ArticlePDF Available

Human imprinted retrogenes exhibit non-canonical imprint chromatin signatures and reside in non-imprinted host genes

Authors:
  • University Clermont- Auvergne (UCA)

Abstract and Figures

Imprinted retrotransposed genes share a common genomic organization including a promoter-associated differentially methylated region (DMR) and a position within the intron of a multi-exonic 'host' gene. In the mouse, at least one transcript of the host gene is also subject to genomic imprinting. Human retrogene orthologues are imprinted and we reveal that human host genes are not imprinted. This coincides with genomic rearrangements that occurred during primate evolution, which increase the separation between the retrogene DMRs and the host genes. To address the mechanisms governing imprinted retrogene expression, histone modifications were assayed at the DMRs. For the mouse retrogenes, the active mark H3K4me2 was associated with the unmethylated paternal allele, while the methylated maternal allele was enriched in repressive marks including H3K9me3 and H4K20me3. Two human retrogenes showed monoallelic enrichment of active, but not of repressive marks suggesting a partial uncoupling of the relationship between DNA methylation and repressive histone methylation, possibly due to the smaller size and lower CpG density of these DMRs. Finally, we show that the genes immediately flanking the host genes in mouse and human are biallelically expressed in a range of tissues, suggesting that these loci are distinct from large imprinted clusters.
Content may be subject to copyright.
Human imprinted retrogenes exhibit non-canonical
imprint chromatin signatures and reside in
non-imprinted host genes
David Monk
1,
*, Philippe Arnaud
2
, Jennifer M. Frost
3,4
, Andrew J. Wood
4
,
Michael Cowley
4
, Alejandro Martin-Trujillo
1
, Amy Guillaumet-Adkins
1
,
Isabel Iglesias Platas
5
, Cristina Camprubi
1
, Deborah Bourc’his
6
, Robert Feil
2
,
Gudrun E. Moore
3
and Rebecca J. Oakey
4
1
Imprinting and Cancer Group, Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical
Research (IDIBELL), L’Hospitalet de Llobregat, 08907, Barcelona, Spain,
2
Institute of Molecular Genetics
(IGMM), CNRS UMR5535 and University of Montpellier, 1919 route de Mende, 34293 Montpellier, France,
3
Clinical and Molecular Genetics Unit, Institute of Child Health, University College London,
4
Department of
Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London, SE1 9RT, UK,
5
Servicio de
Neonatologı
´a, Hospital Sant Joan de De
´u, Barcelona, Spain and
6
UMR 3215/Inserm U934, Unite
´de ge
´ne
´tique
et biologie du de
´veloppement, Institut Curie, 26 rue d’Ulm, Paris Cedex 05, France
Received October 7, 2010; Revised November 5, 2010; Accepted November 12, 2010
ABSTRACT
Imprinted retrotransposed genes share a common
genomic organization including a promoter-
associated differentially methylated region (DMR)
and a position within the intron of a multi-exonic
‘host’ gene. In the mouse, at least one transcript
of the host gene is also subject to genomic imprint-
ing. Human retrogene orthologues are imprinted
and we reveal that human host genes are not
imprinted. This coincides with genomic rearrange-
ments that occurred during primate evolution, which
increase the separation between the retrogene
DMRs and the host genes. To address the mech-
anisms governing imprinted retrogene expression,
histone modifications were assayed at the DMRs.
For the mouse retrogenes, the active mark
H3K4me2 was associated with the unmethylated
paternal allele, while the methylated maternal allele
was enriched in repressive marks including
H3K9me3 and H4K20me3. Two human retrogenes
showed monoallelic enrichment of active, but not
of repressive marks suggesting a partial uncoupling
of the relationship between DNA methylation and
repressive histone methylation, possibly due to the
smaller size and lower CpG density of these DMRs.
Finally, we show that the genes immediately
flanking the host genes in mouse and human are
biallelically expressed in a range of tissues, sug-
gesting that these loci are distinct from large
imprinted clusters.
INTRODUCTION
Genomic imprinting is a form of epigenetic gene regula-
tion that results in allelic expression dictated by parental
origin (1). Differential DNA methylation is a major com-
ponent in regulating this process. Discrete differentially
methylated cis-acting regions, known as imprinting
control regions (ICRs), orchestrate the monoallelic expres-
sion of numerous genes within imprinted domains and are
established while the maternal and paternal genomes are
physically separated in their respective germ lines. To
date, all imprinted domains are known to contain
regions of differential DNA methylation (DMRs) that
are deposited in CpG-rich sequences during oogenesis or
spermatogenesis by the DNMT3A/DNMT3L de novo
methyltransferase complex (2–4). A subset of maternally
DNA-methylated germline DMRs require the activity
of the amine oxidase domain 1 containing histone
demethylase AOF1/KDM1. This demethylase is presum-
ably needed to remove any permissive histone H3 lysine 4
(H3K4) methylation present at these CpG islands in the
growing oocytes (5). After fertilization, these regions of
differential DNA methylation are maintained in somatic
*To whom correspondence should be addressed. Tel: +34 93260 7500 (ext. 3165); Fax: +34 93260 7219; Email: dmonk@iconcologia.net
Published online 7 February 2011 Nucleic Acids Research, 2011, Vol. 39, No. 11 4577–4586
doi:10.1093/nar/gkq1230
ßThe Author(s) 2011. Published by Oxford University Press.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/
by-nc/2.5), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
tissues by DNMT1 (6), and are associated with numerous
histone modifications.
It has previously been shown that DMRs have a con-
stitutional histone signature that comprises histone H3
lysine 9 trimethylation (H3K9me3), Histone H4 lysine
20 trimethylation (H4K20me3) and symmetrical histone
H2A/H4 arginine 3 dimethylation (H2A/H4R3me2s)
on the DNA methylated allele (7,8). This is in contrast
to the enrichment of the transcriptionally permissive
H3K4me2/3 mark on the unmethylated allele (9). A
number of genes that are imprinted solely in the mouse
placenta, have been shown recently to require allelic re-
pressive histone modifications at their own DNA-
unmethylated promoters to maintain allelic expression
(10–13).
Imprinted genes have diverse evolutionary origins.
Some imprinted genes are products of retrotransposition
from parental genes on the X chromosome. Four im-
printed retrogenes in the mouse—Mcts2,Nap1l5,U2af1-
rs1 (also called Zrsr1) and Inpp5f_v2—are associated with
DMRs at their promoters, and reside in introns of multi-
exonic host genes. In all cases, at least one transcript of the
host is also subject to imprinting. We have previously
shown that the mouse retrogene Mcts2 influences the
choice of polyadenylation (polyA) site for transcripts of
the host gene H13 in an allele-specific manner (14).
Expression of Mcts2 from the paternal allele causes H13
transcripts to terminate upstream of the retrogene. On the
maternal chromosome, H13 utilizes downstream polyA
sites because the Mcts2 DMR is methylated and the
retrogene silenced. A recent transcriptome-wide analysis,
using the ultra sensitive RNA-seq technology which is cap-
able of detecting subtle biases in allelic transcription, has
suggested that one transcript variant of Inpp5f, the host
gene of Inpp5f_v2, and Herc3, the Nap1l5 host gene, are
also subject to isoform-specific allelic expression (15).
To investigate whether the human orthologues of the
X-derived imprinted retrogenes influence allele-specific ex-
pression of their respective host genes, and whether this
influence extends to neighboring genes, we have analyzed
the allelic expression of retrogenes, host and flanking
genes in humans. The U2af1-rs1 gene does not have a
human counterpart. We find that the human orthologues
INPP5F_V2,NAP1L5 and MCTS2 are paternally ex-
pressed in a wide range of fetal tissues, and that their
promoters are embedded in maternally DNA-methylated
regions. In humans, the host genes are not subject to im-
printing, probably due to differing exon/30-UTR positions
in relation to the retrogene integration sites. The genes
immediately flanking the host genes are biallelically ex-
pressed in both mice and humans, showing retrogene-host
pairs do not form parts of larger imprinted clusters. In
mice, the allelic chromatin of these DMRs conforms to
the constitutional histone modification signature with
the repressive modifications H3K9me3, H4K20me3 and
H2A/H4R3me2s enriched on the DNA methylated
allele, and the permissive modification H3K4me2
enriched on the unmethylated allele. These patterns of
histone modifications are not conserved at the human
NAP1L5 and MCTS2 promoters, correlating with
reduced CpG content and CpG island size, which we
speculate may influence the recruitment of the histone
methyltransferases (HMTs).
MATERIALS AND METHODS
Human tissues
A cohort comprising 65 fetal tissue sets (8–18 weeks) with
corresponding maternal blood sample and 96-term placen-
tal samples are from the Moore Tissue bank and is
described elsewhere (16). An additional 96 human
placenta samples were obtained from the Hospital
St Joan De Deu collection (Barcelona, Spain). Normal
peripheral blood was collected from adult volunteers
aged between 19–60-years old. DNA and RNA extraction
and cDNA synthesis were carried out as previously
described (11). Ethical approval for adult blood and
fetal tissue collection was granted by the Hammersmith,
Queen Charlotte’s and Chelsea and Acton Hospital
Research Ethics Committee (Project Registration 2001/
6029 and 2001/6028); Collection of the HSJD placental
cohort was granted by the ethical committee of Hospital
St Joan De Deu Ethics Committee (Study number 35/07).
Cell lines and mouse crosses
Wild-type mouse embryos and placentas were produced
by crossing C57BL/6 females with either Mus musculus
molosinus (JF1) or Mus musculus castaneus (C) male
mice. RNA and DNA from Dnmt3l
/+
mice (BxC,
C57BL/6 mother and Castaneus father) was isolated and
extracted as previously described (2). The E9.5 Dnmt3l
/+
embryos (BxJ) used to assess Nap1l5 expression were a
kind gift from Dr Kenichiro Hata (NRICHD, Okura,
Tokyo, Japan). The human TCL1 and 2 placental tropho-
blast cell lines were grown in DMEM supplemented with
10% FCS and antibiotics.
Allelic expression analysis
Genotypes of DNA were obtained for exonic SNPs
identified in the UCSC browser (NCBI36/hg18,
Assembly 2006) by PCR. Sequences were interrogated
using Sequencher v4.6 (Gene Codes Corporation, MI,
USA) to distinguish informative heterozygote samples.
Informative samples were analysed by RT–PCR in corres-
ponding cDNA using, where possible, intron-crossing
primers that incorporated the heterozygous SNP in the
resulting amplicon (Supplementary Table S1). RT–PCRs
were performed using cycle numbers determined to be
within the exponential phase of the PCR, which varied
for each gene, but was between 32–40 cycles. The RT–
PCRs for HERC3A,HERC3C and both isoforms of
Abcg2 were analyzed by nested RT–PCR, with the first
PCR amplified for 25 cycles, with 5 ml of this product
used as template for the second round PCR which was
limited to 30 cycles.
Real-time qRT–PCR
All PCRs were run in triplicate from the same sample on
either an ABI Prism 7700 sequence detector or a 7900 Fast
real-time PCR machine (Applied Biosystems) following
4578 Nucleic Acids Research, 2011, Vol. 39, No. 11
the manufacturer’s protocol. All primers were optimized
using SYBR Green amplification followed by melt curve
analysis to ensure that amplicons were free of primer
dimer products. Thermal cycling parameters included
Taq polymerase activation at 95C for 10 min for one
cycle, repetitive denaturation at 95C for 15 s, and anneal-
ing at 60C for 1 min for 40 cycles. All resulting triplicate
cycle threshold (C
t
) values had to be within one C
t
of each
other. The quantitative values for each triplicate were
determined as a ratio with the level of Gapdh, measured
in the same sample, with the mean providing relative
expression values.
Analysis of allelic DNA methylation
Approximately 1 mg DNA was subjected to sodium
bisulphite treatment and purified using the EZ GOLD
methylation kit (ZYMO, Orange, CA, USA). Bisulphite
specific primers for each region were used with Hotstar
Taq polymerase (Qiagen, West Sussex, UK) at 45 cycles
and the resulting PCR product cloned into pGEM-T Easy
vector (Promega) for subsequent sequencing. MeDIP was
performed on 6 mg sonicated (Diagenode Bioruptor)
genomic DNA with an average size of 150 bp. Samples
were denatured and incubated with a monoclonal
antibody against 5-methylcytidine (Eurogentec).
Immunoprecipitated DNA was then isolated using IgG
Dynabeads (Dynal Biotech), digested with proteinase K
and phenol–chloroform extraction was followed by
ethanol precipitation. MeDIP enrichment was verified by
duplexed PCR for the methylated SERPIN B5 promoter
and the unmethylated UBE2B promoter. Southern
blotting was performed following standard protocols
using methylation-sensitive restriction enzymes. Digested
DNA was subjected to agarose gel electrophoresis and
transferred to Hybond N+ membrane (Amersham).
Radio-labeled PCR product probes were hybridized
over-night at 65C, and subsequently washed in increasing
stringency SSC/0.1% SDS washes. PCR primers used
to generate probes are listed in Supplementary Table S1.
Chromatin immunoprecipitation
Two adult leukocyte samples and the TCL1 and TCL2 cell
lines were used in addition to E18.5 mouse embryos
for chromatin immunoprecipitation (ChIP). ChIP was
carried out as previously described (11,13) using the fol-
lowing Upstate Biotechnology antisera directed against
H3K4me2 (07-030), H3K9me2 (07-441), H3K9me3
(060904589), H3K9ac (07-352), H3K27me3 (07-449),
H4K20me3 (07-463) (Upstate Biotechnology) and H2A/
H4R3me2s (Abcam ab5823 97454/520317). ChIPed DNA
was subjected to allele-specific PCR. Polymorphisms
within 1 kb of the CpG island were identified by
interrogating SNP databases or genomic sequencing (see
Supplementary Table S1 for primer sequences and
location). Only ChIP sample sets that showed enrichment
for additional ICRs were used in the analysis.
Precipitation levels in the ChIP samples were
determined by real-time PCR amplification, using SYBR
Green PCR kit (Applied Biosystems). Each PCR was run
in triplicate and results are presented as fold enrichment
(comparison to mock) and normalized to the level of pre-
cipitation at the SNURF-ICR, a control for both active
and repressive histone modifications located on human
chromosome 15.
RESULTS
MCTS2 does not influence allelic expression of HM13
The H13 gene on mouse chromosome 2 is known to
generate at least five transcripts, all originating from a
single promoter, but differing in polyA site usage (14).
The utilization of these alternative polyA sites is
influenced by the paternally expressed Mcts2 imprinted
retrogene and the CpG island that comprises its DMR.
The short H13d and etranscripts are paternally expressed,
whereas the H13a,band ctranscripts, that extend through
Mcts2 and the DMR to the canonical polyA site are ma-
ternally expressed (Figure 1A). To assess allelic expression
in humans we identified transcribed SNPs unique to each
isoform, and allele-specific assays were carried out in a
selection of human first trimester fetal tissues and term
placentas. The human MCTS2 gene (also known as
PSIMCT-1, MCTS1-pseudogene) is imprinted in a
variety of fetal tissues (Figure 1B and Supplementary
Table S2). MCTS2 differs from its mouse orthologue as
it can splice into the last eight exons of the HM13 host
gene (Figure 1B). Sequence analyses revealed that in
addition to the highly conserved MCTS2 open reading
frame, this RNA has the potential to be bi-cistronic,
encoding for a chimeric protein lacking the first 151
amino acids of HM13, but sharing 243 amino acids in
the C-terminus.
The MCTS2 promoter is embedded within a DMR
(Figure 1B and Supplementary Figure S1A), while the
HM13 host gene originates from a CpG island that is
unmethylated in all the tissues analysed. Alignment of
human expressed sequence tags (ESTs) revealed a
number of transcripts. The expression of the human
short HM13D isoform (Genbank NM_178982), which ter-
minates prior to the transcriptional start site (TSS) of
MCTS2, and the full-length transcript HM13C
(Genbank NM_030789) are biallelic in all fetal tissues
analysed (Figure 1B and Supplementary Table S2).
The NAP1L5 promoter is not within a CpG island,
but is a DMR
The paternally expressed Nap1l5 retrogene was first
identified in a genome-wide screen for differential DNA
methylation, and was reported to be predominantly pater-
nally expressed in mouse brain (17). The host gene, Herc3,
gives rise to a number of transcript isoforms. At least two
short isoforms (Herc3b and c) are expressed from the
paternal allele in mouse brain [A.J. Wood and R.J.
Oakey, unpublished data, (15)]. The full length Herc3a
transcript has a maternal expression bias, similar to the
full length H13 isoforms (14). We detect paternal expres-
sion of NAP1L5 in all fetal tissues analyzed. The human
HERC3 gene also contains one long (HERC3A, Genbank
NM_014606) and two short isoforms (HERC3B,
Genbank BC038960; HERC3C, Genbank AK296397).
Nucleic Acids Research, 2011, Vol. 39, No. 11 4579
A
B
C
D
E
F
Figure 1. (A) Map of the H13 locus located on mouse chromosome 2, showing the location of the various imprinted transcripts and CpG islands
(red transcripts are maternally expressed, blue are paternally expressed and grey are expressed from both parental alleles. Arrows represent direction
of transcription). (B) Schematic of the human HM13 gene on chromosome 20, showing the distribution of exons and insertion of MCTS2 into intron
4. The methylation status of the HM13 promoter CpG island and MCTS2 CpG island were examined by bisulphite PCR. Each circle represents a
single CpG dinucleotide and the strand. Filled circle, a methylated cytosine; open circle, unmethylated cytosine. The sequence traces show allelic
expression for MCTS2 and HM13 isoforms (for clarity only sequence traces for MCTS2 BC053868 are shown). (C) A map of the Herc3 domain on
mouse chromosome 6. (D) The human NAP1L5 gene and the insertion into intron 22 of HERC3.(E) A schematic map of the Inpp5f gene on mouse
chromosome 7, and (F) the orthologous region on human chromosome 10.
4580 Nucleic Acids Research, 2011, Vol. 39, No. 11
Our allele-specific assays are suggestive of biallelic expres-
sion of all isoforms in all tissues including brain (Figure
1C and D, and Supplementary Table S2), although our
methodology is not directly comparable with RNA-seq,
which can detect subtle expression biases. The HERC3
transcripts initiate from an unmethylated CpG island,
whereas the NAP1L5 promoter is DNA methylated on
the maternal allele, even though the region is not statistic-
ally a CpG island in humans (Figure 1D).
INPP5F_V2 is imprinted in numerous human tissues
We previously identified a neural-specific, paternally ex-
pressed Inpp5f_v2 transcript using expression microarrays
(18). Like the other murine imprinted retrogene loci, the
host gene Inpp5f exhibits isoform-specific expression, with
a maternal expression bias in a truncated shorter tran-
script (Genbank AK039468) (15). In addition, another
transcript, Inppf5_v3, arising from a different promoter,
is paternally expressed in mouse brain (19,20). The
genomic organization of the human locus resembles that
of the mouse, however, there is no evidence for a human
Inpp5f_v3 orthologue (Figure 1E and F). The promoter of
the human INPPF5_V2 transcript is embedded within a
maternally DNA-methylated DMR (Figure 1F and
Supplementary Figure S1B), resulting in paternal expres-
sion in a wide range of tissues (Supplementary Table S2).
The full-length host gene transcripts (Genbank
AB023183), and the truncated isoform (Genbank
BC052367), originate from an unmethylated CpG island
and are biallelically expressed.
Retrogenes-host pairs do not form part of larger
imprinting clusters
In order to determine the boundaries of imprinting at each
retrogene-host locus, we investigated the allele-specific ex-
pression of the genes flanking the host genes in both mice
and humans. We assessed expression in various embryonic
tissues and placenta using allele-specific assays between
crosses of mouse strains C57BL/6 (B) x Mus musculus
castaneus (C). The genes immediately adjacent to Mcts2/
H13,Id1 and Remi, are biallelically expressed in all tissues
at embryonic day E18.5, as are 111007A13RIK and Bag3
flanking Inpp5f_v2/Inpp5f. The Fam13a gene, telomeric to
Nap1l5/Herc3 is expressed from both alleles, while Abcg2,
centromeric to Nap1l5/Herc3, is monoallelically expressed
in the placenta. This reflects a bias in expression from the
C57BL/6 allele and the gene is therefore an expressed
quantitative trait locus (eQTL) and not imprinted
(Figure 2A and Supplementary Figure S2). This conclu-
sion is supported by the persistence of Abcg2 monoallelic
expression in Dnmt3l
/+
placental trophoblast, despite the
loss of imprinting of Nap1l5 (Figure 2B).
A
B
C
Figure 2. (A) A schematic map of the Abcg2 gene, with the location of the alternative promoter regions. The methylation status of the CpG island
associated with isoform 1 was examined in placenta-derived DNA. The allelic expression of Abcg2 is assessed in various fetal tissues in reciprocal
mouse crosses. (B) The allelic expression of Abcg2 and Nap1l5 in placental trophoblasts from Dnmt3l
/+
mice. (C) The allelic expression of the
ABCG2 gene in human term placenta.
Nucleic Acids Research, 2011, Vol. 39, No. 11 4581
To assess the allelic origin of expression for the
orthologous flanking genes in the human, we assessed
the expression of REMI,ID1,BAG3,CORF119 and
FAM13A/FAM13AOS. These genes are expressed
biallelically in all fetal tissues (Supplementary Table S3
and Figure S3). In higher primates, including rhesus
monkey, orangutan, chimps and humans, the organization
of the genes centromeric to NAP1L5/HERC3 is different
to that of mouse and rat, resulting in the gene PIGY
being immediately centromeric to NAP1L5/HERC3, and
ABCG2 300 kb away. In all human fetal tissues
analyzed, including placenta, both PIGY and ABCG2
are biallelic (Figure 2C and Supplementary Figure S3).
These finding strongly suggest that imprinted
retrogene-host pairs do not form part of larger imprinting
clusters.
Histone modification and allelic repression at imprinted
retrogene DMRs
In mouse the imprinted expression of Nap1l5 and
Inpp5f_v2 is restricted to the brain (17–19), whereas in
humans, imprinted expression is observed in a wider
variety of fetal tissues (Figure 1, Supplementary Table
S1 and Figure S4). To date all germline DMRs are
differentially methylated in all somatic tissues, indicating
that DNA methylation on its own is not responsible
for tissues-specific differences in expression observed at
imprinted loci. To investigate whether the discrepancy
in expression profiles we observe between species could
be attributed to histone modifications, we analyzed the
allelic enrichment of both permissive and repressive
histone modifications. Our analysis focused on modifica-
tions on histone H3 and H4, including acetylation of
lysine-9 (H3K9ac) and H3K4me2 as markers of active
chromatin; and the repressive marks of H3K9me3 and
H3K27me3 of histone H3, along with the histone H4
modifications H4K20me3 and H2A/H4R3me2s.
ChIP was performed on native chromatin from brain
and decapitated embryos for both BxC and BxJF1 crosses.
We ascertained allelic enrichment using polymorphisms
mapping within the DMRs of Nap1l5, Inpp5f_v2 and
Mcts2. The active modification H3K4me2 was strongly
enriched specifically on the unmethylated paternal allele
for all three DMRs in both brain and embryo, while
most H3K9ac precipitation was predominantly in brain
(Figure 3), the tissue in which these genes are expressed.
The same regions showed precipitation of the repressive
marks H3K9me3, H4K20me3 and H2A/H4R3me2s on
Mouse ChIP
*
Brain
Input
B6 BxJ JF1
H3K9ac H3K4me2 H3K27me3 H4K20me3
UBUBUBUBUB UB
H3K9me3 H4R3me2s
*
** *
I
npp5f_v2
DMR
Embryo
*
**
mat
pat
mat
pat
Nap1l5
DMR
**
*
*
Input
B6 BxJ JF1
H3K9ac H3K4me2 H3K27me3 H4K20me3
UBUBUBUBUB UB
H3K9me3 H4R3me2s
***
**
*
*
**
Embryo
mat
pat
Brain
mat
pat
Mcts2
DMR
**
Input
B6 BxC Cast
H3K9ac H3K4me2 H3K27me3 H4K20me3
UBUBUBUBUBUB
H3K9me3 H4R3me2s
*
*
*
*
*
** *
*
Embryo
mat
pat
Brain
mat
pat
Figure 3. (A) The allelic precipitation of the mouse retrogene DMRs in embryo and brain tissues. Native ChIP followed by PCR and restriction
digest-mediated allelic discrimination of the input, antibody bound (B) and unbound (U) chromatin fractions on BxJ embryos and brains for Nap1l5
and Inpp5f_v2, and on BxC embryos and brains for Mcts2. The asterisks represent a relative allelic enrichment of >3-fold compared to the unbound
fraction.
4582 Nucleic Acids Research, 2011, Vol. 39, No. 11
the DNA methylated maternal allele (Figure 3). The re-
pressive H3K27me3 mark showed different profiles for
the three mouse DMRs. This modification did not show
allelic enrichment at the Inpp5f_v2 DMR in either brain
or embryo, but was precipitated on the DNA methylated
maternal allele at the Mcts2 DMR. At the Nap1l5 DMR,
we observed that H3K27me3 was precipitated on the
unmethylated paternal allele in embryo but not in brain
(Figure 3). This pattern of enrichment is reminiscent of the
monoallelic bivalent chromatin domain reported at the
Grb10/GRB10 gene (21,22). This monoallelic bivalent
conformation is not detected at the Nap1l5 DMR in
brain, suggesting that the removal of H3K27me3 is con-
comitant with the paternal expression observed for Nap1l5
in mouse brain.
Extensive genotyping of the human NAP1L5,MCTS2
and INPP5F_V2 DMRs revealed that SNPs in these regu-
latory regions are rare. However, we were able to identify
heterozygous samples that allowed us to discriminate
between alleles. The SNP rs2972011 is located 200 bp
from the TSS of NAP1L5, whereas rs7907781 and
rs1115713 are 600 bp and 50 bp from the TSS of
INPP5F_V2 and MCTS2, respectively. To ensure that
these SNPs mapped within the DMRs, we performed
DNA methylation immunoprecipitation (meDIP) using
antisera directed against 5-methylcytosine. This was due
to the difficultly in amplifying bisulphite converted DNA
in the vicinity of SNPs rs7907781 and rs1115713. For all
three regions we observed monoallelic enrichment in
heterozygous placental DNA samples, and where inform-
ative, the DNA methylation was detected on the maternal
allele (Figure 4A). Using these same amplification condi-
tions, we performed ChIP on native chromatin isolated
from adult peripheral blood leukocytes and from two
human placental cell lines, TCL1 and TCL2 (23) for the
NAP1L5 and MCTS2 DMRs. Unfortunately, no hetero-
zygous cell lines could be found that were informative for
INPP5F_V2, despite genotyping of over 140 leukocyte
samples and normal tissue cell lines. Similar to the
mouse, we observe strong monoallelic enrichment for
H3K4me2 at the NAP1L5 and MCTS2 DMRs, but since
no parental DNA samples were available, allelic origin
could not be assigned. Unexpectedly, we did not observe
allelic precipitation for any of the repressive histone marks
at these DMRs, despite strong allelic enrichment at the
SNURF/SNRPN,H19 and MEST DMRs (Figure 4B;
data not shown). To confirm that the histone modifications
were present at the NAP1L5 and MCTS2 DMRs, we per-
formed quantitative ChIP analysis on the placental cell
line TCL1 (Figure 4C). The precipitation values obtained
were normalized to those for the SNURF/SNRPN DMR,
which revealed that H3K4me2 is more abundant at the
MCTS2 and NAP1L5 promoters, whereas, the repressive
histone modifications were precipitated several fold less.
Interrogation of human histone maps [http://dir.nhlbi.nih
.gov/papers/lmi/epigenomes/hgtcell.aspx, and (24)] con-
firmed the absence of significant enrichment for these
repressive marks at 1–2 nucleosomes resolution
A
BC
Figure 4. (A) Methylation-immunoprecipitation was performed on placental DNA using anti 5mC antibody. The efficiency of IP was assessed by
PCR specific for the methylated SERPIN-B5 promoter and the unmethylated UBE2B promoter. The precipitations were subsequent used to assess
the methylation at the human MCTS2,INPP5F_V2 and NAP1L5 DMRs. (B) Using the same PCR primer combinations, allelic-ChIP was performed
on human placental cell lines (for clarity, only ChIP-bound fractions are shown). (C) qPCR on ChIP-bound material from the TCL1 cell
immunoprecipitations. Levels of precipitation are compared to the SNURF DMR (red line, equal to one).
Nucleic Acids Research, 2011, Vol. 39, No. 11 4583
(the approximate size of the MCTS2 and NAP1L5 DMRs),
despite strong H3K4me2 enrichment. Together, these data
suggest that these repressive marks are not present, or very
low, in these two regions.
DISCUSSION
In this study, we have shown the paternal allele-specific
expression of INPP5F_V2,MCTS2 and NAP1L5 in a
wide range of human tissues. This is in contrast to the
mouse, where Inpp5f_v2 and Nap1l5 show spatial expres-
sion restricted to brain. We show that the TSSs for these
three genes are embedded in regions of differential DNA
methylation, similar to their mouse orthologues.
In the mouse, Mcts2 and Nap1l5, like Inpp5f_v2 are
organized such that the imprinted ‘intronic retrogene’
and its DMR/promoter reside in the intron of another
gene known as the ‘host’. In each case the retrogene
originated from an ancestoral gene on the X chromosome
some time in early eutheraian evolution, since Inppf5_v2,
Mcts2 and Nap1l5 are absent in marsupials (19).
Retrotransposition has also been linked to the imprinting
of the RB1 gene, however this event occurred much later
in mammalian evolution as only humans, chimpanzee and
rhesus monkeys, but not mice and rat have the processed
RB1/KIAA0649 pseudogene (25). A recent high-resolution
analysis of parent-of-origin allelic expression in mouse
brain has revealed that the host genes Herc3 and Inpp5f
show evidence for allele-specific alternative polyA choice
similar to that of H13 [A.J. Wood and R.J. Oakey,
unpublished data and (14,15)].
To assess whether the human orthologues MCTS2,
NAP1L5 and INPP5F_V2 are also associated with im-
printed host genes, we analyzed the allelic expression of
the host genes in a wide range of fetal tissues and term
placentas. We observe in all cases, that the host transcripts
are biallelically expressed. The genomic location of the
host gene exons are different. In the mouse, the polyA
signal for the paternally expressed H13d isoform maps
to within 100 bp of the Mcts2 DMR, whereas these two
features are separated by >7 kb in humans. Genomic re-
arrangements are more pronounced at the Nap1l5/
NAP1L5 locus, where Herc3b and Nap1l5 are separated
by 1.5 kb in the mouse, and by more that 39 kb in humans.
In addition, the mouse Herc3c isoform initiates from
within the Nap1l5 DMR, whereas the human promoter
for this isoform is adjacent to the HERC3B polyA
(Figure 1D). These differences in exon distribution could
explain the lack of host gene imprinting observed in
humans. We have previously proposed that transcription-
al interference may be involved in alternative polyA choice
at H13, due to the high expression of Mcts2 in brain
directly inhibiting the transcription of H13 on the
paternal allele in co-expressing cells. Alternative models,
including the recruitment of methylation-sensitive polyA
factors, or the association with the CTCF/cohesin
boundary complex to the unmethylated paternal allele of
the Mcts2, could result in similar allelic-termination of the
host genes on the maternal allele (14). For any of the
models, the imprinting of host gene transcripts may
require a close physical proximity of the host gene exons
with the retrogene, which is not observed in humans.
Genes flanking the retrogene-host pairs are biallelically
expressed
Many imprinted genes are clustered in the genome, such
that their expression is influenced by shared control
elements, such as DMRs. The imprinted retrogene-host
pairs are located outside of characterized clusters. To
confirm this, we assayed flanking genes for expression
status and found all to be biallelic, with the exception of
Abcg2 which is monoallelically expressed in the placentae
from reciprocal BxC and CxB sub-species intercrosses, but
not subject to imprinting.
It has recently become evident that non-imprinted
monoallelic expression can result from SNP-associated
DNA methylation (26). To assess whether the Abcg2
eQTL is due to monoallelic DNA methylation similar to
that described in humans, we showed that the promoter
CpG island of Abcg2 is fully unmethylated (Figure 2 and
data not shown) suggesting that another mechanism is
regulating the allelic expression. Recently, Brideau et al.
(27) reported that the AK006067 transcript next to the
imprinted Rasgrf1 gene is expressed >100-fold higher
from the C57BL/6 allele than the PWK allele. Both our
finding of the Abcg2 eQTL and the observations of
Brideau et al. (27), emphasize the necessity to study recip-
rocal F1 crosses.
Histone modification signatures at imprinted retrogene
DMRs
Recent studies have suggested that there is a link between
DNA and histone methylation at imprinted DMRs. A
comprehensive analysis of allelic histone modifications in
Dnmt3l
/+
conceptuses revealed that without oocyte-
derived DNA-methylation imprints, there is a dramatic
effect on the presence of repressive histone modifications,
with maternally DNA methylated DMRs adopting a
paternal epigenotype (7). We explored whether the chro-
matin at Nap1l5,Mcts2 and Inpp5f_v2 is associated with
the plethora of histone modifications known to be
enriched at DMRs. In agreement with our previous obser-
vations, we found that the DNA-methylated alleles of
each DMR are enriched for the repressive histone marks
H3K9me3, H4K20me3 and H2A/H4R3me2s. Unlike
these three modifications, H3K27me3 was not consistently
associated with the DNA-methylated allele, which agrees
with earlier chromatin studies on ICRs. At the Nap1l5
DMR, we confirm the previous observation of bivalent
chromatin in mouse embryos (7), with H3K4me2 and
H3K27me3 both enriched on the paternal allele. This
monoallelic bivalent domain behaves in a similar fashion
to the recently described monoallelic bivalent chromatin at
the Grb10 DMR (21). Like their non-imprinted bivalent
counterparts, these genes are associated with ‘poised’
lineage-specific transcription in mouse and human ES
cells (28). In brain, we observe absence of allelic enrich-
ment for H3K27me3, and this correlates with acquired
expression of Nap1l5, a mechanism comparable to
what we have reported previously at the Grb10 (21,22).
4584 Nucleic Acids Research, 2011, Vol. 39, No. 11
These observations suggest that monoallelic bivalent
chromatin could be a common mechanism conferring
brain-specific imprinted gene expression.
To confirm the conservation of these histone modifica-
tions at the regions we identified as DMRs in humans, we
performed ChIP on leukocytes and placenta cell lines. To
our surprise we did not find significant enrichment of the
repressive histone marks H3K9me3 and H4K20me3 on
the DNA-methylated allele. This uncoupled action of the
DNA-methylation machinery and the H3K9me3 and
H4K20me3 HMTs may be partially explained by the pro-
gressive decrease in CpG island size and CpG density at
the NAP1L5 and MCTS2 DMRs compared to other im-
printed DMRs in the genome (Supplementary Table S4).
Throughout mammalian evolution, the NAP1L5 and
MCTS2 DMRs have lost approximately half of their
CpG dinucleotides compared to mouse. Both DMRs
have diminished in size, with a reduction from >420 bp
to 216 bp for MCTS2. The human NAP1L5 DMR fails to
reach standard CpG island criteria (GC content >50%;
Obs CpG/Exp CpG >0.6; min length 200 bp). The loss in
CpG island size at the NAP1L5 DMR is due to a com-
bination of CpG deamination and the integration of
numerous CpG low-density repeat elements, including
DNA-MER115, LINE-1 and low-complexity CT-repeats
immediately downstream of the transcription start site.
The human and mouse genomes have recently been
shown to contain a similar number of CpG islands (29).
Our observation of an evolutionary loss of CpG density at
the NAP1L5 and MCTS2 DMRs goes against the general
trend. Additionally, for many imprinted DMRs analyzed
in humans (KvDMR1, GRB10,MEST,ZAC1,NDN,
GNAS EX1A,GNAS XL,PEG3,PEG10,NNAT and
IGF2R/AIR), the size of the CpG islands comprising the
DMRs are all larger in humans than in mouse
(Supplementary Table S4). The low-CpG density within
the promoters of NAP1L5 and MCTS2 may mean that
these discrete DMRs go unrecognized by the non-histone
proteins including the HMTs for H4K20me3 and
H3K9me3, respectively (8,12). Overall these observations
are in agreement with the recent studies suggesting that
DNA methylation at functional imprints require DNA
methylation before the acquisition of repressive histone
methylation (7), and that deficiencies in repressive
histone marks do not have a direct role in the regulation
of DNA methylation at ICRs (12,13,30).
CONCLUSIONS
In summary, we have shown that the human orthologues
of mouse imprinted retrogenes are paternally expressed in
a wide range of fetal tissues. In mice, the host genes are
subject to alternative polyadenylation, presumably as a
consequence of retrogene integrations that acquired im-
printing in proximity to weak polyA signals. In humans,
we show that retrogene promoters are subject to
allele-specific CpG methylation, but internal polyA sites
of host genes are situated further upstream of the DMRs
and thereby escape their influence. In mice, these DMRs
are associated with allelic repressive histone modifications.
At the mouse Nap1l5 promoter, monoallelic bivalent
chromatin i.e. the enrichment of both H3K4me2 and
H3K27me3 on the same allele, is associated with the
unmethylated paternal allele. In humans, an evolutionary
deterioration in CpG island size correlates with a lack
of allelic H3K9me3 and H4K20me3 precipitation at
NAP1L5 and MCTS2 DMRs indicating imprinted gene
expression in the absence of the an ICR-specific histone
signature.
SUPPLEMENTARY DATA
Supplementary Data are available at NAR Online.
ACKNOWLEDGEMENTS
We thank R. Schulz for critical reading of the article and
C. Landles for technical assistance with Southern blotting.
We thank K. Hata for Dnmt3l
/+
embryos on the BxJ
background and Dr M. Sullivan for the TCL cell lines.
FUNDING
Spanish Ministerio de Educacion y Ciencia (grant number
SAF2008-1578 to D.M.); Association Pour La Recherche
sur le Cancer (grant number 4980; 2010 to P.A.); La Ligue
contre le Cancer (grant ‘subvention comite’ Herault: 2010
to P.A.); CNRS ‘Projects for International Scientific
Cooperation’ (to P.A. and D.M.); Agence National de la
Recherche; ‘Institut National du Cancer’; Association for
International Cancer Research; ‘Ligue Contre le Cancer’
(to R.F.); Medical Research Council (to G.E.M.);
SPARKs; Wellbeing for Women (to G.E.M.); The
Wellcome Trust (to R.J.O., J.M.F. and M.C.); Ramon y
Cajal research fellowship (to D.M.); FPU studentship (to
A.G.A.). Funding for open access charge: Internal
funding.
Conflict of interest statement. None declared.
REFERENCES
1. Verona,R.I., Mann,M.R. and Bartolomei,M.S. (2003) Genomic
imprinting: intricacies of epigenetic regulation in clusters.
Ann. Rev. Cell Dev. Biol.,19, 237–259.
2. Bourc’his,D., Xu,G.L., Lin,C.S., Bollman,B. and Bestor,T.H.
(2003) Dnmt3L and the establishment of maternal genomic
imprints. Science,21, 2536–2539.
3. Kaneda,M., Okano,M., Hata,K., Sado,T., Tsujimoto,N., Li,E.
and Sasaki,H. (2004) Essential role for de novo DNA
methyltransferase Dnmt3a in paternal and maternal imprinting.
Nature,24, 900–903.
4. Kato,Y., Kaneda,M., Hata,K., Kumaki,K., Hisano,M.,
Kohara,Y., Okano,M., Li,E., Nozaki,M. and Sasaki,H. (2007)
Role of the Dnmt3 family in de novo methylation of imprinted
and repetitive sequences during male germ cell development in the
mouse. Hum. Mol. Genet.,16, 2272–2280.
5. Ciccone,D.N., Su,H., Hevi,S., Gay,F., Lei,H., Bajko,J., Xu,G.,
Li,E. and Chen,T. (2009) KDM1B is a histone H3K4
demethylase required to establish maternal genomic imprints.
Nature,461, 415–418.
6. Cirio,M.C., Ratnam,S., Ding,F., Reinhart,B., Navara,C. and
Chaillet,J.R. (2008) Preimplantation expression of the somatic
Nucleic Acids Research, 2011, Vol. 39, No. 11 4585
form of Dnmt1 suggests a role in the inheritance of genomic
imprints. BMC Dev. Biol.,8,9.
7. Henckel,A., Nakabayashi,K., Sanz,L.A., Feil,R., Hata,K. and
Arnaud,P. (2009) Histone methylation is mechanistically linked to
DNA methylation at imprinting control regions in mammals.
Hum. Mol. Genet.,18, 3375–3383.
8. Pannetier,M., Julien,E., Schotta,G., Tardat,M., Sardet,C.,
Jenuwein,T. and Feil,R. (2008) PR-SET7 and SUV4-20H regulate
H4 lysine-20 methylation at imprinting control regions in the
mouse. EMBO Reports,9, 998–1005.
9. Fournier,C., Goto,Y., Ballestar,E., Delaval,K., Hever,A.M.,
Esteller,M. and Feil,R. (2002) Allele-specific histone lysine
methylation marks regulatory regions at imprinted mouse genes.
EMBO J.,21, 6560–6570.
10. Umlauf,D., Goto,Y., Cao,R., Cerqueira,F., Wagschal,A.,
Zhang,Y. and Feil,R. (2004) Imprinting along the Kcnq1 domain
on mouse chromosome 7 involves repressive histone methylation
and recruitment of Polycomb group complexes. Nat. Genet.,36,
1296–1300.
11. Monk,D., Arnaud,P., Apostolidou,S., Hills,F.A., Kelsey,G.,
Stanier,P., Feil,R. and Moore,G.E. (2006) Limited evolutionary
conservation of imprinting in the human placenta. Proc. Natl
Acad. Sci. USA,103, 6623–6628.
12. Wagschal,A., Sutherland,H.G., Woodfine,K., Henckel,A.,
Chebli,K., Schulz,R., Oakey,R.J., Bickmore,W.A. and Feil,R.
(2007) G9a histone methyltransferase contributes to imprinting in
the mouse placenta. Mol. Cell. Biol.,28, 1104–1113.
13. Monk,D., Wagschal,A., Arnaud,P., Mu
¨ller,P.S., Parker-
Katiraee,L., Bourc’his,D., Scherer,S.W., Feil,R., Stanier,P. and
Moore,G.E. (2008) Comparative analysis of human chromosome
7q21 and mouse proximal chromosome 6 reveals a
placental-specific imprinted gene, TFPI2/Tfpi2, which requires
EHMT2 and EED for allelic-silencing. Genome Res.,18,
1270–1281.
14. Wood,A.J., Schulz,R., Woodfine,K., Koltowska,K., Beechey,C.V.,
Peters,J., Bourc’his,D. and Oakey,R.J. (2008) Regulation of
alternative polyadenylation by genomic imprinting. Genes Dev.,
22, 1141–1146.
15. Gregg,C., Zhang,J., Weissbourd,B., Luo,S., Schroth,G.P., Haig,D.
and Dulac,C. (2010) High-resolution analysis of parent-of-origin
allelic expression in the mouse brain. Science,329, 643–648.
16. Apostolidou,S., Abu-Amero,S., O’Donoghue,K., Frost,J.,
Olafsdottir,O., Chavele,K.M., Whittaker,J.C., Loughna,P.,
Stanier,P. and Moore,G.E. (2007) Elevated placental expression of
the imprinted PHLDA2 gene is associated with low birth weight.
J. Mol. Med.,85, 379–387.
17. Smith,R.J., Dean,W., Konfortova,G. and Kelsey,G. (2003)
Identification of novel imprinted genes in a genome-wide screen
for maternal methylation. Genome Res.,13, 558–569.
18. Choi,J.D., Underkoffler,L.A., Wood,A.J., Collins,J.N.,
Williams,P.T., Golden,J.A., Schuster,E.F. Jr, Loomes,K.M. and
Oakey,R.J. (2005) A novel variant of Inpp5f is imprinted in
brain, and its expression is correlated with differential
methylation of an internal CpG island. Mol. Cell. Biol.,25,
5514–5522.
19. Wood,A.J., Roberts,R.G., Monk,D., Moore,G.E., Schulz,R. and
Oakey,R.J. (2007) A screen for retrotransposed imprinted
genes reveals an association between X chromosome
homology and maternal germ-line methylation. PLoS Genet.,
3, e20.
20. Wood,A.J., Bourc’his,D., Bestor,T.H. and Oakey,R.J. (2007)
Allele-specific demethylation at an imprinted mammalian
promoter. Nucleic Acids Res.,35, 7031–7039.
21. Sanz,L.A., Chamberlain,S., Sabourin,J.C., Henckel,A.,
Magnuson,T., Hugnot,J.P., Feil,R. and Arnaud,P. (2008) A
mono-allelic bivalent chromatin domain controls tissue-specific
imprinting at Grb10. EMBO J.,227, 2523–2532.
22. Monk,D., Arnaud,P., Frost,J., Hills,F.A., Stanier,P., Feil,R. and
Moore,G.E. (2009) Reciprocal imprinting of human
GRB10 in placental trophoblast and brain: evolutionary
conservation of reversed allelic expression. Hum. Mol. Genet.,18,
3066–3074.
23. Takeda,S. and Nakano,H. (2003) TNFalpha-induced apoptosis
and integrin switching in human extravillous trophoblast cell line.
Biol. Reprod.,68, 1771–1778.
24. Barski,A., Cuddapah,S., Cui,K., Roh,T.Y., Schones,D.E.,
Wang,Z., Wei,G., Chepelev,I. and Zhao,K. (2007) High-resolution
profiling of histone methylations in the human genome. Cell,129,
823–837.
25. Kanber,D., Berulava,T., Ammerpohl,O., Mitter,D., Richter,J.,
Siebert,R., Horsthemke,B., Lohman,D. and Buiting,K. (2009) The
human retinoblastoma gene is imprinted. PLoS Genet.,12,
e1000790.
26. Kerkel,K., Spadola,A., Yuan,E., Kosek,J., Jiang,L., Hod,E.,
Li,K., Murty,V.V., Schupf,N., Vilain,E. et al. (2008) Genomic
surveys by methylation-sensitive SNP analysis identify
sequence-dependent allele-specific DNA methylation. Nat. Genet.,
40, 904–908.
27. Brideau,C.M., Eilertson,K.E., Hagarman,J.A., Bustamante,C.D.
and Soloway,P.D. (2010) Successful computational prediction of
novel imprinted genes from epigenomic features. Mol. Cell. Biol.,
30, 3357–3370.
28. Bernstein,B.E., Mikkelsen,T.S., Xie,X., Kamal,M., Huebert,D.J.,
Cuff,J., Fry,B., Meissner,A., Wernig,M., Plath,K. et al. (2006) A
bivalent chromatin structure marks key developmental genes in
embryonic stem cells. Cell,125, 315–326.
29. Illingworth,R.S., Gruenewald-Schneider,U., Webb,S., Kerr,A.R.,
James,K.D., Turner,D.J., Smith,C., Harrison,D.J., Andrews,R.
and Bird,A. (2010) Orphan CpG islands identify numerous
conserved promoters in the mammalian genome. PLoS Genet.,6,
e1001134.
30. Mager,J., Montgomery,N.D., de Villena,F.P. and Magnuson,T.
(2003) Genome imprinting regulated by the mouse Polycomb
group protein Eed. Nat. Genet.,33, 502–507.
4586 Nucleic Acids Research, 2011, Vol. 39, No. 11
... Interestingly, Miranda et al. [145] found methylation of this gene to be a part of a gene signature predictive of gemcitabine response in the GDSC dataset, whereas in our gene region-focused analysis we did not find any associations of methylation of any regions of HM13 with drug response (Additional file 9: Table S6). The genome location of HM13 overlaps with that of the imprinted pseudogene PCIMST-1 [26,146]. Interestingly, in breast cancer, most prominently in the luminal B subtype, overexpression of HM13 was found to be caused by its biallelic expression due to the loss of imprinting, and it was independent of the copy number gain [26]. ...
Article
Full-text available
Background Parent of origin-specific allelic expression of imprinted genes is epigenetically controlled. In cancer, imprinted genes undergo both genomic and epigenomic alterations, including frequent copy number changes. We investigated whether copy number loss or gain of imprinted genes in cancer cell lines is associated with response to chemotherapy treatment. Results We analyzed 198 human imprinted genes including protein-coding genes and noncoding RNA genes using data from tumor cell lines from the Cancer Cell Line Encyclopedia and Genomics of Drug Sensitivity in Cancer datasets. We examined whether copy number of the imprinted genes in 35 different genome locations was associated with response to cancer drug treatment. We also analyzed associations of pretreatment expression and DNA methylation of imprinted genes with drug response. Higher copy number of BLCAP, GNAS, NNAT, GNAS-AS1, HM13, MIR296, MIR298, and PSIMCT-1 in the chromosomal region 20q11-q13.32 was associated with resistance to multiple antitumor agents. Increased expression of BLCAP and HM13 was also associated with drug resistance, whereas higher methylation of gene regions of BLCAP, NNAT, SGK2, and GNAS was associated with drug sensitivity. While expression and methylation of imprinted genes in several other chromosomal regions was also associated with drug response and many imprinted genes in different chromosomal locations showed a considerable copy number variation, only imprinted genes at 20q11-q13.32 had a consistent association of their copy number with drug response. Copy number values among the imprinted genes in the 20q11-q13.32 region were strongly correlated. They were also correlated with the copy number of cancer-related non-imprinted genes MYBL2, AURKA, and ZNF217 in that chromosomal region. Expression of genes at 20q11-q13.32 was associated with ex vivo drug response in primary tumor samples from the Beat AML 1.0 acute myeloid leukemia patient cohort. Association of the increased copy number of the 20q11-q13.32 region with drug resistance may be complex and could involve multiple genes. Conclusions Copy number of imprinted and non-imprinted genes in the chromosomal region 20q11-q13.32 was associated with cancer drug resistance. The genes in this chromosomal region may have a modulating effect on tumor response to chemotherapy.
... Thus, the intragenic island regulates imprinted Inpp5f_ v2 expression [100]. Similarly, the promotor of human INPPF5_V2 transcript is embedded within a maternally methylated DMR [101]. ...
Article
Full-text available
Background In mammals, Imprinting Control Regions (ICRs) regulate a subset of genes in a parent-of-origin-specific manner. In both human and mouse, previous studies identified a set of CpG-rich motifs occurring as clusters in ICRs and germline Differentially Methylated Regions (gDMRs). These motifs consist of the ZFP57 binding site (ZFBS) overlapping a subset of MLL binding units known as MLL morphemes. MLL or MLL1 (Mixed Lineage Leukemia 1) is a relatively large multidomain protein that plays a central role in the regulation of transcription. The structures of both MLL1 and MLL2 include a domain (MT) that binds CpG-rich DNA and a conserved domain (SET) that methylates lysine 4 in histone H3 producing H3K4me3 marks in chromatin. Results Since genomic imprinting impacts many developmental and key physiological processes, we followed a previous bioinformatics strategy to pinpoint ICR positions in the Bos taurus genome. Initial genome-wide analyses involved finding the positions of ZFP57 binding sites, and the CpG-rich motifs (ZFBS-morph overlaps) along cattle chromosomal DNA. By creating plots displaying the density of ZFBS-morph overlaps, we removed background noise and thus improved signal detection. With the density-plots, we could view the positions of peaks locating known and candidate ICRs in cattle DNA. Our evaluations revealed the correspondence of peaks in plots to reported known and inferred ICRs/DMRs in cattle. Beside peaks pinpointing such ICRs, the density-plots also revealed additional peaks. Since evaluations validated the robustness of our approach, we inferred that the additional peaks may correspond to candidate ICRs for imprinted gene expression. Conclusion Our bioinformatics strategy offers the first genome-wide approach for systematically localizing candidate ICRs. Furthermore, we have tailored our datasets for upload onto the UCSC genome browser so that researchers could find known and candidate ICRs with respect to a wide variety of annotations at all scales: from the positions of Single Nucleotide Polymorphisms (SNPs), to positions of genes, transcripts, and repeated DNA elements. Furthermore, the UCSC genome browser offers tools to produce enlarged views: to uncover the genes in the vicinity of candidate ICRs and thus discover potential imprinted genes for experimental validations.
... H4K20me3 is typically a mark of silenced heterochromatin, which is also characterized by other histone modifications, such as H3K9me3 and H3K64me3, and is mainly found near chromosome centromeres and telomeres, where it ensures the correct segregation and integrity of the genome [10,11]. H4K20me3 and H3K9me3 have also been found to control imprinted gene expression by localizing to silenced imprinted gene promoters and non-expressed pseudogenes [12,13]. SUV4-20H1 and SUV4-20H2 are highly homologous and partially redundant, and have been investigated While PHF8 (KDM7B) and LSD1 (KDM1A) have been reported as H4K20me1 demethylases in mammals [16,17], two DNA repair proteins, hHR23A/B, have recently been demonstrated to be demethylases for H4K20me1/2/3 both in vivo and in vitro [18]. ...
Article
Full-text available
The post-translational modification of histone tails is a dynamic process that provides chromatin with high plasticity. Histone modifications occur through the recruitment of nonhistone proteins to chromatin and have the potential to influence fundamental biological processes. Many recent studies have been directed at understanding the role of methylated lysine 20 of histone H4 (H4K20) in physiological and pathological processes. In this review, we will focus on the function and regulation of the histone methyltransferases SUV4-20H1 and SUV4-20H2, which catalyze the di- and tri-methylation of H4K20 at H4K20me2 and H4K20me3, respectively. We will highlight recent studies that have elucidated the functions of these enzymes in various biological processes, including DNA repair, cell cycle regulation, and DNA replication. We will also provide an overview of the pathological conditions associated with H4K20me2/3 misregulation as a result of mutations or the aberrant expression of SUV4-20H1 or SUV4-20H2. Finally, we will critically analyze the data supporting these functions and outline questions for future research.
... Thus, the intragenic island regulates imprinted Inpp5f_v2 expression [82]. Similarly, the promotor of human INPPF5_V2 transcript is embedded within a maternally methylated DMR [83]. ...
Preprint
In mammals, Imprinting Control Regions (ICRs) regulate a subset of genes in a parent-of-origin-specific manner. In both human and mouse, previous studies identified a set of CpG-rich motifs that occurred as clusters in ICRs and germline Differentially Methylated Regions (gDMRs). These motifs consist of the ZFP57 binding site (ZFBS) overlapping a subset of MLL binding units known as MLL morphemes. Furthermore, by creating plots for displaying the density of these overlaps, it became possible to locate known and candidate ICRs in mouse and human genomic DNA. Since genomic imprinting impacts many developmental and key physiological processes, we performed genome-wide analyses to create plots displaying the density of the CpG-rich motifs (ZFBS-morph overlaps) along Bos Taurus chromosomal DNA. We tailored our datasets so that they could be displayed on the UCSC genome browser (the build bosTau8). On the genome browser, we could view the ZFP57 binding sites, the ZFBS-morph overlaps, and peaks in the density-plots in the context of cattle RefSeq Genes, Non-Cow RefSeq Genes, CpG islands, and Single nucleotide polymorphisms (SNPs). Our datasets revealed the correspondence of peaks in plots to known and deduced ICRs in Bos Taurus genomic DNA. We illustrate that by uploading our datasets onto the UCSC genome browser, we could discover candidate ICRs in cattle DNA. In enlarged views, we could pinpoint the genes in the vicinity of candidate ICRs and thus discover potential imprinted genes.
... Utilization of the intronic H13 PAS on the paternal allele results in expression of a truncated and likely non-functional H13 protein (Wood et al., 2008). A similar mode of regulation was reported for the imprinted retrogene Nap1l5 in mouse brain, which is expressed from the paternally inherited allele (Monk et al., 2011;Cowley et al., 2012). Nap1l5 is located within an intron of the Herc3 gene and downstream of two intronic Herc3 PAS. ...
Article
Full-text available
The majority of eukaryotic genes produce multiple mRNA isoforms by using alternative poly(A) sites in a process called alternative polyadenylation (APA). APA is a dynamic process that is highly regulated in development and in response to extrinsic or intrinsic stimuli. Mis-regulation of APA has been linked to a wide variety of diseases, including cancer, neurological and immunological disorders. Since the first example of APA was described 40 years ago, the regulatory mechanisms of APA have been actively investigated. Conventionally, research in this area has focused primarily on the roles of regulatory cis-elements and trans-acting RNA-binding proteins. Recent studies, however, have revealed important functions for epigenetic mechanisms, including DNA and histone modifications and higher-order chromatin structures, in APA regulation. Here we will discuss these recent findings and their implications for our understanding of the crosstalk between epigenetics and mRNA 3'-end processing.
... Consistent with expectation, of all genes containing an ASE, 170 were identified on the X chromosome (equating to 1.96% unidirectional within ≥ 10 individuals) were located within genes known to be imprinted (as reported in www.geneimprint.com or within the literature [27][28][29][30] ) compared with 1-5% of consistent signals (Fig. 6b). ...
Article
Full-text available
Genome-wide association studies have generated an increasing number of common genetic variants associated with neurological and psychiatric disease risk. An improved understanding of the genetic control of gene expression in human brain is vital considering this is the likely modus operandum for many causal variants. However, human brain sampling complexities limit the explanatory power of brain-related expression quantitative trait loci (eQTL) and allele-specific expression (ASE) signals. We address this, using paired genomic and transcriptomic data from putamen and substantia nigra from 117 human brains, interrogating regulation at different RNA processing stages and uncovering novel transcripts. We identify disease-relevant regulatory loci, find that splicing eQTLs are enriched for regulatory information of neuron-specific genes, that ASEs provide cell-specific regulatory information with evidence for cellular specificity, and that incomplete annotation of the brain transcriptome limits interpretation of risk loci for neuropsychiatric disease. This resource of regulatory data is accessible through our web server, http://braineacv2.inf.um.es/.
... Note that annotation of the SNPs to specific genes was often difficult as many overlapping genes were found (Supplementary Note 1). For example, MCTS2P is a retrogene copy and located in HM13, making it uncertain in which gene the detected SNP was located 31 . Similarly, for MTRNR2L1, imprinted SNPs with Ensembl annotation for this locus were found upstream of the gene, making correct annotation uncertain. ...
Article
Full-text available
Genomic imprinting plays an important role in growth and development. Loss of imprinting (LOI) has been found in cancer, yet systematic studies are impeded by data-analytical challenges. We developed a methodology to detect monoallelically expressed loci without requiring genotyping data, and applied it on The Cancer Genome Atlas (TCGA, discovery) and Genotype-Tissue expression project (GTEx, validation) breast tissue RNA-seq data. Here, we report the identification of 30 putatively imprinted genes in breast. In breast cancer (TCGA), HM13 is featured by LOI and expression upregulation, which is linked to DNA demethylation. Other imprinted genes typically demonstrate lower expression in cancer, often associated with copy number variation and aberrant DNA methylation. Downregulation in cancer frequently leads to higher relative expression of the (imperfectly) silenced allele, yet this is not considered canonical LOI given the lack of (absolute) re-expression. In summary, our novel methodology highlights the massive deregulation of imprinting in breast cancer.
... Therefore, they may become targets of epigenetic silencing by repressive chromatin. The association of retrogenes with specific chromatin states has been proposed (Boutanaev et al., 2002;Marques et al., 2005), but only a few have been characterized as to their chromatin states so far (Monk et al., 2011;Pei et al., 2012). ...
Article
Full-text available
Duplications allow for gene functional diversification and accelerate genome evolution. Occasionally, the transposon amplification machinery reverse transcribes the mRNA of a gene, integrates it into the genome, and forms an RNA-duplicated copy: the retrogene. Although retrogenes have been found in plants, their biology and evolution are poorly understood. Here, we identified 251 (216 novel) retrogenes in Arabidopsis thaliana, corresponding to 1% of protein-coding genes. Arabidopsis retrogenes are derived from ubiquitously transcribed parents and reside in gene-rich chromosomal regions. Approximately 25% of retrogenes are cotranscribed with their parents and 3% with head-to-head oriented neighbors. This suggests transcription by novel promoters for 72% of Arabidopsis retrogenes. Many retrogenes reach their transcription maximum in pollen, the tissue analogous to animal spermatocytes, where upregulation of retrogenes has been found previously. This implies an evolutionarily conserved mechanism leading to this transcription pattern of RNA-duplicated genes. During transcriptional repression, retrogenes are depleted of permissive chromatin marks without an obvious enrichment for repressive modifications. However, this pattern is common to many other pollen-transcribed genes independent of their evolutionary origin. Hence, retroposition plays a role in plant genome evolution, and the developmental transcription pattern of retrogenes suggests an analogous regulation of RNA-duplicated genes in plants and animals.
... For three genes (KCNQ1, INPP5F and ZNF331), the monoallelic expression was confined to specific isoforms (Fig. 3). For INPP5F, we found a pattern similar to findings in mice and in human fetus (43,44), where only the short isoforms of INPP5F are imprinted. Another gene, ZNF331, which was recently reported to be imprinted (45,46) showed a more complex pattern. ...
Article
Full-text available
Monoallelic expression, including genomic imprinting, X-chromosome inactivation and random monoallelic expression of autosomal genes are epigenetic phenomena. Genes that are expressed in a monoallelic way may be more vulnerable to genetic or epigenetic mutations. Thus, comprehensive exploration of monoallelic expression in human brains may shed light on complex brain disorders. Autism-related disorders are known to be associated with imprinted genes on chromosome 15. However, it is not clear if other imprinted regions or other types of monoallelic expression are associated with autism spectrum disorder (ASD). Here, we performed a genome-wide survey of allele expression imbalance (AEI) in the human brain using single nucleotide polymorphisms (SNPs), in 18 individuals with ASD and 15 controls. Individuals with ASD had the most extreme number of monoallelic expressed SNPs in both the autosomes and the X chromosome. In two cases that were studied in detail the monoallelic expression was confined to specific brain region or cell type. Using these data we were also able to define the allelic expression status of known imprinted genes in the human brain, and to identify abnormal imprinting in an individual with ASD. Lastly, we developed an analysis of individual level expression, focusing on the difference of each individual from the mean. We found that individuals with ASD had more genes that were up or down-regulated in an individual-specific manner. We also identified pathways perturbed in specific individuals. These results underline the heterogeneity in gene regulation in ASD, at the level of both allelic and total expression.
Article
Full-text available
The whole set of human imprinted genes, termed imprintome, is here analysed by means of a reasonable, valid application of the Semantic Web and Linked Data approaches to a few structured datasets in order to provide a comprehensive collection of imprinted genes in the human genome. Thus, we have stored, organised, filtered, and analysed massive amounts of existing data on human imprinted genes towards compiling, structuring and linking data to comprise a sharing resource for genome and epigenome interrogated studies. Our datasets of linked data are the actual research outcome of this human imprintome analysis because as genomics become more and more data intensive, due to huge amounts of biological data, so does our needs for more structured data to be easier mined and shared. We present the resulting first version of the Linked Human Imprintome as a project within Linked Open Data (LOD) initiative (http://lod-cloud.net/) through Data Hub (http:// thedatahub.org/en/dataset/a-draft-version-of-the-linked-human-imprintome).
Article
Full-text available
Complementary sets of genes are epigenetically silenced in male and female gametes in a process termed genomic imprinting. TheDnmt3L gene is expressed during gametogenesis at stages where genomic imprints are established. Targeted disruption ofDnmt3L caused azoospermia in homozygous males, and heterozygous progeny of homozygous females died before midgestation. Bisulfite genomic sequencing of DNA from oocytes and embryos showed that removal of Dnmt3L prevented methylation of sequences that are normally maternally methylated. The defect was specific to imprinted regions, and global genome methylation levels were not affected. Lack of maternal methylation imprints in heterozygous embryos derived from homozygous mutant oocytes caused biallelic expression of genes that are normally expressed only from the allele of paternal origin. The key catalytic motifs characteristic of DNA cytosine methyltransferases have been lost from Dnmt3L, and the protein is more likely to act as a regulator of imprint establishment than as a DNA methyltransferase.
Article
Full-text available
Genomic imprinting is a developmental mechanism that mediates parent-of-origin-specific expression in a subset of genes. How the tissue specificity of imprinted gene expression is controlled remains poorly understood. As a model to address this question, we studied Grb10, a gene that displays brain-specific expression from the paternal chromosome. Here, we show in the mouse that the paternal promoter region is marked by allelic bivalent chromatin enriched in both H3K4me2 and H3K27me3, from early embryonic stages onwards. This is maintained in all somatic tissues, but brain. The bivalent domain is resolved upon neural commitment, during the developmental window in which paternal expression is activated. Our data indicate that bivalent chromatin, in combination with neuronal factors, controls the paternal expression of Grb10 in brain. This finding highlights a novel mechanism to control tissue-specific imprinting.
Article
Full-text available
CpG islands (CGIs) are vertebrate genomic landmarks that encompass the promoters of most genes and often lack DNA methylation. Querying their apparent importance, the number of CGIs is reported to vary widely in different species and many do not co-localise with annotated promoters. We set out to quantify the number of CGIs in mouse and human genomes using CXXC Affinity Purification plus deep sequencing (CAP-seq). We also asked whether CGIs not associated with annotated transcripts share properties with those at known promoters. We found that, contrary to previous estimates, CGI abundance in humans and mice is very similar and many are at conserved locations relative to genes. In each species CpG density correlates positively with the degree of H3K4 trimethylation, supporting the hypothesis that these two properties are mechanistically interdependent. Approximately half of mammalian CGIs (>10,000) are "orphans" that are not associated with annotated promoters. Many orphan CGIs show evidence of transcriptional initiation and dynamic expression during development. Unlike CGIs at known promoters, orphan CGIs are frequently subject to DNA methylation during development, and this is accompanied by loss of their active promoter features. In colorectal tumors, however, orphan CGIs are not preferentially methylated, suggesting that cancer does not recapitulate a developmental program. Human and mouse genomes have similar numbers of CGIs, over half of which are remote from known promoters. Orphan CGIs nevertheless have the characteristics of functional promoters, though they are much more likely than promoter CGIs to become methylated during development and hence lose these properties. The data indicate that orphan CGIs correspond to previously undetected promoters whose transcriptional activity may play a functional role during development.
Article
Full-text available
Genomic imprinting results in preferential expression of the paternal or maternal allele of certain genes. We have performed a genome-wide characterization of imprinting in the mouse embryonic and adult brain. This approach uncovered parent-of-origin allelic effects of more than 1300 loci. We identified parental bias in the expression of individual genes and of specific transcript isoforms, with differences between brain regions. Many imprinted genes are expressed in neural systems associated with feeding and motivated behaviors, and parental biases preferentially target genetic pathways governing metabolism and cell adhesion. We observed a preferential maternal contribution to gene expression in the developing brain and a major paternal contribution in the adult brain. Thus, parental expression bias emerges as a major mode of epigenetic regulation in the brain.
Article
Full-text available
Approximately 100 mouse genes undergo genomic imprinting, whereby one of the two parental alleles is epigenetically silenced. Imprinted genes influence processes including development, X chromosome inactivation, obesity, schizophrenia, and diabetes, motivating the identification of all imprinted loci. Local sequence features have been used to predict candidate imprinted genes, but rigorous testing using reciprocal crosses validated only three, one of which resided in previously identified imprinting clusters. Here we show that specific epigenetic features in mouse cells correlate with imprinting status in mice, and we identify hundreds of additional genes predicted to be imprinted in the mouse. We used a multitiered approach to validate imprinted expression, including use of a custom single nucleotide polymorphism array and traditional molecular methods. Of 65 candidates subjected to molecular assays for allele-specific expression, we found 10 novel imprinted genes that were maternally expressed in the placenta.
Article
Full-text available
Genomic imprinting is an epigenetic process leading to parent-of-origin-specific DNA methylation and gene expression. To date, approximately 60 imprinted human genes are known. Based on genome-wide methylation analysis of a patient with multiple imprinting defects, we have identified a differentially methylated CpG island in intron 2 of the retinoblastoma (RB1) gene on chromosome 13. The CpG island is part of a 5'-truncated, processed pseudogene derived from the KIAA0649 gene on chromosome 9 and corresponds to two small CpG islands in the open reading frame of the ancestral gene. It is methylated on the maternal chromosome 13 and acts as a weak promoter for an alternative RB1 transcript on the paternal chromosome 13. In four other KIAA0649 pseudogene copies, which are located on chromosome 22, the two CpG islands have deteriorated and the CpG dinucleotides are fully methylated. By analysing allelic RB1 transcript levels in blood cells, as well as in hypermethylated and 5-aza-2'-deoxycytidine-treated lymphoblastoid cells, we have found that differential methylation of the CpG island skews RB1 gene expression in favor of the maternal allele. Thus, RB1 is imprinted in the same direction as CDKN1C, which operates upstream of RB1. The imprinting of two components of the same pathway indicates that there has been strong evolutionary selection for maternal inhibition of cell proliferation.
Article
Full-text available
Differential DNA methylation of the paternal and maternal alleles regulates the parental origin-specific expression of imprinted genes in mammals. The methylation imprints are established in male and female germ cells during gametogenesis, and the de novo DNA methyltransferase DNMT3A and its cofactor DNMT3L are required in this process. However, the mechanisms underlying locus- and parental-specific targeting of the de novo DNA methylation machinery in germline imprinting are poorly understood. Here we show that amine oxidase (flavin-containing) domain 1 (AOF1), a protein related to the lysine demethylase KDM1 (also known as LSD1), functions as a histone H3 lysine 4 (H3K4) demethylase and is required for de novo DNA methylation of some imprinted genes in oocytes. AOF1, now renamed lysine demethylase 1B (KDM1B) following a new nomenclature, is highly expressed in growing oocytes where genomic imprints are established. Targeted disruption of the gene encoding KDM1B had no effect on mouse development and oogenesis. However, oocytes from KDM1B-deficient females showed a substantial increase in H3K4 methylation and failed to set up the DNA methylation marks at four out of seven imprinted genes examined. Embryos derived from these oocytes showed biallelic expression or biallelic suppression of the affected genes and died before mid-gestation. Our results suggest that demethylation of H3K4 is critical for establishing the DNA methylation imprints during oogenesis.
Article
Full-text available
Mono-allelic expression of imprinted genes from either the paternal or the maternal allele is mediated by imprinting control regions (ICRs), which are epigenetically marked in an allele-specific fashion. Although, in somatic cells, these epigenetic marks comprise both DNA methylation and histone methylation, the relationship between these two modifications in imprint acquisition and maintenance remains unclear. To address this important question, we analyzed histone modifications at ICRs in mid-gestation embryos that were obtained from Dnmt3L(-/-) females, in which DNA methylation imprints at ICRs are not established during oogenesis. The absence of maternal DNA methylation imprints in these conceptuses led to a marked decrease and loss of allele-specificity of the repressive H3K9me3, H4K20me3 and H2A/H4R3me2 histone modifications, providing the first evidence of a mechanistic link between DNA and histone methylation at ICRs. The existence of this relationship was strengthened by the observation that when DNA methylation was still present at the Snrpn and Peg3 ICRs in some of the progeny of Dnmt3L(-/-) females, these ICRs were associated with the usual patterns of histone methylation. Combined, our data establish that DNA methylation is involved in the acquisition and/or maintenance of histone methylation at ICRs.
Article
In different eukaryotic model systems, chromatin and gene expression are modulated by post-translational modification of histone tails. In this in vivo study, histone methylation and acetylation are investigated along the imprinted mouse genes Snrpn, Igf2r and U2af1-rs1. These imprinted genes all have a CpG-rich regulatory element at which methylation is present on the maternal allele, and originates from the female germ line. At these ‘differentially methylated regions’ (DMRs), histone H3 on the paternal allele has lysine-4 methylation and is acetylated. On the maternally inherited allele, in contrast, chromatin is marked by hypermethylation on lysine-9 of H3. Allele-specific patterns of lysine-4 and lysine-9 methylation are also detected at other regions of the imprinted loci. For the DMR at the U2af1-rs1 gene, we establish that the methyl-CpG-binding-domain (MBD) proteins MeCP2, MBD1 and MBD3 are associated with the maternal allele. These data support the hypothesis that MBD protein-associated histone deacetylase/chromatin-remodelling complexes are recruited to the parental allele that has methylated DNA and H3-K9 methylation, and are prevented from binding to the opposite allele by H3 lysine-4 methylation.
Article
An intriguing characteristic of imprinted genes is that they often cluster in large chromosomal domains, raising the possibility that gene-specific and domain-specific mechanisms regulate imprinting. Several common features emerged from comparative analysis of four imprinted domains in mice and humans: (a) Certain genes appear to be imprinted by secondary events, possibly indicating a lack of gene-specific imprinting marks; (b) some genes appear to resist silencing, predicting the presence of cis-elements that oppose domain-specific imprinting control; (c) the nature of the imprinting mark remains incompletely understood. In addition, common silencing mechanisms are employed by the various imprinting domains, including silencer elements that nucleate and propagate a silent chromatin state, insulator elements that prevent promoter-enhancer interactions when hypomethylated on one parental allele, and antisense RNAs that function in silencing the overlapping sense gene and more distantly located genes. These commonalities are reminiscent of the behavior of genes subjected to, and the mechanisms employed in, dosage compensation.