Biochimica Et Biophysica Acta: Gang Yuan, Bing Zhu

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Biochimica et Biophysica Acta 1819 (2012) 222–229

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / b b a g r m

Review

Histone variants and epigenetic inheritance ☆


Gang Yuan a, b, Bing Zhu b,⁎
a
Life Science College, Beijing Normal University, Beijing, 100875, China
b
National Institute of Biological Sciences, Beijing, 102206, China

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

Article history: Nucleosome particles, which are composed of core histones and DNA, are the basic unit of eukaryotic
Received 4 May 2011 chromatin. Histone modifications and histone composition determine the structure and function of the
Received in revised form 8 June 2011 chromatin; this genome packaging, often referred to as “epigenetic information”, provides additional
Accepted 9 June 2011
information beyond the underlying genomic sequence. The epigenetic information must be transmitted from
Available online 17 June 2011
mother cells to daughter cells during mitotic division to maintain the cell lineage identity and proper gene
Keywords:
expression. However, the mechanisms responsible for mitotic epigenetic inheritance remain largely
Histone variant unknown. In this review, we focus on recent studies regarding histone variants and discuss the assembly
Epigenetics pathways that may contribute to epigenetic inheritance. This article is part of a Special Issue entitled: Histone
Chromatin chaperones and Chromatin assembly.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction For mitotic inheritance of histone modification-mediated epigenetic


information, numerous models have been discussed in review papers
The eukaryotic genome is compacted into chromatin, which is [9,26–34], and several interesting investigations have been recently
composed of basic units termed “nucleosomes” [1]. Each nucleosome published [35–41]. Although increasing knowledge has been obtained
consists of an octameric particle of four core histone proteins (H2A, for histone variants, little information is known regarding whether
H2B, H3 and H4) that bind and wrap 146 base pairs of DNA around the and/or which histone variants may carry epigenetic information; and
histone octamer [2]. Histones can be covalently modified by reactions how such epigenetic information passes to the daughter cells during
such as acetylation, methylation, phosphorylation, and ubiquitylation mitotic division. In this review, we will discuss the deposition of
[3,4]. These chromatin modifications are often recognized by effector variant histones and their potential roles in mediating the inheritance
proteins through which they exert their functions [5,6]. In addition to of epigenetic information.
chromatin post-translational modifications, the importance of histone
variants has been increasingly demonstrated in recent years [7–9]. 2. H3 variants
Epigenetics is usually defined as the study of mitotically and/or
meiotically heritable changes in gene function that cannot be The number of H3 histone variants differs among species. All
explained by changes in the DNA sequence [10]. However, the eukaryotes have a centromere specific H3 (CenH3, or CENP-A in
mechanisms responsible for the inheritance of epigenetic information mammals), which contains an amino acid sequence that differs
during mitotic division remain unknown. The inheritance of DNA CpG significantly from the other H3 histone variants [7]. In addition to
methylation is the most well understood mechanism; several studies CENP-A, mammals have three ubiquitously expressed H3 variants
have indicated that there is a semi-conservative segregation of the (H3.1, H3.2, and H3.3) as well as an H3 isoform that is specifically
symmetric CpG methylation and maintenance of the DNA methylation expressed in the testis (H3t) [7]. Recently, two primate-specific H3
by DNMT1 [11–13] with the assistance of PCNA [14] and UHRF1 variants (H3.X and H3.Y) [42] and a hominid-specific variant H3.5
[15,16]. Because several histone modifications regulate well-estab- were identified [43]. Other higher eukaryotes have two non-
lished epigenetic phenomena, including position effect variegation centromeric H3 variants, H3.3 and H3.1 (identical to mammalian
[17–19], Polycomb silencing [20–23], and X inactivation [24,25], H3.2). Yeast has only one non-centromeric H3, which is similar to
researchers are particularly interested in studying the mitotic H3.3 in higher eukaryotes [7].
inheritance of histone modification-mediated epigenetic information.
2.1. CenH3
☆ This article is part of a Special Issue entitled: Histone chaperones and Chromatin
assembly.
CENP-A is the first histone variant that was shown to identify
⁎ Corresponding author. Tel.: + 86 10 80728458. specific chromatin regions, namely the centromeres [27]. CENP-A is
E-mail address: zhubing@nibs.ac.cn (B. Zhu). essential for kinetochore formation and chromosome segregation

1874-9399/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbagrm.2011.06.007
G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229 223

[7,44,45]. CENP-A was identified as an H3 variant during co- Because CENP-A is exclusively incorporated at the centromeres,
purification with other core histones [46], although it shares minimal the protein must be removed after incorrect incorporation at non-
sequence similarity with the other H3 histone variants. The other H3 centromere chromatin regions. The Cse4 specific E3 ubiquitin ligase
variants, such as H3.1, H3.2 and H3.3, are at the same molecular Psh1 mediates the degradation of non-centromeric Cse4 [67,68].
weight and differ by only four to five amino acid residues [9]. In Although the data clearly indicates that Psh1 plays a role in preventing
contrast, CENP-A has a variable N-terminus, with no sequence Cse4 mislocalization, parallel mechanisms must exist because Psh1
similarity to the N-terminal region of the other H3 variants [45]; deletion displayed a very mild phenotype unless Cse4 was massively
moreover, CENP-A shares only 50% identity in the histone fold domain over expressed [67,68].
with the other H3 histones [47]. The small GTPase and E3 ubiquitin ligase additionally help to
maintain the epigenetic state of centromeres, although CENP-A appears
2.1.1. CenH3 deposition and genomic distribution to be the most important factor. CENP-A's relatively long loop 1 region in
Centromeric DNA is duplicated during the S phase of the cell cycle. the histone fold domain makes a large area of contact with the
However, unlike canonical histones that are incorporated into centromeric DNA and contributes to the binding specificity [69–71]. The
chromatin during the S phase, newly synthesized CENP-A is deposited structure of the human (CENP-A–H4)2 heterotetramer shows a rotated
during a discrete time period from telophase to the G1 phase in CENP-A–CENP-A interface and a strong hydrophobic interaction
mammals [48]. between CENP-A and H4, which contributes to the unconventional
In yeast, CenH3 is called “Cse4”, and the specific Cse4 chaperone is shape of the CENP-A-containing nucleosomes [72]. Interestingly, human
Scm3 [49]. A crystal structure of the Scm3–Cse4–H4 fusion protein neocentromeres can be formed at genomic regions that lack the satellite
suggests that the complex may form a hexamer (Scm3–Cse4–H4)2 DNA sequences that are normally present at the centromeres [73], and
[50]; correspondingly, the formation of a (Scm3–Cse4–H4)2 hexamer similar events have been observed in fission yeast [74] and plants [75].
was observed in another study under high salt conditions (2 M NaCl) Thus, CENP-A binding, rather than the DNA sequence, appears to be a
[49]. This suggests that Scm3 may bind and deposit (Cse4–H4)2 more important determinant for defining the centromere [76]. Unlike
tetramers. This is in sharp contrast to Asf1, which is a key histone other histones, CENP-A is not replaced by protamines during mamma-
chaperone for the other H3 histone variants; Asf1 only forms a lian spermatogenesis, which also suggests that CENP-A plays an
complex with the H3-H4 dimers, not with the (H3–H4)2 tetramers important role in defining the centromere [77].
[51–55]. However, a recent report demonstrated a trimeric crystal
structure of a Scm3 fragment associated with a Cse4–H4 dimer, which 2.1.3. Models for restoring CenH3 nucleosomes or the epigenetic
argues against the (Scm3–Cse4–H4)2 model [56]. inheritance of centromeres
Studies have shown that in mammals, Mis16 and Mis18 function Extensive investigations regarding CenH3 have suggested several
as upstream factors that bind and recruit CENP-A to the kinetochore; a models for CenH3-containing nucleosome structures, including: 1) an
double knockdown of the two homologous Mis proteins in HeLa cells octameric nucleosome containing two copies of H2A, H2B, Cen-H3 and
abolished localization of CENP-A to the kinetochore [57]. Although H4, with DNA wrapped in left-handed orientation [62,70,72,78–80]; 2) a
Mis16 and Mis18 participate in CENP-A deposition, the direct tetrasome containing two copies of CenH3 and H4 but lacking the H2A–
chaperone that is specific for CENP-A is HJURP [58,59]. The transient H2B dimers [81]; 3) a hexasome containing two copies of Scm3, CenH3
appearance of HJURP precisely coincides with the discrete time period and H4 [49]; and 4) a hemisome that contains only one copy of H2A, H2B,
for CENP-A deposition, and down regulation of HJURP decreases CenH3 and H4, with DNA wrapped in a right-handed orientation [82,83].
CENP-A at the centromeres [58,59]. Because HJURP appears at the Despite the uncertainty regarding CenH3 nucleosome composition and
centromeres slightly later than Mis18, the Mis18 complex may structure, it is generally agreed that CenH3-containing nucleosomes
function as the licensing factor for CENP-A deposition, while HJURP must be restored to maintain the identity of the centromeres after DNA
likely acts as the direct loading factor for CENP-A [60]. Unlike the Scm3 duplication. The incorporation of newly synthesized CenH3 begins
and Cse4–H4 paradigm in yeast, structural analysis of the human during telophase, which occurs much later than centromeric DNA
HJURP–CENP-A–H4 trimeric complex indicates that binding of HJURP duplication. Therefore, CenH3 is in shortage for a considerably long
and CENP-A–H4 dimer prevents the formation of a (CENP-A–H4)2 period of time. Several models have been proposed to explain the
tetramer, which suggests that CENP-A–H4 is deposited as a dimer chromatin states of the centromeres during this period and define the
rather than a tetramer in mammals [61]. mechanisms by which the CenH3-containing nucleosomes are restored.
A number of proteins are physically associated with centromeres,
such as the NAC complex (including CENP-B, CENP-C, CENP-H, CENP- 1) After DNA replication, part of the centromeric region is temporarily
M, CENP-N, CENP-T and CENP-U) [62] and the CENP-H/CENP-I occupied by H3.1 nucleosomes, and CenH3 nucleosomes replace
complex [63]; these protein complexes are important for kinetochore these transient nucleosomes during the later stages in of the cell
assembly and may act upstream or downstream of CENP-A incorpo- cycle (Fig. 1, left). Based upon this model, the most critical problem is
ration. Recently, CENP-C and CENP-T have been reported to act how to ensure that the CenH3 nucleosomes only replace the H3.1
downstream of CENP-A incorporation and ectopic tethering of these nucleosomes at the centromeres. Distinct modification patterns on
proteins can induce ectopic kinetochore assembly in the absence of the interspersed H3.1 nucleosomes at the centromeric regions
CENP-A incorporation [64]. [84,85] and the remaining CenH3 nucleosomes at the centromeric
regions may identify these regions for CenH3 replacement. In
2.1.2. Defining centromere identity mammals, CENP-A and H3.1 nucleosomes are interspaced at the
Once the CenH3 histones are deposited on the centromere, this centromeric regions; however, the CENP-A nucleosomes are
centromeric state must be stably maintained. A number of proteins, spatially continuous and are often oriented in the same direction,
including the CENP-A licensing factor HsKNL2 interacting protein which aids in kinetochore formation [86]; therefore, epigenetic
MgcRacGAP (a Rho family activating protein), Ect2 (a Rho family information may be provided by the spatially proximal CENP-A
guanine nucleotide exchange factor), and Cdc42 and Rac (two small nucleosomes to guide the incorporation of newly synthesized CENP-
GTPases) are required to stabilize the newly incorporated CENP-A at A. If this hypothesis is correct, the CenH3 deposition machinery may
the centromeres [65,66]. Because CENP-A occupancy is typically used be able to recognize this genomic signal. Although we generally
to define a centromere, these proteins may help to ensure the prefer this model, we must point out that it is incompatible with the
epigenetic inheritance of the centromeric state by stabilizing CENP-A data for budding yeast, where each centromere is only composed of
binding. one nucleosome [87].
224 G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229

Fig. 1. Models for CENP-A inheritance at the centromere. Left: H3.1 nucleosomes are incorporated into the centromeric regions during DNA replication, and the CENP-A nucleosomes
are later incorporated and replace the H3.1 nucleosomes. Middle: DNA replication forms gaps that are devoid of nucleosomes during the S phase, and these gaps are later filled with
newly synthesized CENP-A nucleosomes. Right: DNA replication leads to the formation of hemisomes at the centromeric regions, and newly synthesized CENP-A-H4 dimers pair
with the existing hemisomes to form hybrid nucleosomes. Note: the H2A-H2B dimers were omitted from this illustration for simplicity.

2) During DNA replication, gaps are created at the centromeric regions acetylated. The histone chaperone Asf1 is also present in both
that are not filled with canonical nucleosomes and only become complexes, which agrees with previous studies that indicated that
occupied after the incorporation of CenH3 nucleosomes (Fig. 1, Asf1 participates in chromatin assembly [95–98].
middle). However, long stretches of DNA with reduced nucleosome Recently, several independent studies have shown that the ATRX-
protection at the centromeric regions may not be preferable [60]. In DAXX complex and DEK are chaperone proteins that play a role in
addition, this model is also incompatible with the data for budding H3.3 deposition at discrete chromatin regions, including telomeres
yeast for the same reason mentioned with model 1. and pericentromeric regions [91–94]. These studies are particularly
3) During DNA replication, the CenH3 nucleosomes split into two- interesting because they indicate that H3.3, although traditionally
halves, where each half occupies one sister chromatid and can thought to be a marker of euchromatin [8], is also present at hetero-
easily incorporate newly synthesized CenH3 molecules. Consistent chromatic regions.
with this model, CenH3 nucleosomes were reported to be at the The H3.3 chaperones that have been identified play a role in H3.3
half height of the canonical nucleosomes [81,82], which suggests a replication-independent deposition. H3.3 can also be deposited in a
“hemisome” model for CenH3 nucleosome structure. In this model, replication-dependent manner [99,100], although the chaperone
epigenetic information may be maintained at the mono-nucleo- proteins responsible for replication-dependent H3.3 deposition are
some level (Fig. 1, right), which agrees well with the data for unknown. It is possible that the CAF complex can deposit both H3.1
budding yeast centromere identity. and H3.3 histones in a replication-dependent manner because the
yeast CAF complex can deposit the only non-centromeric H3, which is
2.2. H3.3 similar to H3.3 [8].

Histone variant H3.3 differs from the canonical H3 by only four


amino acid residues, where three of these residues are clustered in 2.2.2. H3.3 mediated epigenetic inheritance
the α2 helix of the histone fold domain and the other residue is in the The potential of H3.3 to mediate epigenetic inheritance can be
N-terminal tail [9]. Unlike H3.1, which is strictly expressed and divided into two questions: 1) whether or how H3.3 histone
deposited during S phase, H3.3 is expressed and deposited throughout modifications are maintained in a mitotically heritable manner and
the cell cycle [88]. The three distinct amino acid residues in the α2 2) whether or how H3.3 histones are incorporated at specific
helix are responsible for the replication-independent incorporation of chromatin regions in a mitotically heritable manner.
H3.3 [88], and H3.3 histones are enriched at transcriptionally active Histone variants differ from each other not only in their chromatin
regions [88–90] as well as telomeres and pericentromeric regions location, but also in their post-translational modifications [101]. For
[91–94]. example, compared with the canonical H3 histone, H3.3 histones are
enriched in post-translational modifications that correlate with gene
2.2.1. H3.3 deposition and genomic distribution expression, such as H3K4 and H3K36 methylation, as well as H3K9,
Biochemical purification of non-chromatic H3 histones in HeLa H3K18, and H3K23 acetylation [54,102–106]. This is consistent with
cells showed that the canonical H3 histones are deposited by the CAF- previous observations that the H3.3 histones are enriched at
1 complex in a replication-dependent manner, whereas H3.3 histones transcriptionally active regions of the genome [88–90]. However,
are deposited by the HIRA complex in a replication-independent there is little evidence to suggest that the H3.3 histones are more
manner [51]. Both complexes contain HAT1, which corresponds with favorable substrates for the enzymes that mediate these modifica-
previous observations that pre-deposited histones are transiently tions. The enrichment of these active modifications on H3.3 histone
G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229 225

may simply be because H3.3 deposition and active histone modifica- H2B dimers to the SWR1 complex for final deposition [119]. A recent
tions are both independently correlated with transcriptional activity. study has presented a detailed molecular mechanism of SWR1-
A semi-conservative segregation of the (H3–H4)2 tetramers, mediated H2A.Z deposition [120]. SWR1 replaces H2A–H2B with
followed by templated modification copying events, was once a H2A.Z–H2B in a stepwise manner, one copy of the H2A.Z–H2B dimer
model favored for the mitotic inheritance of histone modification- at a time, which leads to the formation of intermediate hybrid
mediated epigenetic information. However, most lysine methylations nucleosomes that contain both H2A and H2A.Z [120]. Interestingly,
do not have to be symmetrical within each nucleosome [41], and nucleosome-dependent SWR1 ATPase activity is dependent upon the
canonical (H3–H4)2 tetramers do not split during replication- substrate composition, which indicates that the SWR1 complex can
dependent chromatin assembly [35]. These recent findings negate sense the substrate status and receive a “mission accomplished” signal
this model as the general mechanism for mitotic inheritance of to terminate its activity [120].
histone modification-mediated epigenetic information [33,34,107]. Genome-wide nucleosome occupancy studies in yeast and flies
Although a fraction of (H3.3–H4)2 tetramers split in mammalian cells have been performed to assess the distribution of H2A.Z nucleosomes.
[35] and similar events occur for the H3.3-like histones in budding The H2A.Z nucleosomes are localized to both sides of “nucleosome-
yeast [108], it is unlikely that these events mediate a faithful free regions (NFR)” in yeast near the transcription start sites (− 1, + 1
duplication of histone modifications within certain subpopulations nucleosome) [121,122], while the fly H2A.Z nucleosomes are
of mono-nucleosomes. predominantly enriched just downstream of the transcription start
On the other hand, the H3.3 histones are clearly enriched at certain sites (+1 nucleosome) [123]. Because the NFRs in yeast are precisely
chromatin regions, including actively transcribed genes [88–90] and located between two well-positioned H2A.Z nucleosomes, they may
heterochromatic regions, such as the telomeres and pericentromeric play a role in the H2A.Z localization pattern. In yeast, the RSC
regions [91,92,94,109]. However, it remains unclear whether H3.3 chromatin-remodeling complex mediates NFR formation at pro-
histone localization contributes to the transcriptional status of the moters by displacing nucleosomes at the NFR, and the RSC is required
underlying DNA. A number of studies suggest that H3.3 plays a role in for deposition of the flanking H2A.Z nucleosomes [124]. A previous
maintaining the transcriptional status of the target loci. Nuclear study has suggested that in mammals, the “NFRs” at the transcription
transfer experiments in Xenopus laevis demonstrated that the start sites were suggested to be consequence of losing the highly labile
expression of the donor-originated MyoD gene can persist over 24 nucleosomes consisted of both H2A.Z and H3.3 located at NFRs [125].
mitotic divisions and that this epigenetic memory is dependent upon The specific localization pattern of H2A.Z nucleosomes near the
H3.3 lysine 4 methylation [110,111]. However, the loss of both H3.3 transcription start site has been proposed to be an epigenetic marker
genes in flies leads to a mild transcriptional defect with partial for directing or regulating the positioning of downstream nucleo-
lethality, and most of the surviving H3.3-null flies develop normally, somes [126–128]. Therefore, ensuring the correct deposition of H2A.Z
except for the male germline [112]. These results suggest that H3.3 nucleosomes is crucial for the maintenance of epigenetic modifica-
histones are not essential for maintaining the epigenetic status of tions at the transcription start site. Recently, the INO80 complex,
transcriptionally active genes, which is consistent with the idea that which is another ATP-dependent chromatin remodeling complex that
H3.3 enrichment at euchromatic regions is a consequence, rather than is closely related to the SWR1 complex, was reported to mediate the
a determinant, of transcriptional activity. In contrast, H3.3 histones replacement of H2A.Z with canonical H2A to ensure proper H2A.Z
were reported to be critical for the formation of pericentromeric distribution at transcription start sites in yeast [129].
heterochromatin [109] and the maintenance of the telomeres [88,93].
Thus, the data indicate that H3.3 histones may play a role in epigenetic 3.1.2. H2A.Z mediated epigenetic inheritance
silencing. Compared to the (H3–H4)2 tetramers, the H2A–H2B dimers are
much more mobile [130]. Nucleosomes frequently exchange their
3. H2A variants H2A–H2B dimers, but rarely exchange their (H3–H4)2 tetramers [35].
Additionally, lysine methylations, which are thought to be the most
The H2A and H3 histones are structurally similar [113] and have stable histone modification [131], predominantly occur on the H3 or
several variant forms [7,47]. In addition to the canonical histone H2A, H4 histones [3,4]. Thus, the H2A and H2B histones may not be good
four H2A variants have been reported in mammals (H2A.Z, H2A.X, candidates for the propagation of epigenetic information. However, a
marcoH2A and H2A.Bbd) [47]. number of studies discussed below suggest that H2A.Z may carry
important epigenetic information and maintain the chromatin
3.1. H2A.Z transcriptional status.
Transcriptional memory. The yeast genes INO1 and GAL1 localize to
H2A.Z is highly conserved throughout evolution, it has a single the nuclear periphery upon transcriptional induction. After a short
evolutionary origin and remains distinct from all other H2A variants period of transcriptional repression, the nuclear peripheral INO1 and
[47]. This specialization suggests that H2A.Z has a distinct function GAL1 genes are rapidly reactivated, but not in Htz1 mutant strains
from all other H2A variants. H2A.Z differs from canonical H2A and (which cannot encode H2A.Z), which suggests that H2A.Z mediates
other H2A variants mainly in its “docking” domain in the C-terminus epigenetic memory of the previous transcriptional state [132].
and in the L1 loop where two H2A molecules contact each other Interestingly, transcriptional memory of the yeast GAL genes requires
[2,69,114]. SWI/SNF activity [133], and SWI/SNF recruitment is dependent upon
H2A.Z [134], which again suggests that H2A.Z mediates epigenetic
3.1.1. H2A.Z deposition and genomic distribution transcriptional memory.
The SWR1 complex, which is an ATP-dependent chromatin- Heterochromatin formation and maintenance. Upon differentiation
remodeling complex, was the first identified H2A.Z chaperone that of cells at the murine inner cell mass (ICM), H2A.Z is first enriched at
mediates H2A.Z deposition [115–117]. Subsequently, the Nap1 and pericentromeric heterochromatin and subsequently enriched at other
Chz1 histone chaperones also associate with H2A.Z–H2B dimers, and chromatin regions. Moreover, H2A.Z directly interacts with the
these chaperones were proposed to transfer the H2A.Z–H2B dimers to pericentromeric heterochromatin protein INCENP in vivo[135].
the SWR1 complex to exchange chromatin H2A-H2B dimers with H2A.Z depletion caused genome instability and disruption of HP1α
H2A.Z–H2B dimers [118]. Recent studies have indicated that Nap1 and localization at the pericentromeric regions, which suggests that HP1α
Chz1 are not functionally redundant. Nap1 mediates the nuclear function and pericentromeric heterochromatin identity are regulated
import of cytosolic H2A.Z–H2B, whereas Chz1 presents the H2A.Z– by H2A.Z during early embryonic development in mice [136]. In yeast,
226 G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229

Htz1 acts together with a boundary element to prevent the spread of macroH2A is enriched at developmental genes in human pluripotent
heterochromatin. As in htz1Δ cells, Sir2 and Sir3 spread from cells in males and regulates the timing of HoxA activation. The
telomeric regions to flanking eukaryotic regions and lead to ectopic macroH2A distribution pattern at the Hox loci overlaps with PRC2;
heterochromatin formation [137]. therefore, marcoH2A is potentially an epigenetic regulator of key
developmental genes and may cooperate with PRC2 [152].
3.2. H2A.X
3.4. H2A.Bbd
H2A.X is present in nearly all eukaryotes, except nematodes [47].
This histone variant has a histone fold domain that is similar to the H2A.Bbd is the most recently discovered H2A variant; this histone
canonical H2A, although it has a unique C-terminal motif termed variant has a truncated C-terminal docking domain [153] and was
SQ(E/D)Ф (where Ф represents a hydrophobic residue) [47,138]. The named because of its unique genomic distribution, where it is
H2A.X serine residue at the γ-position of the C terminus can be deficient in inactive X chromosomes (bar body deficient). The
phosphorylated (termed γ-H2A.X) after DNA double-strand breaks H2A.Bbd nucleosome binds only 116 base pairs of DNA and is less
(DSB) occur, which has been previously described [139,140]. stable than the canonical nucleosome [154,155]. H2A.Bbd lacks a
small acidic region on the nucleosome surface that is involved in
3.2.1. H2A.X deposition and genomic distribution transcriptional repression and also lacks K119, which is often
Previous studies have shown that H2A.X plays a role in DSB repair; ubiquitinated on canonical H2A at transcriptionally inactive regions
therefore, H2A.X is viewed as the “histone guardian of the genome” [156]. The unique subnuclear localization and key features of H2A.Bbd
[141]. Because DSB can potentially occur anywhere in the genome, suggest that it is involved in gene activation.
H2A.X deposition should be random. Although the FACT protein
complex can mediate the exchange of γ-H2A.X-H2B with unmodified
4. H4 and H2B histones
H2A.X-H2B [142], no specific de novo deposition chaperone for H2A.X
has been identified. Because of the overall sequence similarity between
Unlike the H3 and H2A histones, which have several variants with
H2A.X and canonical H2A [47,138], we hypothesize that the same
different functions, no ubiquitously expressed H4 and H2B variants
assembly factor mediates H2A.X and canonical H2A deposition.
have been reported thus far. Nevertheless, a few tissue-specific H2B
isoforms have been reported, including sperm specific H2B variant
3.2.2. The role of H2A.X in meiotic silencing
(spH2B), testis specific H2B variants TH2B and H2BFWT [157].
Aside from the well known function of H2A.X in DSB repair, H2A.X
Interestingly, TH2B was proposed to be a platform in specifying
has also been implicated to play a role in meiosis, growth, tumor
pericentric heterochromatin during late spermiogenesis [158], which
suppression and immune receptor rearrangements [141]. Interest-
maybe a good target in studying the inheritance of epigenetic states of
ingly, H2A.X knockout mice are sterile [143]. In wild type mice, during
pericentric heterochromatin during meiosis. On the other hand,
the pachytene stage of spermatogenesis γ-H2A.X is enriched at the
H2BFWT appears to be enriched at telomere interstitial blocks, and it
sex chromosomes to initiate meiotic sex chromosome inactivation
has been proposed that H2BFWT may serve as an epigenetic marker
(MSCI) and X and Y chromosome condensation, which leads to the
telomeric identity in testis [159].
formation of a partially paired sex body. In H2A.X-null mice, this
entire process was impaired [143]. Additionally, previous studies have
indicated that γ-H2A.X also plays a role in meiotic silencing of 5. Perspectives
unpaired chromosomes in female mice [144].
H3.3-deficient male flies have decreased fertility [112], and it is Histone variants are currently the new frontier of chromatin
interesting to speculate whether these meiotic defects are the results biology. However, several important questions regarding the mech-
of impaired meiotic silencing and whether H2A.X and H3.3 play anisms behind epigenetic inheritance and stability remain.
cooperative roles in these events. More studies are needed to clarify which histone variants are
involved in epigenetic regulation. Because a key criterion of epigenetics
3.3. macroH2A is that the information is heritable, it is critical to discover which histone
variants play a role in this process and which of their mediated functions
macroH2A is a vertebrate-specific H2A variant that contains two are inherited during mitotic/meiotic division.
distinct domains [145]. Although the N terminal region of macroH2A The post-translational histone modifications often recruit the
is similar to canonical H2A, macroH2A contains a large (200 residue) C associated effector proteins to exert a function [5,6]. However, few
terminal domain termed “the macro domain” that shares no sequence proteins have been discovered that can bind to and specifically
similarity with any other histone [47]. In mammals, macroH2A is recognize the variant histones in chromatin context. Several pressing
enriched on the inactive X chromosomes in females [146]. Interest- questions remain, including “How do the histone variants function once
ingly, macroH2A is re-localized from centrosomes to inactive X they are deposited?” and “Are the histone variants recognized by
chromosomes, and this re-localization is dependent on Xist expres- proteins factors like the histone modifications, or do they function solely
sion after X inactivation initiation [147,148]; however, X inactivation by altering the intrinsic properties of the underlying chromatin?”
initiation is not dependent on macroH2A. Therefore, macroH2A is
thought to be an epigenetic marker of X inactivation and may Acknowledgments
contribute to the maintenance of an inactive X chromosome. Several
studies have suggested potential mechanisms where macroH2A The research of B. Z. is supported by grants from the Chinese Ministry
represses gene expression. A previous study has shown that the C- of Science and Technology (2011CB965300 and 2007AA02Z1A6).
terminal macro domain of macroH2A interferes with the binding of
transcription factors, and the N-terminal domain impedes chromatin
References
remodeling via SWI/SNF [149]. In addition, macroH2A can mediate
gene silencing by inhibiting the catalytic activity and substrate [1] R.D. Kornberg, J.O. Thomas, Chromatin structure; oligomers of the histones,
binding capacity of PARP1, which is a nuclear enzyme that is involved Science 184 (1974) 865–868.
[2] K. Luger, A.W. Mader, R.K. Richmond, D.F. Sargent, T.J. Richmond, Crystal
in gene activation [150,151]. Moreover, additional studies have structure of the nucleosome core particle at 2.8 A resolution, Nature 389 (1997)
indicated that macroH2A is involved in autosomal gene silencing. 251–260.
G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229 227

[3] T. Kouzarides, Chromatin modifications and their function, Cell 128 (2007) polycomb protein EED in the propagation of repressive histone marks, Nature
693–705. 461 (2009) 762–767.
[4] A.J. Bannister, T. Kouzarides, Regulation of chromatin by histone modifications, [37] K.H. Hansen, A.P. Bracken, D. Pasini, N. Dietrich, S.S. Gehani, A. Monrad, J.
Cell Res. 21 (2011) 381–395. Rappsilber, M. Lerdrup, K. Helin, A model for transmission of the H3K27me3
[5] A.J. Ruthenburg, H. Li, D.J. Patel, C.D. Allis, Multivalent engagement of chromatin epigenetic mark, Nat. Cell Biol. 10 (2008) 1291–1300.
modifications by linked binding modules, Nat. Rev. Mol. Cell Biol. 8 (2007) [38] J.J. Pesavento, H. Yang, N.L. Kelleher, C.A. Mizzen, Certain and progressive
983–994. methylation of histone H4 at lysine 20 during the cell cycle, Mol. Cell. Biol. 28
[6] M. Yun, J. Wu, J.L. Workman, B. Li, Readers of histone modifications, Cell Res (2008) 468–486.
(2011). [39] A.N. Scharf, T.K. Barth, A. Imhof, Establishment of histone modifications after
[7] S. Henikoff, K. Ahmad, Assembly of variant histones into chromatin, Annu. Rev. chromatin assembly, Nucleic Acids Res. 37 (2009) 5032–5040.
Cell Dev. Biol. 21 (2005) 133–153. [40] S.M. Sweet, M. Li, P.M. Thomas, K.R. Durbin, N.L. Kelleher, Kinetics of re-
[8] E. Szenker, D. Ray-Gallet, G. Almouzni, The double face of the histone variant establishing H3K79 methylation marks in global human chromatin, J. Biol. Chem.
H3.3, Cell Res. 21 (2011) 421–434. 285 (2010) 32778–32786.
[9] S.B. Hake, C.D. Allis, Histone H3 variants and their potential role in indexing [41] X. Chen, J. Xiong, M. Xu, S. Chen, B. Zhu, Symmetrical modification within a
mammalian genomes: the “H3 barcode hypothesis”, Proc. Natl. Acad. Sci. U.S.A. nucleosome is not required globally for histone lysine methylation, EMBO Rep.
103 (2006) 6428–6435. 12 (2011) 244–251.
[10] V.E.A. Russo, R.A. Martienssen, A.D. Riggs, Epigenetic Mechanisms of Gene [42] S.M. Wiedemann, S.N. Mildner, C. Bonisch, L. Israel, A. Maiser, S. Matheisl, T.
Regulation, Cold Spring Harbor Laboratory Press, 1996. Straub, R. Merkl, H. Leonhardt, E. Kremmer, L. Schermelleh, S.B. Hake,
[11] R. Holliday, J.E. Pugh, DNA modification mechanisms and gene activity during Identification and characterization of two novel primate-specific histone H3
development, Science 187 (1975) 226–232. variants, H3.X and H3.Y, J. Cell Biol. 190 (2010) 777–791.
[12] T.H. Bestor, Activation of mammalian DNA methyltransferase by cleavage of a Zn [43] R. Schenk, A. Jenke, M. Zilbauer, S. Wirth, J. Postberg, H3.5 is a novel hominid-
binding regulatory domain, EMBO J. 11 (1992) 2611–2617. specific histone H3 variant that is specifically expressed in the seminiferous
[13] H. Leonhardt, A.W. Page, H.U. Weier, T.H. Bestor, A targeting sequence directs tubules of human testes, Chromosoma 120 (2011) 275–285.
DNA methyltransferase to sites of DNA replication in mammalian nuclei, Cell 71 [44] R.C. Allshire, G.H. Karpen, Epigenetic regulation of centromeric chromatin: old
(1992) 865–873. dogs, new tricks? Nat. Rev. Genet. 9 (2008) 923–937.
[14] L.S. Chuang, H.I. Ian, T.W. Koh, H.H. Ng, G. Xu, B.F. Li, Human DNA-(cytosine-5) [45] K. Sarma, D. Reinberg, Histone variants meet their match, Nat. Rev. Mol. Cell Biol.
methyltransferase-PCNA complex as a target for p21WAF1, Science 277 (1997) 6 (2005) 139–149.
1996–2000. [46] D.K. Palmer, K. O'Day, M.H. Wener, B.S. Andrews, R.L. Margolis, A 17-kD
[15] M. Bostick, J.K. Kim, P.O. Esteve, A. Clark, S. Pradhan, S.E. Jacobsen, UHRF1 plays a centromere protein (CENP-A) copurifies with nucleosome core particles and
role in maintaining DNA methylation in mammalian cells, Science 317 (2007) with histones, J. Cell Biol. 104 (1987) 805–815.
1760–1764. [47] H.S. Malik, S. Henikoff, Phylogenomics of the nucleosome, Nat. Struct. Biol. 10
[16] J. Sharif, M. Muto, S. Takebayashi, I. Suetake, A. Iwamatsu, T.A. Endo, J. Shinga, Y. (2003) 882–891.
Mizutani-Koseki, T. Toyoda, K. Okamura, S. Tajima, K. Mitsuya, M. Okano, H. [48] L.E. Jansen, B.E. Black, D.R. Foltz, D.W. Cleveland, Propagation of centromeric
Koseki, The SRA protein Np95 mediates epigenetic inheritance by recruiting chromatin requires exit from mitosis, J. Cell Biol. 176 (2007) 795–805.
Dnmt1 to methylated DNA, Nature 450 (2007) 908–912. [49] G. Mizuguchi, H. Xiao, J. Wisniewski, M.M. Smith, C. Wu, Nonhistone Scm3 and
[17] S. Rea, F. Eisenhaber, D. O'Carroll, B.D. Strahl, Z.W. Sun, M. Schmid, S. Opravil, K. histones CenH3-H4 assemble the core of centromere-specific nucleosomes, Cell
Mechtler, C.P. Ponting, C.D. Allis, T. Jenuwein, Regulation of chromatin structure 129 (2007) 1153–1164.
by site-specific histone H3 methyltransferases, Nature 406 (2000) 593–599. [50] Z. Zhou, H. Feng, B.R. Zhou, R. Ghirlando, K. Hu, A. Zwolak, L.M. Miller Jenkins, H.
[18] M. Lachner, D. O'Carroll, S. Rea, K. Mechtler, T. Jenuwein, Methylation of histone Xiao, N. Tjandra, C. Wu, Y. Bai, Structural basis for recognition of centromere
H3 lysine 9 creates a binding site for HP1 proteins, Nature 410 (2001) 116–120. histone variant CenH3 by the chaperone Scm3, Nature 472 (2011) 234–237.
[19] A.J. Bannister, P. Zegerman, J.F. Partridge, E.A. Miska, J.O. Thomas, R.C. Allshire, T. [51] H. Tagami, D. Ray-Gallet, G. Almouzni, Y. Nakatani, Histone H3.1 and H3.3
Kouzarides, Selective recognition of methylated lysine 9 on histone H3 by the complexes mediate nucleosome assembly pathways dependent or independent
HP1 chromo domain, Nature 410 (2001) 120–124. of DNA synthesis, Cell 116 (2004) 51–61.
[20] R. Cao, L. Wang, H. Wang, L. Xia, H. Erdjument-Bromage, P. Tempst, R.S. Jones, Y. [52] C.M. English, N.K. Maluf, B. Tripet, M.E. Churchill, J.K. Tyler, ASF1 binds to a
Zhang, Role of histone H3 lysine 27 methylation in Polycomb-group silencing, heterodimer of histones H3 and H4: a two-step mechanism for the assembly of
Science 298 (2002) 1039–1043. the H3-H4 heterotetramer on DNA, Biochemistry 44 (2005) 13673–13682.
[21] B. Czermin, R. Melfi, D. McCabe, V. Seitz, A. Imhof, V. Pirrotta, Drosophila [53] C.M. English, M.W. Adkins, J.J. Carson, M.E. Churchill, J.K. Tyler, Structural basis
enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity for the histone chaperone activity of Asf1, Cell 127 (2006) 495–508.
that marks chromosomal Polycomb sites, Cell 111 (2002) 185–196. [54] L.J. Benson, Y. Gu, T. Yakovleva, K. Tong, C. Barrows, C.L. Strack, R.G. Cook, C.A.
[22] J. Muller, C.M. Hart, N.J. Francis, M.L. Vargas, A. Sengupta, B. Wild, E.L. Miller, M.B. Mizzen, A.T. Annunziato, Modifications of H3 and H4 during chromatin
O'Connor, R.E. Kingston, J.A. Simon, Histone methyltransferase activity of a replication, nucleosome assembly, and histone exchange, J. Biol. Chem. 281
Drosophila Polycomb group repressor complex, Cell 111 (2002) 197–208. (2006) 9287–9296.
[23] A. Kuzmichev, K. Nishioka, H. Erdjument-Bromage, P. Tempst, D. Reinberg, Histone [55] R. Natsume, M. Eitoku, Y. Akai, N. Sano, M. Horikoshi, T. Senda, Structure and
methyltransferase activity associated with a human multiprotein complex contain- function of the histone chaperone CIA/ASF1 complexed with histones H3 and H4,
ing the enhancer of Zeste protein, Genes Dev. 16 (2002) 2893–2905. Nature 446 (2007) 338–341.
[24] K. Plath, J. Fang, S.K. Mlynarczyk-Evans, R. Cao, K.A. Worringer, H. Wang, C.C. de [56] U.S. Cho, S.C. Harrison, Recognition of the centromere-specific histone Cse4 by
la Cruz, A.P. Otte, B. Panning, Y. Zhang, Role of histone H3 lysine 27 methylation the chaperone Scm3, Proc. Natl. Acad. Sci. U.S.A. 108 (2011) 9367–9371.
in X inactivation, Science 300 (2003) 131–135. [57] Y. Fujita, T. Hayashi, T. Kiyomitsu, Y. Toyoda, A. Kokubu, C. Obuse, M. Yanagida,
[25] I. Okamoto, A.P. Otte, C.D. Allis, D. Reinberg, E. Heard, Epigenetic dynamics of Priming of centromere for CENP-A recruitment by human hMis18alpha,
imprinted X inactivation during early mouse development, Science 303 (2004) hMis18beta, and M18BP1, Dev. Cell 12 (2007) 17–30.
644–649. [58] E.M. Dunleavy, D. Roche, H. Tagami, N. Lacoste, D. Ray-Gallet, Y. Nakamura, Y.
[26] Y. Nakatani, D. Ray-Gallet, J.P. Quivy, H. Tagami, G. Almouzni, Two distinct Daigo, Y. Nakatani, G. Almouzni-Pettinotti, HJURP is a cell-cycle-dependent
nucleosome assembly pathways: dependent or independent of DNA synthesis maintenance and deposition factor of CENP-A at centromeres, Cell 137 (2009)
promoted by histone H3.1 and H3.3 complexes, Cold Spring Harb. Symp. Quant. 485–497.
Biol. 69 (2004) 273–280. [59] D.R. Foltz, L.E. Jansen, A.O. Bailey, J.R. Yates III, E.A. Bassett, S. Wood, B.E. Black,
[27] S. Henikoff, T. Furuyama, K. Ahmad, Histone variants, nucleosome assembly and D.W. Cleveland, Centromere-specific assembly of CENP-a nucleosomes is
epigenetic inheritance, Trends in genetics : TIG 20 (2004) 320–326. mediated by HJURP, Cell 137 (2009) 472–484.
[28] A.T. Annunziato, Split decision: what happens to nucleosomes during DNA [60] B.E. Black, D.W. Cleveland, Epigenetic centromere propagation and the nature of
replication? J. Biol. Chem. 280 (2005) 12065–12068. CENP-a nucleosomes, Cell 144 (2011) 471–479.
[29] A. Groth, W. Rocha, A. Verreault, G. Almouzni, Chromatin challenges during DNA [61] H. Hu, Y. Liu, M. Wang, J. Fang, H. Huang, N. Yang, Y. Li, J. Wang, X. Yao, Y. Shi, G.
replication and repair, Cell 128 (2007) 721–733. Li, R.M. Xu, Structure of a CENP-A-histone H4 heterodimer in complex with
[30] C. Martin, Y. Zhang, Mechanisms of epigenetic inheritance, Curr. Opin. Cell Biol. chaperone HJURP, Genes Dev. 25 (2011) 901–906.
19 (2007) 266–272. [62] D.R. Foltz, L.E. Jansen, B.E. Black, A.O. Bailey, J.R. Yates III, D.W. Cleveland, The
[31] S. Henikoff, Nucleosome destabilization in the epigenetic regulation of gene human CENP-A centromeric nucleosome-associated complex, Nat. Cell Biol.
expression, Nat. Rev. Genet. 9 (2008) 15–26. 8 (2006) 458–469.
[32] A.V. Probst, E. Dunleavy, G. Almouzni, Epigenetic inheritance during the cell [63] M. Okada, I.M. Cheeseman, T. Hori, K. Okawa, I.X. McLeod, J.R. Yates III, A. Desai, T.
cycle, Nat. Rev. Mol. Cell Biol. 10 (2009) 192–206. Fukagawa, The CENP-H-I complex is required for the efficient incorporation of
[33] M. Xu, B. Zhu, Nucleosome assembly and epigenetic inheritance, Protein Cell 1 newly synthesized CENP-A into centromeres, Nat. Cell Biol. 8 (2006) 446–457.
(2010) 820–829. [64] K.E. Gascoigne, K. Takeuchi, A. Suzuki, T. Hori, T. Fukagawa, I.M. Cheeseman,
[34] B. Zhu, D. Reinberg, Epigenetic inheritance: uncontested? Cell Res. 21 (2011) Induced ectopic kinetochore assembly bypasses the requirement for CENP-A
435–441. nucleosomes, Cell 145 (2011) 410–422.
[35] M. Xu, C. Long, X. Chen, C. Huang, S. Chen, B. Zhu, Partitioning of histone H3-H4 [65] A. Lagana, J.F. Dorn, V. De Rop, A.M. Ladouceur, A.S. Maddox, P.S. Maddox, A small
tetramers during DNA replication-dependent chromatin assembly, Science 328 GTPase molecular switch regulates epigenetic centromere maintenance by
(2010) 94–98. stabilizing newly incorporated CENP-A, Nat. Cell Biol. 12 (2010) 1186–1193.
[36] R. Margueron, N. Justin, K. Ohno, M.L. Sharpe, J. Son, W.J. Drury III, P. Voigt, S.R. [66] L. Prendergast, K.F. Sullivan, A GTPase switch maintains CENP-A at centromeric
Martin, W.R. Taylor, V. De Marco, V. Pirrotta, D. Reinberg, S.J. Gamblin, Role of the chromatin, Nat. Cell Biol. 12 (2010) 1128–1130.
228 G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229

[67] P. Ranjitkar, M.O. Press, X. Yi, R. Baker, M.J. MacCoss, S. Biggins, An E3 ubiquitin [96] F. Hu, A.A. Alcasabas, S.J. Elledge, Asf1 links Rad53 to control of chromatin
ligase prevents ectopic localization of the centromeric histone H3 variant via the assembly, Genes Dev. 15 (2001) 1061–1066.
centromere targeting domain, Mol. Cell 40 (2010) 455–464. [97] J.A. Sharp, E.T. Fouts, D.C. Krawitz, P.D. Kaufman, Yeast histone deposition
[68] G. Hewawasam, M. Shivaraju, M. Mattingly, S. Venkatesh, S. Martin-Brown, L. protein Asf1p requires Hir proteins and PCNA for heterochromatic silencing,
Florens, J.L. Workman, J.L. Gerton, Psh1 is an E3 ubiquitin ligase that targets the Curr. Biol. 11 (2001) 463–473.
centromeric histone variant Cse4, Mol. Cell 40 (2010) 444–454. [98] J.K. Tyler, K.A. Collins, J. Prasad-Sinha, E. Amiott, M. Bulger, P.J. Harte, R.
[69] G. Arents, R.W. Burlingame, B.C. Wang, W.E. Love, E.N. Moudrianakis, The Kobayashi, J.T. Kadonaga, Interaction between the Drosophila CAF-1 and ASF1
nucleosomal core histone octamer at 3.1 A resolution: a tripartite protein chromatin assembly factors, Mol. Cell. Biol. 21 (2001) 6574–6584.
assembly and a left-handed superhelix, Proc. Natl. Acad. Sci. U.S.A. 88 (1991) [99] G.A. Orsi, P. Couble, B. Loppin, Epigenetic and replacement roles of histone
10148–10152. variant H3.3 in reproduction and development, Int. J. Dev. Biol. 53 (2009)
[70] R.D. Shelby, O. Vafa, K.F. Sullivan, Assembly of CENP-A into centromeric chromatin 231–243.
requires a cooperative array of nucleosomal DNA contact sites, J. Cell Biol. 136 [100] B. Cui, Y. Liu, M.A. Gorovsky, Deposition and function of histone H3 variants in
(1997) 501–513. Tetrahymena thermophila, Mol. Cell. Biol. 26 (2006) 7719–7730.
[71] D. Vermaak, H.S. Hayden, S. Henikoff, Centromere targeting element within the [101] A. Loyola, G. Almouzni, Marking histone H3 variants: how, when and why?
histone fold domain of Cid, Mol. Cell. Biol. 22 (2002) 7553–7561. Trends Biochem. Sci. 32 (2007) 425–433.
[72] N. Sekulic, E.A. Bassett, D.J. Rogers, B.E. Black, The structure of (CENP-A-H4)(2) [102] A. Loyola, T. Bonaldi, D. Roche, A. Imhof, G. Almouzni, PTMs on H3 variants before
reveals physical features that mark centromeres, Nature 467 (2010) 347–351. chromatin assembly potentiate their final epigenetic state, Mol. Cell 24 (2006)
[73] R. Saffery, H. Sumer, S. Hassan, L.H. Wong, J.M. Craig, K. Todokoro, M. Anderson, 309–316.
A. Stafford, K.H. Choo, Transcription within a functional human centromere, Mol. [103] J.H. Waterborg, Sequence analysis of acetylation and methylation in two histone
Cell 12 (2003) 509–516. H3 variants of alfalfa, J. Biol. Chem. 265 (1990) 17157–17161.
[74] M.D. Blower, B.A. Sullivan, G.H. Karpen, Conserved organization of centromeric [104] E. McKittrick, P.R. Gafken, K. Ahmad, S. Henikoff, Histone H3.3 is enriched in
chromatin in flies and humans, Dev. Cell 2 (2002) 319–330. covalent modifications associated with active chromatin, Proc. Natl. Acad. Sci.
[75] K. Nagaki, Z. Cheng, S. Ouyang, P.B. Talbert, M. Kim, K.M. Jones, S. Henikoff, C.R. U.S.A. 101 (2004) 1525–1530.
Buell, J. Jiang, Sequencing of a rice centromere uncovers active genes, Nat. Genet. [105] L. Johnson, S. Mollah, B.A. Garcia, T.L. Muratore, J. Shabanowitz, D.F. Hunt, S.E.
36 (2004) 138–145. Jacobsen, Mass spectrometry analysis of Arabidopsis histone H3 reveals distinct
[76] B.E. Black, L.E. Jansen, P.S. Maddox, D.R. Foltz, A.B. Desai, J.V. Shah, D.W. combinations of post-translational modifications, Nucleic Acids Res. 32 (2004)
Cleveland, Centromere identity maintained by nucleosomes assembled with 6511–6518.
histone H3 containing the CENP-A targeting domain, Mol. Cell 25 (2007) [106] S.B. Hake, B.A. Garcia, E.M. Duncan, M. Kauer, G. Dellaire, J. Shabanowitz, D.P.
309–322. Bazett-Jones, C.D. Allis, D.F. Hunt, Expression patterns and post-translational
[77] D.K. Palmer, K. O'Day, R.L. Margolis, The centromere specific histone CENP-A is modifications associated with mammalian histone H3 variants, J. Biol. Chem. 281
selectively retained in discrete foci in mammalian sperm nuclei, Chromosoma (2006) 559–568.
100 (1990) 32–36. [107] H. Wu, B. Zhu, Split decision: why it matters? Front. Biol. 6 (2011) 88–92.
[78] R. Camahort, M. Shivaraju, M. Mattingly, B. Li, S. Nakanishi, D. Zhu, A. Shilatifard, [108] Y. Katan-Khaykovich, K. Struhl, Splitting of H3-H4 tetramers at transcriptionally
J.L. Workman, J.L. Gerton, Cse4 is part of an octameric nucleosome in budding active genes undergoing dynamic histone exchange, Proc. Natl. Acad. Sci. U.S.A.
yeast, Mol. Cell 35 (2009) 794–805. 108 (2011) 1296–1301.
[79] N. Conde e Silva, B.E. Black, A. Sivolob, J. Filipski, D.W. Cleveland, A. Prunell, [109] A. Santenard, C. Ziegler-Birling, M. Koch, L. Tora, A.J. Bannister, M.E. Torres-
CENP-A-containing nucleosomes: easier disassembly versus exclusive centro- Padilla, Heterochromatin formation in the mouse embryo requires critical
meric localization, J Mol Biol 370 (2007) 555–573. residues of the histone variant H3.3, Nat. Cell Biol. 12 (2010) 853–862.
[80] D.K. Palmer, R.L. Margolis, Kinetochore components recognized by human [110] N. Lacoste, G. Almouzni, Epigenetic memory: H3.3 steps in the groove, Nat. Cell
autoantibodies are present on mononucleosomes, Mol. Cell. Biol. 5 (1985) Biol. 10 (2008) 7–9.
173–186. [111] R.K. Ng, J.B. Gurdon, Epigenetic memory of an active gene state depends on
[81] J.S. Williams, T. Hayashi, M. Yanagida, P. Russell, Fission yeast Scm3 mediates histone H3.3 incorporation into chromatin in the absence of transcription, Nat.
stable assembly of Cnp1/CENP-A into centromeric chromatin, Mol. Cell 33 Cell Biol. 10 (2008) 102–109.
(2009) 287–298. [112] A. Sakai, B.E. Schwartz, S. Goldstein, K. Ahmad, Transcriptional and developmental
[82] Y. Dalal, H. Wang, S. Lindsay, S. Henikoff, Tetrameric structure of centromeric functions of the H3.3 histone variant in Drosophila, Curr. Biol. 19 (2009) 1816–1820.
nucleosomes in interphase Drosophila cells, PLoS Biol. 5 (2007) e218. [113] S.L. Pereira, J.N. Reeve, Histones and nucleosomes in Archaea and Eukarya: a
[83] T. Furuyama, S. Henikoff, Centromeric nucleosomes induce positive DNA comparative analysis, Extremophiles 2 (1998) 141–148.
supercoils, Cell 138 (2009) 104–113. [114] R.K. Suto, M.J. Clarkson, D.J. Tremethick, K. Luger, Crystal structure of a
[84] B.A. Sullivan, G.H. Karpen, Centromeric chromatin exhibits a histone modifica- nucleosome core particle containing the variant histone H2A.Z, Nat. Struct.
tion pattern that is distinct from both euchromatin and heterochromatin, Nat. Biol. 7 (2000) 1121–1124.
Struct. Mol. Biol. 11 (2004) 1076–1083. [115] G. Mizuguchi, X. Shen, J. Landry, W.H. Wu, S. Sen, C. Wu, ATP-driven exchange of
[85] H.P. Cam, T. Sugiyama, E.S. Chen, X. Chen, P.C. FitzGerald, S.I. Grewal, histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex,
Comprehensive analysis of heterochromatin- and RNAi-mediated epigenetic Science 303 (2004) 343–348.
control of the fission yeast genome, Nat. Genet. 37 (2005) 809–819. [116] N.J. Krogan, M.C. Keogh, N. Datta, C. Sawa, O.W. Ryan, H. Ding, R.A. Haw, J.
[86] I.M. Cheeseman, A. Desai, Molecular architecture of the kinetochore-microtubule Pootoolal, A. Tong, V. Canadien, D.P. Richards, X. Wu, A. Emili, T.R. Hughes, S.
interface, Nat. Rev. Mol. Cell Biol. 9 (2008) 33–46. Buratowski, J.F. Greenblatt, A Snf2 family ATPase complex required for
[87] K.S. Bloom, J. Carbon, Yeast centromere DNA is in a unique and highly ordered recruitment of the histone H2A variant Htz1, Mol. Cell 12 (2003) 1565–1576.
structure in chromosomes and small circular minichromosomes, Cell 29 (1982) [117] M.S. Kobor, S. Venkatasubrahmanyam, M.D. Meneghini, J.W. Gin, J.L. Jennings,
305–317. A.J. Link, H.D. Madhani, J. Rine, A protein complex containing the conserved
[88] K. Ahmad, S. Henikoff, The histone variant H3.3 marks active chromatin by Swi2/Snf2-related ATPase Swr1p deposits histone variant H2A.Z into euchro-
replication-independent nucleosome assembly, Mol. Cell 9 (2002) 1191–1200. matin, PLoS Biol. 2 (2004) E131.
[89] Y. Mito, J.G. Henikoff, S. Henikoff, Genome-scale profiling of histone H3.3 [118] E. Luk, N.D. Vu, K. Patteson, G. Mizuguchi, W.H. Wu, A. Ranjan, J. Backus, S. Sen, M.
replacement patterns, Nat. Genet. 37 (2005) 1090–1097. Lewis, Y. Bai, C. Wu, Chz1, a nuclear chaperone for histone H2AZ, Mol. Cell 25
[90] S. Henikoff, J.G. Henikoff, A. Sakai, G.B. Loeb, K. Ahmad, Genome-wide profiling of (2007) 357–368.
salt fractions maps physical properties of chromatin, Genome Res. 19 (2009) [119] K. Straube, J.S. Blackwell Jr., L.F. Pemberton, Nap1 and Chz1 have separate Htz1
460–469. nuclear import and assembly functions, Traffic 11 (2010) 185–197.
[91] A.D. Goldberg, L.A. Banaszynski, K.M. Noh, P.W. Lewis, S.J. Elsaesser, S. Stadler, S. [120] E. Luk, A. Ranjan, P.C. Fitzgerald, G. Mizuguchi, Y. Huang, D. Wei, C. Wu, Stepwise
Dewell, M. Law, X. Guo, X. Li, D. Wen, A. Chapgier, R.C. DeKelver, J.C. Miller, Y.L. histone replacement by SWR1 requires dual activation with histone H2A.Z and
Lee, E.A. Boydston, M.C. Holmes, P.D. Gregory, J.M. Greally, S. Rafii, C. Yang, P.J. canonical nucleosome, Cell 143 (2010) 725–736.
Scambler, D. Garrick, R.J. Gibbons, D.R. Higgs, I.M. Cristea, F.D. Urnov, D. Zheng, [121] T.N. Mavrich, I.P. Ioshikhes, B.J. Venters, C. Jiang, L.P. Tomsho, J. Qi, S.C. Schuster, I.
C.D. Allis, Distinct factors control histone variant H3.3 localization at specific Albert, B.F. Pugh, A barrier nucleosome model for statistical positioning of
genomic regions, Cell 140 (2010) 678–691. nucleosomes throughout the yeast genome, Genome Res. 18 (2008) 1073–1083.
[92] P. Drane, K. Ouararhni, A. Depaux, M. Shuaib, A. Hamiche, The death-associated [122] G.C. Yuan, Y.J. Liu, M.F. Dion, M.D. Slack, L.F. Wu, S.J. Altschuler, O.J. Rando,
protein DAXX is a novel histone chaperone involved in the replication- Genome-scale identification of nucleosome positions in S. cerevisiae, Science
independent deposition of H3.3, Genes Dev. 24 (2010) 1253–1265. 309 (2005) 626–630.
[93] L.H. Wong, J.D. McGhie, M. Sim, M.A. Anderson, S. Ahn, R.D. Hannan, A.J. George, [123] T.N. Mavrich, C. Jiang, I.P. Ioshikhes, X. Li, B.J. Venters, S.J. Zanton, L.P. Tomsho, J.
K.A. Morgan, J.R. Mann, K.H. Choo, ATRX interacts with H3.3 in maintaining Qi, R.L. Glaser, S.C. Schuster, D.S. Gilmour, I. Albert, B.F. Pugh, Nucleosome
telomere structural integrity in pluripotent embryonic stem cells, Genome Res. organization in the Drosophila genome, Nature 453 (2008) 358–362.
20 (2010) 351–360. [124] P.D. Hartley, H.D. Madhani, Mechanisms that specify promoter nucleosome
[94] L.H. Wong, H. Ren, E. Williams, J. McGhie, S. Ahn, M. Sim, A. Tam, E. Earle, M.A. location and identity, Cell 137 (2009) 445–458.
Anderson, J. Mann, K.H. Choo, Histone H3.3 incorporation provides a unique and [125] C. Jin, C. Zang, G. Wei, K. Cui, W. Peng, K. Zhao, G. Felsenfeld, H3.3/H2A.Z double
functionally essential telomeric chromatin in embryonic stem cells, Genome Res. variant-containing nucleosomes mark ‘nucleosome-free regions’ of active
19 (2009) 404–414. promoters and other regulatory regions, Nat. Genet. 41 (2009) 941–945.
[95] J.A. Mello, H.H. Sillje, D.M. Roche, D.B. Kirschner, E.A. Nigg, G. Almouzni, Human [126] S. Gupta, J. Dennis, R.E. Thurman, R. Kingston, J.A. Stamatoyannopoulos, W.S.
Asf1 and CAF-1 interact and synergize in a repair-coupled nucleosome assembly Noble, Predicting human nucleosome occupancy from primary sequence, PLoS
pathway, EMBO Rep. 3 (2002) 329–334. Comput. Biol. 4 (2008) e1000134.
G. Yuan, B. Zhu / Biochimica et Biophysica Acta 1819 (2012) 222–229 229

[127] I.P. Ioshikhes, I. Albert, S.J. Zanton, B.F. Pugh, Nucleosome positions predicted [145] J.R. Pehrson, V.A. Fried, MacroH2A, a core histone containing a large nonhistone
through comparative genomics, Nat. Genet. 38 (2006) 1210–1215. region, Science 257 (1992) 1398–1400.
[128] O.J. Rando, K. Ahmad, Rules and regulation in the primary structure of chromatin, [146] C. Costanzi, J.R. Pehrson, Histone macroH2A1 is concentrated in the inactive X
Curr. Opin. Cell Biol. 19 (2007) 250–256. chromosome of female mammals, Nature 393 (1998) 599–601.
[129] M. Papamichos-Chronakis, S. Watanabe, O.J. Rando, C.L. Peterson, Global [147] T.P. Rasmussen, M.A. Mastrangelo, A. Eden, J.R. Pehrson, R. Jaenisch, Dynamic
regulation of H2A.Z localization by the INO80 chromatin-remodeling enzyme relocalization of histone MacroH2A1 from centrosomes to inactive X chromo-
is essential for genome integrity, Cell 144 (2011) 200–213. somes during X inactivation, J. Cell Biol. 150 (2000) 1189–1198.
[130] H. Kimura, P.R. Cook, Kinetics of core histones in living human cells: little exchange [148] J.E. Mermoud, C. Costanzi, J.R. Pehrson, N. Brockdorff, Histone macroH2A1.2 relocates
of H3 and H4 and some rapid exchange of H2B, J. Cell Biol. 153 (2001) 1341–1353. to the inactive X chromosome after initiation and propagation of X-inactivation, J.
[131] B.M. Zee, R.S. Levin, B. Xu, G. LeRoy, N.S. Wingreen, B.A. Garcia, In vivo residue- Cell Biol. 147 (1999) 1399–1408.
specific histone methylation dynamics, J. Biol. Chem. 285 (2010) 3341–3350. [149] D. Angelov, A. Molla, P.Y. Perche, F. Hans, J. Cote, S. Khochbin, P. Bouvet, S.
[132] D.G. Brickner, I. Cajigas, Y. Fondufe-Mittendorf, S. Ahmed, P.C. Lee, J. Widom, J.H. Dimitrov, The histone variant macroH2A interferes with transcription factor
Brickner, H2A.Z-mediated localization of genes at the nuclear periphery confers binding and SWI/SNF nucleosome remodeling, Mol. Cell 11 (2003) 1033–1041.
epigenetic memory of previous transcriptional state, PLoS Biol. 5 (2007) e81. [150] D.A. Nusinow, I. Hernandez-Munoz, T.G. Fazzio, G.M. Shah, W.L. Kraus, B.
[133] S. Kundu, P.J. Horn, C.L. Peterson, SWI/SNF is required for transcriptional Panning, Poly(ADP-ribose) polymerase 1 is inhibited by a histone H2A variant,
memory at the yeast GAL gene cluster, Genes Dev. 21 (2007) 997–1004. MacroH2A, and contributes to silencing of the inactive X chromosome, J. Biol.
[134] K. Lemieux, M. Larochelle, L. Gaudreau, Variant histone H2A.Z, but not the HMG Chem. 282 (2007) 12851–12859.
proteins Nhp6a/b, is essential for the recruitment of Swi/Snf, Mediator, and SAGA to [151] K. Ouararhni, R. Hadj-Slimane, S. Ait-Si-Ali, P. Robin, F. Mietton, A. Harel-Bellan,
the yeast GAL1 UAS(G), Biochem. Biophys. Res. Commun. 369 (2008) 1103–1107. S. Dimitrov, A. Hamiche, The histone variant mH2A1.1 interferes with
[135] D. Rangasamy, L. Berven, P. Ridgway, D.J. Tremethick, Pericentric heterochro- transcription by down-regulating PARP-1 enzymatic activity, Genes Dev. 20
matin becomes enriched with H2A.Z during early mammalian development, (2006) 3324–3336.
EMBO J. 22 (2003) 1599–1607. [152] M. Buschbeck, I. Uribesalgo, I. Wibowo, P. Rue, D. Martin, A. Gutierrez, L. Morey,
[136] D. Rangasamy, I. Greaves, D.J. Tremethick, RNA interference demonstrates a R. Guigo, H. Lopez-Schier, L. Di Croce, The histone variant macroH2A is an
novel role for H2A.Z in chromosome segregation, Nat. Struct. Mol. Biol. 11 (2004) epigenetic regulator of key developmental genes, Nat. Struct. Mol. Biol. 16
650–655. (2009) 1074–1079.
[137] M.D. Meneghini, M. Wu, H.D. Madhani, Conserved histone variant H2A.Z protects [153] B.P. Chadwick, H.F. Willard, A novel chromatin protein, distantly related to
euchromatin from the ectopic spread of silent heterochromatin, Cell 112 (2003) histone H2A, is largely excluded from the inactive X chromosome, J. Cell Biol. 152
725–736. (2001) 375–384.
[138] P.B. Talbert, S. Henikoff, Histone variants—ancient wrap artists of the epigenome, [154] Y. Bao, K. Konesky, Y.J. Park, S. Rosu, P.N. Dyer, D. Rangasamy, D.J. Tremethick, P.J.
Nat. Rev. Mol. Cell Biol. 11 (2010) 264–275. Laybourn, K. Luger, Nucleosomes containing the histone variant H2A.Bbd
[139] J.A. Downs, N.F. Lowndes, S.P. Jackson, A role for Saccharomyces cerevisiae organize only 118 base pairs of DNA, EMBO J. 23 (2004) 3314–3324.
histone H2A in DNA repair, Nature 408 (2000) 1001–1004. [155] T. Gautier, D.W. Abbott, A. Molla, A. Verdel, J. Ausio, S. Dimitrov, Histone variant
[140] E.P. Rogakou, W. Nieves-Neira, C. Boon, Y. Pommier, W.M. Bonner, Initiation of H2ABbd confers lower stability to the nucleosome, EMBO Rep. 5 (2004)
DNA fragmentation during apoptosis induces phosphorylation of H2AX histone 715–720.
at serine 139, J. Biol. Chem. 275 (2000) 9390–9395. [156] J. Zhou, J.Y. Fan, D. Rangasamy, D.J. Tremethick, The nucleosome surface
[141] O. Fernandez-Capetillo, A. Lee, M. Nussenzweig, A. Nussenzweig, H2AX: the regulates chromatin compaction and couples it with transcriptional repression,
histone guardian of the genome, DNA Repair (Amst) 3 (2004) 959–967. Nat. Struct. Mol. Biol. 14 (2007) 1070–1076.
[142] K. Heo, H. Kim, S.H. Choi, J. Choi, K. Kim, J. Gu, M.R. Lieber, A.S. Yang, W. An, FACT- [157] A.A. Gineitis, I.A. Zalenskaya, P.M. Yau, E.M. Bradbury, A.O. Zalensky, Human
mediated exchange of histone variant H2AX regulated by phosphorylation of sperm telomere-binding complex involves histone H2B and secures telomere
H2AX and ADP-ribosylation of Spt16, Mol. Cell 30 (2008) 86–97. membrane attachment, J. Cell Biol. 151 (2000) 1591–1598.
[143] O. Fernandez-Capetillo, S.K. Mahadevaiah, A. Celeste, P.J. Romanienko, R.D. [158] J. Govin, E. Escoffier, S. Rousseaux, L. Kuhn, M. Ferro, J. Thevenon, R. Catena, I.
Camerini-Otero, W.M. Bonner, K. Manova, P. Burgoyne, A. Nussenzweig, H2AX is Davidson, J. Garin, S. Khochbin, C. Caron, Pericentric heterochromatin repro-
required for chromatin remodeling and inactivation of sex chromosomes in male gramming by new histone variants during mouse spermiogenesis, J. Cell Biol.
mouse meiosis, Dev. Cell 4 (2003) 497–508. 176 (2007) 283–294.
[144] J.M. Turner, S.K. Mahadevaiah, O. Fernandez-Capetillo, A. Nussenzweig, X. Xu, [159] D. Churikov, J. Siino, M. Svetlova, K. Zhang, A. Gineitis, E. Morton Bradbury, A.
C.X. Deng, P.S. Burgoyne, Silencing of unsynapsed meiotic chromosomes in the Zalensky, Novel human testis-specific histone H2B encoded by the interrupted
mouse, Nat. Genet. 37 (2005) 41–47. gene on the X chromosome, Genomics 84 (2004) 745–756.

You might also like