Sie sind auf Seite 1von 12

MINIREVIEW

MicroRNAs and epigenetics


Fumiaki Sato1, Soken Tsuchiya1, Stephen J. Meltzer2 and Kazuharu Shimizu1
1 Department of Nanobio Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan
2 Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA

Keywords MicroRNAs (miRNAs) comprise species of short noncoding RNA that


DNA methylation; epigenetics; histone regulate gene expression post-transcriptionally. Recent studies have demon-
modification; microRNA
strated that epigenetic mechanisms, including DNA methylation and his-
Correspondence
tone modication, not only regulate the expression of protein-encoding
F. Sato, Department of Nanobio Drug genes, but also miRNAs, such as let-7a, miR-9, miR-34a, miR-124, miR-
Discovery, Graduate School of 137, miR-148 and miR-203. Conversely, another subset of miRNAs con-
Pharmaceutical Sciences, Kyoto University, trols the expression of important epigenetic regulators, including DNA
4629 Shimoadachicho Yoshida Sakyoku, methyltransferases, histone deacetylases and polycomb group genes. This
Main Building A320, Kyoto 606-8501, Kyoto, complicated network of feedback between miRNAs and epigenetic path-
Japan
ways appears to form an epigeneticsmiRNA regulatory circuit, and to
Fax: +81 75 753 9557
Tel: +81 75 753 9559
organize the whole gene expression prole. When this regulatory circuit is
E-mail: fsato@pharm.kyoto-u.ac.jp disrupted, normal physiological functions are interfered with, contributing
to various disease processes. The present minireview details recent discover-
(Received 10 November 2010, revised 6 ies involving the epigeneticsmiRNA regulatory circuit, suggesting possible
February 2011, accepted 1 March 2011) biological insights into gene-regulatory mechanisms that may underlie a
variety of diseases.
doi:10.1111/j.1742-4658.2011.08089.x

Introduction
MicroRNAs (miRNA) comprise a class of short non- diseases [12] and carcinogenesis [13]. The biogenesis
coding RNAs with 1825 nucleotides in length that are and RNAi functions of miRNA (i.e. how miRNAs are
found in animal and plant cells. In 1993, the rst miR- generated and processed into a mature form, and how
NAs were recognized in Caenorhabditis elegans by Lee they regulate gene expression) have been intensively
et al. [1]. In 2001, various small regulatory RNAs were investigated and well-described [10]. Furthermore,
discovered in plants and mammals [24] and desig- developments in miRNA-related technologies, such as
nated microRNA [5]. Currently, 1100 human miR- miRNA expression proling and synthetic oligoRNA,
NAs are registered in the miRBase database (release have contributed to the identication of miRNAs
16, September 2010) [69]. miRNAs are involved in involved in a number of physiological and pathological
RNA interference (RNAi) machinery to regulate gene phenotypes. However, some questions remain largely
expression post-transcriptionally, and they contribute unanswered, such as how miRNA expression is con-
to diverse physiological and pathophysiological func- trolled and which genes are regulated by each miRNA.
tions, including the regulation of developmental timing Recently, accumulating studies have shown that a sub-
and pattern formation [2], restriction of differentiation group of miRNAs is regulated epigenetically. Although
potential [10], cell signaling [11], cardiovascular epigenetics and miRNAs have been frequently

Abbreviations
DGCR8, DiGeorge syndrome critical region gene 8; DNMT, DNA methyltransferase; EMT, epithelialmesenchymal transition; HDAC, histone
deacetylase; miRNA, microRNA; NF-jB, nuclear factor kappa B; PRC, polycomb repressor complex; RISC, RNA-induced silencer complex;
RLC, RISC-loading complex; RNAi, RNA interference; SNP, single nucleotide polymorphism; TGIF2, TGFb-inducing factor 2; VNTR, variable
nucleotide tandem repeat; YY1, Yin Yang 1.

1598 FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS
F. Sato et al. MicroRNAs and epigenetics

reviewed [1418], few reviews have focused upon the X mental retardation-related protein 1 [23]. Thus,
relationship between epigenetics and miRNA. In the molecular mechanisms of the RISC in translational
present minireview, we illustrate the current knowledge regulation remain to be claried. At the same time,
regarding the epigeneticsmiRNA regulatory networks turnover of miRNAs is mediated by the XRN2 gene
aiming to provide biological insights for a wide range in C. elegans [24]. However, the mechanisms underly-
of biomedical researchers. ing miRNA turnover in human cells also remain
unclear.
Biogenesis and RNAi functions of
miRNAs Epigenetically-regulated miRNAs
As illustrated in Fig. 1, in the nucleus, mainly RNA As described above, the biogenesis of miRNA has been
polymerase II initially transcribes miRNAs into long intensively studied and is well-described. However, the
segments of coding or noncoding RNA, known as regulation of miRNA expression remains largely
pri-miRNAs, which are usually capped and polyaden- unclear. In early studies, promoter regions had been
ylated. Portions in the pri-miRNAs measuring determined for only a small portion of miRNAs.
approximately 70100 nucleotides in length and con- Although several in silico studies attempted to predict
taining a stem-loop, are captured and extracted from the promoter regions of miRNAs [2527], most of
pri-miRNAs by a complex containing RNase type III, these predicted miRNA promoters were not conrmed
Drosha and the dsRNA binding protein DiGeorge in wet-laboratory experiments.
syndrome critical region gene 8 (DGCR8) (also called miRNAs can be classied as either intragenic and
Pasha) [19]. These short stem-loop-shaped RNAs are intergenic, according to whether the miRNA is local-
called pre-miRNAs, and the protein complex of ized in a genome region transcribed by a gene, or not.
RNase, Drosha and DGCR8 is known as the micro- Our in silico analysis (see Materials and methods)
processor complex. Pre-miRNAs form a complex with revealed that, among 939 miRNAs, 293 (31.2%) of
exportin-5 and RAN-GTP, and are then exported miRNAs were intergenic, whereas 317 (44.4%), 119
from the nucleus to the cytoplasm. The pre-miRNAs (12.7%) and 110 (11.7%) were overlapped by RNA
are further processed to a double-stranded miRNA transcripts in the same, opposite and both directions,
duplex by a dsRNase type III, Dicer. This double- respectively. Localization of promoters for intergenic
stranded miRNA duplex is incorporated into a RNA- and inversely-directed intragenic miRNAs is largely
induced silencer complex (RISC)-loading complex unknown, whereas promoters for overlapping primary
(RLC) in an ATP-dependent manner [20]. Next, one genes are considered to be promoters for the intragenic
strand (the passenger strand) of the miRNA is miRNAs that are localized in the same direction as the
removed from the RLC, whereas the other strand (the primary gene. However, some studies have identied
guide strand) remains in the complex to form a that an independent promoter within the intron in
mature RNA-induced silencer complex (mature RISC) which a miRNA is embedded can also regulate miR-
and serves as a template for capturing target mRNAs. NA expression [28]. Additionally, as shown in one
Under most conditions, the mature RISC represses study [29], a single member of a miRNA cluster,
gene expression post-transcriptionally. For highly although ordinarily expressed from the same pri-miR-
complementary target mRNAs, the mature RISC NA, can alternatively be regulated independently by its
complex cleaves target mRNAs via a catalytic domain own promoter in certain scenarios. Furthermore, the
(RNase III domain) of Argonaute proteins, a core total amount of miRNAs contained within a given
component of the RISC complex, and degrades them quantity of total RNA can be reduced in cancer cells
by the SKI complex and XRN1 [21]. For partially and rapidly proliferating cells [13,30], a nding for
complementary targets, the RISC complex decaps and which the underlying mechanism is still unknown.
deadenylates target mRNAs via the DCP1-DCP2 and Thus, the means by which miRNA expression is regu-
CAF1-CCR4-NOT complexes, respectively, to reduce lated appears somewhat complicated.
the stability of the target mRNAs [22]. In addition, Recently, Saito et al. [29] established that the expres-
the RISC complex also represses the translation of sion of miR-127 is regulated epigenetically. In their
target genes under most conditions. However, not all study, pharmacological unmasking of epigenetically
miRNAs work in translational repression. Under silenced miRNAs activated 17 of 313 miRNAs investi-
serum-starved conditions, miR-369-3 activates transla- gated in the bladder cancer cell line T24 and the nor-
tion of tumor necrosis factor-a by binding to AU-rich mal broblast cell line LD419. The gene for miR-127
elements in the 3 UTR of the transcript with fragile was upregulated the most in epigenetically unmasked

FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS 1599
MicroRNAs and epigenetics F. Sato et al.

Fig. 1. EpigeneticsmiRNA regulatory circuit. Epigenetics and miRNAs regulate whole gene expression pattern transcriptionally and post-
transcriptionally, respectively. At the same time, epigenetics and miRNAs controll each other to form a regulatory circuit and to maintain nor-
mal physiological functions. A disruption of this regulatory circuit may cause various diseases, such as cardiovascular diseases and cancers.
PABP, poly(A) binding protein; TF, transcriptional factors; TRBP, Tar RNA binding protein.

cancer cells. DNA methylation level and histone modi- acute lymphoblastic leukemia [3134]. In breast cancer,
cation status at identied promoter regions of the miR-9-1 locus is highly methylated not only in
miR-127 correlated signicantly with mature miR-127 invasive ductal carcinoma, but also in ductal carci-
expression. Subsequent to this initial report, the num- noma in situ and the intraductal component of invasive
ber of studies documenting the epigenetic regulation of ductal carcinoma [34]. In addition, an in vitro experi-
miRNAs has increased dramatically (Table 1). We mental study showed that xenoestrogen exposure may
summarize the ndings regarding some of the more induce aberrant epigenetic patterns at various miRNA
intensively studied miRNAs for which expression is gene loci, including miR-9-3 [35]. These ndings sug-
regulated by epigenetic mechanisms. gest that epigenetic silencing of miR-9 loci constitutes
an early event in breast carcinogenesis. Furthermore,
the miR-9 DNA methylation signature is correlated
miR-9
with cancer metastasis [33]. Target genes of mature
miR-9 is expressed from three genomic loci, miR-9-1, miR-9 responsible for carcinogenesis and cancer metas-
miR-9-2 and miR-9-3, all of which are associated with tasis remain largely unknown. However, a recent study
CpG islands. Hypermethylation of miR-9 loci is demonstrated that mature miR-9 targets nuclear factor
observed in various malignant tissues, including breast, kappa B (NF-jB), which is overexpressed in a number
lung, colon, head and neck cancers, melanoma and of different cancers [36].

1600 FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS
F. Sato et al. MicroRNAs and epigenetics

Table 1. Epigenetically-regulated miRNAs. The numbers in the binding sites column represent the distance (bp) between the stop codon
and binding sites of seed sequences in the miRNAs. The letters c and p with respect to miRNA binding site numbers indicate that the
miRNA binding sites are the conserved region and poorly conserved region among vertebrates, according to the TargetScan database
(http://www.targetscan.org ).

miRNA genes Inter- intra genic Locus Host gene Target genes Binding sites References

let-7a-3 Intergenic 22q13.31 IGF2BP1-3 1632c, 1651c, 4269c, [55,83]


4923c, 5568c
miR-1-1 Intragenic 20q13.33 C20orf166 FoxP1 772c, 819ca, 965p, 3447p [84]
MET 499p, 811c
HDAC4 2333c, 3513ca,3546ca
miR-9-1 Intragenic 1q22 C1orf61 NFKB1 29pa [3135]
miR-9-2 Intragenic 5q14.3 CR612213
miR-9-3 Intragenic 15q26.1 FLJ30369
miR-10a Intragenic 21q21.32 HOX3B HOXA3 299c [31,85]
HOXD10 276c
miR-34a Intragenic 1p36.23 EF570048 CDK6 1087c, 6941p, 9172c [39]
miR-34b c Intragenic 11q23.1 BC021736 CDK6 1087c, 6941p, 9172c [28,31,33,40,41]
Intragenic MYC 138pa
E2F3 2714c
CREB 3259p, 3317c
miR-107 Intragenic 10q23.31 PANK1 CDK6 308c, 1815p [86]
miR-124-1 Intergenic 8p23.1 1532p, 1647p, 7788p, 8004p [31,34,4448]
miR-124-2 Intragenic 8q12.3 AK124256 CDK6
miR-124-3 Intergenic 20q13.33 FLJ42262 C EBPa 283c, 340c, 981c
VIM 81c
SMYD3 43p
miR-126 Intragenic 9q34.3 EGFL7 [87]
miR-127 Intergenic 14q32.31 BCL6 584c [29,47]
miR-129-2 Intragenic 11p11.2 EST [32]
miR-132 212 Intergenic 17p13.2 [31]
miR-137 Intragenic 1p21.3 AK311400 CDK6 4214p, 7114p, 7133c [32,40,47]
E2F6 79c
NCOA2 1244c
miR-148a Intergenic 7p15.2 TGIF2 159c, 566pa, 2288c [33,34]
miR-152 Intragenic 17q21.32 COPZ2 [34]
miR-181a b-2 Intragenic 9q33.3 NR6A1 PLAG1 391p, 3501c, 4389c [88]
miR-193a Intergenic 17q11.2 E2F6 127c [40,47]
PTK2 545p
MCL1 315ca
miR-196a-2 Intragenic 12q13.13 EST [89]
miR-196b Intragenic 7p15.2 EST [31]
miR-199a*-1 Intragenic 19p13.2 DNM2 MET 1425ca [90]
miR-199a*-2 Intragenic 1q24.3 DNM3
miR-141 200c Intergenic 12p13.31 ZEB2 207c, 733p, 774c [5153]
miR-200a b 429 Intergenic 1p36.33 ZEB1 369c, 463c
ZEB2 391ca, 454ca, 812c, 897c,
1028c, 1362ca
SOX2 477ca
KLF4 42ca
miR-203 Intergenic 14q32.33 ABL1 1074c [31,40,48,51,54]
BCR-ABL1 1074c
Bmi-1 1443c
miR-342 Intragenic 14q32.2 EVL [91]
miR-370 Intragenic 14q32.31 EST MAP3K8 567p [92]
miR-512-5p Intergenic 19q13.41 Mcl-1 1631p [93]
miR-663 Intragenic 20p11.1 BC036544 [34]
a
SNPs are located within the miRNA binding sites (not only the seed sequence regions, but also an approximately 23 bp region), which may
affect the affinity of miRNA with the binding sites.

FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS 1601
MicroRNAs and epigenetics F. Sato et al.

loci are also hypermethylated in precancerous lesions.


miR-34 (a and b c)
Methylation levels at miR-124 loci in the gastric muco-
The net level of miR-34 reects the expression of three sae of healthy volunteers infected by Helicobact-
separate genes for miR-34: miR-34a, miR-34b and er pylori are markedly elevated compared to healthy
miR-34c. miR-34a is monocistronic, whereas miRs- individuals without H. pylori infection [47]. Thus,
34b c are polycistronic. Promoter regions of both loci H. pylori infection appears to induce aberrant epige-
contain p53-binding sites, and are regulated by the p53 netic patterns at miRNA loci in normal gastric muco-
signal. Likely as a result of this feature, the expression sae, which may contribute to gastric carcinogenesis as
of mature miR-34a species is induced by DNA damage a eld effect. Targets of mature miR-124 include the
and oncogenic stress, as well as other p53-related 3 UTR of CDK6, an oncogene. Epigenetically mask-
events that control the cell cycle, induce apoptosis and ing of miR-124 induces activation of CDK6 and conse-
suppress tumor formation [37,38]. The host or mother quent phosphorylation of Rb at serine residues 807
gene (FLJ41150) of miR-34a is associated with a CpG and 811, the targets of CDK6, resulting in an accelera-
island surrounding its transcriptional start site, which tion of cell growth. Notably, in acute lymphoblastic
is frequently methylated in various malignancies [39]. leukemia, epigenetic silencing of miR-124 loci is linked
The epigenetic mechanism underlying miR-34b c tran- to both disease-free and overall survival [31].
scriptional regulation was described in detail by Toy-
ota et al. [28]. The miR-34b c host gene (BC021736)
miR-137
contains a CpG island, not within its own promoter
region, but also located at the rst intronsecond exon Physiologically, miR-137 is involved in neurogenesis
boundary. The latter CpG island also happens to lie by targeting CDK6, analogous to miR-124 [43], as well
within the promoter region of the oppositely-oriented as in melanocyte function by targeting microphthal-
BTG4 gene, thus exerting bidirectional promoter activ- mia-associated transcription factor [49]. miR-137 is an
ity for both the BTG4 gene and the miR-34b c polycis- intragenic miRNA that is directly overlapped by a
toron [28]. Thus, miR-34b c expression may be CpG island. The CpG island is specically hyperme-
regulated by both the promoter of the host gene and thylated in cancer tissues [32,40,47]. Overexpression of
the promoter in the latter CpG island. The methyla- miR-137 in cancer cells induces cell cycle G1 arrest
tion levels of the CpG island are inversely correlated and apoptosis [40]. Furthermore, a 15 nucleotide
with mature miR-34b c expression levels in various variable tandem repeat (VNTR) (5-TAGCAGCGGC
cancers [28,31,33,40,41]. In colorectal cancer cell lines, AGCGG-3) is located just 5 to pre-miR-137, and
in which the miR-34b c locus is epigenetically silenced, extending the length of this VNTR impairs the matu-
the p53 signal alone does not induce miR-34b c ration of miR-137. Specically, pri-miR-137 with three
expression [28]. This nding suggests that hypermethy- VNTRs is more efciently processed to mature miR-
lation of the CpG island modulates p53-mediated 137 than is pri-miR-137 with 12 VNTRs. Thus, both
miR-34b c expression. In terms of the functions of genomic and epigenetic variations affect mature miR-
miR-34 species, mature miR-34 miRNAs target vari- 137 expression levels and may contribute to disease
ous genes related to the cell cycle, oncogenesis and formation.
cancer metastasis, including MYC, CDK4, CDK6,
E2F3, CREB and MET [33,37,41]. Ectopic expression
miR-148
of miR-34 species induces cell-cycle arrest and apopto-
sis and suppresses cell growth and metastasis, possibly Lujambio et al. [33] screened cancer metastasis-related
by silencing these target genes [28,33,37,3941]. miRNAs that are epigenetically inactivated, using a
pharmacological epigenetic reversal technique in meta-
static cancer cell lines, which identied three miRNAs,
miR-124
one of which is miR-148. The miR-148 locus is more
Many studies have shown that mature miR-124 is the heavily methylated in metastatic than in non-metastatic
most abundant miRNA in the adult brain, and that it cancer tissues. Cancer cells that stably express exoge-
plays a key role in neurogenesis [42]. Conversely, nous miR-148 exhibit reduced invasiveness, cell motility
epigenetic silencing of three miR-124 loci (miR-124-1 and metastatic propensity in an in vivo model [33]. In
to -3) is frequently observed not only in brain tumors, addition, miR-148 targets TGFb-inducing factor 2
but also in a variety of other cancer types [4348], such (TGIF2), which is overexpressed in highly malignant
as colon (prevalence: 75%), breast (3250%), lung ovarian cancers [50]. Thus, epigenetic inactivation
(48%), leukemia (36%) and lymphoma (41%). miR-124 of miR-148 would be expected to enhance TGIF2

1602 FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS
F. Sato et al. MicroRNAs and epigenetics

activation. In addition, several isoforms of DNA methy- as a result of polycomb-mediated modication in epi-
transferase (DNMT)3b are targeted by miR-148 within genetic patterns.
their coding region (described in detail below). There-
fore, although being targeted epigenetically, miR-148
let-7a-3
may itself exert effects on DNA methylation in cells.
Epigenetic control of let-7a-3 expression was discovered
by a comparison between parent and DNMT1-3B dou-
The miR-200 family
ble-knockout HCT116 colon cancer cells [55]. The let-
The miR-200 family consists of miR-141, 200a b c and 7a-3 locus is generally methylated in normal tissues but
429, which share similar seed sequences. miRs- hypomethylated in some types of cancers, such as colon
141 200c and miRs-200a b 429 comprise multicistronic and lung cancer [55]. Methylation levels of let-7a-3
miRs whose genomic loci are located in close proximity correlate inversely with let-7a-3 pri-miRNA expression
to each other. Several studies have established that the levels [55]. However, the effect of let-7a-3 methylation
miR-200 family is involved in epithelialmesenchymal status on mature let-7a expression level is unclear
transition (EMT). EMT occurrence in cancer cells com- because levels of mature let-7a reect the expression of
prises a phenomenon in which these cells obtain pheno- three let-7a genes, let-7a-1, let-7a-2 and let-7a-3.
types characteristic of mesenchymal cells, such as Indeed, let-7a-3 methylation levels in ovarian cancer
spindle-shaped morphology, activated cell motility and correlate with mature let-7a levels. In the context of
invasiveness. Therefore, EMT research is important for miRNA function, let-7a-3 has oncogenic potential. The
understanding the molecular mechanisms underlying introduction of let-7a-3 enhanced the colony-forming
the malignant potential of cancer cells. Recently, Well- ability of A549 lung adenocarcinoma cells. In addition,
ner et al. [51] demonstrated that an EMT activator, let-7a may regulate IGF-II via targeting of IGF2-bind-
ZEB1, suppresses miR-200c, whereas miR-200c targets ing proteins (IMP-1 and 2). Methylation levels at the
ZEB1. This nding suggests that miR200c and ZEB1 let-7a-3 locus correlate inversely with IGF-II levels,
form a feedback loop regulatory mechanism that main- and are also linked to the survival of ovarian cancer
tains EMT [51]. Additional studies showed that both patients. In general, the let-7 family is considered to
the miR-141 200c [52,53] and miR-200a b 429 [53] comprise tumor suppressor miRNAs [5658]. Diversity
clusters are epigenetically regulated. Thus, EMT could in functions among let-7 family members may cause
conceivably be regulated by epigenetic events targeting apparently contradictory observations.
the miR-200 family. Table 1 shows that miR-
200a b 429 binding sites in the 3 UTR of ZEB2 have
Imprinting and miRNAs
several single nucleotide polymorphism (SNP) sites.
However, to date, no study is available demonstrating Genomic imprinting is an epigenetic process by which
the clinical signicance of these SNPs. a small proportion of genes (< 1% of all genes in
mammals) are expressed in a parent-of-origin-specic
manner [59]. In genomic imprinting, DNA methylation
miR-203
and histone modication regulate monoallelic expres-
In hematopoietic malignancies, 12% of miRNAs are sion. These epigenetic patterns are established in germ-
located in fragile genomic regions that encompass only line cells, and are inherited through somatic cells. For
seven megabases (0.2% of whole genome). miR-203 is example, at the well-investigated IGF2 H19 locus, the
one of these regions, and it targets ABL1 and BCR- IGF2 gene is expressed from the paternal allele,
ABL1, an oncogenic fusion gene generated by the Phil- whereas the H19 gene is expressed from the maternal
adelphia translocation [54]. Epigenetic silencing of allele. Abnormal genomic imprinting is associated with
miR-203 enhances activation of the BCR-ABL1 fusion several diseases. Some inheritable disorders, such as
gene, resulting in an elevation of tumor cell growth PraderWilli syndrome and Angelman syndrome, are
rate. Epigenetic inactivation of miR-203 is frequently caused by aberrant imprinting. Furthermore, the phe-
observed in other types of malignancies, including oral nomenon known as loss of imprinting, in which the
cancer, hepatocellular carcinoma, etc. [40,48]. Another normally inactivated allele becomes reactivated as a
candidate target gene of miR-203 is Bmi-1, a member result of hypomethylation or histone abnormalities, is
of the polycomb repressor complex 1 [51], which is a frequently observed in cancers [60].
histone modier complex regulating gene expression. Several miRNAs are located within imprinting-asso-
Introduction of ectopic miR-203 into cancer cells ciated regions, including miR-296 and miR-298 at the
induces apoptosis and represses cell growth [48], possibly GNAS NESP locus, miR-483 and miR-675 at the

FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS 1603
MicroRNAs and epigenetics F. Sato et al.

IGF2 H19 locus, and miR-335, miR-29a and miR-29b corresponding normal tissues, with the highest EZH2
at the MEST KLF14 locus [61]. However, the imprint- levels correlating with advanced disease stages and
ing and expression status of such miRNAs remains lar- poor prognosis. In some cases, EZH2 overabundance
gely unknown. is paralleled by DNA amplication of the gene [63]. A
second mechanism of EZH2 overexpression is post-
transcriptional regulation by miRNAs. EZH2 expres-
miRNAs regulating epigenetic pathway-
sion is controlled by miR-26a, miR-101, miR-205 and
related genes
miR-214 [6468]. Cancer-specic downregulation of
miRNAs themselves are capable of targeting genes that these miRNAs results in overexpression of EZH2.
control epigenetic pathways. As shown in Table 2, var-
ious miRNAs may control chromatin structure by reg-
Bmi-1
ulating histone modier molecules, such as polycomb
group-related genes and histone deacetylase (HDAC). In a subsequent step, PRC2 and the H3K27 methyla-
The polycomb group proteins are transcriptional tion recruit PRC1 binding to chromatin to maintain
repressors that regulate lineage choices occurring dur- stable gene silencing. PRC1 catalyzes ubiquitinylation
ing development and differentiation. There are two of histone H2A and remains anchored to chromatin
polycomb repressor complexes (PRCs), PRC1 and after its modication by the cooperation between
PRC2. The PRC1 core complex contains Cbx, Mph, PRC2 and PRC1. Bmi-1, a component of PRC1, plays
Ring, Bmi-1 and Me118, whereas the PCR2 core com- an important role in gene silencing and is overexpres-
plex consists of Ezh2, Suz12 and Eed [62]. In an initial sed in several cancers, including nonsmall cell lung
step, PRC2 initiates silencing by catalyzing histone H3 cancer and colorectal cancer. Bmi-1 overexpression
Lysine-27 (H3K27) methylation. Recent studies have contributes to self-renewal in some types of cancer
advanced our understanding of the means by which stem cells, including those of the pancreas [69], breast
epigenomic dysregulation potentially contributes to [70], brain [71] and white blood cell lineage [72].
various diseases. Downregulation of miR-128 in glioma tissue causes
elevated expression of Bmi-1, which consequently
enhances self-renewal of the cancer stem cell popula-
EZH2
tion via chromatin remodeling [71]. In addition,
Expression levels of EZH2, a conserved catalytic sub- recently, Wellner et al. [51] recently demonstrated that
unit within PRC2, are elevated in cancers relative to an EMT-related miRNA, miR-203, targets Bmi-1. This
nding suggests that EMT mechanisms include the reg-
Table 2. miRNAs targeting genes that are involved in epigenetic ulation of epigenetic regulators by miRNAs.
regulatory pathways. The letters c and p with respect to miRNA
binding site numbers indicate that the miRNA binding sites are the
conserved region and poorly conserved region among verte- Yin Yang 1 (YY1)
brates, according to the TargetScan database (http://www.target-
scan.org ). YY1 is a transcription factor that contributes to vari-
ous biological processes, including embryogenesis, the
Target genes miRNAs Binding sites References cell cycle, apoptosis, inammation, carcinogenesis and
EZH2 miR-26a 249c [6466,68] epigenetics. In the epigenetic context, YY1 is a PRC-
miR101 58p, 113ca binding protein that recruits PRC2 and HDAC to a
miR-214 172p specic genome locus to induce chromatin remodeling.
Bmi1 miR-128 481c [51,71] NF-jB-mediated miR-29b c repression reactivates
miR-203 1443c YY1 protein expression from post-transcriptional
YY1 miR-29b 774c [73]
silencing induced by these two miRs. In addition, YY1
HDAC1 miR-449 459p [94]
HDAC4 miR-1 2333c, 3513ca, 3546ca [95]
also represses miR-29b c. This NF-jB-miR-29-YY1
DNMT3A miR-29 855c [79,80] regulatory circuit is also involved in myogenesis and
DNMT3B miR-29 1202c [7981] tumorigenesis, probably via chromatin remodeling [73].
miR-148 1424c and 2384c in
coding region
MeCP2 miR-132 6886c [96] HDACs
a
SNPs are located within the miRNA binding sites (not only the In human cells, PRC2 physically associates with
seed sequence regions, but also an approximately 23 bp region), HDACs 1 and 2 [74]. If H3K27 is pre-acetylated,
which may affect the affinity of miRNA with the binding sites. methylation at an H3K27 residue by PRC2 may

1604 FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS
F. Sato et al. MicroRNAs and epigenetics

require deacetylation by HDACs. Thus, both acetyla- indicates that miRNAs can regulate gene expression
tion and deacetylation of histones is involved in the uniquely among different gene isoforms by targeting a
transcriptional regulation of target genes. In addition, coding exon.
recent studies have demonstrated that HDACs target As described above and illustrated in Fig. 1, a num-
not only histone proteins, but also nonhistone pro- ber of miRNAs are regulated epigenetically. At the
teins: p53 and Myo-D are targeted by HDAC-1, same time, a variety of miRNAs regulate epigenetic
whereas Bcl-6, Stat3 and YY1 are targeted by HDAC- pathway-related molecules, most notably polycomb
2. By regulating both histone and nonhistone proteins, group proteins, HDACs and DNA methyltransferases.
HDACs 1 and 2, classied as class I HDACs, are Taken together, post-transcriptional regulation by
implicated in cell proliferation, apoptosis and chemore- miRNAs and transcriptional control machinery by epi-
sistance. The expression of HDACs 1 and 2 is elevated genetics cooperate with each other to organize the
in various cancers [75]. However, the mechanism of whole gene expression prole and to maintain physio-
HDAC overexpression remains unclear. Dysregulation logical functions in cells. Once this miRNAepigenetics
of miRNAs may contribute to the overexpression of regulatory circuit is disrupted, normal physiological
HDACs observed in cancer cells. In prostate cancer, functions are interfered with, contributing to various
HDAC-1 is a direct target of miR-449a, and downre- disease processes. A comprehensive elucidation of this
gulation of miR-449a causes overexpression of regulatory network still remains to be completed.
HDAC-1. Thus, aberrant expression of miR-449a may Therefore, continual studies on dysregulation of the
contribute to the abnormal epigenetic patterns occur- miRNAepigenetics regulatory circuitry would be
ring in prostate cancer. highly benecial for deepening our understanding of
diseases.
DNMT 3A and 3B
Materials and methods
DNMTs 1, 3A, and 3B are key DNA methylation
enzymes. Recent studies in human cells have demon-
Typing of miRNAs by positional relationship to
strated that PRC2 and DNMTs are physically and
mRNA transcripts
functionally linked [76], and that DNMT-mediated
DNA methylation lies downstream of PRC2-mediated Information about the localization and strand direction of
H3K27 methylation [76,77]. Thus, these two key epige- 939 miRNAs, 35245 Refseq genes and 283708 mRNAs
netic repression systems cooperate in the silencing of was retrieved from the genome browser of University of
target genes. Dysregulation of DNMTs has been California Santa Cruz [82] on 31 January 2011. Because the
linked to various disease processes, including cancer original data table of refseq genes included miRNA genes,
and congenital disorders. These DNMTs are predicted these miRNA data were excluded from the Refseq data set.
to be potential targets of miRNAs [78]. Fabbri et al. Using matlab, version 2011a (Mathworks, Natick, MA,
[79] showed that members of the miR-29 family USA), we compared localization and strand direction
between miRNAs and transcripts (Refseq genes and
directly target DNMTs 3A and 3B, and that exoge-
mRNAs). Intragenic and intergenic miRNAs were dened
nous miR-29 species can reactivate methylation-
by whether the miRNAs are overlapped by transcripts, or
silenced tumor suppressor genes by restoring normal
not, respectively. In addition, intragenic miRNAs were
patterns of DNA methylation in nonsmall cell lung
divided into three different types, which are overlapped by
cancer cells. Another study reported similar ndings in
transcripts only in the same strand direction, only in oppo-
acute myeloid leukemia [80]. Thus, miRNAs may be site direction, or in both directions, respectively. The com-
involved in the establishment and or maintenance of plete results of this typing analysis are provided in
DNA methylation. In addition, some isoforms of Table S1.
DNMT3B are targeted at the penultimate exon of their
coding regions by miR-148 [81]. DNMT3B exhibits
several splicing isoforms, of which DNMT3B-1 and -3 References
are the most abundant. DNMT3B-1 possesses a cata- 1 Lee RC, Feinbaum RL & Ambros V (1993) The
lytic domain and a miR-148 target site. Thus, C. elegans heterochronic gene lin-4 encodes small
DNMT3B-1 is a miR-148-sensitive isoform. By con- RNAs with antisense complementarity to lin-14. Cell
trast, DNMT3B-3 lacks a catalytic domain and the 75, 843854.
miR-148 target site, and remains miR-148 resistant. 2 Lagos-Quintana M, Rauhut R, Lendeckel W & Tuschl
The biological roles of different DNMT3B isoforms T (2001) Identication of novel genes coding for small
are not yet fully understood. However, this nding expressed RNAs. Science 294, 853858.

FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS 1605
MicroRNAs and epigenetics F. Sato et al.

3 Lau NC, Lim LP, Weinstein EG & Bartel DP (2001) enylase and DCP1:DCP2 decapping complexes. Genes
An abundant class of tiny RNAs with probable regula- Dev 20, 18851898.
tory roles in Caenorhabditis elegans. Science 294, 858 23 Vasudevan S, Tong Y & Steitz JA (2007) Switching
862. from repression to activation: microRNAs can up-regu-
4 Lee RC & Ambros V (2001) An extensive class of small late translation. Science 318, 19311934.
RNAs in Caenorhabditis elegans. Science 294, 862864. 24 Chatterjee S & Grosshans H (2009) Active turnover
5 Ambros V, Bartel B, Bartel DP, Burge CB, Carrington modulates mature microRNA activity in Caenorhabd-
JC, Chen X, Dreyfuss G, Eddy SR, Grifths-Jones S, itis elegans. Nature 461, 546549.
Marshall M et al. (2003) A uniform system for 25 Wang X, Xuan Z, Zhao X, Li Y & Zhang MQ (2009)
microRNA annotation. RNA 9, 277279. High-resolution human core-promoter prediction with
6 Bartel DP (2004) MicroRNAs: genomics, biogenesis, CoreBoost_HM. Genome Res 19, 266275.
mechanism, and function. Cell 116, 281297. 26 Zhou X, Ruan J, Wang G & Zhang W (2007) Charac-
7 Grifths-Jones S (2004) The microRNA Registry. terization and identication of microRNA core promot-
Nucleic Acids Res 32, D109D111. ers in four model species. PLoS Comput Biol 3, e37.
8 Kim VN & Nam JW (2006) Genomics of microRNA. 27 Corcoran DL, Pandit KV, Gordon B, Bhattacharjee A,
Trends Genet 22, 165173. Kaminski N & Benos PV (2009) Features of mamma-
9 Zamore PD & Haley B (2005) Ribo-gnome: the big lian microRNA promoters emerge from polymerase II
world of small RNAs. Science 309, 15191524. chromatin immunoprecipitation data. PLoS ONE 4,
10 Nakahara K & Carthew RW (2004) Expanding roles e5279.
for miRNAs and siRNAs in cell regulation. Curr Opin 28 Toyota M, Suzuki H, Sasaki Y, Maruyama R, Imai K,
Cell Biol 16, 127133. Shinomura Y & Tokino T (2008) Epigenetic silencing of
11 Ichimura A, Ruike Y, Terasawa K & Tsujimoto G microRNA-34b c and B-cell translocation gene 4 is
(2011) MicoRNAs and regulation of cell signaling. associated with CpG island methylation in colorectal
FEBS J 278, 16101618. cancer. Cancer Res 68, 41234132.
12 Ono K, Kuwabara Y & Han J (2011) MicroRNAs 29 Saito Y, Liang G, Egger G, Friedman JM, Chuang JC,
and cardiovascular diseases. FEBS J 278, 16191633. Coetzee GA & Jones PA (2006) Specic activation of
13 Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, microRNA-127 with downregulation of the proto-onco-
Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferran- gene BCL6 by chromatin-modifying drugs in human
do AA et al. (2005) MicroRNA expression proles cancer cells. Cancer Cell 9, 435443.
classify human cancers. Nature 435, 834838. 30 Sato F, Tsuchiya S, Terasawa K & Tsujimoto G (2009)
14 Berdasco M & Esteller M (2010) Aberrant epigenetic Intra-platform repeatability and inter-platform compa-
landscape in cancer: how cellular identity goes awry. rability of microRNA microarray technology. PLoS
Dev Cell 19, 698711. ONE 4, e5540.
15 Taby R & Issa J-PJ (2010) Cancer epigenetics. CA 31 Roman-Gomez J, Agirre X, Jimenez-Velasco A,
Cancer J Clin 60, 376392. Arqueros V, Vilas-Zornoza A, Rodriguez-Otero P,
16 Guil S & Esteller M (2009) DNA methylomes, histone Martin-Subero I, Garate L, Cordeu L, San Jose-Eneriz
codes and miRNAs: tying it all together. Int J Biochem E et al. (2009) Epigenetic regulation of microRNAs in
Cell Biol 41, 8795. acute lymphoblastic leukemia. J Clin Oncol 27, 1316
17 Chuang JC & Jones PA (2007) Epigenetics and microR- 1322.
NAs. Pediatr Res 61, 24R29R. 32 Bandres E, Agirre X, Bitarte N, Ramirez N, Zarate R,
18 Veeck J & Esteller M (2010) Breast cancer epigenetics: Roman-Gomez J, Prosper F & Garcia-Foncillas J
from DNA methylation to microRNAs. J Mammary (2009) Epigenetic regulation of microRNA expression
Gland Biol Neoplasia 15, 517. in colorectal cancer. Int J Cancer 125, 27372743.
19 Kim VN, Han J & Siomi MC (2009) Biogenesis of 33 Lujambio A, Calin GA, Villanueva A, Ropero S, San-
small RNAs in animals. Nat Rev Mol Cell Biol 10, 126 chez-Cespedes M, Blanco D, Montuenga LM, Rossi S,
139. Nicoloso MS, Faller WJ et al. (2008) A microRNA
20 Yoda M, Kawamata T, Paroo Z, Ye X, Iwasaki S, Liu DNA methylation signature for human cancer
Q & Tomari Y (2010) ATP-dependent human RISC metastasis. Proc Natl Acad Sci USA 105, 1355613561.
assembly pathways. Nat Struct Mol Biol 17, 1723. 34 Lehmann U, Hasemeier B, Christgen M, Muller M,
21 Orban TI & Izaurralde E (2005) Decay of mRNAs tar- Romermann D, Langer F & Kreipe H (2008) Epigenetic
geted by RISC requires XRN1, the Ski complex, and inactivation of microRNA gene hsa-mir-9-1 in human
the exosome. RNA 11, 459469. breast cancer. J Pathol 214, 1724.
22 Behm-Ansmant I, Rehwinkel J, Doerks T, Stark A, 35 Hsu PY, Deatherage DE, Rodriguez BA, Liyanarachchi
Bork P & Izaurralde E (2006) mRNA degradation by S, Weng YI, Zuo T, Liu J, Cheng AS & Huang TH
miRNAs and GW182 requires both CCR4:NOT dead- (2009) Xenoestrogen-induced epigenetic repression of

1606 FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS
F. Sato et al. MicroRNAs and epigenetics

microRNA-9-3 in breast epithelial cells. Cancer Res 69, 47 Ando T, Yoshida T, Enomoto S, Asada K, Tatematsu
59365945. M, Ichinose M, Sugiyama T & Ushijima T (2009) DNA
36 Guo LM, Pu Y, Han Z, Liu T, Li YX, Liu M, Li X & methylation of microRNA genes in gastric mucosae of
Tang H (2009) MicroRNA-9 inhibits ovarian cancer cell gastric cancer patients: its possible involvement in the
growth through regulation of NF-kappaB1. FEBS J formation of epigenetic eld defect. Int J Cancer 124,
276, 55375546. 23672374.
37 He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang 48 Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I & In-
Y, Xue W, Zender L, Magnus J, Ridzon D et al. (2007) azawa J (2010) miR-124 and miR-203 are epigenetically
A microRNA component of the p53 tumour suppressor silenced tumor-suppressive microRNAs in hepatocellu-
network. Nature 447, 11301134. lar carcinoma. Carcinogenesis 31, 766776.
38 Chang TC, Wentzel EA, Kent OA, Ramachandran K, 49 Bemis LT, Chen R, Amato CM, Classen EH, Robinson
Mullendore M, Lee KH, Feldmann G, Yamakuchi M, SE, Coffey DG, Erickson PF, Shellman YG & Robin-
Ferlito M, Lowenstein CJ et al. (2007) Transactivation son WA (2008) MicroRNA-137 targets microphthalmia-
of miR-34a by p53 broadly inuences gene expression associated transcription factor in melanoma cell lines.
and promotes apoptosis. Mol Cell 26, 745752. Cancer Res 68, 13621368.
39 Lodygin D, Tarasov V, Epanchintsev A, Berking C, 50 Imoto I, Pimkhaokham A, Watanabe T, Saito-Ohara F,
Knyazeva T, Korner H, Knyazev P, Diebold J & Her- Soeda E & Inazawa J (2000) Amplication and overex-
meking H (2008) Inactivation of miR-34a by aberrant pression of TGIF2, a novel homeobox gene of the
CpG methylation in multiple types of cancer. Cell Cycle TALE superclass, in ovarian cancer cell lines. Biochem
7, 25912600. Biophys Res Commun 276, 264270.
40 Kozaki K, Imoto I, Mogi S, Omura K & Inazawa J 51 Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu
(2008) Exploration of tumor-suppressive microRNAs F, Sonntag A, Waldvogel B, Vannier C, Darling D, zur
silenced by DNA hypermethylation in oral cancer. Can- Hausen A et al. (2009) The EMT-activator ZEB1 pro-
cer Res 68, 20942105. motes tumorigenicity by repressing stemness-inhibiting
41 Pigazzi M, Manara E, Baron E & Basso G (2009) microRNAs. Nat Cell Biol 11, 14871495.
miR-34b targets cyclic AMP-responsive element binding 52 Vrba L, Jensen TJ, Garbe JC, Heimark RL, Cress AE,
protein in acute myeloid leukemia. Cancer Res 69, Dickinson S, Stampfer MR & Futscher BW (2010) Role
24712478. for DNA methylation in the regulation of miR-200c
42 Cao X, Pfaff SL & Gage FH (2007) A functional study and miR-141 expression in normal and cancer cells.
of miR-124 in the developing neural tube. Genes Dev PLoS ONE 5, e8697.
21, 531536. 53 Wiklund ED, Bramsen JB, Hulf T, Dyrskjot L, Rama-
43 Silber J, Lim DA, Petritsch C, Persson AI, Maunakea nathan R, Hansen TB, Villadsen SB, Gao S, Ostenfeld
AK, Yu M, Vandenberg SR, Ginzinger DG, James CD, MS, Borre M et al. (2011) Coordinated epigenetic
Costello JF et al. (2008) miR-124 and miR-137 inhibit repression of the miR-200 family and miR-205 in inva-
proliferation of glioblastoma multiforme cells and sive bladder cancer. Int J Cancer 128, 13271334.
induce differentiation of brain tumor stem cells. BMC 54 Bueno MJ, Perez de Castro I, Gomez de Cedron M,
Med 6, 14. Santos J, Calin GA, Cigudosa JC, Croce CM, Fernan-
44 Agirre X, Vilas-Zornoza A, Jimenez-Velasco A, Martin- dez-Piqueras J & Malumbres M (2008) Genetic and epi-
Subero JI, Cordeu L, Garate L, San Jose-Eneriz E, genetic silencing of microRNA-203 enhances ABL1 and
Abizanda G, Rodriguez-Otero P, Fortes P et al. (2009) BCR-ABL1 oncogene expression. Cancer Cell 13, 496
Epigenetic silencing of the tumor suppressor microRNA 506.
Hsa-miR-124a regulates CDK6 expression and confers 55 Brueckner B, Stresemann C, Kuner R, Mund C, Musch
a poor prognosis in acute lymphoblastic leukemia. Can- T, Meister M, Sultmann H & Lyko F (2007) The
cer Res 69, 44434453. human let-7a-3 locus contains an epigenetically regu-
45 Hackanson B, Bennett KL, Brena RM, Jiang J, Claus lated microRNA gene with oncogenic function. Cancer
R, Chen SS, Blagitko-Dorfs N, Maharry K, Whitman Res 67, 14191423.
SP, Schmittgen TD et al. (2008) Epigenetic modica- 56 Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch
tion of CCAAT enhancer binding protein alpha E, Yendamuri S, Shimizu M, Rattan S, Bullrich F,
expression in acute myeloid leukemia. Cancer Res 68, Negrini M et al. (2004) Human microRNA genes are
31423151. frequently located at fragile sites and genomic regions
46 Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato involved in cancers. Proc Natl Acad Sci USA 101,
C, Setien F, Casado S, Suarez-Gauthier A, Sanchez- 29993004.
Cespedes M, Git A et al. (2007) Genetic unmasking of 57 Takamizawa J, Konishi H, Yanagisawa K, Tomida S,
an epigenetically silenced microRNA in human cancer Osada H, Endoh H, Harano T, Yatabe Y, Nagino M,
cells. Cancer Res 67, 14241429. Nimura Y et al. (2004) Reduced expression of the let-7

FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS 1607
MicroRNAs and epigenetics F. Sato et al.

microRNAs in human lung cancers in association with Chiocca EA & Lawler S (2008) Targeting of the Bmi-1
shortened postoperative survival. Cancer Res 64, 3753 oncogene stem cell renewal factor by microRNA-128
3756. inhibits glioma proliferation and self-renewal. Cancer
58 Johnson SM, Grosshans H, Shingara J, Byrom M, Jar- Res 68, 91259130.
vis R, Cheng A, Labourier E, Reinert KL, Brown D & 72 Lessard J & Sauvageau G (2003) Bmi-1 determines the
Slack FJ (2005) RAS is regulated by the let-7 microR- proliferative capacity of normal and leukaemic stem
NA family. Cell 120, 635647. cells. Nature 423, 255260.
59 Wilkinson LS, Davies W & Isles AR (2007) Genomic 73 Wang H, Garzon R, Sun H, Ladner KJ, Singh R,
imprinting effects on brain development and function. Dahlman J, Cheng A, Hall BM, Qualman SJ, Chandler
Nat Rev Neurosci 8, 832843. DS et al. (2008) NF-kappaB-YY1-miR-29 regulatory
60 Steenman MJ, Rainier S, Dobry CJ, Grundy P, Horon circuitry in skeletal myogenesis and rhabdomyosar-
IL & Feinberg AP (1994) Loss of imprinting of IGF2 is coma. Cancer Cell 14, 369381.
linked to reduced expression and abnormal methylation 74 van der Vlag J & Otte AP (1999) Transcriptional
of H19 in Wilms tumour. Nat Genet 7, 433439. repression mediated by the human polycomb-group
61 Peters J & Robson JE (2008) Imprinted noncoding protein EED involves histone deacetylation. Nat Genet
RNAs. Mamm Genome 19, 493502. 23, 474478.
62 Simon JA & Kingston RE (2009) Mechanisms of poly- 75 Witt O, Deubzer HE, Milde T & Oehme I (2009)
comb gene silencing: knowns and unknowns. Nat Rev HDAC family: what are the cancer relevant targets?
Mol Cell Biol 10, 697708. Cancer Lett 277, 821.
63 Simon JA & Lange CA (2008) Roles of the EZH2 his- 76 Vire E, Brenner C, Deplus R, Blanchon L, Fraga M,
tone methyltransferase in cancer epigenetics. Mutat Res Didelot C, Morey L, Van Eynde A, Bernard D, Van-
647, 2129. derwinden JM et al. (2006) The Polycomb group pro-
64 Sander S, Bullinger L, Klapproth K, Fiedler K, Kestler tein EZH2 directly controls DNA methylation. Nature
HA, Barth TF, Moller P, Stilgenbauer S, Pollack JR & 439, 871874.
Wirth T (2008) MYC stimulates EZH2 expression by 77 Lehnertz B, Ueda Y, Derijck AA, Braunschweig U,
repression of its negative regulator miR-26a. Blood 112, Perez-Burgos L, Kubicek S, Chen T, Li E, Jenuwein T
42024212. & Peters AH (2003) Suv39h-mediated histone H3 lysine
65 Varambally S, Cao Q, Mani RS, Shankar S, Wang X, 9 methylation directs DNA methylation to major satel-
Ateeq B, Laxman B, Cao X, Jing X, Ramnarayanan K lite repeats at pericentric heterochromatin. Curr Biol 13,
et al. (2008) Genomic loss of microRNA-101 leads to 11921200.
overexpression of histone methyltransferase EZH2 in 78 Rajewsky N (2006) microRNA target predictions in
cancer. Science 322, 16951699. animals. Nat Genet 38(Suppl), S8S13.
66 Friedman JM, Liang G, Liu CC, Wolff EM, Tsai YC, 79 Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N,
Ye W, Zhou X & Jones PA (2009) The putative tumor Callegari E, Liu S, Alder H, Costinean S, Fernandez-
suppressor microRNA-101 modulates the cancer epige- Cymering C et al. (2007) MicroRNA-29 family reverts
nome by repressing the polycomb group protein EZH2. aberrant methylation in lung cancer by targeting DNA
Cancer Res 69, 26232629. methyltransferases 3A and 3B. Proc Natl Acad Sci USA
67 Gandellini P, Folini M, Longoni N, Pennati M, Binda 104, 1580515810.
M, Colecchia M, Salvioni R, Supino R, Moretti R, Li- 80 Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Calle-
monta P et al. (2009) miR-205 Exerts tumor-suppressive gari E, Schwind S, Pang J, Yu J, Muthusamy N et al.
functions in human prostate through down-regulation (2009) MicroRNA-29b induces global DNA hypome-
of protein kinase Cepsilon. Cancer Res 69, 22872295. thylation and tumor suppressor gene reexpression in
68 Juan AH, Kumar RM, Marx JG, Young RA & Sartor- acute myeloid leukemia by targeting directly DNMT3A
elli V (2009) Mir-214-dependent regulation of the poly- and 3B and indirectly DNMT1. Blood 113, 64116418.
comb protein Ezh2 in skeletal muscle and embryonic 81 Duursma AM, Kedde M, Schrier M, le Sage C &
stem cells. Mol Cell 36, 6174. Agami R (2008) miR-148 targets human DNMT3b pro-
69 Lee CJ, Dosch J & Simeone DM (2008) Pancreatic can- tein coding region. RNA 14, 872877.
cer stem cells. J Clin Oncol 26, 28062812. 82 Fujita PA, Rhead B, Zweig AS, Hinrichs AS, Karolchik
70 Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson D, Cline MS, Goldman M, Barber GP, Clawson H,
KW, Suri P & Wicha MS (2006) Hedgehog signaling Coelho A et al. (2010) The UCSC genome browser
and Bmi-1 regulate self-renewal of normal and malig- database: update 2011. Nucleic Acids Res 39, D876
nant human mammary stem cells. Cancer Res 66, 6063 D882.
6071. 83 Lu L, Katsaros D, de la Longrais IA, Sochirca O & Yu
71 Godlewski J, Nowicki MO, Bronisz A, Williams S, H (2007) Hypermethylation of let-7a-3 in epithelial
Otsuki A, Nuovo G, Raychaudhury A, Newton HB, ovarian cancer is associated with low insulin-like growth

1608 FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS
F. Sato et al. MicroRNAs and epigenetics

factor-II expression and favorable prognosis. Cancer gene EVL in colorectal cancer. Oncogene 27, 3880
Res 67, 1011710122. 3888.
84 Datta J, Kutay H, Nasser MW, Nuovo GJ, Wang B, 92 Meng F, Wehbe-Janek H, Henson R, Smith H & Patel
Majumder S, Liu CG, Volinia S, Croce CM, Schmitt- T (2008) Epigenetic regulation of microRNA-370 by
gen TD et al. (2008) Methylation mediated silencing of interleukin-6 in malignant human cholangiocytes. Onco-
MicroRNA-1 gene and its role in hepatocellular carci- gene 27, 378386.
nogenesis. Cancer Res 68, 50495058. 93 Saito Y, Suzuki H, Tsugawa H, Nakagawa I, Matsu-
85 Han L, Witmer PD, Casey E, Valle D & Sukumar S zaki J, Kanai Y & Hibi T (2009) Chromatin remodeling
(2007) DNA methylation regulates MicroRNA expres- at Alu repeats by epigenetic treatment activates silenced
sion. Cancer Biol Ther 6, 12841288. microRNA-512-5p with downregulation of Mcl-1 in
86 Lee KH, Lotterman C, Karikari C, Omura N, Feld- human gastric cancer cells. Oncogene 28, 27382744.
mann G, Habbe N, Goggins MG, Mendell JT & Maitra 94 Noonan EJ, Place RF, Pookot D, Basak S, Whitson
A (2009) Epigenetic silencing of MicroRNA miR-107 JM, Hirata H, Giardina C & Dahiya R (2009) miR-
regulates cyclin-dependent kinase 6 expression in pan- 449a targets HDAC-1 and induces growth arrest in
creatic cancer. Pancreatology 9, 293301. prostate cancer. Oncogene 28, 17141724.
87 Saito Y, Friedman JM, Chihara Y, Egger G, Chuang 95 Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE,
JC & Liang G (2009) Epigenetic therapy upregulates Hammond SM, Conlon FL & Wang DZ (2006) The
the tumor suppressor microRNA-126 and its host gene role of microRNA-1 and microRNA-133 in skeletal
EGFL7 in human cancer cells. Biochem Biophys Res muscle proliferation and differentiation. Nat Genet 38,
Commun 379, 726731. 228233.
88 Pallasch CP, Patz M, Park YJ, Hagist S, Eggle D, 96 Mann J, Chu DC, Maxwell A, Oakley F, Zhu NL, Tsu-
Claus R, Debey-Pascher S, Schulz A, Frenzel LP, kamoto H & Mann DA (2010) MeCP2 controls an epi-
Claasen J et al. (2009) miRNA deregulation by epige- genetic pathway that promotes myobroblast
netic silencing disrupts suppression of the oncogene transdifferentiation and brosis. Gastroenterology 138,
PLAG1 in chronic lymphocytic leukemia. Blood 114, 705714.
32553264.
89 Hoffman AE, Zheng T, Yi C, Leaderer D, Weidhaas J,
Slack F, Zhang Y, Paranjape T & Zhu Y (2009) micr-
Supporting information
oRNA miR-196a-2 and breast cancer: a genetic and epi- The following supplementary material is available:
genetic association study and functional analysis. Table S1. miRNAs and overlapping transcripts.
Cancer Res 69, 59705977. This supplementary material can be found in the
90 Kim S, Lee UJ, Kim MN, Lee EJ, Kim JY, Lee MY, online version of this article.
Choung S, Kim YJ & Choi YC (2008) MicroRNA Please note: As a service to our authors and readers,
miR-199a* regulates the MET proto-oncogene and the
this journal provides supporting information supplied
downstream extracellular signal-regulated kinase 2
by the authors. Such materials are peer-reviewed and
(ERK2). J Biol Chem 283, 1815818166.
may be re-organized for online delivery, but are not
91 Grady WM, Parkin RK, Mitchell PS, Lee JH, Kim
copy-edited or typeset. Technical support issues arising
YH, Tsuchiya KD, Washington MK, Paraskeva C,
from supporting information (other than missing les)
Willson JK, Kaz AM et al. (2008) Epigenetic silencing
of the intronic microRNA hsa-miR-342 and its host
should be addressed to the authors.

FEBS Journal 278 (2011) 15981609 2011 The Authors Journal compilation 2011 FEBS 1609

Das könnte Ihnen auch gefallen