Está en la página 1de 9

Basic concepts of epigenetics

Michal Inbar-Feigenberg, M.D.,a,b Sanaa Choufani, Ph.D.,a Darci T. Butcher, Ph.D.,a Maian Roifman, M.D.,b
and Rosanna Weksberg, M.D., Ph.D.a,b,c
a
Genetics and Genome Biology and b Division of Clinical and Metabolic Genetics, The Hospital for Sick Children; and
c
Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada

Several types of epigenetic marks facilitate the complex patterning required for normal human development. These epigenetic marks
include DNA methylation at CpG dinucleotides, covalent modications of histone proteins, and noncoding RNAs (ncRNAs). They
function in a highly orchestrated manner, regulating mitotically heritable differences in gene expression potential without altering
the primary DNA sequence. In germ cells and the developing embryo, genome-wide epigenetic reprogramming drives the erasure
and reestablishment of correct epigenetic patterns at critical developmental time periods and in specic cell types. Two specic types
of epigenetic regulation established in early development include X-chromosome inactivation and genomic imprinting; they regulate
gene expression in a dosage-dependent and parent-of-origin-specic manner, respectively. Both genetic and environmental factors
impact epigenetic marks, generating phenotypic variation that ranges from normal variation to human disease. Aberrant epigenetic
patterning can lead to a variety of human disorders, including subfertility and imprinting
disorders. (Fertil Steril 2013;99:60715. 2013 by American Society for Reproductive
Medicine.) Use your smartphone
Key Words: Epigenetics, DNA methylation, histone modication, microRNA, epigenetic to scan this QR code
reprogramming, X inactivation, genomic imprinting and connect to the
discussion forum for
this article now.*
Discuss: You can discuss this article with its authors and with other ASRM members at http://
fertstertforum.com/inbar-feigenbergm-basic-concepts-of-epigenetics/ * Download a free QR code scanner by searching for QR
scanner in your smartphones app store or app marketplace.

A
ll cells in the human body carry developing embryo and fetus. Several females. Genomic imprinting refers to
the same DNA complement, types of epigenetic marks work in a process by which particular genes, car-
which originates from a single concert to drive appropriate gene rying parent-of-origin-specic epige-
cell at conception. Highly orchestrated expression (Fig. 1). These include DNA netic marks, have the potential to drive
epigenetic mechanisms facilitate the methylation at CpG dinucleotides, monoallelic parent-of-origin-specic
complex patterning required to ensure covalent modications of histone gene expression in certain cell types
normal human development and sup- proteins, ncRNAs, and other comple- at specic times in development. In
port stable regulation of appropriate mentary mechanisms controlling germ cells and in the developing
patterns of gene expression in diverse higher order chromatin organization embryo, genome-wide epigenetic
cell types. Epigenetic mechanisms within the cell nucleus. reprogramming underpins the erasure
dene mitotically heritable differences Two unique epigenetic mecha- and reestablishment of correct epige-
in gene expression potential without nisms, X-chromosome inactivation netic patterns. These naturally
altering the primary DNA sequence and genomic imprinting, will be dis- occurring processes are distinct from
(1). These mechanisms are highly regu- cussed as examples of the importance the in vitro reprogramming process
lated by a large number of proteins that of epigenetic regulation in maintaining recently developed to induce pluripo-
establish, read, and erase specic correct patterns of gene expression in tent stem cells from somatic cells (2).
epigenetic modications, thereby early development. X-chromosome While some epigenetic marks are
dening where and when the transcrip- inactivation represents a paradigm for stable over time in particular tissues,
tional machinery can access the dosage compensation in females, others demonstrate developmental
primary DNA sequences to drive nor- resulting in monoallelic expression of plasticity. Epigenetic alterations or
mal growth and differentiation in the large numbers of X-linked genes in epimutations that arise via a number
of different mechanisms can lead to
Received November 21, 2012; revised January 16, 2013; accepted January 17, 2013; published online a variety of human disorders, including
January 26, 2013. subfertility and imprinting disorders.
M.I.-F. has nothing to disclose. S.C. has nothing to disclose. D.T.B. has nothing to disclose. M.R. has
nothing to disclose. R.W. has nothing to disclose. Both genetic and environmental factors
Reprint requests: Rosanna Weksberg, M.D., Ph.D., Division of Clinical and Metabolic Genetics, The impact epigenetic marks, generating
Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada (E-mail:
rweksb@sickkids.ca). phenotypic variation ranging from
normal variation to human disease
Fertility and Sterility Vol. 99, No. 3, March 1, 2013 0015-0282/$36.00 (3). Environmental factors such as
Copyright 2013 American Society for Reproductive Medicine, Published by Elsevier Inc.
http://dx.doi.org/10.1016/j.fertnstert.2013.01.117 maternal starvation and the use of

VOL. 99 NO. 3 / MARCH 1, 2013 607


VIEWS AND REVIEWS

a methyl group to the cytosine of the CpG dinucleotide (13).


FIGURE 1
The vast majority of mammalian DNA methylation occurs at
CpG dinucleotides (14, 15). CpGs are distributed
nonrandomly in the genome. They are concentrated in
genomic regions called CpG islands ranging in size from 200
bp to several kilobases. These CpG islands, often
unmethylated, are typically located within gene promoters of
actively transcribed housekeeping genes and tumor
suppressor genes (16). In contrast, CpG islands of silent genes
are predominantly methylated (17).
Specic proteins, such as DNA methyltransferases, can
establish or maintain DNA methylation patterns. Studies in
mice demonstrate that DNA methyltransferases are essential
for normal embryonic development (18, 19). DNA
Epigenetic mechanisms affecting gene expression. Epigenetic methylation is established de novo by the DNA
patterns are established by a number of mechanisms. Epigenetic methyltranferase (DNMT) enzymes DNMT3a and DNMT3b
marks include DNA methylation and covalent modications of and maintained through mitosis primarily by the DNMT1
histone proteins. DNA methylation is established and maintained by
the DNMT enzymes. DNA is wrapped around histone protein cores
enzyme (12). Another member of the DNMT3 family,
composed of an octamer containing two copies of each core DNMT3L, has no catalytic activity but can activate
histone: H2A, H2B, H3, and H4. Together, these form the basic unit DNMT3A to establish allele-specic methylation in imprinted
of chromatin, the nucleosome. Histone modications are regulated regions of the genome (20). DNMT1 is the primary mainte-
by several enzymes including histone acetyltransferases (HATs) and
deacetylases (HDACs). Acetylation of histone proteins by HAT is nance methyltransferase with a high afnity for hemimethy-
commonly found in euchromatin (relaxed state of chromatin) and is lated DNA (21). Its primary function is to copy the
associated with active transcription. Deacetylation of histone methylation patterns during replication. DNMT1o, one devel-
proteins by HDAC and methylation of DNA by DNMTs is a hallmark
of heterochromatin (condensed state of chromatin), which is
opmental stage-specic isoform of DNMT1, is an
associated with transcriptional repression. oocyte-derived protein that enters nuclei at the eight-cell
Inbar-Feigenberg. Basic concepts of epigenetics. Fertil Steril 2013. stage of early embryos and has an essential role in
maintenance of epigenetic marks (22).
DNA demethylation is also critical during primordial
assisted reproductive technologies (ART) have been demon- germ cell (PGC) and early embryo development (23). This
strated to impact the epigenome of the embryo. Some of the can occur via passive demethylation that is associated with
epigenetic alterations associated with maternal starvation in cell division or via active demethylation using excision repair
fetal life persist through to adulthood, likely contributing to mechanisms. The active pathway requires hydroxylation of
late-onset disorders such as cardiovascular disorder and the 5-methylcytosine to 5-hydroxymethylcytosine by the
type 2 diabetes (48). enzymes TET1 and TET2, followed by deamination by AID
and APOBEC1 before base or nucleotide excision repair (23).
All the enzymes in this pathway are expressed in mouse
Epigenetic Marks PGCs, suggesting a role in gametic epigenetic reprogramming
DNA methylation, histone modications, and ncRNAs are (24, 25).
described below as independent mechanisms, but it is impor-
tant to note that there is cross-talk between the different
epigenetic marks to regulate the epigenome (9, 10). The Histone Modications
ENCODE Project Consortium, a large collaborative effort The basic unit of chromatin consists of an octamer of histone
developed to dene all of the functional elements in the proteins, two each of H2A, H2B, H3, and H4. DNA wraps
human genome, has recently published large data sets of around this core, which provides structural stability and the
transcription, histone modications, and additional protein capacity to regulate gene expression (Fig. 1). Each core histone
binding data. These data annotate both global and regional within the nucleosome contains a globular domain and
overlapping epigenetic features, which in combination a highly dynamic N-terminal tail extending from the globular
regulate gene expression (11). domains (Fig. 1). Histone proteins have tails that can have
a number of post-translational modications including
acetylation, methylation, phosphorylation, ubiquitylation,
DNA Methylation sumoylation, ADP-ribosylation, proline isomerization, citrul-
Currently, one of the best studied epigenetic mechanisms is lination, butyrylation, propionylation, and glycosylation (26).
DNA methylation (12). DNA methylation is typically associated Recently published data from the ENCODE Project Consortium
with gene silencing through binding of methylation-sensitive analyzed 11 such histone post-translational modications in-
DNA binding proteins and/or by interacting with various cluding acetylation and methylation, which mark active and
modications of histone proteins that modulate access of repressive chromatin, as well as modications associated
gene promoters to transcriptional machinery (13). In with transcription. Assessing various histone modications
eukaryotic species, DNA methylation involves transfer of in a number of tissues, that data set identied different

608 VOL. 99 NO. 3 / MARCH 1, 2013


Fertility and Sterility

chromatin states including inactive, bimodal, and active, each through a process referred to as X-chromosome inactivation
of which has different functional properties (27). Bimodal (40). Embryos containing more than one X-chromosome
states, in which a combination of active and repressive marks (XX, XXX females and XXY males) undergo random
are present in the chromatin of a promoter region of a gene, X-chromosome inactivation at the blastocyst stage in early
facilitate rapid changes in gene expression, as might be ex- embryogenesis (41). X-chromosome inactivation is regulated
pected during early development, when differentiation and by a master switch locus, the X inactivation center (XIC),
specication occur (23). which regulates in cis the expression of the lincRNA gene
XIST (X-inactive specic transcript) and its antisense
Regulatory ncRNAs transcription unit TSIX. In mouse, the XIC senses the number
of X-chromosomes in the cell and randomly silences all but
ncRNAs are also required for epigenetic regulation of gene one of the X-chromosomes. This involves coating of all future
expression. Although eukaryotic genomes transcribe up to inactive X-chromosomes by Xist RNA, followed by polycomb
75% of genomic DNA, approximately 3% of these transcripts repressor complex 2 recruitment and the addition of silencing
encode for proteins; the majority are ncRNAs, which can be chromatin marks such as histone H3 and H4 hypoacetylation,
classied according to size and function (11, 27, 28). H3 lysine27 methylation, and DNA methylation at CpG-rich
Regulatory ncRNAs including small interfering RNAs promoters (42, 43). This process, the regulation of which is
(siRNAs), microRNAs (miRNAs), and long ncRNAs still incompletely understood, restricts each diploid cell to
(lncRNAs) play important roles in gene expression one active copy of the X-chromosome.
regulation at several levels: transcription, mRNA
degradation, splicing, and translation (29). SiRNAs are
double-stranded RNAs (dsRNA) that mediate Genomic Imprinting
post-transcriptional silencing, in part by inducing hetero- Genomic imprinting is an epigenetic process by which the
chromatin to recruit histone deacetylase complexes (30). male and female germ lines confer specic marks or
MiRNAs comprise a novel class of endogenous, small imprints onto certain chromosomal regions (44).
(1824 nucleotides in length); single-stranded RNAs gener- These imprints provide the potential for monoallelic
ated from precursor RNA cleaved by two RNA polymerase parent-of-origin-specic expression at certain times in
III enzymes DROSHA and DICER to produce mature miRNA. development or in specic cell types.
These miRNAs can control gene expression by targeting Normal imprinted gene expression is dependent on
specic mRNAs for degradation and/or translational a normal relative number of specic parental alleles and
repression (31, 32). They can also control gene expression requires contributions from the alleles of both parents. That
by recruiting chromatin-modifying complexes to DNA is, changing the number or proportion of parental alleles leads
through binding to DNA regulatory regions, thereby altering to imprint deregulation. The most extreme example of change
chromatin conformation (33, 34). Expression of miRNA in in the relative parental contributions occurs in conceptuses
human blastocysts correlates with maintenance of that contain only maternal or only paternal genomic contri-
pluripotency in embryo development (35). butions. Only maternally derived chromosomes are found in
LincRNAs, a subset of lncRNA, exhibit high conservation mature cystic ovarian teratomas (MCT), which arise from
across different species. They have been shown to guide a meiotic error during oocyte maturation (gynogenetic
chromatin-modifying complexes to specic genomic loci, conceptus). The result is the formation of a cyst containing
thereby participating in the establishment of cell typespecic tissues from each of the three embryonic germ cell layers. In
epigenetic states (36, 37). In embryonic development, contrast, an androgenetic conceptus, carrying two paternal
expression of lncRNAs, regulated by the pluripotent genomes and no maternal genome in all cells, fails to drive
transcription factors OCT4 and NANOG, facilitates cell embryonic/fetal development and results in a hydatidiform
lineagespecic gene expression (38). LincRNAs also play mole (AnCHM). MCT and AnCHM display the serious
an important role in developmental processes such as biological consequences of uniparental inheritance (45), dem-
X-chromosome inactivation and genomic imprinting (39). onstrating that both parental genomes are required for
normal development of an embryo, with the paternal genome
Specic Types of Epigenetic Regulation: X being required for extraembryonic tissues and the maternal
Inactivation and Genomic Imprinting genome being required for embryonic development.
X inactivation and genomic imprinting are specialized forms Most autosomal genes are normally expressed from both
of epigenetic regulation essential for correct dosage control of paternal and maternal alleles, whereas imprinted genes are
gene expression for the X-chromosome and for genes with expressed predominantly, or exclusively, from either the
parent-of-origin-specic expression. The study of these maternal or paternal allele in a parent-of-origin-specic
complex regulatory mechanisms has been important in manner. That is, an imprinted gene is expressed from the
elucidating how the entire epigenome is regulated. paternal allele while the maternal copy is silenced, or,
conversely, it is expressed from the maternal allele while
the paternal allele is silenced (46). Imprinted genes do not
X Inactivation conform to Mendelian genetics and do not predict
To compensate for gene dosage disparities of X-linked genes, parent-of-origin specicity for gene expression (47). About
females silence most of one of their two X-chromosomes 100 human genes have been reported to be imprinted

VOL. 99 NO. 3 / MARCH 1, 2013 609


VIEWS AND REVIEWS

(http://igc.otago.ac.nz/home.html). Many such genes play


FIGURE 2
vital roles in embryonic, fetal, and placental growth, as well
as in neurodevelopment (4749).
Imprinted genes tend to cluster together to form
imprinted domains. In humans, imprinted domains have
been found on chromosomes 6, 7, 11, 14, 15, and 20
(4951). Within these domains, imprinted genes are
regulated by imprinting centers (ICs). ICs are characterized
by the presence of differentially methylated regions (DMRs).
Such DMRs carry parent-of-origin-specic DNA methylation
and histone modication marks. These cis-acting DMRs,
along with trans-acting factors, form the basis of the
parent-of-origin-specic gene expression of imprinted genes.
For example, the insulin growth factor2 (IGF2/H19) IC core-
gulates the paternal expression of IGF2 with the maternal
expression of H19, two genes located in the same imprinted
DNA methylation reprogramming in preimplantation embryos. Soon
domain 90 kb apart (52). after fertilization both paternal (blue) and maternal (pink) pronuclei
undergo genome-wide demethylation. Although demethylation of
the maternal genome lags behind the paternal genome, both are
Epigenetic Reprogramming in Mammalian remethylated around the time of implantation (yellow arrow). In
Development contrast to most other genes, epigenetic marks, such as DNA
methylation, at imprinted loci (ICs) are established early in germ line
Our current knowledge about how the epigenome is reprog- development and are protected from the global wave of
rammed in early human development comes largely from demethylation early in embryonic development by maintenance
DNMT isoforms Dnmt1o, Dnmt1, and several other genes such as
extensive research on mouse embryos. Genome-wide epigenetic NLPR2, NLPR7, and Zfp57. The epigenetic reprogramming of the
reprogramming takes place at two pivotal developmental stages: preimplantation embryo also includes X inactivation. Notably, ART
during gametogenesis and during early embryogenesis. The rst takes place during developmental time periods that involve
event occurs in the PGCs, where epigenetic marks, both epigenetic reprogramming. Adapted from reference (109).
Inbar-Feigenberg. Basic concepts of epigenetics. Fertil Steril 2013.
tissue-specic and parent-of-origin-specic imprints, are erased.
New epigenetic marks, including parent-of-origin-specic
imprints, are established at specic developmental time points
both before and after fertilization. After fertilization, a second
wave of reprogramming of epigenetic marks occurs in the early X-chromosomes may participate in meiosis for the
embryo, including global demethylation followed by de novo production of female gametes.
DNA methylation to allow for totipotency and subsequent De novo methylation in PGCs is established by recruitment
cell-specic differentiation and lineage commitment (Fig. 2). of Dnmt3a and Dnmt3l (58). The KRAB zinc nger protein
Notably, epigenetic marks at ICs are protected from this wave ZFP57 also appears to play an important role specically in oo-
of genome-wide epigenetic reprogramming. cyte imprint establishment (50, 59). Interestingly, the timing of
gamete development differs between male and female embryos.
Spermatogenesis occurs prenatally, and the associated
Epigenetic Reprogramming in PGCs paternal-specic DNA methylation programming is complete
Genome-wide erasure and reprogramming of epigenetic marks by birth (60). The epigenetic reprogramming of oocytes occurs
is initiated in the PGCs. The sex of the developing embryo much later than in sperm. It begins at puberty and is nearly
dictates which gamete, the egg or the sperm, will develop complete in each oocyte at the time of ovulation (60).
from such early precursors. This sex-specic differentiation
includes the establishment of parent-of-origin-specic
imprints in the sperm and oocytes. Developmental Epigenetic Reprogramming in the
Studies in mice have shown that PGCs begin their Early Embryo
migration toward the developing genital ridge at day 8.5 of Before fertilization, the sperm and the egg are highly special-
embryonic life in mouse (E8.5), reaching their end point by ized, differing in their gene expression as dictated by distinct
E11.5 (53). Genome-wide demethylation of the PGCs is patterns of DNA methylation and chromatin organization
thought to occur mainly between E11.5 and E13.5, at the (61). DNA in the sperm is tightly condensed by protamines.
site of the future gonads (54). The decrease in methylation Early embryo reprogramming begins with paternal genome
is global; only 7% of CpGs remain methylated compared decondensing as these protamines are replaced by mater-
with 70%80% in embryonic stem cells (ES) and somatic cells nally derived histones (61). Soon after fertilization, the
(25). Germ cellspecic gene promoters are methylated in paternal pronuclear genome undergoes rapid demethylation
early PGCs, become demethylated, and are expressed during (Fig. 2) (6268). Demethylation in the maternal genome
reprogramming (55). At this time as well, PGCs of female follows, such that at the 4-cell stage of the embryo,
embryos down-regulate Xist RNA expression from the the DNA methylation status of the two parental genomes
inactive X-chromosome (Xi) (56, 57) so that two equivalent are equalized (68). Epigenetic marks at imprinted genes are

610 VOL. 99 NO. 3 / MARCH 1, 2013


Fertility and Sterility

protected by specic proteins (Fig. 2) from epigenetic reprog- by deregulation of epigenetic control mechanisms. Mutations
ramming throughout the preimplantation development of the in proteins that establish, erase, or read epigenetic marks can
embryo (54). negatively affect the development of multiple organ systems,
as can be seen in a variety of human genetic syndromes
such as ATRX (OMIM#301040; alpha thalassemia, mental re-
Lineage Commitment
tardation) (12), Rubinstein-Taybi (OMIM#180849; CREBBP/
Lineage commitment is well underway in the blastocyst, with EP300) (76), CHARGE (OMIM#214800; CHD7) (77), and Rett
the formation of the inner cell mass (ICM) and trophectoderm syndromes (OMIM#312750; MECP2) (78).
(TE), each carrying a unique epigenetic signature (61). An epigenetic alteration or epimutation refers to an
Declining levels of the 5-hydroxymethylcytosine (5hmC) aberrant DNA methylation or histone modication pattern.
and TET oxidases accompany progressive differentiation Such alterations occur in the absence or presence of an
(69, 70). As a result, 5-methylcytosine reaccumulates, possibly underlying genomic alteration and are called primary and
silencing the genes responsible for early developmental regu- secondary epimutations, respectively. A primary epimutation
lation (69, 70). By the time cells become fully committed to can result from aberrant erasure, establishment, or mainte-
their lineage, they will have attained a distinct epigenetic nance of epigenetic marks (50).
signature reecting their phenotype, developmental history, In nonimprinted genes, somatic epimutations caused by
and environmental inuences (61). Interestingly, placental mitotic errors in the normal maintenance of epigenetic marks
cells, derived from the trophectoderm, remain relatively can result in abnormal growth regulation. For example,
hypomethylated, compared with the differentiated deriva- approximately 20% of sporadic breast cancer displays hyper-
tives of the ICM (71), in keeping with the role of the methylation of the BRCA1 promoter, often in combination
placenta in endometrial invasion as well as its limited need with an inherited or sporadic mutation on the second allele
for extensive differentiation and longevity (61). (79). Reduced growth potential can also result from an
epimutation. For example, promoter methylation of WNT2
(Wingless-type MMTV integration site family, member 2) in
Epigenetic Mark Maintenance
placenta is associated with reduced birth weight percentile
Once established, methylation and other epigenetic marks in in the neonate (80), demonstrating that a single epigenetic
both somatic and germ line cells must be maintained over hit may impact human growth phenotypes.
the course of future cell divisions. Several genes have been
found to play a major role in this maintenance process in
Imprinting and Human Disease
addition to DNMT1 and DNMT1o; these include NLPR2,
NLPR7, and ZFP57 (59, 7275). As mentioned earlier, the Imprinted genes are subject to more complex epigenetic
ZFP57 protein functions in establishing new imprints in regulation than nonimprinted genes, providing more oppor-
oocytes; it is also needed for maintenance of methylation tunities to acquire epigenetic errors. The relative parental
more globally (59). contributions of specic imprinted regions across the genome
can be disrupted by a number of different mechanisms,
including genetic and/or epigenetic alterations (Table 1).
Epigenetic Deregulation and Human Disease Primary epimutations (i.e., those without associated
Epigenetic deregulation can result from disturbances in epige- genomic alterations) can arise from stochastic errors in
netic control mechanisms or from alterations in the epigenetic imprint reprogramming. For example, failure to erase imprints
marks themselves, either in imprinted or nonimprinted genes. will result in a gamete of one sex carrying the imprints of the
The development of multiple organ systems can be disrupted other sex. When this gamete proceeds to fertilization, the

TABLE 1

Genetic and epigenetic alterations in BWS.


Genetic
alterations Sequence changes Cytogenetic abnormalities
Mutation of the Mutations in Submicroscopic genomic Duplication, inversion, or
maternalCDKN1C NLRP2 at 19q13.42 alteration (duplication/deletion) translocation of 11p15.5
allele chromosome within chromosome 11p15.5
11p15.5
Epigenetic
alterations Methylation abnormalities
Loss of methylation at IC2 Gain of methylation at IC1 on the Methylation alterations at multiple
on the maternal chromosome maternal chromosome imprinted loci
Combined
alteration Parent of origin changes of gene copies
Paternal UPD of 11p15
Inbar-Feigenberg. Basic concepts of epigenetics. Fertil Steril 2013.

VOL. 99 NO. 3 / MARCH 1, 2013 611


VIEWS AND REVIEWS

resulting zygote will carry uniparental imprints on both BWS demonstrate important epigenotype-phenotype
parental chromosomes. Failure to establish appropriate correlations. For example, the highest tumor risks occur in
imprints in the sperm or oocyte may lead to various disorders BWS cases with molecular abnormalities that include
of growth and development, including subfertility (81). deregulation of IC1.
Genomic alterations that contribute to imprinting disorders Russell-Silver syndrome (RSS) is characterized by poor
include deletions and duplications of imprinted regions. Also as- pre- and postnatal growth, dysmorphic features, and variable
sociated with imprinting disorders is uniparental disomy (UPD), presence of limb/body asymmetry and developmental delay.
in which both homologues of a chromosomal region/segment To date, two different epigenetic defects have been associated
are inherited from only one parent (46, 82), that is, there are with RSS (89). The rst is maternal UPD of chromosome 7
two copies of an imprinted region from one parent and none occurring in about 10% of RSS cases (90).
from the other. UPD of nonimprinted genomic regions is The second is hypomethylation at the IC1 on the paternal
generally not associated with disease, whereas UPD at specic chromosome 11p15.5, which occurs in 45% of cases (89,
genomic locations containing imprinted regions can result in 9193). The latter form is essentially opposite, both in
imprinting disorders. The extent of UPD may range from phenotype and epigenotype, to cases of BWS with gain of
a small genomic segment to the entire chromosome (83). The methylation of the same region. Interestingly, some men
incidence of UPD of any chromosome is estimated to be about with oligospermia have been found to lack paternal
1:3,500 live births (84). Since deletion or duplication of an methylation at the IGF2/H19 IC1 on chromosome 11p15.5
imprinted genomic region changes the relative parental in their sperm (94). Children who inherit this chromosome
contributions, it represents another mechanism by which from their fathers are at risk for developing RSS (89).
imprinted genes can be regulated. Imprinting disorders can include imprinting errors at
Many imprinted genes are expressed in the placenta and multiple imprinted domains. For example, in both BWS and
also function in fetal growth regulation and brain RSS, some patients exhibit loss of methylation not only at
development (44). Epigenetic and associated genetic the IC on chromosome 11p15-IC1 for RSS and IC2 for BWS
aberrations at imprinted genes result in human diseases that but also at other imprinted loci. The molecular basis of this
often reect aberrant growth and/or aberrant neurodevelop- multilocus loss of methylation (MLOM) is not known for these
ment. As outlined below, it is very common for individual two conditions. However, MLOM also occurs in the imprint-
imprinting disorders to demonstrate etiologic heterogeneity. ing disorder transient neonatal diabetes mellitus (74), where
Further, different molecular etiologies may demonstrate the primary imprinting defect occurs at PLAGL1 DMR on
epigenotype/genotype-phenotype correlations. chromosome 6q24; associated LOM at other maternal DMRs
Neurologic and psychiatric disorders in which deregulation have been reported (74, 75). The cause of this MLOM
of imprinted genes are involved and parent-of-origin effects are condition has been shown to be homozygous mutations in
observed (85) include Prader-Willi syndrome (PWS) and the ZFP57 gene. This gene normally encodes an oocyte-
Angelman syndrome (AS), two distinct neurodevelopmental derived maternal factor that participates in preimplantation
syndromes that both map to the imprinted gene cluster on maintenance of imprints at multiple loci.
chromosome 15q11-q13 (86). These syndromes serve as Mutations in NLRP genes can also be associated with
an important example of how parental origin of the imprinting errors at multiple genomic loci. NLRP proteins are
genomic/epigenomic lesion denes which imprinting disorder members of the CATERPILLER protein family that is involved
occurs. A paternal deletion of chromosome 15q11-13 or maternal in inammation and apoptosis (50). Mutations of NLRP7 in the
UPD15q11-13 leads to PWS, whereas a maternal deletion of chro- oocyte are associated with aberrant imprints at multiple loci
mosome 15q11-13 or paternal UPD15q11-13 leads to AS (50). (72), causing formation of partial hydatidiform mole, which
One of the best known pediatric growth disorders caused is termed biparental due to its oocyte and sperm origins.
by abnormal imprinting is Beckwith-Wiedemann syndrome
(BWS). BWS is characterized by macrosomia, macroglossia,
and visceromegaly; embryonal tumors such as Wilms, Epigenetic Deregulation in Fetal Development
hepatoblastoma, neuroblastoma, and rhabdomyosarcoma; Early embryonic development has been shown to be a critical
omphalocele, neonatal hypoglycemia, ear creases/pits, period of susceptibility to epigenetic deregulation. Indeed,
adrenocortical cytomegaly, and renal abnormalities (46). studies of children born after maternal starvation have shown
Pregnancies in which the fetus has BWS may be complicated that it is the rst trimester of pregnancy that is a critical period
by polyhydramnios, enlarged placenta, a long thickened for epigenetic changes (3, 5, 94). Interestingly, children born
umbilical cord, increased risk of premature delivery (87), after the use of ART have also been identied to have
and placental mesenchymal dysplasia (88). The molecular changes in epigenetic modications, which are described
etiology of BWS includes two different primary epimutations: below (95, 96).
hypomethylation of the proximal IC (IC2) on the maternal
chromosome and hypermethylation at the distal IC (IC1) on
the maternal chromosome. Hypermethylation at IC2 can ART
also occur as a secondary epimutation when there is an ART accounts for 1%2% of live births in developed countries
underlying deletion of the maternal IC2 region. In cases of (95). In general, such technologies are highly successful, with
BWS with paternal UPD of chromosome 11p15.5, genomic few reports of adverse outcomes. An increased incidence of
imprints at both IC1 and IC2 are affected (51). Patients with epigenetic errors leading to imprinting disorders such as

612 VOL. 99 NO. 3 / MARCH 1, 2013


Fertility and Sterility

BWS, RSS (96), and AS (95) is reported in some studies of programming; uncovering the effects of medication and
children conceived by ART. However, not all studies have nutrition; and assessing risks for adult-onset disorders.
supported such associations (97). The variability in the
outcomes of such studies is likely due to the large numbers REFERENCES
of variable factors that could affect epigenetic reprogram- 1. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental
ming. It has been shown that subfertility itself could be origins hypothesis. Annu Rev Nutr 2007;27:36388.
associated with epigenetic errors in the sperm of oligospermic 2. Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery JR, Hon G, et al. Hotspots
males (81). Data from mice shows that epigenetic errors can of aberrant epigenomic reprogramming in human induced pluripotent
stem cells. Nature 2011;471:6873.
also arise from ovulation induction or in vitro culture of
3. Huang C, Li Z, Wang M, Martorell R. Early life exposure to the 19591961
embryos (98). We expect that future investigations of new re- Chinese famine has long-term health consequences. J Nutr;140:18748.
productive technologies will elucidate specic risk factors as- 4. Mathers JC. Early nutrition: impact on epigenetics. Forum Nutr 2007;60:
sociated with ART. In addition, we expect that only some 428.
individuals who undertake ART will be genetically susceptible 5. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, et al.
to such environmental risk factors. Persistent epigenetic differences associated with prenatal exposure to
famine in humans. Proc Natl Acad Sci U S A 2008;105:170469.
6. Chawla RK, Watson WH, Jones DP. Effect of hypoxia on hepatic DNA
Environmental Effects methylation and tRNA methyltransferase in rat: similarities to effects of
methyl-decient diets. J Cell Biochem 1996;61:7280.
Environmental factors can alter primary epigenetic marks, im- 7. Wellmann S, Bettkober M, Zelmer A, Seeger K, Faigle M, Eltzschig HK, et al.
pacting not only the developing embryo but also the next gen- Hypoxia upregulates the histone demethylase JMJD1A via HIF-1. Biochem
eration of offspring. Delineating such transgenerational Biophys Res Commun 2008;372:8927.
environmental effects will be an important focus of future 8. Yang J, Ledaki I, Turley H, Gatter KC, Montero JC, Li JL, et al. Role of
hypoxia-inducible factors in epigenetic regulation via histone demethy-
research. A documented example of the importance of
lases. Ann N Y Acad Sci 2009;1177:18597.
environment on the evolution of epigenetically based disease 9. Otani J, Nankumo T, Arita K, Inamoto S, Ariyoshi M, Shirakawa M.
arises from the Dutch and Chinese famine studies (3, 5, 94). Structural basis for recognition of H3K4 methylation status by the DNA
For example, male offspring who were exposed to the Dutch methyltransferase 3A ATRX-DNMT3-DNMT3L domain. EMBO Rep 2009;
famine of 194445 within the rst 10 weeks after conception 10:123541.
demonstrated persistent methylation changes at the imprinted 10. Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, et al.
IGF2 gene six decades later (6, 100). The historical famines Distribution, silencing potential and evolutionary impact of promoter
DNA methylation in the human genome. Nat Genet 2007;39:45766.
provide powerful natural experiments in humans that
11. The ENCODE (ENCyclopedia Of DNA Elements) Project. Science 2004;306:
resulted in the discovery of epigenetic marks that may be 63640.
modied by the prenatal environment (99). Prenatal exposure 12. Maunakea AK, Chepelev I, Zhao K. Epigenome mapping in normal and
to the famine was associated with various adverse metabolic disease States. Circ Res 2010;107:32739.
and mental phenotypes later in life, including a higher body 13. Lande-Diner L, Zhang J, Ben-Porath I, Amariglio N, Keshet I, Hecht M, et al.
mass index (100102), elevated plasma lipids (103), increased Role of DNA methylation in stable gene repression. J Biol Chem 2007;282:
12194200.
risk of schizophrenia (100102, 104, 105), and possible
14. Ibrahim AE, Thorne NP, Baird K, Barbosa-Morais NL, Tavare S, Collins VP,
cardiovascular disease (106). et al. MMASS: an optimized array-based method for assessing CpG island
Exposure to nutritional factors, environmental pollut- methylation. Nucleic Acids Res 2006;34:e136.
ants, and medications during pregnancy may also alter fetal 15. Ziller MJ, Muller F, Liao J, Zhang Y, Gu H, Bock C, et al. Genomic distribu-
epigenetic marks. For example, choline intake during preg- tion and inter-sample variation of non-CpG methylation across human cell
nancy was found to increase placental promoter methylation types. PLoS Genet 2011;7:e1002389.
of the cortisol regulation genes CRH and NR31C, leading to 16. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev
2002;16:621.
improved stress response in children by lowering cortisol
17. Costello JF, Plass C. Methylation matters. J Med Genet 2001;38:285303.
levels via the hypothalamic-pituitary-adrenal axis (107). 18. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and
Exposure to toxicants such as the pesticide vinclozolin, Dnmt3b are essential for de novo methylation and mammalian develop-
plastics, dioxin, or jet fuel were found to affect fetal and ment. Cell 1999;99:24757.
postnatal development in rats, resulting in changes consistent 19. Li E, Bestor TH, Jaenisch R. Targeted mutation of the DNA methyltransfer-
with the biology of primary ovarian insufciency and ase gene results in embryonic lethality. Cell 1992;69:91526.
polycystic ovarian syndrome in humans (108). Some of 20. Bourchis D, Xu GL, Lin CS, Bollman B, Bestor TH. Dnmt3L and the estab-
lishment of maternal genomic imprints. Science 2001;294:25369.
these epigenetic changes were transmitted to the next
21. Pradhan S, Bacolla A, Wells RD, Roberts RJ. Recombinant human
generation (109). DNA (cytosine-5) methyltransferase. I. Expression, purication, and com-
The role of epigenetic marks in translating the primary parison of de novo and maintenance methylation. J Biol Chem 1999;
genomic sequence has now moved to the forefront of human 274:3300210.
genetic studies. Current epigenetic research is addressing 22. Howell CY, Bestor TH, Ding F, Latham KE, Mertineit C, Trasler JM, et al.
a number of topics that will add to our understanding of the Genomic imprinting disrupted by a maternal effect mutation in the
Dnmt1 gene. Cell 2001;104:82938.
role of epigenetic mechanisms in human health and disease,
23. Cedar H, Bergman Y. Programming of DNA methylation patterns. Ann Rev
including characterization of cis- and trans-acting inuences Biochem 2012;81:97117.
on the genetic background; delineating cell and tissue- 24. Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani MA. Genome-
specic epigenetic marks; determining the interactions wide reprogramming in the mouse germ line entails the base excision
between epigenome and environment, with a focus on fetal repair pathway. Science 2010;329:7882.

VOL. 99 NO. 3 / MARCH 1, 2013 613


VIEWS AND REVIEWS

25. Popp C, Dean W, Feng S, Cokus SJ, Andrews S, Pellegrini M, et al. Genome- 52. Gabory A, Jammes H, Dandolo L. The H19 locus: role of an imprinted non-
wide erasure of DNA methylation in mouse primordial germ cells is coding RNA in growth and development. Bioessays 2010;32:47380.
affected by AID deciency. Nature 2010;463:11015. 53. McLaren A. Primordial germ cells in the mouse. Dev Biol 2003;262:115.
26. Gardner KE, Allis CD, Strahl BD. Operating on chromatin, a colorful 54. Feng S, Jacobsen SE, Reik W. Epigenetic reprogramming in plant and ani-
language where context matters. J Mol Biol 2011;409:3646. mal development. Science 2010;330:6227.
27. An integrated encyclopedia of DNA elements in the human genome. 55. Maatouk DM, Kellam LD, Mann MR, Lei H, Li E, Bartolomei MS, et al. DNA
Nature 2012;489:5774. methylation is a primary mechanism for silencing postmigratory primordial
28. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, germ cell genes in both germ cell and somatic cell lineages. Development
et al. Landscape of transcription in human cells. Nature 2012;489: 2006;133:34118.
1018. 56. de Napoles M, Nesterova T, Brockdorff N. Early loss of Xist RNA expression
29. Kaikkonen MU, Lam MT, Glass CK. Non-coding RNAs as regulators of gene and inactive X chromosome associated chromatin modication in develop-
expression and epigenetics. Cardiovasc Res 2011;90:43040. ing primordial germ cells. PLoS ONE 2007;2:e860.
30. Grewal SI. RNAi-dependent formation of heterochromatin and its diverse 57. Sugimoto M, Abe K. X chromosome reactivation initiates in nascent pri-
functions. Curr Opin Genet Dev 2010;20:13441. mordial germ cells in mice. PLoS Genet 2007;3:e116.
31. Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, et al. The nuclear RNase III Drosha 58. Kaneda M, Okano M, Hata K, Sado T, Tsujimoto N, Li E, et al. Essential role
initiates microRNA processing. Nature 2003;425:4159. for de novo DNA methyltransferase Dnmt3a in paternal and maternal im-
32. Hutvagner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme printing. Nature 2004;429:9003.
complex. Science 2002;297:205660. 59. Li X, Ito M, Zhou F, Youngson N, Zuo X, Leder P, et al. A maternal-zygotic
33. Chuang JC, Jones PA. Epigenetics and microRNAs. Pediatr Res 2007;61: effect gene, Zfp57, maintains both maternal and paternal imprints. Dev
24R9R. Cell 2008;15:54757.
34. Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and 60. Saitou M, Kagiwada S, Kurimoto K. Epigenetic reprogramming in mouse
siRNAs. Cell 2009;136:64255. pre-implantation development and primordial germ cells. Development
35. Rosenbluth EM, Shelton DN, Sparks AET, Devor E, Christenson L, Van 2012;139:1531.
Voorhis BJ. MicroRNA expression in the human blastocyst. Fertil Steril 61. Morgan HD, Santos F, Green K, Dean W, Reik W. Epigenetic reprogram-
2012; http://dx.doi.org/10.1016/j.fertnstert.2012.11.001. ming in mammals. Hum Mol Genet 2005;14 Spec No 1:R4758.
36. Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, et al. 62. Santos F, Hendrich B, Reik W, Dean W. Dynamic reprogramming of DNA
Many human large intergenic noncoding RNAs associate with chromatin- methylation in the early mouse embryo. Dev Biol 2002;241:17282.
modifying complexes and affect gene expression. Proc Natl Acad Sci U S A 63. Mayer W, Niveleau A, Walter J, Fundele R, Haaf T. Demethylation of the
2009;106:1166772. zygotic paternal genome. Nature 2000;403:5012.
37. Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, et al. Chromatin 64. Dean W, Santos F, Stojkovic M, Zakhartchenko V, Walter J, Wolf E, et al.
signature reveals over a thousand highly conserved large non-coding RNAs Conservation of methylation reprogramming in mammalian development:
in mammals. Nature 2009;458:2237. aberrant reprogramming in cloned embryos. Proc Natl Acad Sci U S A
38. Sheik Mohamed J, Gaughwin PM, Lim B, Robson P, Lipovich L. Conserved 2001;98:137348.
long noncoding RNAs transcriptionally regulated by Oct4 and Nanog 65. Beaujean N, Hartshorne G, Cavilla J, Taylor J, Gardner J, Wilmut I, et al.
modulate pluripotency in mouse embryonic stem cells. Rna 2010;16: Non-conservation of mammalian preimplantation methylation dynamics.
32437. Curr Biol 2004;14:R2667.
39. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding 66. Fulka H, Mrazek M, Tepla O, Fulka J Jr. DNA methylation pattern in human
RNAs. Cell 2009;136:62941. zygotes and developing embryos. Reproduction 2004;128:7038.
40. Heard E, Disteche CM. Dosage compensation in mammals: ne-tuning the 67. Lepikhov K, Wossidlo M, Arand J, Walter J. DNA methylation reprogram-
expression of the X chromosome. Genes Dev 2006;20:184867. ming and DNA repair in the mouse zygote. Int J Dev Biol 2010;54:156574.
41. Pessia E, Makino T, Bailly-Bechet M, McLysaght A, Marais GA. Mammalian 68. Hales BF, Grenier L, Lalancette C, Robaire B. Epigenetic programming: from
X chromosome inactivation evolved as a dosage-compensation mecha- gametes to blastocyst. Birth Defects Res A Clin Mol Teratol 2011;91:
nism for dosage-sensitive genes on the X chromosome. Proc Natl Acad 65265.
Sci U S A 2012;109:534651. 69. Ficz G, Branco MR, Seisenberger S, Santos F, Krueger F, Hore TA, et al. Dy-
42. Morey C, Avner P. Genetics and epigenetics of the X chromosome. Ann N namic regulation of 5-hydroxymethylcytosine in mouse ES cells and during
Y Acad Sci 2010;1214:E1833. differentiation. Nature 2011;473:398402.
43. Okamoto I, Heard E. Lessons from comparative analysis of X-chromosome 70. Koh KP, Yabuuchi A, Rao S, Huang Y, Cunniff K, Nardone J, et al. Tet1 and
inactivation in mammals. Chromosome Res 2009;17:65969. Tet2 regulate 5-hydroxymethylcytosine production and cell lineage speci-
44. Reik W, Walter J. Genomic imprinting: parental inuence on the genome. cation in mouse embryonic stem cells. Cell Stem Cell 2011;8:20013.
Nat Rev Gen 2001;2:2132. 71. Chapman V, Forrester L, Sanford J, Hastie N, Rossant J. Cell lineage-specic
45. Mutter GL. Role of imprinting in abnormal human development. Mutat Res undermethylation of mouse repetitive DNA. Nature 1984;307:2846.
1997;396:1417. 72. Judson H, Hayward BE, Sheridan E, Bonthron DT. A global disorder of im-
46. Choufani S, Shuman C, Weksberg R. Beckwith-Wiedemann syndrome. Am printing in the human female germ line. Nature 2002;416:53942.
J Med Genet C Semin Med Genet 2010;154C:34354. 73. Meyer E, Lim D, Pasha S, Tee LJ, Rahman F, Yates JR, et al. Germline muta-
47. Tycko B, Morison IM. Physiological functions of imprinted genes. J Cell tion in NLRP2 (NALP2) in a familial imprinting disorder (Beckwith-Wiede-
Physiol 2002;192:24558. mann Syndrome). PLoS Genet 2009;5:e1000423.
48. Reik W, Constancia M, Fowden A, Anderson N, Dean W, Ferguson- 74. Mackay DJ, Callaway JL, Marks SM, White HE, Acerini CL, Boonen SE, et al.
Smith A, et al. Regulation of supply and demand for maternal nutrients Hypomethylation of multiple imprinted loci in individuals with transient
in mammals by imprinted genes. J Physiol 2003;547:3544. neonatal diabetes is associated with mutations in ZFP57. Nat Genet
49. Weksberg R. Imprinted genes and human disease. Am J Med Genet C 2008;40:94951.
Semin Med Genet 2010;154C:31720. 75. Mackay DJ, Temple IK. Transient neonatal diabetes mellitus type 1. Am J
50. Horsthemke B. Mechanisms of imprint dysregulation. Am J Med Genet C Med Genet C Semin Med Genet 2010;154C:33542.
Semin Med Genet 2010;154C:3218. 76. Munshi A, Sha G, Aliya N, Jyothy A. Histone modications dictate specic
51. Choufani S, Shapiro JS, Susiarjo M, Butcher DT, Grafodatskaya D, biological readouts. J Genet Genom 2009;36:7588.
Lou Y, et al. A novel approach identies new differentially methylated 77. Schnetz MP, Bartels CF, Shastri K, Balasubramanian D, Zentner GE, Balaji R,
regions (DMRs) associated with imprinted genes. Genome Res 2011; et al. Genomic distribution of CHD7 on chromatin tracks H3K4 methylation
21:46576. patterns. Genome research 2009;19:590601.

614 VOL. 99 NO. 3 / MARCH 1, 2013


Fertility and Sterility

78. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett Silver-Russell syndrome (SRS): results from a large cohort of patients with
syndrome is caused by mutations in X-linked MECP2, encoding methyl- SRS and SRS-like phenotypes. J Med Genet 2009;46:1927.
CpG-binding protein 2. Nat Genet 1999;23:1858. 94. Tobi EW, Slagboom PE, van Dongen J, Kremer D, Stein AD, Putter H, et al.
79. Hansmann T, Pliushch G, Leubner M, Kroll P, Endt D, Gehrig A, et al. Prenatal famine and genetic variation are independently and additively
Constitutive promoter methylation of BRCA1 and RAD51C in patients associated with DNA methylation at regulatory loci within IGF2/H19.
with familial ovarian cancer and early-onset sporadic breast cancer. Hum PLoS ONE 2012;7:e37933.
Mol Genet 2012;21:466979. 95. Hiura H, Okae H, Miyauchi N, Sato F, Sato A, Van De Pette M, et al.
80. Ferreira JC, Choufani S, Grafodatskaya D, Butcher DT, Zhao C, Chitayat D, Characterization of DNA methylation errors in patients with imprinting
et al. WNT2 promoter methylation in human placenta is associated with disorders conceived by assisted reproduction technologies. Hum Reprod
low birthweight percentile in the neonate. Epigenetics 2011;6:4409. 2012;27:25418.
81. Poplinski A, Tuttelmann F, Kanber D, Horsthemke B, Gromoll J. Idiopathic 96. Eroglu A, Layman LC. Role of ART in imprinting disorders. Semin Reprod
male infertility is strongly associated with aberrant methylation of MEST Med 2012;30:92104.
and IGF2/H19 ICR1. Int J Androl;33:6429. 97. Zheng HY, Shi XY, Wu FR, Wu YQ, Wang LL, Chen SL. Assisted reproduc-
82. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syndrome. Genet tive technologies do not increase risk of abnormal methylation of PEG1/M-
Med 2012;14:1026. EST in human early pregnancy loss. Fertil Steril;96:849.e2.
83. Yamazawa K, Ogata T, Ferguson-Smith AC. Uniparental disomy and 98. Denomme MM, Zhang L, Mann MR. Embryonic imprinting perturbations
human disease: an overview. Am J Med Genet C Semin Med Genet do not originate from superovulation-induced defects in DNA methylation
2010;154C:32934. acquisition. Fertil Steril;96:7348.e2.
84. Robinson WP. Mechanisms leading to uniparental disomy and their clinical 99. Heijmans BT, Tobi EW, Lumey LH, Slagboom PE. The epigenome: archive of
consequences. Bioessays 2000;22:4529. the prenatal environment. Epigenetics 2009;4:52631.
85. Isles AR, Wilkinson LS. Imprinted genes, cognition and behaviour. Trends 100. Li Y, He Y, Qi L, Jaddoe VW, Feskens EJ, Yang X, et al. Exposure to the
Cogn Sci 2000;4:30918. Chinese famine in early life and the risk of hyperglycemia and type 2 dia-
86. Buiting K. Prader-Willi syndrome and Angelman syndrome. Am J Med betes in adulthood. Diabetes;59:24006.
Genet C Semin Med Genet 2010;154C:36576. 101. Ravelli AC, van Der Meulen JH, Osmond C, Barker DJ, Bleker OP. Obesity at
87. Weng EY, Moeschler JB, Graham JM Jr. Longitudinal observations on 15 the age of 50 y in men and women exposed to famine prenatally. Am J Clin
children with Wiedemann-Beckwith syndrome. Am J Med Genet 1995; Nutr 1999;70:8116.
56:36673. 102. Stein AD, Kahn HS, Rundle A, Zybert PA, van der Pal-de Bruin K, Lumey LH.
88. Wilson M, Peters G, Bennetts B, McGillivray G, Wu ZH, Poon C, et al. Anthropometric measures in middle age after exposure to famine during
The clinical phenotype of mosaicism for genome-wide paternal unipa- gestation: evidence from the Dutch famine. Am J Clin Nutr 2007;85:
rental disomy: two new reports. Am J Med Genet A 2008;146A: 86976.
13748. 103. Lumey LH, Stein AD. Transgenerational effects of prenatal exposure to the
89. Eggermann T. Russell-Silver syndrome. Am J Med Genet C Semin Med Dutch famine. Br J Obstet Gynecol 2009;116:868. author reply.
Genet 2010;154C:35564. 104. Susser EB, Brown A, Matte TD. Prenatal factors and adult mental and phys-
90. Monk D, Bentley L, Hitchins M, Myler RA, Clayton-Smith J, Ismail S, ical health. Can J Psychiatry 1999;44:32634.
et al. Chromosome 7p disruptions in Silver Russell syndrome: delineat- 105. Xu MQ, Sun WS, Liu BX, Feng GY, Yu L, Yang L, et al. Prenatal malnutrition
ing an imprinted candidate gene region. Hum Genet 2002;111: and adult schizophrenia: further evidence from the 1959-1961 Chinese
37687. famine. Schizophr Bull 2009;35:56876.
91. Horike S, Ferreira JC, Meguro-Horike M, Choufani S, Smith AC, Shuman C, 106. Painter RC, de Rooij SR, Bossuyt PM, Simmers TA, Osmond C, Barker DJ,
et al. Screening of DNA methylation at the H19 promoter or the distal et al. Early onset of coronary artery disease after prenatal exposure to
region of its ICR1 ensures efcient detection of chromosome 11p15 the Dutch famine. Am J Clin Nutr 2006;84:3227, quiz 4667.
epimutations in Russell-Silver syndrome. Am J Med Genet A 2009;149A: 107. Jiang X, Yan J, West AA, Perry CA, Malysheva OV, Devapatla S, et al.
241523. Maternal choline intake alters the epigenetic state of fetal cortisol-
92. Netchine I, Rossignol S, Dufourg MN, Azzi S, Rousseau A, Perin L, et al. regulating genes in humans. Faseb J 2012;26:356374.
11p15 imprinting center region 1 loss of methylation is a common and 108. Nilsson EE, Anway MD, Staneld J, Skinner MK. Transgenerational
specic cause of typical Russell-Silver syndrome: clinical scoring system epigenetic effects of the endocrine disruptor vinclozolin on preg-
and epigenetic-phenotypic correlations. J Clin Endocrinol Metab 2007; nancies and female adult onset disease. Reproduction 2008;135:
92:314854. 71321.
93. Bartholdi D, Krajewska-Walasek M, Ounap K, Gaspar H, Chrzanowska KH, 109. Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian devel-
Ilyana H, et al. Epigenetic mutations of the imprinted IGF2-H19 domain in opment. Science 2001;293:108993.

VOL. 99 NO. 3 / MARCH 1, 2013 615

También podría gustarte