates histone acetylation in transcrip-

Page 4: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 973

tion regulation, dosage compensation, the cell cycle,and nucleosome assembly.

All HATs are divided into two large classes, depend-ing on the cell compartment where they function. TypeB enzymes function in the cytoplasm, while type AHATs are nuclear. In addition, several families andsuperfamilies of HATs are recognized by the presenceof conserved domains and association with certain pro-tein complexes (GNAT, MYST, p300/CBP, nuclearreceptor coactivators, TAFII250, and TFIIIC) [69, 70].All HATs have homologous catalytic domains. Thespatial structure and the mechanism of action of the cat-alytic domain were studied in detail for PCAF andGCN5 of various organisms. The domain contains sev-eral motifs. The greatest conservation is characteristicof motif A (R/G–x–x–G–x–G/A), which is involved inrecognizing and binding Ac-CoA, which serves as adonor of the acetyl group [73–78].

HDACs are divided into four classes based on theirhomology and nuclear or cytoplasmic localization[79–83]. Enzymes of classes I, II, and III are respec-tively homologous to

S. cerevisiae

RPD3, HDA1, andSir2. Only class III enzymes utilize NAD

+

as a cofactor[84, 85]. HDAC11 is close to class I enzymes, but isisolated in an individual class (IV) because of the lowhomology [83, 86]. Class I and II enzymes possess a cata-lytic domain that consists of approximately 390 aminoacid residues and includes Zn

2+

as an essential compo-nent [87]. In vivo, HDACs are active only in complexwith cofactors [79].

The bromodomain is necessary for recognizing acety-lated lysine in histones and other proteins [88–90]. Thisdomain is approximately 110 residues in size and con-tains four long and one short

α

-helices. About tenamino acid residues of the domain are conserved; theseresidues form a hydrophobic pocket for the binding ofthe side chain of lysine and a charged region within thepocket for hydrogen bonding with the acetyl group[91, 92]. Other residues of the domain determine itsgeneral configuration and specific binding with the sub-strate. In addition, an important role in specificity isplayed by the context of acetylated lysine. For instance,positions K + 2 and K + 3 are crucial for the binding ofthe bromodomain of

S. cerevisiae

GCN5 with acety-lated H4K16 of a histone molecule, while positions K – 1and K + 1 does not affect binding [93]. Some proteinssuch as TAFII250 and Brd4 have double bromo-domains. It is believed that these proteins are capable ofbinding simultaneously with two acetylated lysines(K5/K12 or K8/K16) in histone H4 [94, 95].

PHOSPHORYLATION

Phosphorylation is a labile modification and isknown for all histones (Fig. 2). Phosphorylation isinvolved in two structurally opposite processes: decon-densation of chromatin upon transcription activationand condensation of chromosomes during cell division

or apoptosis [96–98]. A necessary balance betweenphosphorylation and dephosphorylation is maintainedby kinases and phosphatases. Different processesinvolve different sets of enzymes. For instance phos-phorylation of histone H3 during the cell cycle is regu-lated by kinases Ipl1/Aurora and phosphatasesGlc7/PP1 [99–101]. Kinases RSK2, MSK1, and MSK2transfer phosphate groups to histone H3 to activatetranscription [102–104]. The enzyme phosphorylatingH3S10 upon heat shock in

Drosophila

is still unknown,and JIL-1 is regarded as a possible candidate [105].PP2A probably acts as phosphatase that removes phos-phate groups from all other sites during this process [106].

UBIQUITYLATION

In addition to small chemical groups, relativelylarge molecules are utilized for modifying histones. Forinstance, lysine residues can be linked with ubiquitin, apolypeptide of 76 amino acid residues [107]. This mod-ification was demonstrated for only a few positions inhistones H2A and H2B. In contrast to the overwhelm-ing majority of other modification positions, the ubiq-uitylation sites are at the C ends of molecules. HistoneH2A can be ubiquitylated at K119, and histone H2B, atK120/K123 [108–110] (Fig. 2). Ubiquitin is added orremoved by ubiquitin ligases and proteases [111].

Thus, the four types of the best-studied covalentmodifications of histones have been considered above.The following sections focus on the role of thesemolecular marks in the fundamentally differing pro-cesses of transcriptional suppression and activation.

TRANSCRIPTIONAL SUPPRESSION (SILENCING)

The common term silencing implies various pro-cesses that lead to heritable epigenetic suppression oftranscription, including the formation of pericentricheterochromatin, dosage compensation in mammals,gene inactivation in yeast

S. cerevisiae

, and regulationof homeotic genes in

Drosophila.

In general, these pro-cesses are characterized at least by the absence of labilemodification (such as acetylation), the presence of sta-ble marks (e.g., methylation of H3K9, H3K27, andH4K20), and DNA methylation. Since a lysine residuecan bear one to three methyl groups, every silent regionhas a specific combination of epigenetic marks. Suchmarks are epigenetic indeed, being inherited throughcell generations.

The main modification characterizing mammalianpericentric heterochromatin is H3K9 trimethylationcatalyzed by Suv39h1 [112]. Trimethylation of H4K20and monomethylation of H3K27 are two other modifi-cations that prevail in these chromosome regions. WhenH3K9 trimethylation is lacking, a trimethylated, ratherthan monomethylated, form of H3K27 accumulatesand H4K20 trimethylation disappears [113, 114].

Page 5: Histone modification and regulation of chromatin function

974

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

KORYAKOV

Unlike in mammals, the main epigenetic mark of

Drosophila

pericentric heterochromatin is H3K9 dime-thylation catalized by Su(var)3-9, which is homologousto Suv39h1 [115, 116]. Trimethylated H3K9 andH3K27 are few in

Drosophila

heterochromatin, whilemonomethylated and dimethylated H3K27 residues arerelatively abundant [117].

A specific set of epigenetic marks is generated in themammalian X chromosome upon its inactivation by adosage compensation system [118]. Inactivation is anintricate process, and its initiation and maintenancerequire different marks. During initiation, synthesis ofthe

Xist

RNA stimulates trimethylation of H3K27 andmonomethylation of H4K20, but these marks do notlead to genetic inactivation [119–121]. Transcriptionalsilencing results from dimethylation of H3K9 [122–125]. H3K9-specific HMTs functioning in pericentricheterochromatin and in the X chromosome differ andare not interchangeable. The X-chromosomal enzymeis G9a [124–126]. In addition, important features of theinactive X chromosome are DNA methylation [127]and histone hypoacetylation [128].

The process of H3K9 methylation and its conse-quences have been studied in detail in various organ-isms. It was believed until recently that H3K9 is theonly residue whose acetyl group can be replaced bymethyl groups, while other lysine residues can bearonly one of these modifications. In

Drosophila,

acety-lated H3K9 is deacetylated by RPD3, which provides asignal for methylation by Su(var)3-9, which functionsin complex with RPD3 [129] (Figs. 3a–3d). A similarsituation was observed for

S. pombe

: methylation of

H3K9 by Clr4 is preceded by its deacetylation by Clr6[130, 131]. It is possible that

Drosophila

Su(var)3-9and HDAC1 are not the only enzymes involved in theprocess, because Su(var)3-9 is responsible for approxi-mately 80% of dimethylation and, probably, for all100% of trimethylation. The presence of a dimethy-lated form in chromosome 4 and monomethylation areindependent of this HMT, suggesting the presence ofanother H3K9-specific enzyme (or two enzymes) in

Drosophila

[116, 117].

After this, methylated H3K9 is recognized by vari-ous proteins that trigger processes associated withsilencing. For instance, the Pdd1 and Pdd3 proteinsinteract with methylated H3K9 and trigger chromatindiminution in

Tetrahymena

[132]. The interaction withH3K9 has been studied in most detail for heterochro-matin protein 1 (HP1), which, in turn, is capable ofrecruiting many other proteins [65, 130, 133–136]. Forinstance, HP1 binding to H3K9 is essential for therecruitment of H4K20-specific HMT Suv4-20 andH4K20 methylation in mammals and

Drosophila

[114](Figs. 3e–3i). There is ample evidence that HP1 corre-lates with genetic inactivation [137–140]. However, theprocesses involving HP1 are greatly diverse and can beassociated with transcription as well [141–146].

Methylation of H3K9 is necessary for the recruit-ment of DNA methyltransferases. This process is medi-ated by HP1 or other proteins binding to H3K9 [147–155]. An opposite dependence has also been observed[156, 157]. Methylated cytosines occurring in DNAbind with proteins having the methyl-binding domain(MBD). In turn, these proteins recruit HDACs and

HDACRPD3

HMT ?

HMTSu(var)3-9

HMTSET8

HMT ?

HP1

HMTSuv4-20

HP1

H4K20

H4

H4

H4 H3

H3

H3

H3

H3

H3 (a)

(b)

(c)

(d)

(e)

(f)

(g)

(h)

(i)

Fig. 3.

Processes occurring during the formation of pericentric heterochromatin in

Drosophila.

Filled bars with horizontal lines, thecore regions and N tails of histones H3 and H4. Black squares show one, two, or three methyl groups at H3K9 and H4K20 (c–f, h, i);gray triangles show the acetyl group at H3K9 (a, b). See text for comments.

H3K9

Page 6: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 975

HMTs for transferring the methyl group to H3K9.Moreover, some HMTs (e.g., ESET and CLLL8) har-bor MBD and, possibly, are capable of binding directlyto DNA [48, 158, 159]. Such a bi-directional interac-tion probably serves for memorization of the histonemodification pattern during DNA replication andassembly of new nucleosomes, thereby playing animportant role in the inheritance of epigenetic marks.

Methylation of H3K27 is involved in another inacti-vation system. As with H3K9-dependent repression,the relevant system is conserved and has many commonfeatures in different organisms such as

Drosopila

andmammals, although some differences are also known[160, 161]. The system involved about two dozens ofproteins, which belong to the Polycomb group (PcG).The PcG proteins form individual interacting com-plexes. Two such complexes, PRC1 and PRC2 (Poly-comb repressive complex) are known in

Drosophila

,and four (PRC1–PRC4), in mammals.

Drosophila

PRC1 includes proteins that recognize specific DNAsequences known as PcG response elements (PREs)and determine the inactivation site. Note that PREshave so far been identified in

Drosophila

but not inmammals. In addition, PRC1 includes the Polycombprotein (PC) in

Drosophila

and its homolog HPC inmammals [162–165]. PCR2 possesses histone methyl-transferase activity due to a specific subunit, which isknown as E(z) in

Drosophila

and EZH2 in humans[166–168]. In addition to PRC2, two other human com-plexes, PRC3 and PRC4, contain EZH2 and, conse-quently, are capable of methylating lysine residues inhistones. The site specificity of the complexes differsand depends mostly on the context of the target lysine.For instance, E(z) contained in

Drosophila

PRC2 meth-ylates mostly H3K27 and, to a lesser extent, H3K9in vitro [167]. These residues belong to the commonmotif ARKS. In vivo, a lack of E(z) affects the propor-tion of methylated H3K27, but not H3K9 [117]. As acomponent of PRC2, human EZH2 methylates bothH3K27 and H1K26, the latter also belonging to theARKS motif.

Drosophila

histone H1 is not methylatedby E(z), which can be explained by the lack of thismotif. As a component of PRC3, EZH2 methylates onlyH3K27 and only in the absence of histone H1 [169].PRC4 contains not only EZH2, but also NAD

+

-depen-dent deacetylase SirT1, and is possibly capable todeacetylate and then methylate H1K26 [169–171].

The exact function has so far been established foronly some components involved in PC-dependentsilencing. To simplify, this process can be reduced toH3K27 methylation by E(z) contained in PRC2. In

Drosophila

, methylation affects nucleosomes that sur-round PREs. Modified H3K27 binds with PRC1through the chromodomain of the PC protein [172,173]. This interaction is necessary for Ring1/ESC, aPCR1 component that has a ubiquitin ligase activityand is capable of modifying H2AK119 [174, 175]. Inaddition, EZH2 contained in PCR2/3 interacts withDNA methyltransferases, which methylate DNA in

PRC-binding sites. This possibly ensures the epigeneticinheritance of PC-dependent repression [176].

The

S. cerevisiae

silencing system is of particularinterest, since it substantially differs from the corre-sponding systems of other organisms. Pericentric het-erochromatin is absent from

S. cerevisiae

; its analogsare mating-type loci, rRNA genes, and telomeres [177].There is no H3K9-specific HMT, H3K9 methylation,and HP1 homologs in

S. cerevisiae

lacks [178]. More-over, H3K27 can be acetylated but not methylated in

S. cerevisiae

, while the situation is opposite in otherorganisms. The

S. cerevisiae

silencing system employsproteins known as silent information regulators (Sir1–Sir4) [179, 180]. Like with the

Drosophila

PcG pro-teins, their binding requires certain DNA sequencesknown as silencers. All four Sir proteins function tosilence the mating-type loci, while telomere silencingdoes not require Sir1 [181]. A lack of Sir3 and Sir4 doesnot affect silencing of the rRNA gene cluster [182].Thus, only Sir2, acting as histone deacetylase, func-tions in all silent regions [183]. Sir2 is a class IIIenzyme, while deacetylases involved in silencing in

Drosophila

and

S. pombe

belong to class I. Sir2deacetylates H4K16, which leads to subsequentdeacetylation of other lysine residues. Deacetylation,rather than methylation, of lysine residues is the mainprerequisite to transcriptional suppression of all silentregions in

S. cerevisiae

as opposed to other organisms.In telomeres and mating-type loci, deacetylated H4binds a complex containing Sir3, which recruits newmolecules of Sir2 and Sir4 and thereby causes a spread-ing of silencing [184, 185].

TRANSCRIPTION

Transcription is a multistep process and includes ini-tiation, elongation, and termination, which are controlledby different protein complexes. The situation is evenmore intricate, since genes differ in the pattern of theirexpression. For instance, housekeeping genes are alwaysactive in all tissues. The heat shock genes are potentiallyactive: they may never function and can be rapidly acti-vated in response to certain environmental factors. Geneslocated in heterochromatin require a specific regulatorysystem, differing from that of euchromatic genes. Thus,the intricate process of transcription requires an intricatesystem of marks, involving acetylation, methylation,phosphorylation, and ubiquitylation. Modificationsshould be intensely changed during transcription, andsuch changes should be initiated anew in every cell gen-eration. Thus, the marks that regulate transcription can-not be regarded as epigenetic. The process has been stud-ied best in

S. cerevisiae

; however, the regulatory modifi-cations are much the same in different organisms onexperimental evidence.

Transcription initiation in yeast

S. cerevisiae

andhuman cells require acetylation of H3K9, H3K14, andhistone H4 and nucleosome remodeling in the promoterand 5'-terminal regions of a gene. Such modifications

Page 7: Histone modification and regulation of chromatin function

976

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

KORYAKOV

are far fewer in the coding region [186, 187]. Acetyla-tion was effected by acetyltransferase complexes, e.g.,GCN5 and ESA1, the highest number of which was alsoat the promoter region [188, 189]. It is important to notethat the proportion of acetylated nucleosomes is maximalin the 5'-coding region rather than in the promoter. Thisagrees with the fact that nucleosomes are virtually absentfrom the promoters of active genes [187, 190, 191].

The group of immediate-early response genesrequires a special initiation system. These genes lack acertain temporal expression profile and are rapidly acti-vated in response to environmental stimuli such as hor-mones, protein synthesis inhibitors, heat shock, andirradiation. Their activation involves phosphorylationof H3S10. This modification appears early after expo-sure to an external stimulus and is short-lived.

In

S. cerevisiae

and mammals, H3S10 modificationcorrelates with hyperacetylation of the same nucleo-some, suggesting a co-operation of these modifications[192–194]. Two hypotheses have been advanced toexplain the role of phosphorylation in activation. Accord-ing to one hypothesis, phosphorylation is an essential pre-requisite to subsequent acetylation. In

S. cerevisiae

, SNF1kinase functions together with GCN5 phosphorylase onthe

INO1

promoter and phosphorylates H3S10, whichprovides a binding site for GCN5 for subsequent acety-lation of H3K14 [195, 196]. Other data suggest thatacetylation and phosphorylation act independently. Forinstance, activation of immediate-early response genesin cultured mammalian cells is accompanied by H3phosphorylation but does not increase the extent of his-tone acetylation [104, 197]. Heat shock leads to a com-plete redistribution of H3S10 phosphorylation sites onpolytene chromosomes in

Drosophila

larvae. At a nor-mal temperature, this modification is present in allactively transcribed regions. As temperature increases,activation of the heat shock genes is accompanied by denovo phosphorylation of nucleosomes associated withtheir loci, while this modification disappears from allother regions of the genome. These changes are notaccompanied by any changes in the extent of acetyla-tion of H3K14 and H4K8 [198].

The transition from transcription initiation to elon-gation is associated with methylation of H3K4, whichcan bear one to three methyl groups. The distribution ofdi- and monomethylated forms of H3K4 through chro-matin is relatively homogeneous, while a peak of trim-ethylated H3K4 corresponds to the promoter and 5'-ter-minal regions of active genes in

S. cerevisiae,

human,mouse, and chicken cells, clearly distinguishing thesegenes from inactive genes [187, 199–201]. The firstmethyl group is added by HMT homologous to mam-malian Set9 [202]; the second and third groups areadded by an enzyme homologous to yeast Set1 [178,203, 204]. Phosphorylation of RNA polymerase II bythe TFIIH complex is an essential prerequisite to Set1binding to chromatin in

S. cerevisiae

and, consequently,trimethylation of H3K4. This chain of events results in

elongation; trimethylation of H3K4 in the promoterregion persists for a long time, providing a molecularmark of recent transcriptional activity and, possibly,playing an important role in reinitiation of transcription[205]. It cannot be excluded that trimethylation ofH3K4 in the 5' region of a gene is involved in RNA pro-cessing. Set1 contains an RNA-recognizing motif andis associated with a multisubunit complex that containsthe Swd2 protein. In turn, Swd2 is involved in matura-tion of the 3' end of newly synthesized RNA [203].

There is evidence that ubiquitylation is necessaryfor transcriptional activation. In

S. cerevisiae

cells, acti-vation of the

GAL1

and

SUC2

genes is accompanied byubiquitylation of H2BK123 in nucleosomes of the pro-moter regions. This modification is short-lived andserves to recruit the SAGA acetyltransferase complex.The complex contains the Ubp8 protein, whichremoves ubiquitin residues [206]. In addition, ubiquity-lation is associated with methylation of H3K4 andH3K79. Modification of the latter correlates with activetranscription, although its role in the process is stillunclear. Human UbcH6 and hBRE1 ubiquitylateH2BK120, which is essential for methylation of K4 andK79 and expression of some genes. Suppression ofubiquitin-binding proteins exerts an opposite effect[207, 208]. In

S. cerevisiae

, H2BK123 ubiquitylation isimportant for addition of the second and third methylgroups to H3K4 and H3K79, rather than for monome-thylation of these residues [209].

Transcription elongation is accompanied by methy-lation of K36 in the core region of histone H3 in yeast

S. cerevisiae

and chicken cells. Unlike with H3K4, apeak of di- and trimethylated forms of H3K36 is in the3' part of the coding region rather than in the promoterregion of a gene [187, 210]. This modification dependson Set2, which is also detectable only in the codingregions of genes [211, 212]. Like with H3K4, methyla-tion of H3K26 requires phosphorylation of RNA poly-merase II, but at another site. Set2 contains the specificSRI (Set2 Rpb1 interacting) domain, which binds onlywith phosphorylated RNA polymerase [212–215]. Amutation of the SRI domain of Set2 or a lack of phos-phorylation of RNA polymerase blocks the interactionof the two enzymes, prevents methylation of H3K36,and eventually distorts elongation. During elongationin

S. cerevisiae

, methylated H3K36 binds with a com-plex containing RPD3 histone deacetylase. This enzymeremoves acetyl groups from the sequence that hasalready been transcribed, which possibly prevents tran-scription initiation on cryptic promoters located withinthe coding region [216, 217]. Note that, in

S. cerevisiae

,H3K36 methylation is independent of H2B ubiquityla-tion, while methylation of H3K4 and H3K79 is associ-ated with this modification [218].

CONCLUSIONS

Studies with model subjects of various taxonomicgroups have shown that histone modification is a uni-

Page 8: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS

Vol. 42

No. 9

2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 977

versal system regulating the function of chromatin. Dif-ferent organisms have common principles of the his-tone code, and homologous enzymes modify the sameresidues in most cases. Yet every organism has its spe-cifics, as is the case with gene silencing in

S. cerevisiae.

Moreover, dinoflagellates (

Dinophyta

) provide anexception to eukaryotic rules. These unicellular organ-isms lack nucleosomes, histones, and their modifica-tions in their common form. The packing of dinoflagel-late chromosomes involves proteins that have nohomologs in other organisms. The histone–DNAweight ratio is approximately 1 : 1 in the eukaryoticnucleus, while histone-like proteins amount to only10% of the DNA weight in dinoflagellates [219, 220].This exception indicates that, universal as it is, the his-tone code is not the only system regulating the functionof chromatin.

As evident from ample data, the histone code isintricate and various modifications are involved in hier-archic interactions known as cross-talk. Some chemicalmodifications always correlate with each other, whilesome others are antagonistic [44, 221]. Moreover, thehistone code is to an extent multivalued. A modificationcharacteristic of heterochromatin can play a role ingene activation in some cases. A modification involvedin transcription can be involved in transcriptional sup-pression under certain conditions. For instance, Ash1(HMT) is capable of methylating simultaneouslyH3K4, H3K9, and H4K20. On genetic evidence, thefunction of the ash1 gene is necessary for activation ofthe Ubx gene in imaginal disks of the third leg pair inDrosophila [222]. This activation requires simulta-neous methylation of the three lysine residues innucleosomes located in the Ubx promoter and a conse-quent recruitment of the Brm transcriptional activator[223]. Thus, the modifications that are usually alterna-tive occur together in this case. Although H3K4 methy-lation is necessary for the transition from transcriptioninitiation to elongation in S. cerevisiae, this modifica-tion causes silencing in the ribosomal gene cluster andtelomeres [178, 224]. Thus, combinations of modifica-tions, rather than individual modifications alone, gov-ern the function of chromatin, and the role of a particu-lar modification depends on its context.

ACKNOWLEDGMENTS

I am grateful to A.A. Gorchakov and T.D. Kolesnikovafor help in searching for literature sources, advice, andfruitful discussion.

This work was supported by the program “Molecu-lar and Cell Biology” of the Presidium of the RussianAcademy of Sciences (project no. 10.1), the Interdisci-plinary Integration Project of the Siberian Division ofthe Russian Academy of Sciences (no. 45), and the Rus-sian Foundation for Basic Research (project no. 06-04-48387a).

REFERENCES1. Kornberg, R.D., Chromatin Structure: A Repeating Unit

of Histones and DNA, Science, 1974, vol. 184, pp. 868–871.

2. Oudet, P., Gross-Bellard, M., and Chambon, P., Elec-tron Microscopic and Biochemical Evidence ThatChromatin Structure Is a Repeating Unit, Cell (Cam-bridge, Mass.), 1975, vol. 4, pp. 281–300.

3. Finch, J.T., Lutter, L.C., Rhodes, D., et al., Structure ofNucleosome Core Particles of Chromatin, Nature,1977, vol. 269, pp. 29–36.

4. Thomas, J.O. and Kornberg, R.D., An Octamer of His-tones in Chromatin and Free in Solution, Proc. Natl.Acad. Sci. USA, 1975, vol. 72, pp. 2626–2630.

5. Klug, A., Rhodes, D., Smith, J., et al., A Low ResolutionStructure for the Histone Core of the Nucleosome,Nature, 1980, vol. 287, pp. 509–516.

6. Arents, G., Burlingame, R.W., Wang, B., et al., TheNucleosomal Core Histone Octamer at 3.1 Å Resolu-tion: A Tripartite Protein Assembly and a Left-HandedSuperhelix, Proc. Natl. Acad. Sci. USA, 1991, vol. 88,pp. 10 148–10 152.

7. Lilley, D.M.J., Pardon, J.F., and Richards, B.M., Struc-tural Investigations of Chromatin Core Protein by NuclearMagnetic Resonance, Biochemistry, 1977, vol. 16,pp. 2853–2860.

8. Luger, K., Mader, A.W., Richmond, R.K., et al., CrystalStructure of the Nucleosome Core Particle at 2.8 Å Res-olution, Nature, 1997, vol. 389, pp. 251–260.

9. Varshavsky, A.J., Bakayev, V.V., and Georgiev, G.P.,Heterogeneity of Chromatin Subunits In vitro and Loca-tion of Histone H1, Nucleic Acids Res., 1976, vol. 3,pp. 477–492.

10. Hayashi, K., Hofstaetter, T., and Yakuwa, N., Asymme-try of Chromatin Subunits Probed with Histone H1 in anH1–DNA Complex, Biochemistry, 1978, vol. 17,pp. 1880–1883.

11. Simpson, R.T., Structure of the Chromatosome, a Chro-matin Particle Containing 160 Base Pairs of DNA andAll the Histones, Biochemistry, 1978, vol. 17, pp. 5524–5531.

12. Sperling, J. and Sperling, R., Photochemical Cross-Linking of Histones to DNA Nucleosomes, NucleicAcids Res., 1978, vol. 5, pp. 2755–2773.

13. Thoma, F., Koller, T.H., and Klug, A., Involvement ofHistone H1 in the Organization of the Nucleosome andof the Salt-Dependent Superstructures of Chromatin, J.Cell Biol., 1979, vol. 83, pp. 403–427.

14. Allan, J., Hartman, P.G., Crane-Robinson, C., andAviles, F.X., The Structure of Histone H1 and Its Loca-tion in Chromatin, Nature, 1980, vol. 288, pp. 675–679.

15. Appels, R. and Wells, J.R., Synthesis and Turnover ofDNA-Bound Histone during Maturation of Avian RedBlood Cells, J. Mol. Biol., 1972, vol. 70, pp. 425–434.

16. Hewish, D.R. and Burgoyne, L.A., Chromatin Sub-Structure: The Digestion of Chromatin DNA at Regu-larly Spaced Sites by a Nuclear Deoxyribonuclease,Biochem. Biophys. Res. Communs., 1973, vol. 52,pp. 504–510.

17. Noll, M., Internal Structure of the Chromatin Subunit,Nucleic Acids Res., 1974, vol. 1, pp. 1573–1578.

Page 9: Histone modification and regulation of chromatin function

978

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

KORYAKOV

18. Mirzabekov, A.D., Shick, V.V., Belyavsky, A.V., andBavykin, S.G., Primary Organization of NucleosomeCore Particle of Chromatin: Sequence of HistoneArrangement along DNA, Proc. Natl. Acad. Sci. USA,1978, vol. 75, pp. 4184–4188.

19. Lutter, L.C., Precise Location of DNase I Cutting Sitesin the Nucleosome Core Determined by High Resolu-tion Gel Electrophoresis, Nucleic Acids Res., 1979, vol. 6,pp. 41–56.

20. DeLange, R.J., Fambrough, D.M., Smith, E.L., andBonner, J., Calf and Pea Histone IV: II. The CompleteAmino Acid Sequence of Calf Thymus Histone IV;Presence of ε-N-Acetyllysine, J. Biol. Chem., 1969,vol. 244, pp. 319–334.

21. DeLange, R.J., Fambrough, D.M., Smith, E.L., andBonner, J., Calf and Pea Histone IV: III. CompleteAmino Acid Sequence of Pea Seedling Histone IV;Comparison with the Homologous Calf Thymus His-tone, J. Biol. Chem., 1969, vol. 244, pp. 5664–5679.

22. Sullivan, S., Sink, D.W., Trout, K.L., et al., The HistoneDatabase, Nucleic Acid Res., 2002, vol. 30, pp. 341–342.

23. Thatcher, T.H. and Gorovsky, M.A., Phylogenetic Anal-ysis of the Core Histones H2A, H2B, H3, and H4,Nucleic Acids Res., 1994, vol. 22, pp. 2174–2179.

24. Redon, C., Pilch, D., Rogakou, E., et al., Histone H2AVariants H2AX and H2AZ, Curr. Opin. Gen. Dev.,2002, vol. 12, pp. 162–169.

25. Henikoff, S. and Ahmad, K., Assembly of Variant His-tones into Chromatin, Annu. Rev. Cell. Dev. Biol., 2005,vol. 21, pp. 133–153.

26. Kamakaka, R.T. and Biggins, S., Histone Variants:Deviants?, Genes Dev., 2005, vol. 19, pp. 295–310.

27. Pusarla, R. and Bhargava, P., Histones in FunctionalDiversification: Core Histone Variants, FEBS J., 2005,vol. 272, pp. 5149–5168.

28. Phillips, D.M.P., The Presence of Acetyl Groups in His-tones, Biochem. J., 1963, vol. 87, pp. 258–263.

29. Allfrey, V.G., Faulkner, R., and Mirsky, A.E., Acetyla-tion and Methylation of Histones and Their PossibleRole in the Regulation of RNA Synthesis, Proc. Natl.Acad. Sci. USA, 1964, vol. 51, pp. 786–794.

30. Murray, K., The Occurrence of ε-N-Methyl Lysine inHistones, Biochemistry, 1964, vol. 3, pp. 10–15.

31. Roth, S.Y. and Allis, C.D., Chromatin Condensation:Does Histone H1 Dephosphorylation Play a Role?,Trends Biochem. Sci., 1992, vol. 17, pp. 93–98.

32. Tse, C., Sera, T., Wolffe, A.P., and Hansen, J.C., Disrup-tion of Higher-Order Folding by Core Histone Acetyla-tion Dramatically Enhances Transcription of Nucleoso-mal Arrays by RNA Polymerase III, Mol. Cell. Biol.,1998, vol. 18, pp. 4629–4638.

33. Horn, P.J. and Peterson, C.L., Chromatin Higher OrderFolding-Wrapping up Transcription, Science, 2002,vol. 297, pp. 1824–1827.

34. Strahl, B.D. and Allis, C.D., The Language of CovalentHistone Modifications, Nature, 2000, vol. 403, pp. 41–45.

35. Jenuwein, T. and Allis, C.D., Translating the HistoneCode, Science, 2001, vol. 293, pp. 1074–1080.

36. Paik, W.K. and Kim, S., Enzymatic Demethylation ofCalf Thymus Histones, Biochem. Biophys. Res. Com-mun., 1973, vol. 51, pp. 781–788.

37. Kubicek, S. and Jenuwein, T., A Crack in HistoneLysine Methylation, Cell (Cambridge, Mass.), 2004,vol. 119, pp. 903–906.

38. Rice, J.C. and Allis, C.D., Histone Methylation VersusHistone Acetylation: New Insights into Epigenetic Reg-ulation, Curr. Opin. Cell Biol., 2001, vol. 13, pp. 263–273.

39. Stallcup, M.R., Role of Protein Methylation in Chroma-tin Remodeling and Transcriptional Regulation, Onco-gene, 2001, vol. 20, pp. 3014–3020.

40. Zhang, Y. and Reinberg, D., Transcription Regulationby Histone Methylation: Interplay between DifferentCovalent Modifications of the Core Histone Tails,Genes Dev., 2001, vol. 15, pp. 2343–2360.

41. Lachner, M., O’Sullivan, R.J., and Jenuwein, T., AnEpigenetic Road Map for Histone Lysine Methylation,J. Cell Sci., 2003, vol. 116, pp. 2117–2124.

42. Sims, R.J. III, Nishioka, K., and Reinberg, D., HistoneLysine Methylation: A Signature for Chromatin Func-tion, Trends Genet., 2003, vol. 19, pp. 629–639.

43. Zhang, L., Eugeni, E.E., Parthun, M.R., and Freitas, M.A.,Identification of Novel Histone Post-TranslationalModifications by Peptide Mass Fingerprinting, Chro-mosoma, 2003, vol. 112, pp. 77–86.

44. Margueron, R., Trojer, P., and Reinberg, D., The Key toDevelopment: Interpreting the Histone Code?, Curr.Opin. Genet. Dev., 2005, vol. 15, pp. 163–176.

45. Bannister, A.J. and Kouzarides, T., Reversing HistoneMethylation, Nature, 2005, vol. 436, pp. 1103–1106.

46. Jenuwein, T., Laible, G., Dorn, R., and Reuter, G., SETDomain Proteins Modulate Chromatin Domains in Eu-and Heterochromatin, Cell. Mol. Life Sci., 1998, vol. 54,pp. 80–93.

47. Alvarez-Venegasa, R. and Avramova, Z., SET-DomainProteins of the Su(var)3-9, E(z) and Trithorax Families,Gene, 2002, vol. 285, pp. 25–37.

48. Kouzarides, T., Histone Methylation in TranscriptionalControl, Curr. Opin. Genet. Dev., 2002, vol. 12,pp. 198–209.

49. Feng, Q., Wang, H., Ng, H.H., et al., Methylation of H3-Lysine 79 Is Mediated by a New Family of HMTaseswithout a SET Domain, Curr. Biol., 2002, vol. 12,pp. 1052–1058.

50. McBride, A.E. and Silver, P.A., State of the Arg: ProteinMethylation at Arginine Comes of Age, Cell (Cam-bridge, Mass.), 2001, vol. 106, pp. 5–8.

51. Schluckebier, G., O’Gara, M., Saenger, W., and Cheng, X.,Universal Catalytic Domain Structure of AdoMet-Dependent Methyltransferases, J. Mol. Biol., 1995,vol. 247, pp. 16–20.

52. Niewmierzycka, A. and Clarke, S., S-Adenosylme-thionine-Dependent Methylation in Saccharomycescerevisiae: Identification of a Novel Protein ArginineMethyltransferase, J. Biol. Chem., 1999, vol. 274,pp. 814–824.

53. Xiao, B., Wilson, J.R., and Gamblin, S.J., SET Domainsand Histone Methylation, Curr. Opin. Struct. Biol.,2003, vol. 13, pp. 699–705.

Page 10: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 979

54. Marmorstein, R., Structure of SET Domain Proteins: ANew Twist on Histone Methylation, Trends Biochem.Sci., 2003, vol. 28, pp. 59–62.

55. Shi, Y., Lan, F., Matson, C., et al., Histone Demethyla-tion Mediated by the Nuclear Amine Oxidase HomologLSD1, Cell (Cambridge, Mass.), 2004, vol. 119,pp. 941–953.

56. Wysocka, J., Milne, T.A., and Allis, C.D., Taking LSD1to a New High, Cell (Cambridge, Mass.), 2005, vol. 122,pp. 654–658.

57. Trewick, S.C., McLaughlin, P.J., and Allshire, R.C.,Methylation: Lost in Hydroxylation?, EMBO Rep.,2005, vol. 6, pp. 315–320.

58. Tsukada, Y., Fang, J., Erdjument-Bromage, H., et al.,Histone Demethylation by a Family of JmjC Domain-Containing Proteins, Nature, 2006, vol. 439, pp. 811–816.

59. Wang, Y., Wysocka, J., Sayegh, J., et al., Human PAD4Regulates Histone Arginine Methylation Levels ViaDemethylimination, Science, 2004, vol. 306, pp. 279–283.

60. Cuthbert, G.L., Daujat, S., Snowden, A.W., et al., His-tone Deimination Antagonizes Arginine Methylation,Cell (Cambridge, Mass.), 2004, vol. 118, pp. 545–553.

61. Lee, M.G., Wynder, C., Cooch, N., and Shiekhattar, R.,An Essential Role for CoREST in Nucleosomal Histone3 Lysine 4 Demethylation, Nature, 2005, vol. 437,pp. 432–435.

62. Metzger, E., Wissmann, M., Yin, N., et al., LSD1 Dem-ethylates Repressive Histone Marks to Promote Andro-gen-Receptor-Dependent Transcription, Nature, 2005,vol. 437, pp. 436–439.

63. Shi, Y.J., Matson, C., Lan, F., et al., Regulation of LSD1Histone Demethylase Activity by Its Associated Fac-tors, Mol. Cell, 2005, vol. 19, pp. 857–864.

64. Eissenberg, J.C., Molecular Biology of the ChromoDomain: An Ancient Chromatin Module Comes of Age,Gene, 2001, vol. 275, pp. 19–29.

65. Nielsen, P.R., Nietlispach, D., Mott, H.R., et al., Struc-ture of the HP1 Chromodomain Bound to Histone H3Methylated at Lysine 9, Nature, 2002, vol. 416,pp. 103–107.

66. Huyen, Y., Zgheib, O., Di Tullio, R.A., Jr., et al., Meth-ylated Lysine 79 of Histone H3 Targets 53BP1 to DNADouble-Strand Breaks, Nature, 2004, vol. 432, pp. 406–411.

67. Sanders, S.L., Portoso, M., Mata, J., et al., Methylationof Histone H4 Lysine 20 Controls Recruitment of Crb2to Sites of DNA Damage, Cell (Cambridge, Mass.),2004, vol. 119, pp. 603–614.

68. Ball, L.J., Murzina, N.V., Broadhurst, R.W., et al.,Structure of the Chromatin Binding (Chromo) Domainfrom Mouse Modifier Protein 1, EMBO J., 1997, vol. 16,pp. 2473–2481.

69. Sterner, D.E. and Berger, S.L., Acetylation of Histonesand Transcription-Related Factors, Microbiol. Mol.Biol. Rev., 2000, vol. 64, pp. 435–459.

70. Roth, S.Y., Denu, J.M., and Allis, C.D., Histone Acetyl-transferases, Annu. Rev. Biochem., 2001, vol. 70,pp. 81–120.

71. Turner, B.M., Histone Acetylation and an EpigeneticCode, BioEssays, 2000, vol. 22, pp. 836–845.

72. Turner, B.M., Cellular Memory and the Histone Code,Cell (Cambridge, Mass.), 2002, vol. 111, pp. 285–291.

73. Dutnall, R.N., Tafrov, S.T., Sternglanz, R., andRamakrishnan, V., Structure of the Histone Acetyltrans-ferase Hat1: A Paradigm for the GCN5-Related N-Acetyltransferase Superfamily, Cell (Cambridge,Mass.), 1998, vol. 94, pp. 427–438.

74. Wolf, E., Vassilev, A., Makino, Y., et al., Crystal Struc-ture of a GCN5-Related N-Acetyltransferase: Serratiamarcescens Aminoglycoside 3-N-Acetyltransferase,Cell (Cambridge, Mass.), 1998, vol. 94, pp. 439–449.

75. Clements, A., Rojas, J.R., Trievel, R.C., et al., CrystalStructure of the Histone Acetyltransferase Domain ofthe Human PCAF Transcriptional Regulator Bound toCoenzyme A, EMBO J., 1999, vol. 18, pp. 3521–3532.

76. Lin, Y., Fletcher, C.M., Zhou, J., et al., Solution Struc-ture of the Catalytic Domain of GCN5 Histone Acetyl-transferase Bound to Coenzyme A, Nature, 1999,vol. 400, pp. 86–89.

77. Rojas, J.R., Trievel, R.C., Zhou, J., et al., Structure ofTetrahymena GCN5 Bound to Coenzyme A and a His-tone H3 Peptide, Nature, 1999, vol. 401, pp. 93–98.

78. Trievel, R.C., Rojas, J.R., Sterner, D.E., et al., CrystalStructure and Mechanism of Histone Acetylation of theYeast GCN5 Transcriptional Coactivator, Proc. Natl.Acad. Sci. USA, 1999, vol. 96, pp. 8931–8936.

79. de Ruijter, A.J.M., van Gennip, A.H., Caron, H.N., et al.,Histone Deacetylases (HDACs): Characterization of theClassical HDAC Family, Biochem. J., 2003, vol. 370,pp. 737–749.

80. Verdin, E., Dequiedt, F., and Kasler, H.G., Class II His-tone Deacetylases: Versatile Regulator, Trends Genet.,2003, vol. 19, pp. 286–293.

81. Blander, G. and Guarente, L., The Sir2 Family of Pro-tein Deacetylases, Annu. Rev. Biochem., 2004, vol. 73,pp. 417–435.

82. Hisahara, S., Chiba, S., Matsumoto, H., and Horio, Y.,Transcriptional Regulation of Neuronal Genes and ItsEffect on Neural Functions: NAD-Dependent HistoneDeacetylase SIRT1 (Sir2), J. Pharmacol. Sci., 2005,vol. 98, pp. 200–204.

83. Yang, X. and Gregoire, S., Class II Histone Deacety-lases: From Sequence to Function, Regulation, andClinical Implication, Mol. Cell. Biol., 2005, vol. 25,pp. 2873–2884.

84. Bjerling, P., Silverstein, R.A., Thon, G., et al., Func-tional Divergence between Histone Deacetylases in Fis-sion Yeast by Distinct Cellular Localization and In vivoSpecificity, Mol. Cell. Biol., 2002, vol. 22, pp. 2170–2181.

85. Imai, S., Armstrong, C.M., Kaeberlein, M., andGuarente, L., Transcriptional Silencing and LongevityProtein Sir2 Is an NAD-Dependent Histone Deacety-lase, Nature, 2000, vol. 403, pp. 795–800.

86. Gao, L., Cueto, M.A., Asselbergs, F., and Atadja, P.,Cloning and Functional Characterization of HDAC11, aNovel Member of the Human Histone DeacetylaseFamily, J. Biol. Chem., 2002, vol. 277, pp. 25 748–25 755.

Page 11: Histone modification and regulation of chromatin function

980

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

KORYAKOV

87. Finnin, M.S., Donigian, J.R., Cohen, A., et al., Struc-tures of a Histone Deacetylase Homologue Bound to theTSA and SAHA Inhibitors, Nature, 1999, vol. 401,pp. 188–193.

88. Dhalluin, C., Carlson, J.E., Zeng, L., et al., Structureand Ligand of a Histone Acetyltransferase Bromo-domain, Nature, 1999, vol. 399, pp. 491–496.

89. Ornaghi, P., Ballario, P., Lena, A.M., et al., The Bromo-domain of GCN5p Interacts In vitro with Specific Res-idues in the N Terminus of Histone H4, J. Mol. Biol.,1999, vol. 287, pp. 1–7.

90. Winston, F. and Allis, C.D., The Bromodomain: AChromatin-Targeting Module?, Nat. Struct. Biol., 1999,vol. 6, pp. 601–604.

91. Marmorstein, R. and Berger, S.L., Structure and Func-tion of Bromodomains in Chromatin-Regulating Com-plexes, Gene, 2001, vol. 272, pp. 1–9.

92. Zeng, L. and Zhou, M., Bromodomain: An Acetyl-Lysine Binding Domain, FEBS Lett., 2002, vol. 513,pp. 124–128.

93. Owen, D.J., Ornaghi, P., Yang, J.C., et al., The Struc-tural Basis for the Recognition of Acetylated HistoneH4 by the Bromodomain of Histone AcetyltransferaseGCN5p, EMBO J., 2000, vol. 19, pp. 6141–6149.

94. Jacobson, R.H., Ladurner, A.G., King, D.S., and Tjian, R.,Structure and Function of a Human TAFII250 DoubleBromodomain Module, Science, 2000, vol. 288,pp. 1422–1425.

95. Dey, A., Chitsaz, F., Abbasi, A., et al., The Double Bro-modomain Protein Brd4 Binds to Acetylated Chromatinduring Interphase and Mitosis, Proc. Natl. Acad. Sci.USA, 2003, vol. 100, pp. 8758–8763.

96. Hans, F. and Dimitrov, S., Histone H3 Phosphorylationand Cell Division, Oncogene, 2001, vol. 20, pp. 3021–3027.

97. Nowak, S.J. and Corces, V.G., Phosphorylation of His-tone H3: A Balancing Act between Chromosome Con-densation and Transcriptional Activation, TrendsGenet., 2004, vol. 20, pp. 214–220.

98. Ahn, S., Henderson, K.A., Keeney, S., and Allis, C.D.,H2B (Ser10) Phosphorylation Is Induced during Apop-tosis and Meiosis in S. cerevisiae, Cell Cycle, 2005,vol. 4, pp. 780–783.

99. Bischoff, J.R. and Plowman, G.D., The Aurora/Ipl1pKinase Family: Regulators of Chromosome Segrega-tion and Cytokinesis, Trends Cell Biol., 1999, vol. 9,pp. 454–459.

100. Hsu, J.Y., Sun, Z.W., Li, X., et al., Mitotic Phosphory-lation of Histone H3 Is Governed by Ipl1/Aurora Kinaseand Glc7/PP1 Phosphatase in Budding Yeast andNematodes, Cell (Cambridge, Mass.), 2000, vol. 102,pp. 279–291.

101. Demidov, D., van Damme, D., Geelen, D., et al., Identi-fication and Dynamics of Two Classes of Aurora-LikeKinases in Arabidopsis and Other Plants, Plant Cell,2005, vol. 17, pp. 836–848.

102. Sassone-Corsi, P., Mizzen, C.A., Cheung, P., et al.,Requirement of Rsk-2 for Epidermal Growth Factor-Activated Phosphorylation of Histone H3, Science,1999, vol. 285, pp. 886–891.

103. Strelkov, I.S. and Davie, J.R., Ser-10 Phosphorylationof Histone H3 and Immediate Early Gene Expression in

Oncogene-Transformed Mouse Fibroblasts, CancerRes., 2002, vol. 62, pp. 75–78.

104. Soloaga, A., Thomson, S., Wiggin, G.R., et al., MSK2and MSK1 Mediate the Mitogen- and Stress-InducedPhosphorylation of Histone H3 and HMG-14, EMBO J.,2003, vol. 22, pp. 2788–2797.

105. Wang, Y., Zhang, W., Jin, Y., et al., The JIL-1 TandemKinase Mediates Histone H3 Phosphorylation and IsRequired for Maintenance of Chromatin Structure inDrosophila, Cell (Cambridge, Mass.), 2001, vol. 105,pp. 433–443.

106. Nowak, S.J. and Corces, V.G., Protein Phosphatase 2AActivity Affects Histone H3 Phosphorylation and Tran-scription in Drosophila melanogaster, Mol. Cell. Biol.,2003, vol. 23, pp. 6129–6138.

107. Goldknopf, I.L., Taylor, C.W., Baum, R.M., et al., Iso-lation and Characterization of Protein A24, a “Histone-Like” Non-Histone Chromosomal Protein, J. Biol.Chem., 1975, vol. 250, pp. 7182–7187.

108. Goldknopf, I.L. and Busch, H., Isopeptide Linkagebetween Nonhistone and Histone 2A Polypeptides ofChromosomal Conjugate-Protein A24, Proc. Natl.Acad. Sci. USA, 1977, vol. 74, pp. 864–868.

109. Thorne, A.W., Sautiere, P., Briand, G., and Crane-Rob-inson, C., The Structure of Ubiquitinated Histone H2B,EMBO J., 1987, vol. 6, pp. 1005–1010.

110. Robzyk, K., Recht, J., and Osley, M.A., Rad6-Depen-dent Ubiquitination of Histone H2B in Yeast, Science,2000, vol. 287, pp. 501–504.

111. Hershko, A. and Ciechanover, A., The Ubiquitin Sys-tem, Annu. Rev. Biochem., 1998, vol. 67, pp. 425–479.

112. Rea, S., Eisenhaber, F., O’Carroll, D., et al., Regulationof Chromatin Structure by Site-Specific Histone H3Methyltransferases, Nature, 2000, vol. 406, pp. 593–599.

113. Peters, A.H.F.M., Kubicek, S., Mechtler, K., et al., Par-titioning and Plasticity of Repressive Histone Methyla-tion States in Mammalian Chromatin, Mol. Cell, 2003,vol. 12, pp. 1577–1589.

114. Schotta, G., Lachner, M., Sarma, K., et al., A SilencingPathway to Induce H3–K9 and H4–K20 Tri-Methyla-tion at Constitutive Heterochromatin, Genes Dev.,2004, vol. 18, pp. 1251–1262.

115. Cowell, I.G., Aucott, R., Mahadevaiah, S.K., et al., Het-erochromatin, HP1 and Methylation at Lysine 9 of His-tone H3 in Animals, Chromosoma, 2002, vol. 111,pp. 22–36.

116. Schotta, G., Ebert, A., Krauss, V., et al., Central Role ofDrosophila SU(VAR)3-9 in Histone H3–K9 Methyla-tion and Heterochromatic Gene Silencing, EMBO J.,2002, vol. 21, pp. 1121–1131.

117. Ebert, A., Schotta, G., Lein, S., et al., Su(var) GenesRegulate the Balance between Euchromatin and Het-erochromatin in Drosophila, Genes Dev., 2004, vol. 18,pp. 2973–2983.

118. Brockdorff, N., X-Chromosome Inactivation: Closingin on Proteins That Bind Xist RNA, Trends Genet.,2002, vol. 18, pp. 352–358.

119. Plath, K., Fang, J., Mlynarczyk-Evans, S.K., et al., Roleof Histone H3 Lysine 27 Methylation in X Inactivation,Science, 2003, vol. 300, pp. 131–135.

Page 12: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 981

120. Silva, J., Mak, W., Zvetkova, I., et al., Establishment ofHistone H3 Methylation on the Inactive X ChromosomeRequires Transient Recruitment of Eed–Enx1 Poly-comb Group Complexes, Dev. Cell, 2003, vol. 4,pp. 481–495.

121. Kohlmaier, A., Savarese, F., Lachner, M., et al., A Chro-mosomal Memory Triggered by Xist Regulates HistoneMethylation in X Inactivation, PLoS Biol., 2004, vol. 2,p. E171.

122. Heard, E., Rougeulle, C., Arnaud, D., et al., Methyla-tion of Histone H3 at Lys-9 Is an Early Mark on theX Chromosome during X Inactivation, Cell (Cam-bridge, Mass.), 2001, vol. 107, pp. 727–738.

123. Mermoud, J.E., Popova, B., Peters, A.H., et al., HistoneH3 Lysine 9 Methylation Occurs Rapidly at the Onsetof Random X Chromosome Inactivation, Curr. Biol.,2002, vol. 12, pp. 247–251.

124. Peters, A.H.F.M., Mermoud, J.E., O’Carroll, D., et al.,Histone H3 Lysine 9 Methylation Is an EpigeneticImprint of Facultative Heterochromatin, Nat. Genet.,2002, vol. 30, pp. 77–80.

125. Rougeulle, C., Chaumeil, J., Sarma, K., et al., Differen-tial Histone H3 Lys-9 and Lys-27 Methylation Profileson the X Chromosome, Mol. Cell. Biol., 2004, vol. 24,pp. 5475–5484.

126. Rice, J.C., Briggs, S.D., Ueberheide, B., et al., HistoneMethyltransferases Direct Different Degrees of Methy-lation to Define Distinct Chromatin Domains, Mol.Cell, 2003, vol. 12, pp. 1591–1598.

127. Norris, D.P., Brockdorff, N., and Rastan, S., Methyla-tion Status of CpG-Rich Islands on Active and InactiveMouse X Chromosomes, Mamm. Genome, 1991, vol. 1,pp. 78–83.

128. Jeppesen, P. and Turner, B.M., The Inactive X Chromo-some in Female Mammals Is Distinguished by a Lackof Histone H4 Acetylation, a Cytogenetic Marker forGene Expression, Cell (Cambridge, Mass.), 1993,vol. 74, pp. 281–289.

129. Czermin, B., Schotta, G., Hulsmann, B.B., et al., Phys-ical and Functional Association of SU(VAR)3-9 andHDAC1 in Drosophila, EMBO Rep., 2001, vol. 2,pp. 915–919.

130. Nakayama, J., Rice, J.C., Strahl, B.D., et al., Role ofHistone H3 Lysine 9 Methylation in Epigenetic Controlof Heterochromatin Assembly, Science, 2001, vol. 292,pp. 110–113.

131. Kim, H.S., Choi, E.S., Shin, J.A., et al., Regulation ofSwi6/HP1-Dependent Heterochromatin Assembly byCooperation of Components of the Mitogen-ActivatedProtein Kinase Pathway and a Histone DeacetylaseClr6, J. Biol. Chem., 2004, vol. 279, pp. 42850–42859.

132. Taverna, S.D., Coyne, R.S., and Allis, C.D., Methyla-tion of Histone H3 at Lysine 9 Targets ProgrammedDNA Elimination in Tetrahymena, Cell (Cambridge,Mass.), 2002, vol. 110, pp. 701–711.

133. Bannister, A.J., Zegerman, P., Partridge, J.F., et al.,Selective Recognition of Methylated Lysine 9 on His-tone H3 by the HP1 Chromo Domain, Nature, 2001,vol. 410, pp. 120–124.

134. Lachner, M., O’Carroll, D., Rea, S., et al., Methylationof Histone H3 Lysine 9 Creates a Binding Site for HP1Proteins, Nature, 2001, vol. 410, pp. 116–120.

135. Nielsen, A.L., Oulad-Abdelghani, M., Ortiz, J.A., et al.,Heterochromatin Formation in Mammalian Cells: Inter-action between Histones and HP1 Proteins, Mol. Cell,2001, vol. 7, pp. 729–739.

136. Li, Y., Kirschmann, D.A., and Wallrath, L.L., Does Het-erochromatin Protein 1 Always Follow Code?, Proc.Natl. Acad. Sci. USA, 2002, vol. 99, suppl. 4, pp. 16462–16 469.

137. Reuter, G., Dorn, R., Wustmann, G., et al., Third Chro-mosome Suppressor of Position-Effect Variegation Lociin Drosophila melanogaster, Mol. Gen. Genet., 1986,pp. 481–487.

138. Wustmann, G., Szidonya, J., Taubert, H., and Reuter, G.,The Genetics of Position-Effect Variegation ModifyingLoci in Drosophila melanogaster, Mol. Gen. Genet.,1989, vol. 217, pp. 520–527.

139. Hwang, K., Eissenberg, J.C., and Worman, H.J., Tran-scriptional Repression of Euchromatic Genes by Droso-phila Heterochromatin Protein 1 and Histone Modifiers,Proc. Natl. Acad. Sci. USA, 2001, vol. 98, pp. 11 423–11 427.

140. Greil, F., van der Kraan, I., Delrow, J., et al., DistinctHP1 and Su(var)3-9 Complexes Bind to Sets of Devel-opmentally Coexpressed Genes Depending on Chromo-somal Location, Genes Dev., 2003, vol. 17, pp. 2825–2838.

141. Hearn, M.G., Hedrick, A., Grigliatti, T.A., and Waki-moto, B.T., The Effect of Modifiers of Position-EffectVariegation on the Variegation Heterochromatic Genesof Drosophila melanogaster, Genetics, 1991, vol. 128,pp. 785–797.

142. Lu, B.Y., Emtage, P.C.R., Duyf, B.J., et al., Heterochro-matin Protein 1 Is Required for the Normal Expressionof Two Heterochromatin Genes in Drosophila, Genet-ics, 2000, vol. 155, pp. 699–708.

143. Piacentini, L., Fanti, L., Berloco, M., et al., Heterochro-matin Protein 1 (HP1) Is Associated with Induced GeneExpression in Drosophila Euchromatin, J. Cell Biol.,2003, vol. 161, pp. 707–714.

144. Schulze, S.R., Sinclair, D.A.R., Fitzpatrick, K.A., andHonda, B.M., A Genetic and Molecular Characteriza-tion of Two Proximal Heterochromatic Genes on Chro-mosome 3 of Drosophila melanogaster, Genetics, 2005,vol. 169, pp. 2165–2177.

145. Cryderman, D.E., Grade, S.K., Li, Y., et al., Role ofDrosophila HP1 in Euchromatic Gene Expression, Dev.Dyn., 2005, vol. 232, pp. 767–774.

146. Vakoc, C.R., Mandat, S.A., Olenchock, B.A., and Blo-bel, G.A., Histone H3 Lysine 9 Methylation and HP1Are Associated with Transcription Elongation throughMammalian Chromatin, Mol. Cell, 2005, vol. 19,pp. 381–391.

147. Jackson, J.P., Lindroth, A.M., Cao, X., and Jacobsen, S.E.,Control of CpNpG DNA Methylation by the KRYPTO-NITE Histone H3 Methyltransferase, Nature, 2002,vol. 416, pp. 556–560.

148. Gendrel, A.V., Lippman, Z., Yordan, C., et al., Depen-dence of Heterochromatic Histone H3 Methylation Pat-terns on the Arabidopsis Gene DDM1, Science, 2002,vol. 297, pp. 1871–1873.

Page 13: Histone modification and regulation of chromatin function

982

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

KORYAKOV

149. Tamaru, H. and Selker, E.U., A Histone H3 Methyl-transferase Controls DNA Methylation in Neurosporacrassa, Nature, 2001, vol. 414, pp. 277–283.

150. Tamaru, H., Zhang, X., McMillen, D., et al., Trimethy-lated Lysine 9 of Histone H3 Is a Mark for DNA Meth-ylation in Neurospora crassa, Nat. Genet., 2003,vol. 34, pp. 75–79.

151. Malagnac, F., Bartee, L., and Bender, J., An ArabidopsisSET Domain Protein Required for Maintenance but NotEstablishment of DNA Methylation, EMBO J., 2002,vol. 21, pp. 6842–6852.

152. Fuks, F., Hurd, P.J., Deplus, R., and Kouzarides, T., TheDNA Methyltransferases Associate with HP1 and theSUV39H1 Histone Methyltransferase, Nucleic AcidsRes., 2003, vol. 31, pp. 2305–2312.

153. Lehnertz, B., Ueda, Y., Derijck, A.A.H.A., et al.,Suv39h-Mediated Histone H3 Lysine 9 MethylationDirects DNA Methylation to Major Satellite Repeats atPericentric Heterochromatin, Curr. Biol., 2003, vol. 13,pp. 1192–1200.

154. Weissmann, F., Muyrers-Chen, I., Musch, T., et al.,DNA Hypermethylation in Drosophila melanogasterCauses Irregular Chromosome Condensation and Dys-regulation of Epigenetic Histone Modifications, Mol.Cell. Biol., 2003, vol. 23, pp. 2577–2586.

155. Freitag, M., Hickey, P.C., Khlafallah, T.K., et al., HP1 IsEssential for DNA Methylation in Neurospora, Mol.Cell, 2004, vol. 13, pp. 427–434.

156. Tariq, M., Saze, H., Probst, A.V., et al., Erasure of CpGMethylation in Arabidopsis Alters Patterns of HistoneH3 Methylation in Heterochromatin, Proc. Natl. Acad.Sci. USA, 2003, vol. 100, pp. 8823–8827.

157. Espada, J., Ballestar, E., Fraga, M.F., et al., HumanDNA Methyltransferase 1 Is Required for Maintenanceof the Histone H3 Modification Pattern, J. Biol. Chem.,2004, vol. 279, pp. 37 175–37184.

158. Ng, H. and Bird, A., DNA Methylation and ChromatinModification, Curr. Opin. Genet. Dev., 1999, vol. 9,pp. 158–163.

159. Hung, M. and Shen, C.J., Eukaryotic Methyl-CpG-Binding Domain Proteins and Chromatin Modification,Eukar. Cell, 2003, vol. 2, pp. 841–846.

160. Otte, A.P. and Kwaks, T.H.J., Gene Repression by Poly-comb Group Protein Complexes: A Distinct Complexfor Every Occasion?, Curr. Opin. Genet. Dev., 2003,vol. 13, pp. 448–454.

161. Ringrose, L. and Paro, R., Epigenetic Regulation ofCellular Memory by the Polycomb and Trithorax GroupProteins, Annu. Rev. Genet., 2004, vol. 38, pp. 413–443.

162. Shao, Z., Raible, F., Mollaaghababa, R., et al., Stabili-zation of Chromatin Structure by PRC1, a PolycombComplex, Cell (Cambridge, Mass.), 1999, vol. 98,pp. 37–46.

163. Francis, N.J., Saurin, A.J., Shao, Z., and Kingston, R.E.,Reconstitution of a Functional Core Polycomb Repres-sive Complex, Mol. Cell, 2001, vol. 8, pp. 545–556.

164. Huang, D.H., Chang, Y.L., Yang, C.C., et al., PipsqueakEncodes a Factor Essential for Sequence-Specific Tar-geting of a Polycomb Group Protein Complex, Mol.Cell. Biol., 2002, vol. 22, pp. 6261–6271.

165. Mulholland, N.M., King, I.F., and Kingston, R.E., Reg-ulation of Polycomb Group Complexes by the

Sequence-Specific DNA-Binding Proteins Zeste andGAGA, Genes Dev., 2003, vol. 17, pp. 2741–2746.

166. Cao, R., Wang, L., Wang, H., et al., Role of Histone H3Lysine 27 Methylation in Polycomb-Group Silencing,Science, 2002, vol. 298, pp. 1039–1043.

167. Czermin, B., Melfi, R., McCabe, D., et al., DrosophilaEnhancer of Zeste/ESC Complexes Have a Histone H3Methyltransferase Activity That Marks ChromosomalPolycomb Sites, Cell (Cambridge, Mass.), 2002, vol. 111,pp. 185–196.

168. Kuzmichev, A., Nishioka, K., Erdjument-Bromage, H.,et al., Histone Methyltransferase Activity Associatedwith a Human Multiprotein Complex Containing theEnhancer of Zeste Protein, Genes Dev., 2002, vol. 16,pp. 2893–2905.

169. Kuzmichev, A., Jenuwein, T., Tempst, P., and Rein-berg, D., Different Ezh2-Containing Complexes TargetMethylation of Histone H1 or Nucleosomal Histone H3,Mol. Cell, 2004, vol. 14, pp. 183–193.

170. Vaquero, A., Scher, M., Lee, D., et al., Human SirT1Interacts with Histone H1 and Promotes Formation ofFacultative Heterochromatin, Mol. Cell, 2004, vol. 16,pp. 93–105.

171. Kuzmichev, A., Margueron, R., Vaquero, A., et al.,Composition and Histone Substrates of PolycombRepressive Group Complexes Change during CellularDifferentiation, Proc. Natl. Acad. Sci. USA, 2005,vol. 102, pp. 1859–1864.

172. Fischle, W., Wang, Y., Jacobs, S.A., et al., MolecularBasis for the Discrimination of Repressive Methyl-Lysine Marks in Histone H3 by Polycomb and HP1Chromodomains, Genes Dev., 2003, vol. 17, pp. 1870–1881.

173. Wang, L., Brown, J.L., Cao, R., et al., HierarchicalRecruitment of Polycomb Group Silencing Complexes,Mol. Cell, 2004, vol. 14, pp. 637–646.

174. Wang, H., Wang, L., Erdjument-Bromage, H., et al.,Role of Histone H2A Ubiquitination in PolycombSilencing, Nature, 2004, vol. 431, pp. 873–878.

175. Cao, R., Tsukada, Y., and Zhang, Y., Role of Bmi-1 andRing1A in H2A Ubiquitylation and Hox Gene Silenc-ing, Mol. Cell, 2005, vol. 20, pp. 845–854.

176. Vire, E., Brenner, C., Deplus, R., et al., The PolycombGroup Protein EZH2 Directly Controls DNA Methyla-tion, Nature, 2006, vol. 439, pp. 871–874.

177. Rusche, L.N., Kirchmaier, A.L., and Rine, J., TheEstablishment, Inheritance, and Function of SilencedChromatin in Saccharomyces cerevisiae, Annu. Rev.Biochem., 2003, vol. 72, pp. 481–516.

178. Briggs, S.D., Bryk, M., Strah, B.D., et al., Histone H3Lysine 4 Methylation Is Mediated by Set1 and Requiredfor Cell Growth and rDNA Silencing in Saccharomycescerevisiae, Genes Dev., 2001, vol. 15, pp. 3286–3295.

179. Ivy, J.M., Klar, A.J.S., and Hicks, J.B., Cloning andCharacterization of Four SIR Genes of Saccharomycescerevisiae, Mol. Cell. Biol., 1986, vol. 6, pp. 688–702.

180. Rine, J. and Herskowitz, I., Four Genes Responsible fora Position Effect on Expression from HML and HMR inSaccharomyces cerevisiae, Genetics, 1987, vol. 116,pp. 9–22.

181. Aparicio, O.M., Billington, B.L., and Gottschling, D.E.,Modifiers of Position Effect Are Shared between Telo-

Page 14: Histone modification and regulation of chromatin function

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

HISTONE MODIFICATION AND REGULATION OF CHROMATIN FUNCTION 983

meric and Silent Mating-Type Loci in S. cerevisiae, Cell(Cambridge, Mass.), 1991, vol. 66, pp. 1279–1287.

182. Smith, J.S. and Boeke, J.D., An Unusual Form of Tran-scriptional Silencing in Yeast Ribosomal DNA, GenesDev., 1997, vol. 11, pp. 241–254.

183. Landry, J., Sutton, A., Tafrov, S.T., et al., The SilencingProtein SIR2 and Its Homologs Are NAD-DependentProtein Deacetylases, Proc. Natl. Acad. Sci. USA, 2000,vol. 97, pp. 5807–5811.

184. Carmen, A.A., Milne, L., and Grunstein, M., Acetyla-tion of the Yeast Histone H4 N-Terminus Regulates ItsBinding to Heterochromatin Protein SIR3, J. Biol.Chem., 2002, vol. 277, pp. 4778–4781.

185. Rusche, L.N., Kirchmaier, A.L., and Rine, J., OrderedNucleation and Spreading of Silenced Chromatin inSaccharomyces cerevisiae, Mol. Biol. Cell, 2002, vol. 13,pp. 2207–2222.

186. Agalioti, T., Chen, G., and Thanos, D., Deciphering theTranscriptional Histone Acetylation Code for a HumanGene, Cell (Cambridge, Mass.), 2002, vol. 111, pp. 381–392.

187. Pokholok, D.K., Harbison, C.T., Levine, S., et al.,Genome-Wide Map of Nucleosome Acetylation andMethylation in Yeast, Cell (Cambridge, Mass.), 2005,vol. 122, pp. 517–527.

188. Suka, N., Suka, Y., Carmen, A.A., et al., Highly SpecificAntibodies Determine Histone Acetylation Site Usagein Yeast Heterochromatin and Euchromatin, Mol. Cell,2001, vol. 8, pp. 473–479.

189. Robert, F., Pokholok, D.K., Hannett, N.M., et al., Glo-bal Position and Recruitment of HATs and HDACs inthe Yeast Genome, Mol. Cell, 2004, vol. 16, pp. 199–209.

190. Roh, T., Ngau, W.C., Cui, K., et al., High-ResolutionGenome-Wide Mapping of Histone Modifications, Nat.Biotechnol., 2004, vol. 22, pp. 1013–1016.

191. Liu, C.L., Kaplan, T., Kim, M., et al., Single-Nucleo-some Mapping of Histone Modifications in S. cerevi-siae, PLoS Biol., 2005, vol. 3, p. E328.

192. Barratt, M.J., Hazzalin, C.A., Cano, E., and Mahade-van, L.C., Mitogen-Stimulated Phosphorylation of His-tone H3 Is Targeted to a Small Hyperacetylation-Sensi-tive Fraction, Proc. Natl. Acad. Sci. USA, 1994, vol. 91,pp. 4781–4785.

193. Cheung, P., Allis, C.D., and Sassone-Corsi, P., Signal-ing to Chromatin through Histone Modifications, Cell(Cambridge, Mass.), 2000, vol. 103, pp. 263–271.

194. Clayton, A.L., Rose, S., Barratt, M.J., and Mahade-van, L.C., Phosphoacetylation of Histone H3 on c-fos-and c-jun-Associated Nucleosomes upon Gene Activa-tion, EMBO J., 2000, vol. 19, pp. 3714–3726.

195. Lo, W.S., Trievel, R.C., Rojas, J.R., et al., Phosphoryla-tion of Serine 10 in Histone H3 Is Functionally LinkedIn vitro and In vivo to GCN5-Mediated Acetylation atLysine 14, Mol. Cell, 2000, vol. 5, pp. 917–926.

196. Lo, W.S., Duggan, L., Emre, N.C., et al., Snf1—A His-tone Kinase That Works in Concert with the HistoneAcetyltransferase GCN5 to Regulate Transcription, Sci-ence, 2001, vol. 293, pp. 1142–1146.

197. Thomson, S., Clayton, A.L., and Mahadevan, L.C.,Independent Dynamic Regulation of Histone Phospho-

rylation and Acetylation during Immediate-Early GeneInduction, Mol. Cell, 2001, vol. 8, pp. 1231–1241.

198. Nowak, S.J. and Corces, V.G., Phosphorylation of His-tone H3 Correlates with Transcriptionally Active Loci,Genes Dev., 2000, vol. 14, pp. 3003–3013.

199. Santos-Rosa, H., Schneider, R., Bannister, A.J., et al.,Active Genes Are Tri-Methylated at K4 of Histone H3,Nature, 2002, vol. 419, pp. 407–411.

200. Schneider, R., Bannister, A.J., Myers, F.A., et al., His-tone H3 Lysine 4 Methylation Patterns in HigherEukaryotic Genes, Nature Cell. Biol., 2004, vol. 6,pp. 73–77.

201. Bernstein, B.E., Kamal, M., Lindblad-Toh, K., et al.,Genomic Maps and Comparative Analysis of HistoneModifications in Human and Mouse, Cell (Cambridge,Mass.), 2005, vol. 120, pp. 169–181.

202. Nishioka, K., Chuikov, S., Sarma, K., et al., Set9, aNovel Histone H3 Methyltransferase That FacilitatesTranscription by Precluding Histone Tail ModificationsRequired for Heterochromatin Formation, Genes Dev.,2002, vol. 16, pp. 479–489.

203. Roguev, A., Schaft, D., Shevchenko, A., et al., The Sac-charomyces cerevisiae Set1 Complex Includes an Ash2Homologue and Methylates Histone 3 Lysine 4, EMBO J.,2001, vol. 20, pp. 7137–7148.

204. Miller, T., Krogan, N.J., Dover, J., et al., COMPASS: AComplex of Proteins Associated with a Trithorax-Related SET Domain Protein, Proc. Natl. Acad. Sci.USA, 2001, vol. 98, pp. 12902–12907.

205. Ng, H., Robert, F., Young, R.A., and Struhl, K., TargetedRecruitment of Set1 Histone Methylase by ElongatingPol II Provides a Localized Mark and Memory ofRecent Transcriptional Activity, Mol. Cell, 2003,vol. 11, pp. 709–719.

206. Henry, K.W., Wyce, A., Lo, W., et al., TranscriptionalActivation Via Sequential Histone H2B Ubiquitylationand Deubiquitylation, Mediated by SAGA-AssociatedUbp8, Genes Dev., 2003, vol. 17, pp. 2648–2663.

207. Kim, J., Hake, S.B., and Roeder, R.G., The HumanHomolog of Yeast BRE1 Functions As a TranscriptionalCoactivator through Direct Activator Interactions, Mol.Cell, 2005, vol. 20, pp. 759–770.

208. Zhu, B., Zheng, Y., Pham, A.D., et al., Monoubiquitina-tion of Human Histone H2B: The Factors Involved andTheir Roles in HOX Gene Regulation, Mol. Cell, 2005,vol. 20, pp. 601–611.

209. Shahbazian, M.D., Zhang, K., and Grunstein, M., His-tone H2B Ubiquitylation Controls Processive Methyla-tion but Not Monomethylation by Dot1 and Set1, Mol.Cell, 2005, vol. 19, pp. 271–277.

210. Bannister, A.J., Schneider, R., Myers, F.A., et al., Spa-tial Distribution of Di- and Tri-Methyl Lysine 36 of His-tone H3 at Active Genes, J. Biol. Chem., 2005, vol. 280,pp. 17732–17736.

211. Strahl, B.D., Grant, P.A., Briggs, S.D., et al., Set2 Is aNucleosomal Histone H3-Selective MethyltransferaseThat Mediates Transcriptional Repression, Mol. Cell.Biol., 2002, vol. 22, pp. 1298–1306.

212. Schaft, D., Roguev, A., Kotovic, K.M., et al., The His-tone 3 Lysine 36 Methyltransferase, SET2, Is Involvedin Transcriptional Elongation, Nucleic Acids Res., 2003,vol. 31, pp. 2475–2482.

Page 15: Histone modification and regulation of chromatin function

984

RUSSIAN JOURNAL OF GENETICS Vol. 42 No. 9 2006

KORYAKOV

213. Xiao, T., Hall, H., Kizer, K.O., et al., Phosphorylation ofRNA Polymerase II CTD Regulates H3 Methylation inYeast, Genes Dev., 2003, vol. 17, pp. 654–663.

214. Krogan, N.J., Kim, M., Tong, A., et al., Methylation ofHistone H3 by Set2 in Saccharomyces cerevisiae IsLinked to Transcriptional Elongation by RNA Poly-merase II, Mol. Cell. Biol., 2003, vol. 23, pp. 4207–4218.

215. Kizer, K.O., Phatnani, H.P., Shibata, Y., et al., A NovelDomain in Set2 Mediates RNA Polymerase II Interac-tion and Couples Histone H3 K36 Methylation withTranscript Elongation, Mol. Cell. Biol., 2005, vol. 25,pp. 3305–3316.

216. Carrozza, M.J., Li, B., Florens, L., et al., Histone H3Methylation by Set2 Directs Deacetylation of CodingRegions by Rpd3S to Suppress Spurious IntragenicTranscription, Cell (Cambridge, Mass.), 2005, vol. 123,pp. 581–592.

217. Keogh, M.C., Kurdistani, S.K., Morris, S.A., et al.,Cotranscriptional Set2 Methylation of Histone H3Lysine 36 Recruits a Repressive Rpd3 Complex, Cell(Cambridge, Mass.), 2005, vol. 123, pp. 593–605.

218. Briggs, S.D., Xiao, T., Sun, Z.W., et al., Gene Silencing:Trans-Histone Regulatory Pathway in Chromatin,Nature, 2002, vol. 418, p. 498.

219. Wargo, M.J. and Rizzo, P.J., Exception to EukaryoticRules, Science, 2001, vol. 294, p. 2477.

220. Rizzo, P.J., Those Amazing Dinoflagellate Chromo-somes, Cell Res., 2002, vol. 13, pp. 215–217.

221. Fischle, W., Wang, Y., and Allis, C.D., Histone andChromatin Cross-Talk, Curr. Opin. Cell Biol., 2003,vol. 15, pp. 172–183.

222. LaJeunesse, D. and Shearn, A., Trans-Regulation ofThoracic Homeotic Selector Genes of the Antennapediaand Bithorax Complexes by the trithorax Group Genes:Absent, Small, and Homeotic Discs 1 and 2, Mech.Dev., 1995, vol. 53, pp. 123–139.

223. Beisel, C., Imhof, A., Greene, J., et al., Histone Methy-lation by the Drosophila Epigenetic TranscriptionalRegulator Ash1, Nature, 2002, vol. 419, pp. 857–862.

224. Krogan, N.J., Dover, J., Khorrami, S., et al., COM-PASS, a Histone H3 (Lysine 4) MethyltransferaseRequired for Telomeric Silencing of Gene Expression,J. Biol. Chem., 2002, vol. 277, pp. 10753–10 755.