SlideShare ist ein Scribd-Unternehmen logo
1 von 36
Histone demethylation enzymes and dynamic cell biology
Leanne Stalker and Christopher Wynder2
Introduction
In order to maintain structure and organization within the nucleus of a eukaryotic
cell, the large DNA macromolecule is structured in to chromosomes. To provide
an additional layer of organization, these chromosomes are wrapped around
protein complexes containing proteins known as histones to form the basic unit of
chromatin, the nucleosome (Kornberg, 1974; Kornberg & Lorch, 1999). Each
nucleosome is comprised of an octameric core containing two each of Histone
H2A, H2B, H3 and H4 around which 146bp of DNA is wound. This DNA is then
secured to the core by an additional histone, histone H1(Kornberg & Lorch, 1999;
Kouzarides, 2007; Sims et al, 2003; Volkel & Angrand, 2007). This DNA/protein
complex provides a mechanism by which to conform the large DNA molecule to
the confined space of the nucleus, allows protection from DNA damage during
cell division, and plays a pertinent role in transcriptional regulation(Kooistra &
Helin, 2012; Kouzarides, 2007). Each individual histone protein contains two
highly conserved protein domains including a large globular core and an amino
terminal tail that protrudes from both the histone individually and the nucleosomal
structure as a whole(Luger et al, 1997). From a gene regulation perspective,
these N-terminal tails represent an infinite ability for the nucleosomal structure to
become modified.
Histone tail modifications
Due to their availability outside of the core nucleosome, many amino acid
residues on histone tails are targets of extensive post transcriptional
modifications. These occur on specific amino acid residues and include
acetylations, phosphorylations, SUMOylations, ubiquitinations and methylations.
The result of the addition of these molecular groups is varied and depends highly
on both the specific amino acid modified and the modification itself (Kouzarides,
2007). The addition of these various groups tends to result in one of two
possible consequences. First, it may change the interaction between DNA and
the histone directly leading to an alteration of the chromatin structure as a whole.
This activity is observed mostly when a posttranscriptional modification, such as
an acetylation, alters the charge of an amino acid on the histone tail. Acetylation
of a lysine (K) residue acts to neutralize its basic charge. This loosens the
interaction between the histone and DNA, increasing the accessibility of the DNA
and generally resulting in transcriptional activation(Shogren-Knaak et al, 2006;
Workman & Kingston, 1998). Acetylation is the most extensively studied of the
post-transcriptional modifications and occurs most frequently on residues K9,
K14, K18 and K56 of Histone H3. The enzymes responsible for both the addition
of the acetyl group, Histone Aceytl Transferases (HATs) and the enzymes
responsible for the removal of the acetyl group, Histone Deacetylases (HDACs)
have been increasingly popular targets for drug discovery(Khan & Khan, 2010;
Kuo & Allis, 1998).
The second consequence of histone modification is the alteration of non-
histone protein recruitment to histone tails. For example, histone phosphorylating
enzymes MSK1/2 and RSK2 tend to target serine residues at H3S10.
Phosphorylation of this residue is found to attract the phospho-binding protein
14-3-3, which is thought to activate NFB-regulated genes(Banerjee &
Chakravarti, 2011; Kouzarides, 2007). Greater understanding of the role of
histone phosphorylation is yet to be determined. Ubiquitylation and SUMOylation
differ from the aforementioned mechanisms because they require the addition of
large moieties(Berger, 2007). The function of ubiquitylation remains unclear but
its mechanism of action is believed to either act to recruit supplementary proteins
to histone tails or physically “wedge” chromatin open due to its size. Functional
effects of ubiquitylation appear to vary depending on the residue to which the
moiety is added. For example, ubiquitylation of H2BK123 is associated with the
activation of transcription while ubiquitylation of H2AK119 by NSPc1 has been
found to cooperate with DNA methylation correlate with the transcriptional
silencing of Hox genes.(Wright et al, 2011; Wu et al, 2008). Conversely, the
result of sumoylation is believed to be mainly transcriptionally repressive(Nathan
et al, 2006).
Histone Methylation
Recently, much interest has been placed on the regulation of histone tail
methylation. Unlike the previously mentioned modifications, methylation can
occur on both lysine and arginine (R) residues on amino terminal histone
tails(Shilatifard, 2006; Sims et al, 2003). This modification has also been found to
be processive, suggesting that unlike acetylation, which is either present or
absent, methylation potentially allows for an increased ability to fine tune
regulation. An arginine can become mono or dimethylated, the latter of which can
be either symmetrical or asymmetrical. Whereas a lysine can be modified in a
mono- or di- and tri-methylated form, each of which has been found to have a
differing effect (Cloos et al, 2008; Santos-Rosa et al, 2002). Methylation does
not alter the charge of the histone tail. Therefore, this modification is not thought
to play a direct role in DNA/ histone interactions. Rather, methylation can result in
a modulation of chromatin structure, altering the accessibility to chromatin to
effector proteins, or may act as a recruitment signal for regulatory factors(Cloos
et al, 2008). This results in transcriptional alterations due to changes in the
chromatin landscape as a whole (Bannister et al, 2002; Lachner et al, 2001).
Histone methylations have been found to be associated with both transcriptional
activation and repression with methylation of K4 and K36 of H3 being generally
ascribed to gene activation, whereas association with K9 and K27 of the same
histone are generally thought to be involved with transcriptional
repression(Berger, 2007). The enzymes responsible, known as lysine
methyltransferases (HMTs) are unique in the sense that they are residue
specific. For instance, the Set1/COMPASS or MLL class of histone
methyltransferases are specific for the methylation (mono-, di-, and tri-) of H3K4,
while the Su(var)3-9 family is restricted to methylation of H3K9 (Kouzarides,
2007)
Demethylation of the histone tail
Historically, methylation was considered to be a mark of permanence.
Without the discovery of an enzyme class capable of the removal of methylation,
it was thought that these marks were static. The discovery of the enzyme KDM1a
(also known as LSD1, BHC110) in 2004, changed this notion. KDM1a was found
to have the ability to catalyze the demethylation of histone residues by a flavin
adenine dinucleotide (FAD)-dependent amine oxidase reaction. However, the
enzymology of this demethylase requires a protonated methyl -ammonium in its
substrate. This is absent in the trimethylated version of methylation, resulting in
the conclusion that this enzyme was restricted to mono and dimethylated
modifications(Shi et al, 2004). Since then a more novel, larger protein group
named the Jumonji (JMJC) domain family has been discovered. These enzymes
catalyze the removal of methylation marks utilizing a hydroxylation reaction
through their JMJC domain. This reaction no longer requires a protonated
methyl -ammonium, allowing for the demethylation of all three methyl states. In
several cases, the trimethylated version is actually the preferred
substrate(Christensen et al, 2007; Fodor et al, 2006; Klose et al, 2006; Tsukada
et al, 2006; Whetstine et al, 2006). Historically, F-Box and Leu-rich repeat protein
11 (FBXL11) was the first enzyme discovered in this class; it has demethylase
activity towards both the mono and dimethylated versions of H3K36 (Tsukada et
al, 2006).
To date, JMJC enzymes of this class have been found to be active on
H3K4(Iwase et al, 2007; Klose et al, 2007; Lee et al, 2007; Secombe &
Eisenman, 2007; Seward et al, 2007; Tahiliani et al, 2007; Yamane et al, 2007) ;
H3K9(Yamane et al, 2006), H3K27(Agger et al, 2007; De Santa et al, 2007; Lan
et al, 2007), H3K36(Fodor et al, 2006) and H4K20(Liu et al, 2010). This has led
to the current understanding that methylation represents an extremely flexible
and dynamic modification state resulting in the active modulation of transcription.
Though the JMJC class of demethylases as a whole is an expansive protein
family (the human genome encodes 30 different JMJC containing proteins, 18 of
which have been proven to show demethylase acitivity on both arginine and
lysine residues(Kooistra & Helin, 2012) phylogeny has suggested that within this
family there are several clusters of proteins which appear to group together in
both structure and function. The KDM5 family of demethylases, known to target
all three methylation states of H3K4, represents one such cluster(Cloos et al,
2008)
Specific function of the KDM5 family of HDM enzymes
The KDM5 family of JMJC demethylases includes four known members:
KDM5a, KDM5b, KDM5c and KDM5d (previously known as Jarid1a, Jarid1b,
Jarid1c and Jarid1d respectively). As seen in Figure 1; these demethylases are
highly conserved structurally and are characterized by the presence of five
protein domains:JmjN and JmjC domains required for demethylation activity a
BRIGHT/ARID domain for A/T DNA binding, and both a C5HC2-Zinc finger
domain and several PHD (plant homeobox domains) involved in the enzymes
ability to recognize and bind methylated residues and regulate protein-protein
interactions(Cloos et al, 2008). This review will concentrate on the known roles of
KDM5 proteins in transcriptional regulation, development and disease. For a
recent review encompassing all histone demethylases, please see Kooistra et
al.(Kooistra & Helin, 2012)
H3K4 methylation: a fine balancing act
The KDM5 family of histone demethylases act specifically on H3K4
methylation marks, with a preference for trimethylated H3K4 (H3K4me3). Studies
of H3K4 methylation and its biological roles have been vast and the majority of
studies report the presence of methylated H3K4 as a sign of transcriptional
activation(Barski et al, 2007; Pokholok et al, 2005; Schubeler et al, 2004);.
H3K4me3 localized to gene promoters allows for transcriptional activation by
binding a subunit of TFIID, which then leads to the formation of the initiation
complex(Sims et al, 2003; Vermeulen et al, 2007). Though both mono- and di-
methylated versions of H3K4 span further into the transcribed protein and have
even been found at enhancer elements(Heintzman et al, 2009; Robertson et al,
2008) H3K4me3 remains strongly conserved to the transcriptional start site
(TSS)(Cloos et al, 2008; Kooistra & Helin, 2012; Santos-Rosa et al, 2002). As
expected due to their conserved enzymatic targets, KDM5 demethylases have
been suggested as potent transcriptional repressors through their known ability
to remove this activating mark. Recent genome studies have suggested
however, that the presence of H3K4me3 at the transcriptional start site is not
sufficient to assume active transcription (Guenther et al, 2007). Within
embryonic stem cells (ESC) for example, a very high proportion of transcriptional
start sites possess marks of both transcriptionally active, and transcriptionally
silent chromatin. These sites are said to be bivalent and represent the ability of a
non-committed cell to be poised for commitment and development(Azuara et al,
2006; Bernstein et al, 2006). This phenomenon has also been observed lower
on the evolutionary scale, with C. elegans showing H3K4me3 and H3K27me3
co-occupying promoters early in development (Wang et al, 2011).This suggests
that modification of this methyl mark may represent an ability of the cell to tweak
transcription in one direction or the other, without requiring an absolute condition
of “On” or “Off”. Studies of both KDM5a and KDM5b have suggested that these
demethylases actually co-localize with their substrate, with target genes showing
expression of both the enzyme and H3K4me3(Lopez-Bigas et al, 2008; Schmitz
et al, 2011). Though expression of H3K4me3 was generally found to be lower at
sites of demethylase recruitment, the methylation mark was not completely
absent, suggesting that these enzymes function to maintain low levels of
H3K4me3 but not to abolish the mark completely. This also suggests that
recruitment of additional factors may be required for full demethylase activity of
the enzyme, or that the context of the protein complex in which the KDM5
demethylase is present may alter its enzymology.
Roles for KDM5 outside of the transcriptional start site
Additional groups have suggested a role for KDM5 family members in
intragenic regions of the genome. Liefe et al. suggest that KDM5a plays a role in
Notch-mediated silencing and that demethylation at specific regulator elements
rather than entire promoter TSS regions, is sufficient to result in gene
silencing(Liefke et al, 2010) where Xie et al. have also recently suggested that
KDM5b may play a role in intragenic transcription and elongation of KDM5b
target genes, though these results are currently under debate (Schmitz et al,
2011; Xie et al, 2011). This adds an additional layer of regulation, suggesting
that the accuracy of these enzymes for transcriptional regulation is most likely
extremely pertinent to sensitive biological functions within the cell, with potentially
significant impact on processes including development and differentiation, and
that even the smallest of perturbations could wholly or in part give rise to disease
or transformation.
H3K4me3 and Cellular Identity
Previous studies in D. melanogaster have shown that the KDM5
homologue Little Imaginal Disc (LID) is required for normal development to
proceed through the regulation of homeotic genes (Gildea et al, 2000)
Additionally, the homologue of KDM5 in C.elegans, rbr-2, has been found to be
both an active demethylase and to play a role in the normal development of the
nematode, dependent upon this enzymology (Christensen et al, 2007). Knock
down of rbr-2 was found to result in an increase in H3K4me3 expression and
resulted in a disruption to normal vulval development. Most recently, rbr-2 has
also been implicated in regulation over C.elegans lifespan (Greer et al, 2010).
As both flies and worms only possess one copy of the KDM5 homologue, there is
no chance for functional redundancy. Within higher eukaryotes however, the role
of these proteins in development becomes increasingly complex.
Roles for KDM5 in higher order organisms
Though higher order organisms possess four KDM5 family members, their
roles appear, in many cases, to be functionally distinct. Knock out studies of
KDM5c in a zebrafish model leads to impaired neuronal development. Similar
phenomena are observed in rats where dendritic development becomes
impaired(Iwase et al, 2007). This suggests that any functional redundancy
exhibited by KDM5 family members does not include the role of KDM5c in neural
development. This is of interest considering how similar KDM5c and KDM5d, in
specific, are, and reiterates the importance of target specificity and expression
profile differences between the four family members.
Knock out studies in mice continue to support functionally distinct roles for
these enzymes. Though viable and possessing only mild behavioural
abnormalities, KDM5a -/- mice have been found to have altered transcription of
several cytokine genes known to be KDM5a targets. This has been shown to
lead to aberrant hematology, altered cell cycle and a resistance to apoptosis of
hematopoietic cancers(Wang et al, 2009b). Knockout of KDM5b in mice
however, in contrast to family member KDM5a, has been reported to be
embryonic lethal around E4.5(Catchpole et al, 2011). This suggests that KDM5b
is required in early embryonic development and that this role cannot be taken
over by another KDM5 family member. This early functional importance of
KDM5b is somewhat to be expected due to differences in KDM5 family member
expression profiles. Where KDM5a appears to be widely expressed through all
tissues showing high expression in the haematopoetic system(Christensen et al,
2007; Cloos et al, 2008; Klose et al, 2007; Lopez-Bigas et al, 2008) KDM5c, an X
linked gene which escapes X linked inactivation(Wu et al, 1994a; Wu et al,
1994b) appears to have more limited expression, showing neuronal expression
patterns and playing a role in neuronal development (Iwase et al, 2007). KDM5b
shows a completely different profile, widely expressed in ESCs and
undifferentiated progenitors(Dey et al, 2008), but limited in adult tissues:
restricted to the testis and differentiating mammary gland(Barrett et al, 2002; Lu
et al, 1999). Of interest however, KDM5b is highly expressed in several forms of
cancer(Barrett et al, 2002; Barrett et al, 2007; Madsen et al, 2003; Roesch et al,
2006; Roesch et al, 2010; Xiang et al, 2007). Catchpole et al. additionally report
the creation of a KDM5b mouse strain containing a mutation in which the ARID
domain is removed. This mutation has previously been documented to
completely obliterate the demethylase activity of KDM5b(Tan et al, 2003;
Yamane et al, 2007) though Catchpole et al. suggest that some residual activity
is a possibility (Catchpole et al, 2011). Interestingly, though these mice display
what is referred to as a “mammary phenotype” they are both viable and fertile
suggesting that the role of KDM5b in embryonic development may not hinge
completely on its enzymology (Catchpole et al, 2011). To increase the
complexity of the KDM5b knockout story, Schmitz et al. have recently suggested
that they were successful in creating a KDM5b knock out mouse that is both
viable and fertile and suggest that compensation by other family members may
rescue the knockout phenotype previously described (Schmitz et al, 2011)
KDM5; master regulators of differentiation and development
Though KDM5 family knockout mice may remain viable, distinct and
numerous defects in differentiation and development are frequently noted. This is
suggestive of a protein family involved in the regulation of differentiation control.
In 2005, Benevolenskaya et al. found the first evidence of pRB-KDM5a
complexes in cells and determined that KDM5a was a key regulator of
differentiation control by demonstrating that pRB must displace KDM5a from key
promoters in order to promote differentiation (Benevolenskaya et al, 2005). This
work was completed previous to the knowledge of KDM5a enzymology. Further
study in ESC suggests that during differentation the removal of KDM5a from Hox
genes correlates with increased levels of H3K4me3 (Christensen et al, 2007),
consistent with its role in cellular differentation and development. Previous work
on KDM5b has found that this family member can also repress several target
genes important to differentiation including HOXA5(Yamane et al, 2007), Brain
Factor-1 (BF-1) and Pax9 (Tan et al, 2003).
Recently, work in our laboratory has suggested that KDM5b plays a role in
mouse embryonic stem cells (mESC) to maintain a population of uncommitted
progenitors. Overexpression of KDM5b in mESC was additionally found to impair
specification, and delay or destroy neural differentiation (Dey et al, 2008). More
recent studies have supported this work, suggesting that KDM5b is required for
neural differentiation, most specifically, the generation of neural progenitors
(NPC) from ESC (Schmitz et al, 2011). KDM5b was found to occupy
developmental regulator genes in ESC, and as seen previously (Dey et al, 2008)
plays a pertinent role in gene regulation in this cell type. Their findings however,
suggest that KDM5b is dispensable for the self-renewal capacity of ESC, but
absolutely required for differentiation. Of interest, the modulation of KDM protein
expression in most cell types results in no change in global H3K4me3 levels,
including neural stem cells (NSC) (Schmitz et al, 2011) and MCF7 (Yamane et al,
2007) after the knockdown of KDM5b; and MEFs after the knockdown of KDM5a
(Klose et al, 2007). This is however different in ESC where alteration to KDM5b
levels appears to have a direct effect on global H3K4me3 levels (Dey et al, 2008;
Schmitz et al, 2011). Genome wide chromatin studies have suggested that the
global levels of H3K4me3 decrease from the ESC stage over the course of
differentiation (Ang et al, 2011) with bivalency being removed through
demethylation of H3K4me3 positive promoters (Bernstein et al, 2006).
H3K27me3 expression however, appears to remain present. This suggests that
the presence of H3K4me3 may be required for early development, although its
removal may also represent a required checkpoint for certain stages of
differentiation. This selective removal of H3K4me3 seems to be required for
appropriate cell fate determination to occur.
Studies in C. elegans demonstrate that the appearance of H3K4me3 is both
regulated according to cell lineage and that the deposit of this tri-methylation is
extremely dynamic (Wang et al, 2011) lending credence to the theory that both
the presence and absence of this mark may represent significant methods of
gene regulation during development. Interestingly, recent studies categorizing
the role of H3K4 methylation in fully differentiated cells such as the
cardiomyocyte adds to this work, suggesting that maintenance of H3K4me3 is
required to maintain cellular integrity even in a non dividing, fully committed cell
type (Stein et al, 2011). This also supports an ideal where though the expression
of H3K4me3 may be required to be reduced at certain developmental check-
points, that re-expression of this mark does occur at later stages of development.
All these data together paint a picture where a fine balance between methylation
and demethylation must be maintained in both a lineage and commitment
dependent manner. Slight alterations to the expression level or localization of,
enzymes required to maintain this balance may result in changes in levels of
H3K4me3 in either a global, or gene specific manner which, in turn, could easily
result in disease or abnormal cellular phenotypes.
Demethylation and disease; a fine balance disrupted
Known for their potent roles in development, it is of no surprise that
misregulation of several KDM5 family members has been found to play role in
several developmental diseases. Mostly targeted to the neurological system,
where several KDM5 family members have been studied as developmental
regulators, KDM5 family member involvement in diseases other than cancer has
been a target of recent study.
Past studies of KDM5c have resulted in the striking conclusion that
KDM5c regulation is pertinent to appropriate neural development. Though it is
known as an H3K4me3 demethylase, KDM5c has also been found to recognize
Histone 3 Lysine 9 trimethylation (H3K9me3) (Iwase et al, 2007), and to play a
role in RE1 silencing transcription factor (REST) mediated repression, as it has
been found to co occupy several REST target genes (Ballas & Mandel, 2005).
Loss of KDM5c causes de-repression and increases in H3K43me at key REST
targets leading to an impairment of neuronal gene regulation (Tahiliani et al,
2007). Strikingly, KDM5c has been found to be involved in several diseases of
neurodevelopment including X linked mental retardation/X linked Intellectual
Disability (XLMR/XLID), epilepsy, and autism spectrum disorders (ASD). Many
mutations, currently a total of more than 21, to KDM5c have been found and
continue to be found associated only with cases of XLMR (Abidi et al, 2008;
Jensen et al, 2010; Santos-Reboucas et al, 2011; Tzschach et al, 2006) several
of these mutations resulting in a decrease in the ability KDM5c to recognize
H3K9me3, or to demethylate H3K4me3; suggesting that the enzyomology of
KDM5c may be linked to pathology. One novel mutation was found to alter the
start site of KDM5C, presumably resulting in a complete lack of translation
(Ounap et al, 2012). Additionally, mutations to KDM5c have been connected to
distinct symptomology within XLMR such as memory loss (Simensen et al,
2012). This suggests that specific areas of the brain may be targeted by KDM5c
misregulation. In 2008, KDM5c was connected to another neurocognitive
phenotype when a missense mutation in exon 16 was found connected to ASD.
Though several KDM5c target genes such as BDNF and SCN2A had previously
been known to show altered expression in patients presenting with ASD, KDM5c
itself had never been implicated (Adegbola et al, 2008). KDM5c is not the only
family member with a neurodevelopmental phenotype.
KDM5b, another KDM5 family member which is a known regulator of
neurological development (Schmitz et al, 2011) has also recently been implicated
as a possible player in a congenital variant of Rett Syndrome*, a severe
neurodevelopmental disease. Molecular causes of Rett syndrome include the
persistent expression of early developmental genes (Urdinguio et al, 2008).
Although Rett syndrome is normally classified by a mutation in the X-linked
methyl-CpG-binding protein MeCP2 (Kramer & van Bokhoven, 2009), a
congenital variant showing FOXG1 truncation has recently been discovered
(Ariani et al, 2008; Bahi-Buisson et al, 2010; Mencarelli et al, 2009; Papa et al,
2008) Further analysis of the FOXG1 truncation shows that in both (of the two)
observed truncation events, the domain known as the JBD or the KDM5b binding
domain, is missing, suggesting that the interaction between FOXG1 and KDM5b
is pertinent to the regulation of this disease. A reduction in KDM5b binding would
result in a series of downstream effects, causing a reduction in the ability of
FOXG1 to repress transcription. This transcriptional change would, in turn, result
in a reduction of MeCP2 binding due to a delay in neural differentiation. This
may mimic what occurs when MeCP2 itself is mutated, resulting in a similar
disease phenotype.
Taken together, this data supports the conclusion that alterations to KDM5
proteins result not only in impaired development at the embryonic level, but that
these alterations and mutations may translate into long term disabilities- either
through functional deficits in the demethylase itself, or through downstream
effects on interacting proteins. This also provides additional evidence that each
KDM5 family member plays a unique role in the regional and temporal control of
chromatin structure, and that compensation by additional family members may
not be sufficient to result in phenotypic rescue. (Figure 2)
Though examples of KDM5 demethylases in disease appear limited to
diseases of a neuro-developmental decree, an increased understanding of these
enzymes and how they are regulated will undoubtedly uncover a wide range of
diseases in which they contribute to pathogenesis. Research efforts have, until
recently, concentrated on understanding the roles of these enzymes in various
types of cancer, as detailed below. In many cases KDM5s appear to play a role
in turning on correct genes at an incorrect time. This leads us to question
whether these enzymes may also play a role in degeneration in disorders such
as Alzheimers and Huntington’s disease, by encouraging incorrect signaling,
leading to alterations in neural regulatory networks later in life.
Demethylation and Cancer; a fine balance turned back on incorrectly?
Though they are currently know as transcriptional repressors through
their demethylase activity, several KDM5 family members first garnered the
attention of researchers long before their enzymology was discovered. KDM5a,
for example, was originally identified as an interaction partner for retinoblastoma
protein (pRB). As such, it was originally named Retinoblastoma Binding Protein
2 (RBP2) (Benevolenskaya et al, 2005). Further work on KDM5a showed that it
binds to genes known to be involved in pluripotency and is active in CD34+ and
CD105+ cell populations (known to be markers of HSCs and mesenchymal stem
cells (MSCs) respectively (Wang et al, 2009a). KDM5a target gene activation and
repression may therefore play a key role in the determination of differentiation
profiles in HSCs vs MSCs. Paired with the information gathered from KDM5a
null mice, mentioned earlier, this presents a strong case that KDM5a may play a
key role in the regulation of the haemotopoetic system including the modulation
of haematopoietic cell resistance to apoptosis, a hallmark of several blood
cancers. Multitudinous KDM5a target genes are preferentially expressed in
leukemia and lymphoma and interestingly, KDM5a has recently been found to be
a gene partner involved in Acute Myeloid Leukemia (Wang et al, 2009a). This
suggests that its involvement in cancer may not be limited to retinoblastoma and
that the pRB/KDM5a axis may be a pertinent player in leukemia pathogenesis as
well as a regulator of differentiation and development. Additional studies have
supported the role of KDM5a in a tumour suppressor role, including a recent
study by Liefke et al. Here they suggest that the switch that regulates Notch
target genes includes KDM5a and that through this target specific role, KDM5a
may act as a potent tumour suppressor in Notch mediated carcinogenesis (Liefke
et al, 2010).
KDM5b was additionally recognized prior to its enzymology becoming apparent.
Originally known as Plu-1, this protein was first discovered as a target up-
regulated in response to Her2/c-ErbB2 in breast cancer cell lines and primary
breast cancers (Lu et al, 1999). Of limited expression in most adult tissues,
KDM5b shows consistent up regulation in breast and prostate cancers in both
human and mice, and has been suggested as a possible oncogene in multiple
cancer types. (Barrett et al, 2002; Hayami et al, 2010; Lu et al, 1999; Roesch et
al, 2010; Xiang et al, 2007; Yamane et al, 2007) Hayami et al. draw on previous
work completed in breast (Yamane) and Prostate (Xiang) cancers and
demonstrate that KDM5b is directly involved in the proliferative rate and ability of
both lung and bladder cancer cells to escape apoptosis. In concordance with
other groups, they demonstrate that reduction of KDM5b level results in
alterations to the cell cycle of tumour cells, and a reduction in oncogenic potential
(Hayami et al, 2010). Delineating the exact role that KDM5b exerts in cancer has
become complex and more and more evidence points towards the theory that
cancer should be categorized as a group of diseases, rather than a single
dysfunction. KDM5b is known to be a regulator of both oncogenes and tumour
suppressors through direct interaction with their promoters, such as BRCA1 in
breast cancer (Yamane et al, 2007). It has also been associated with cell cycle
control in both an accelerating (breast cancer)(Yamane et al, 2007) and
decelerating (Melanoma) (Roesch et al, 2010) fashion and has been found to
increase the invasive potential of non invasive cell types through repression of
the tumour suppressor KAT5 (Yoshida et al, 2011). Recently, KDM5b has also
been demonstrated to promote cell cycle progression in breast cancer cells by
the epigenetic modulation of the expression of micro RNA let7e suggesting an
additional, indirect method to regulate of gene expression (Mitra et al, 2011).
In the recent years, another role of KDM5b in tumor survival has
surfaced, suggesting that KDM5b may be required for the adaptation of cells to
hypoxia. Solid tumors are considered to be highly hypoxic compared to
surrounding tissue, and adaptation to this state is pertinent for tumor survival
(Semenza, 2003). Adaptation to hypoxia is driven through Hypoxia inducible
factor-1 (HIF-1) and is largely mediated through transcriptional repression.
Recent screens for proteins that facilitate this adaptation noted several Jumonji
family demethylases, including KDM5b. KDM5b was found to be a direct HIF
target and shows increased expression under hypoxic conditions (Xia et al,
2009). Previous work has shown that reduced H3K4 methylation is linked to poor
prognosis in cancer patients (Seligson et al, 2005), suggesting that an ability to
demethylate H3K4 is important for tumor survival. Due to the requirement of
dioxygenases such as KDM5b, for molecular oxygen, Xia et al. propose that the
increased expression level of these enzymes may represent a compensatory
mechanism in response to decreasing oxygen availability (Xia et al, 2009).
Without this compensatory mechanism, H3K4me3 levels would be expected to
increase as tumors increase in size and oxygen levels decrease, leading to the
death of the hypoxic tumor cells. An increase in the expression level of
demethylases such as KDM5b may provide a mechanism for the tumor to
maintain low H3K4me3 levels even in situations where decreased oxygen levels
are present. This novel mechanism may allow tumors to literally skirt death and
continue to proliferate.
KDM5c, a demethylase more commonly thought to exert control
over neuronal identity, is over expressed in both prostate tumors and
seminomas, and has been shown to act as a co-repressor to Smad3. Binding of
KDM5c to Smad3 blocks its transactivation ability, thus reducing its ability to act
as an effector of the TGF-B pathway. Blockage of this pathway is apparent in
several cancer types suggesting that KDM5c may possess oncogenic potential
through its ability to block Smad3. Most interestingly, this appears to be
independent of its demethylase activity (Kim et al, 2008). Recently, Niu et al.
explored the role of KDM5c in clear cell renal cell carcinoma (ccRCC). A high
proportion of ccRCCs show inactivation of the tumour suppressor von Hippel-
Lindau (VHL). Additionally, VHL-/- tumours show decreased levels of H3K4me3
compared to their VHL +/+ counterparts. Interestingly, this was also shown to be
Hypoxia inducible factor- (HIF1-) dependent. Previous work, demonstrating gene
alterations in patient samples of ccRCC, had provided evidence that mutations to
KDM5c were higher than would be expected by chance in ccRCC patients
(Dalgliesh et al, 2010), suggesting a connection between KDM5c alterations and
aberrant levels of H3K4me3. Niu et al. have shown that KDM5c is responsible for
suppressing HIF response genes by removal of H3K4me3, and that mutations to
KDM5c are promote tumour growth. This tumour suppressor role of KDM5c is
specific to this family member as loss of KDM5c (but not KDM5a or KDM5b)
abolished the difference between VHL-/- and +/+ tumors (Niu et al, 2012).
Given their role in stem cell biology and development, we are left
to question whether KDM5s simply do the “right” job at the “wrong” time in
cancers; exerting control similar to non-pathogenic contexts during differentiation
and development, but with aberrant results within a fully developed tissue. The
roles of KDM5s during carcinogenesis appear to focus on helping tumour cells to
survive in contexts when appropriate cellular signaling would lead to cell death;
survival of hypoxia, escaping apoptosis, increasing potential for invasion, and
alterations to cell cycle leading to over proliferation and the development of
inappropriate cell types. However, information on the roles of these proteins are
often contradictory, with several being classified as proteins with both oncogenic
and tumour suppressor abilities depending on cellular context. Though, as
previously mentioned, reduced H3K4 methylation levels appear to be linked to
poor prognosis in cancer patients (Seligson et al, 2005), in the case presented
above, increased H3K4 in the context of HIF response genes in ccRCC appears
to be tumour-promoting. This again draws attention to the fine balance of
H3K4me3 expression and the regulation of the enzymes that control this
methylation, both are highly dependent upon cellular context.
KDM5s in tumour sub populations
Several groups have now suggested that KDM5 family members
exert control in specific subsets of a tumour population to maintain or promote
growth. Sharma et al. noted a population of “reversibly drug tolerant” cells within
several human cancers which maintain viability through an altered chromatin
state requiring KDM5a. These cells appear absolutely required to protect tumors
from eradication (Sharma et al, 2010). Roesch et al. show another angle of the
KDM5 cancer story, using the expression of KDM5b as a biomarker to flag a
small population of slow cycling cells within the heterogeneous population of a
melanoma (Roesch et al, 2010). These “slow” cells appear to be required for
tumour maintenance, giving rise to progeny which express low levels of KDM5b,
and knock down of KDM5b results in an exhaustion of tumour growth.
Interestingly the same group has also proposed that KDM5b has a tumour
suppressor role (Roesch et al, 2006; Roesch et al, 2008). It has been
suggested that the acceleration of cell cycle in these melanocytes after KDM5b
expression decrease may be due to a derepression of E2F-target genes, thus
accelerating cell cycle. Both KDM5b and KDM5a have been shown to be
members of the Rb repression complex, required for the repression of E2F target
genes during senescence (Chicas et al, 2012; Nijwening et al, 2011). Though
repression of E2F targets would generally be considered a tumour suppressive
function, mutations to Rb are common in cancer progression, allowing pro-
proliferative effects to override normal suppression and could lead to increased
oncogenic potential. Following in this theory, loss of KDM5a in a pRb defective
tumour context promotes senescence and differentiation, suggestive of an
oncogenic role in the absence of Rb (Lin et al, 2011). As noted by Chicas et al.,
this highlights the context- dependent role of these demethylases (Chicas et al,
2012). These results together suggest that though they are involved in
oncogenesis, KDM5s appear to exert their “tumourogenic potential” in different
ways, depending on cellular context and may respond differently depending on
which upstream cellular cues become activated (Figure 3).
These aspects of KDM5 demethylases, though complex, make
them potentially lucrative targets for pharmaceutical intervention. Enzymes are
known to provide excellent drug targets and KDM5b in particular, due to its low
expression level in most adult human tissues, may provide a potentially safe
target for pharmaceuticals. Immunotherapy approaches against KDM5b have
been investigated recently with results suggesting that KDM5b may represent a
tumour associated antigen (TAA) for breast cancer (Coleman et al, 2010).
The major question that remains for future clinical use of KDM5
targeting therapeutics is: How can we utilize this knowledge of KDM5 biology to
combat cancer and disease? Histone deacetylase inhibitors have long been the
“king” of the epigenetic pharmaceutical industry, with drugs such as Valproic
acid, Entinostat and Romadepsin showing large potential in the clinic and earning
FDA approval (Song et al, 2011). However, little has been done targeting
demethylase enzymes as possible treatment options. Recent studies have
demonstrated the release of therapeutic agents against KDM1 and studies of
agents against JMJD2 demethylases (Hamada et al, 2010), and novel assays
are being developed to screen and identify novel candidates against these
targets (Yu et al, 2012). The KDM5 family is not special in this contextual activity.
The importance of context and the flexibility that KDMs in general bring to
transcriptional control is the key to a variety of processes. Understanding how
and when the KDMs interact with both each other and the basal transcriptional
machinery will likely provide clues into a myriad of diseases.
1. Abidi FE, Holloway L, Moore CA, Weaver DD, Simensen RJ, Stevenson RE,
Rogers RC, Schwartz CE (2008) Mutations in JARID1C are associated with X-
linked mental retardation, short stature and hyperreflexia. J Med Genet 45:
787-793
2. Adegbola A, Gao H, Sommer S, Browning M (2008) A novel mutation in
JARID1C/SMCX in a patient with autism spectrum disorder (ASD). Am J Med
Genet A 146A: 505-511
3. Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rappsilber J, Issaeva I,
Canaani E, Salcini AE, Helin K (2007) UTX and JMJD3 are histone H3K27
demethylases involved in HOX gene regulation and development. Nature
449: 731-734
4. Ang YS, Tsai SY, Lee DF, Monk J, Su J, Ratnakumar K, Ding J, Ge Y, Darr H,
Chang B, Wang J, Rendl M, Bernstein E, Schaniel C, Lemischka IR (2011)
Wdr5 mediates self-renewal and reprogramming via the embryonic stem cell
core transcriptional network. Cell 145: 183-197
5. Ariani F, Hayek G, Rondinella D, Artuso R, Mencarelli MA, Spanhol-Rosseto A,
Pollazzon M, Buoni S, Spiga O, Ricciardi S, Meloni I, Longo I, Mari F, Broccoli
V, Zappella M, Renieri A (2008) FOXG1 is responsible for the congenital
variant of Rett syndrome. Am J Hum Genet 83: 89-93
6. Azuara V, Perry P, Sauer S, Spivakov M, Jorgensen HF, John RM, Gouti M,
Casanova M, Warnes G, Merkenschlager M, Fisher AG (2006) Chromatin
signatures of pluripotent cell lines. Nat Cell Biol 8: 532-538
7. Bahi-Buisson N, Nectoux J, Girard B, Van Esch H, De Ravel T, Boddaert N,
Plouin P, Rio M, Fichou Y, Chelly J, Bienvenu T (2010) Revisiting the
phenotype associated with FOXG1 mutations: two novel cases of congenital
Rett variant. Neurogenetics 11: 241-249
8. Ballas N, Mandel G (2005) The many faces of REST oversee epigenetic
programming of neuronal genes. Curr Opin Neurobiol 15: 500-506
9. Banerjee T, Chakravarti D (2011) A peek into the complex realm of histone
phosphorylation. Mol Cell Biol 31: 4858-4873
10. Bannister AJ, Schneider R, Kouzarides T (2002) Histone methylation:
dynamic or static? Cell 109: 801-806
11. Barrett A, Madsen B, Copier J, Lu PJ, Cooper L, Scibetta AG, Burchell J, Taylor-
Papadimitriou J (2002) PLU-1 nuclear protein, which is upregulated in breast
cancer, shows restricted expression in normal human adult tissues: a new
cancer/testis antigen? Int J Cancer 101: 581-588
12. Barrett A, Santangelo S, Tan K, Catchpole S, Roberts K, Spencer-Dene B, Hall
D, Scibetta A, Burchell J, Verdin E, Freemont P, Taylor-Papadimitriou J (2007)
Breast cancer associated transcriptional repressor PLU-1/JARID1B interacts
directly with histone deacetylases. Int J Cancer 121: 265-275
13. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I,
Zhao K (2007) High-resolution profiling of histone methylations in the
human genome. Cell 129: 823-837
14. Benevolenskaya EV, Murray HL, Branton P, Young RA, Kaelin WG, Jr. (2005)
Binding of pRB to the PHD protein RBP2 promotes cellular differentiation.
Mol Cell 18: 623-635
15. Berger SL (2007) The complex language of chromatin regulation during
transcription. Nature 447: 407-412
16. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B,
Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL,
Lander ES (2006) A bivalent chromatin structure marks key developmental
genes in embryonic stem cells. Cell 125: 315-326
17. Catchpole S, Spencer-Dene B, Hall D, Santangelo S, Rosewell I, Guenatri M,
Beatson R, Scibetta AG, Burchell JM, Taylor-Papadimitriou J (2011) PLU-
1/JARID1B/KDM5B is required for embryonic survival and contributes to
cell proliferation in the mammary gland and in ER+ breast cancer cells. Int J
Oncol 38: 1267-1277
18. Chicas A, Kapoor A, Wang X, Aksoy O, Evertts AG, Zhang MQ, Garcia BA,
Bernstein E, Lowe SW (2012) H3K4 demethylation by Jarid1a and Jarid1b
contributes to retinoblastoma-mediated gene silencing during cellular
senescence. Proc Natl Acad Sci U S A 109: 8971-8976
19. Christensen J, Agger K, Cloos PA, Pasini D, Rose S, Sennels L, Rappsilber J,
Hansen KH, Salcini AE, Helin K (2007) RBP2 belongs to a family of
demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell
128: 1063-1076
20. Cloos PA, Christensen J, Agger K, Helin K (2008) Erasing the methyl mark:
histone demethylases at the center of cellular differentiation and disease.
Genes Dev 22: 1115-1140
21. Coleman JA, Correa I, Cooper L, Bohnenkamp HR, Poulsom R, Burchell JM,
Taylor-Papadimitriou J (2010) T cells reactive with HLA-A*0201 *peptides
from the histone demethylase JARID1B are found in the circulation of breast
cancer patients. Int J Cancer
22. Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, Davies H,
Edkins S, Hardy C, Latimer C, Teague J, Andrews J, Barthorpe S, Beare D, Buck
G, Campbell PJ, Forbes S, Jia M, Jones D, Knott H, Kok CY, Lau KW, Leroy C, Lin
ML, McBride DJ, Maddison M, Maguire S, McLay K, Menzies A, Mironenko T,
Mulderrig L, Mudie L, O'Meara S, Pleasance E, Rajasingham A, Shepherd R,
Smith R, Stebbings L, Stephens P, Tang G, Tarpey PS, Turrell K, Dykema KJ,
Khoo SK, Petillo D, Wondergem B, Anema J, Kahnoski RJ, Teh BT, Stratton MR,
Futreal PA (2010) Systematic sequencing of renal carcinoma reveals
inactivation of histone modifying genes. Nature 463: 360-363
23. De Santa F, Totaro MG, Prosperini E, Notarbartolo S, Testa G, Natoli G (2007)
The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition
of polycomb-mediated gene silencing. Cell 130: 1083-1094
24. Dey BK, Stalker L, Schnerch A, Bhatia M, Taylor-Papidimitriou J, Wynder C
(2008) The histone demethylase KDM5b/JARID1b plays a role in cell fate
decisions by blocking terminal differentiation. Mol Cell Biol 28: 5312-5327
25. Fodor BD, Kubicek S, Yonezawa M, O'Sullivan RJ, Sengupta R, Perez-Burgos L,
Opravil S, Mechtler K, Schotta G, Jenuwein T (2006) Jmjd2b antagonizes
H3K9 trimethylation at pericentric heterochromatin in mammalian cells.
Genes Dev 20: 1557-1562
26. Gildea JJ, Lopez R, Shearn A (2000) A screen for new trithorax group genes
identified little imaginal discs, the Drosophila melanogaster homologue of
human retinoblastoma binding protein 2. Genetics 156: 645-663
27. Greer EL, Maures TJ, Hauswirth AG, Green EM, Leeman DS, Maro GS, Han S,
Banko MR, Gozani O, Brunet A (2010) Members of the H3K4 trimethylation
complex regulate lifespan in a germline-dependent manner in C. elegans.
Nature 466: 383-387
28. Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007) A chromatin
landmark and transcription initiation at most promoters in human cells. Cell
130: 77-88
29. Hamada S, Suzuki T, Mino K, Koseki K, Oehme F, Flamme I, Ozasa H, Itoh Y,
Ogasawara D, Komaarashi H, Kato A, Tsumoto H, Nakagawa H, Hasegawa M,
Sasaki R, Mizukami T, Miyata N (2010) Design, synthesis, enzyme-inhibitory
activity, and effect on human cancer cells of a novel series of jumonji domain-
containing protein 2 histone demethylase inhibitors. J Med Chem 53: 5629-
5638
30. Hayami S, Yoshimatsu M, Veerakumarasivam A, Unoki M, Iwai Y, Tsunoda T,
Field HI, Kelly JD, Neal DE, Yamaue H, Ponder BA, Nakamura Y, Hamamoto R
(2010) Overexpression of the JmjC histone demethylase KDM5B in human
carcinogenesis: involvement in the proliferation of cancer cells through the
E2F/RB pathway. Mol Cancer 9: 59
31. Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, Ye Z,
Lee LK, Stuart RK, Ching CW, Ching KA, Antosiewicz-Bourget JE, Liu H, Zhang
X, Green RD, Lobanenkov VV, Stewart R, Thomson JA, Crawford GE, Kellis M,
Ren B (2009) Histone modifications at human enhancers reflect global cell-
type-specific gene expression. Nature 459: 108-112
32. Iwase S, Lan F, Bayliss P, de la Torre-Ubieta L, Huarte M, Qi HH, Whetstine JR,
Bonni A, Roberts TM, Shi Y (2007) The X-linked mental retardation gene
SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell
128: 1077-1088
33. Jensen LR, Bartenschlager H, Rujirabanjerd S, Tzschach A, Numann A, Janecke
AR, Sporle R, Stricker S, Raynaud M, Nelson J, Hackett A, Fryns JP, Chelly J, de
Brouwer AP, Hamel B, Gecz J, Ropers HH, Kuss AW (2010) A distinctive gene
expression fingerprint in mentally retarded male patients reflects disease-
causing defects in the histone demethylase KDM5C. Pathogenetics 3: 2
34. Khan SN, Khan AU (2010) Role of histone acetylation in cell physiology and
diseases: An update. Clin Chim Acta 411: 1401-1411
35. Kim TD, Shin S, Janknecht R (2008) Repression of Smad3 activity by histone
demethylase SMCX/JARID1C. Biochem Biophys Res Commun 366: 563-567
36. Klose RJ, Kallin EM, Zhang Y (2006) JmjC-domain-containing proteins and
histone demethylation. Nat Rev Genet 7: 715-727
37. Klose RJ, Yan Q, Tothova Z, Yamane K, Erdjument-Bromage H, Tempst P,
Gilliland DG, Zhang Y, Kaelin WG, Jr. (2007) The retinoblastoma binding
protein RBP2 is an H3K4 demethylase. Cell 128: 889-900
38. Kooistra SM, Helin K (2012) Molecular mechanisms and potential functions
of histone demethylases. Nat Rev Mol Cell Biol 13: 297-311
39. Kornberg RD (1974) Chromatin structure: a repeating unit of histones and
DNA. Science 184: 868-871
40. Kornberg RD, Lorch Y (1999) Twenty-five years of the nucleosome,
fundamental particle of the eukaryote chromosome. Cell 98: 285-294
41. Kouzarides T (2007) Chromatin modifications and their function. Cell 128:
693-705
42. Kramer JM, van Bokhoven H (2009) Genetic and epigenetic defects in mental
retardation. Int J Biochem Cell Biol 41: 96-107
43. Kuo MH, Allis CD (1998) Roles of histone acetyltransferases and deacetylases
in gene regulation. Bioessays 20: 615-626
44. Lachner M, O'Carroll D, Rea S, Mechtler K, Jenuwein T (2001) Methylation of
histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410: 116-
120
45. Lan F, Bayliss PE, Rinn JL, Whetstine JR, Wang JK, Chen S, Iwase S, Alpatov R,
Issaeva I, Canaani E, Roberts TM, Chang HY, Shi Y (2007) A histone H3 lysine
27 demethylase regulates animal posterior development. Nature 449: 689-
694
46. Lee MG, Norman J, Shilatifard A, Shiekhattar R (2007) Physical and functional
association of a trimethyl H3K4 demethylase and Ring6a/MBLR, a polycomb-
like protein. Cell 128: 877-887
47. Liefke R, Oswald F, Alvarado C, Ferres-Marco D, Mittler G, Rodriguez P,
Dominguez M, Borggrefe T (2010) Histone demethylase KDM5A is an integral
part of the core Notch-RBP-J repressor complex. Genes Dev 24: 590-601
48. Lin W, Cao J, Liu J, Beshiri ML, Fujiwara Y, Francis J, Cherniack AD, Geisen C,
Blair LP, Zou MR, Shen X, Kawamori D, Liu Z, Grisanzio C, Watanabe H,
Minamishima YA, Zhang Q, Kulkarni RN, Signoretti S, Rodig SJ, Bronson RT,
Orkin SH, Tuck DP, Benevolenskaya EV, Meyerson M, Kaelin WG, Jr., Yan Q
(2011) Loss of the retinoblastoma binding protein 2 (RBP2) histone
demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1. Proc
Natl Acad Sci U S A 108: 13379-13386
49. Liu W, Tanasa B, Tyurina OV, Zhou TY, Gassmann R, Liu WT, Ohgi KA, Benner
C, Garcia-Bassets I, Aggarwal AK, Desai A, Dorrestein PC, Glass CK, Rosenfeld
MG (2010) PHF8 mediates histone H4 lysine 20 demethylation events
involved in cell cycle progression. Nature 466: 508-512
50. Lopez-Bigas N, Kisiel TA, Dewaal DC, Holmes KB, Volkert TL, Gupta S, Love J,
Murray HL, Young RA, Benevolenskaya EV (2008) Genome-wide analysis of
the H3K4 histone demethylase RBP2 reveals a transcriptional program
controlling differentiation. Mol Cell 31: 520-530
51. Lu PJ, Sundquist K, Baeckstrom D, Poulsom R, Hanby A, Meier-Ewert S, Jones
T, Mitchell M, Pitha-Rowe P, Freemont P, Taylor-Papadimitriou J (1999) A
novel gene (PLU-1) containing highly conserved putative DNA/chromatin
binding motifs is specifically up-regulated in breast cancer. J Biol Chem 274:
15633-15645
52. Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ (1997) Crystal
structure of the nucleosome core particle at 2.8 A resolution. Nature 389:
251-260
53. Madsen B, Tarsounas M, Burchell JM, Hall D, Poulsom R, Taylor-
Papadimitriou J (2003) PLU-1, a transcriptional repressor and putative
testis-cancer antigen, has a specific expression and localisation pattern
during meiosis. Chromosoma 112: 124-132
54. Mencarelli MA, Kleefstra T, Katzaki E, Papa FT, Cohen M, Pfundt R, Ariani F,
Meloni I, Mari F, Renieri A (2009) 14q12 Microdeletion syndrome and
congenital variant of Rett syndrome. Eur J Med Genet 52: 148-152
55. Mitra D, Das PM, Huynh FC, Jones FE (2011) Jumonji/ARID1 B (JARID1B)
protein promotes breast tumor cell cycle progression through epigenetic
repression of microRNA let-7e. J Biol Chem 286: 40531-40535
56. Nathan D, Ingvarsdottir K, Sterner DE, Bylebyl GR, Dokmanovic M, Dorsey JA,
Whelan KA, Krsmanovic M, Lane WS, Meluh PB, Johnson ES, Berger SL (2006)
Histone sumoylation is a negative regulator in Saccharomyces cerevisiae and
shows dynamic interplay with positive-acting histone modifications. Genes
Dev 20: 966-976
57. Nijwening JH, Geutjes EJ, Bernards R, Beijersbergen RL (2011) The histone
demethylase Jarid1b (Kdm5b) is a novel component of the Rb pathway and
associates with E2f-target genes in MEFs during senescence. PLoS One 6:
e25235
58. Niu X, Zhang T, Liao L, Zhou L, Lindner DJ, Zhou M, Rini B, Yan Q, Yang H
(2012) The von Hippel-Lindau tumor suppressor protein regulates gene
expression and tumor growth through histone demethylase JARID1C.
Oncogene 31: 776-786
59. Ounap K, Puusepp-Benazzouz H, Peters M, Vaher U, Rein R, Proos A, Field M,
Reimand T (2012) A novel c.2T > C mutation of the KDM5C/JARID1C gene in
one large family with X-linked intellectual disability. Eur J Med Genet 55: 178-
184
60. Papa FT, Mencarelli MA, Caselli R, Katzaki E, Sampieri K, Meloni I, Ariani F,
Longo I, Maggio A, Balestri P, Grosso S, Farnetani MA, Berardi R, Mari F,
Renieri A (2008) A 3 Mb deletion in 14q12 causes severe mental retardation,
mild facial dysmorphisms and Rett-like features. Am J Med Genet A 146A:
1994-1998
61. Pokholok DK, Harbison CT, Levine S, Cole M, Hannett NM, Lee TI, Bell GW,
Walker K, Rolfe PA, Herbolsheimer E, Zeitlinger J, Lewitter F, Gifford DK,
Young RA (2005) Genome-wide map of nucleosome acetylation and
methylation in yeast. Cell 122: 517-527
62. Robertson AG, Bilenky M, Tam A, Zhao Y, Zeng T, Thiessen N, Cezard T, Fejes
AP, Wederell ED, Cullum R, Euskirchen G, Krzywinski M, Birol I, Snyder M,
Hoodless PA, Hirst M, Marra MA, Jones SJ (2008) Genome-wide relationship
between histone H3 lysine 4 mono- and tri-methylation and transcription
factor binding. Genome Res 18: 1906-1917
63. Roesch A, Becker B, Schneider-Brachert W, Hagen I, Landthaler M, Vogt T
(2006) Re-expression of the retinoblastoma-binding protein 2-homolog 1
reveals tumor-suppressive functions in highly metastatic melanoma cells. J
Invest Dermatol 126: 1850-1859
64. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA,
Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M (2010) A temporarily distinct
subpopulation of slow-cycling melanoma cells is required for continuous
tumor growth. Cell 141: 583-594
65. Roesch A, Mueller AM, Stempfl T, Moehle C, Landthaler M, Vogt T (2008)
RBP2-H1/JARID1B is a transcriptional regulator with a tumor suppressive
potential in melanoma cells. Int J Cancer 122: 1047-1057
66. Santos-Reboucas CB, Fintelman-Rodrigues N, Jensen LR, Kuss AW, Ribeiro
MG, Campos M, Jr., Santos JM, Pimentel MM (2011) A novel nonsense
mutation in KDM5C/JARID1C gene causing intellectual disability, short
stature and speech delay. Neurosci Lett 498: 67-71
67. Santos-Rosa H, Schneider R, Bannister AJ, Sherriff J, Bernstein BE, Emre NC,
Schreiber SL, Mellor J, Kouzarides T (2002) Active genes are tri-methylated
at K4 of histone H3. Nature 419: 407-411
68. Schmitz SU, Albert M, Malatesta M, Morey L, Johansen JV, Bak M, Tommerup
N, Abarrategui I, Helin K (2011) Jarid1b targets genes regulating
development and is involved in neural differentiation. EMBO J 30: 4586-4600
69. Schubeler D, MacAlpine DM, Scalzo D, Wirbelauer C, Kooperberg C, van
Leeuwen F, Gottschling DE, O'Neill LP, Turner BM, Delrow J, Bell SP, Groudine
M (2004) The histone modification pattern of active genes revealed through
genome-wide chromatin analysis of a higher eukaryote. Genes Dev 18: 1263-
1271
70. Secombe J, Eisenman RN (2007) The function and regulation of the JARID1
family of histone H3 lysine 4 demethylases: the Myc connection. Cell Cycle 6:
1324-1328
71. Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunstein M, Kurdistani SK (2005)
Global histone modification patterns predict risk of prostate cancer
recurrence. Nature 435: 1262-1266
72. Semenza GL (2003) Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3:
721-732
73. Seward DJ, Cubberley G, Kim S, Schonewald M, Zhang L, Tripet B, Bentley DL
(2007) Demethylation of trimethylated histone H3 Lys4 in vivo by JARID1
JmjC proteins. Nat Struct Mol Biol 14: 240-242
74. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, McDermott
U, Azizian N, Zou L, Fischbach MA, Wong KK, Brandstetter K, Wittner B,
Ramaswamy S, Classon M, Settleman J (2010) A chromatin-mediated
reversible drug-tolerant state in cancer cell subpopulations. Cell 141: 69-80
75. Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA (2004)
Histone demethylation mediated by the nuclear amine oxidase homolog
LSD1. Cell 119: 941-953
76. Shilatifard A (2006) Chromatin modifications by methylation and
ubiquitination: implications in the regulation of gene expression. Annu Rev
Biochem 75: 243-269
77. Shogren-Knaak M, Ishii H, Sun JM, Pazin MJ, Davie JR, Peterson CL (2006)
Histone H4-K16 acetylation controls chromatin structure and protein
interactions. Science 311: 844-847
78. Simensen RJ, Rogers RC, Collins JS, Abidi F, Schwartz CE, Stevenson RE
(2012) Short-term memory deficits in carrier females with KDM5C
mutations. Genet Couns 23: 31-40
79. Sims RJ, 3rd, Nishioka K, Reinberg D (2003) Histone lysine methylation: a
signature for chromatin function. Trends Genet 19: 629-639
80. Song SH, Han SW, Bang YJ (2011) Epigenetic-based therapies in cancer:
progress to date. Drugs 71: 2391-2403
81. Stein AB, Jones TA, Herron TJ, Patel SR, Day SM, Noujaim SF, Milstein ML, Klos
M, Furspan PB, Jalife J, Dressler GR (2011) Loss of H3K4 methylation
destabilizes gene expression patterns and physiological functions in adult
murine cardiomyocytes. J Clin Invest 121: 2641-2650
82. Tahiliani M, Mei P, Fang R, Leonor T, Rutenberg M, Shimizu F, Li J, Rao A, Shi
Y (2007) The histone H3K4 demethylase SMCX links REST target genes to X-
linked mental retardation. Nature 447: 601-605
83. Tan K, Shaw AL, Madsen B, Jensen K, Taylor-Papadimitriou J, Freemont PS
(2003) Human PLU-1 Has transcriptional repression properties and interacts
with the developmental transcription factors BF-1 and PAX9. J Biol Chem
278: 20507-20513
84. Tsukada Y, Fang J, Erdjument-Bromage H, Warren ME, Borchers CH, Tempst
P, Zhang Y (2006) Histone demethylation by a family of JmjC domain-
containing proteins. Nature 439: 811-816
85. Tzschach A, Lenzner S, Moser B, Reinhardt R, Chelly J, Fryns JP, Kleefstra T,
Raynaud M, Turner G, Ropers HH, Kuss A, Jensen LR (2006) Novel
JARID1C/SMCX mutations in patients with X-linked mental retardation. Hum
Mutat 27: 389
86. Urdinguio RG, Lopez-Serra L, Lopez-Nieva P, Alaminos M, Diaz-Uriarte R,
Fernandez AF, Esteller M (2008) Mecp2-null mice provide new neuronal
targets for Rett syndrome. PLoS One 3: e3669
87. Vermeulen M, Mulder KW, Denissov S, Pijnappel WW, van Schaik FM, Varier
RA, Baltissen MP, Stunnenberg HG, Mann M, Timmers HT (2007) Selective
anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4.
Cell 131: 58-69
88. Volkel P, Angrand PO (2007) The control of histone lysine methylation in
epigenetic regulation. Biochimie 89: 1-20
89. Wang P, Lin C, Smith ER, Guo H, Sanderson BW, Wu M, Gogol M, Alexander T,
Seidel C, Wiedemann LM, Ge K, Krumlauf R, Shilatifard A (2009a) Global
analysis of H3K4 methylation defines MLL family member targets and points
to a role for MLL1-mediated H3K4 methylation in the regulation of
transcriptional initiation by RNA polymerase II. Mol Cell Biol 29: 6074-6085
90. Wang S, Fisher K, Poulin GB (2011) Lineage specific trimethylation of H3 on
lysine 4 during C. elegans early embryogenesis. Dev Biol 355: 227-238
91. Wang Z, Zang C, Cui K, Schones DE, Barski A, Peng W, Zhao K (2009b)
Genome-wide mapping of HATs and HDACs reveals distinct functions in
active and inactive genes. Cell 138: 1019-1031
92. Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S, Chen Z, Spooner E, Li E,
Zhang G, Colaiacovo M, Shi Y (2006) Reversal of histone lysine trimethylation
by the JMJD2 family of histone demethylases. Cell 125: 467-481
93. Workman JL, Kingston RE (1998) Alteration of nucleosome structure as a
mechanism of transcriptional regulation. Annu Rev Biochem 67: 545-579
94. Wright DE, Wang CY, Kao CF (2011) Flickin' the ubiquitin switch: the role of
H2B ubiquitylation in development. Epigenetics 6: 1165-1175
95. Wu J, Ellison J, Salido E, Yen P, Mohandas T, Shapiro LJ (1994a) Isolation and
characterization of XE169, a novel human gene that escapes X-inactivation.
Hum Mol Genet 3: 153-160
96. Wu J, Salido EC, Yen PH, Mohandas TK, Heng HH, Tsui LC, Park J, Chapman
VM, Shapiro LJ (1994b) The murine Xe169 gene escapes X-inactivation like
its human homologue. Nat Genet 7: 491-496
97. Wu X, Gong Y, Yue J, Qiang B, Yuan J, Peng X (2008) Cooperation between
EZH2, NSPc1-mediated histone H2A ubiquitination and Dnmt1 in HOX gene
silencing. Nucleic Acids Res 36: 3590-3599
98. Xia X, Lemieux ME, Li W, Carroll JS, Brown M, Liu XS, Kung AL (2009)
Integrative analysis of HIF binding and transactivation reveals its role in
maintaining histone methylation homeostasis. Proc Natl Acad Sci U S A 106:
4260-4265
99. Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, Ma Y, Yu Y, Lin H, Chen AP, Chen CD
(2007) JARID1B is a histone H3 lysine 4 demethylase up-regulated in
prostate cancer. Proc Natl Acad Sci U S A 104: 19226-19231
100. Xie L, Pelz C, Wang W, Bashar A, Varlamova O, Shadle S, Impey S
(2011) KDM5B regulates embryonic stem cell self-renewal and represses
cryptic intragenic transcription. EMBO J 30: 1473-1484
101. Yamane K, Tateishi K, Klose RJ, Fang J, Fabrizio LA, Erdjument-
Bromage H, Taylor-Papadimitriou J, Tempst P, Zhang Y (2007) PLU-1 is an
H3K4 demethylase involved in transcriptional repression and breast cancer
cell proliferation. Mol Cell 25: 801-812
102. Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P,
Wong J, Zhang Y (2006) JHDM2A, a JmjC-containing H3K9 demethylase,
facilitates transcription activation by androgen receptor. Cell 125: 483-495
103. Yoshida M, Ishimura A, Terashima M, Enkhbaatar Z, Nozaki N, Satou K,
Suzuki T (2011) PLU1 histone demethylase decreases the expression of
KAT5 and enhances the invasive activity of the cells. Biochem J 437: 555-564
104. Yu V, Fisch T, Long AM, Tang J, Lee JH, Hierl M, Chen H, Yakowec P,
Schwandner R, Emkey R (2012) High-throughput TR-FRET assays for
identifying inhibitors of LSD1 and JMJD2C histone lysine demethylases. J
Biomol Screen 17: 27-38

Weitere ähnliche Inhalte

Was ist angesagt?

MSBMB 2016 conference poster
MSBMB 2016 conference posterMSBMB 2016 conference poster
MSBMB 2016 conference poster
Nasiru Abdullahi
 
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Daire Murphy
 
PROTEINS AND MOLECULAR CHAPERONES
PROTEINS AND MOLECULAR CHAPERONESPROTEINS AND MOLECULAR CHAPERONES
PROTEINS AND MOLECULAR CHAPERONES
AnaCano10
 
post translational modifications of protein
post translational modifications of proteinpost translational modifications of protein
post translational modifications of protein
Anandhan Ctry
 
PhD Federica III anno
PhD Federica III annoPhD Federica III anno
PhD Federica III anno
lab13unisa
 
Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...
Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...
Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...
Maciej Luczynski
 

Was ist angesagt? (20)

Brief introduction of post-translational modifications (PTMs)
Brief introduction of post-translational modifications (PTMs)Brief introduction of post-translational modifications (PTMs)
Brief introduction of post-translational modifications (PTMs)
 
Protein folding and aggregation
Protein folding and aggregationProtein folding and aggregation
Protein folding and aggregation
 
Presentation P5
Presentation P5Presentation P5
Presentation P5
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
MCP2009
MCP2009MCP2009
MCP2009
 
PIIS2211124716313158
PIIS2211124716313158PIIS2211124716313158
PIIS2211124716313158
 
Alzheimer’s disease
Alzheimer’s disease Alzheimer’s disease
Alzheimer’s disease
 
MSBMB 2016 conference poster
MSBMB 2016 conference posterMSBMB 2016 conference poster
MSBMB 2016 conference poster
 
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
Characterising the Interactome of EZH2 in Embryonic Stem Cells (3)
 
Strategies for Post-translational Modification (PTM)
Strategies for Post-translational Modification (PTM)Strategies for Post-translational Modification (PTM)
Strategies for Post-translational Modification (PTM)
 
2006 O'Leary et al MBC
2006 O'Leary et al  MBC2006 O'Leary et al  MBC
2006 O'Leary et al MBC
 
Dna modifications
Dna modificationsDna modifications
Dna modifications
 
PROTEINS AND MOLECULAR CHAPERONES
PROTEINS AND MOLECULAR CHAPERONESPROTEINS AND MOLECULAR CHAPERONES
PROTEINS AND MOLECULAR CHAPERONES
 
Protein folding and proteostasis
Protein folding and proteostasisProtein folding and proteostasis
Protein folding and proteostasis
 
post translational modifications of protein
post translational modifications of proteinpost translational modifications of protein
post translational modifications of protein
 
Biotech 2012 spring_8_post-trans
Biotech 2012 spring_8_post-transBiotech 2012 spring_8_post-trans
Biotech 2012 spring_8_post-trans
 
Protein stability(molecular biology)
Protein stability(molecular biology)Protein stability(molecular biology)
Protein stability(molecular biology)
 
PhD Federica III anno
PhD Federica III annoPhD Federica III anno
PhD Federica III anno
 
Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...
Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...
Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-hom...
 
Hemoglobina
HemoglobinaHemoglobina
Hemoglobina
 

Andere mochten auch

Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...
Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...
Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...
shevchuk_conf
 
parents participation
parents participationparents participation
parents participation
anlio
 

Andere mochten auch (20)

Doc16 2.7 guión de programación
Doc16 2.7 guión de programaciónDoc16 2.7 guión de programación
Doc16 2.7 guión de programación
 
1. cover isal 2
1. cover isal 21. cover isal 2
1. cover isal 2
 
Módulo 2.2 doc12 análisis puesto trabajo
Módulo 2.2 doc12 análisis puesto trabajoMódulo 2.2 doc12 análisis puesto trabajo
Módulo 2.2 doc12 análisis puesto trabajo
 
Doc1
Doc1Doc1
Doc1
 
2
22
2
 
Rec08 cuarto. módulo de métodos y materiales
Rec08 cuarto. módulo de métodos y materialesRec08 cuarto. módulo de métodos y materiales
Rec08 cuarto. módulo de métodos y materiales
 
Rec02 elementos de comunicación
Rec02 elementos de comunicaciónRec02 elementos de comunicación
Rec02 elementos de comunicación
 
Laserfiche Empower 2017 information managementaas
Laserfiche Empower 2017 information managementaasLaserfiche Empower 2017 information managementaas
Laserfiche Empower 2017 information managementaas
 
Og fp
Og fpOg fp
Og fp
 
Circle of influence
Circle of influenceCircle of influence
Circle of influence
 
Виталий Колосов — «Ключевые особенности работы по СРА в арабском регионе (на ...
Виталий Колосов — «Ключевые особенности работы по СРА в арабском регионе (на ...Виталий Колосов — «Ключевые особенности работы по СРА в арабском регионе (на ...
Виталий Колосов — «Ключевые особенности работы по СРА в арабском регионе (на ...
 
Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...
Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...
Александр Борняков — «Как заработать на видеорекламе в США», доклад на конфер...
 
WebCamp 2016: Python. Левон Авакян: Архитектура мета игры Wargaming. Глобальн...
WebCamp 2016: Python. Левон Авакян: Архитектура мета игры Wargaming. Глобальн...WebCamp 2016: Python. Левон Авакян: Архитектура мета игры Wargaming. Глобальн...
WebCamp 2016: Python. Левон Авакян: Архитектура мета игры Wargaming. Глобальн...
 
7.2 la evaluación en la fpe
7.2 la evaluación en la fpe7.2 la evaluación en la fpe
7.2 la evaluación en la fpe
 
The Four Candles
The Four CandlesThe Four Candles
The Four Candles
 
Think
ThinkThink
Think
 
Plano de aula prosa e poesia (4° bimestre)
Plano de aula prosa e poesia (4° bimestre) Plano de aula prosa e poesia (4° bimestre)
Plano de aula prosa e poesia (4° bimestre)
 
Visual resume
Visual resumeVisual resume
Visual resume
 
The Ultimate Guide To Event Insurance
The Ultimate Guide To Event InsuranceThe Ultimate Guide To Event Insurance
The Ultimate Guide To Event Insurance
 
parents participation
parents participationparents participation
parents participation
 

Ähnlich wie Histone demethylase and it srole in cell biology review

Epigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digestEpigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digest
Boobalan Pachaiyappan
 
Patten DA et al. - EMBO J - 2014
Patten DA et al. - EMBO J - 2014Patten DA et al. - EMBO J - 2014
Patten DA et al. - EMBO J - 2014
Jacob Wong
 
Types of histones, histone modifications and their effects
Types of histones, histone modifications and their effectsTypes of histones, histone modifications and their effects
Types of histones, histone modifications and their effects
AnuKiruthika
 
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
SSR Institute of International Journal of Life Sciences
 

Ähnlich wie Histone demethylase and it srole in cell biology review (20)

AlexGriffith432
AlexGriffith432AlexGriffith432
AlexGriffith432
 
Epigenetics
Epigenetics  Epigenetics
Epigenetics
 
Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2
 
Lecture_Chromatin remodelling_slideshare.pdf
Lecture_Chromatin remodelling_slideshare.pdfLecture_Chromatin remodelling_slideshare.pdf
Lecture_Chromatin remodelling_slideshare.pdf
 
Epigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digestEpigenetic modulators - review - BMCL digest
Epigenetic modulators - review - BMCL digest
 
Patten DA et al. - EMBO J - 2014
Patten DA et al. - EMBO J - 2014Patten DA et al. - EMBO J - 2014
Patten DA et al. - EMBO J - 2014
 
Histone modification in living cells
Histone modification in living cellsHistone modification in living cells
Histone modification in living cells
 
Epigenetics role in diseases
Epigenetics role in diseasesEpigenetics role in diseases
Epigenetics role in diseases
 
cancer nice paper.pdf
cancer nice paper.pdfcancer nice paper.pdf
cancer nice paper.pdf
 
Chromatin remodeling
Chromatin remodelingChromatin remodeling
Chromatin remodeling
 
Histone modifications
Histone modificationsHistone modifications
Histone modifications
 
EngenuitySC's Science Cafe - March with Dr. Patrick Woster
EngenuitySC's Science Cafe - March with Dr. Patrick WosterEngenuitySC's Science Cafe - March with Dr. Patrick Woster
EngenuitySC's Science Cafe - March with Dr. Patrick Woster
 
Examples Of Epigenetics
Examples Of EpigeneticsExamples Of Epigenetics
Examples Of Epigenetics
 
epigenetics.pptx
epigenetics.pptxepigenetics.pptx
epigenetics.pptx
 
Types of histones, histone modifications and their effects
Types of histones, histone modifications and their effectsTypes of histones, histone modifications and their effects
Types of histones, histone modifications and their effects
 
HOXD13 I34L
HOXD13 I34LHOXD13 I34L
HOXD13 I34L
 
Epigenetics- Transcription regulation of gene expression
Epigenetics- Transcription regulation of gene expressionEpigenetics- Transcription regulation of gene expression
Epigenetics- Transcription regulation of gene expression
 
Epigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plantsEpigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plants
 
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
Effect of Environmental Chemical Exposures on Epigenetics of Diseases: A Syst...
 
Epigenetic regulation of rice flowering and reproduction
Epigenetic regulation of rice flowering and reproductionEpigenetic regulation of rice flowering and reproduction
Epigenetic regulation of rice flowering and reproduction
 

Mehr von Christopher Wynder

Mehr von Christopher Wynder (20)

Opentext brings intelligence to Pharma and biotech
Opentext brings intelligence to Pharma and biotechOpentext brings intelligence to Pharma and biotech
Opentext brings intelligence to Pharma and biotech
 
Preparing for a digital future in healthcare
Preparing for a digital future in healthcarePreparing for a digital future in healthcare
Preparing for a digital future in healthcare
 
Ensuring document control for healthcare vendors
Ensuring document control for healthcare vendorsEnsuring document control for healthcare vendors
Ensuring document control for healthcare vendors
 
Automating Patient Management with ApplicationXtender Workflow
Automating Patient Management with ApplicationXtender WorkflowAutomating Patient Management with ApplicationXtender Workflow
Automating Patient Management with ApplicationXtender Workflow
 
OpenText Content Services for healthcare
OpenText Content Services for healthcareOpenText Content Services for healthcare
OpenText Content Services for healthcare
 
OpenText ApplicationXtender -the basics
OpenText ApplicationXtender -the basicsOpenText ApplicationXtender -the basics
OpenText ApplicationXtender -the basics
 
Open text applicationxtender 16.3 highlights customerfacing
Open text applicationxtender 16.3 highlights customerfacingOpen text applicationxtender 16.3 highlights customerfacing
Open text applicationxtender 16.3 highlights customerfacing
 
ECNO 2017 Paperless Student Administration
ECNO 2017 Paperless Student AdministrationECNO 2017 Paperless Student Administration
ECNO 2017 Paperless Student Administration
 
Information Management aaS AIIM First Canadian presentation
Information Management aaS AIIM First Canadian presentationInformation Management aaS AIIM First Canadian presentation
Information Management aaS AIIM First Canadian presentation
 
Information management as a Service
Information management as a ServiceInformation management as a Service
Information management as a Service
 
ECNO 2016-Using ECM to gain administrative efficiency for school boards
ECNO 2016-Using ECM to gain administrative efficiency for school boardsECNO 2016-Using ECM to gain administrative efficiency for school boards
ECNO 2016-Using ECM to gain administrative efficiency for school boards
 
Digital signatures whitepaper_thinkdox
Digital signatures whitepaper_thinkdoxDigital signatures whitepaper_thinkdox
Digital signatures whitepaper_thinkdox
 
ARMA Southwest Ontario InfoSpeaks
ARMA Southwest Ontario InfoSpeaks ARMA Southwest Ontario InfoSpeaks
ARMA Southwest Ontario InfoSpeaks
 
ThinkDox ECM implmentation and support services
ThinkDox ECM implmentation and support servicesThinkDox ECM implmentation and support services
ThinkDox ECM implmentation and support services
 
Laserfiche10 highlights- how the new features can benefit your mobile and wor...
Laserfiche10 highlights- how the new features can benefit your mobile and wor...Laserfiche10 highlights- how the new features can benefit your mobile and wor...
Laserfiche10 highlights- how the new features can benefit your mobile and wor...
 
Laserfiche empowercon302 2016
Laserfiche empowercon302 2016Laserfiche empowercon302 2016
Laserfiche empowercon302 2016
 
Gov civilworkshop
Gov civilworkshopGov civilworkshop
Gov civilworkshop
 
Bring IT together_2015_ECOOandOASBO
Bring IT together_2015_ECOOandOASBOBring IT together_2015_ECOOandOASBO
Bring IT together_2015_ECOOandOASBO
 
Ecm implementation planning_workshop_hospital_sample
Ecm implementation planning_workshop_hospital_sampleEcm implementation planning_workshop_hospital_sample
Ecm implementation planning_workshop_hospital_sample
 
AMCTO presentation on moving from records managment to information management
AMCTO presentation on moving from records managment to information managementAMCTO presentation on moving from records managment to information management
AMCTO presentation on moving from records managment to information management
 

Kürzlich hochgeladen

Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.
Silpa
 
Digital Dentistry.Digital Dentistryvv.pptx
Digital Dentistry.Digital Dentistryvv.pptxDigital Dentistry.Digital Dentistryvv.pptx
Digital Dentistry.Digital Dentistryvv.pptx
MohamedFarag457087
 
Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.
Silpa
 
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
?#DUbAI#??##{{(☎️+971_581248768%)**%*]'#abortion pills for sale in dubai@
 
Module for Grade 9 for Asynchronous/Distance learning
Module for Grade 9 for Asynchronous/Distance learningModule for Grade 9 for Asynchronous/Distance learning
Module for Grade 9 for Asynchronous/Distance learning
levieagacer
 
Cyathodium bryophyte: morphology, anatomy, reproduction etc.
Cyathodium bryophyte: morphology, anatomy, reproduction etc.Cyathodium bryophyte: morphology, anatomy, reproduction etc.
Cyathodium bryophyte: morphology, anatomy, reproduction etc.
Silpa
 
CYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptxCYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptx
Silpa
 
POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.
Silpa
 
The Mariana Trench remarkable geological features on Earth.pptx
The Mariana Trench remarkable geological features on Earth.pptxThe Mariana Trench remarkable geological features on Earth.pptx
The Mariana Trench remarkable geological features on Earth.pptx
seri bangash
 
Human genetics..........................pptx
Human genetics..........................pptxHuman genetics..........................pptx
Human genetics..........................pptx
Silpa
 
development of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virusdevelopment of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virus
NazaninKarimi6
 

Kürzlich hochgeladen (20)

FAIRSpectra - Enabling the FAIRification of Analytical Science
FAIRSpectra - Enabling the FAIRification of Analytical ScienceFAIRSpectra - Enabling the FAIRification of Analytical Science
FAIRSpectra - Enabling the FAIRification of Analytical Science
 
GBSN - Microbiology (Unit 3)Defense Mechanism of the body
GBSN - Microbiology (Unit 3)Defense Mechanism of the body GBSN - Microbiology (Unit 3)Defense Mechanism of the body
GBSN - Microbiology (Unit 3)Defense Mechanism of the body
 
Selaginella: features, morphology ,anatomy and reproduction.
Selaginella: features, morphology ,anatomy and reproduction.Selaginella: features, morphology ,anatomy and reproduction.
Selaginella: features, morphology ,anatomy and reproduction.
 
Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.
 
Digital Dentistry.Digital Dentistryvv.pptx
Digital Dentistry.Digital Dentistryvv.pptxDigital Dentistry.Digital Dentistryvv.pptx
Digital Dentistry.Digital Dentistryvv.pptx
 
Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.Porella : features, morphology, anatomy, reproduction etc.
Porella : features, morphology, anatomy, reproduction etc.
 
Proteomics: types, protein profiling steps etc.
Proteomics: types, protein profiling steps etc.Proteomics: types, protein profiling steps etc.
Proteomics: types, protein profiling steps etc.
 
Call Girls Ahmedabad +917728919243 call me Independent Escort Service
Call Girls Ahmedabad +917728919243 call me Independent Escort ServiceCall Girls Ahmedabad +917728919243 call me Independent Escort Service
Call Girls Ahmedabad +917728919243 call me Independent Escort Service
 
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptxPSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
 
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
 
Module for Grade 9 for Asynchronous/Distance learning
Module for Grade 9 for Asynchronous/Distance learningModule for Grade 9 for Asynchronous/Distance learning
Module for Grade 9 for Asynchronous/Distance learning
 
Cyathodium bryophyte: morphology, anatomy, reproduction etc.
Cyathodium bryophyte: morphology, anatomy, reproduction etc.Cyathodium bryophyte: morphology, anatomy, reproduction etc.
Cyathodium bryophyte: morphology, anatomy, reproduction etc.
 
CYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptxCYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptx
 
Site Acceptance Test .
Site Acceptance Test                    .Site Acceptance Test                    .
Site Acceptance Test .
 
POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.POGONATUM : morphology, anatomy, reproduction etc.
POGONATUM : morphology, anatomy, reproduction etc.
 
FAIRSpectra - Enabling the FAIRification of Spectroscopy and Spectrometry
FAIRSpectra - Enabling the FAIRification of Spectroscopy and SpectrometryFAIRSpectra - Enabling the FAIRification of Spectroscopy and Spectrometry
FAIRSpectra - Enabling the FAIRification of Spectroscopy and Spectrometry
 
The Mariana Trench remarkable geological features on Earth.pptx
The Mariana Trench remarkable geological features on Earth.pptxThe Mariana Trench remarkable geological features on Earth.pptx
The Mariana Trench remarkable geological features on Earth.pptx
 
Bhiwandi Bhiwandi ❤CALL GIRL 7870993772 ❤CALL GIRLS ESCORT SERVICE In Bhiwan...
Bhiwandi Bhiwandi ❤CALL GIRL 7870993772 ❤CALL GIRLS  ESCORT SERVICE In Bhiwan...Bhiwandi Bhiwandi ❤CALL GIRL 7870993772 ❤CALL GIRLS  ESCORT SERVICE In Bhiwan...
Bhiwandi Bhiwandi ❤CALL GIRL 7870993772 ❤CALL GIRLS ESCORT SERVICE In Bhiwan...
 
Human genetics..........................pptx
Human genetics..........................pptxHuman genetics..........................pptx
Human genetics..........................pptx
 
development of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virusdevelopment of diagnostic enzyme assay to detect leuser virus
development of diagnostic enzyme assay to detect leuser virus
 

Histone demethylase and it srole in cell biology review

  • 1. Histone demethylation enzymes and dynamic cell biology Leanne Stalker and Christopher Wynder2 Introduction In order to maintain structure and organization within the nucleus of a eukaryotic cell, the large DNA macromolecule is structured in to chromosomes. To provide an additional layer of organization, these chromosomes are wrapped around protein complexes containing proteins known as histones to form the basic unit of chromatin, the nucleosome (Kornberg, 1974; Kornberg & Lorch, 1999). Each nucleosome is comprised of an octameric core containing two each of Histone H2A, H2B, H3 and H4 around which 146bp of DNA is wound. This DNA is then secured to the core by an additional histone, histone H1(Kornberg & Lorch, 1999; Kouzarides, 2007; Sims et al, 2003; Volkel & Angrand, 2007). This DNA/protein complex provides a mechanism by which to conform the large DNA molecule to the confined space of the nucleus, allows protection from DNA damage during cell division, and plays a pertinent role in transcriptional regulation(Kooistra & Helin, 2012; Kouzarides, 2007). Each individual histone protein contains two highly conserved protein domains including a large globular core and an amino terminal tail that protrudes from both the histone individually and the nucleosomal structure as a whole(Luger et al, 1997). From a gene regulation perspective, these N-terminal tails represent an infinite ability for the nucleosomal structure to become modified.
  • 2. Histone tail modifications Due to their availability outside of the core nucleosome, many amino acid residues on histone tails are targets of extensive post transcriptional modifications. These occur on specific amino acid residues and include acetylations, phosphorylations, SUMOylations, ubiquitinations and methylations. The result of the addition of these molecular groups is varied and depends highly on both the specific amino acid modified and the modification itself (Kouzarides, 2007). The addition of these various groups tends to result in one of two possible consequences. First, it may change the interaction between DNA and the histone directly leading to an alteration of the chromatin structure as a whole. This activity is observed mostly when a posttranscriptional modification, such as an acetylation, alters the charge of an amino acid on the histone tail. Acetylation of a lysine (K) residue acts to neutralize its basic charge. This loosens the interaction between the histone and DNA, increasing the accessibility of the DNA and generally resulting in transcriptional activation(Shogren-Knaak et al, 2006; Workman & Kingston, 1998). Acetylation is the most extensively studied of the post-transcriptional modifications and occurs most frequently on residues K9, K14, K18 and K56 of Histone H3. The enzymes responsible for both the addition of the acetyl group, Histone Aceytl Transferases (HATs) and the enzymes responsible for the removal of the acetyl group, Histone Deacetylases (HDACs) have been increasingly popular targets for drug discovery(Khan & Khan, 2010; Kuo & Allis, 1998).
  • 3. The second consequence of histone modification is the alteration of non- histone protein recruitment to histone tails. For example, histone phosphorylating enzymes MSK1/2 and RSK2 tend to target serine residues at H3S10. Phosphorylation of this residue is found to attract the phospho-binding protein 14-3-3, which is thought to activate NFB-regulated genes(Banerjee & Chakravarti, 2011; Kouzarides, 2007). Greater understanding of the role of histone phosphorylation is yet to be determined. Ubiquitylation and SUMOylation differ from the aforementioned mechanisms because they require the addition of large moieties(Berger, 2007). The function of ubiquitylation remains unclear but its mechanism of action is believed to either act to recruit supplementary proteins to histone tails or physically “wedge” chromatin open due to its size. Functional effects of ubiquitylation appear to vary depending on the residue to which the moiety is added. For example, ubiquitylation of H2BK123 is associated with the activation of transcription while ubiquitylation of H2AK119 by NSPc1 has been found to cooperate with DNA methylation correlate with the transcriptional silencing of Hox genes.(Wright et al, 2011; Wu et al, 2008). Conversely, the result of sumoylation is believed to be mainly transcriptionally repressive(Nathan et al, 2006). Histone Methylation Recently, much interest has been placed on the regulation of histone tail methylation. Unlike the previously mentioned modifications, methylation can occur on both lysine and arginine (R) residues on amino terminal histone
  • 4. tails(Shilatifard, 2006; Sims et al, 2003). This modification has also been found to be processive, suggesting that unlike acetylation, which is either present or absent, methylation potentially allows for an increased ability to fine tune regulation. An arginine can become mono or dimethylated, the latter of which can be either symmetrical or asymmetrical. Whereas a lysine can be modified in a mono- or di- and tri-methylated form, each of which has been found to have a differing effect (Cloos et al, 2008; Santos-Rosa et al, 2002). Methylation does not alter the charge of the histone tail. Therefore, this modification is not thought to play a direct role in DNA/ histone interactions. Rather, methylation can result in a modulation of chromatin structure, altering the accessibility to chromatin to effector proteins, or may act as a recruitment signal for regulatory factors(Cloos et al, 2008). This results in transcriptional alterations due to changes in the chromatin landscape as a whole (Bannister et al, 2002; Lachner et al, 2001). Histone methylations have been found to be associated with both transcriptional activation and repression with methylation of K4 and K36 of H3 being generally ascribed to gene activation, whereas association with K9 and K27 of the same histone are generally thought to be involved with transcriptional repression(Berger, 2007). The enzymes responsible, known as lysine methyltransferases (HMTs) are unique in the sense that they are residue specific. For instance, the Set1/COMPASS or MLL class of histone methyltransferases are specific for the methylation (mono-, di-, and tri-) of H3K4, while the Su(var)3-9 family is restricted to methylation of H3K9 (Kouzarides, 2007)
  • 5. Demethylation of the histone tail Historically, methylation was considered to be a mark of permanence. Without the discovery of an enzyme class capable of the removal of methylation, it was thought that these marks were static. The discovery of the enzyme KDM1a (also known as LSD1, BHC110) in 2004, changed this notion. KDM1a was found to have the ability to catalyze the demethylation of histone residues by a flavin adenine dinucleotide (FAD)-dependent amine oxidase reaction. However, the enzymology of this demethylase requires a protonated methyl -ammonium in its substrate. This is absent in the trimethylated version of methylation, resulting in the conclusion that this enzyme was restricted to mono and dimethylated modifications(Shi et al, 2004). Since then a more novel, larger protein group named the Jumonji (JMJC) domain family has been discovered. These enzymes catalyze the removal of methylation marks utilizing a hydroxylation reaction through their JMJC domain. This reaction no longer requires a protonated methyl -ammonium, allowing for the demethylation of all three methyl states. In several cases, the trimethylated version is actually the preferred substrate(Christensen et al, 2007; Fodor et al, 2006; Klose et al, 2006; Tsukada et al, 2006; Whetstine et al, 2006). Historically, F-Box and Leu-rich repeat protein 11 (FBXL11) was the first enzyme discovered in this class; it has demethylase activity towards both the mono and dimethylated versions of H3K36 (Tsukada et al, 2006).
  • 6. To date, JMJC enzymes of this class have been found to be active on H3K4(Iwase et al, 2007; Klose et al, 2007; Lee et al, 2007; Secombe & Eisenman, 2007; Seward et al, 2007; Tahiliani et al, 2007; Yamane et al, 2007) ; H3K9(Yamane et al, 2006), H3K27(Agger et al, 2007; De Santa et al, 2007; Lan et al, 2007), H3K36(Fodor et al, 2006) and H4K20(Liu et al, 2010). This has led to the current understanding that methylation represents an extremely flexible and dynamic modification state resulting in the active modulation of transcription. Though the JMJC class of demethylases as a whole is an expansive protein family (the human genome encodes 30 different JMJC containing proteins, 18 of which have been proven to show demethylase acitivity on both arginine and lysine residues(Kooistra & Helin, 2012) phylogeny has suggested that within this family there are several clusters of proteins which appear to group together in both structure and function. The KDM5 family of demethylases, known to target all three methylation states of H3K4, represents one such cluster(Cloos et al, 2008) Specific function of the KDM5 family of HDM enzymes The KDM5 family of JMJC demethylases includes four known members: KDM5a, KDM5b, KDM5c and KDM5d (previously known as Jarid1a, Jarid1b, Jarid1c and Jarid1d respectively). As seen in Figure 1; these demethylases are highly conserved structurally and are characterized by the presence of five protein domains:JmjN and JmjC domains required for demethylation activity a
  • 7. BRIGHT/ARID domain for A/T DNA binding, and both a C5HC2-Zinc finger domain and several PHD (plant homeobox domains) involved in the enzymes ability to recognize and bind methylated residues and regulate protein-protein interactions(Cloos et al, 2008). This review will concentrate on the known roles of KDM5 proteins in transcriptional regulation, development and disease. For a recent review encompassing all histone demethylases, please see Kooistra et al.(Kooistra & Helin, 2012) H3K4 methylation: a fine balancing act The KDM5 family of histone demethylases act specifically on H3K4 methylation marks, with a preference for trimethylated H3K4 (H3K4me3). Studies of H3K4 methylation and its biological roles have been vast and the majority of studies report the presence of methylated H3K4 as a sign of transcriptional activation(Barski et al, 2007; Pokholok et al, 2005; Schubeler et al, 2004);. H3K4me3 localized to gene promoters allows for transcriptional activation by binding a subunit of TFIID, which then leads to the formation of the initiation complex(Sims et al, 2003; Vermeulen et al, 2007). Though both mono- and di- methylated versions of H3K4 span further into the transcribed protein and have even been found at enhancer elements(Heintzman et al, 2009; Robertson et al, 2008) H3K4me3 remains strongly conserved to the transcriptional start site (TSS)(Cloos et al, 2008; Kooistra & Helin, 2012; Santos-Rosa et al, 2002). As
  • 8. expected due to their conserved enzymatic targets, KDM5 demethylases have been suggested as potent transcriptional repressors through their known ability to remove this activating mark. Recent genome studies have suggested however, that the presence of H3K4me3 at the transcriptional start site is not sufficient to assume active transcription (Guenther et al, 2007). Within embryonic stem cells (ESC) for example, a very high proportion of transcriptional start sites possess marks of both transcriptionally active, and transcriptionally silent chromatin. These sites are said to be bivalent and represent the ability of a non-committed cell to be poised for commitment and development(Azuara et al, 2006; Bernstein et al, 2006). This phenomenon has also been observed lower on the evolutionary scale, with C. elegans showing H3K4me3 and H3K27me3 co-occupying promoters early in development (Wang et al, 2011).This suggests that modification of this methyl mark may represent an ability of the cell to tweak transcription in one direction or the other, without requiring an absolute condition of “On” or “Off”. Studies of both KDM5a and KDM5b have suggested that these demethylases actually co-localize with their substrate, with target genes showing expression of both the enzyme and H3K4me3(Lopez-Bigas et al, 2008; Schmitz et al, 2011). Though expression of H3K4me3 was generally found to be lower at sites of demethylase recruitment, the methylation mark was not completely absent, suggesting that these enzymes function to maintain low levels of H3K4me3 but not to abolish the mark completely. This also suggests that recruitment of additional factors may be required for full demethylase activity of
  • 9. the enzyme, or that the context of the protein complex in which the KDM5 demethylase is present may alter its enzymology. Roles for KDM5 outside of the transcriptional start site Additional groups have suggested a role for KDM5 family members in intragenic regions of the genome. Liefe et al. suggest that KDM5a plays a role in Notch-mediated silencing and that demethylation at specific regulator elements rather than entire promoter TSS regions, is sufficient to result in gene silencing(Liefke et al, 2010) where Xie et al. have also recently suggested that KDM5b may play a role in intragenic transcription and elongation of KDM5b target genes, though these results are currently under debate (Schmitz et al, 2011; Xie et al, 2011). This adds an additional layer of regulation, suggesting that the accuracy of these enzymes for transcriptional regulation is most likely extremely pertinent to sensitive biological functions within the cell, with potentially significant impact on processes including development and differentiation, and that even the smallest of perturbations could wholly or in part give rise to disease or transformation. H3K4me3 and Cellular Identity Previous studies in D. melanogaster have shown that the KDM5 homologue Little Imaginal Disc (LID) is required for normal development to
  • 10. proceed through the regulation of homeotic genes (Gildea et al, 2000) Additionally, the homologue of KDM5 in C.elegans, rbr-2, has been found to be both an active demethylase and to play a role in the normal development of the nematode, dependent upon this enzymology (Christensen et al, 2007). Knock down of rbr-2 was found to result in an increase in H3K4me3 expression and resulted in a disruption to normal vulval development. Most recently, rbr-2 has also been implicated in regulation over C.elegans lifespan (Greer et al, 2010). As both flies and worms only possess one copy of the KDM5 homologue, there is no chance for functional redundancy. Within higher eukaryotes however, the role of these proteins in development becomes increasingly complex. Roles for KDM5 in higher order organisms Though higher order organisms possess four KDM5 family members, their roles appear, in many cases, to be functionally distinct. Knock out studies of KDM5c in a zebrafish model leads to impaired neuronal development. Similar phenomena are observed in rats where dendritic development becomes impaired(Iwase et al, 2007). This suggests that any functional redundancy exhibited by KDM5 family members does not include the role of KDM5c in neural development. This is of interest considering how similar KDM5c and KDM5d, in specific, are, and reiterates the importance of target specificity and expression profile differences between the four family members.
  • 11. Knock out studies in mice continue to support functionally distinct roles for these enzymes. Though viable and possessing only mild behavioural abnormalities, KDM5a -/- mice have been found to have altered transcription of several cytokine genes known to be KDM5a targets. This has been shown to lead to aberrant hematology, altered cell cycle and a resistance to apoptosis of hematopoietic cancers(Wang et al, 2009b). Knockout of KDM5b in mice however, in contrast to family member KDM5a, has been reported to be embryonic lethal around E4.5(Catchpole et al, 2011). This suggests that KDM5b is required in early embryonic development and that this role cannot be taken over by another KDM5 family member. This early functional importance of KDM5b is somewhat to be expected due to differences in KDM5 family member expression profiles. Where KDM5a appears to be widely expressed through all tissues showing high expression in the haematopoetic system(Christensen et al, 2007; Cloos et al, 2008; Klose et al, 2007; Lopez-Bigas et al, 2008) KDM5c, an X linked gene which escapes X linked inactivation(Wu et al, 1994a; Wu et al, 1994b) appears to have more limited expression, showing neuronal expression patterns and playing a role in neuronal development (Iwase et al, 2007). KDM5b shows a completely different profile, widely expressed in ESCs and undifferentiated progenitors(Dey et al, 2008), but limited in adult tissues: restricted to the testis and differentiating mammary gland(Barrett et al, 2002; Lu et al, 1999). Of interest however, KDM5b is highly expressed in several forms of cancer(Barrett et al, 2002; Barrett et al, 2007; Madsen et al, 2003; Roesch et al, 2006; Roesch et al, 2010; Xiang et al, 2007). Catchpole et al. additionally report
  • 12. the creation of a KDM5b mouse strain containing a mutation in which the ARID domain is removed. This mutation has previously been documented to completely obliterate the demethylase activity of KDM5b(Tan et al, 2003; Yamane et al, 2007) though Catchpole et al. suggest that some residual activity is a possibility (Catchpole et al, 2011). Interestingly, though these mice display what is referred to as a “mammary phenotype” they are both viable and fertile suggesting that the role of KDM5b in embryonic development may not hinge completely on its enzymology (Catchpole et al, 2011). To increase the complexity of the KDM5b knockout story, Schmitz et al. have recently suggested that they were successful in creating a KDM5b knock out mouse that is both viable and fertile and suggest that compensation by other family members may rescue the knockout phenotype previously described (Schmitz et al, 2011) KDM5; master regulators of differentiation and development Though KDM5 family knockout mice may remain viable, distinct and numerous defects in differentiation and development are frequently noted. This is suggestive of a protein family involved in the regulation of differentiation control. In 2005, Benevolenskaya et al. found the first evidence of pRB-KDM5a complexes in cells and determined that KDM5a was a key regulator of differentiation control by demonstrating that pRB must displace KDM5a from key promoters in order to promote differentiation (Benevolenskaya et al, 2005). This work was completed previous to the knowledge of KDM5a enzymology. Further
  • 13. study in ESC suggests that during differentation the removal of KDM5a from Hox genes correlates with increased levels of H3K4me3 (Christensen et al, 2007), consistent with its role in cellular differentation and development. Previous work on KDM5b has found that this family member can also repress several target genes important to differentiation including HOXA5(Yamane et al, 2007), Brain Factor-1 (BF-1) and Pax9 (Tan et al, 2003). Recently, work in our laboratory has suggested that KDM5b plays a role in mouse embryonic stem cells (mESC) to maintain a population of uncommitted progenitors. Overexpression of KDM5b in mESC was additionally found to impair specification, and delay or destroy neural differentiation (Dey et al, 2008). More recent studies have supported this work, suggesting that KDM5b is required for neural differentiation, most specifically, the generation of neural progenitors (NPC) from ESC (Schmitz et al, 2011). KDM5b was found to occupy developmental regulator genes in ESC, and as seen previously (Dey et al, 2008) plays a pertinent role in gene regulation in this cell type. Their findings however, suggest that KDM5b is dispensable for the self-renewal capacity of ESC, but absolutely required for differentiation. Of interest, the modulation of KDM protein expression in most cell types results in no change in global H3K4me3 levels, including neural stem cells (NSC) (Schmitz et al, 2011) and MCF7 (Yamane et al, 2007) after the knockdown of KDM5b; and MEFs after the knockdown of KDM5a (Klose et al, 2007). This is however different in ESC where alteration to KDM5b levels appears to have a direct effect on global H3K4me3 levels (Dey et al, 2008; Schmitz et al, 2011). Genome wide chromatin studies have suggested that the
  • 14. global levels of H3K4me3 decrease from the ESC stage over the course of differentiation (Ang et al, 2011) with bivalency being removed through demethylation of H3K4me3 positive promoters (Bernstein et al, 2006). H3K27me3 expression however, appears to remain present. This suggests that the presence of H3K4me3 may be required for early development, although its removal may also represent a required checkpoint for certain stages of differentiation. This selective removal of H3K4me3 seems to be required for appropriate cell fate determination to occur. Studies in C. elegans demonstrate that the appearance of H3K4me3 is both regulated according to cell lineage and that the deposit of this tri-methylation is extremely dynamic (Wang et al, 2011) lending credence to the theory that both the presence and absence of this mark may represent significant methods of gene regulation during development. Interestingly, recent studies categorizing the role of H3K4 methylation in fully differentiated cells such as the cardiomyocyte adds to this work, suggesting that maintenance of H3K4me3 is required to maintain cellular integrity even in a non dividing, fully committed cell type (Stein et al, 2011). This also supports an ideal where though the expression of H3K4me3 may be required to be reduced at certain developmental check- points, that re-expression of this mark does occur at later stages of development. All these data together paint a picture where a fine balance between methylation and demethylation must be maintained in both a lineage and commitment dependent manner. Slight alterations to the expression level or localization of,
  • 15. enzymes required to maintain this balance may result in changes in levels of H3K4me3 in either a global, or gene specific manner which, in turn, could easily result in disease or abnormal cellular phenotypes. Demethylation and disease; a fine balance disrupted Known for their potent roles in development, it is of no surprise that misregulation of several KDM5 family members has been found to play role in several developmental diseases. Mostly targeted to the neurological system, where several KDM5 family members have been studied as developmental regulators, KDM5 family member involvement in diseases other than cancer has been a target of recent study. Past studies of KDM5c have resulted in the striking conclusion that KDM5c regulation is pertinent to appropriate neural development. Though it is known as an H3K4me3 demethylase, KDM5c has also been found to recognize Histone 3 Lysine 9 trimethylation (H3K9me3) (Iwase et al, 2007), and to play a role in RE1 silencing transcription factor (REST) mediated repression, as it has been found to co occupy several REST target genes (Ballas & Mandel, 2005). Loss of KDM5c causes de-repression and increases in H3K43me at key REST targets leading to an impairment of neuronal gene regulation (Tahiliani et al, 2007). Strikingly, KDM5c has been found to be involved in several diseases of neurodevelopment including X linked mental retardation/X linked Intellectual
  • 16. Disability (XLMR/XLID), epilepsy, and autism spectrum disorders (ASD). Many mutations, currently a total of more than 21, to KDM5c have been found and continue to be found associated only with cases of XLMR (Abidi et al, 2008; Jensen et al, 2010; Santos-Reboucas et al, 2011; Tzschach et al, 2006) several of these mutations resulting in a decrease in the ability KDM5c to recognize H3K9me3, or to demethylate H3K4me3; suggesting that the enzyomology of KDM5c may be linked to pathology. One novel mutation was found to alter the start site of KDM5C, presumably resulting in a complete lack of translation (Ounap et al, 2012). Additionally, mutations to KDM5c have been connected to distinct symptomology within XLMR such as memory loss (Simensen et al, 2012). This suggests that specific areas of the brain may be targeted by KDM5c misregulation. In 2008, KDM5c was connected to another neurocognitive phenotype when a missense mutation in exon 16 was found connected to ASD. Though several KDM5c target genes such as BDNF and SCN2A had previously been known to show altered expression in patients presenting with ASD, KDM5c itself had never been implicated (Adegbola et al, 2008). KDM5c is not the only family member with a neurodevelopmental phenotype. KDM5b, another KDM5 family member which is a known regulator of neurological development (Schmitz et al, 2011) has also recently been implicated as a possible player in a congenital variant of Rett Syndrome*, a severe neurodevelopmental disease. Molecular causes of Rett syndrome include the persistent expression of early developmental genes (Urdinguio et al, 2008).
  • 17. Although Rett syndrome is normally classified by a mutation in the X-linked methyl-CpG-binding protein MeCP2 (Kramer & van Bokhoven, 2009), a congenital variant showing FOXG1 truncation has recently been discovered (Ariani et al, 2008; Bahi-Buisson et al, 2010; Mencarelli et al, 2009; Papa et al, 2008) Further analysis of the FOXG1 truncation shows that in both (of the two) observed truncation events, the domain known as the JBD or the KDM5b binding domain, is missing, suggesting that the interaction between FOXG1 and KDM5b is pertinent to the regulation of this disease. A reduction in KDM5b binding would result in a series of downstream effects, causing a reduction in the ability of FOXG1 to repress transcription. This transcriptional change would, in turn, result in a reduction of MeCP2 binding due to a delay in neural differentiation. This may mimic what occurs when MeCP2 itself is mutated, resulting in a similar disease phenotype. Taken together, this data supports the conclusion that alterations to KDM5 proteins result not only in impaired development at the embryonic level, but that these alterations and mutations may translate into long term disabilities- either through functional deficits in the demethylase itself, or through downstream effects on interacting proteins. This also provides additional evidence that each KDM5 family member plays a unique role in the regional and temporal control of chromatin structure, and that compensation by additional family members may not be sufficient to result in phenotypic rescue. (Figure 2) Though examples of KDM5 demethylases in disease appear limited to diseases of a neuro-developmental decree, an increased understanding of these
  • 18. enzymes and how they are regulated will undoubtedly uncover a wide range of diseases in which they contribute to pathogenesis. Research efforts have, until recently, concentrated on understanding the roles of these enzymes in various types of cancer, as detailed below. In many cases KDM5s appear to play a role in turning on correct genes at an incorrect time. This leads us to question whether these enzymes may also play a role in degeneration in disorders such as Alzheimers and Huntington’s disease, by encouraging incorrect signaling, leading to alterations in neural regulatory networks later in life. Demethylation and Cancer; a fine balance turned back on incorrectly? Though they are currently know as transcriptional repressors through their demethylase activity, several KDM5 family members first garnered the attention of researchers long before their enzymology was discovered. KDM5a, for example, was originally identified as an interaction partner for retinoblastoma protein (pRB). As such, it was originally named Retinoblastoma Binding Protein 2 (RBP2) (Benevolenskaya et al, 2005). Further work on KDM5a showed that it binds to genes known to be involved in pluripotency and is active in CD34+ and CD105+ cell populations (known to be markers of HSCs and mesenchymal stem cells (MSCs) respectively (Wang et al, 2009a). KDM5a target gene activation and repression may therefore play a key role in the determination of differentiation profiles in HSCs vs MSCs. Paired with the information gathered from KDM5a null mice, mentioned earlier, this presents a strong case that KDM5a may play a
  • 19. key role in the regulation of the haemotopoetic system including the modulation of haematopoietic cell resistance to apoptosis, a hallmark of several blood cancers. Multitudinous KDM5a target genes are preferentially expressed in leukemia and lymphoma and interestingly, KDM5a has recently been found to be a gene partner involved in Acute Myeloid Leukemia (Wang et al, 2009a). This suggests that its involvement in cancer may not be limited to retinoblastoma and that the pRB/KDM5a axis may be a pertinent player in leukemia pathogenesis as well as a regulator of differentiation and development. Additional studies have supported the role of KDM5a in a tumour suppressor role, including a recent study by Liefke et al. Here they suggest that the switch that regulates Notch target genes includes KDM5a and that through this target specific role, KDM5a may act as a potent tumour suppressor in Notch mediated carcinogenesis (Liefke et al, 2010). KDM5b was additionally recognized prior to its enzymology becoming apparent. Originally known as Plu-1, this protein was first discovered as a target up- regulated in response to Her2/c-ErbB2 in breast cancer cell lines and primary breast cancers (Lu et al, 1999). Of limited expression in most adult tissues, KDM5b shows consistent up regulation in breast and prostate cancers in both human and mice, and has been suggested as a possible oncogene in multiple cancer types. (Barrett et al, 2002; Hayami et al, 2010; Lu et al, 1999; Roesch et al, 2010; Xiang et al, 2007; Yamane et al, 2007) Hayami et al. draw on previous work completed in breast (Yamane) and Prostate (Xiang) cancers and demonstrate that KDM5b is directly involved in the proliferative rate and ability of
  • 20. both lung and bladder cancer cells to escape apoptosis. In concordance with other groups, they demonstrate that reduction of KDM5b level results in alterations to the cell cycle of tumour cells, and a reduction in oncogenic potential (Hayami et al, 2010). Delineating the exact role that KDM5b exerts in cancer has become complex and more and more evidence points towards the theory that cancer should be categorized as a group of diseases, rather than a single dysfunction. KDM5b is known to be a regulator of both oncogenes and tumour suppressors through direct interaction with their promoters, such as BRCA1 in breast cancer (Yamane et al, 2007). It has also been associated with cell cycle control in both an accelerating (breast cancer)(Yamane et al, 2007) and decelerating (Melanoma) (Roesch et al, 2010) fashion and has been found to increase the invasive potential of non invasive cell types through repression of the tumour suppressor KAT5 (Yoshida et al, 2011). Recently, KDM5b has also been demonstrated to promote cell cycle progression in breast cancer cells by the epigenetic modulation of the expression of micro RNA let7e suggesting an additional, indirect method to regulate of gene expression (Mitra et al, 2011). In the recent years, another role of KDM5b in tumor survival has surfaced, suggesting that KDM5b may be required for the adaptation of cells to hypoxia. Solid tumors are considered to be highly hypoxic compared to surrounding tissue, and adaptation to this state is pertinent for tumor survival (Semenza, 2003). Adaptation to hypoxia is driven through Hypoxia inducible factor-1 (HIF-1) and is largely mediated through transcriptional repression.
  • 21. Recent screens for proteins that facilitate this adaptation noted several Jumonji family demethylases, including KDM5b. KDM5b was found to be a direct HIF target and shows increased expression under hypoxic conditions (Xia et al, 2009). Previous work has shown that reduced H3K4 methylation is linked to poor prognosis in cancer patients (Seligson et al, 2005), suggesting that an ability to demethylate H3K4 is important for tumor survival. Due to the requirement of dioxygenases such as KDM5b, for molecular oxygen, Xia et al. propose that the increased expression level of these enzymes may represent a compensatory mechanism in response to decreasing oxygen availability (Xia et al, 2009). Without this compensatory mechanism, H3K4me3 levels would be expected to increase as tumors increase in size and oxygen levels decrease, leading to the death of the hypoxic tumor cells. An increase in the expression level of demethylases such as KDM5b may provide a mechanism for the tumor to maintain low H3K4me3 levels even in situations where decreased oxygen levels are present. This novel mechanism may allow tumors to literally skirt death and continue to proliferate. KDM5c, a demethylase more commonly thought to exert control over neuronal identity, is over expressed in both prostate tumors and seminomas, and has been shown to act as a co-repressor to Smad3. Binding of KDM5c to Smad3 blocks its transactivation ability, thus reducing its ability to act as an effector of the TGF-B pathway. Blockage of this pathway is apparent in several cancer types suggesting that KDM5c may possess oncogenic potential
  • 22. through its ability to block Smad3. Most interestingly, this appears to be independent of its demethylase activity (Kim et al, 2008). Recently, Niu et al. explored the role of KDM5c in clear cell renal cell carcinoma (ccRCC). A high proportion of ccRCCs show inactivation of the tumour suppressor von Hippel- Lindau (VHL). Additionally, VHL-/- tumours show decreased levels of H3K4me3 compared to their VHL +/+ counterparts. Interestingly, this was also shown to be Hypoxia inducible factor- (HIF1-) dependent. Previous work, demonstrating gene alterations in patient samples of ccRCC, had provided evidence that mutations to KDM5c were higher than would be expected by chance in ccRCC patients (Dalgliesh et al, 2010), suggesting a connection between KDM5c alterations and aberrant levels of H3K4me3. Niu et al. have shown that KDM5c is responsible for suppressing HIF response genes by removal of H3K4me3, and that mutations to KDM5c are promote tumour growth. This tumour suppressor role of KDM5c is specific to this family member as loss of KDM5c (but not KDM5a or KDM5b) abolished the difference between VHL-/- and +/+ tumors (Niu et al, 2012). Given their role in stem cell biology and development, we are left to question whether KDM5s simply do the “right” job at the “wrong” time in cancers; exerting control similar to non-pathogenic contexts during differentiation and development, but with aberrant results within a fully developed tissue. The roles of KDM5s during carcinogenesis appear to focus on helping tumour cells to survive in contexts when appropriate cellular signaling would lead to cell death; survival of hypoxia, escaping apoptosis, increasing potential for invasion, and
  • 23. alterations to cell cycle leading to over proliferation and the development of inappropriate cell types. However, information on the roles of these proteins are often contradictory, with several being classified as proteins with both oncogenic and tumour suppressor abilities depending on cellular context. Though, as previously mentioned, reduced H3K4 methylation levels appear to be linked to poor prognosis in cancer patients (Seligson et al, 2005), in the case presented above, increased H3K4 in the context of HIF response genes in ccRCC appears to be tumour-promoting. This again draws attention to the fine balance of H3K4me3 expression and the regulation of the enzymes that control this methylation, both are highly dependent upon cellular context. KDM5s in tumour sub populations Several groups have now suggested that KDM5 family members exert control in specific subsets of a tumour population to maintain or promote growth. Sharma et al. noted a population of “reversibly drug tolerant” cells within several human cancers which maintain viability through an altered chromatin state requiring KDM5a. These cells appear absolutely required to protect tumors from eradication (Sharma et al, 2010). Roesch et al. show another angle of the KDM5 cancer story, using the expression of KDM5b as a biomarker to flag a small population of slow cycling cells within the heterogeneous population of a melanoma (Roesch et al, 2010). These “slow” cells appear to be required for tumour maintenance, giving rise to progeny which express low levels of KDM5b,
  • 24. and knock down of KDM5b results in an exhaustion of tumour growth. Interestingly the same group has also proposed that KDM5b has a tumour suppressor role (Roesch et al, 2006; Roesch et al, 2008). It has been suggested that the acceleration of cell cycle in these melanocytes after KDM5b expression decrease may be due to a derepression of E2F-target genes, thus accelerating cell cycle. Both KDM5b and KDM5a have been shown to be members of the Rb repression complex, required for the repression of E2F target genes during senescence (Chicas et al, 2012; Nijwening et al, 2011). Though repression of E2F targets would generally be considered a tumour suppressive function, mutations to Rb are common in cancer progression, allowing pro- proliferative effects to override normal suppression and could lead to increased oncogenic potential. Following in this theory, loss of KDM5a in a pRb defective tumour context promotes senescence and differentiation, suggestive of an oncogenic role in the absence of Rb (Lin et al, 2011). As noted by Chicas et al., this highlights the context- dependent role of these demethylases (Chicas et al, 2012). These results together suggest that though they are involved in oncogenesis, KDM5s appear to exert their “tumourogenic potential” in different ways, depending on cellular context and may respond differently depending on which upstream cellular cues become activated (Figure 3). These aspects of KDM5 demethylases, though complex, make them potentially lucrative targets for pharmaceutical intervention. Enzymes are known to provide excellent drug targets and KDM5b in particular, due to its low
  • 25. expression level in most adult human tissues, may provide a potentially safe target for pharmaceuticals. Immunotherapy approaches against KDM5b have been investigated recently with results suggesting that KDM5b may represent a tumour associated antigen (TAA) for breast cancer (Coleman et al, 2010). The major question that remains for future clinical use of KDM5 targeting therapeutics is: How can we utilize this knowledge of KDM5 biology to combat cancer and disease? Histone deacetylase inhibitors have long been the “king” of the epigenetic pharmaceutical industry, with drugs such as Valproic acid, Entinostat and Romadepsin showing large potential in the clinic and earning FDA approval (Song et al, 2011). However, little has been done targeting demethylase enzymes as possible treatment options. Recent studies have demonstrated the release of therapeutic agents against KDM1 and studies of agents against JMJD2 demethylases (Hamada et al, 2010), and novel assays are being developed to screen and identify novel candidates against these targets (Yu et al, 2012). The KDM5 family is not special in this contextual activity. The importance of context and the flexibility that KDMs in general bring to transcriptional control is the key to a variety of processes. Understanding how and when the KDMs interact with both each other and the basal transcriptional machinery will likely provide clues into a myriad of diseases.
  • 26. 1. Abidi FE, Holloway L, Moore CA, Weaver DD, Simensen RJ, Stevenson RE, Rogers RC, Schwartz CE (2008) Mutations in JARID1C are associated with X- linked mental retardation, short stature and hyperreflexia. J Med Genet 45: 787-793 2. Adegbola A, Gao H, Sommer S, Browning M (2008) A novel mutation in JARID1C/SMCX in a patient with autism spectrum disorder (ASD). Am J Med Genet A 146A: 505-511 3. Agger K, Cloos PA, Christensen J, Pasini D, Rose S, Rappsilber J, Issaeva I, Canaani E, Salcini AE, Helin K (2007) UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature 449: 731-734 4. Ang YS, Tsai SY, Lee DF, Monk J, Su J, Ratnakumar K, Ding J, Ge Y, Darr H, Chang B, Wang J, Rendl M, Bernstein E, Schaniel C, Lemischka IR (2011) Wdr5 mediates self-renewal and reprogramming via the embryonic stem cell core transcriptional network. Cell 145: 183-197 5. Ariani F, Hayek G, Rondinella D, Artuso R, Mencarelli MA, Spanhol-Rosseto A, Pollazzon M, Buoni S, Spiga O, Ricciardi S, Meloni I, Longo I, Mari F, Broccoli V, Zappella M, Renieri A (2008) FOXG1 is responsible for the congenital variant of Rett syndrome. Am J Hum Genet 83: 89-93 6. Azuara V, Perry P, Sauer S, Spivakov M, Jorgensen HF, John RM, Gouti M, Casanova M, Warnes G, Merkenschlager M, Fisher AG (2006) Chromatin signatures of pluripotent cell lines. Nat Cell Biol 8: 532-538 7. Bahi-Buisson N, Nectoux J, Girard B, Van Esch H, De Ravel T, Boddaert N, Plouin P, Rio M, Fichou Y, Chelly J, Bienvenu T (2010) Revisiting the phenotype associated with FOXG1 mutations: two novel cases of congenital Rett variant. Neurogenetics 11: 241-249 8. Ballas N, Mandel G (2005) The many faces of REST oversee epigenetic programming of neuronal genes. Curr Opin Neurobiol 15: 500-506 9. Banerjee T, Chakravarti D (2011) A peek into the complex realm of histone phosphorylation. Mol Cell Biol 31: 4858-4873 10. Bannister AJ, Schneider R, Kouzarides T (2002) Histone methylation: dynamic or static? Cell 109: 801-806 11. Barrett A, Madsen B, Copier J, Lu PJ, Cooper L, Scibetta AG, Burchell J, Taylor- Papadimitriou J (2002) PLU-1 nuclear protein, which is upregulated in breast cancer, shows restricted expression in normal human adult tissues: a new cancer/testis antigen? Int J Cancer 101: 581-588
  • 27. 12. Barrett A, Santangelo S, Tan K, Catchpole S, Roberts K, Spencer-Dene B, Hall D, Scibetta A, Burchell J, Verdin E, Freemont P, Taylor-Papadimitriou J (2007) Breast cancer associated transcriptional repressor PLU-1/JARID1B interacts directly with histone deacetylases. Int J Cancer 121: 265-275 13. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K (2007) High-resolution profiling of histone methylations in the human genome. Cell 129: 823-837 14. Benevolenskaya EV, Murray HL, Branton P, Young RA, Kaelin WG, Jr. (2005) Binding of pRB to the PHD protein RBP2 promotes cellular differentiation. Mol Cell 18: 623-635 15. Berger SL (2007) The complex language of chromatin regulation during transcription. Nature 447: 407-412 16. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL, Lander ES (2006) A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125: 315-326 17. Catchpole S, Spencer-Dene B, Hall D, Santangelo S, Rosewell I, Guenatri M, Beatson R, Scibetta AG, Burchell JM, Taylor-Papadimitriou J (2011) PLU- 1/JARID1B/KDM5B is required for embryonic survival and contributes to cell proliferation in the mammary gland and in ER+ breast cancer cells. Int J Oncol 38: 1267-1277 18. Chicas A, Kapoor A, Wang X, Aksoy O, Evertts AG, Zhang MQ, Garcia BA, Bernstein E, Lowe SW (2012) H3K4 demethylation by Jarid1a and Jarid1b contributes to retinoblastoma-mediated gene silencing during cellular senescence. Proc Natl Acad Sci U S A 109: 8971-8976 19. Christensen J, Agger K, Cloos PA, Pasini D, Rose S, Sennels L, Rappsilber J, Hansen KH, Salcini AE, Helin K (2007) RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell 128: 1063-1076 20. Cloos PA, Christensen J, Agger K, Helin K (2008) Erasing the methyl mark: histone demethylases at the center of cellular differentiation and disease. Genes Dev 22: 1115-1140 21. Coleman JA, Correa I, Cooper L, Bohnenkamp HR, Poulsom R, Burchell JM, Taylor-Papadimitriou J (2010) T cells reactive with HLA-A*0201 *peptides from the histone demethylase JARID1B are found in the circulation of breast cancer patients. Int J Cancer
  • 28. 22. Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, Davies H, Edkins S, Hardy C, Latimer C, Teague J, Andrews J, Barthorpe S, Beare D, Buck G, Campbell PJ, Forbes S, Jia M, Jones D, Knott H, Kok CY, Lau KW, Leroy C, Lin ML, McBride DJ, Maddison M, Maguire S, McLay K, Menzies A, Mironenko T, Mulderrig L, Mudie L, O'Meara S, Pleasance E, Rajasingham A, Shepherd R, Smith R, Stebbings L, Stephens P, Tang G, Tarpey PS, Turrell K, Dykema KJ, Khoo SK, Petillo D, Wondergem B, Anema J, Kahnoski RJ, Teh BT, Stratton MR, Futreal PA (2010) Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463: 360-363 23. De Santa F, Totaro MG, Prosperini E, Notarbartolo S, Testa G, Natoli G (2007) The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell 130: 1083-1094 24. Dey BK, Stalker L, Schnerch A, Bhatia M, Taylor-Papidimitriou J, Wynder C (2008) The histone demethylase KDM5b/JARID1b plays a role in cell fate decisions by blocking terminal differentiation. Mol Cell Biol 28: 5312-5327 25. Fodor BD, Kubicek S, Yonezawa M, O'Sullivan RJ, Sengupta R, Perez-Burgos L, Opravil S, Mechtler K, Schotta G, Jenuwein T (2006) Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev 20: 1557-1562 26. Gildea JJ, Lopez R, Shearn A (2000) A screen for new trithorax group genes identified little imaginal discs, the Drosophila melanogaster homologue of human retinoblastoma binding protein 2. Genetics 156: 645-663 27. Greer EL, Maures TJ, Hauswirth AG, Green EM, Leeman DS, Maro GS, Han S, Banko MR, Gozani O, Brunet A (2010) Members of the H3K4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature 466: 383-387 28. Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007) A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130: 77-88 29. Hamada S, Suzuki T, Mino K, Koseki K, Oehme F, Flamme I, Ozasa H, Itoh Y, Ogasawara D, Komaarashi H, Kato A, Tsumoto H, Nakagawa H, Hasegawa M, Sasaki R, Mizukami T, Miyata N (2010) Design, synthesis, enzyme-inhibitory activity, and effect on human cancer cells of a novel series of jumonji domain- containing protein 2 histone demethylase inhibitors. J Med Chem 53: 5629- 5638 30. Hayami S, Yoshimatsu M, Veerakumarasivam A, Unoki M, Iwai Y, Tsunoda T, Field HI, Kelly JD, Neal DE, Yamaue H, Ponder BA, Nakamura Y, Hamamoto R
  • 29. (2010) Overexpression of the JmjC histone demethylase KDM5B in human carcinogenesis: involvement in the proliferation of cancer cells through the E2F/RB pathway. Mol Cancer 9: 59 31. Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, Ye Z, Lee LK, Stuart RK, Ching CW, Ching KA, Antosiewicz-Bourget JE, Liu H, Zhang X, Green RD, Lobanenkov VV, Stewart R, Thomson JA, Crawford GE, Kellis M, Ren B (2009) Histone modifications at human enhancers reflect global cell- type-specific gene expression. Nature 459: 108-112 32. Iwase S, Lan F, Bayliss P, de la Torre-Ubieta L, Huarte M, Qi HH, Whetstine JR, Bonni A, Roberts TM, Shi Y (2007) The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell 128: 1077-1088 33. Jensen LR, Bartenschlager H, Rujirabanjerd S, Tzschach A, Numann A, Janecke AR, Sporle R, Stricker S, Raynaud M, Nelson J, Hackett A, Fryns JP, Chelly J, de Brouwer AP, Hamel B, Gecz J, Ropers HH, Kuss AW (2010) A distinctive gene expression fingerprint in mentally retarded male patients reflects disease- causing defects in the histone demethylase KDM5C. Pathogenetics 3: 2 34. Khan SN, Khan AU (2010) Role of histone acetylation in cell physiology and diseases: An update. Clin Chim Acta 411: 1401-1411 35. Kim TD, Shin S, Janknecht R (2008) Repression of Smad3 activity by histone demethylase SMCX/JARID1C. Biochem Biophys Res Commun 366: 563-567 36. Klose RJ, Kallin EM, Zhang Y (2006) JmjC-domain-containing proteins and histone demethylation. Nat Rev Genet 7: 715-727 37. Klose RJ, Yan Q, Tothova Z, Yamane K, Erdjument-Bromage H, Tempst P, Gilliland DG, Zhang Y, Kaelin WG, Jr. (2007) The retinoblastoma binding protein RBP2 is an H3K4 demethylase. Cell 128: 889-900 38. Kooistra SM, Helin K (2012) Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol 13: 297-311 39. Kornberg RD (1974) Chromatin structure: a repeating unit of histones and DNA. Science 184: 868-871 40. Kornberg RD, Lorch Y (1999) Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome. Cell 98: 285-294 41. Kouzarides T (2007) Chromatin modifications and their function. Cell 128: 693-705
  • 30. 42. Kramer JM, van Bokhoven H (2009) Genetic and epigenetic defects in mental retardation. Int J Biochem Cell Biol 41: 96-107 43. Kuo MH, Allis CD (1998) Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays 20: 615-626 44. Lachner M, O'Carroll D, Rea S, Mechtler K, Jenuwein T (2001) Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410: 116- 120 45. Lan F, Bayliss PE, Rinn JL, Whetstine JR, Wang JK, Chen S, Iwase S, Alpatov R, Issaeva I, Canaani E, Roberts TM, Chang HY, Shi Y (2007) A histone H3 lysine 27 demethylase regulates animal posterior development. Nature 449: 689- 694 46. Lee MG, Norman J, Shilatifard A, Shiekhattar R (2007) Physical and functional association of a trimethyl H3K4 demethylase and Ring6a/MBLR, a polycomb- like protein. Cell 128: 877-887 47. Liefke R, Oswald F, Alvarado C, Ferres-Marco D, Mittler G, Rodriguez P, Dominguez M, Borggrefe T (2010) Histone demethylase KDM5A is an integral part of the core Notch-RBP-J repressor complex. Genes Dev 24: 590-601 48. Lin W, Cao J, Liu J, Beshiri ML, Fujiwara Y, Francis J, Cherniack AD, Geisen C, Blair LP, Zou MR, Shen X, Kawamori D, Liu Z, Grisanzio C, Watanabe H, Minamishima YA, Zhang Q, Kulkarni RN, Signoretti S, Rodig SJ, Bronson RT, Orkin SH, Tuck DP, Benevolenskaya EV, Meyerson M, Kaelin WG, Jr., Yan Q (2011) Loss of the retinoblastoma binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1. Proc Natl Acad Sci U S A 108: 13379-13386 49. Liu W, Tanasa B, Tyurina OV, Zhou TY, Gassmann R, Liu WT, Ohgi KA, Benner C, Garcia-Bassets I, Aggarwal AK, Desai A, Dorrestein PC, Glass CK, Rosenfeld MG (2010) PHF8 mediates histone H4 lysine 20 demethylation events involved in cell cycle progression. Nature 466: 508-512 50. Lopez-Bigas N, Kisiel TA, Dewaal DC, Holmes KB, Volkert TL, Gupta S, Love J, Murray HL, Young RA, Benevolenskaya EV (2008) Genome-wide analysis of the H3K4 histone demethylase RBP2 reveals a transcriptional program controlling differentiation. Mol Cell 31: 520-530 51. Lu PJ, Sundquist K, Baeckstrom D, Poulsom R, Hanby A, Meier-Ewert S, Jones T, Mitchell M, Pitha-Rowe P, Freemont P, Taylor-Papadimitriou J (1999) A novel gene (PLU-1) containing highly conserved putative DNA/chromatin binding motifs is specifically up-regulated in breast cancer. J Biol Chem 274: 15633-15645
  • 31. 52. Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ (1997) Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 389: 251-260 53. Madsen B, Tarsounas M, Burchell JM, Hall D, Poulsom R, Taylor- Papadimitriou J (2003) PLU-1, a transcriptional repressor and putative testis-cancer antigen, has a specific expression and localisation pattern during meiosis. Chromosoma 112: 124-132 54. Mencarelli MA, Kleefstra T, Katzaki E, Papa FT, Cohen M, Pfundt R, Ariani F, Meloni I, Mari F, Renieri A (2009) 14q12 Microdeletion syndrome and congenital variant of Rett syndrome. Eur J Med Genet 52: 148-152 55. Mitra D, Das PM, Huynh FC, Jones FE (2011) Jumonji/ARID1 B (JARID1B) protein promotes breast tumor cell cycle progression through epigenetic repression of microRNA let-7e. J Biol Chem 286: 40531-40535 56. Nathan D, Ingvarsdottir K, Sterner DE, Bylebyl GR, Dokmanovic M, Dorsey JA, Whelan KA, Krsmanovic M, Lane WS, Meluh PB, Johnson ES, Berger SL (2006) Histone sumoylation is a negative regulator in Saccharomyces cerevisiae and shows dynamic interplay with positive-acting histone modifications. Genes Dev 20: 966-976 57. Nijwening JH, Geutjes EJ, Bernards R, Beijersbergen RL (2011) The histone demethylase Jarid1b (Kdm5b) is a novel component of the Rb pathway and associates with E2f-target genes in MEFs during senescence. PLoS One 6: e25235 58. Niu X, Zhang T, Liao L, Zhou L, Lindner DJ, Zhou M, Rini B, Yan Q, Yang H (2012) The von Hippel-Lindau tumor suppressor protein regulates gene expression and tumor growth through histone demethylase JARID1C. Oncogene 31: 776-786 59. Ounap K, Puusepp-Benazzouz H, Peters M, Vaher U, Rein R, Proos A, Field M, Reimand T (2012) A novel c.2T > C mutation of the KDM5C/JARID1C gene in one large family with X-linked intellectual disability. Eur J Med Genet 55: 178- 184 60. Papa FT, Mencarelli MA, Caselli R, Katzaki E, Sampieri K, Meloni I, Ariani F, Longo I, Maggio A, Balestri P, Grosso S, Farnetani MA, Berardi R, Mari F, Renieri A (2008) A 3 Mb deletion in 14q12 causes severe mental retardation, mild facial dysmorphisms and Rett-like features. Am J Med Genet A 146A: 1994-1998
  • 32. 61. Pokholok DK, Harbison CT, Levine S, Cole M, Hannett NM, Lee TI, Bell GW, Walker K, Rolfe PA, Herbolsheimer E, Zeitlinger J, Lewitter F, Gifford DK, Young RA (2005) Genome-wide map of nucleosome acetylation and methylation in yeast. Cell 122: 517-527 62. Robertson AG, Bilenky M, Tam A, Zhao Y, Zeng T, Thiessen N, Cezard T, Fejes AP, Wederell ED, Cullum R, Euskirchen G, Krzywinski M, Birol I, Snyder M, Hoodless PA, Hirst M, Marra MA, Jones SJ (2008) Genome-wide relationship between histone H3 lysine 4 mono- and tri-methylation and transcription factor binding. Genome Res 18: 1906-1917 63. Roesch A, Becker B, Schneider-Brachert W, Hagen I, Landthaler M, Vogt T (2006) Re-expression of the retinoblastoma-binding protein 2-homolog 1 reveals tumor-suppressive functions in highly metastatic melanoma cells. J Invest Dermatol 126: 1850-1859 64. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, Basu D, Gimotty P, Vogt T, Herlyn M (2010) A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 141: 583-594 65. Roesch A, Mueller AM, Stempfl T, Moehle C, Landthaler M, Vogt T (2008) RBP2-H1/JARID1B is a transcriptional regulator with a tumor suppressive potential in melanoma cells. Int J Cancer 122: 1047-1057 66. Santos-Reboucas CB, Fintelman-Rodrigues N, Jensen LR, Kuss AW, Ribeiro MG, Campos M, Jr., Santos JM, Pimentel MM (2011) A novel nonsense mutation in KDM5C/JARID1C gene causing intellectual disability, short stature and speech delay. Neurosci Lett 498: 67-71 67. Santos-Rosa H, Schneider R, Bannister AJ, Sherriff J, Bernstein BE, Emre NC, Schreiber SL, Mellor J, Kouzarides T (2002) Active genes are tri-methylated at K4 of histone H3. Nature 419: 407-411 68. Schmitz SU, Albert M, Malatesta M, Morey L, Johansen JV, Bak M, Tommerup N, Abarrategui I, Helin K (2011) Jarid1b targets genes regulating development and is involved in neural differentiation. EMBO J 30: 4586-4600 69. Schubeler D, MacAlpine DM, Scalzo D, Wirbelauer C, Kooperberg C, van Leeuwen F, Gottschling DE, O'Neill LP, Turner BM, Delrow J, Bell SP, Groudine M (2004) The histone modification pattern of active genes revealed through genome-wide chromatin analysis of a higher eukaryote. Genes Dev 18: 1263- 1271
  • 33. 70. Secombe J, Eisenman RN (2007) The function and regulation of the JARID1 family of histone H3 lysine 4 demethylases: the Myc connection. Cell Cycle 6: 1324-1328 71. Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunstein M, Kurdistani SK (2005) Global histone modification patterns predict risk of prostate cancer recurrence. Nature 435: 1262-1266 72. Semenza GL (2003) Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3: 721-732 73. Seward DJ, Cubberley G, Kim S, Schonewald M, Zhang L, Tripet B, Bentley DL (2007) Demethylation of trimethylated histone H3 Lys4 in vivo by JARID1 JmjC proteins. Nat Struct Mol Biol 14: 240-242 74. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, McDermott U, Azizian N, Zou L, Fischbach MA, Wong KK, Brandstetter K, Wittner B, Ramaswamy S, Classon M, Settleman J (2010) A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141: 69-80 75. Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA (2004) Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119: 941-953 76. Shilatifard A (2006) Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu Rev Biochem 75: 243-269 77. Shogren-Knaak M, Ishii H, Sun JM, Pazin MJ, Davie JR, Peterson CL (2006) Histone H4-K16 acetylation controls chromatin structure and protein interactions. Science 311: 844-847 78. Simensen RJ, Rogers RC, Collins JS, Abidi F, Schwartz CE, Stevenson RE (2012) Short-term memory deficits in carrier females with KDM5C mutations. Genet Couns 23: 31-40 79. Sims RJ, 3rd, Nishioka K, Reinberg D (2003) Histone lysine methylation: a signature for chromatin function. Trends Genet 19: 629-639 80. Song SH, Han SW, Bang YJ (2011) Epigenetic-based therapies in cancer: progress to date. Drugs 71: 2391-2403 81. Stein AB, Jones TA, Herron TJ, Patel SR, Day SM, Noujaim SF, Milstein ML, Klos M, Furspan PB, Jalife J, Dressler GR (2011) Loss of H3K4 methylation destabilizes gene expression patterns and physiological functions in adult murine cardiomyocytes. J Clin Invest 121: 2641-2650
  • 34. 82. Tahiliani M, Mei P, Fang R, Leonor T, Rutenberg M, Shimizu F, Li J, Rao A, Shi Y (2007) The histone H3K4 demethylase SMCX links REST target genes to X- linked mental retardation. Nature 447: 601-605 83. Tan K, Shaw AL, Madsen B, Jensen K, Taylor-Papadimitriou J, Freemont PS (2003) Human PLU-1 Has transcriptional repression properties and interacts with the developmental transcription factors BF-1 and PAX9. J Biol Chem 278: 20507-20513 84. Tsukada Y, Fang J, Erdjument-Bromage H, Warren ME, Borchers CH, Tempst P, Zhang Y (2006) Histone demethylation by a family of JmjC domain- containing proteins. Nature 439: 811-816 85. Tzschach A, Lenzner S, Moser B, Reinhardt R, Chelly J, Fryns JP, Kleefstra T, Raynaud M, Turner G, Ropers HH, Kuss A, Jensen LR (2006) Novel JARID1C/SMCX mutations in patients with X-linked mental retardation. Hum Mutat 27: 389 86. Urdinguio RG, Lopez-Serra L, Lopez-Nieva P, Alaminos M, Diaz-Uriarte R, Fernandez AF, Esteller M (2008) Mecp2-null mice provide new neuronal targets for Rett syndrome. PLoS One 3: e3669 87. Vermeulen M, Mulder KW, Denissov S, Pijnappel WW, van Schaik FM, Varier RA, Baltissen MP, Stunnenberg HG, Mann M, Timmers HT (2007) Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4. Cell 131: 58-69 88. Volkel P, Angrand PO (2007) The control of histone lysine methylation in epigenetic regulation. Biochimie 89: 1-20 89. Wang P, Lin C, Smith ER, Guo H, Sanderson BW, Wu M, Gogol M, Alexander T, Seidel C, Wiedemann LM, Ge K, Krumlauf R, Shilatifard A (2009a) Global analysis of H3K4 methylation defines MLL family member targets and points to a role for MLL1-mediated H3K4 methylation in the regulation of transcriptional initiation by RNA polymerase II. Mol Cell Biol 29: 6074-6085 90. Wang S, Fisher K, Poulin GB (2011) Lineage specific trimethylation of H3 on lysine 4 during C. elegans early embryogenesis. Dev Biol 355: 227-238 91. Wang Z, Zang C, Cui K, Schones DE, Barski A, Peng W, Zhao K (2009b) Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138: 1019-1031
  • 35. 92. Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S, Chen Z, Spooner E, Li E, Zhang G, Colaiacovo M, Shi Y (2006) Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell 125: 467-481 93. Workman JL, Kingston RE (1998) Alteration of nucleosome structure as a mechanism of transcriptional regulation. Annu Rev Biochem 67: 545-579 94. Wright DE, Wang CY, Kao CF (2011) Flickin' the ubiquitin switch: the role of H2B ubiquitylation in development. Epigenetics 6: 1165-1175 95. Wu J, Ellison J, Salido E, Yen P, Mohandas T, Shapiro LJ (1994a) Isolation and characterization of XE169, a novel human gene that escapes X-inactivation. Hum Mol Genet 3: 153-160 96. Wu J, Salido EC, Yen PH, Mohandas TK, Heng HH, Tsui LC, Park J, Chapman VM, Shapiro LJ (1994b) The murine Xe169 gene escapes X-inactivation like its human homologue. Nat Genet 7: 491-496 97. Wu X, Gong Y, Yue J, Qiang B, Yuan J, Peng X (2008) Cooperation between EZH2, NSPc1-mediated histone H2A ubiquitination and Dnmt1 in HOX gene silencing. Nucleic Acids Res 36: 3590-3599 98. Xia X, Lemieux ME, Li W, Carroll JS, Brown M, Liu XS, Kung AL (2009) Integrative analysis of HIF binding and transactivation reveals its role in maintaining histone methylation homeostasis. Proc Natl Acad Sci U S A 106: 4260-4265 99. Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, Ma Y, Yu Y, Lin H, Chen AP, Chen CD (2007) JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. Proc Natl Acad Sci U S A 104: 19226-19231 100. Xie L, Pelz C, Wang W, Bashar A, Varlamova O, Shadle S, Impey S (2011) KDM5B regulates embryonic stem cell self-renewal and represses cryptic intragenic transcription. EMBO J 30: 1473-1484 101. Yamane K, Tateishi K, Klose RJ, Fang J, Fabrizio LA, Erdjument- Bromage H, Taylor-Papadimitriou J, Tempst P, Zhang Y (2007) PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation. Mol Cell 25: 801-812 102. Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y (2006) JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell 125: 483-495
  • 36. 103. Yoshida M, Ishimura A, Terashima M, Enkhbaatar Z, Nozaki N, Satou K, Suzuki T (2011) PLU1 histone demethylase decreases the expression of KAT5 and enhances the invasive activity of the cells. Biochem J 437: 555-564 104. Yu V, Fisch T, Long AM, Tang J, Lee JH, Hierl M, Chen H, Yakowec P, Schwandner R, Emkey R (2012) High-throughput TR-FRET assays for identifying inhibitors of LSD1 and JMJD2C histone lysine demethylases. J Biomol Screen 17: 27-38