You are on page 1of 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/38075441

A moment's pause: Putative nucleosome-based influences on MeCP2 regulation

Article  in  Biochemistry and Cell Biology · October 2009


DOI: 10.1139/O09-054 · Source: PubMed

CITATIONS READS
3 104

2 authors, including:

Juan Ausio
University of Victoria
328 PUBLICATIONS   10,821 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Reproductive health assessment View project

Nuclear chaperones View project

All content following this page was uploaded by Juan Ausio on 01 June 2014.

The user has requested enhancement of the downloaded file.


791

MINIREVIEW / MINISYNTHÈSE

A moment’s pause: putative nucleosome-based


influences on MeCP2 regulation1
Anita A. Thambirajah and Juan Ausió

Abstract: There has been a hotbed of activity surrounding MeCP2 research in the past number of years. Despite better
characterizing the functions and nature of this protein, it has become abundantly clear that MeCP2 is involved in far more
complex activities than perhaps initially anticipated. Recent publications have shown that MeCP2 is dynamically post-
translationally modified, and it is possible that these marks permit MeCP2 to inhabit very diverse chromatin environments.
It is also of interest to consider how nucleosome composition differs in these varying chromatin regions, and how the
chromatin template itself contributes to diversifying the regulatory roles of MeCP2. These will be critical points to exam-
ine when seeking to understand how MeCP2 behaviour differentiates in tissues other than the brain. By understanding the
chromatin and (or) tissue context in which MeCP2 interacts, it may be possible to discern the specific etiology of diseases
linked to MeCP2 dysfunction.
Key words: MeCP2, Rett syndrome, histone variants, post-translational modifications, chromatin.
Résumé : Il y a eu un foisonnement d’activités entourant la recherche sur MeCP2 au cours des dernières années. Malgré
une meilleure caractérisation des fonctions et de la nature de cette protéine, il est apparu très clairement que MeCP2 est
impliquée dans des activités beaucoup plus complexes que peut-être d’abord anticipé. Des publications récentes ont montré
que MeCP2 est modifiée après sa traduction de façon dynamique et qu’il est possible que ces marques puissent permettre
à MeCP2 de séjourner dans différents environnement chromatiniens. Il est aussi d’un intérêt supplémentaire de considérer
comment la composition du nucléosome diffère dans ces différentes régions de la chromatine et comment la matrice chro-
matinienne elle-même peut contribuer à diversifier les rôles régulateurs de MeCP2. Ceci constituera les points critiques à
examiner en cherchant à comprendre comment le comportement de MeCP2 se différencie dans les tissus autres que le cer-
veau. En comprenant le contexte chromatinien ou tissulaire dans lequel MeCP2 interagit, il peut être possible de discerner
l’étiologie spécifique de maladies liées à la dysfonction de MeCP2.
Mots-clés : MeCP2, syndrome de Rett, variants d’histone, modifications post-traductionnelles, chromatine.
[Traduit par la Rédaction]

Introduction ger thought to provide only a structural scaffolding role;


rather, increasing evidence demonstrates that these proteins
Current understanding of chromatin structure has changed have a tremendous informational capacity and have an in-
diametrically from historical perspectives, which considered trinsic ability to modulate the control of gene expression
chromatin to be a static, unchanging entity. Chromatin, a (Jenuwein and Allis 2001; Shogren-Knaak et al. 2006). Part
complex macromolecular nucleoprotein entity, consists of of this is exerted by a combination of post-translational
DNA associated with both histone and nonhistone proteins. modifications (PTMs) to histones and (or) the inclusion of
A highly plastic environment, the chromatin landscape is an histone variants within the nucleosome structure. However,
undulating one, constantly changing in response to transcrip- these are not the only ways in which chromatin conforma-
tional requirements or external stimuli. Histones are no lon- tions are modulated. Chemical modification of DNA through
Received 11 March 2009. Revision received 29 May 2009. Accepted 2 June 2009. Published on the NRC Research Press Web site at
bcb.nrc.ca on 9 September 2009.
Abbreviations: BDNF, brain-derived neurotrophic factor; CaMK, calcium/calmodulin-dependent kinases; MBD, methyl-CpG binding
domain; MeCP2, methyl-CpG binding protein 2; PTM, post-translational modification; RTT, Rett syndrome; SUMO, small ubiquitin-
related modifier; SWI/SNF, switch mating type/sucrose nonfermenting.
A.A. Thambirajah and J. Ausió.2 Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 3P6,
Canada.
1This paper is one of a selection of papers published in this Special Issue, entitled 30th Annual International Asilomar Chromatin and
Chromosomes Conference, and has undergone the Journal’s usual peer review process.
2Corresponding author (e-mail: jausio@uvic.ca).

Biochem. Cell Biol. 87: 791–798 (2009) doi:10.1139/O09-054 Published by NRC Research Press
792 Biochem. Cell Biol. Vol. 87, 2009

methylation is another means, as is the concerted direct (or activity (Fuks et al. 2003). Further to this, confocal micro-
indirect) action of trans-acting factors and chromatin-associ- scopy of MeCP2 showed that it associated with (pericentro-
ated proteins on the chromatin template (Probst et al. 2009; meric) heterochromatin regions in a variety of mouse
Tost 2009; Wu et al. 2009). tissues, including, but not limited to, the cortex and cerebel-
As we begin to recognize the importance of developmen- lum, cells undergoing myogenic differentiation, and fibro-
tally timed and regulated gene expression, it will be impera- blasts (Agarwal et al. 2007; Brero et al. 2005; Collins et al.
tive to understand the chromatin context that facilitates these 2004; Kumar et al. 2008; Marchi et al. 2007). Similar inves-
changes. This will require discernment of not just the nucle- tigations using MCF-7 cells, a human breast cancer cell line,
osomal architecture, but also of the nonhistone chromatin showed that MeCP2 had a granular distribution throughout
proteins that mediate changes to chromatin condensation. the nucleus (Koch and Strätling 2004). These cytological
This minireview will restrict its attention to the chromatin- variations may reflect differences in the cell type, as the
binding protein methyl-CpG binding protein 2 (MeCP2) and murine and human MeCP2 share a high sequence similarity
some outstanding questions that reflect upon the chromatin (Thambirajah et al. 2009a).
composition factors that influence and differentiate MeCP2 A new perspective on MeCP2 behaviour began to emerge
interactions. at the end of 2007 and the beginning of 2008 when 2 groups
independently showed that the majority of MeCP2-bound
The early years: MeCP2 promoters were, in fact, from active genes (Chahrour et al.
2008; Yasui et al. 2007). In addition to this, a small percent-
During the 1990s, Adrian Bird’s group in Edinburgh dis- age of promoters associated with MeCP2 were transcription-
covered a then-unique family of proteins that were able to ally silent (Yasui et al. 2007). Indeed, fractionation of HeLa
bind to methylated DNA (Hendrich and Bird 1998; Lewis S3 chromatin showed that the majority of MeCP2 is present
et al. 1992). Termed the methyl-CpG binding domain in the more nuclease-accessible, active regions of chromatin
(MBD) family of proteins, these proteins had not been pre- (Ishibashi et al. 2008). Furthermore, a smaller portion of
viously characterized. Since then, 1 member, MeCP2, has MeCP2 was associated with heterochromatin (Ishibashi et
perhaps become perhaps the most well-studied MBD protein al. 2008). Although these recent findings may seem dichoto-
(Meehan et al. 1992). Early work in vitro demonstrated that mous, compared with earlier findings, the association of
MeCP2 is able to bind to reconstituted chromatin templates MeCP2 with active promoters and its purported role in gene
through its MBD, and compete with H1 binding (Nan et al. repression are not necessarily mutually exclusive. It may be
1997, 1993). MeCP2 has independently been shown to bind that MeCP2 represses gene expression in a temporally de-
to the DNA linker region, and does so preferentially within pendent, cyclical manner. The latter seems to be the case at
10 base pairs of the DNA entry and exit site of the nucleo- the promoters of inducible genes (Métivier et al. 2008).
some (Ishibashi et al. 2008; Nikitina et al. 2007). The pref- What is clear, however, is that MeCP2 does play a role in
erence for binding to a longer DNA linker was shown in the dynamic regulation of specific gene activity. Possible
vivo through the sedimentation of native mononucleosomes support of this proposed idea is illustrated in the following
to which MeCP2 was bound (Ishibashi et al. 2008). While paragraph.
MeCP2 can bind to unmethylated DNA, it preferentially Perhaps one of the best examples of MeCP2 target genes
binds to (reconstituted) methylated DNA templates in the is brain-derived neurotrophic factor (BDNF). MeCP2 binds
presence of competitor DNA (Ishibashi et al. 2008; Meehan to the third promoter of BDNF and, upon (calcium influx
et al. 1992). MeCP2 binds to its methylated target with a or) KCl-triggered membrane depolarization, MeCP2 be-
variable affinity and can exchange with chromatin, in a way comes phosphorylated via a CaMK II-dependent pathway
similar to linker histone H1 (Klose et al. 2005; Meehan et (Chen et al. 2003; Martinowich et al. 2003). MeCP2 is sub-
al. 1992; Nan et al. 1993). Because the 2 proteins compete sequently released from the promoter, and transcription en-
for the same region of the nucleosome, it is not surprising sues. Certainly, given the abundance of information on
that MeCP2 can displace H1 (Nan et al. 1997). BDNF and its importance in neuronal development and
Coimmunoprecipitation studies showed that MeCP2 inter- maintenance, this is not a target that one would expect
acts with histone deacetylases and Sin3a through its tran- MeCP2 to permanently silence. Gene repression would need
scriptional repressor domain, and this helped establish its to be relieved in response to specific stimuli and at particu-
role as a transcriptional repressor, through its ability to re- lar times. But does MeCP2 always regulate in this manner?
cruit the aforementioned interacting partners (Jones et al.
1998; Nan et al. 1998; Suzuki et al. 2003). MeCP2 has sim-
ilarly been shown to interact with heterochromatin protein 1
MeCP2 binding requirements and dynamics
(Agarwal et al. 2007). However, in the absence of salt, Any discourse on how MeCP2 may differentially regulate
MeCP2 was able to condense chromatin fibres in vitro, inde- genes, particularly those located in distinct chromatin re-
pendent of any other factors (Georgel et al. 2003). MeCP2 gions (euchromatin or heterochromatin), should be prefaced
has also been shown to interact with the Brahma subunit of by a discussion of what is known about the MeCP2-chroma-
the SWI/SNF complex, although this interaction can vary, tin binding constraints. As mentioned, MeCP2 binds sym-
depending on the cell type investigated (Harikrishnan et al. metrically methylated 5’-CpG regions that are proximal to
2005; Hu et al. 2006). Interactions between MeCP2 and at least 4 adenine or thymine nucleotides in the adjacent se-
other chromatin proteins, such as histone methyltransferases quence (Klose et al. 2005). However, the necessity for me-
and DNA methyltransferases, also support the notion that thylated DNA is not an absolute requirement. MeCP2 can
MeCP2 activities are associated with the repression of gene bind to (unmethylated) DNA 4-way junctions or cruciform-

Published by NRC Research Press


Thambirajah and Ausió 793

type structures, which approximates well the observations It is evident that the way MeCP2 is post-translationally
that MeCP2 binds to the linker DNA close to the nucleo- modified strongly influences MeCP2 regulatory behaviour
some dyad axis of symmetry (Galvão and Thomas 2005). in specific chromatin locales and in response to various
A solution structure of the MeCP2 MBD, determined us- stimuli impinging on different signalling pathway cascades.
ing nuclear magnetic resonance spectroscopy, showed that it It is possible that S80 phosphorylation may allow MeCP2
formed an asymmetric wedge-shaped structure comprising an to bind chromatin in resting-state neurons or cells, while
anti-parallel b-sheet with 4 strands on one side; the other side S421 phosphorylation facilitates MeCP2 discharge following
was more helical in nature (Wakefield et al. 1999). Based on depolarization and transcriptional induction (Fig. 1). When
the nature of the structure, it was suggested that the symme- considering the array of other phosphorylation sites identi-
try of the binding target was not key in recognition. Further- fied, it is possible that a plethora of other PTM combina-
more, it was postulated that a conserved hydrophobic pocket tions exist, which would further diversify the portfolio of
within the b-sheet was in close proximity to the methyl MeCP2 epigenetic effects. These effects may not be limited
groups of the cytosines (Wakefield et al. 1999). Similarly, to MeCP2-based interactions with chromatin, but may ex-
the structure of the MBD for chicken MeCP2 has been deter- tend into other known (or unknown) functions, including
mined (Heitmann et al. 2003). However, a recent publication the involvement of MeCP2 in messenger RNA splicing
suggested that recognition of the MeCP2 MBD for methy- (Buschdorf and Strätling 2004; Young et al. 2005).
lated DNA involves the hydrated DNA major groove, and PTM of MeCP2 is not the only means by which specific
not the methylated cytosines themselves (Ho et al. 2008). Us- MeCP2 behaviour may be modulated in relation to chroma-
ing methylated DNA-MBD cocrystals and X-ray analysis, the tin. The development of the histone code hypothesis (Jenu-
hydrophilic MBD region, in the presence of water molecules, wein and Allis 2001; Jiang et al. 2008; Tsanev and Sendov
associates with the methyl groups (Ho et al. 2008). The crys- 1971) and the emergence of inter-related epigenetic effects
tal structure of the full-length MeCP2 has not yet been deter- (Fischle 2008) underscore yet another level of complexity
mined. This is due in part, perhaps, to the fact that MeCP2 in our understanding of how MeCP2 elicits its own out-
exhibits a high degree of intrinsic disorder (Adams et al. comes. A lot of focus has been paid to the methylation sta-
2007). Despite this lack of detailed information, it is of inter- tus of the DNA that MeCP2 recognizes, but little attention
est to understand what other factors influence and modulate has been paid to the local nucleosome composition. Using a
the binding of MeCP2 to its target sequences. mouse model containing a genetically disrupted MeCP2 lo-
cus, no global changes in H3 or H4 acetylation or methyla-
Places to go, genes to regulate: the tion were observed (Urdinguio et al. 2007). Specific
modifications investigated include trimethylation of histone
ubiquitous MeCP2 H3 at lysine 9 (H3K9me3), H4K20me3, and H3K79me2, for
Early studies showed that phosphorylation of MeCP2 re- which no differences for any PTM were observed between
sulted in its release from its target sequence, resulting in wild-type and MeCP2-null mice. Despite this, the local dis-
gene transcription. It has also been shown that phosphoryla- tribution of these PTMs at pertinent gene promoters remains
tion of MeCP2 at S421, in response to neuron depolariza- uncharacterized. It is well appreciated that the presence of
tion, releases it from chromatin, permitting transcription PTMs on histone tail regions, or the incorporation of histone
(Zhou et al. 2006) (Fig. 1). The ability to phosphorylate this variants, can modulate local chromatin structure through ei-
residue is important for dendrite branching and neuron ma- ther direct structural changes (Ausio 2006) or the recruit-
turation, and appears to occur exclusively in the brain. Other ment of remodelling machinery and other chromatin-
phosphorylated residues have been observed, such as S80 interacting partners (Wu et al. 2009). By doing so, the nu-
and S229; more recently, the list has expanded to include cleosome can respond to a wide array of replication- or tran-
T148/S149, S164, and S424 (Tao et al. 2009; Zhou et al. scription-dependent requirements. Is there a nucleosomal
2006). Like S421, the functional relevance of phosphoryla- signature, based on variable histone composition, that
tion at S80 has been characterized. Interestingly, in contrast MeCP2 recognizes? How do PTMs of MeCP2 influence this
to S421, S80 becomes dephosphorylated following depolari- identification? Through the synergistic combination of varia-
zation, and this is correlated to a response in neuronal activ- ble DNA methylation, histone PTM or variant incorporation,
ity. S80 dephosphorylation also attenuates the association of and MeCP2 PTMs, a broad range of possibilities exist to
MeCP2 with chromatin (Tao et al. 2009) (Fig. 1). These 2 fine-tune the response of MeCP2 to specific stimuli or tran-
polar phosphorylation events do not appear to directly affect scriptional needs in either particular chromatin locations or
each other; in fact, they are likely the result of 2 separate cell types. This regulation may become even more complex
signalling cascades or regulatory events. While inhibition of when the 2 splice variant isoforms of MeCP2, MeCP2e1 and
CaMK II and CaMK IV activities ablated S421 phosphoryla- MeCPe2, which are differentially expressed in the brain,
tion, S80 dephosphorylation was not affected (Tao et al. compared with other tissues, are considered (Kriaucionis
2009). The kinase responsible for S80 phosphorylation has and Bird 2004; Mnatzakanian et al. 2004).
not been ascertained as yet, nor has the potential phospha- There has been a strong neurocentric focus to MeCP2 re-
tase responsible for its dephosphorylation. The analysis of search, particularly in the functional studies of MeCP2.
S80A mutants of MeCP2 revealed the presence of locomotor However, MeCP2 is a ubiquitously expressed protein and is
deficits in affected mice and the reduced association of expressed in many, if not most, tissues of the body (Shahba-
MeCP2 with several euchromatin gene promoters in resting zian et al. 2002; Zhou et al. 2006; A.A. Thambirajah, un-
neurons. The latter did not alter the association of MeCP2 published results). Presuming that MeCP2 regulates the
with heterochromatin (Tao et al. 2009). same set of genes, regardless of tissue origin, the question

Published by NRC Research Press


794 Biochem. Cell Biol. Vol. 87, 2009

Fig. 1. Potential nucleosome composition factors that could influence methyl-CpG binding protein 2 (MeCP2) association and regulatory
behaviour. (A) A transcriptionally inert state involving MeCP2 requires the binding of the protein to its methylated target sequence, and also
the phosphorylation of MeCP2 at S80 (Tao et al. 2009). As MeCP2 has been shown to interact with the histone methyltransferase responsi-
ble for methylation of histone H3 at lysine 9 (H3K9) (Fuks et al. 2003), and as the H3 N-terminal tail binds to the linker DNA (Leuba et al.
1998), it is possible that this mark may be in close association with MeCP2. Similarly, other post-translational modifications (PTMs) mark-
ing inactive states, such as H3K27 trimethylation (H3K27me3) for facultative heterochromatin, may also mark nucleosomes proximal to the
MeCP2 binding site. The H3 tail regions and potential PTM sites (small ovals) are indicated. It is also possible that nucleosomes close to
the binding site of MeCP2 contain histone variants, such as H2A.X or H2A.Z. The latter variant has been shown to be present in the 5’ ends
of promoters (Raisner et al. 2005). The potential H2A variant C-terminal tail is shown. (B) Following the initiation of transcriptional acti-
vation, MeCP2 is phosphorylated at S421 and is released from its binding site (Zhou et al. 2006). The loss of MeCP2 may also occur con-
current with the demethylation of the previously modified 5’-CpG (Métivier et al. 2008). The H3K4me2 PTM, which marks active
chromatin regions (Benevolenskaya 2007), may now be incorporated into these nucleosomes. Furthermore, as demethylation has been
shown to occur through base excision, it is possible that H2A.X, if nearby, may become phosphorylated. Also, although not necessarily in
the same nucleosome, H2A.Z may become acetylated; this would then be associated with the active state of the promoter region. Potential
H2A PTMs are indicated by ovals.

of how MeCP2 differentially regulates or silences these patients. One study found an increase in H4K16 acetylation
genes in different tissues arises. In nonbrain tissues, does with no changes in H3 acetylation, while another study
MeCP2 terminally silence brain-specific genes that it would found no changes in global acetylation of histone H3 or H4
otherwise transiently repress in the brain? Are there different (Balmer et al. 2002; Wan et al. 2001). Adding to the com-
nucleosomal cues to signal or facilitate such transcriptional plexity of these findings is another study, which detected no
outcomes? It is likely that seemingly ‘‘brain-specific’’ genes changes in H4 PTMs, but did detect differences in H3 meth-
are not exclusively expressed in the brain; rather, they may ylation and acetylation (Kaufmann et al. 2005). Whether
be expressed in other tissues for alternative functions. This these changes are directly dependent upon a MeCP2 defi-
has been shown for certain transcripts and protein forms of ciency, or even the type of MeCP2 mutation, is of particular
BDNF and other neurotropins (Lomen-Hoerth and Shooter interest, but they may not necessarily be the cause. Perhaps
1995; Prakash et al. 2009), but such expression patterns the more relevant question is: What are the histone PTMs
may not apply to all potential tissue-specific genes regulated that influence MeCP2 interaction?
by MeCP2. It is also conceivable that MeCP2 may not regu- What is (are) the nucleosomal signature(s) that MeCP2
late or even associate with the same gene(s) in different tis- recognizes? Is MeCP2 able to mediate post-translational
sues as it does in the brain. Regardless of how and what changes to its environment? MeCP2 has been shown to in-
target genes MeCP2 binds to in a given tissue, the PTMs of teract with the histone methyltransferase responsible for me-
MeCP2 and the local nucleosome environment may be fun- diating the methylation of H3K9. By doing so, H3K9
damental in refining the regulatory role of MeCP2. becomes methylated within the chromatin region that
MeCP2 is associated with (Fuks et al. 2003). But are there
MeCP2 and its nucleosomal signature other modified or variant histones that MeCP2 recognizes?
Although no others have yet been thoroughly explored, there
As mentioned earlier, no studies have yet addressed what are some strong potential candidates. In consideration of re-
parameters of nucleosome composition are critical for cent findings that MeCP2 is predominantly associated with
MeCP2 binding, other than DNA methylation. Some studies active genes, it may be prudent to consider what chromatin
have investigated the effects of disease (Rett syndrome conditions prime particular gene regions for activation. Bi-
(RTT))-relevant mutations of MeCP2 on histone PTMs, but valent modification of histone tails have been observed to
no clear trends were outstanding. A comparison of different predispose the proximal gene to either activation or silenc-
brain regions (midbrain, cortex, and cerebellum) of MeCP2- ing, depending on the transcriptional cue received. H3K27
knockout and wild-type mice did not reveal in any signifi- trimethylation (H3K27me3, silencing) and H3K4 dimethyla-
cant differences in histone H3 or H4 acetylation or methyla- tion (H3K4me2, activation) is one such example (Delcuve et
tion (Urdinguio et al. 2007). However, contrasting results al. 2009; Gan et al. 2007; Soshnikova and Duboule 2008). If
were observed from samples or cell lines derived from RTT the gene becomes poised for activation, the H3K4me2 mark

Published by NRC Research Press


Thambirajah and Ausió 795

is perpetuated, while the H3K27me3 PTM is lost; the oppo- acterized; these include significant deficits in dendritic and
site is true for the converse scenario (Fig. 1). As research major neuron morphology (Armstrong et al. 1998; Jugloff et
progresses into the study of histone PTMs, it is apparent al. 2005). As well studied as the effects of MeCP2 muta-
that there is an interplay and cross-talk between histone tions, overexpression, and knockouts on neurons have been,
PTMs and DNA methylation (Fischle 2008). It is quite there has been little attention paid to other cell types, much
likely that the nature of the chromatin environment that less other tissue types. However, a recent paper showed that
MeCP2 interacts with will be more complex than what this glia cells harboured mutated MeCP2 forms (Ballas et al.
discussion anticipates. 2009). Moreover, mutant MeCP2 astrocytes affected the
The other consideration is the histone variant partners that dendritic morphology of both wild-type and mutated
MeCP2 may have. A recent study demonstrated the cycling MeCP2 neurons grown under in vitro conditions. It was sug-
of MeCP2 and MBD2 deposition at activated promoters, gested that this conferred damage to otherwise healthy neu-
which was accompanied by the demethylation of these pro- rons was the result of aberrantly secreted factors (Ballas et
moter regions. Of interest was that this demethylation oc- al. 2009). This recent finding underscores an important
curred through base excision of the modified nucleotides point: it is not only neurons that are affected by abnormal-
(Métivier et al. 2008). With well-documented roles in dou- ities arising from MeCP2; other cell types and potentially
ble-strand break repair and in neuronal base-excision repair different tissues are also affected. Should the latter prove to
(Crowe et al. 2006; Vasko et al. 2005), is it possible that be true, it may prompt a reconsideration of the way RTT eti-
H2A.X, and particularly phosphorylated H2A.X, may be lo- ology is perceived. For instance, the constellation of symp-
calized within these regions (Thambirajah et al. 2009b). toms experienced by girls with RTT, including respiratory
Moreover, is there a close association between MeCP2 and and gastrointestinal difficulties, may not be just a corollary
H2A.X? Another histone variant candidate could be H2A.Z. of brain dysfunction (Isaacs et al. 2003). Rather, such symp-
Although there is a multitude of putative roles ascribed to toms may arise from a localized dysregulation within the af-
this histone variant, H2A.Z is incorporated into nucleosomes fected tissues due to MeCP2 mutations. Of course, it is also
located within the boundaries between euchromatin and het- possible that these irregularities may ensue from improper
erochromatin, and also at the promoters of active genes autonomous function. Regardless of the primary cause of
(Meneghini et al. 2003; Raisner et al. 2005). With the latter, these other symptoms, it is quite possible that, if combined,
H2A.Z is often found to be acetylated (Bruce et al. 2005). Is both factors could exacerbate the symptom severity.
it possible that such nucleosomes may be the sites of
MeCP2 interactions or may be localized proximal to these Conclusion
binding sequences (Fig. 1)? Because of active research efforts into MeCP2 and RTT,
It is likely that the differential association of MeCP2 with much has been elucidated about the way this protein oper-
nucleosomes containing a combination of different PTMs ates at the molecular level, and of its downstream effects on
and histone variants may specialize the transcriptional role a larger physiological scale. However, as more information
that MeCP2 takes on. This may pertain to its partitioning emerges about the involvement of MeCP2 in chromatin-
within different chromatin fractions (i.e., euchromatin or based phenomena and cellular development, it is apparent
heterochromatin), which would strongly correlate to whether that this protein does not behave in a clear-cut manner, as
or not its bound genes are transiently active or terminally re- was once anticipated. While this protein might continue to
pressed (Henikoff et al. 2009). Furthermore, this differentia- surprise, it may be that, in the coming years, further work
tion would have critical implications for the temporal- and in this area could become a gateway through which several
tissue-dependent regulation of gene activity. Thus, it is con- new areas of research endeavours coalesce. It is apparent
ceivable that by simply changing the PTM status of MeCP2 that, even at the molecular level, the interplay between the
and the nucleosome composition, genes that are active in PTMs of MeCP2, the status of DNA methylation, and the
one tissue could be silenced in another. SUMOylation of nucleosome composition may all strongly influence the tran-
MeCP2 has been demonstrated (Miyake and Nagai 2007), scriptional states of particular genes, depending on the cues
and a recent publication has proposed a hypothesis regarding received and the tissue in question. To better understand the
the regulation of MeCP2 turnover through PTM (Thambir- context of MeCP2 function, it will be imperative to fathom
ajah et al. 2009a). There are potentially other yet-to-be-de- the interplay between these superimposed layers, and not
termined PTMs of MeCP2 that could further diversify the only to consider them in isolation.
nuanced regulatory roles MeCP2 undertakes.
References
Implications for Rett syndrome Adams, V.H., McBryant, S.J., Wade, P.A., Woodcock, C.L., and
Hansen, J.C. 2007. Intrinsic disorder and autonomous domain
As alluded to earlier, mutations in MeCP2 result in ap- function in the multifunctional nuclear protein, MeCP2. J. Biol.
proximately 90% of all diagnosed incidences of RTT. RTT, Chem. 282(20): 15057–15064. doi:10.1074/jbc.M700855200.
perhaps one of the best-studied diseases of the broad spec- PMID:17371874.
trum of autism disorders, predominantly afflicts young girls Agarwal, N., Hardt, T., Brero, A., Nowak, D., Rothbauer, U.,
who develop a diverse and wide range of postnatal cognitive Becker, A., et al. 2007. MeCP2 interacts with HP1 and modu-
and physical disabilities (Hagberg et al. 1983; Rett 1966). lates its heterochromatin association during myogenic differen-
RTT is considered a neurodevelopmental disorder, in part tiation. Nucleic Acids Res. 35(16): 5402–5408. doi:10.1093/nar/
based on the presenting clinical pathology. Neuronal abnor- gkm599. PMID:17698499.
malities, arising from MeCP2 dysfunction, have been char- Armstrong, D.D., Dunn, K., and Antalffy, B. 1998. Decreased den-

Published by NRC Research Press


796 Biochem. Cell Biol. Vol. 87, 2009

dritic branching in frontal, motor and limbic cortex in Rett syn- MeCP2 to four-way junction DNA through its methyl CpG-
drome compared with trisomy 21. J. Neuropathol. Exp. Neurol. binding domain. Nucleic Acids Res. 33(20): 6603–6609. doi:10.
57(11): 1013–1017. doi:10.1097/00005072-199811000-00003. 1093/nar/gki971. PMID:16314321.
PMID:9825937. Gan, Q., Yoshida, T., McDonald, O.G., and Owens, G.K. 2007.
Ausio, J. 2006. Histone variants—the structure behind the function. Concise review: epigenetic mechanisms contribute to pluripo-
Brief. Funct. Genomic. Proteomic. 5(3): 228–243. doi:10.1093/ tency and cell lineage determination of embryonic stem cells.
bfgp/ell020. PMID:16772274. Stem Cells, 25(1): 2–9. doi:10.1634/stemcells.2006-0383.
Ballas, N., Lioy, D.T., Grunseich, C., and Mandel, G. 2009. Non- PMID:17023513.
cell autonomous influence of MeCP2-deficient glia on neuronal Georgel, P.T., Horowitz-Scherer, R.A., Adkins, N., Woodcock,
dendritic morphology. Nat. Neurosci. 12(3): 311–317. doi:10. C.L., Wade, P.A., and Hansen, J.C. 2003. Chromatin compac-
1038/nn.2275. PMID:19234456. tion by human MeCP2. Assembly of novel secondary chromatin
Balmer, D., Arredondo, J., Samaco, R.C., and LaSalle, J.M. 2002. structures in the absence of DNA methylation. J. Biol. Chem.
MECP2 mutations in Rett syndrome adversely affect lympho- 278(34): 32181–32188. doi:10.1074/jbc.M305308200. PMID:
cyte growth, but do not affect imprinted gene expression in 12788925.
blood or brain. Hum. Genet. 110(6): 545–552. doi:10.1007/ Hagberg, B., Aicardi, J., Dias, K., and Ramos, O. 1983. A progres-
s00439-002-0724-4. PMID:12107440. sive syndrome of autism, dementia, ataxia, and loss of purpose-
Benevolenskaya, E.V. 2007. Histone H3K4 demethylases are es- ful hand use in girls: Rett’s syndrome: report of 35 cases. Ann.
sential in development and differentiation. Biochem. Cell Biol. Neurol. 14(4): 471–479. doi:10.1002/ana.410140412. PMID:
85(4): 435–443. doi:10.1139/O07-057. PMID:17713579. 6638958.
Brero, A., Easwaran, H.P., Nowak, D., Grunewald, I., Cremer, T., Harikrishnan, K.N., Chow, M.Z., Baker, E.K., Pal, S., Bassal, S.,
Leonhardt, H., and Cardoso, M.C. 2005. Methyl CpG-binding Brasacchio, D., et al. 2005. Brahma links the SWI/SNF chroma-
proteins induce large-scale chromatin reorganization during tin-remodeling complex with MeCP2-dependent transcriptional
terminal differentiation. J. Cell Biol. 169(5): 733–743. doi:10. silencing. Nat. Genet. 37(3): 254–264. doi:10.1038/ng1516.
1083/jcb.200502062. PMID:15939760. PMID:15696166.
Bruce, K., Myers, F.A., Mantouvalou, E., Lefevre, P., Greaves, I., Heitmann, B., Maurer, T., Weitzel, J.M., Strätling, W.H., Kalbitzer,
Bonifer, C., et al. 2005. The replacement histone H2A.Z in a hy- H.R., and Brunner, E. 2003. Solution structure of the matrix at-
peracetylated form is a feature of active genes in the chicken. tachment region-binding domain of chicken MeCP2. Eur. J. Bio-
Nucleic Acids Res. 33(17): 5633–5639. doi:10.1093/nar/gki874. chem. 270(15): 3263–3270. doi:10.1046/j.1432-1033.2003.
PMID:16204459. 03714.x. PMID:12869202.
Buschdorf, J.P., and Strätling, W.H. 2004. A WW domain binding Hendrich, B., and Bird, A. 1998. Identification and characterization
region in methyl-CpG-binding protein MeCP2: impact on Rett of a family of mammalian methyl-CpG binding proteins. Mol.
syndrome. J. Mol. Med. 82(2): 135–143. doi:10.1007/s00109- Cell. Biol. 18(11): 6538–6547. PMID:9774669.
003-0497-9. PMID:14618241. Henikoff, S., Henikoff, J.G., Sakai, A., Loeb, G.B., and Ahmad, K.
Chahrour, M., Jung, S.Y., Shaw, C., Zhou, X., Wong, S.T., Qin, J., 2009. Genome-wide profiling of salt fractions maps physical
and Zoghbi, H.Y. 2008. MeCP2, a key contributor to neurologi- properties of chromatin. Genome Res. 19(3): 460–469. doi:10.
cal disease, activates and represses transcription. Science, 1101/gr.087619.108. PMID:19088306.
320(5880): 1224–1229. doi:10.1126/science.1153252. PMID: Ho, K.L., McNae, I.W., Schmiedeberg, L., Klose, R.J., Bird, A.P.,
18511691. and Walkinshaw, M.D. 2008. MeCP2 binding to DNA depends
Chen, W.G., Chang, Q., Lin, Y., Meissner, A., West, A.E., Griffith, upon hydration at methyl-CpG. Mol. Cell, 29(4): 525–531.
E.C., et al. 2003. Derepression of BDNF transcription involves doi:10.1016/j.molcel.2007.12.028. PMID:18313390.
calcium-dependent phosphorylation of MeCP2. Science, Hu, K., Nan, X., Bird, A., and Wang, W. 2006. Testing for associa-
302(5646): 885–889. doi:10.1126/science.1086446. PMID: tion between MeCP2 and the brahma-associated SWI/SNF chro-
14593183. matin-remodeling complex. Nat. Genet. 38(9): 962–964, author
Collins, A.L., Levenson, J.M., Vilaythong, A.P., Richman, R., reply 964–967. doi:10.1038/ng0906-962. PMID:16940996.
Armstrong, D.L., Noebels, J.L., et al. 2004. Mild overexpression Isaacs, J.S., Murdock, M., Lane, J., and Percy, A.K. 2003. Eating
of MeCP2 causes a progressive neurological disorder in mice. difficulties in girls with Rett syndrome compared with other de-
Hum. Mol. Genet. 13(21): 2679–2689. doi:10.1093/hmg/ velopmental disabilities. J. Am. Diet. Assoc. 103(2): 224–230.
ddh282. PMID:15351775. doi:10.1053/jada.2003.50026. PMID:12589330.
Crowe, S.L., Movsesyan, V.A., Jorgensen, T.J., and Kondratyev, A. Ishibashi, T., Thambirajah, A.A., and Ausió, J. 2008. MeCP2 pre-
2006. Rapid phosphorylation of histone H2A.X following iono- ferentially binds to methylated linker DNA in the absence of the
tropic glutamate receptor activation. Eur. J. Neurosci. 23(9): terminal tail of histone H3 and independently of histone acetyla-
2351–2361. doi:10.1111/j.1460-9568.2006.04768.x. PMID: tion. FEBS Lett. 582(7): 1157–1162. doi:10.1016/j.febslet.2008.
16706843. 03.005. PMID:18339321.
Delcuve, G.P., Rastegar, M., and Davie, J.R. 2009. Epigenetic con- Jenuwein, T., and Allis, C.D. 2001. Translating the histone code.
trol. J. Cell. Physiol. 219(2): 243–250. doi:10.1002/jcp.21678. Science, 293(5532): 1074–1080. doi:10.1126/science.1063127.
PMID:19127539. PMID:11498575.
Fischle, W. 2008. Talk is cheap–cross-talk in establishment, main- Jiang, Y., Langley, B., Lubin, F.D., Renthal, W., Wood, M.A., Ya-
tenance, and readout of chromatin modifications. Genes Dev. sui, D.H., et al. 2008. Epigenetics in the nervous system. J. Neu-
22(24): 3375–3382. doi:10.1101/gad.1759708. PMID:19141469. rosci. 28(46): 11753–11759. doi:10.1523/JNEUROSCI.3797-08.
Fuks, F., Hurd, P.J., Wolf, D., Nan, X., Bird, A.P., and Kouzarides, 2008. PMID:19005036.
T. 2003. The methyl-CpG-binding protein MeCP2 links DNA Jones, P.L., Veenstra, G.J., Wade, P.A., Vermaak, D., Kass, S.U.,
methylation to histone methylation. J. Biol. Chem. 278(6): Landsberger, N., et al. 1998. Methylated DNA and MeCP2 re-
4035–4040. doi:10.1074/jbc.M210256200. PMID:12427740. cruit histone deacetylase to repress transcription. Nat. Genet.
Galvão, T.C., and Thomas, J.O. 2005. Structure-specific binding of 19(2): 187–191. doi:10.1038/561. PMID:9620779.

Published by NRC Research Press


Thambirajah and Ausió 797

Jugloff, D.G., Jung, B.P., Purushotham, D., Logan, R., and Eu- tion during neuronal cell differentiation. Neurochem. Int. 50(1):
banks, J.H. 2005. Increased dendritic complexity and axonal 264–270. doi:10.1016/j.neuint.2006.08.018. PMID:17052801.
length in cultured mouse cortical neurons overexpressing Mnatzakanian, G.N., Lohi, H., Munteanu, I., Alfred, S.E., Yamada,
methyl-CpG-binding protein MeCP2. Neurobiol. Dis. 19(1-2): T., MacLeod, P.J., et al. 2004. A previously unidentified
18–27. doi:10.1016/j.nbd.2004.11.002. PMID:15837557. MECP2 open reading frame defines a new protein isoform rele-
Kaufmann, W.E., Jarrar, M.H., Wang, J.S., Lee, Y.J., Reddy, S., vant to Rett syndrome. Nat. Genet. 36(4): 339–341. doi:10.1038/
Bibat, G., and Naidu, S. 2005. Histone modifications in Rett ng1327. PMID:15034579.
syndrome lymphocytes: a preliminary evaluation. Brain Dev. Nan, X., Meehan, R.R., and Bird, A. 1993. Dissection of the
27(5): 331–339. doi:10.1016/j.braindev.2004.09.005. PMID: methyl-CpG binding domain from the chromosomal protein
16023547. MeCP2. Nucleic Acids Res. 21(21): 4886–4892. doi:10.1093/
Klose, R.J., Sarraf, S.A., Schmiedeberg, L., McDermott, S.M., nar/21.21.4886. PMID:8177735.
Stancheva, I., and Bird, A.P. 2005. DNA binding selectivity of Nan, X., Campoy, F.J., and Bird, A. 1997. MeCP2 is a transcrip-
MeCP2 due to a requirement for A/T sequences adjacent to tional repressor with abundant binding sites in genomic chroma-
methyl-CpG. Mol. Cell, 19(5): 667–678. doi:10.1016/j.molcel. tin. Cell, 88(4): 471–481. doi:10.1016/S0092-8674(00)81887-5.
2005.07.021. PMID:16137622. PMID:9038338.
Koch, C., and Strätling, W.H. 2004. DNA binding of methyl-CpG- Nan, X., Ng, H.H., Johnson, C.A., Laherty, C.D., Turner, B.M., Ei-
binding protein MeCP2 in human MCF7 cells. Biochemistry, senman, R.N., and Bird, A. 1998. Transcriptional repression by
43(17): 5011–5021. doi:10.1021/bi0359271. PMID:15109260. the methyl-CpG-binding protein MeCP2 involves a histone dea-
Kriaucionis, S., and Bird, A. 2004. The major form of MeCP2 has cetylase complex. Nature, 393(6683): 386–389. doi:10.1038/
a novel N-terminus generated by alternative splicing. Nucleic 30764. PMID:9620804.
Acids Res. 32(5): 1818–1823. doi:10.1093/nar/gkh349. PMID: Nikitina, T., Ghosh, R.P., Horowitz-Scherer, R.A., Hansen, J.C., Gri-
15034150. goryev, S.A., and Woodcock, C.L. 2007. MeCP2-chromatin inter-
Kumar, A., Kamboj, S., Malone, B.M., Kudo, S., Twiss, J.L., actions include the formation of chromatosome-like structures
Czymmek, K.J., et al. 2008. Analysis of protein domains and and are altered in mutations causing Rett syndrome. J. Biol.
Rett syndrome mutations indicate that multiple regions influence Chem. 282(38): 28237–28245. doi:10.1074/jbc.M704304200.
chromatin-binding dynamics of the chromatin-associated protein PMID:17660293.
MECP2 in vivo. J. Cell Sci. 121(Pt 7): 1128–1137. doi:10.1242/ Prakash, Y.S., Thompson, M.A., and Pabelick, C.M. 2009. Brain
jcs.016865. PMID:18334558. derived neurotrophic factor in TNFa modulation of Ca2+ in hu-
Leuba, S.H., Bustamante, C., van Holde, K., and Zlatanova, J. man airway smooth muscle. Am J Respir Cell Mol Biol. doi:10.
1998. Linker histone tails and N-tails of histone H3 are redun- 1074/jbc.M704304200. PMID:19213875.
dant: scanning force microscopy studies of reconstituted fibers. Probst, A.V., Dunleavy, E., and Almouzni, G. 2009. Epigenetic in-
Biophys. J. 74(6): 2830–2839. doi:10.1016/S0006-3495(98) heritance during the cell cycle. Nat. Rev. Mol. Cell Biol. 10(3):
77990-1. PMID:9635737. 192–206. doi:10.1038/nrm2640. PMID:19234478.
Lewis, J.D., Meehan, R.R., Henzel, W.J., Maurer-Fogy, I., Jeppe- Raisner, R.M., Hartley, P.D., Meneghini, M.D., Bao, M.Z., Liu,
sen, P., Klein, F., and Bird, A. 1992. Purification, sequence, C.L., Schreiber, S.L., et al. 2005. Histone variant H2A.Z marks
and cellular localization of a novel chromosomal protein that the 5’ ends of both active and inactive genes in euchromatin.
binds to methylated DNA. Cell, 69(6): 905–914. doi:10.1016/ Cell, 123(2): 233–248. doi:10.1016/j.cell.2005.10.002. PMID:
0092-8674(92)90610-O. PMID:1606614. 16239142.
Lomen-Hoerth, C., and Shooter, E.M. 1995. Widespread neurotro- Rett, A. 1966. [On a unusual brain atrophy syndrome in hyperam-
phin receptor expression in the immune system and other non- monemia in childhood]. Wien. Med. Wochenschr. 116(37): 723–
neuronal rat tissues. J. Neurochem. 64(4): 1780–1789. PMID: 726. PMID:5300597.
7891106. Shahbazian, M.D., Antalffy, B., Armstrong, D.L., and Zoghbi, H.Y.
Marchi, M., Guarda, A., Bergo, A., Landsberger, N., Kilstrup-Niel- 2002. Insight into Rett syndrome: MeCP2 levels display tissue-
sen, C., Ratto, G.M., and Costa, M. 2007. Spatio-temporal dy- and cell-specific differences and correlate with neuronal matura-
namics and localization of MeCP2 and pathological mutants in tion. Hum. Mol. Genet. 11(2): 115–124. doi:10.1093/hmg/11.2.
living cells. Epigenetics, 2(3): 187–197. PMID:17965612. 115. PMID:11809720.
Martinowich, K., Hattori, D., Wu, H., Fouse, S., He, F., Hu, Y., et Shogren-Knaak, M., Ishii, H., Sun, J.M., Pazin, M.J., Davie, J.R.,
al. 2003. DNA methylation-related chromatin remodeling in ac- and Peterson, C.L. 2006. Histone H4-K16 acetylation controls
tivity-dependent BDNF gene regulation. Science, 302(5646): chromatin structure and protein interactions. Science,
890–893. doi:10.1126/science.1090842. PMID:14593184. 311(5762): 844–847. doi:10.1126/science.1124000. PMID:
Meehan, R.R., Lewis, J.D., and Bird, A.P. 1992. Characterization 16469925.
of MeCP2, a vertebrate DNA binding protein with affinity for Soshnikova, N., and Duboule, D. 2008. Epigenetic regulation of
methylated DNA. Nucleic Acids Res. 20(19): 5085–5092. Hox gene activation: the waltz of methyls. Bioessays, 30(3):
doi:10.1093/nar/20.19.5085. PMID:1408825. 199–202. doi:10.1002/bies.20724. PMID:18293357.
Meneghini, M.D., Wu, M., and Madhani, H.D. 2003. Conserved Suzuki, M., Yamada, T., Kihara-Negishi, F., Sakurai, T., and Oi-
histone variant H2A.Z protects euchromatin from the ectopic kawa, T. 2003. Direct association between PU.1 and MeCP2
spread of silent heterochromatin. Cell, 112(5): 725–736. doi:10. that recruits mSin3A-HDAC complex for PU.1-mediated tran-
1016/S0092-8674(03)00123-5. PMID:12628191. scriptional repression. Oncogene, 22(54): 8688–8698. doi:10.
Métivier, R., Gallais, R., Tiffoche, C., Le Péron, C., Jurkowska, 1038/sj.onc.1207182. PMID:14647463.
R.Z., Carmouche, R.P., et al. 2008. Cyclical DNA methylation Tao, J., Hu, K., Chang, Q., Wu, H., Sherman, N.E., Martinowich,
of a transcriptionally active promoter. Nature, 452(7183): 45– K., et al. 2009. Phosphorylation of MeCP2 at Serine 80 regu-
50. doi:10.1038/nature06544. PMID:18322525. lates its chromatin association and neurological function. Proc
Miyake, K., and Nagai, K. 2007. Phosphorylation of methyl-CpG Natl Acad Sci U S A. 106(12): 4882–4887. PMID:19225110.
binding protein 2 (MeCP2) regulates the intracellular localiza- Thambirajah, A.A., Eubanks, J.H., and Ausió, J. 2009 a. MeCP2

Published by NRC Research Press


798 Biochem. Cell Biol. Vol. 87, 2009

post-translational regulation through PEST domains: two novel MeCP2 that binds to methylated DNA. J. Mol. Biol. 291(5):
hypotheses: potential relevance and implications for Rett syn- 1055–1065. doi:10.1006/jmbi.1999.3023. PMID:10518942.
drome. Bioessays, 31(5): 561–569. doi:10.1002/bies.200800220. Wan, M., Zhao, K., Lee, S.S., and Francke, U. 2001. MECP2 trun-
PMID:19319913. cating mutations cause histone H4 hyperacetylation in Rett syn-
Thambirajah, A.A., Li, A., Ishibashi, T., and Ausió, J. 2009b. New drome. Hum. Mol. Genet. 10(10): 1085–1092. doi:10.1093/hmg/
developments in post-translational modifications and functions 10.10.1085. PMID:11331619.
of histone H2A variants. Biochem. Cell Biol. 87(1): 7–17. Wu, J.I., Lessard, J., and Crabtree, G.R. 2009. Understanding the
doi:10.1139/O08-103. PMID:19234519. words of chromatin regulation. Cell, 136(2): 200–206. doi:10.
Tost, J. 2009. DNA methylation: an introduction to the biology and 1016/j.cell.2009.01.009. PMID:19167321.
the disease-associated changes of a promising biomarker. Meth- Yasui, D.H., Peddada, S., Bieda, M.C., Vallero, R.O., Hogart, A.,
ods Mol Biol. 507: 3–20. doi:10.1016/0022-5193(71)90059-2. Nagarajan, R.P., et al. 2007. Integrated epigenomic analyses of
PMID:5555592. neuronal MeCP2 reveal a role for long-range interaction with
Tsanev, R., and Sendov, B. 1971. Possible molecular mechanism active genes. Proc. Natl. Acad. Sci. U.S.A. 104(49): 19416–
for cell differentiation in multicellular organisms. J. Theor. 19421. doi:10.1073/pnas.0707442104. PMID:18042715.
Biol. 30(2): 337–393. doi:10.1016/0022-5193(71)90059-2. Young, J.I., Hong, E.P., Castle, J.C., Crespo-Barreto, J., Bowman,
PMID:5555592. A.B., Rose, M.F., et al. 2005. Regulation of RNA splicing by
Urdinguio, R.G., Pino, I., Ropero, S., Fraga, M.F., and Esteller, M. the methylation-dependent transcriptional repressor methyl-CpG
2007. Histone H3 and H4 modification profiles in a Rett syn- binding protein 2. Proc. Natl. Acad. Sci. U.S.A. 102(49): 17551–
drome mouse model. Epigenetics, 2(1): 11–14. PMID:17965622. 17558. doi:10.1073/pnas.0507856102. PMID:16251272.
Vasko, M.R., Guo, C., and Kelley, M.R. 2005. The multifunctional Zhou, Z., Hong, E.J., Cohen, S., Zhao, W.N., Ho, H.Y., Schmidt,
DNA repair/redox enzyme Ape1/Ref-1 promotes survival of L., et al. 2006. Brain-specific phosphorylation of MeCP2 regu-
neurons after oxidative stress. DNA Repair (Amst.), 4(3): 367– lates activity-dependent Bdnf transcription, dendritic growth,
379. doi:10.1016/j.dnarep.2004.11.006. PMID:15661660. and spine maturation. Neuron, 52(2): 255–269. doi:10.1016/j.
Wakefield, R.I., Smith, B.O., Nan, X., Free, A., Soteriou, A., Uh- neuron.2006.09.037. PMID:17046689.
rin, D., et al. 1999. The solution structure of the domain from

Published by NRC Research Press

View publication stats

You might also like