SlideShare a Scribd company logo
1 of 9
Download to read offline
Send Orders for Reprints to reprints@benthamscience.ae
Epigenetic Diagnosis & Therapy, 2015, 1, 5-13 5
2214-0840/15 $58.00+.00 © 2015 Bentham Science Publishers
DNA Methylation: A Possible Target for Current and Future Studies on
Cancer?
Sahar Olsadat Sajadian and Andreas K. Nussler*
University of Tübingen, BG Trauma Center, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076
Tübingen, Germany
Abstract: The field of epigenetics encompasses studies about the ways in which genes are influenced
by various external stimuli, such as drugs, chemicals, and the environment. Over the last few years, it
has become an increasingly important area of research. Recently, substantial evidence has been
presented for the claim that certain diseases, including cancer, exhibit epigenetic changes. In the
present overview, we will give a brief summary of recent observations on epigenetics and cancer as
well as of the ways in which various small molecules may influence the epigenetic state of cells.
Keywords: Cancer, CpG islands, DNA methylation, epigenetic drugs, epigenetics, TET proteins.
DEFINITION OF EPIGENETICS
Epigenetics is defined ‘as a permanent change of gene
expression during cell proliferation and development, which
does not cause a change of the gene sequences’. Epigenetic
regulation takes place in three separate stages: 1)
nucleosome positioning, 2) histone modification, and 3)
DNA methylation (Fig 1).
DNA METHYLATION
In the past years, epigenetic studies have focused on
DNA methylation in mammals. Particular interest was
directed towards genomic imprinting in the inactivation
pattern of X-chromosome, in silencing of the
retrotransposon, known as repetitive elements, and in tissue-
specific genes [1, 2], These epigenetic changes have
demonstrated playing an important role in many pathologies
and gene therapies [3-5].
During the DNA methylation process, a methyl group
(CH3) is added to the phenyl ring of the cytosine at the 5-
carbon position. Somatic cell methylation can take place at
all CpG (cytosine- phosphate-guanin) dinucleotides positions
except at the CpG island [6]. CpG islands are parts of the
DNA with more than 500 bp and over 55% of GC content
[7]. These islands accommodate approximately 60% of the
mammalian gene promoters [8]. When DNA methylation
occurs in these promoters, some crucial transcription factors
(TF) are down-regulated with subsequent inhibition of the
corresponding protein formation. However, it has been
proven that even in regions that are poor in CpGs, such as
the Oct4 promoter, DNA methylation plays an important role
in gene regulation [9, 10].
*Address correspondence to this author at the University of Tübingen, BG
Trauma Center, Siegfried Weller Institute, Schnarrenbergstraße 95, D-72076
Tübingen, Germany, Tel: +49 7071 606-1065; Fax: +49 7071 606-1978;
E-mail: andreas.nuessler@gmail.com
DNA methylation is supported by a special group of
enzymes; the so called DNA methyltransferases (DNMT).
Up to now, the following DNMTs have been discovered:
DNMT1, DNMT1b, DNMT1o, DNMT1p, DNMT2,
DNMT3A, DNMT3B, and DNMT3L.
DNMTs such as DNMT3A, DNMT3B, and DNMT3L
are apparently responsible for the de novo DNA methylation
in early developmental stages. In order to maintain the
methylation in daughter cells, the so called UHRF1 protein,
a ubiquitin-like protein with PHD and RING finger domain
1, binds to hemi-methylated DNA. During replications,
UHRF1 causes DNMT1 to methylate new cytosine from
synthesized DNA strands. There exists also growing
evidence which suggests that DNMT3A/B are also involved
in maintaining the DNA methylation pattern in somatic cells,
specifically in imprinted genes and repeated elements [10].
The significance of DNMTs was demonstrated in various
animal studies, proving that mice lacking DNMTs died
during their early development or shortly after birth [11].
Nevertheless, the expression of these enzymes differs
between certain types of cancer and it is not always
associated with hyper- or hypomethylation, as had been
expected. Sometimes, the expression of DNMTs is regulated
by miRNAs, which may explain the differences in the
expression of DNMTs in various tumors [12]. For instance,
it has been demonstrated in several studies that DNA
demethylase is post-transcriptionally regulated by mir29 and
mir148 [13]. Furthermore, So et al. have claimed that DNA
methyltransferase controls stem cell aging through
microRNA. They have shown that DNMT inhibition leads to
a decrease of the expression of Polycomb groups (PcG) and
to an increase of the expression of microRNAs (miRNAs)
that target PcG [14].
DNA DEMETHYLATION
In early embryogenesis, many researchers have observed
a loss of DNA methylation and subsequently have postulated
the existence of several DNA demethylases and various
6 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler
mechanisms of the DNA demethylation [15, 16]. One
potential DNA demethylation pathway is enabled by the ten-
eleven-translocation proteins (TETs), which are named after
the ten-eleven-translocation (q22, q23) (10, 11) that occurs
in rare cases of acute myeloid and lymphocytic leukemia
[17, 18]. 5-mC is oxidized by TET proteins and thereby
converted to 5-hmC, 5-fC, and 5-CaC, which all belong to
the family of Fe (II)/2-oxygluttarate-dependent dioxgenases
[19]. DNA methylation is reversible either through passive
demethylation via DNA replication or by active
demethylation via DNA repair followed by base excision
repair of modified nucleotides or DNA glycosidase [15, 17]
(Fig. 2).
TET proteins play different roles in diverse biological
processes, including epigenetic regulation of gene
transcription, embryonic development, stem cell functions,
and cancer. They mediate the DNA demethylation process
and change the epigenetic state of DNA. A loss of 5-hmC,
together with a down-regulation or mutation of TET
proteins, has been reported in numerous types of cancer, like
melanoma, breast, prostate, myeloid, brain, and liver cancer
[20]. It has been demonstrated that in patients suffering from
acute myeloid leukemia (AML), TET1 protein influences
mixed lineage leukemia (MLL) development [16].
Furthermore, several changes in TET2 proteins, like e.g.
deletion and deactivation of TET2, have been identified in
30% of all patients suffering from different kinds of myeloid
malignancies. Several work groups have reported an
alteration of a TET2 gene in a variety of myeloid
malignancies and the majority of the TET2 alterations
include single or double copy defects and nonsense/frame
shift mutation suggesting that TET2 lesions cause a loss of
function. Therefore, it has been suggested that TET2 is a
possible tumor suppressor gene that is responsible for
myeloid malignancies [21]. Furthermore, the loss of 5-hmC
in correlation with clinical progression and relapse of
melanoma has been reported [20].
DNA METHYLATION AND CANCER
Numerous studies have reported that tumor cells exhibit a
major disruption in the regulation of the DNA methylation
pattern in cancer. Therefore, over the last few years, studies
have particularly focused on changes in DNA methylation.
[22]. Earlier studies have demonstrated that the
hypomethylation of interspersed repetitive DNA is a
common feature of numerous types of cancer that causes
genomic instability and aberrant transcription initiation.
However, researchers have proven that the hypermethylation
of focal CpG islands is elevated in cancer and may result in
Fig. (1). Epigenetic machinery and interplay between epigenetic factors (modified from [91]). Epigenetic regulation depends on the three
following epigenetic factors: (a) DNA methylation, (b) histone modification, and (c) nucleosome positioning. The interaction between these
factors results in the final outcome, but disturbances of this balance lead to several diseases. Abbreviations: DNMT: DNA Methyl
Transferase, HAT: Histone Acetylation Transferase, HDAC: Histone Deacetylase, HMT: Histone Methyltransferase, HDM: Histone
Demethyl Transferase.
MethylationMethylation DemethylationDemethylation
DNMT1 TETs:
Me
DNA
Methylation
(maintenance DNMT)
DNMT3A & DNMT3B
(de novo DNMT)
TET1, TET2,
TET3
5hmC
Epigenetic
machinery
Epigenetic
machinery
AcetylationAcetylation
HAT & HDAC
Histone
modifications
Histone
modifications
Chromatin
remodeling
Chromatin
remodeling
MethylationMethylation
HAT & HDAC
HMT & HDM
Disfunc
Phosphor-
tylation
Phosphor-
tylation
Kinase & Phosphor
SWI/SNF, ISWI
MI-2, INO80
Disease
ctioning
Ac
Me
Me
P
Disease
• Cancer
• Autoimmune disorder
• Neural disorder
• Aging
Ub
DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 7
inhibition of various miRNAs, tumor suppressor and
functional genes [23, 24].
Moreover, it has become evident that different pathways
target DNMT at the specific CpG Island. Moreover, four
different mechanisms target DNA methyltransferase. Firstly,
DNMT3A and DNMT3L recognize DNA or chromatin with
specific domains and they interact with the LYS4 of histone
3 (H3K4). Secondly, DNMT interacts with different
chromatin modifiers such as KMT and HDACs. Thirdly, the
Piwi-PiRNA complex, a class of ncRNAs, guides DNMTs to
transposon elements. Finally, the direct interaction of
promoter-associated RNA (pRNA) with the promoter of the
rRNA gene might recruit DNMT3b and enable methylation
[25]. In addition, it has been shown that, in an alteration
between methylation and phosphorylation, an adjacent lysine
and serine determine the stability of DNMT1 thus preserving
the methylation pattern during cell division [26].
However, the detailed mechanism(s) leading to hyper- or
hypomethylation shall be decorticated in the future.
HYPERMETHYLATION IN CANCER
The CpG islands are protected by various defense
mechanisms by accessing the DNA methyltransferase, such
as DNA replication timing, active demethylation, or active
transcription. Most genes that are susceptible to
hypermethylation are associated with the regulation of DNA
repair, apoptosis, cell cycle, drug resistance, metastasis, and
differentiation [10]. Many tumors are hypermethylated in
one or more genes. For instance, in lung cancer, it has been
demonstrated that alterations of DNA methylation have been
found in more than 40 genes [27].
Regarding breast cancer, it has been shown that several
critical genes, which seem to be important in tumorigenesis,
are hypermethylated. Among those genes, cell adhesion
genes, inhibitors of matrix metalloproteinases, as well as the
ER and PR steroid receptor genes can be found and have
been linked to breast cancer development [28].
It has also been reported that DNA methylation is one of
the reasons for the inactivation of BRCA1 in breast cancer,
which is one of the genes most commonly associated with
breast cancer [29]. Moreover, the hypermethylation of other
cell cycle genes may result in acute lymphoblastic leukemia
which is an undesirable clinical outcome [30].
HYPOMETHYLATION IN MALIGNANCIES
Hypomethylation is an alteration in DNA methylation
which is frequently seen in solid tumors [31-33], and is a
sign of a progressive malignancy [34]. Furthermore, it has
been reported that the hypomethylation status contributes to
oncogenesis through up-regulation of some oncogenes, like
H-RAs and c-MYC [31], as well as by chromosome
instability [35, 36].
During cancer development, aberrations not only occur in
the DNA methylation, but also in the regulation of histone
modifications [37]. For instance, numerous studies have
demonstrated that histone deacytylases (HDACs) are
overexpressed in different types of cancer. This
overexpression leads to a deacetylation of the Transcription
Starting Site (TSS) of important genes and subsequently to
the assembly of silenced genes into more compact structures
[38, 39].
Fig. (2). Active demethylation pathway through the conversion of 5mC to 5hmC by recruiting TET proteins. Following 5mC oxidation,
demethylation occurs by dilution of oxidized base or by removal through DNA glycosidase- and DNA repair-mediated excision of modified
nucleotides (modified from [15]). Abbreviations: C: Cytosine, 5mC: 5-methylcytosine, 5hmC: 5-hydroxymethylcytosine; 5fC: 5-
formylcytosine, 5caC: 5-carboxylcytosine, TDG: Thymine DNA Glucosidase, BER: Base Excision Repair.
8 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler
In summary, there exists an epigenetic crosstalk between
histone modification, micro-RNAs, and DNA methylation.
Post-transcriptionally, miR-29 and miR-148 regulate the
DNA demethylases [40, 41]. Moreover, it has been proven
that DNMTs interact with HDAC [28]. In particular, the
combination of DNMT and HDAC inhibitors is a primary
target for potential therapeutics against cancer [42, 43],
because these inhibitors are capable of targeting the
epigenetic status of DNA and histones simultaneously,
which may enable the development of more effective
anticancer drugs.
EPIGENETICS AND CANCER STEM CELL
HYPOTHESIS
Recently, the concept has arisen that cancer stem cells
are to be classified as a subpopulation of cancer cells with a
different tumorigenic potential. This side population, which
is usually named cancer-initiating cells or cancer stem cells
(CSC), has a characteristic potential for self-renewal that
may sustain and perpetuate tumors and is known to dispose
of a higher drug resistance. Firstly, it has been observed that
CD34+CD38- leukemic stem cells disposing of a
differentiation and proliferation capacity are able to trigger
human leukemia in NOD/SCID mice [44]. Furthermore, it
has been proven that a landscape of solid tumors, such as
glioblastoma [45], breast cancer [46], prostate cancer [47],
and hepatocellular carcinoma [48], contain CSCs. A growing
number of studies prove that the cancer-initiating cells are
mostly resistant to chemotherapy or radiotherapy, which
results in the recurrence of tumors [49], and in the formation
of metastases [50].
Studies on the epigenetic modifications in stem cells and
in cancer cells have directed interest to stem/progenitor cells,
which are at the origin of cancer. It is believed that the
genetic and epigenetic changes in these cells provide the
ability for their survival, protecting them from toxic
environment, which then contributes to tumor initiation [51].
Interestingly, the Polycomb repressive complex, which
causes abnormal gene silencing in cancer, represses CpG
island genes in Emberyonic Stem Cells (ESCs) in the same
way as in cancer [52, 53]. Following this abnormal
hypermethylation due to DNA damage, inflammation or
stress caused by ROS, tumor suppressor genes become
silenced. This may lead to the activation of stem cell
pathways and provide the ability of self-renewal for cancer
cells [54]. For example, in stem cells, like e.g. the CD133+
cells, promoter hypermethylation of some tumor suppressor
genes, such as SFRP1 and SOX17, was identified [55],
proving the stem cell origin of epigenetic abnormalities in
cancer [56].
APPLICATION OF EPIGENETIC DRUGS
As was described above, the application of epigenetic
drugs is being increasingly focused on in the field of
biomedicine and drug development research. Like numerous
other diseases, cancer causes aberrancies in the epigenetic
regulation. Most importantly, the pattern of DNA
demethylation and histone modification has been related to
tumor genesis and metastasis [10, 57]. Therefore, it is
believed that these two biomechanical processes have a high
potential for being used in clinical applications, such as e.g.
in cancer detection, tumor prognosis, and
pharmacoepigenetics (prediction of treatment response). For
example, hypermethylation of glutathione transferase
(GSTP1) constitutes a promising epigenetic marker in
prostate cancer [58]. It has to be pointed out that 80-90% of
men suffering from prostate cancer exhibit a
hypermethylation of GSTP1 and that this hypermethylation
does not occur in benign hyperplastic prostate tissue [59].
Therefore, GSTP1 methylation is a helpful prognostic
marker, which enables the discrimination of benign tumor
tissue in prostate cancer. As a matter of fact, GSTP1 is also
hypermethylated in other cancers such as in kindney, liver
and breast cancer [60]. Interestingly, the screening of single
markers through global DNA methylation offers great
clinical potential, as for example, it allows for the prediction
of metastases in colorectal cancer, recurrence in lung cancer,
and progression in virus-associated neoplasms [57, 61]. In
addition to DNA methylation markers, histone-based
markers have been suggested as epigenetic biomarkers. In
particular, aberrant histone modifications have been linked to
recurring prostate cancer [62].
Nowadays, most studies focus on the development of
epigenetic drugs, which can reverse the abnormal epigenetic
state to the normal epigenetic state in cancer cells. Up to
now, the US Food and Drug Administration (FDA) has
approved four epigenetic small molecule drugs for the
treatment of cancer: 5-Azacytidine (5-Aza) and 5-Aza-2´-
deoxycytidine (DNMT inhibitors) which have been used for
patients suffering from myelodyplastic syndrome and from
acute leukemia, as well as Varinostat and Romidepsin
(HDAC inhibitors) which are used for treatment of T-cell
lymphoma and some hematological tumors [63]. It has to be
pointed out that each DNMT inhibitor possesses its own way
of action on the gene transcription.
Although 5-Aza and 5-Aza-2´-deoxycytidine have the
same therapeutic effect, their mode of action in tumor cells is
different. It has been demonstrated that 5-Aza-2´-
deoxycytidine is able to induce senescence in tumor cells,
while 5-Aza induces cell death in tumor cells in vitro and in
vivo. These two DNMT inhibitors differ from each other on
the molecular and the cellular level: For instance, 5-Aza can
be integrated in both DNA and RNA (20-40% and 60-80%
respectively), while 5-Aza-2´-deoxycytidine can only be
incorporated in DNA. Moreover, 5-Aza-2´-deoxycytidine
induces more DNA damage and micronuclei formation than
5-Aza [64].
Furthermore, it has been rported that Azacytidine
increases the antitumor effect sensitivity of Docetaxel (DTX)
and Cisplation in aggressive and chemoresistant prostate
cancer [65]. In addition, it has been shown that Pyrazofurin
(PF), an inhibitor of orotidylate decarboxylase improved the
anti-tumoral activity of 5-Aza in human leukemia cells [66].
The combination of PF and 5-Aza may provide a useful
treatment for patients who do not respond to standard anti-
leukemic therapies [67]. Furthermore, it has been proven that
5-Aza-2´-deoxycytidine inhibits telomerase activity and
reactivates p16 and c-Myc in hepatocellular carcinoma cells
(HCC), such as the MMC-7721 and the HepG2 cell line [68].
Other DNMT inhibitors, such as 5-fuoro-2´-
deoxycytidine, in combination with other drugs are currently
DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 9
undergoing clinical trials regarding the treatment of different
types of cancers [69]. The DNA methyltransferase inhibitor
Zebularin provoked the demethylation of tumor suppressor
genes by inducing apoptosis, cell cycle regulation in HCC
cell lines (Huh7, KMCH, and HepG2), and in vivo inhibition
of tumor growth in a xenograft model [70].
It has been discussed if the expression of specific
DNMTs might influence the differentiation status in
Fig. (3). Epigenetic drugs for cancer therapy. It has been described that scores of small molecules participate in the fight against cancer cells
by inducing changes in the epigenetic machineries. This figure illustrates some of the most important epigenetic drugs, which are classified
in relation to their epigenetic target. Abbreviations: DNMT1: DNA Methyltransferase 1, HDAC: Histone Deacetylase, PcG: Polycomb
Group, Me: Methylated, HAT: Histone Acetylase.
Repressive
complex
Repressive
complex
DNMT1DNMT1
PcG
SirtuinsSirtuins
HDACHDAC
PcG
MeDNMT
Inhibitors:
Vidaza
Decitabine
HMT Inhibitors
DZNep
BIX-01294
Me
Decitabine
Zelbularine
Procainamide
HDAC Inhibitors:HDAC Inhibitors: SIRTSIRT
BIX 01294
Chaetocin
DNMT1DNMT1
HDACHDAC
Me
VPA
TSA
Vorinostat
Panobinostat
Belinostat
VPA
TSA
Vorinostat
Panobinostat
Belinostat
Inhibitors
Sirtinol
Cambinol
Salermide
Inhibitors
Sirtinol
Cambinol
Salermide
PcG
SirtuinsSirtuins
Transcription
HATHAT
TETsTETs
factors/
coactivator
Pol IIPol II
5-methylcytosine5 methylcytosine
5-hydroxymethylcytosine
5-formylcytosine
5-carboxylcytosine
Histone acetylated
10 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler
developing and dedifferentiating cells. It has been shown
that 5-Aza triggers differentiation in mesenchymal stem cells
(MSCs) [8, 71, 72]. Furthermore, we have reported that
pretreatment of fat-derived old MSCs with 5-Aza improves
osteogenic differentiation by DNA demethylation. We have
observed a reduction of 5hmC in Ad-MSCs from older
donors (> 60 years) compared to young donors (< 45 years).
After stimulation of cells with 5-Azacytidine the osteogenic
potential of the cells improved. It has been observed that the
increase of AP activity and matrix mineralization is
associated with an inducement of the expression of 5hmC as
well as with the expression of TET2 and TET3 [8].
Therefore, we have concluded that 5-Azacytidine induces the
rejuvenating process of older Ad-MSCs (Fig. 4).
Furthermore, it has been demonstrated in various studies that
the treatment of human Adipose Stem Cells (ADSC) and
human bone marrow MSCs with 20 µM 5-Azacytidine
improves their hepatic differentiation [73-75]. In the same
line of evidence, it has been shown that the incubation of
human bone marrow MSCs with 1 µM TSA which had been
pretreated with hepatogenic-stimulating agents, improves the
hepatogenic differentiation of these cells [76]. Moreover, a
combination of TSA and 5-Aza-dc has improved the
differentiation of human CD34+ to CD34/ CD90 cells [77].
Regarding the influence of the epigenetic status on the
reprogramming mechanism of the cells, it has been reported
that vitamin C induces the active demethylation pathway
through the induction TET enzyme activity in mouse
embryonic stem cells (ES) [78]. In the same line of evidence,
it has been demonstrated that TET2 facilitates the
differentiation of hematopoietic stem cells [79]. Therefore,
vitamin C can be used for reprogramming stem cells, as it
induces TET-dependent demethylation.
OTHER EPIGENETIC MODIFYING DRUGS
In the past years, many anticancer drugs targeting the
acetylation or methylation status of histones through
different mechanisms have been developed, such as HDAC
inhibitors class I, II, III (sirtuins), and IV, HATs, HMTs,
HDMs, and various kinds of kinases. Based on their
chemical properties, the HDACs are branched into the four
Fig. (4). Reprogrammation and differentiation of cells through a DNA methylation pattern. Changes in the methylation status of cells may
lead to differentiation either through passive demethylation [8, 72] by inhibition of DNMT or through active demethylation by inducing TET
proteins to produce 5hmC [78, 79]. Abbreviations: 5mC: 5-methylcytosine 5hmC: 5-hydroxymethylcytosine; 5fC: 5-formylcytosine; 5caC:
5-carboxylcytosine; TDG: Thymine DNA Glucosidase; BER: Base Excision Repair, TET: Ten eleven translocation, DNMT: DNA
Methyltransferase, SAH: S-adenosylhomocysteine, Hcy: Homocysteine SAM: S-Adenosylmethionin.
Progenitor /
stem Cells
Differentiated Cells
DNMT Inhibitors:
5-AZA
stem Cells
MethionineSAH5mC
5caC
C
Pol II
Progenitor
Cells
HcySAM
5hm
C
5fC
5caC
Pol IICells
Differentiated
Cells
DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 11
following groups: hydroxamic acid, short fatty acids, cyclic
peptide and benzamide enzymes [57]. HDACs inhibitors
mainly arrest the cell cycle in G1 and G2-M phases, which,
in turn, induces apoptosis and cell differentiation. However,
they are able to inhibit angiogenesis and metastasis, and to
decrease the resistance of tumor cells to chemotherapy [80].
Recently, a new epigenetic drug called Suberoylanilide
hydroxamic acid (SAHA) has been approved for treating T-
cell cutaneous lymphoma by the Federal Drug
Administration (FDA) [81]. Furthermore, a large number of
HDAC inhibitors, like e.g. TSA (Tricostatin A), PXD101
(Belinostat), and LBH589 (Panobinostat), are currently being
developed and are undergoing clinical trials [82, 83].
Class III HDAC inhibitors, so called Sirtuins (SIRT), are
important for the regulation of senescence, survival, and cell
proliferation. An increasing number of effective SIRT
inhibitors have recently been generated [57]. SIRT1 and
SIRT2 inhibitors are likely to stop p53 activity, thereby
inducing growth arrest in cancer cells and increasing
sensitivity of tumor cells towards anticancer drugs [84].
Salermide - another SIRT1 and SIRT2 inhibitor - induces p-
53-independent apoptosis in tumor cells and reactivates
proapoptotic genes by deacytelation of H4K16Ac at the
epigenetic level which is mediated by SIRT1 [85].
It has been shown that curcumin, garcinol, and anacaridic
acid may act as HAT inhibitors with an anti-cancer effect in
ovarian and colon cancer. It has been demonstrated that all
three substances are able to inhibit EP300-mediated p53
acetylation [86, 87].
DZNep, Chaetocin, and BIX-01294 are known to be
lysine HMT inhibitors. It has been shown that DZNep
promotes apoptosis in the MCF7 cell line (breast cancer) and
in the HCT116 cell line (colorectal cancer) [88]. Chaetocine
has an anti-cancer effect on multiple myeloma (MM) [89].
BIX-01294 deactivates the histone H3 lysine 9 (H3K9)
methyltransferases G9a and G9a-like protein by promoting
the apoptotic cell death [90].
It has been demonstrated that BIX-01294 induces
endothelial differentiation in Ad-MSCs. We have been able
to demonstrate that BIX modifies DNA and histone
methylation and remarkably increases the expression of
various endothelial markers that are required for blood vessel
formation, such as VCAM-1, PECAM, VEGFR-2, and
PDGF [72]. From our studies, we conclude that drugs that
modify the DNA or histone methylation also induce changes
in the pluripotency [8, 72].
SUMMARY AND FUTURE PERSPECTIVES
In the present overview, we have presented numerous
encouraging findings demonstrating that so called small
molecules exert anti-tumoral activity through epigenetic
changes within target cells. Interestingly, even molecules,
like e.g. polyphenols, which are considered to be anti-
oxidative drugs, dispose of a potent epigenetic modifying
activity, and therefore slow down tumor growth. However,
the large variety among the structures of different molecules
that exert epigenetic modifying activity bears a high risk.
The mechanisms by which epigenetically modifying drugs
reduce growth or damage tumor cells are not very specific
and may therefore damage normal/healthy cells as well. In
this avenue, it is mandatory to develop new and more
specific small molecules against specific enzymes of the
epigenetic machinery in order to reduce damage on healthy
tissue.
CONFLICT OF INTEREST
The authors declare that they do not have any conflict of
interest.
ACKNOWLEDGEMENTS
We like to thank Dr. Luc Koster for editing the
manuscript. The authors would also like to thank the
Ministry of Rural Affairs, and Consumer Protection Baden-
Württemberg for supporting our study and hence promoting
important research projects.
REFERENCES
[1] Laird PW. The power and the promise of DNA methylation
markers. Nat Rev Cancer 2003; 3(4): 253-66.
[2] Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U,
Zoghbi HY. Rett syndrome is caused by mutations in X-linked
MECP2, encoding methyl-CpG-binding protein 2. Nat Gen 1999;
23(2): 185-8.
[3] Jones PA, Baylin SB. The fundamental role of epigenetic events in
cancer. Nat. Rev Gen 2002; 3(6): 415-28.
[4] Baylin SB. Tying it all together: epigenetics, genetics, cell cycle,
and cancer. Science 1997; 277(5334): 1948-9.
[5] Costello JF, Plass C. Methylation matters. J Med Gen 2001; 38(5):
285-303.
[6] Suzuki MM, Bird A. DNA methylation landscapes: provocative
insights from epigenomics. Nat Rev Gen 2008; 9(6): 465-76.
[7] Takai D, Jones PA. Comprehensive analysis of CpG islands in
human chromosomes 21 and 22. Proc Nat Acad Sci 2002; 99(6):
3740-5.
[8] Yan X, Ehnert S, Culmes M, et al. 5-Azacytidine improves the
osteogenic differentiation potential of aged human adipose-derived
mesenchymal stem cells by DNA demethylation. PloS one 2014;
9(3): e90846.
[9] Irizarry RA, Ladd-Acosta C, Wen B, et al. The human colon cancer
methylome shows similar hypo-and hypermethylation at conserved
tissue-specific CpG island shores. Nat Gen 2009; 41(2): 178-86.
[10] Das PM, Singal R. DNA methylation and cancer. J Clin Oncol
2004; 22(22): 4632-42.
[11] Robertson KD. DNA methylation and chromatin-unraveling the
tangled web. Oncogene 2002; 21(35): 5361-79.
[12] Blair LP, Yan Q. Epigenetic mechanisms in commonly occurring
cancers. DNA Cell Biol 2012; 31(S1): S-49-S-61.
[13] Pan W, Zhu S, Yuan M, et al. MicroRNA-21 and microRNA-148a
contribute to DNA hypomethylation in lupus CD4+ T cells by
directly and indirectly targeting DNA methyltransferase 1. J
Immunol 2010; 184(12): 6773-81.
[14] So A-Y, Jung J-W, Lee S, Kim H-S, Kang K-S. DNA
methyltransferase controls stem cell aging by regulating BMI1 and
EZH2 through microRNAs. PLoS One 2011; 6(5): e19503.
[15] Kohli RM, Zhang Y. TET enzymes, TDG and the dynamics of
DNA demethylation. Nature 2013; 502(7472): 472-9.
[16] Wu SC, Zhang Y. Active DNA demethylation: many roads lead to
Rome. Nat Rev Mol Cell Biol 2010; 11(9): 607-20.
[17] Tahiliani M, Koh KP, Shen Y, et al. Conversion of 5-
methylcytosine to 5-hydroxymethylcytosine in mammalian DNA
by MLL partner TET1. Science 2009; 324(5929): 930-5.
[18] Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y.
Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-
renewal and inner cell mass specification. Nature 2010; 466(7310):
1129-33.
[19] Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5-
methylcytosine to 5-formylcytosine and 5-carboxylcytosine.
Science 2011; 333(6047): 1300-3.
12 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler
[20] Lian CG, Xu Y, Ceol C, et al. Loss of 5-hydroxymethylcytosine is
an epigenetic hallmark of melanoma. Cell 2012; 150(6):1135-46.
[21] Li Z, Cai X, Cai C-L, et al. Deletion of Tet2 in mice leads to
dysregulated hematopoietic stem cells and subsequent development
of myeloid malignancies. Blood 2011; 118(17): 4509-18.
[22] Baylin SB, Herman JG. DNA hypermethylation in tumorigenesis:
epigenetics joins genetics. Trends Genet 2000; 16(4): 168-74.
[23] Esteller M. CpG island hypermethylation and tumor suppressor
genes: a booming present, a brighter future. Oncogene 2002;
21(35): 5427-40.
[24] Ehrlich M. DNA methylation in cancer: too much, but also too
little. Oncogene 2002; 21(35): 5400-13.
[25] Denis H, Fuks F. Regulation of mammalian DNA
methyltransferases: a route to new mechanisms. EMBO Rep 2011;
12(7): 647-56.
[26] Estève P-O, Chang Y, Samaranayake M, et al. A methylation and
phosphorylation switch between an adjacent lysine and serine
determines human DNMT1 stability. Nat Struc Mol Biol 2011;
18(1): 42-8.
[27] Tsou JA, Hagen JA, Carpenter CL, Laird-Offringa IA. DNA
methylation analysis: a powerful new tool for lung cancer
diagnosis. Oncogene 2002; 21(35): 5450-61.
[28] Yang X, Yan L, Davidson N. DNA methylation in breast cancer.
Endocrine-Related Cancer 2001; 8(2): 115-27.
[29] Catteau A, Morris JR, editors. BRCA1 methylation: a significant
role in tumour development? Semin Cancer Biol 2002; 12(5): 359-
371.
[30] Leone G, Teofili L, Voso MT, Lubbert M. DNA methylation and
demethylating drugs in myelodysplastic syndromes and secondary
leukemias. Haematologica 2002; 87(12): 1324-41.
[31] Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes
of some human cancers from their normal counterparts. Nature
1983; 301(5895): 89-92.
[32] Kim YI, Giuliano A, Hatch KD, et al. Global DNA
hypomethylation increases progressively in cervical dysplasia and
carcinoma. Cancer 1994; 74(3): 893-9.
[33] Lin C-H, Hsieh S-Y, Sheen I-S, et al. Genome-wide
hypomethylation in hepatocellular carcinogenesis. Cancer Res
2001; 61(10): 4238-43.
[34] Bedford MT, Van Helden PD. Hypomethylation of DNA in
pathological conditions of the human prostate. Cancer Res 1987;
47(20): 5274-6.
[35] Alves G, Tatro A, Fanning T. Differential methylation of human
LINE-1 retrotransposons in malignant cells. Gene 1996; 176(1):
39-44.
[36] Tuck-Muller C, Narayan A, Tsien F, et al. DNA hypomethylation
and unusual chromosome instability in cell lines fromICF
syndrome patients. Cytogenet Cell Genet 2000; 89(1-2): 121-8.
[37] Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism
to therapy. Cell. 2012;150(1):12-27.
[38] Halkidou K, Gaughan L, Cook S, Leung HY, Neal DE, Robson
CN. Upregulation and nuclear recruitment of HDAC1 in hormone
refractory prostate cancer. Prostate 2004; 59(2): 177-89.
[39] Song J, Noh JH, Lee JH, et al. Increased expression of histone
deacetylase 2 is found in human gastric cancer. APMIS 2005;
113(4): 264-8.
[40] Fabbri M, Garzon R, Cimmino A, et al. MicroRNA-29 family
reverts aberrant methylation in lung cancer by targeting DNA
methyltransferases 3A and 3B. Proc Nat Acad Sci 2007; 104(40):
15805-10.
[41] Duursma AM, Kedde M, Schrier M, Le Sage C, Agami R. miR-148
targets human DNMT3b protein coding region. RNA 2008; 14(5):
872-7.
[42] Zhu W-G, Otterson GA. The interaction of histone deacetylase
inhibitors and DNA methyltransferase inhibitors in the treatment of
human cancer cells. Curr Med Chem Anticancer Agents 2003; 3(3):
187-99.
[43] Unoki M. Current and potential anticancer drugs targeting
members of the UHRF1 complex including epigenetic modifiers.
Recent Pat Anticancer Drug Discov 2011; 6(1): 116-30.
[44] Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute
myeloid leukaemia after transplantation into SCID mice. 1994;
367(6464):645-8.
[45] Singh SK, Hawkins C, Clarke ID, et al. Identification of human
brain tumour initiating cells. Nature. 2004; 432(7015): 396-401.
[46] Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke
MF. Prospective identification of tumorigenic breast cancer cells.
Proceedings of the National Academy of Sciences. 2003; 100(7):
3983-8.
[47] Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ.
Prospective identification of tumorigenic prostate cancer stem cells.
Cancer research. 2005; 65(23): 10946-51.
[48] Suetsugu A, Nagaki M, Aoki H, Motohashi T, Kunisada T,
Moriwaki H. Characterization of CD133+
hepatocellular carcinoma
cells as cancer stem/progenitor cells. Biochem Biophys Res
Commun 2006; 351(4): 820-4.
[49] Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma
progenitors, stem cell properties, and drug resistance. Cancer Res
2008; 68(1): 190-7.
[50] Bao S, Wu Q, McLendon RE, et al. Glioma stem cells promote
radioresistance by preferential activation of the DNA damage
response. Nature 2006; 444(7120): 756-60.
[51] Ohm JE, Baylin SB. Stem cell chromatin patterns--an instructive
mechanism for DNA hypermethylation? Cell cycle (Georgetown,
Tex) 2007; 6(9): 1040.
[52] Schlesinger Y, Straussman R, Keshet I, et al. Polycomb-mediated
methylation on Lys27 of histone H3 pre-marks genes for de novo
methylation in cancer. Nature Gen 2006; 39(2): 232-6.
[53] Widschwendter M, Fiegl H, Egle D, et al. Epigenetic stem cell
signature in cancer. Nature Gen 2006; 39(2): 157-8.
[54] Baylin SB, Ohm JE. Epigenetic gene silencing in cancer-a
mechanism for early oncogenic pathway addiction? Nat Rev
Cancer 2006; 6(2): 107-16.
[55] Zhang W, Glöckner SC, Guo M, et al. Epigenetic inactivation of
the canonical Wnt antagonist SRY-box containing gene 17 in
colorectal cancer. Cancer Res 2008; 68(8): 2764-72.
[56] Tsai H-C, Baylin SB. Cancer epigenetics: linking basic biology to
clinical medicine. Cell Res 2011; 21(3): 502-17.
[57] Rodríguez-Paredes M, Esteller M. Cancer epigenetics reaches
mainstream oncology. Nat Med 2011; 330-9.
[58] Henrique R, Jerónimo C. Molecular Detection of Prostate Cancer:
A Role for< i> GSTP1</i> Hypermethylation. European urology.
2004; 46(5):660-9.
[59] Lee W-H, Morton RA, Epstein JI, et al. Cytidine methylation of
regulatory sequences near the pi-class glutathione S-transferase
gene accompanies human prostatic carcinogenesis. Proc Nat Acad
Sci 1994; 91(24): 11733-7.
[60] Esteller M, Corn PG, Urena JM, Gabrielson E, Baylin SB, Herman
JG. Inactivation of glutathione S-transferase P1 gene by promoter
hypermethylation in human neoplasia. Cancer Res 1998; 58(20):
4515-8.
[61] Esteller M. Epigenetics in cancer. New Engl J Med 2008; 358(11):
1148-59.
[62] Seligson DB, Horvath S, Shi T, et al. Global histone modification
patterns predict risk of prostate cancer recurrence. Nature 2005;
435(7046):1262-6.
[63] Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and
pharmacodynamic study of depsipeptide (FK228) in chronic
lymphocytic leukemia and acute myeloid leukemia. Blood 2005;
105(3): 959-67.
[64] Venturelli S, Berger A, Weiland T, et al. Differential induction of
apoptosis and senescence by the DNA methyltransferase inhibitors
5-azacytidine and 5-aza-2′-deoxycytidine in solid tumor cells. Mol.
Cancer Ther 2013; 12(10): 2226-36.
[65] Festuccia C, Gravina GL, D'Alessandro AM, et al. Azacitidine
improves antitumor effects of docetaxel and cisplatin in aggressive
prostate cancer models. Endocr Relat Cancer. 2009; 16(2): 401-13.
[66] Cadman E, Eiferman F, Heimer R, Davis L. Pyrazofurin
enhancement of 5-azacytidine antitumor activity in L5178Y and
human leukemia cells. Cancer Res 1978; 38(12): 4610-7.
[67] Lancet JE, Karp JE. Novel postremission strategies in adults with
acute myeloid leukemia. Curr Opin Hematol 2009; 16(2): 105.
[68] Tao S-F, Zhang C-S, Guo X-L, et al. Anti-tumor effect of 5-aza-2'-
deoxycytidine by inhibiting telomerase activity in hepatocellular
carcinoma cells. World J Gastroenterol 2012; 18(19): 2334.
[69] Yang X, Lay F, Han H, Jones PA. Targeting DNA methylation for
epigenetic therapy. Trends Pharmacol Sci 2010; 31(11): 536-46.
[70] Nakamura K, Aizawa K, Nakabayashi K, et al. DNA
methyltransferase inhibitor zebularine inhibits human hepatic
carcinoma cells proliferation and induces apoptosis. PloS One
2013; 8(1): e54036.
DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 13
[71] Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat
bone marrow mesenchymal stem cells exposed to 5-azacytidine.
Muscle & nerve. 1995; 18(12): 1417-26.
[72] Culmes M, Eckstein H-H, Burgkart R, et al. Endothelial
differentiation of adipose-derived mesenchymal stem cells is
improved by epigenetic modifying drug BIX-01294. Eur J Cell
Biol 2013; 92(2): 70-9.
[73] Zhou G-S, Zhang X-L, Wu J-P, et al. 5-Azacytidine facilitates
osteogenic gene expression and differentiation of mesenchymal
stem cells by alteration in DNA methylation. Cytotechnology 2009;
60(1-3): 11-22.
[74] Stock P, Staege M, Müller L, et al., editors. Hepatocytes derived
from adult stem cells. Transplant Proc 2008; 40(2): 620-3.
[75] Snykers S, Henkens T, De Rop E, et al. Role of epigenetics in
liver-specific gene transcription, hepatocyte differentiation and
stem cell reprogrammation. J Hepatol 2009; 51(1): 187-211.
[76] Snykers S, Vanhaecke T, De Becker A, et al. Chromatin
remodeling agent trichostatin A: a key-factor in the hepatic
differentiation of human mesenchymal stem cells derived of adult
bone marrow. BMC Dev Biol 2007; 7(1): 24.
[77] Milhem M, Mahmud N, Lavelle D, et al. Modification of
hematopoietic stem cell fate by 5aza 2′ deoxycytidine and
trichostatin A. Blood 2004; 103(11): 4102-10.
[78] Blaschke K, Ebata KT, Karimi MM, et al. Vitamin [thinsp]
C induces Tet-dependent DNA demethylation and a blastocyst-
like state in ES cells. Nature 2013; 500(7461): 222-6.
[79] Monfort A, Wutz A. Breathing-in epigenetic change with vitamin
C. EMBO Rep 2013; 14(4): 337-46.
[80] Joseph J, Mudduluru G, Antony S, Vashistha S, Ajitkumar P,
Somasundaram K. Expression profiling of sodium butyrate (NaB)-
treated cells: identification of regulation of genes related to
cytokine signaling and cancer metastasis by NaB. Oncogene 2004;
23(37): 6304-15.
[81] Duvic M, Talpur R, Ni X, et al. Phase 2 trial of oral vorinostat
(suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous
T-cell lymphoma (CTCL). Blood 2007; 109(1): 31-9.
[82] Khan O, La Thangue NB. HDAC inhibitors in cancer biology:
emerging mechanisms and clinical applications. Immunol Cell Biol
2011; 90(1): 85-94.
[83] Cang S, Ma Y, Liu D. New clinical developments in histone
deacetylase inhibitors for epigenetic therapy of cancer. J Hematol
Oncol 2009; 2: 22.
[84] Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M,
Sinclair DA. Inhibition of silencing and accelerated aging by
nicotinamide, a putative negative regulator of yeast sir2 and human
SIRT1. J Biol Chem 2002; 277(47): 45099-107.
[85] Lara E, Mai A, Calvanese V, et al. Salermide, a Sirtuin inhibitor
with a strong cancer-specific proapoptotic effect. Oncogene 2008;
28(6): 781-91.
[86] Mukhopadhyay A, Banerjee S, Stafford LJ, Xia C, Liu M,
Aggarwal BB. Curcumin-induced suppression of cell proliferation
correlates with down-regulation of cyclin D1 expression and
CDK4-mediated retinoblastoma protein phosphorylation.
Oncogene 2002; 21(57): 8852-61.
[87] Shishodia S, Amin HM, Lai R, Aggarwal BB. Curcumin
(diferuloylmethane) inhibits constitutive NF-κB activation, induces
G1/S arrest, suppresses proliferation, and induces apoptosis in
mantle cell lymphoma. Biochem Pharmacol 2005; 70(5): 700-13.
[88] Tan J, Yang X, Zhuang L, et al. Pharmacologic disruption of
Polycomb-repressive complex 2-mediated gene repression
selectively induces apoptosis in cancer cells. Genes Dev 2007;
21(9): 1050-63.
[89] Greiner D, Bonaldi T, Eskeland R, Roemer E, Imhof A.
Identification of a specific inhibitor of the histone
methyltransferase SU (VAR) 3-9. Nat Chem Biol 2005; 1(3): 143-
5.
[90] Choi J-Y, Lee S, Hwang S, et al. Histone H3 lysine 27 and 9
hypermethylation within the Bad promoter region mediates 5-Aza-
2′-deoxycytidine-induced Leydig cell apoptosis: implications of 5-
Aza-2′-deoxycytidine toxicity to male reproduction. Apoptosis
2013; 18(1): 99-109.
[91] Portela A, Esteller M. Epigenetic modifications and human disease.
Nature Biotechnology. 2010; 28(10): 1057-68.
Received: September 17, 2014 Revised: December 3, 2014 Accepted: December 8, 2014

More Related Content

What's hot

2015 07 09__epigenetic_profiling_environmental_health_sciences_v42
2015 07 09__epigenetic_profiling_environmental_health_sciences_v422015 07 09__epigenetic_profiling_environmental_health_sciences_v42
2015 07 09__epigenetic_profiling_environmental_health_sciences_v42Prof. Wim Van Criekinge
 
Ibica2014 p(8) visualizing and identifying the dna methylation
Ibica2014 p(8) visualizing and identifying the dna methylationIbica2014 p(8) visualizing and identifying the dna methylation
Ibica2014 p(8) visualizing and identifying the dna methylationAboul Ella Hassanien
 
Epigenetics : overview and concepts
Epigenetics : overview and conceptsEpigenetics : overview and concepts
Epigenetics : overview and conceptsPrabhash Bhavsar
 
Epigenetic role in plant
Epigenetic role in plant Epigenetic role in plant
Epigenetic role in plant harshdeep josan
 
Efficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatmentEfficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatmentjanelle_leggere
 
Introduction to CpG island power point presentation
Introduction to CpG island power point presentationIntroduction to CpG island power point presentation
Introduction to CpG island power point presentationjkhdfhk
 
Inheritence of dna methylation
Inheritence of dna methylationInheritence of dna methylation
Inheritence of dna methylationsonam mahawar
 
002 & 003 Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14002 & 003   Dna Methyl And Human Disease 14
002 & 003 Dna Methyl And Human Disease 14guest0fa715
 
Unit Presentation Dec 08 Short
Unit Presentation Dec 08 ShortUnit Presentation Dec 08 Short
Unit Presentation Dec 08 ShortGilesOElliott
 
Dna methylation field guide 20130806
Dna methylation field guide 20130806Dna methylation field guide 20130806
Dna methylation field guide 20130806abizarl
 
An introduction to population based data for studies of DNA methylation
An introduction to population based data for studies of DNA methylationAn introduction to population based data for studies of DNA methylation
An introduction to population based data for studies of DNA methylationDavid Rehkopf
 
Dna methylation
Dna methylationDna methylation
Dna methylationAkshay Raj
 
Dna methylation signature
Dna methylation signatureDna methylation signature
Dna methylation signatureNikunj tyagi
 
Genomics epigenetic modifications & epigenomic technologies
Genomics epigenetic modifications & epigenomic technologiesGenomics epigenetic modifications & epigenomic technologies
Genomics epigenetic modifications & epigenomic technologiessciencelearning123
 

What's hot (20)

2015 07 09__epigenetic_profiling_environmental_health_sciences_v42
2015 07 09__epigenetic_profiling_environmental_health_sciences_v422015 07 09__epigenetic_profiling_environmental_health_sciences_v42
2015 07 09__epigenetic_profiling_environmental_health_sciences_v42
 
Ibica2014 p(8) visualizing and identifying the dna methylation
Ibica2014 p(8) visualizing and identifying the dna methylationIbica2014 p(8) visualizing and identifying the dna methylation
Ibica2014 p(8) visualizing and identifying the dna methylation
 
DNA_methylation
DNA_methylationDNA_methylation
DNA_methylation
 
DNA Methylation
DNA MethylationDNA Methylation
DNA Methylation
 
Dna methylation and epigenetics
Dna methylation and epigeneticsDna methylation and epigenetics
Dna methylation and epigenetics
 
Epigenetics : overview and concepts
Epigenetics : overview and conceptsEpigenetics : overview and concepts
Epigenetics : overview and concepts
 
Epigenetic role in plant
Epigenetic role in plant Epigenetic role in plant
Epigenetic role in plant
 
Efficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatmentEfficacy of epigenetic therapy as cancer treatment
Efficacy of epigenetic therapy as cancer treatment
 
Introduction to CpG island power point presentation
Introduction to CpG island power point presentationIntroduction to CpG island power point presentation
Introduction to CpG island power point presentation
 
Inheritence of dna methylation
Inheritence of dna methylationInheritence of dna methylation
Inheritence of dna methylation
 
002 & 003 Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14002 & 003   Dna Methyl And Human Disease 14
002 & 003 Dna Methyl And Human Disease 14
 
Unit Presentation Dec 08 Short
Unit Presentation Dec 08 ShortUnit Presentation Dec 08 Short
Unit Presentation Dec 08 Short
 
Dna methylation field guide 20130806
Dna methylation field guide 20130806Dna methylation field guide 20130806
Dna methylation field guide 20130806
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
An introduction to population based data for studies of DNA methylation
An introduction to population based data for studies of DNA methylationAn introduction to population based data for studies of DNA methylation
An introduction to population based data for studies of DNA methylation
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Epigenetics diet and cancer
Epigenetics diet and cancerEpigenetics diet and cancer
Epigenetics diet and cancer
 
DNA Methylation
DNA MethylationDNA Methylation
DNA Methylation
 
Dna methylation signature
Dna methylation signatureDna methylation signature
Dna methylation signature
 
Genomics epigenetic modifications & epigenomic technologies
Genomics epigenetic modifications & epigenomic technologiesGenomics epigenetic modifications & epigenomic technologies
Genomics epigenetic modifications & epigenomic technologies
 

Viewers also liked

Friend Collabsum 20130131
Friend Collabsum 20130131Friend Collabsum 20130131
Friend Collabsum 20130131Sage Base
 
New Cayman Chemical Products - Sept 24th, 2013
New Cayman Chemical Products - Sept 24th, 2013New Cayman Chemical Products - Sept 24th, 2013
New Cayman Chemical Products - Sept 24th, 2013Cayman Chemical
 
Bba 301 Assignment 8.2 Fret
Bba 301 Assignment 8.2 FretBba 301 Assignment 8.2 Fret
Bba 301 Assignment 8.2 Fretnmanuel
 
Computational molecular biophysics of ribonucleic acids and ribozymes
Computational molecular biophysics of ribonucleic acids and ribozymesComputational molecular biophysics of ribonucleic acids and ribozymes
Computational molecular biophysics of ribonucleic acids and ribozymeschrisltang
 
Sergi Fluorescence
Sergi FluorescenceSergi Fluorescence
Sergi Fluorescencepoutreda
 
Fluorescence recovery after photo bleaching
Fluorescence recovery after photo bleachingFluorescence recovery after photo bleaching
Fluorescence recovery after photo bleachinganasshokor
 
FRET BIOL 497 Presentation
FRET BIOL 497 PresentationFRET BIOL 497 Presentation
FRET BIOL 497 PresentationSaki Mihori
 
Fluorescence recovery after photobleaching
Fluorescence recovery after photobleachingFluorescence recovery after photobleaching
Fluorescence recovery after photobleaching1aditi1
 
Making organelles visible - in planta and in societas
Making organelles visible - in planta and in societasMaking organelles visible - in planta and in societas
Making organelles visible - in planta and in societasAnne Osterrieder
 
PEPTIDE NUCLEIC ACID
PEPTIDE NUCLEIC ACID PEPTIDE NUCLEIC ACID
PEPTIDE NUCLEIC ACID darwin1988
 
Energy Resonance Study, France 2005
Energy Resonance Study, France 2005Energy Resonance Study, France 2005
Energy Resonance Study, France 2005Thina3
 
Isothermal Nucleic Acid Amplification Techniques
Isothermal Nucleic Acid Amplification TechniquesIsothermal Nucleic Acid Amplification Techniques
Isothermal Nucleic Acid Amplification TechniquesAref Farokhi Fard
 
Fluorescence(Forster) Resonance Energy Transfer
Fluorescence(Forster) Resonance Energy TransferFluorescence(Forster) Resonance Energy Transfer
Fluorescence(Forster) Resonance Energy Transfersavvysahana
 
''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke
''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke
''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom DeebekeWisdom Deebeke Kate
 
Lecture on nucleic acid and proteins
Lecture on nucleic acid and proteinsLecture on nucleic acid and proteins
Lecture on nucleic acid and proteinsMarilen Parungao
 

Viewers also liked (20)

Friend Collabsum 20130131
Friend Collabsum 20130131Friend Collabsum 20130131
Friend Collabsum 20130131
 
New Cayman Chemical Products - Sept 24th, 2013
New Cayman Chemical Products - Sept 24th, 2013New Cayman Chemical Products - Sept 24th, 2013
New Cayman Chemical Products - Sept 24th, 2013
 
A Peek at Dpharm 2012
A Peek at Dpharm 2012A Peek at Dpharm 2012
A Peek at Dpharm 2012
 
Bba 301 Assignment 8.2 Fret
Bba 301 Assignment 8.2 FretBba 301 Assignment 8.2 Fret
Bba 301 Assignment 8.2 Fret
 
Computational molecular biophysics of ribonucleic acids and ribozymes
Computational molecular biophysics of ribonucleic acids and ribozymesComputational molecular biophysics of ribonucleic acids and ribozymes
Computational molecular biophysics of ribonucleic acids and ribozymes
 
Sergi Fluorescence
Sergi FluorescenceSergi Fluorescence
Sergi Fluorescence
 
Prashantha_Docking
Prashantha_DockingPrashantha_Docking
Prashantha_Docking
 
Calorimeter
CalorimeterCalorimeter
Calorimeter
 
Fluorescence recovery after photo bleaching
Fluorescence recovery after photo bleachingFluorescence recovery after photo bleaching
Fluorescence recovery after photo bleaching
 
FRET BIOL 497 Presentation
FRET BIOL 497 PresentationFRET BIOL 497 Presentation
FRET BIOL 497 Presentation
 
Fluorescence recovery after photobleaching
Fluorescence recovery after photobleachingFluorescence recovery after photobleaching
Fluorescence recovery after photobleaching
 
Making organelles visible - in planta and in societas
Making organelles visible - in planta and in societasMaking organelles visible - in planta and in societas
Making organelles visible - in planta and in societas
 
PEPTIDE NUCLEIC ACID
PEPTIDE NUCLEIC ACID PEPTIDE NUCLEIC ACID
PEPTIDE NUCLEIC ACID
 
Peptide nucleic acid
Peptide nucleic acidPeptide nucleic acid
Peptide nucleic acid
 
Energy Resonance Study, France 2005
Energy Resonance Study, France 2005Energy Resonance Study, France 2005
Energy Resonance Study, France 2005
 
Isothermal Nucleic Acid Amplification Techniques
Isothermal Nucleic Acid Amplification TechniquesIsothermal Nucleic Acid Amplification Techniques
Isothermal Nucleic Acid Amplification Techniques
 
Fluorescence(Forster) Resonance Energy Transfer
Fluorescence(Forster) Resonance Energy TransferFluorescence(Forster) Resonance Energy Transfer
Fluorescence(Forster) Resonance Energy Transfer
 
G7 ch2.1 cells and life
G7 ch2.1   cells and lifeG7 ch2.1   cells and life
G7 ch2.1 cells and life
 
''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke
''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke
''Electrophoretic Mobility Shift Assay'' by KATE, Wisdom Deebeke
 
Lecture on nucleic acid and proteins
Lecture on nucleic acid and proteinsLecture on nucleic acid and proteins
Lecture on nucleic acid and proteins
 

Similar to DNA methylation

DNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomas
DNA Methylation and Epigenetic Events Underlying Renal Cell CarcinomasDNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomas
DNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomaskomalicarol
 
Regulation of deoxynucleotide metabolism
Regulation of deoxynucleotide metabolismRegulation of deoxynucleotide metabolism
Regulation of deoxynucleotide metabolismdharmendra maurya
 
cancer nice paper.pdf
cancer nice paper.pdfcancer nice paper.pdf
cancer nice paper.pdfasthabisht789
 
Cancer Epigenetics: Concepts, Challenges and Promises
Cancer Epigenetics: Concepts, Challenges and PromisesCancer Epigenetics: Concepts, Challenges and Promises
Cancer Epigenetics: Concepts, Challenges and PromisesMrinmoy Pal
 
DNA methylation in lung cancer 1 (1) (2).pptx
DNA methylation in lung cancer 1 (1) (2).pptxDNA methylation in lung cancer 1 (1) (2).pptx
DNA methylation in lung cancer 1 (1) (2).pptx9596276530AMIN
 
Hipermetilação MTHFR e complicações em diabetes
Hipermetilação MTHFR e complicações em diabetesHipermetilação MTHFR e complicações em diabetes
Hipermetilação MTHFR e complicações em diabetesDarleneCamatiPersuhn
 
Epigenomes as a therapeutic target
Epigenomes as a therapeutic targetEpigenomes as a therapeutic target
Epigenomes as a therapeutic targetAhmed Wasif
 
Gallbladder cancer patient analysis
Gallbladder cancer patient analysisGallbladder cancer patient analysis
Gallbladder cancer patient analysisAnton Yuryev
 
Epigenetics and small molecule
Epigenetics and small moleculeEpigenetics and small molecule
Epigenetics and small moleculeFei Xiang
 
Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...
Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...
Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...Kazi Manir
 
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...arman170701
 
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .dr_ekbalabohashem
 
Cancer Genetics - Denise Sheer
Cancer Genetics - Denise SheerCancer Genetics - Denise Sheer
Cancer Genetics - Denise SheerDenise Sheer
 
The Role of DNA Methylation in Coronary Artery Disease
 The Role of DNA Methylation in Coronary Artery Disease The Role of DNA Methylation in Coronary Artery Disease
The Role of DNA Methylation in Coronary Artery DiseaseBardia Farivar
 

Similar to DNA methylation (20)

DNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomas
DNA Methylation and Epigenetic Events Underlying Renal Cell CarcinomasDNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomas
DNA Methylation and Epigenetic Events Underlying Renal Cell Carcinomas
 
Regulation of deoxynucleotide metabolism
Regulation of deoxynucleotide metabolismRegulation of deoxynucleotide metabolism
Regulation of deoxynucleotide metabolism
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Cancer epigenetics
Cancer epigenetics Cancer epigenetics
Cancer epigenetics
 
cancer nice paper.pdf
cancer nice paper.pdfcancer nice paper.pdf
cancer nice paper.pdf
 
Cancer Epigenetics: Concepts, Challenges and Promises
Cancer Epigenetics: Concepts, Challenges and PromisesCancer Epigenetics: Concepts, Challenges and Promises
Cancer Epigenetics: Concepts, Challenges and Promises
 
DNA methylation in lung cancer 1 (1) (2).pptx
DNA methylation in lung cancer 1 (1) (2).pptxDNA methylation in lung cancer 1 (1) (2).pptx
DNA methylation in lung cancer 1 (1) (2).pptx
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Hipermetilação MTHFR e complicações em diabetes
Hipermetilação MTHFR e complicações em diabetesHipermetilação MTHFR e complicações em diabetes
Hipermetilação MTHFR e complicações em diabetes
 
Epigenomes as a therapeutic target
Epigenomes as a therapeutic targetEpigenomes as a therapeutic target
Epigenomes as a therapeutic target
 
Gallbladder cancer patient analysis
Gallbladder cancer patient analysisGallbladder cancer patient analysis
Gallbladder cancer patient analysis
 
Epigenetics and small molecule
Epigenetics and small moleculeEpigenetics and small molecule
Epigenetics and small molecule
 
Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...
Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...
Anindya seminar 1 growth factors and cell cycle signalling in pathogenesis of...
 
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
Epigenetic silencing of MGMT (O6-methylguanine DNA methyltransferase) gene in...
 
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
Epigenetic Changes in Hepatocellular Carcinoma (HCC) .
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Cancer Genetics - Denise Sheer
Cancer Genetics - Denise SheerCancer Genetics - Denise Sheer
Cancer Genetics - Denise Sheer
 
Targeted Therapy
Targeted TherapyTargeted Therapy
Targeted Therapy
 
The Role of DNA Methylation in Coronary Artery Disease
 The Role of DNA Methylation in Coronary Artery Disease The Role of DNA Methylation in Coronary Artery Disease
The Role of DNA Methylation in Coronary Artery Disease
 

DNA methylation

  • 1. Send Orders for Reprints to reprints@benthamscience.ae Epigenetic Diagnosis & Therapy, 2015, 1, 5-13 5 2214-0840/15 $58.00+.00 © 2015 Bentham Science Publishers DNA Methylation: A Possible Target for Current and Future Studies on Cancer? Sahar Olsadat Sajadian and Andreas K. Nussler* University of Tübingen, BG Trauma Center, Siegfried Weller Institute, Schnarrenbergstraße 95, 72076 Tübingen, Germany Abstract: The field of epigenetics encompasses studies about the ways in which genes are influenced by various external stimuli, such as drugs, chemicals, and the environment. Over the last few years, it has become an increasingly important area of research. Recently, substantial evidence has been presented for the claim that certain diseases, including cancer, exhibit epigenetic changes. In the present overview, we will give a brief summary of recent observations on epigenetics and cancer as well as of the ways in which various small molecules may influence the epigenetic state of cells. Keywords: Cancer, CpG islands, DNA methylation, epigenetic drugs, epigenetics, TET proteins. DEFINITION OF EPIGENETICS Epigenetics is defined ‘as a permanent change of gene expression during cell proliferation and development, which does not cause a change of the gene sequences’. Epigenetic regulation takes place in three separate stages: 1) nucleosome positioning, 2) histone modification, and 3) DNA methylation (Fig 1). DNA METHYLATION In the past years, epigenetic studies have focused on DNA methylation in mammals. Particular interest was directed towards genomic imprinting in the inactivation pattern of X-chromosome, in silencing of the retrotransposon, known as repetitive elements, and in tissue- specific genes [1, 2], These epigenetic changes have demonstrated playing an important role in many pathologies and gene therapies [3-5]. During the DNA methylation process, a methyl group (CH3) is added to the phenyl ring of the cytosine at the 5- carbon position. Somatic cell methylation can take place at all CpG (cytosine- phosphate-guanin) dinucleotides positions except at the CpG island [6]. CpG islands are parts of the DNA with more than 500 bp and over 55% of GC content [7]. These islands accommodate approximately 60% of the mammalian gene promoters [8]. When DNA methylation occurs in these promoters, some crucial transcription factors (TF) are down-regulated with subsequent inhibition of the corresponding protein formation. However, it has been proven that even in regions that are poor in CpGs, such as the Oct4 promoter, DNA methylation plays an important role in gene regulation [9, 10]. *Address correspondence to this author at the University of Tübingen, BG Trauma Center, Siegfried Weller Institute, Schnarrenbergstraße 95, D-72076 Tübingen, Germany, Tel: +49 7071 606-1065; Fax: +49 7071 606-1978; E-mail: andreas.nuessler@gmail.com DNA methylation is supported by a special group of enzymes; the so called DNA methyltransferases (DNMT). Up to now, the following DNMTs have been discovered: DNMT1, DNMT1b, DNMT1o, DNMT1p, DNMT2, DNMT3A, DNMT3B, and DNMT3L. DNMTs such as DNMT3A, DNMT3B, and DNMT3L are apparently responsible for the de novo DNA methylation in early developmental stages. In order to maintain the methylation in daughter cells, the so called UHRF1 protein, a ubiquitin-like protein with PHD and RING finger domain 1, binds to hemi-methylated DNA. During replications, UHRF1 causes DNMT1 to methylate new cytosine from synthesized DNA strands. There exists also growing evidence which suggests that DNMT3A/B are also involved in maintaining the DNA methylation pattern in somatic cells, specifically in imprinted genes and repeated elements [10]. The significance of DNMTs was demonstrated in various animal studies, proving that mice lacking DNMTs died during their early development or shortly after birth [11]. Nevertheless, the expression of these enzymes differs between certain types of cancer and it is not always associated with hyper- or hypomethylation, as had been expected. Sometimes, the expression of DNMTs is regulated by miRNAs, which may explain the differences in the expression of DNMTs in various tumors [12]. For instance, it has been demonstrated in several studies that DNA demethylase is post-transcriptionally regulated by mir29 and mir148 [13]. Furthermore, So et al. have claimed that DNA methyltransferase controls stem cell aging through microRNA. They have shown that DNMT inhibition leads to a decrease of the expression of Polycomb groups (PcG) and to an increase of the expression of microRNAs (miRNAs) that target PcG [14]. DNA DEMETHYLATION In early embryogenesis, many researchers have observed a loss of DNA methylation and subsequently have postulated the existence of several DNA demethylases and various
  • 2. 6 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler mechanisms of the DNA demethylation [15, 16]. One potential DNA demethylation pathway is enabled by the ten- eleven-translocation proteins (TETs), which are named after the ten-eleven-translocation (q22, q23) (10, 11) that occurs in rare cases of acute myeloid and lymphocytic leukemia [17, 18]. 5-mC is oxidized by TET proteins and thereby converted to 5-hmC, 5-fC, and 5-CaC, which all belong to the family of Fe (II)/2-oxygluttarate-dependent dioxgenases [19]. DNA methylation is reversible either through passive demethylation via DNA replication or by active demethylation via DNA repair followed by base excision repair of modified nucleotides or DNA glycosidase [15, 17] (Fig. 2). TET proteins play different roles in diverse biological processes, including epigenetic regulation of gene transcription, embryonic development, stem cell functions, and cancer. They mediate the DNA demethylation process and change the epigenetic state of DNA. A loss of 5-hmC, together with a down-regulation or mutation of TET proteins, has been reported in numerous types of cancer, like melanoma, breast, prostate, myeloid, brain, and liver cancer [20]. It has been demonstrated that in patients suffering from acute myeloid leukemia (AML), TET1 protein influences mixed lineage leukemia (MLL) development [16]. Furthermore, several changes in TET2 proteins, like e.g. deletion and deactivation of TET2, have been identified in 30% of all patients suffering from different kinds of myeloid malignancies. Several work groups have reported an alteration of a TET2 gene in a variety of myeloid malignancies and the majority of the TET2 alterations include single or double copy defects and nonsense/frame shift mutation suggesting that TET2 lesions cause a loss of function. Therefore, it has been suggested that TET2 is a possible tumor suppressor gene that is responsible for myeloid malignancies [21]. Furthermore, the loss of 5-hmC in correlation with clinical progression and relapse of melanoma has been reported [20]. DNA METHYLATION AND CANCER Numerous studies have reported that tumor cells exhibit a major disruption in the regulation of the DNA methylation pattern in cancer. Therefore, over the last few years, studies have particularly focused on changes in DNA methylation. [22]. Earlier studies have demonstrated that the hypomethylation of interspersed repetitive DNA is a common feature of numerous types of cancer that causes genomic instability and aberrant transcription initiation. However, researchers have proven that the hypermethylation of focal CpG islands is elevated in cancer and may result in Fig. (1). Epigenetic machinery and interplay between epigenetic factors (modified from [91]). Epigenetic regulation depends on the three following epigenetic factors: (a) DNA methylation, (b) histone modification, and (c) nucleosome positioning. The interaction between these factors results in the final outcome, but disturbances of this balance lead to several diseases. Abbreviations: DNMT: DNA Methyl Transferase, HAT: Histone Acetylation Transferase, HDAC: Histone Deacetylase, HMT: Histone Methyltransferase, HDM: Histone Demethyl Transferase. MethylationMethylation DemethylationDemethylation DNMT1 TETs: Me DNA Methylation (maintenance DNMT) DNMT3A & DNMT3B (de novo DNMT) TET1, TET2, TET3 5hmC Epigenetic machinery Epigenetic machinery AcetylationAcetylation HAT & HDAC Histone modifications Histone modifications Chromatin remodeling Chromatin remodeling MethylationMethylation HAT & HDAC HMT & HDM Disfunc Phosphor- tylation Phosphor- tylation Kinase & Phosphor SWI/SNF, ISWI MI-2, INO80 Disease ctioning Ac Me Me P Disease • Cancer • Autoimmune disorder • Neural disorder • Aging Ub
  • 3. DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 7 inhibition of various miRNAs, tumor suppressor and functional genes [23, 24]. Moreover, it has become evident that different pathways target DNMT at the specific CpG Island. Moreover, four different mechanisms target DNA methyltransferase. Firstly, DNMT3A and DNMT3L recognize DNA or chromatin with specific domains and they interact with the LYS4 of histone 3 (H3K4). Secondly, DNMT interacts with different chromatin modifiers such as KMT and HDACs. Thirdly, the Piwi-PiRNA complex, a class of ncRNAs, guides DNMTs to transposon elements. Finally, the direct interaction of promoter-associated RNA (pRNA) with the promoter of the rRNA gene might recruit DNMT3b and enable methylation [25]. In addition, it has been shown that, in an alteration between methylation and phosphorylation, an adjacent lysine and serine determine the stability of DNMT1 thus preserving the methylation pattern during cell division [26]. However, the detailed mechanism(s) leading to hyper- or hypomethylation shall be decorticated in the future. HYPERMETHYLATION IN CANCER The CpG islands are protected by various defense mechanisms by accessing the DNA methyltransferase, such as DNA replication timing, active demethylation, or active transcription. Most genes that are susceptible to hypermethylation are associated with the regulation of DNA repair, apoptosis, cell cycle, drug resistance, metastasis, and differentiation [10]. Many tumors are hypermethylated in one or more genes. For instance, in lung cancer, it has been demonstrated that alterations of DNA methylation have been found in more than 40 genes [27]. Regarding breast cancer, it has been shown that several critical genes, which seem to be important in tumorigenesis, are hypermethylated. Among those genes, cell adhesion genes, inhibitors of matrix metalloproteinases, as well as the ER and PR steroid receptor genes can be found and have been linked to breast cancer development [28]. It has also been reported that DNA methylation is one of the reasons for the inactivation of BRCA1 in breast cancer, which is one of the genes most commonly associated with breast cancer [29]. Moreover, the hypermethylation of other cell cycle genes may result in acute lymphoblastic leukemia which is an undesirable clinical outcome [30]. HYPOMETHYLATION IN MALIGNANCIES Hypomethylation is an alteration in DNA methylation which is frequently seen in solid tumors [31-33], and is a sign of a progressive malignancy [34]. Furthermore, it has been reported that the hypomethylation status contributes to oncogenesis through up-regulation of some oncogenes, like H-RAs and c-MYC [31], as well as by chromosome instability [35, 36]. During cancer development, aberrations not only occur in the DNA methylation, but also in the regulation of histone modifications [37]. For instance, numerous studies have demonstrated that histone deacytylases (HDACs) are overexpressed in different types of cancer. This overexpression leads to a deacetylation of the Transcription Starting Site (TSS) of important genes and subsequently to the assembly of silenced genes into more compact structures [38, 39]. Fig. (2). Active demethylation pathway through the conversion of 5mC to 5hmC by recruiting TET proteins. Following 5mC oxidation, demethylation occurs by dilution of oxidized base or by removal through DNA glycosidase- and DNA repair-mediated excision of modified nucleotides (modified from [15]). Abbreviations: C: Cytosine, 5mC: 5-methylcytosine, 5hmC: 5-hydroxymethylcytosine; 5fC: 5- formylcytosine, 5caC: 5-carboxylcytosine, TDG: Thymine DNA Glucosidase, BER: Base Excision Repair.
  • 4. 8 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler In summary, there exists an epigenetic crosstalk between histone modification, micro-RNAs, and DNA methylation. Post-transcriptionally, miR-29 and miR-148 regulate the DNA demethylases [40, 41]. Moreover, it has been proven that DNMTs interact with HDAC [28]. In particular, the combination of DNMT and HDAC inhibitors is a primary target for potential therapeutics against cancer [42, 43], because these inhibitors are capable of targeting the epigenetic status of DNA and histones simultaneously, which may enable the development of more effective anticancer drugs. EPIGENETICS AND CANCER STEM CELL HYPOTHESIS Recently, the concept has arisen that cancer stem cells are to be classified as a subpopulation of cancer cells with a different tumorigenic potential. This side population, which is usually named cancer-initiating cells or cancer stem cells (CSC), has a characteristic potential for self-renewal that may sustain and perpetuate tumors and is known to dispose of a higher drug resistance. Firstly, it has been observed that CD34+CD38- leukemic stem cells disposing of a differentiation and proliferation capacity are able to trigger human leukemia in NOD/SCID mice [44]. Furthermore, it has been proven that a landscape of solid tumors, such as glioblastoma [45], breast cancer [46], prostate cancer [47], and hepatocellular carcinoma [48], contain CSCs. A growing number of studies prove that the cancer-initiating cells are mostly resistant to chemotherapy or radiotherapy, which results in the recurrence of tumors [49], and in the formation of metastases [50]. Studies on the epigenetic modifications in stem cells and in cancer cells have directed interest to stem/progenitor cells, which are at the origin of cancer. It is believed that the genetic and epigenetic changes in these cells provide the ability for their survival, protecting them from toxic environment, which then contributes to tumor initiation [51]. Interestingly, the Polycomb repressive complex, which causes abnormal gene silencing in cancer, represses CpG island genes in Emberyonic Stem Cells (ESCs) in the same way as in cancer [52, 53]. Following this abnormal hypermethylation due to DNA damage, inflammation or stress caused by ROS, tumor suppressor genes become silenced. This may lead to the activation of stem cell pathways and provide the ability of self-renewal for cancer cells [54]. For example, in stem cells, like e.g. the CD133+ cells, promoter hypermethylation of some tumor suppressor genes, such as SFRP1 and SOX17, was identified [55], proving the stem cell origin of epigenetic abnormalities in cancer [56]. APPLICATION OF EPIGENETIC DRUGS As was described above, the application of epigenetic drugs is being increasingly focused on in the field of biomedicine and drug development research. Like numerous other diseases, cancer causes aberrancies in the epigenetic regulation. Most importantly, the pattern of DNA demethylation and histone modification has been related to tumor genesis and metastasis [10, 57]. Therefore, it is believed that these two biomechanical processes have a high potential for being used in clinical applications, such as e.g. in cancer detection, tumor prognosis, and pharmacoepigenetics (prediction of treatment response). For example, hypermethylation of glutathione transferase (GSTP1) constitutes a promising epigenetic marker in prostate cancer [58]. It has to be pointed out that 80-90% of men suffering from prostate cancer exhibit a hypermethylation of GSTP1 and that this hypermethylation does not occur in benign hyperplastic prostate tissue [59]. Therefore, GSTP1 methylation is a helpful prognostic marker, which enables the discrimination of benign tumor tissue in prostate cancer. As a matter of fact, GSTP1 is also hypermethylated in other cancers such as in kindney, liver and breast cancer [60]. Interestingly, the screening of single markers through global DNA methylation offers great clinical potential, as for example, it allows for the prediction of metastases in colorectal cancer, recurrence in lung cancer, and progression in virus-associated neoplasms [57, 61]. In addition to DNA methylation markers, histone-based markers have been suggested as epigenetic biomarkers. In particular, aberrant histone modifications have been linked to recurring prostate cancer [62]. Nowadays, most studies focus on the development of epigenetic drugs, which can reverse the abnormal epigenetic state to the normal epigenetic state in cancer cells. Up to now, the US Food and Drug Administration (FDA) has approved four epigenetic small molecule drugs for the treatment of cancer: 5-Azacytidine (5-Aza) and 5-Aza-2´- deoxycytidine (DNMT inhibitors) which have been used for patients suffering from myelodyplastic syndrome and from acute leukemia, as well as Varinostat and Romidepsin (HDAC inhibitors) which are used for treatment of T-cell lymphoma and some hematological tumors [63]. It has to be pointed out that each DNMT inhibitor possesses its own way of action on the gene transcription. Although 5-Aza and 5-Aza-2´-deoxycytidine have the same therapeutic effect, their mode of action in tumor cells is different. It has been demonstrated that 5-Aza-2´- deoxycytidine is able to induce senescence in tumor cells, while 5-Aza induces cell death in tumor cells in vitro and in vivo. These two DNMT inhibitors differ from each other on the molecular and the cellular level: For instance, 5-Aza can be integrated in both DNA and RNA (20-40% and 60-80% respectively), while 5-Aza-2´-deoxycytidine can only be incorporated in DNA. Moreover, 5-Aza-2´-deoxycytidine induces more DNA damage and micronuclei formation than 5-Aza [64]. Furthermore, it has been rported that Azacytidine increases the antitumor effect sensitivity of Docetaxel (DTX) and Cisplation in aggressive and chemoresistant prostate cancer [65]. In addition, it has been shown that Pyrazofurin (PF), an inhibitor of orotidylate decarboxylase improved the anti-tumoral activity of 5-Aza in human leukemia cells [66]. The combination of PF and 5-Aza may provide a useful treatment for patients who do not respond to standard anti- leukemic therapies [67]. Furthermore, it has been proven that 5-Aza-2´-deoxycytidine inhibits telomerase activity and reactivates p16 and c-Myc in hepatocellular carcinoma cells (HCC), such as the MMC-7721 and the HepG2 cell line [68]. Other DNMT inhibitors, such as 5-fuoro-2´- deoxycytidine, in combination with other drugs are currently
  • 5. DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 9 undergoing clinical trials regarding the treatment of different types of cancers [69]. The DNA methyltransferase inhibitor Zebularin provoked the demethylation of tumor suppressor genes by inducing apoptosis, cell cycle regulation in HCC cell lines (Huh7, KMCH, and HepG2), and in vivo inhibition of tumor growth in a xenograft model [70]. It has been discussed if the expression of specific DNMTs might influence the differentiation status in Fig. (3). Epigenetic drugs for cancer therapy. It has been described that scores of small molecules participate in the fight against cancer cells by inducing changes in the epigenetic machineries. This figure illustrates some of the most important epigenetic drugs, which are classified in relation to their epigenetic target. Abbreviations: DNMT1: DNA Methyltransferase 1, HDAC: Histone Deacetylase, PcG: Polycomb Group, Me: Methylated, HAT: Histone Acetylase. Repressive complex Repressive complex DNMT1DNMT1 PcG SirtuinsSirtuins HDACHDAC PcG MeDNMT Inhibitors: Vidaza Decitabine HMT Inhibitors DZNep BIX-01294 Me Decitabine Zelbularine Procainamide HDAC Inhibitors:HDAC Inhibitors: SIRTSIRT BIX 01294 Chaetocin DNMT1DNMT1 HDACHDAC Me VPA TSA Vorinostat Panobinostat Belinostat VPA TSA Vorinostat Panobinostat Belinostat Inhibitors Sirtinol Cambinol Salermide Inhibitors Sirtinol Cambinol Salermide PcG SirtuinsSirtuins Transcription HATHAT TETsTETs factors/ coactivator Pol IIPol II 5-methylcytosine5 methylcytosine 5-hydroxymethylcytosine 5-formylcytosine 5-carboxylcytosine Histone acetylated
  • 6. 10 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler developing and dedifferentiating cells. It has been shown that 5-Aza triggers differentiation in mesenchymal stem cells (MSCs) [8, 71, 72]. Furthermore, we have reported that pretreatment of fat-derived old MSCs with 5-Aza improves osteogenic differentiation by DNA demethylation. We have observed a reduction of 5hmC in Ad-MSCs from older donors (> 60 years) compared to young donors (< 45 years). After stimulation of cells with 5-Azacytidine the osteogenic potential of the cells improved. It has been observed that the increase of AP activity and matrix mineralization is associated with an inducement of the expression of 5hmC as well as with the expression of TET2 and TET3 [8]. Therefore, we have concluded that 5-Azacytidine induces the rejuvenating process of older Ad-MSCs (Fig. 4). Furthermore, it has been demonstrated in various studies that the treatment of human Adipose Stem Cells (ADSC) and human bone marrow MSCs with 20 µM 5-Azacytidine improves their hepatic differentiation [73-75]. In the same line of evidence, it has been shown that the incubation of human bone marrow MSCs with 1 µM TSA which had been pretreated with hepatogenic-stimulating agents, improves the hepatogenic differentiation of these cells [76]. Moreover, a combination of TSA and 5-Aza-dc has improved the differentiation of human CD34+ to CD34/ CD90 cells [77]. Regarding the influence of the epigenetic status on the reprogramming mechanism of the cells, it has been reported that vitamin C induces the active demethylation pathway through the induction TET enzyme activity in mouse embryonic stem cells (ES) [78]. In the same line of evidence, it has been demonstrated that TET2 facilitates the differentiation of hematopoietic stem cells [79]. Therefore, vitamin C can be used for reprogramming stem cells, as it induces TET-dependent demethylation. OTHER EPIGENETIC MODIFYING DRUGS In the past years, many anticancer drugs targeting the acetylation or methylation status of histones through different mechanisms have been developed, such as HDAC inhibitors class I, II, III (sirtuins), and IV, HATs, HMTs, HDMs, and various kinds of kinases. Based on their chemical properties, the HDACs are branched into the four Fig. (4). Reprogrammation and differentiation of cells through a DNA methylation pattern. Changes in the methylation status of cells may lead to differentiation either through passive demethylation [8, 72] by inhibition of DNMT or through active demethylation by inducing TET proteins to produce 5hmC [78, 79]. Abbreviations: 5mC: 5-methylcytosine 5hmC: 5-hydroxymethylcytosine; 5fC: 5-formylcytosine; 5caC: 5-carboxylcytosine; TDG: Thymine DNA Glucosidase; BER: Base Excision Repair, TET: Ten eleven translocation, DNMT: DNA Methyltransferase, SAH: S-adenosylhomocysteine, Hcy: Homocysteine SAM: S-Adenosylmethionin. Progenitor / stem Cells Differentiated Cells DNMT Inhibitors: 5-AZA stem Cells MethionineSAH5mC 5caC C Pol II Progenitor Cells HcySAM 5hm C 5fC 5caC Pol IICells Differentiated Cells
  • 7. DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 11 following groups: hydroxamic acid, short fatty acids, cyclic peptide and benzamide enzymes [57]. HDACs inhibitors mainly arrest the cell cycle in G1 and G2-M phases, which, in turn, induces apoptosis and cell differentiation. However, they are able to inhibit angiogenesis and metastasis, and to decrease the resistance of tumor cells to chemotherapy [80]. Recently, a new epigenetic drug called Suberoylanilide hydroxamic acid (SAHA) has been approved for treating T- cell cutaneous lymphoma by the Federal Drug Administration (FDA) [81]. Furthermore, a large number of HDAC inhibitors, like e.g. TSA (Tricostatin A), PXD101 (Belinostat), and LBH589 (Panobinostat), are currently being developed and are undergoing clinical trials [82, 83]. Class III HDAC inhibitors, so called Sirtuins (SIRT), are important for the regulation of senescence, survival, and cell proliferation. An increasing number of effective SIRT inhibitors have recently been generated [57]. SIRT1 and SIRT2 inhibitors are likely to stop p53 activity, thereby inducing growth arrest in cancer cells and increasing sensitivity of tumor cells towards anticancer drugs [84]. Salermide - another SIRT1 and SIRT2 inhibitor - induces p- 53-independent apoptosis in tumor cells and reactivates proapoptotic genes by deacytelation of H4K16Ac at the epigenetic level which is mediated by SIRT1 [85]. It has been shown that curcumin, garcinol, and anacaridic acid may act as HAT inhibitors with an anti-cancer effect in ovarian and colon cancer. It has been demonstrated that all three substances are able to inhibit EP300-mediated p53 acetylation [86, 87]. DZNep, Chaetocin, and BIX-01294 are known to be lysine HMT inhibitors. It has been shown that DZNep promotes apoptosis in the MCF7 cell line (breast cancer) and in the HCT116 cell line (colorectal cancer) [88]. Chaetocine has an anti-cancer effect on multiple myeloma (MM) [89]. BIX-01294 deactivates the histone H3 lysine 9 (H3K9) methyltransferases G9a and G9a-like protein by promoting the apoptotic cell death [90]. It has been demonstrated that BIX-01294 induces endothelial differentiation in Ad-MSCs. We have been able to demonstrate that BIX modifies DNA and histone methylation and remarkably increases the expression of various endothelial markers that are required for blood vessel formation, such as VCAM-1, PECAM, VEGFR-2, and PDGF [72]. From our studies, we conclude that drugs that modify the DNA or histone methylation also induce changes in the pluripotency [8, 72]. SUMMARY AND FUTURE PERSPECTIVES In the present overview, we have presented numerous encouraging findings demonstrating that so called small molecules exert anti-tumoral activity through epigenetic changes within target cells. Interestingly, even molecules, like e.g. polyphenols, which are considered to be anti- oxidative drugs, dispose of a potent epigenetic modifying activity, and therefore slow down tumor growth. However, the large variety among the structures of different molecules that exert epigenetic modifying activity bears a high risk. The mechanisms by which epigenetically modifying drugs reduce growth or damage tumor cells are not very specific and may therefore damage normal/healthy cells as well. In this avenue, it is mandatory to develop new and more specific small molecules against specific enzymes of the epigenetic machinery in order to reduce damage on healthy tissue. CONFLICT OF INTEREST The authors declare that they do not have any conflict of interest. ACKNOWLEDGEMENTS We like to thank Dr. Luc Koster for editing the manuscript. The authors would also like to thank the Ministry of Rural Affairs, and Consumer Protection Baden- Württemberg for supporting our study and hence promoting important research projects. REFERENCES [1] Laird PW. The power and the promise of DNA methylation markers. Nat Rev Cancer 2003; 3(4): 253-66. [2] Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Gen 1999; 23(2): 185-8. [3] Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat. Rev Gen 2002; 3(6): 415-28. [4] Baylin SB. Tying it all together: epigenetics, genetics, cell cycle, and cancer. Science 1997; 277(5334): 1948-9. [5] Costello JF, Plass C. Methylation matters. J Med Gen 2001; 38(5): 285-303. [6] Suzuki MM, Bird A. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Gen 2008; 9(6): 465-76. [7] Takai D, Jones PA. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc Nat Acad Sci 2002; 99(6): 3740-5. [8] Yan X, Ehnert S, Culmes M, et al. 5-Azacytidine improves the osteogenic differentiation potential of aged human adipose-derived mesenchymal stem cells by DNA demethylation. PloS one 2014; 9(3): e90846. [9] Irizarry RA, Ladd-Acosta C, Wen B, et al. The human colon cancer methylome shows similar hypo-and hypermethylation at conserved tissue-specific CpG island shores. Nat Gen 2009; 41(2): 178-86. [10] Das PM, Singal R. DNA methylation and cancer. J Clin Oncol 2004; 22(22): 4632-42. [11] Robertson KD. DNA methylation and chromatin-unraveling the tangled web. Oncogene 2002; 21(35): 5361-79. [12] Blair LP, Yan Q. Epigenetic mechanisms in commonly occurring cancers. DNA Cell Biol 2012; 31(S1): S-49-S-61. [13] Pan W, Zhu S, Yuan M, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J Immunol 2010; 184(12): 6773-81. [14] So A-Y, Jung J-W, Lee S, Kim H-S, Kang K-S. DNA methyltransferase controls stem cell aging by regulating BMI1 and EZH2 through microRNAs. PLoS One 2011; 6(5): e19503. [15] Kohli RM, Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 2013; 502(7472): 472-9. [16] Wu SC, Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat Rev Mol Cell Biol 2010; 11(9): 607-20. [17] Tahiliani M, Koh KP, Shen Y, et al. Conversion of 5- methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009; 324(5929): 930-5. [18] Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self- renewal and inner cell mass specification. Nature 2010; 466(7310): 1129-33. [19] Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5- methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 2011; 333(6047): 1300-3.
  • 8. 12 Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 Sajadian and Nussler [20] Lian CG, Xu Y, Ceol C, et al. Loss of 5-hydroxymethylcytosine is an epigenetic hallmark of melanoma. Cell 2012; 150(6):1135-46. [21] Li Z, Cai X, Cai C-L, et al. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood 2011; 118(17): 4509-18. [22] Baylin SB, Herman JG. DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends Genet 2000; 16(4): 168-74. [23] Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene 2002; 21(35): 5427-40. [24] Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene 2002; 21(35): 5400-13. [25] Denis H, Fuks F. Regulation of mammalian DNA methyltransferases: a route to new mechanisms. EMBO Rep 2011; 12(7): 647-56. [26] Estève P-O, Chang Y, Samaranayake M, et al. A methylation and phosphorylation switch between an adjacent lysine and serine determines human DNMT1 stability. Nat Struc Mol Biol 2011; 18(1): 42-8. [27] Tsou JA, Hagen JA, Carpenter CL, Laird-Offringa IA. DNA methylation analysis: a powerful new tool for lung cancer diagnosis. Oncogene 2002; 21(35): 5450-61. [28] Yang X, Yan L, Davidson N. DNA methylation in breast cancer. Endocrine-Related Cancer 2001; 8(2): 115-27. [29] Catteau A, Morris JR, editors. BRCA1 methylation: a significant role in tumour development? Semin Cancer Biol 2002; 12(5): 359- 371. [30] Leone G, Teofili L, Voso MT, Lubbert M. DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica 2002; 87(12): 1324-41. [31] Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 1983; 301(5895): 89-92. [32] Kim YI, Giuliano A, Hatch KD, et al. Global DNA hypomethylation increases progressively in cervical dysplasia and carcinoma. Cancer 1994; 74(3): 893-9. [33] Lin C-H, Hsieh S-Y, Sheen I-S, et al. Genome-wide hypomethylation in hepatocellular carcinogenesis. Cancer Res 2001; 61(10): 4238-43. [34] Bedford MT, Van Helden PD. Hypomethylation of DNA in pathological conditions of the human prostate. Cancer Res 1987; 47(20): 5274-6. [35] Alves G, Tatro A, Fanning T. Differential methylation of human LINE-1 retrotransposons in malignant cells. Gene 1996; 176(1): 39-44. [36] Tuck-Muller C, Narayan A, Tsien F, et al. DNA hypomethylation and unusual chromosome instability in cell lines fromICF syndrome patients. Cytogenet Cell Genet 2000; 89(1-2): 121-8. [37] Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12-27. [38] Halkidou K, Gaughan L, Cook S, Leung HY, Neal DE, Robson CN. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. Prostate 2004; 59(2): 177-89. [39] Song J, Noh JH, Lee JH, et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS 2005; 113(4): 264-8. [40] Fabbri M, Garzon R, Cimmino A, et al. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Nat Acad Sci 2007; 104(40): 15805-10. [41] Duursma AM, Kedde M, Schrier M, Le Sage C, Agami R. miR-148 targets human DNMT3b protein coding region. RNA 2008; 14(5): 872-7. [42] Zhu W-G, Otterson GA. The interaction of histone deacetylase inhibitors and DNA methyltransferase inhibitors in the treatment of human cancer cells. Curr Med Chem Anticancer Agents 2003; 3(3): 187-99. [43] Unoki M. Current and potential anticancer drugs targeting members of the UHRF1 complex including epigenetic modifiers. Recent Pat Anticancer Drug Discov 2011; 6(1): 116-30. [44] Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. 1994; 367(6464):645-8. [45] Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature. 2004; 432(7015): 396-401. [46] Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proceedings of the National Academy of Sciences. 2003; 100(7): 3983-8. [47] Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer research. 2005; 65(23): 10946-51. [48] Suetsugu A, Nagaki M, Aoki H, Motohashi T, Kunisada T, Moriwaki H. Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun 2006; 351(4): 820-4. [49] Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res 2008; 68(1): 190-7. [50] Bao S, Wu Q, McLendon RE, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006; 444(7120): 756-60. [51] Ohm JE, Baylin SB. Stem cell chromatin patterns--an instructive mechanism for DNA hypermethylation? Cell cycle (Georgetown, Tex) 2007; 6(9): 1040. [52] Schlesinger Y, Straussman R, Keshet I, et al. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nature Gen 2006; 39(2): 232-6. [53] Widschwendter M, Fiegl H, Egle D, et al. Epigenetic stem cell signature in cancer. Nature Gen 2006; 39(2): 157-8. [54] Baylin SB, Ohm JE. Epigenetic gene silencing in cancer-a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 2006; 6(2): 107-16. [55] Zhang W, Glöckner SC, Guo M, et al. Epigenetic inactivation of the canonical Wnt antagonist SRY-box containing gene 17 in colorectal cancer. Cancer Res 2008; 68(8): 2764-72. [56] Tsai H-C, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res 2011; 21(3): 502-17. [57] Rodríguez-Paredes M, Esteller M. Cancer epigenetics reaches mainstream oncology. Nat Med 2011; 330-9. [58] Henrique R, Jerónimo C. Molecular Detection of Prostate Cancer: A Role for< i> GSTP1</i> Hypermethylation. European urology. 2004; 46(5):660-9. [59] Lee W-H, Morton RA, Epstein JI, et al. Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Nat Acad Sci 1994; 91(24): 11733-7. [60] Esteller M, Corn PG, Urena JM, Gabrielson E, Baylin SB, Herman JG. Inactivation of glutathione S-transferase P1 gene by promoter hypermethylation in human neoplasia. Cancer Res 1998; 58(20): 4515-8. [61] Esteller M. Epigenetics in cancer. New Engl J Med 2008; 358(11): 1148-59. [62] Seligson DB, Horvath S, Shi T, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature 2005; 435(7046):1262-6. [63] Byrd JC, Marcucci G, Parthun MR, et al. A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia. Blood 2005; 105(3): 959-67. [64] Venturelli S, Berger A, Weiland T, et al. Differential induction of apoptosis and senescence by the DNA methyltransferase inhibitors 5-azacytidine and 5-aza-2′-deoxycytidine in solid tumor cells. Mol. Cancer Ther 2013; 12(10): 2226-36. [65] Festuccia C, Gravina GL, D'Alessandro AM, et al. Azacitidine improves antitumor effects of docetaxel and cisplatin in aggressive prostate cancer models. Endocr Relat Cancer. 2009; 16(2): 401-13. [66] Cadman E, Eiferman F, Heimer R, Davis L. Pyrazofurin enhancement of 5-azacytidine antitumor activity in L5178Y and human leukemia cells. Cancer Res 1978; 38(12): 4610-7. [67] Lancet JE, Karp JE. Novel postremission strategies in adults with acute myeloid leukemia. Curr Opin Hematol 2009; 16(2): 105. [68] Tao S-F, Zhang C-S, Guo X-L, et al. Anti-tumor effect of 5-aza-2'- deoxycytidine by inhibiting telomerase activity in hepatocellular carcinoma cells. World J Gastroenterol 2012; 18(19): 2334. [69] Yang X, Lay F, Han H, Jones PA. Targeting DNA methylation for epigenetic therapy. Trends Pharmacol Sci 2010; 31(11): 536-46. [70] Nakamura K, Aizawa K, Nakabayashi K, et al. DNA methyltransferase inhibitor zebularine inhibits human hepatic carcinoma cells proliferation and induces apoptosis. PloS One 2013; 8(1): e54036.
  • 9. DNA Methylation: Possible Target for Studies on Cancer Epigenetic Diagnosis & Therapy, 2015, Vol. 1, No. 1 13 [71] Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle & nerve. 1995; 18(12): 1417-26. [72] Culmes M, Eckstein H-H, Burgkart R, et al. Endothelial differentiation of adipose-derived mesenchymal stem cells is improved by epigenetic modifying drug BIX-01294. Eur J Cell Biol 2013; 92(2): 70-9. [73] Zhou G-S, Zhang X-L, Wu J-P, et al. 5-Azacytidine facilitates osteogenic gene expression and differentiation of mesenchymal stem cells by alteration in DNA methylation. Cytotechnology 2009; 60(1-3): 11-22. [74] Stock P, Staege M, Müller L, et al., editors. Hepatocytes derived from adult stem cells. Transplant Proc 2008; 40(2): 620-3. [75] Snykers S, Henkens T, De Rop E, et al. Role of epigenetics in liver-specific gene transcription, hepatocyte differentiation and stem cell reprogrammation. J Hepatol 2009; 51(1): 187-211. [76] Snykers S, Vanhaecke T, De Becker A, et al. Chromatin remodeling agent trichostatin A: a key-factor in the hepatic differentiation of human mesenchymal stem cells derived of adult bone marrow. BMC Dev Biol 2007; 7(1): 24. [77] Milhem M, Mahmud N, Lavelle D, et al. Modification of hematopoietic stem cell fate by 5aza 2′ deoxycytidine and trichostatin A. Blood 2004; 103(11): 4102-10. [78] Blaschke K, Ebata KT, Karimi MM, et al. Vitamin [thinsp] C induces Tet-dependent DNA demethylation and a blastocyst- like state in ES cells. Nature 2013; 500(7461): 222-6. [79] Monfort A, Wutz A. Breathing-in epigenetic change with vitamin C. EMBO Rep 2013; 14(4): 337-46. [80] Joseph J, Mudduluru G, Antony S, Vashistha S, Ajitkumar P, Somasundaram K. Expression profiling of sodium butyrate (NaB)- treated cells: identification of regulation of genes related to cytokine signaling and cancer metastasis by NaB. Oncogene 2004; 23(37): 6304-15. [81] Duvic M, Talpur R, Ni X, et al. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood 2007; 109(1): 31-9. [82] Khan O, La Thangue NB. HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications. Immunol Cell Biol 2011; 90(1): 85-94. [83] Cang S, Ma Y, Liu D. New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. J Hematol Oncol 2009; 2: 22. [84] Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M, Sinclair DA. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J Biol Chem 2002; 277(47): 45099-107. [85] Lara E, Mai A, Calvanese V, et al. Salermide, a Sirtuin inhibitor with a strong cancer-specific proapoptotic effect. Oncogene 2008; 28(6): 781-91. [86] Mukhopadhyay A, Banerjee S, Stafford LJ, Xia C, Liu M, Aggarwal BB. Curcumin-induced suppression of cell proliferation correlates with down-regulation of cyclin D1 expression and CDK4-mediated retinoblastoma protein phosphorylation. Oncogene 2002; 21(57): 8852-61. [87] Shishodia S, Amin HM, Lai R, Aggarwal BB. Curcumin (diferuloylmethane) inhibits constitutive NF-κB activation, induces G1/S arrest, suppresses proliferation, and induces apoptosis in mantle cell lymphoma. Biochem Pharmacol 2005; 70(5): 700-13. [88] Tan J, Yang X, Zhuang L, et al. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev 2007; 21(9): 1050-63. [89] Greiner D, Bonaldi T, Eskeland R, Roemer E, Imhof A. Identification of a specific inhibitor of the histone methyltransferase SU (VAR) 3-9. Nat Chem Biol 2005; 1(3): 143- 5. [90] Choi J-Y, Lee S, Hwang S, et al. Histone H3 lysine 27 and 9 hypermethylation within the Bad promoter region mediates 5-Aza- 2′-deoxycytidine-induced Leydig cell apoptosis: implications of 5- Aza-2′-deoxycytidine toxicity to male reproduction. Apoptosis 2013; 18(1): 99-109. [91] Portela A, Esteller M. Epigenetic modifications and human disease. Nature Biotechnology. 2010; 28(10): 1057-68. Received: September 17, 2014 Revised: December 3, 2014 Accepted: December 8, 2014