ChapterPDF Available

Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression, and Associated Pathologies

Authors:

Abstract and Figures

Genomic imprinting is a form of epigenetic regulation whereby some genes are silenced according to their parental origin. The H19/IGF2 locus located in the chromosome 11 in p15.5 is the best characterized imprinted cluster. The locus generates two types of noncoding RNAs: the mRNA-like noncoding RNA H19 and the antisense RNA 91H. The regulation of H19 and its closely linked and reciprocally imprinted neighbor, IGF2, has been studied intensively both as a model for understanding imprinting control mechanisms and because of its role in human diseases. As with all imprinted clusters, H19 imprinted expression is regulated by an Imprinting Control Region (ICR), which controls interactions between promoters and shared enhancers. The locus functions like an “insulator model” in which trans-factors and epigenetic modifications are also required for full expression of genes. It is well assumed that H19 RNA functions as a riboregulator of which, expression is developmentally regulated. Elsewhere, the antisense RNA 91H has been recently discovered as a large and maternally imprinted noncoding RNA. It plays a role in the paternal IGF2 regulation and is overexpressed in breast cancer. This original trans-effect may be due to 91H participation in the three-dimensional organization of the locus, essential for the appropriate expression of genes. In this chapter, we summarize our current understanding of the molecular and biological roles of the ncRNAs expressed at the H19/IGF2 domain, both in terms of their contribution to genomic imprinting control, as well as in terms of cellular targets they might interact with. We also review knowledge of the locus-associated pathologies such as cancers and children syndromes. KeywordsNon coding RNA-Imprinting-H19/IGF2-91H-Cancer-Developmental pathologies
Reciprocal imprinting mechanism of H19 and IGF2 genes. Activation of gene expression is indicated by (þ), repression by (À), and inhibition of the enhancer function is represented by a vertical bar. Relative positions are expressed in kilobase pairs relatively to the H19 transcription start site. Three principal mechanisms intervene in the regulation: the methylation (represented by vertical bars), the enhancer activity, and the insulator activity (Hark et al. 2000). Three DNA regions are differentially methylated according to the allele, the DMR1 and 2 of the IGF2 gene (violet diamond) and the ICR (blue oval) at 2 kb upstream of the H19 gene. The enhancer sequences Enh (Enhancer downstream of the H19 gene, green circles) and Huc (enhancers upstream of the H19 gene, blue circles) represent, respectively, the endodermic and mesodermic enhancers (Ishihara et al. 2000; Drewell et al. 2002). On the maternal allele, the nonmethylated ICR contains four consensus CTCF binding sites (Hark et al. 2000). The CTCF DNA binding produces then a chromatin boundary, which prohibits enhancers to access to the IGF2 gene. The Huc enhancers may also activate the H19 gene (Drewell et al. 2002). The nonmethylated IGF2 DMR1 (violet diamond) acts as a silencer (Constancia et al. 2000). On the paternal chromosome, the methylated ICR does not bind any protein but acts as a H19 expression repressor. The Enh enhancers can then activate the IGF2 promoter and the methylated IGF2 DMR2 also activates gene expression (Murrell et al. 2001). ICR has a role of transcriptional repressor for the H19 gene (Srivastava et al. 2000). In 3 0 of H19, a secondary chromatin boundary independent of the methylation delimits the imprinted domain (Ishihara and Sasaki 2002)
… 
Content may be subject to copyright.
Noncoding RNAs at H19/IGF2 Locus:
Role in Imprinting, Gene Expression,
and Associated Pathologies
Nahalie Berteaux, Nathalie Spruyt, and Eric Adriaenssens
Contents
1 The H19/IGF2 Locus and the Parental Imprinting Model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 420
1.1 Overview and Description of the 11p15.5 Locus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 420
1.2 The Insulator Model of Imprinting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 424
2 The mRNA-Like Noncoding RNA H19 .................................................. 428
2.1 Properties and Expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 429
2.2 Functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 430
2.3 Regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 430
3 The Noncoding Antisense RNA 91H ..................................................... 431
3.1 Characterization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 431
3.2 Hypothesis About 91H Mechanism of Action ...................................... 432
4 H19/IGF2 Locus-Associated Pathologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 433
4.1 Hormone-Dependent Cancers (Breast, Uterus) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 433
4.2 Children Syndromes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 435
5 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 436
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 436
Abstract Genomic imprinting is a form of epigenetic regulatio n whereby some
genes are silenced according to their parental origin. The H19/IGF2 locus loca ted in
the chromosome 11 in p15.5 is the best characterized imprinted cluster. The locus
generates two types of noncoding RNAs: the mRNA-like noncoding RNA H19 and
the antisense RNA 91H. The regulation of H19 and its closely linked and recipro-
cally imprinted neighbor, IGF2, has been studied intensively both as a model for
understanding imprinting control mechanisms and because of its role in human
diseases. As with all imprinted clusters, H19 imprinted expression is regulated by
an Imprinting Control Region (ICR), which controls interactions between promo-
ters and shared enhancers. The locus functions like an “insulator model” in which
N. Berteaux, N. Spruyt, and E. Adriaenssens (*)
Institut de Biology de Lille, CNRS UMR 8161, 1 rue Pr Calmette, BP 447 59021 Lille Cedex,
France
e-mail: eric.adriaenssens@univ-lille1.fr
V.A. Erdmann and J. Barciszewski (eds.), RNA Technologies and Their Applications,
RNA Technologies, DOI 10.1007/978-3-642-12168-5_19,
#
Springer-Verlag Berlin Heidelberg 2010
419
trans-factors and epigenetic modifications are also required for full expression of
genes. It is well assumed that H19 RNA functions as a riboregulator of which,
expression is developmentally regulated. Elsewhere, the antisense RNA 91H has
been recently discovered as a large and maternally imprinted noncoding RNA. It
plays a role in the paternal IGF2 regulation and is overexpressed in breast cancer.
This original trans-effect may be due to 91H participation in the three-dimensional
organization of the locus, essential for the appropriate expression of genes. In this
chapter, we summarize our current understanding of the molecular and biological
roles of the ncRNAs expressed at the H19/IGF2 domain, both in terms of their
contribution to genomic imprinting cont rol, as well as in terms of cellular targets
they might interact with. We also review knowledge of the locu s-associated patho-
logies such as cancers and children syndromes.
Keywords Non coding RNA Imprinting H19/IGF2 91H Cancer Develop-
mental pathologies
1 The H19/IGF2 Locus and the Parental Imprinting Model
1.1 Overview and Description of the 11p15.5 Locus
The mammalian genome contains a small but growing number of genes that are
subject to genomic imprinting (Edwards and Ferguson-Smith 2007; Verona et al.
2003). Genomic imprinting is a form of epigenetic gene regulation that results in
expression from a single allele in a parent-of-origin-depe ndent manner. This form of
monoallelic expression is essential to normal mammalian development. While the
precise natu re of the initial epigenetic imprint remains an intensively investigated
topic, it is assumed that the parental imprint is set in the germline, when genomes are
in distinct compartments and can be differentially modified. After fertilization, the
parental imprints must survive the reprogramming that takes place in the preimplan-
tation embryo, including DNA demethylation and changes in histones modifications
(Reik et al. 2001). Imprinting is maintained throughout development and then erased
before being reestablished in the next generation’s germline. About 90 genes have
been reported to be imprinted even if some of them probably remain to be discovered
(for a complete list, see http://igc.otago.ac.nz/home.html and http://www.har.mrc.
ac.uk/research/genomicimprinting/maps.html). Despite extensive studies and some
major advancement regarding this intriguing phenomenon, we have not yet fully
characterized the underlying molecular mechanisms of genomic imprinting. Never-
theless, some principal hallmarks of imprinted genes can be listed:
l
Gene expression is allele-specific.
l
Gene expression is often tissue or stage-specific.
l
Many of imprinted genes are found in clusters throughout the genome.
l
The clusters contain two or more imprinted genes over a region that can span
1 Mb or more.
420 N. Berteaux et al.
l
Within each cluster, a common regulating region, which is called “imprinting
control region” (ICR, also called IC for imprinting Center or ICE for imprinting
control element), controls the imprinting of all genes in the cluster and can act
over hundreds of kilobases. ICRs are designed as differentially methylated
regions with parental-specific modifications that determine their activity. Deletions
of this region lead to the loss of imprinting of multiple genes of the cluster
(Leighton et al. 1995; Ripoche et al. 1997).
l
More recently, it has been reported that noncodin g RNA were associated with
imprinted clusters and have an essential role in regulating gene expression.
The H19/IGF2 cluster is located on the human chromosome 11 in p15.5 (homolog
to the murine distal chromosome 7). This 1 Mbp-long domain described in Fig. 1
includes nine imprinted genes and two independent imprinting centers.
M
R
PL23
H19
INS
TH
ASCL2 (MASH2)
PHEMX (TSSC6)
CD81 (TAPA1)
TSSC4
KCNQ1 (KvLQT1)
CDKN1C (p57KIP2)
SLC22A1L (Tssc5, IMPT1)
SLC22A1L-AS (Tssc5-AS
)
TSSC3 (Phlda2)
TRPM5 (MTR1)
NAP1L4
P
M
Telomere
Centromere
IGF-2-A
S
IGF-2
IC2 IC1
91H
KCNQ1DN
Expressed allele
Silent allele
Sense of transcription
IC
Imprinting Center
P
M
Paternal allele
Maternal allele
KCNQ1OT1 (KvLQT1-as , Lit1
)
Fig. 1 Representation of the human chromosomic region 11p15.5. This 1 Mb-locus includes nine
imprinted genes and two imprinting centers. It is delimited by the two maternally expressed genes
TSSC3 et H19 flanked by two nonimprinted genes NAP1L4 et MRPL23 (Tsang et al. 1995; Paulsen
et al. 1998). This region is homologous with the distal region of chromosome 7 in mouse. In spite
of the phylogenetic maintenance if the region over species, it exhibits some structural and
functional discrepancies. Indeed, the TSSC4 gene is imprinted in mouse but not in human (Paulsen
et al. 2000). The TRPM5 gene is imprinted in human but not in mouse (Prawitt et al. 2000).
A TSSC5 antisense transcript has been revealed in human but not in mouse and its imprinted status
has not yet been defined (Crider-Miller et al. 1997; Cooper et al. 1998). The CD81 gene is
imprinted in human and the imprinted status of the INS (Insulin gene) gene is not determined
(Maher and Reik 2000). The ASCL2 gene is imprinted with maternal expression in mouse whereas
its expression is biallelic in human (Miyamoto et al. 2002; Westerman et al. 2001). Usual names
are indicated and followed by secondary names in brackets. Name of antisense transcripts are
underlined and arrows indicating the sense of transcription are depicted below the chromosome
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 421
Numerous imprinted genes are associated to long antisense RNAs that overlap
several genes. The best described examples are Igf-2r/Air, Ube3A/Ube3A-as, and
Kcnq1/Kcnq1ot1 (Fig. 2). The first demonstration of a direct implication of an
MKRN3
a
b
c
UBE3A
MAGEL2
UBE3A-as
SNRPN ATP10CNDN
AS-SRO
PWS-SRO
ICE
Slc22a1Igf2rMas1 Slc22a2 Slc22a3
Air
Tssc4Ltrpc5
KnDMR1
Cd81 Tssc6 Ascl2Tssc3 Tssc5 Cdkn1c
Kνlqt1-as, Lit1
Kνlqt1
Fig. 2 Antisense RNA associated to imprinted clusters. Igf-2r/Air, Ube3A/Ube3A-as, and Kcnq1/
Kcnq1ot1 are the three best described examples of long antisense RNA, which take part in
epigenetic modifications and gene silencing within imprinted loci. (a) The region of chromosome
15q11–q13 responsible for the Angelman and Prader–Willi syndromes contains a number of
imprinted genes that are coordinately regulated by an imprinting center that contains two func-
tional elements, the PWS-SRO and the AS-SRO. The 460 kb-long Ube3A-as RNA is initiated in
the imprinting center from the paternal allele. It acts as a host gene for the transcription of several
snoRNA (small nucleolar RNA) and represses the UBE3A gene on the paternal allele (Rougeulle
et al. 1998; Runte et al. 2001; Landers et al. 2004). (b) The IGF2R locus. The imprinting center
(ICE) produces a paternally expressed 108-kb long transcript called Air that is necessary for the
silencing in cis of the genes IGF2R, Slc22a1, Slc22a2, and Slc22a3. Air could be implicated in the
methylation spreading thought the locus. (Rougeulle and Heard 2002; Sleutels et al. 2002).
(c) Within the 11p15.5 region, the Kcnq1 gene contains within its intron 10 the imprinting center
KvDMR1, which harbors bidirectional silencing property. This is linked to a paternal antisense
RNA, Kcnq1ot1 (also called Lit1) initiated from KvDMR1. The Kcnq1ot1 promoter shows a
maternal specific methylation. Kcnq1ot1 transcript has a key role in silencing of genes contained in
the Kcnq1 gene imprinted region and it participates to both silencing activity and methylation
spreading (Pandey et al. 2004; Thakur et al. 2004)
422 N. Berteaux et al.
antisense RNA concerns the Air transcript. It consists of a 108 kb-long transcript
localized in the imprinting center within the IG F2r gene and is necessary for the
paternal repress ion of the gene of the locus (Rougeulle and Heard 2002; Sleutels
et al. 2002). In spite of its well-established role in imprinting process, the molecular
mechanism remains unclear and authors propose hypothesis of methylation propa-
gation from the IGF2R gene or of repressive ARN/protein complexes formation.
The 460 kb-long Ube3A-as RNA is initiated in the imprinting center of the
Prader–Willi syndrome. It acts as a host gene for the transcription of several
snoRNA (small nucleolar RNA) and represses the UBE3A gene on the paternal
allele (Rougeulle et al. 1998; Runte et al. 2001; Landers et al. 2004).
Within the 11p 15.5 regi on, the ICR2 i s located within the intron 10 of the
Kcnq1 gene and harbors bidirectional silencing property. This feature is linked to
an antisense RNA, Kcnq1ot1 (also called Lit1 ), of w h ich the prom oter is
containedinICR2.Expressionofthistranscript is exclusively paternal. Indeed,
the Kcnq1ot1 promoter shows a maternal-specific methylation. This differential
epigenetic mark is lost in patients affected by Beckwith–Wiedemann syndrome
(BWS) with RNA biallelic expressi on (Mitsuya et al. 1999; Lee et al. 1999;
Du et al. 2004). More recently, Pandey et al. (2004) have documented that the
Kcnq1ot transcript has a key role in silencing o f genes contained in the Kcnq1
gene imprinted region and that it participates di rectly or indirectly to the methyl-
ation but without RNA interference mechanisms. Furthermore, interruption of
Kcnq1ot1 RNA pr oduc tion by t he inserti on of a polyade nyl atio n sequenc e down-
stream of the promoter also caused a loss of both silencing activity and methyla-
tion spreading. Thus, the antisense RNA plays a key role in t he silencing function
of the ICR (Thakur et al. 2004).
Elsewhere, a noncoding antisense RNA has also been described in the mouse
and human IGF2 genes (Moore et al. 1997; Okutsu et al. 2000). Like IGF2 , this
2.2 kb mRNA tran script is maternally imprinted and overexpressed in Wilms’
tumors. IG F2 -AS was expressed at levels comparable with IGF2 sense expression
derived from promoters P1 and P2 in normal tissue and in breast, ovarian, and
Wilms’ tumor tissues. It is composed of three exons, which overlap the exons 3 and
4 of the IGF2 gene (Vu et al. 2003). Its function remains unknown and its
involvement in imprinting has not yet been demonstrated, but findings indicate
that it is a good marker of Wilms’ tumor (Okutsu et al. 2000).
Finally, discovery of microRNAs (miRNA) and RNA interference could likely
provide new insights on imprinting mechanism. Then, the group of Cavaille has
identified in mouse a short cluster of maternally expressed miRNA genes (miR-431,
miR-433, miR-127, miR-434, and miR-136) transcribed and processed from a gene
antisense to the paternally expressed Rtl1 gene (Retrotransposon-like gene1) (Seitz
et al. 2003). Rtl1, also called Peg11 in sheep, displays homology with the Ty3/gypsy
retrotransposon family and its function is currently unknown (Charlier et al. 2001;
Youngson et al. 2005). Due to this peculiar sense–antisense organization, the
encoded miRNAs are obviously perfectly complementary to Rtl1 mRNA and thus
were predicted to cleave Rtl1 mRNA via RNAi-like mechanisms (Seitz et al. 2003).
Indeed, the predicted RNAi-mediated cleavage sites in the middle of the RNA
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 423
duplexes have been experimentally mapped by 5
0
-RACE (rapid amplification of
cDNA ends) experiments (Davis et al. 2005). These imprinted miRNAs are there-
fore among the very rar e miRNAs in animals that act as siRNAs (small interfering
RNAs) (Yekta et al. 2004). From an evolutionary point of view, this trans-allelic
RNAi between imprinted genes (for example, maternally expressed miRNAs
silence a paternally expressed gene) can be viewed as a good illustration of the
parent-conflict theory (Wilk ins and Haig 2003; for further discussion, see also
Davis et al. 2005; Lewis and Redrup 2005). In addition, this regulation might
also account for the complex gene regulatory network occurring at the ovine
Dlk1–Gtl2 locus, the so-called polar overdominance phenomena (for further infor-
mation, see Davis et al. 2005; also reviewed in Royo and Cavaille
´
2008).
Interestingly, a 23-nucleotide microRNA miR-675 was shown recently to be
processed from the H19 gene and this may in turn regulate mRNAs in development
and/or in oncogenesis. It is endogenously expressed in human keratinocytes and
neonatal mice and overexpressed in cells transfected with human or mouse H19
expression plasmids. These data demonstrate that H19 can function as a primary
microRNA precursor (Cai and Cullen 2007).
1.2 The Ins ulator Model of Imprinting
The H19 gene is one of the first genes proven to be imprinted. In human, it lies
within 200 kbp downstream of the IGF-2 gene (Zemel et al. 1992). The regulation
of H19 and its closely linked and reciprocally imprinted neighbor, IGF2, has been
studied intensively both as a model for understanding imprinting control mechanisms
and because of its role in human diseases. The two genes are imprinted in an opposite
manner, with the paternal IGF-2 and the maternal H19 alleles being reciprocally
expressed (Giannoukakis et al. 1993; Zhang and Tycko 1992).
1.2.1 DNA Methylation of H19 and IGF2 Genes
The H19 silent paternal allele exhibits several characteristics associated to its
transcriptional repression: it is hypermethylated in the promoter region and in the
5
0
region in embryonic tissue s, the promoter shows a compact chromatin structure
(Bartolomei et al. 1993; Ferguson-Smith et al. 1993) and its histone acetylation rate
is lower than the one of the maternal allele (Grandjean et al. 2001).
Surprisingly, the IGF2 promoter region is not methylated and its chromatin
structure is favorable to a biallelic transcription (Sasaki et al. 1992). However,
two other regions preferentially methylated on the expressed paternal allele have
been identified within the gene: the DMR1 located 3 kbp upstream the P1 promoter
acts as a silencer on the maternal allele when it is unmethylated, and the DMR2,
located within exons 5 and 6 is an activator on the paternal allele when it is
methylated (Feil et al. 1994; Murrell et al. 2001; Constancia et al. 2000). It is
424 N. Berteaux et al.
interesting to notice that both regions acquire this differential methylation after
fecundation (it is then a secondary methylation by opposition to primary methylation
that it is established in gamete and allows to distinguish the two parental alleles), and
this implies that it is not responsible for imprinting establishment.
1.2.2 Histone Modifications at the H19/IGF2 Locus
Analysis of histone acetylation state at the murine locus shows that H4 histone
acetylation discrepancies take place within H19 and IGF2 genes in a parental-
specific manner with expressed alleles being more acetylated than silent alleles.
However, the link between DNA methylat ion, histone hypoacetylation, and gene
expression is established only for the H19 promoter region (Grandjean et al. 2001;
Pedone et al. 1999). Moreover, several studies show that the inhi bition of histone
deacetylases deregulates gene expression at the locus with a repression of the H19
active maternal allele, changes in acetylation patterns of the ICR region (Grandjean
et al. 2001), and an IGF2 biallelic expression (Hu et al. 1998; Yang et al. 2003).
Finally, the HDAC recruitment is directly involved in the repression effect of the
insulator protein CTCF described in the next paragraph (Lutz et al. 2000).
1.2.3 The ICR or Imprinting Control Region
The methylation of promoter regions is not sufficient to explain the reciprocal
expression of the two genes, other elements exhibiting primary methylation hall-
marks have been searched within the locus. Discovery of the ICR, located between
the H19 and IGF2 genes, allows understanding the mechanism of imprinting setting.
Existence of a common regulatory region has been initially suggested by dele-
tions assays of the H19 gene region (Leighton et al. 1995; Ripoche et al. 1997). This
sequence, named ICR, is located within the region comprised between 2 and 4 kb
upstream of the transcription start site of the H19 gene and carries primary
methylation marks (Tremblay et al. 1997). ICR has a long-range action to establish
the H19 and IGF2 imprinting during the embryonic development. It is exclusively
methylated on the paterna l allele and shows nuclease hypersensibility sites on the
maternal allele (Hark and Tilghman 1998; Kanduri et al. 2000a). These epigenetic
hallmarks are a sign of a protein binding to the sequences. ICR deletion leads to loss
of H19 and IGF2 (Thorvaldsen et al. 1998, 2002). ICR is also necessary to the
imprinting of a H19 transgene in mouse (Elson and Bartolomei 1997). These studies
show the pivotal role of ICR in imprint ing.
To understand the long-range and allele specific effect of ICR, deletion and
relocalization assays have been conducted. It revealed the chromatin insulator
activity of the ICR region (Webber et al. 1998; Kaffer et al. 2000; Kanduri et al.
2000b). Indeed, it is able to insulate communication between a promoter and
an enhancer when it is located between the two regions. This activity required a
zinc-finger protein named CTCF (CCTC-binding factor) (Bell et al. 1999). In vitro
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 425
and in v ivo experiments have shown in mouse that CTCF binds unmethylated ICR
via four consensus sites and the binding is abolished by DNA methylation (Bell and
Felsenfeld 2000; Hark et al. 2000; Kanduri et al. 2000b; Szabo et al. 2000;
Holmgren et al. 2001; Ulaner et al. 2003). In human, there are seven binding sites
but the sixth is only the one to possess a differential methylation (Takai et al. 2001).
The H19/IGF2 insulation model is represented in the Fig. 3. Both genes share a
common set of enhancers located downstream from the H19 gene. In the mater nal
allele, the CTCF recruitment on the unmethylated ICR acts as a chromatin bound-
ary and blocks the enhancer access to the IGF2 promoter to prevent its activation.
The H19 gene is then activated (Reik and Murrell 2000; Wolffe 2000).
H19
IGF-2
×
EnhICRHucDMR1
M
P
H19
×
++
+
+
INSULATOR
–2–12–137 +10.5 kpb
CTCF
CTCF
INSULATOR
DMR2
+
IGF-2
CTCF
Fig. 3 Reciprocal imprinting mechanism of H19 and IGF2 genes. Activation of gene expression
is indicated by (þ), repression by (), and inhibition of the enhancer function is represented by a
vertical bar. Relative positions are expressed in kilobase pairs relatively to the H19 transcription
start site. Three principal mechanisms intervene in the regulation: the methylation (represented by
vertical bars), the enhancer activity, and the insulator activity (Hark et al. 2000). Three DNA
regions are differentially methylated according to the allele, the DMR1 and 2 of the IGF2 gene
(violet diamond) and the ICR (blue oval) at 2 kb upstream of the H19 gene. The enhancer
sequences Enh (Enhancer downstream of the H19 gene, green circles) and Huc (enhancers
upstream of the H19 gene, blue circles) represent, respectively, the endodermic and mesodermic
enhancers (Ishihara et al. 2000; Drewell et al. 2002). On the maternal allele, the nonmethylated
ICR contains four consensus CTCF binding sites (Hark et al. 2000). The CTCF DNA binding
produces then a chromatin boundary, which prohibits enhancers to access to the IGF2 gene. The
Huc enhancers may also activate the H19 gene (Drewell et al. 2002). The nonmethylated IGF2
DMR1 (violet diamond ) acts as a silencer (Constancia et al. 2000). On the paternal chromosome,
the methylated ICR does not bind any protein but acts as a H19 expression repressor. The Enh
enhancers can then activate the IGF2 promoter and the methylated IGF2 DMR2 also activates
gene expression (Murrell et al. 2001). ICR has a role of transcriptional repressor for the H19 gene
(Srivastava et al. 2000). In 3
0
of H19, a secondary chromatin boundary independent of the
methylation delimits the imprinted domain (Ishihara and Sasaki 2002)
426 N. Berteaux et al.
On the paternal allele, the ICR methylation does not allow CTCF binding and
leads to activation of the distal IGF2 promoter and gene expression (for a review,
see Lewis and Murrel 2004 ). Finally, the maintenance of the unmethylated state of
maternal ICR is due to the CTCF protein, which prevents the de novo methylation
in this region (Fedoriw et al. 2004; Lewis and Murrel 2004; Szabo et al. 2004). The
mesodermic enhancer activity recently discovered (Drewell et al. 2002), which
intervene in the regulation can be added to this model. Finally, the in vivo CTCF
binding upstream of the Mrpl23 gene could be a chromatin boundary delimiting the
imprinted domain (Ishihara and Sasaki 2002).
1.2.4 Imprinting and Parental Specific Chromatin Loops
The cis elements described previously have a long-range action. They have to
physically interact with each other or with their target to exert their eff ects.
Chromosome conformation capture (3C) analysis in mice, which assay for physical
interactions betwee n chromosomal regions, have suggested that CTCF has a critica l
role in the epigenetic regulation of high-order chromati n structure and gene silenc-
ing over considerable distances in the genome, but the precise nature and function
of the looping is debated (Engel et al. 2008; Kurukuti et al. 2006; Lopes et al. 2003;
Murrell et al. 2004; Yoon et al. 2007). Kurukuti et al. 2006 reported that on the
paternal allele, enhancers interact with the IGF2 promoters whereas on the mater-
nal, this is prevented by CTCF binding within the H19 ICR. They demonstrated that
the maternal-specific silencing of IGF2 results when the ICR interacts with a matrix
attachment region (MAR3) and a differentially methylated region (DMR1) at the
IGF2 locus to generate a tight loop around the IGF2 gene, thereby physically
impeding Igf2 expression. Moreover, CTCF interacts with the three clustered
IGF2 promot ers and recruits polycomb repress ive complexes that lead to the
allele-specific methylation at lysine 27 of histone H3 (H3-K27) and to the suppres-
sion of the maternal IGF2 promoters (Li et al. 2008). Elsewhere, Murrell et al.
(2004) reported that on the maternal allele, the unmethylated ICR binds to the
DMR1 of IGF2 resulting in an inactive domain where IGF2 is away from the
enhancers. On the paternal allele, the methylated ICR associates with the methy-
lated IGF2 DMR2 moving IGF2 into the active chromatin domain (Dekker et al.
2002). More recently, it has been shown that on the maternal allele, the enhancers
make contacts throughout the H19 coding unit and promoter (Engel et al. 2008;
Kato and Sasaki 2005).
Figure 4 provides a simplified overview of available data about chromatin loop
structures at the H19/IGF2 locus (Kurukuti et al. 2006; Murrell et al. 2004; Weber
et al. 2003). Additional mechanisms exist for an imprint mark, such as chromatin
composition, organization, and histone acetylation or methylation state (Fuks 2003;
Grandjean et al. 2001), even if DNA methylation is by far the best candidate
(Bestor 2000).
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 427
2 The mRNA-Like Noncoding RNA H19
H19 encodes a spliced and polyadenylated RNA that lacks conserved open reading
frames (ORFs) but does have a conserved secondary RNA structure (Juan et al.
2000). Even if extensive deletions and/or point mutations of an ectopic human H19
RNA generate a protein, (Joubel et al. 1996) no endogenous transl ation product has
so far been identified (Pachnis et al. 1984, 1988). Therefore, it was quickly proposed
that H19 RNA functions as a riboregulator (Brannan et al. 1990) of which
Maternal allele
Paternal allele
CTCF
H19
DMR2
CTCF
ICR
enhancers
H19
MAR3
IGF2
DMR1
H19
DMR2
ICR
enhancers
MAR3
DMR2
ICR
DMR1
Fig. 4 Chromatin loop structures at the H19/IGF2 locus. Chromosome conformation capture (3C)
assays revealed long-range physical interactions within the H19/IGF2 region. These chromatin
structures are orchestrated by CTCF and regulate epigenetic hallmarks and gene silencing within
the locus. On the maternal allele, CTCF binding to the ICR prevents interaction between enhancers
and IGF2 promoter. ICR interacts with a matrix attachment region MAR3 and with the differen-
tially methylated region DMR1 within the IGF2 region. This leads to the formation of a silencing
loop, which impedes IGF2 expression. Enhancers can interact with the H19 promoter and activate
its expression (Engel et al. 2008; Kato and Sasaki 2005). Moreover, CTCF interacts with the three
clustered IGF2 promoters and recruits polycomb repressive complexes that lead to the allele-
specific histone methylation and to suppression of the maternal IGF2 promoters (Li et al. 2008).
On the paternal allele, enhancers interact with the IGF2 promoters (Kurukuti et al. 2006). The
methylated ICR associates with the methylated IGF2 DMR2 moving IGF2 into the active
chromatin domain (Dekker et al. 2002). ICR methylation spreads to the H19 promoter that impairs
H19 paternal expression
428 N. Berteaux et al.
expression is developmentally regulated. It is abundantly expressed in both extra-
embryonic and fetal tissues and is repressed after bir th except in a few adult organs,
particularly in the mammary gland (Douc-Rasy et al. 1993; Dugimont et al. 1995). It
should be emphasized that the role of the H19 gene in cancer is still a matter of
debate. It has been proposed that H19 functions as a tumor suppressor in some
Wilms’ tumors, embryonic rhabdomyosarcoma, and the Beckwith–Wiedmann cancer
predisposing syndrome (Okamoto et al. 1997; Steenman et al. 1994). Consistently,
some studies conclude that it downregulates the IGF2 factor (Wilkin et al. 2000). By
contrast, other studies including ours argue in favor of an oncogenic role of H19 with
a positive correlation with cell aggressiveness (Lottin et al. 2002a; Rachmilewitz
et al. 1995). H19 activation has also been reported in various cancer tissues:breast
(Douc-Rasy et al. 1993; Dugimont et al. 1995; Adriaenssens et al. 1998), bladder
(Ariel et al. 1995; Elkin et al. 1995), lung (Kondo et al. 1995), and esophageal
cancers (Hibi et al. 1996). Its oncoge nic role has been well documented in the
bladder, since it is considered as an oncodevelopmental marker (Cooper et al. 1996)
and regulates genes involved in metastasis and blood vessel development (Ayesh
et al. 2002). These observations support a H19 role in tumor invasion and angiogen-
esis. In spite of polemic on the H19 role in cancer, we clearly established the
oncogenic role of H19 (Lottin et al. 2002a) in breast cancer, and we demonstrated
that H19 overexpression in breast cancer cells promotes the cell cycle progression,
by increasing S phase entry (Berteaux et al. 2005).
2.1 Properties and Expression
The H19 gene was discovered in the mouse as a gene under coordinate regulation
with (-feto-protein in the liver (Pachnis et al. 1984). More recently, Juan et al.
(2000) brought evidence for evolutionarily conserved secondary structure in the
H19 RNA from several mammalian species. The H19 gene is an unusual gene, in
that it is transcribed by RNA polymerase II, processed by capping, splicing, and
polyadenylation; but it does not appear to encode a protein. Actually, the particu-
larity of the H19 transcript is its inability to be translated when the 5
0
untranslated-
region (5
0
UTR) is not experimentally altered (deletions and/or point mutations)
(Pachnis et al. 1988; Joubel et al. 1996). Furthermore, hypothetical translation of
established sequences from a range of mammalian species shows an absence
of conserved ORFs of any size. Consequently, given the evolutionary conservation
of structure at the RNA level and the absence of conservation at the protein level, it
has been proposed that the mature transcript is the functional product of the H19
gene and that its function requires the ability to fold into a specific secondary
structure. As early as 1990, Brannan et al. (1990) proposed that this gene could act
as a riboregul ator .”
The H19 gene encodes one of the most abundant RNAs in the devel oping mouse
and human embryo (Pachnis et al. 1984; Brannan et al. 1990). It is expressed at the
blastocyst stage of development and accumulates to high levels in tissues of
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 429
endodermal and mesodermal origins (Poirier et al. 1991; Lustig et al. 1994) as well
as ectodermal origin (Ohlsson et al. 1994, Hemberger et al. 1998). H19 is expressed
in the choroid plexus and leptomeninges of the developing mouse fetus (Svensson
et al. 1995) but not in these tissues during human development (Ohlsson et al.
1994). After birth, the gene is repressed in almost all tissues except skeletal muscle
(Pachnis et al. 1984; Leibovitch et al. 1995; Douc-Rasy et al. 1993; Milligan et al.
2000). In other respects, a basal but significant H19 expression is detected at
adulthood in lung, heart, and thymus (Poirier et al. 1991), mammary gland
(Douc-Rasy et al. 1993; Dugimont et al. 1995; Adriaenssens et al. 1999), adrenal
gland (Liu et al. 1995), and uterus (Adriaenssens et al. 1999; Ariel et al. 1997).
2.2 Functions
Since the first mention of H19 in 1984 by Pachnis et al., its functions have only
begun to emerge. It has been reported that H19 RNA was involved in the repression
of the IGF2 oncogene by affecting its transcription (Wilkin et al. 2000) or its
translation (Li et al. 1998). In addition, we brought evidence that the H19 gene
posttranscriptionally upregulates the thioredoxin level, a key protein of the cellular
redox metabolism (Lottin et al. 2002b). Deletion of the H19 gene in mouse
(KO mice) leads to a size and weight increase of about 10% (Ripoche et al.
1997). But the consecutive biallelic IGF2 expression in these animals does not
allow concluding to an H19 direct effect. However, the group of Surani demon-
strated that induction of a targeted H19 biallelic expression via specific silencer
deletion in transge nic mice leads to smaller animals whereas IGF2 expression is not
affected (Drewell et al. 2000). So, the H19 expression pattern suggests that the
transcript assures a major functi on during the development.
2.3 Regulation
Beyond epigenetic regulations, the H19 gene is submitted to local regulations (i.e.,
on the level of the promoter), by hormones, growth factors, or other cytokines.
Indeed, in mammary cells, H19 is activated by HGF-SF (hepatocyte growth factor/
scatter factor), which has been identified as one of the main paracrin mediators of
morphogenetic epithelial/mesenchymal interactions. This factor has potent moto-
genic, mitogenic, and morphogenic effects on epithelial cells in culture, and H19
RNA synthesis is related to the migratory phenotype of cultured cells. EGF and
FGF-2 also activates H19 but less, whereas IGF-2, TGFb1, and TNFa have no
effect on H19 expression in these cells (Adriaenssens et al. 2002). In uterus, during
estrus (proliferative phase) and metestrus (early secretory phase) phases, and in
breast, during puberty and pregnancy, morphologic changes are associated to peak
of H19 expression. Moreover, its expression is regulated by steroid hormones
430 N. Berteaux et al.
estrogen and progesterone, which respectively up- and downregulate the gene
(Adriaenssens et al. 1999). Elsewhere, we also demonstrated a negativ e regulation
of H19 by the tumor suppressor protein P53 (Dugimont et al. 1998).
H19 and AFP (a-fetoprotein) are abundantly transcripted in mammalian fetal
liver but are rapidly repressed after birth. This repression is partly controlled by the
Afr1 locus (a Fetoprotein Regulator 1) (Pachnis et al. 1984). Recently, Perincheri
et al. (2005) have mapped the locus and identified the murine Zhx gene (Zinc
Fingers and Homeoboxes gene), ortholog to the human ZHX2 gene. This factor is
directly responsible for the H19 postnatal repression in liver but also probably in
other organs since it is expressed in ubiquist manner.
In breast cancer, preferential accumulation of H19 transcripts has been observed
at stroma/ epithelium interface (Dugimont et al. 1995; Adriaenssens et al. 1998).
Moreover, scattering and morphogenesis of epithelial cells by a conditioned
medium from fibroblasts induces activation of the H19 gene (Adriaenssens et al.
2002). The H19 gene is then finely regulated by environmental factors and it is
involved in the epithelial/mesenchymal crosstalk, essential during tumorigenesis.
Posttranscriptional regulations have been also reported such as RNA stabilization
by not yet identified proteins (Milligan et al. 2000; Jouvenot et al. 1999) Elsewhere,
a direct association of the human and mouse H19 RNA with the IMP protei n
family have been demonstrated (In mouse: CRD-BP (cMyc mRNA coding Region
instability Determinant Binding Protein) and in human: IMP1, 2, and 3 (IGF-II
mRNA-binding protein)). These proteins are able to regulate H19 RNA localization
and stabilization and are also able to bind the IGF2 RNA (Tessier et al. 2004; Runge
et al. 2000; Nielsen et al. 2001, 2004; Liao et al. 2005).
3 The Noncoding Antisense RNA 91H
3.1 Characterization
A more recently identified characteristic of imprinted genes is their association, in
some cases, with noncoding antisense transcripts (ncRNAs), which have been
suggested to constitute a new epigenetic regulatory system (see Fig. 2). These
ncRNAs are not yet clearly classified, but categories with known gene regulatory
functions are emerging. It includes (1) intergenic transcripts that regulate local
chromatin activity, (2) cis-acting long ncRNAs such as Xist involved in chromo-
some inactivation, and (3) ncRNA expressed within imprinting loci such as Air,
KCNQ1OT1, and UBE3A-as involved in domain silenc ing (Rougeulle and Heard
2002; Rougeulle et al. 1998; Sleutels et al. 2002; Thakur et al. 2004).
These latter transcripts share some characteristics:
l
It is always very long RNA (several hundreds of base pairs).
l
Its expression is allele-specific.
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 431
l
The promoter is often located near an imprinting center and/or in an intron of a
coding gene.
l
The transcript is implicated in gene silencing and epigenetic modifications.
It regulates expression of overlapped (or not) genes in cis.
Apart from the identification of some IGF2 antisenses transcripts, few data are
available on that topic at the H19/IGF2 locus. We recently identified and character-
ized a new transcript within this locus. It consists of a large intergenic 120 kb-long
RNA that we named 91H since it is transcribed antisense to H19. This nuclear and
short-lived RNA is expressed predominantly from the maternal allele in both mouse
and human within the H19 gene region. Moreover, the transcript is stabilized in
breast cancer cells and overexpressed in human breast tumors. Knockdown experi-
ments showed that 91H inhibition downregulates IGF2 expression in trans. Thus,
91H shares the same characteristics as the other similar ncRNAs do, which are
described above, apar t from its trans effect on IGF2 expression. Indee d, we
demonstrated that the maternal 91H transcript is involved in the maintenance of
the paternal IGF2 gene expression (Berteaux et al. 2008).
3.2 Hypothesis About 91H Mechanism of Action
To unravel this possible trans-effect of the 91H RNA, we envisaged a physical
proximity between homologous chromosomes. Recent works using chromosome
conformation capture technology strongly support the notion of epigenetic chro-
mosomal networks. Several different chromosomes converged on the H19 ICR
simultaneously (Ling et al. 2006; Zhao et al. 2006) probably through the CTCF
protein, and these long-range allele-specific chromosomal associations were linked
with epigenetic regulation of transcriptio n in trans. These data support the possi-
bility that the 91H RNA collaborates in vivo with the establishment of chromo-
somal complexes and that this collaboration leads to activation of the IGF2 paternal
allele. The fact that 91H knockdown only slightly affects gene expression from the
maternal allele seems to indicate that 91H effect is not mediated through the ICR/
CTCF complex. However, other candidates such as enhancer DNA regions may be
considered for interacting with the antisense RNA. Indeed, 91H encompasses a
region including the biallelically transcribed HUC sequences. It consists of regu-
latory elements located just upstream of the ICR, whi ch act as strong mesodermal
enhancers and are supposed to activate IGF2 expression on the paternal allele
(Drewell et al. 2002). Accordingly, we proposed a model to explain the 91H
trans-effect on IGF2 expression in which cooperation between mesodermic
(HUC) and endodermic (ENH) enhancer sequences would be required for full
expression of the gene (Fig. 5). Moreover, 91H may attract repressive chromatin
modifications to the maternal allele by trapping factors responsible for DNA or
histone modifications, which become available for the paternal allele when 91H is
disrupted, resulting in IGF2 expression decrease (Rinn et al. 2007; Yu et al. 2008).
432 N. Berteaux et al.
4 H19/IGF2 Locus-Associated Pathologies
4.1 Hormone-Dependent Cancers (Breast, Uterus)
It should be emphasized that the role of the H19 gene in cancer is still a matter
of debate. Investigations in tumor development are delicate on account of the H19
noncoding state, the lack of knowledge of its mechanism of action, and its
imprinted status. Gene expression has been studied in numerous cancer types, but
the gene status appears to be contradictory since it can be either tumor suppressor or
oncogene depending on the studied model. Table 1 provides a nonexhaustive
general survey of bibliographic data available and illus trates their heterogeneity.
Indeed, it has been proposed that H19 functions as a tumor suppressor in some
Wilms’ tumors, embryonic rhabdomyosarcoma, and the Beckwith–Wiedmann can-
cer predisposing syndrome (Okamoto et al. 1997; Steenman et al. 1 994). Consis-
tently, some studies conclude that it downregulates the IGF2 factor (Wilkin et al.
2000). By contrast, other studies including ours argue in favor of an oncogenic role
of H19 with a positive correlation with cell aggressiveness (Lottin et al. 2002a;
Rachmilewitz et al. 1995). H19 activation has also been reported in various cancer
kbp–2–12–137 +10.5
H19
IGF-2
´
CTCF
MrpL23
H19
IGF-2
Limiting
regulatory
factors
?
91H RNA
ICR
HUC
ENH
ICR
HUC ENH
MrpL23
´
Fig. 5 Model for 91H trans-effect on IGF2 expression. Two sets of enhancers (HUC and ENH
sequences that correspond, respectively, to mesodermic and endodermic enhancers) regulate IGF2
expression. Both would be required for full expression of the gene. In addition, the two IGF2
alleles would be competing for a common limited stock of regulatory elements (methylation/
acetylation/transcription?). On the maternal allele, 91H would block the locus and prevent the
HUC sequences from interaction with any regulatory factors. These latter would be then directed
on the paternal allele, in the HUC region and/or in the IGF2 promoter region, and would cooperate
with the cis endodermic enhancers resulting in IGF2 enhanced expression. (Arrows indicates
positive regulations whereas lines with bars correspond to inhibitions; ICR: Imprinting Control
Region, CTCF: CCTC-binding factor)
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 433
tissues: brea st (Douc-Rasy et al. 1993; Dugimont et al. 1995; Adriaenssens et al.
1998), bladder (Ariel et al. 1995; Elkin et al. 1995), lung (Kondo et al. 1995), and
esophageal cancers (Hibi et al. 1996). Its oncogenic role has been well documented
in the bladder, since it is considered as an oncodevelopmental marker (Cooper et al.
1996) and regulates genes involved in metastasis and blood vessel development
(Ayesh et al. 2002). These observations suppor t a H19 role in tumor invasion and
angiogenesis. In spite of polemic on the H19 role in cancer, the oncogenic role of
H19 has been clearly established in some models. In blad der and uterus cancers,
H19 is directly associated to tumor progression and is considered as tumor marker
(Ariel et al. 2000b; Lottin et al. 2005). Some authors consider H19 as an oncofetal
RNA (Ariel et al. 1997, 2000a). Ohana et al. (2002) proposed an approach of gene
therapy based on the use of H19 regulatory sequences driving the expression of the
Table 1 Overview of bibliographic data about oncogene or tumor suppressor status of the H19
gene
Oncogene Tumor suppressor
Organ, model or
type of cancer
Bibliographic references Organ, model or type
of cancer
Bibliographic references
Transgenic mice Leighton et al. (1995),
Ripoche et al. (1997)
Transgenic mice Drewell et al. (2000)
Bladder Ariel et al. (1995, 1997,
2000b), Elkin et al.
(1995), Cooper et al.
(1996), Ayesh et al.
(2002), Ohana et al.
(2002)
SHE cells Wiseman et al. (1991),
Isfort et al. (1997)
Chorion (JEG-3
cells)
Rachmilewitz et al. (1995) G401 cells (WT) Hao et al. (1993)
Breast Douc-Rasy et al. (1993),
Dugimont et al.
(1995), Adriaenssens
et al. (1998), Lottin
et al. (2002a, b)
JEG-3 cells (Chorion) Li et al. (1998)
Lung Kondo et al. (1995) Children liver,
Hepatoblastomas
Li et al. (1998), Wilkin
et al. (2000)
Uterus Tanos et al. (2004), Lottin
et al. (2005)
Wilms’ tumors
(kidney)
Steenman et al. (1994),
Casola et al. (1997)
Esophagus/colon Hibi et al. (1996), Cui
et al. (2002)
Children muscle,
rhabdomyosarcoma
Casola et al. (1997)
Liver Manoharan et al. (2004) Beckwith–Wiedemann
syndrome
Reik et al. (1995)
Ovary Tanos et al. (1999),
Chen et al. (2000)
Adrenals Liu et al. (1995),
Wilkin et al. (2000)
Head/neck Rainho et al. (2001)
Pharynx Ng et al. (2003)
Testicles Verkerk et al. (1997),
Ariel et al. (2000a)
Demonstration of the oncogene/tumor suppressor status in mentioned works is not always clearly
established. Numerous results converge to one or the other of these statuses and from these data,
authors have proposed their hypothesis
434 N. Berteaux et al.
diphtheria toxin. Injection of these constructs in mice-induced tumors derived from
cancerous colon and bladder cells leads to a decrease of the tumor growth (Ohana
et al. 2005). In breast cancer, H19 is overexpressed in 70% of the adenocarcinomas
and its expression is correlated with the “Tumor values,” the pres ence of hormones
receptors, and tumor invasion (Adriaenssens et al. 1998). Overexpression of an
ectopic H19 gene enhances the tumorigenic properties of breast cance r cells (Lottin
et al. 2002a) and promotes the cell cycle progression, by increasing S phase entry
(Berteaux et al. 2005).
In all cases, deregulation of H19 expression results in genetic alterations leading
to a loss of heterozygote (LOH, for example, in the case of uniparental disomy) or
epigenetic alterations leading to a loss of imprinting (LO I by hypermethylation or
hypomethylation of the H19 promoter or the ICR). It is then often difficult to assign
directly the observed phenotypes to the H19 gene only, since it is tightly connected
to the IGF2 gene, and in most of the cases, the expression of both the genes is
modified. To summarize, the H19 contribution in tumorigenesis depends on the
physiological conditions, the considered tissue, and the nature of deregulations.
4.2 Children Syndromes
The demonstration that the H19 transcript is stabilized in breast cancer cells and
overexpressed in human breast tumors led us to propose a link between antisense
transcription and cancer. Furthermore, we know that the deregulation of the H19/
IGF2 locus is associated to several human fetal syndromes such as BWS and
Silver–Russell syndrome (SRS).
BWS is associated with fetal and postnatal overgrowth and is associated with
embryonic tumors such as children kidney tumors named Wilm’s tumors. BWS can
be caused by a range of different defe cts. Several distinct genetics or epigenetics
errors involving 11p15 have been identified in different BWS patients. Some
patients have maternal chromosomal rearrangements of 11p15, meaning that
there is a disruption of the chromosome in this region. Other patients have paternal
uniparental disomy of 11p15, meanin g that the maternal copy of this region is
replaced with an extra paternal copy. Many other patients have abnormal DNA
methylation in different DMR regions of 11p15, meaning that normal epigenetic
marks that regulate imprinted genes in this region are alter ed. In some cases, the
expression of IGF2 gene is doubled and expression of H19 gene is silenced. BWS is
often associated with H19 epigenetic inactivation due to ICR hypermethylation.
This overexpression of IGF2 is responsible for symptoms linked to the pathology.
Nevertheless, in some cases, the specific defe ct causing BWS in an affected patient
may remain unknown. In about one-third of BWS patients, the genetic or epigenetic
mutation is unknown.
SRS is an intrauterine growth delay associated to an altered postnatal growth with
facial dysmorphy and corporal asymmetry. Reasons of SRS are varied, chromosome
mosaic, equilibrated translocation (1; 17 or 17; 20), deletion (8q11–q13). In some
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 435
cases, defect in 11p15-5 region has been observed. For numerous patients, this
syndrome is associated with ICR hypomethylation, IGF2 silencing, and H19 biallelic
expression. As BWS, some cases of SRS remain unexplained. The discovery of 91H
gene and its function on gene regulation in 11p15.5 locus can give research lanes to
explore theses unexplained cases of BWS or SRS.
5 Conclusion
There is growing evidence that noncoding transcripts are functional and take part
in most, if not all, complex genetic phenomena in eukaryotes, including RNA
interference-related processes such as transcriptional and posttranscriptional gene
silencing as well as parental imprinting and allelic exclusion. Noncoding RNAs
intervene in general biological processes and the close connection between non-
coding RNAs and epigenet ic processes suggests that they compose a hitherto
hidden layer of genomic programming in humans. The next frontier is now the
functional characterization of these molecules to understand the interactions
between the regulatory RNAs and their targets. This is a way to better understand
how an RNA molecule can regulate cell cycle, gene expression, methylation
spreading, or even chromosomal network. The particular 91H antisense transcript
that is produced at the H19/IGF2 locus adds further complexity to the cluster of
imprinted genes in the human imprinted 11p15.5 region. 91H RNA is involved in
the control of paternal IGF2 expression suggesting that a distinct mechanism of
imprinting arises in the locus. This innovating type of regulation in trans between
two homologous chromosomes need to be elucidated to understand, at least in part,
the complex regulation that takes place in the imprinted locus (chromosomal
network, methylation...). Functiona l studies would allow using these RNA mole-
cules as tumor markers or therapeutic targets. Particularly, 91H could be a good
candidate to improve the diagnostic and the therapeutic tools for severe children’s
syndromes or adult cancers associated to the 11p15 chromosomal region.
References
Adriaenssens E, Dumont L, Lottin S et al (1998) H19 overexpression in breast adenocarcinoma
stromal cells is associated with tumor values and steroid receptor status but independent of p53
and Ki-67 expression. Am J Pathol 153:1597–1607
Adriaenssens E, Lottin S, Dugimont T et al (1999) Steroid hormones modulate H19 gene
expression in both mammary gland and uterus. Oncogene 18:4460–4473
Adriaenssens E, Lottin S, Berteaux N et al (2002) Cross-talk between mesenchyme and epithelium
increases H19 gene expression during scattering and morphogenesis of epithelial cells. Exp
Cell Res 275:215–229
Ariel I, Lustig O, Schneider T et al (1995) The imprinted H19 gene as a tumor marker in bladder
carcinoma. Urology 45:335–338
436 N. Berteaux et al.
Ariel I, Ayesh S, Perlman EJ et al (1997) The product of the imprinted H19 gene is an oncofetal
RNA. Mol Pathol 50:34–44
Ariel I, de Groot N, Hochberg A (2000a) Imprinted H19 gene expression in embryogenesis and
human cancer: the oncofetal connection. Am J Med Genet 91:46–50
Ariel I, Sughayer M, Fellig Y et al (2000b) The imprinted H19 gene is a marker of early recurrence
in human bladder carcinoma. Mol Pathol 53:320–323
Ayesh S, Matouk I, Schneider T et al (2002) Possible physiological role of H19 RNA. Mol
Carcinog 35:63–74
Bartolomei MS, Webber AL, Brunkow ME et al (1993) Epigenetic mechanisms underlying the
imprinting of the mouse H19 gene. Genes Dev 7:1663–1673
Bell AC, Felsenfeld G (2000) Methylation of a CTCF-dependent boundary controls imprinted
expression of the IGF2 gene. Nature 405:482–485
Bell AC, West AG, Felsenfeld G (1999) The protein CTCF is required for the enhancer blocking
activity of vertebrate insulators. Cell 98:387–396
Berteaux N, Lottin S, Monte
´
D et al (2005) H19 mRNA-like non-coding RNA promotes breast
cancer cell proliferation through its positive control by E2F1. J Biol Chem 280:29625–29636
Berteaux N, Aptel N, Cathala G et al (2008) A novel H19 antisense RNA overexpressed in breast
cancer contributes to paternal IGF2 expression. Mol Cell Biol 28:6731–6745
Bestor TH (2000) The DNA methyltransferases of mammals. Hum Mol Genet 9:2395–2402
Brannan CI, Dees EC, Ingram RS et al (1990) The product of the H19 gene may function as an
RNA. Mol Cell Biol 10:28–36
Cai X, Cullen BR (2007) The imprinted H19 noncoding RNA is a primary microRNA precursor.
RNA 13:313–316
Casola S, Pedone PV, Cavazzana AO et al (1997) Expression and parental imprinting of the H19
gene in human rhabdomyosarcoma. Oncogene 14:1503–1510
Charlier C, Segers K, Wagenaar D et al (2001) Human-ovine comparative sequencing of a 250-kb
imprinted domain encompassing the callipyge (clpg) locus and identification of six imprinted
transcripts: DLK1, DAT, GTL2, PEG11, antiPEG11, and MEG8. Genome Res 11:850–862
Chen CL, Ip SM, Cheng D et al (2000) Loss of imprinting of the IGF-II and H19 genes in epithelial
ovarian cancer. Clin Cancer Res 6:474–479
Constancia M, Dean W, Lopes S et al (2000) Deletion of a silencer element in IGF2 results in loss
of imprinting independent of H19. Nat Genet 26:203–206
Cooper MJ, Fischer M, Komitowski D et al (1996) Developmentally imprinted genes as markers
for bladder tumor progression. J Urol 155:2120–2127
Cooper PR, Smilinich NJ, Day CD et al (1998) Divergently transcribed overlapping genes
expressed in liver and kidney and located in the 11p15.5 imprinted domain. Genomics
49:38–51
Crider-Miller SJ, Reid LH, Higgins MJ et al (1997) Novel transcribed sequences within the BWS/
WT2 region in 11p15.5: tissue-specific expression correlates with cancer type. Genomics
46:355–363
Cui H, Onyango P, Brandenburg S et al (2002) Loss of imprinting in colorectal cancer linked to
hypomethylation of H19 and IGF2. Cancer Res 62:6442–6446
Davis E, Caiment F, Tordoir X et al (2005) RNAi-mediated allelic trans-interaction at the
imprinted Rtl1/Peg11 locus. Curr Biol 15:743–749
Dekker J, Rippe K, Dekker M et al (2002) Capturing chromosome conformation. Science
295:1306–1311
Douc-Rasy S, Coll J, Barrois M et al (1993) Expression of the human fetal BAC/H19 gene in
invasive cancer. Int J Oncol 2:753–758
Drewell RA, Brenton JD, Ainscough JF et al (2000) Deletion of a silencer element disrupts H19
imprinting independently of a DNA methylation epigenetic switch. Development 127:3419–3428
Drewell RA, Arney KL, Arima T et al (2002) Novel conserved elements upstream of the H19 gene
are transcribed and act as mesodermal enhancers. Development 129:1205–1213
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 437
Du M, Zhou W, Beatty LG et al (2004) The KCNQ1OT1 promoter, a key regulator of genomic
imprinting in human chromosome 11p15.5. Genomics 84:288–300
Dugimont T, Curgy JJ, Wernert N et al (1995) The H19 gene is expressed within both epithelial
and stromal components of human invasive adenocarcinomas. Biol Cell 85:117–124
Dugimont T, Montpellier C, Adriaenssens E et al (1998) The H19 TATA-less promoter is
efficiently repressed by wild-type tumor suppressor gene product p53. Oncogene 16:
2395–2401
Edwards CA, Ferguson-Smith AC (2007) Mechanisms regulating imprinted genes in clusters. Curr
Opin Cell Biol 19:281–289
Elkin M, Shevelev A, Schulze E et al (1995) The expression of the imprinted H19 and IGF-2 genes
in human bladder carcinoma. FEBS Lett 374:57–61
Elson DA, Bartolomei MS (1997) A 5
0
differentially methylated sequence and the 3
0
-flanking
region are necessary for H19 transgene imprinting. Mol Cell Biol 17:309–317
Engel N, Raval AK, Thorvaldsen JL et al (2008) Three-dimensional conformation at the H19/
IGF2 locus supports a model of enhancer tracking. Hum Mol Genet 17:3021–3029
Fedoriw AM, Stein P, Svoboda P et al (2004) Transgenic RNAi reveals essential function for
CTCF in H19 gene imprinting. Science 303:238–240
Feil R, Walter J, Allen ND et al (1994) Developmental control of allelic methylation in the
imprinted mouse IGF2 and H19 genes. Development 120:2933–2943
Ferguson-Smith AC, Sasaki H, Cattanach BM et al (1993) Parental-origin-specific epigenetic
modification of the mouse H19 gene. Nature 362:751–755
Fuks F (2003) DNA methyltransferases: from chromatin remodeling to cancer. Med Sci
19:477–480
Giannoukakis N, Deal C, Paquette J et al (1993) Parental genomic imprinting of the human IGF2
gene. Nat Genet 4:98–101
Grandjean V, O’Neill L, Sado T et al (2001) Relationship between DNA methylation, histone H4
acetylation and gene expression in the mouse imprinted IGF2-H19 domain. FEBS Lett
488:165–169
Hao Y, Crenshaw T, Moulton T et al (1993) Tumour-suppressor activity of H19 RNA. Nature
365:764–767
Hark AT, Tilghman SM (1998) Chromatin conformation of the H19 epigenetic mark. Hum Mol
Genet 7:1979–1985
Hark AT, Schoenherr CJ, Katz DJ et al (2000) CTCF mediates methylation-sensitive enhancer-
blocking activity at the H19/IGF2 locus. Nature 405:486–489
Hemberger M, Redies C, Krause R et al (1998) H19 and IGF2 are expressed and differentially
imprinted in neuroectoderm-derived cells in the mouse brain. Dev Genes Evol 208:393–402
Hibi K, Nakamura H, Hirai A et al (1996) Loss of H19 imprinting in esophageal cancer. Cancer
Res 56:480–482
Holmgren C, Kanduri C, Dell G et al (2001) CpG methylation regulates the IGF2/H19 insulator.
Curr Biol 11:1128–1130
Hu JF, Oruganti H, Vu TH et al (1998) The role of histone acetylation in the allelic expression of
the imprinted human insulin-like growth factor II gene. Biochem Biophys Res Commun
251:403–408
Isfort RJ, Cody DB, Kerckaert GA et al (1997) Role of the H19 gene in Syrian hamster embryo cell
tumorigenicity. Mol Carcinog 20:189–193
Ishihara K, Sasaki H (2002) An evolutionarily conserved putative insulator element near the
3’ boundary of the imprinted IGF2/H19 domain. Hum Mol Genet 11:1627–1636
Joubel A, Curgy JJ, Pelczar H et al (1996) The 5
0
part of the human H19 RNA contains cis-acting
elements hampering its translatability. Cell Mol Biol (Noisy-le-grand) 42:1159–1172
Jouvenot Y, Poirier F, Jami J et al (1999) Biallelic transcription of IGF2 and H19 in individual cells
suggests a post-transcriptional contribution to genomic imprinting. Curr Biol 9:1199–1202
Juan V, Crain C, Wilson C (2000) Evidence for evolutionarily conserved secondary structure in
the H19 tumor suppressor RNA. Nucleic Acids Res 28:1221–1227
438 N. Berteaux et al.
Kaffer CR, Srivastava M, Park KY et al (2000) A transcriptional insulator at the imprinted H19/
IGF2 locus. Genes Dev 14:1908–1919
Kanduri C, Holmgren C, Pilartz M et al (2000a) The 5’ flank of mouse H19 in an unusual
chromatin conformation unidirectionally blocks enhancer-promoter communication. Curr
Biol 10:449–457
Kanduri C, Pant V, Loukinov D et al (2000b) Functional association of CTCF with the insulator
upstream of the H19 gene is parent of origin-specific and methylation-sensitive. Curr Biol
10:853–856
Kato Y, Sasaki H (2005) Imprinting and looping: epigenetic marks control interactions between
regulatory elements. Bioessays 27:1–4
Kondo M, Suzuki H, Ueda R et al (1995) Frequent loss of imprinting of the H19 gene is often
associated with its overexpression in human lung cancers. Oncogene 10:1193–1198
Kurukuti S, Tiwari VK, Tavoosidana G et al (2006) CTCF binding at the H19 imprinting control
region mediates maternally inherited higher-order chromatin conformation to restrict enhancer
access to IGF2. Proc Natl Acad Sci USA 103:10684–10689
Landers M, Bancescu DL, Le Meur E et al (2004) Regulation of the large (approximately 1000 kb)
imprinted murine Ube3a antisense transcript by alternative exons upstream of Snurf/Snrpn.
Nucleic Acids Res 32:3480–3492
Lee MP, DeBaun MR, Mitsuya K et al (1999) Loss of imprinting of a paternally expressed
transcript, with antisense orientation to KVLQT1, occurs frequently in Beckwith–Wiedemann
syndrome and is independent of insulin-like growth factor II imprinting. Proc Natl Acad Sci
USA 96:5203–5208
Leibovitch MP, Solhonne B, Guillier M et al (1995) Direct relationship between the expression of
tumor suppressor H19 mRNA and c-mos proto-oncogene during myogenesis. Oncogene
10:251–260
Leighton PA, Ingram RS, Eggenschwiler J et al (1995) Disruption of imprinting caused by deletion
of the H19 gene region in mice. Nature 375:34–39
Lewis A, Redrup L (2005) Genetic imprinting: conflict at the Callipyge locus. Curr Biol 15:
R291–R294
Lewis A, Murrel A (2004) Genomic imprinting: CTCF protects the boundaries. Curr Biol
14:284–286
Li T, Hu JF, Qiu X et al (2008) CTCF regulates allelic expression of IGF2 by orchestrating a
promoter-polycomb repressive complex 2 intrachromosomal loop. Mol Cell Biol 28:6473–6482
Li YM, Franklin G, Cui HM et al (1998) The H19 transcript is associated with polysomes and may
regulate IGF2 expression in trans. J Biol Chem 273:28247–28252
Liao B, Hu Y, Herrick DJ et al (2005) The RNA-binding protein IMP-3 is a translational activator
of insulin-like growth factor II leader-3 mRNA during proliferation of human K562 leukemia
cells. J Biol Chem 280:18517–18524
Ling JQ, Li T, Hu JF et al (2006) CTCF mediates interchromosomal colocalization between Igf2/
H19 and Wsb1/Nf1. Science 312:269–272
Liu J, Kahri AI, Heikkila P et al (1995) H19 and insulin-like growth factor-II gene expression in
adrenal tumors and cultured adrenal cells. J Clin Endocrinol Metab 80:492–496
Lopes S, Lewis A, Hajkova P et al (2003) Epigenetic modifications in an imprinting cluster are
controlled by a hierarchy of DMRs suggesting longrange chromatin interactions. Hum Mol
Genet 12:295–305
Lottin S, Adriaenssens E, Dupressoir T et al (2002a) Overexpression of an ectopic H19 gene
enhances the tumorigenic properties of breast cancer cells. Carcinogenesis 23:1885–1895
Lottin S, Vercoutter-Edouart AS, Adriaenssens E et al (2002b) Thioredoxin post-transcriptional
regulation by H19 provides a new function to mRNA-like non-coding RNA. Oncogene
21:1625–1631
Lottin S, Adriaenssens E, Berteaux N et al (2005) The human H19 gene is frequently over-
expressed in myometrium and stroma during pathological endometrial proliferative events. Eur
J Cancer 41:168–177
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 439
Lustig O, Ariel I, Ilan J et al (1994) Expression of the imprinted gene H19 in the human fetus. Mol
Reprod Dev 38:239–246
Lutz M, Burke LJ, Barreto G et al (2000) Transcriptional repression by the insulator protein CTCF
involves histone deacetylases. Nucleic Acids Res 28:1707–1713
Maher ER and Reik W (2000) Beckwith-Wiedemann syndrome: imprinting in clusters revisited.
J Clin Invest 105:247–252
Manoharan H, Babcock K, Pitot HC (2004) Changes in the DNA methylation profile of the rat H19
gene upstream region during development and transgenic hepatocarcinogenesis and its role in
the imprinted transcriptional regulation of the H19 gene. Mol Carcinog 41:1–16
Milligan L, Antoine E, Bisbal C et al (2000) H19 gene expression is up-regulated exclusively
by stabilization of the RNA during muscle cell differentiation. Oncogene 19:5810–5816
Mitsuya K, Meguro M, Lee MP et al (1999) LIT1, an imprinted antisense RNA in the human
KvLQT1 locus identified by screening for differentially expressed transcripts using mono-
chromosomal hybrids. Hum Mol Genet 8:1209–1217
Miyamoto T, Hasuike S, Jinno Y et al (2002) The human ASCL2 gene escaping genomic
imprinting and its expression pattern. J Assist Reprod Genet 19:240–244
Moore T, Constancia M, Zubair M et al (1997) Multiple imprinted sense and antisense transcripts,
differential methylation and tandem repeats in a putative imprinting control region upstream of
mouse IGF2. Proc Natl Acad Sci USA 94:12509–12514
Murrell A, Heeson S, Reik W (2004) Interaction between differentially methylated regions partitions
the imprinted genes IGF2 and H19 into parent-specific chromatin loops. Nat Genet 36:889–893
Murrell A, Heeson S, Bowden L et al (2001) An intragenic methylated region in the imprinted
IGF2 gene augments transcription. EMBO Rep 2:1101–1106
Nielsen J, Kristensen MA, Willemoe
¨
s M et al (2004) Sequential dimerization of human zipcode-
binding protein IMP1 on RNA: a cooperative mechanism providing RNP stability. Nucleic
Acids Res 32:4368–4376
Nielsen FC, Nielsen J, Christiansen J (2001) A family of IGF-II mRNA binding proteins (IMP)
involved in RNA trafficking. Scand J Clin Lab Invest Suppl 234:93–99
Ng A, Tang JP, Goh CH et al (2003) Regulation of the H19 imprinting gene expression in human
nasopharyngeal carcinoma by methylation. Int J Cancer 104:179–187
Ohana P, Bibi O, Matouk I et al (2002) Use of H19 regulatory sequences for targeted gene therapy
in cancer. Int J Cancer 98:645–650
Ohana P, Schachter P, Ayesh B et al (2005) Regulatory sequences of H19 and IGF2 genes in
DNA-based therapy of colorectal rat liver metastases. J Gene Med 7:366–374
Ohlsson R, Hedborg F, Holmgren L et al (1994) Overlapping patterns of IGF2 and H19 expression
during human development: biallelic IGF2 expression correlates with a lack of H19 expres-
sion. Development 120:361–368
Okamoto K, Morison IM, Taniguchi T et al (1997) Epigenetic changes at the insulin-like growth
factor II/H19 locus in developing kidney is an early event in Wilms tumorigenesis. Proc Natl
Acad Sci USA 94:5367–5371
Okutsu T, Kuroiwa Y, Kagitani F et al (2000) Expression and imprinting status of human PEG8/
IGF2AS, a paternally expressed antisense transcript from the IGF2 locus, in Wilms’ tumors.
J Biochem (Tokyo) 127:475–483
Pachnis V, Belayew A, Tilghman SM (1984) Locus unlinked to alpha-fetoprotein under the
control of the murine raf and Rif genes. Proc Natl Acad Sci USA 81:5523–5527
Pachnis V, Brannan CI, Tilghman SM (1988) The structure and expression of a novel gene
activated in early mouse embryogenesis. EMBO J 7:673–681
Pandey RR, Ceribelli M, Singh PB et al (2004) NF-Y regulates the antisense promoter, bidirec-
tional silencing, and differential epigenetic marks of the Kcnq1 imprinting control region.
J Biol Chem 279:52685–52693
Paulsen M, Davies KR, Bowden LM et al (1998) Syntenic organization of the mouse distal
chromosome 7 imprinting cluster and the Beckwith-Wiedemann syndrome region in chromo-
some 11p15.5. Hum Mol Genet 7:1149–1159
440 N. Berteaux et al.
Paulsen M, El-Maarri O, Engemann S, Strodicke M et al (2000) Sequence conservation and
variability of imprinting in the Beckwith-Wiedemann syndrome gene cluster in human and
mouse. Hum Mol Genet 9:1829–1841
Pedone PV, Pikaart MJ, Cerrato F et al (1999) Role of histone acetylation and DNA methylation in the
maintenance of the imprinted expression of the H19 and IGF2 genes. FEBS Lett 458:45–50
Perincheri S, Dingle RW, Peterson ML et al (2005) Hereditary persistence of alpha-fetoprotein and
H19 expression in liver of BALB/cJ mice is due to a retrovirus insertion in the Zhx2 gene. Proc
Natl Acad Sci USA 102:396–401
Poirier F, Chan CT, Timmons PM et al (1991) The murine H19 gene is activated during embryonic
stem cell differentiation in vitro and at the time of implantation in the developing embryo.
Development 113:1105–1114
Prawitt D, Enklaar T, Klemm G et al (2000) Identification and characterization of MTR1, a novel
gene with homology to melastatin (MLSN1) and the trp gene family located in the BWS-WT2
critical region on chromosome 11p15.5 and showing allele-specific expression. Hum Mol
Genet 9:203–216
Rachmilewitz J, Elkin M, Rosensaft J et al (1995) H19 expression and tumorigenicity of chorio-
carcinoma derived cell lines. Oncogene 11:863–870
Rainho CA, Kowalski LP, Rogatto SR (2001) Loss of imprinting and loss of heterozygosity on
11p15.5 in head and neck squamous cell carcinomas. Head Neck 23:851–859
Reik W, Brown KW, Schneid H et al (1995) Imprinting mutations in the Beckwith–Wiedemann
syndrome suggested by altered imprinting pattern in the IGF2-H19 domain. Hum Mol Genet
4:2379–2385
Reik W, Dean W, Walter J (2001) Epigenetic reprogramming in mammalian development.
Science 293:1089–1093
Reik W, Murrell A (2000) Genomic imprinting silence across the border. Nature 405:408–409
Rinn JL, Kertesz M, Wang JK et al (2007) Functional demarcation of active and silent chromatin
domains in human HOX loci by noncoding RNAs. Cell 129:1311–1323
Ripoche MA, Kress C, Poirier F et al (1997) Deletion of the H19 transcription unit reveals the
existence of a putative imprinting control element. Genes Dev 11:1596–1604
Rougeulle C, Heard E (2002) Antisense RNA in imprinting: spreading silence through air. Trends
Genet 18:434–437
Rougeulle C, Cardoso C, Fontes M et al (1998) An imprinted antisense RNA overlaps UBE3A and
a second maternally expressed transcript. Nat Genet 19:15–16
Royo H, Cavaille
´
J (2008) Non coding RNAs in imprinted gene clusters. J Biol Cell 100:149–166
Runge S, Nielsen FC, Nielsen J et al (2000) H19 RNA binds four molecules of insulin-like growth
factor II mRNA-binding protein. J Biol Chem 275:29562–29569
Runte M, Huttenhofer A, Gross S et al (2001) The IC-SNURF-SNRPN transcript serves as a host
for multiple small nucleolar RNA species and as an antisense RNA for UBE3A. Hum Mol
Genet 10:2687–2700
Sasaki H, Jones PA, Chaillet JR et al (1992) Parental imprinting: potentially active chromatin of
the repressed maternal allele of the mouse insulin-like growth factor II (IGF2) gene. Genes
Dev 6:1843–1856
Seitz H, Youngson N, Lin SP et al (2003) Imprinted microRNA genes transcribed antisense to a
reciprocally imprinted retrotransposon-like gene. Nat Genet 34:261–262
Sleutels F, Zwart R, Barlow DP (2002) The non-coding Air RNA is required for silencing
autosomal imprinted genes. Nature 415:810–813
Steenman MJ, Rainier S, Dobry CJ et al (1994) Loss of imprinting of IGF2 is linked to reduced
expression and abnormal methylation of H19 in Wilms’ tumour. Nat Genet 7:433–439
Svensson K, Walsh C, Fundele R et al (1995) H19 is imprinted in the choroids plexus and
leptomeninges of the mouse fetus. Mech Dev 51:31–37
Szabo P, Tang SH, Rentsendorj A et al (2000) Maternal-specific footprints at putative CTCF sites
in the H19 imprinting control region give evidence for insulator function. Curr Biol
10:607–610
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 441
Szabo PE, Tang SH, Silva FJ et al (2004) Role of CTCF binding sites in the IGF2/H19 imprinting
control region. Mol Cell Biol 24:4791–4800
Takai D, Gonzales FA, Tsai YC et al (2001) Large scale mapping of methylcytosines in CTCF-
binding sites in the human H19 promoter and aberrant hypomethylation in human bladder
cancer. Hum Mol Genet 10:2619–2626
Tanos V, Prus D, Ayesh S et al (1999) Expression of the imprinted H19 oncofetal RNA in
epithelial ovarian cancer. Eur J Obstet Gynecol Reprod Biol 85:7–11
Tanos V, Ariel I, Prus D et al (2004) H19 and IGF2 gene expression in human normal, hyper-
plastic, and malignant endometrium. Int J Gynecol Cancer 14:521–525
Tessier CR, Doyle GA, Clark BA et al (2004) Mammary tumor induction in transgenic mice
expressing an RNA-binding protein. Cancer Res 64:209–214
Thakur N, Tiwari VK, Thomassin H et al (2004) An antisense RNA regulates the bidirectional
silencing property of the Kcnq1 imprinting control region. Mol Cell Biol 24:7855–7862
Thorvaldsen JL, Duran KL, Bartolomei MS (1998) Deletion of the H19 differentially methylated
domain results in loss of imprinted expression of H19 and IGF2. Genes Dev 12:3693–3702
Thorvaldsen JL, Mann MR, Nwoko O et al (2002) Analysis of sequence upstream of the endoge-
nous H19 gene reveals elements both essential and dispensable for imprinting. Mol Cell Biol
22:2450–2462
Tremblay KD, Duran KL, Bartolomei MS (1997) A 5
0
2-kilobase-pair region of the imprinted
mouse H19 gene exhibits exclusive paternal methylation throughout development. Mol Cell
Biol 17:4322–4329
Tsang P, Gilles F, Yuan L et al (1995) A novel L23-related gene 40 kb downstream of the
imprinted H19 gene is biallelically expressed in mid-fetal and adult human tissues. Hum
Mol Genet 4:1499–1507
Ulaner GA, Yang Y, Hu JF et al (2003) CTCF binding at the insulin-like growth factor-II (IGF2)/
H19 imprinting control region is insufficient to regulate IGF2/H19 expression in human
tissues. Endocrinology 144:4420–4426
Verkerk AJ, Ariel I, Dekker MC et al (1997) Unique expression patterns of H19 in human
testicular cancers of different etiology. Oncogene 14:95–107
Verona RI, Mann MR, Bartolomei MS (2003) Genomic imprinting: intricacies of epigenetic
regulation in clusters. Annu Rev Cell Dev Biol 19:237–259
Vu TH, Chuyen NV, Li T et al (2003) Loss of imprinting of IGF2 sense and antisense transcripts in
Wilms’ tumor. Cancer Res 63:1900–1905
Webber AL, Ingram RS, Levorse JM et al (1998) Location of enhancers is essential for the
imprinting of H19 and IGF2 genes. Nature 391:711–715
Weber M, Hage
`
ge H, Murrell A et al (2003) Genomic imprinting controls matrix attachment
regions in the Igf2 gene. Mol Cell Biol 23:8953–8959
Wilkin F, Paquette J, Ledru E et al (2000) H19 sense and antisense transgenes modify insulin-like
growth factor-II mRNA levels. Eur J Biochem 267:4020–4027
Wilkins JF, Haig D (2003) What good is genomic imprinting: the function of parent-specific gene
expression. Nat Rev Genet 4:359–368
Wiseman RW, Montgomery JC, Hosoi J et al (1991) Identification of genes associated with tumor
suppression in Syrian hamster embryo cells. Environ Health Perspect 93:105–109
Westerman BA, Poutsma A, Looijenga LH et al (2001) The Human Achaete Scute Homolog
2 gene contains two promotors, generating overlapping transcripts and encoding two proteins
with different nuclear localization. Placenta 22:511–518
Wolffe AP (2000) Transcriptional control: imprinting insulation. Curr Biol 10:R463–465
Yoon YS, Jeong S, Rong Q et al (2007) Analysis of the H19ICR Insulator. Mol Cell Biol
27:3499–3510
Yang Y, Hu JF, Ulaner GA et al (2003) Epigenetic regulation of IGF2/H19 imprinting at CTCF
insulator binding sites. J Cell Biochem 90:1038–1055
Yekta S, Shih IH, Bartel DP (2004) MicroRNA-directed cleavage of HOXB8 mRNA. Science
304:594–596
442 N. Berteaux et al.
Youngson NA, Kocialkowski S, Peel N et al (2005) A small family of sushi-class retrotransposon-
derived genes in mammals and their relation to genomic imprinting. J Mol Evol 61:481–490
Yu W, Gius D, Onyango P et al (2008) Epigenetic silencing of tumour suppressor gene p15 by its
antisense RNA. Nature 451:202–206
Zemel S, Bartolomei MS, Tilghman SM (1992) Physical linkage of two mammalian imprinted
genes, H19 and insulin-like growth factor 2. Nat Genet 2:61–65
Zhang Y, Tycko B (1992) Monoallelic expression of the human H19 gene. Nat Genet 1:40–44
Zhao Z, Tavoosidana G, Sjolinder M et al (2006) Circular chromosome conformation capture
(4C) uncovers extensive networks of epigenetically regulated intra- and interchromosomal
interactions. Nat Genet 38:1341–1347
Noncoding RNAs at H19/IGF2 Locus: Role in Imprinting, Gene Expression 443
Thesis
With the completion of the Human Genome Project in 2003, it has become clear that only ~2% of the human genome encodes for proteins. The remaining 98% of the genome is de facto noncoding and it was erroneously dubbed “junk DNA” for a long time. Today we know, thanks to the advent of high-throughput sequencing technologies, that the larger fraction of the nonrepetitive portion of the genome is actively transcribed, albeit not encoding for proteins. These transcripts instead exert their functions independently of translation, in the form of RNA, and are therefore termed non-coding RNAs (ncRNAs). Among them, a specific class of ncRNAs known as long non-coding RNAs (lncRNAs), has attracted most of the attention of the scientific community over the past decade. LncRNAs have been involved in gene regulation, both at the transcriptional and post-transcriptional level, through several different mechanisms of action, ranging from the recruitment of chromatin-modifying enzymes to the sponging of microRNAs. With the ability to interact with other macromolecules such as DNA, RNA and proteins, lncRNAs have been proven to be essential in almost any aspect of the cell’s life, from physiological to pathological processes. In this essay, I aim to summarize the most relevant knowledge about lncRNAs from both their structural and functional aspects according to the literature. After describing the general features of lncRNAs, I deepen into their role as epigenetic regulators and their involvement in key processes such as the maintenance of stemness, differentiation, neurogenesis, cancer development and epigenetics.
Article
Full-text available
Genomic imprinting at the Igf2/H19 locus originates from allele-specific DNA methylation, which modifies the affinity of some proteins for their target sequences. Here, we show that AT-rich DNA sequences located in the vicinity of previously characterized differentially methylated regions (DMRs) of the imprinted Igf2 gene are conserved between mouse and human. These sequences have all the characteristics of matrix attachment regions (MARs), which are known as versatile regulatory elements involved in chromatin structure and gene expression. Combining allele-specific nuclear matrix binding assays and real-time PCR quantification, we show that retention of two of these Igf2 MARs (MAR0 and MAR2) in the nuclear matrix fraction depends on the tissue and is specific to the paternal allele. Furthermore, on this allele, the Igf2 MAR2 is functionally linked to the neighboring DMR2 while, on the maternal allele, it is controlled by the imprinting-control region. Our work clearly demonstrates that genomic imprinting controls matrix attachment regions in the Igf2 gene.
Article
Full-text available
Aims—To investigate the expression of the imprinted oncofetal H19 gene in human bladder carcinoma and to examine the possibility of using it as a tumour marker, similar to other oncofetal gene products. Methods—In situ hybridisation for H19 RNA was performed on 61 first biopsies of bladder carcinoma from Hadassah Medical Centre in Jerusalem. The intensity of the reaction and the number of tumour cells expressing H19 in each biopsy were evaluated in 56 patients, excluding biopsies with carcinoma in situ. The medical files were searched for demographic data and disease free survival. Results—More than 5% of cells expressed H19 in 47 of the 56 (84%) biopsies. There was a decrease in the number of cells expressing H19 with increasing tumour grade (loss of differentiation) (p = 0.03). Disease free survival from the first biopsy to first recurrence was significantly shorter in patients with tumours having a larger fraction of H19 expressing cells, controlling for tumour grade. This was also supported by the selective analysis of tumour recurrence in patients with grade I tumours. Conclusions—It might be possible to use H19 as a prognostic tumour marker for the early recurrence of bladder cancer. In addition, for the gene therapy of bladder carcinoma that is based on the transcriptional regulatory sequences of H19, the expression of H19 in an individual biopsy could be considered a predictive tumour marker for selecting those patients who would benefit from this form of treatment.
Article
Marja J. C. Steenman, Shirley Rainier, Craig J. Dobry, Paul Grundy, Isabelle L. Horon & Andrew P. Feinberg Nature Genetics 7,433–439 In Table 1, heading WTs with LOI, third row (NK), the last five digits (0) should be in the appropriate columns 1,2,3,4 and 5. The thin-lined boxes in Table 2 wereleft out.
Article
Imprinted ncRNA (non-coding RNA) genes represent a family of untranslated transcripts that are mono-allelically expressed in a parent-of-origin manner (their expression is restricted to either the maternal or the paternal allele). Although the expression of a few long imprinted ncRNAs act as cis-acting silencers in the epigenetic regulation of chromatin at imprinted gene clusters, many of them fall into the growing class of small regulatory RNAs, namely C/D small nucleolar RNAs, microRNAs and also likely piRNAs (Piwi-interacting RNAs), which are known to act as antisense trans-acting regulators of gene expression (for example, site-specific RNA modifications and RNA-mediated gene silencing). Although their biological functions remain elusive, recent studies have pointed to their functional importance in development, in brain plasticity and also perhaps in some pathological situations, such as cancers or Prader-Willi syndrome. In this review, we summarize our current understanding of the molecular and biological roles of these ncRNAs, both in terms of their contribution to genomic imprinting control, as well as in terms of cellular RNA targets they might interact with.
Article
The highly conserved zinc-finger protein, CTCF, is a candidate tumor suppressor protein that binds to highly divergent DNA sequences. CTCF has been connected to multiple functions in chromatin organization and gene regulation including chromatin insulator activity and transcriptional enhancement and silencing. Here we show that CTCF harbors several autonomous repression domains. One of these domains, the zinc-finger cluster, silences transcription in all cell types tested and binds directly to the co-repressor SIN3A. Two distinct regions of SIN3A, the PAH3 domain and the extreme C-terminal region, bind independently to this zinc-finger cluster. Analysis of nuclear extract from HeLa cells revealed that CTCF is also capable of retaining functional histone deacetylase activity. Furthermore, the ability of regions of CTCF to retain deacetylase activity correlates with the ability to bind to SIN3A and to repress gene activity. We suggest that CTCF driven repression is mediated in part by the recruitment of histone deacetylase activity by SIN3A.
Article
The imprinted domain on human chromosome 15 consists of two oppositely imprinted gene clusters, which are under the coordinated control of an imprinting center (IC) at the 5' end of the SNURF-SNRPN gene. One gene cluster spans the centromeric part of this domain and contains several genes that are transcribed from the paternal chromosome only (MKRN3, MAGEL2, NDN, SNURF-SNRPN, HBII-13, HBII-85 and HBII-52). Apart from the HBII small nucleolar RNA (snoRNA) genes, each of these genes is associated with a 5' differentially methylated region (DMR). The second gene cluster maps to the telomeric part of the imprinted domain and contains two genes (UBE3A and ATP10C), which in some tissues are preferentially expressed from the maternal chromosome. So far, no DMR has been identified at these loci. Instead, maternal-only expression of UBE3A may be regulated indirectly through a paternally expressed antisense transcript. We report here that a processed antisense transcript of UBE3A starts at the IC. The SNURF-SNRPN sense/ UBE3A antisense transcription unit spans more than 460 kb and contains at least 148 exons, including the previously identified IPW exons. It serves as the host for the previously identified HBII-13, HBII-85 and HBII-52 snoRNAs as well as for four additional snoRNAs (HBII-436, HBII-437, HBII-438A and HBII-438B), newly identified in this study. Almost all of those snoRNAs are encoded within introns of this large transcript. Northern blot analysis indicates that most if not all of these snoRNAs are indeed expressed by processing from these introns. As we have not obtained any evidence for other genes in this region, which, from the mouse data appears to be critical for the neonatal Prader-Willi syndrome phenotype, a lack of these snoRNAs may be causally involved in this disease.
Article
The maternally expressed H19 gene is transcribed as an untranslated RNA that serves as a riboregulator. We have previously reported that this transcript accumulates in epithelial cells in ∼10% of breast cancers. To gain further insight on how the overexpression of the H19 gene affects the phenotype of human breast epithelial cells, we investigated the oncogenic potential of RNA that was abundantly expressed from MDA-MB-231 breast cancer cells stably transfected with the genomic sequence of the human H19 gene. The amount of H19 RNA did not affect cell proliferation capacity, timing of cell cycle phases or anchorage-dependent ability of H19-transfected clones in vitro. But in anchorage-independent growth assays the H19-recombined cells formed more and larger colonies in soft-agar versus control cells. To explore this phenotypic change, we analysed tumour development after subcutaneous injection of H19-recombined cells into scid mice. Results showed that H19 overexpression promotes tumour progression. These data support the hypothesis that an overload of H19 transcript is associated with cells exhibiting higher tumorigenic phenotypes and therefore we conclude that the H19 gene has oncogenic properties in breast epithelial cells.
Article
Parent-of-origin-specific expression of the mouse insulin-like growth factor 2 (Igf2) gene and the closely linked H19 gene are regulated by an intervening 2 kb imprinting control region (ICR), which displays parent-specific differential DNA methylation [1,2]. Four 21 bp repeats are embedded within the ICR and are conserved in the putative ICR of human and rat Igf2 and H19, suggesting that the repeats have a function [3,4]. Here, we report that prominent DNA footprints were found in vivo on the unmethylated maternal ICR at all four 21 bp repeats, demonstrating the presence of protein binding. The methylated paternal ICR displayed no footprints. Significantly, the maternal-specific footprints were localized to putative binding sites for CTCF, a highly conserved zinc-finger DNA-binding protein with multiple roles in gene regulation including that of chromatin insulator function [5,6]. These results strongly suggest that the maternal ICR functions as an insulator element in regulating mutually exclusive expression of Igf2 and H19 in cis. The chromatin structure of the Igf2 and H19 ICR in vivo was investigated in mouse primary embryo fibroblasts (PEFs) that carry only maternally or paternally inherited copies of the distal chromosome 7 region on which these genes reside (MatDup.d7 and PatDup.d7, respectively). In MatDup.d7 embryos, on each of the two distal chro-mosome 7 regions, Igf2 is silent whereas H19 is active in cis, and in PatDup.d7 embryos, the opposite is true [7]. PEFs were first treated with DNase I, dimethyl sulfate (DMS) or ultraviolet (UV) light. The modification of DNA by these reagents is sensitive to bound protein, and areas of protein–DNA interaction show as footprints on ligation-mediated-PCR (LMPCR) genomic sequencing ladders. LMPCR analysis of DNA treated with these reagents after its isolation from PEFs, or in vitro analysis, served as a reference.