Two epigenetically labile subsets of genes that link embryonic environmental exposures with adult disease susceptibility are those that are imprinted and those with metastable epialleles. The expression of genes with metastable epialleles, like the agouti gene in Agouti viable yellow (Avy) mice, is highly variable between individuals but uniform in tissues within an individual. We used the Avy mouse to demonstrate that exposure to nutritional supplements, chemical toxicants, and low-dose ionizing radiation during embryogenesis alters adult disease susceptibility by modifying the epigenome. Genomic imprinting is a unique species-dependent epigenetic form of gene regulation that evolved approximately 150 million years ago in a common ancestor to Therian mammals. It resulted in monoallelic parent-of-origin-dependent gene silencing. Thus, imprinted genes are functionally haploid disease susceptibility loci, since only a single genetic or epigenetic event is required to alter their function. Expression of imprinted genes in the human genome is regulated by hemi-methylated imprint control regions (ICRs) in the human imprintome. Furthermore, human imprintome ICRs associated with chronic diseases (e.g., cancer, diabetes, and obesity) and behavioral disorders (e.g., autism, bipolar disorder, psychopathy, and schizophrenia) can now be identified with the use of cells from peripheral samples and the human imprintome array. The importance of metastable epialleles and imprinted genes in the etiology of environmentally induced human chronic diseases is discussed in this review.

Epigenetics was first defined by the English developmental biologist Conrad Waddington [1] as ‘…the interactions of genes with their environment which bring the phenotype into being’. Holliday and Pugh [2] proposed in 1975 that covalent chemical DNA modifications, including methylation of cytosine at CpG dinucleotide sites, were the molecular mechanisms behind Conrad’s hypothesis. Subsequently, epigenetics was defined as the study of changes in gene expression that occur not by changing the DNA sequence but by modifying DNA methylation and remodeling [3]. Only 100 papers were published on epigenetics in 1990, but the number increased to 17,000 in 2024. This 170-fold increase in annual publications in 34 years demonstrates that the field of epigenetics is growing exponentially with a doubling time of four years.

Developmental origin of health and disease

Forty years ago, Barker and Osmond demonstrated that mortality rates from chronic diseases in England and Wales in the 1970s were correlated geographically with increased death rates among newborn babies caused by poor nutrition [4]. These findings resulted in the novel postulate that retardation of growth during critical periods of development in fetal life, and subsequent low birth weight, were associated in adulthood with chronic diseases including cancer, cardiovascular disease, and bronchitis [5]. Subsequent epidemiologic studies of survivors of the Dutch famine during World War II (1944–1945) and the Chinese Great Leap Forward famine (1959–1961) provided additional evidence that significant reduction in food availability, particularly during the first trimester of pregnancy, increases the adult risk of developing a variety of chronic diseases such as cancer, cardiovascular disease, diabetes, kidney disease, obesity, and schizophrenia [6–8]. Moreover, there is evidence that the increased risk of these chronic diseases can be inherited transgenerationally, possibly by changes in the epigenome [8–10].

These results ultimately evolved into the developmental origins of health and disease (DOHaD) hypothesis that posits the intriguing idea that the evolution of developmental plasticity, which enables an organism to adapt to environmental signals during early life, can also increase the risk of developing chronic diseases when there is a mismatch between the perceived environment during gestation and that which is actually encountered in adulthood [11]. This postulate, however, was met with skepticism because at the time, there was no known mechanism linking molecular changes induced by environmental exposures during early development to disease formation decades later.

With the use of the Agouti viable yellow (Avy) mouse, we demonstrated in 2003 that exposure to enhanced methyl donor supplements during embryogenesis induced persistent DNA methylation changes at the Agouti locus that resulted in alterations in adult phenotype [12]. Our study provided a plausible mechanism for DOHaD and provided the first evidence that epigenetic modifications (e.g., DNA methylation) in early life impacts later health [13]. These findings ushered in the era of environmental epigenomics [14], and the first international meeting on this subject was held in 2005; it can be viewed at https://www.geneimprint.com/site/meetings/2005-durham.

There is now evidence that two epigenetically labile targets—imprinted genes and those with metastable epialleles—link environmental exposures during early embryonic development to adult diseases. Their potential role in the etiology of environmentally induced chronic diseases and the strategies to investigate their influence on human health and disease are discussed in this review.

Agouti mouse study—nutritional supplements

The highly variable expression of genes with metastable epialleles results from stochastic allelic changes in the epigenome (e.g., DNA methylation) during early embryonic development rather than from mutations in the genome. Genes with metastable epialleles have large variability in expression between individuals but low variability in gene expression between tissues in an individual. Thus, the ratio of these two gene-expression variances is expected to be large for genes with metastable epialleles.

The formation of the embryonic DNA methylation that regulates metastable epiallele expression is controlled by both the level of methyl donors in the diet (e.g., folic acid, vitamin B12, choline chloride and betaine), and the environmental conditions that modify the efficiency of one-carbon metabolism. Additionally, the activity of DNA methyltransferase 3 alpha (DNMT3A) and DNA methyltransferase 3 beta (DNMT3B) functions in de novo methylation [15], and DNA methyltransferase 1 (DNMT1) functions in maintenance DNA methylation [16]. Thus, any chemical or physical exposure during early development that affects either methyl donor levels or DNMT3A, DNMT3B, and/or DNMT1 activity can modify the DNA methylation of metastable epialleles, thereby altering their expression in adulthood and the pathogenesis of disease formation.

The most actively investigated metastable genes in mice are the agouti gene in the Avy mouse [17], the AxinFu gene [18], and the CabpIAP gene [19], although additional metastable epialleles have been identified in the mouse [20]. Moreover, metastable epialleles or correlated regions of systemic interindividual variation (CoRSIVs) have been identified in cattle [21] and the human [22–24], indicating that these systemic epigenetic variants are common in mammals.

The Avy mouse varies in coat color, ranging from brown (i.e., methylated intracisternal A-particle [IAP]) to yellow (i.e., unmethylated IAP) with intermediate mottled Avy mice that are epigenetically mosaic because it harbors a metastable Agouti gene (Figure 1A). A retroviral IAP is inserted approximately 100 kb upstream of the gene (Figure 1B) [26,27]. The degree of methylation of CpG sites in the IAP at the cryptic promoter region in the proximal end of the IAP varies dramatically among individual isogenic Avy mice, causing a wide distribution in coat color. Hypomethylation of this alternative promoter results in inappropriate Agouti gene expression throughout the Avy mouse. This not only leads to a yellow coat color; it also antagonizes MC4R in the hypothalamus, causing the animals to overeat, thereby becoming obese, and subsequently developing diabetes and cancer at a high frequency. In contrast, the incidence of these diseases is markedly reduced in pseudoagouti (i.e., brown) offspring that develop when this ectopic promoter is hypermethylated, and the developmental expression of the Agouti gene is limited to the hair follicles [28–30].

Agouti mouse nutritional supplement study.

Figure 1:
Agouti mouse nutritional supplement study.

(A) Isogenic Avy/a mouse littermates representing the five coat-color phenotypes. (B) The contraoriented IAP insertion within PS1A of the murine Agouti gene. A cryptic promoter (i.e., short arrow labeled Avy) drives ectopic Agouti expression. CpG sites 1–7 are oriented in the 3´ to 5´ direction with respect to the IAP insertion. Transcription of A and a alleles initiates from a hair-cycle-specific promoter in exon 2 (i.e., short arrow labeled A,a). (C) Coat color distribution of Avy/a offspring born to mothers fed an unsupplemented diet (i.e. yellow bars) and a methyl donor supplemented diet (i.e. brown bars). (D) CpG methylation within the Avy PS1A of Avy/a offspring from mothers fed unsupplemented and methylation supplemented diets; redrawn from [12,25].

Figure 1:
Agouti mouse nutritional supplement study.

(A) Isogenic Avy/a mouse littermates representing the five coat-color phenotypes. (B) The contraoriented IAP insertion within PS1A of the murine Agouti gene. A cryptic promoter (i.e., short arrow labeled Avy) drives ectopic Agouti expression. CpG sites 1–7 are oriented in the 3´ to 5´ direction with respect to the IAP insertion. Transcription of A and a alleles initiates from a hair-cycle-specific promoter in exon 2 (i.e., short arrow labeled A,a). (C) Coat color distribution of Avy/a offspring born to mothers fed an unsupplemented diet (i.e. yellow bars) and a methyl donor supplemented diet (i.e. brown bars). (D) CpG methylation within the Avy PS1A of Avy/a offspring from mothers fed unsupplemented and methylation supplemented diets; redrawn from [12,25].

Close modal

This makes the Avy mouse an excellent, exquisitely sensitive biosensor for identifying environmental exposures, early in development, that alter adult disease susceptibility by modifying the epigenome rather than by mutating the genome. When the Avy mice were exposed during pregnancy to nutritional donor supplements (i.e., folic acid, vitamin B12, choline chloride and betaine), the coat color of their offspring shifted to heavily mottled and brown (Figure 1C) concomitant with a significant increase in DNA methylation of IAP at the Avy locus (Figure 1D) [12]. A plausible mechanism for DOHaD had been identified (ShortCutstv Documentary: The Agouti Mouse Study - https://www.youtube.com/watch?v=VM37fh5ykbg).

Genistein is a major phytoestrogen in soy linked to cancer chemoprevention and decreased adipose deposition. Maternal Avy mouse dietary genistein supplementation during gestation, to levels comparable with humans consuming high-soy diets, also shifted the coat color of the Avy/a offspring toward brown [25]. This marked phenotypic change was likewise associated with increased methylation of the IAP upstream of the transcription start site of the Agouti gene (Figure 1B). The extent of this DNA methylation was similar in tissues from the three germ layers, indicating that genistein acts during early embryonic development. Moreover, this genistein-induced hypermethylation persisted into adulthood, decreasing ectopic agouti expression and protecting the offspring from obesity. Genistein is not a methyl donor, and no evidence was found of enhanced efficiency of one or more steps in the one-carbon metabolism pathway. Thus, the effect of genistein on IAP methylation during early embryonic development may be mediated by modifying the activity of DNMT3A, DNMT3B [15] and/or DNMT1 [16]. This was the first evidence that in utero dietary genistein affects gene expression and alters susceptibility to obesity in adulthood by permanently altering the epigenome.

Agouti mouse study—nongenotoxic agents

Perinatal exposure to the endocrine active compound, bisphenol A (BPA), a chemical used in the manufacture of polycarbonate plastic, epoxy resin, and other polymer materials, is associated with higher body weight, increased breast and prostate cancer, and altered reproductive function [31]. Maternal exposure of Avy mice to this endocrine disruptive, non-genotoxic compound [32] shifted the coat color distribution of the mouse offspring toward yellow by decreasing DNA methylation at the IAP of the Avy locus (Figure 1B); DNA methylation was also decreased at the CabpIAP metastable epiallele locus [33]. DNA methylation at the Avy locus was similar in tissues from the three germ layers, providing evidence that epigenetic patterning during early stem cell development is sensitive to BPA exposure. Moreover, maternal dietary supplementation, with either methyl donors or the phytoestrogen genistein, blocks the negative DNA hypomethylating effect of BPA. These results provide compelling evidence that early developmental exposure of Avy mice to BPA increases offspring disease susceptibility by stably altering the epigenome, an effect that can be counteracted by maternal dietary supplements. Thus, food is medicine!

Agouti mouse study—low-dose ionizing radiation

The negative human health effects resulting from moderate to high-dose exposures (i.e., > 100 cGy) have been well documented since the discovery of ionizing radiation in 1895; however, the majority of human exposures occur in the low-dose range (i.e., < 10 cGy). In addition to inducing genetic mutations, low-dose ionizing radiation (LDIR) may also alter the epigenome.

We first demonstrated over a decade ago that LDIR exposure of the Avy mouse during pregnancy causes positive adaptive changes in the offspring by significantly increasing DNA methylation at the Agouti locus (Figure 1B) [34]. Average DNA methylation was significantly increased in offspring exposed to doses between 0.7 and 7.6 cGy, with maximum effects at 3 cGy; this effect was seen in both male and female mice, but was significantly greater in males. Offspring coat color was concomitantly shifted toward brown, which is linked to reduced obesity, diabetes, and cancer in the Avy mouse. Thus, LDIR exposure of Avy mice in utero results in the offspring being healthier than those that are not exposed to radiation. The phenomenon that occurs when an environmental factor that is toxic at high doses stimulates a beneficial adaptive response in an organism at low doses is called hormesis [35].

Interestingly, maternal dietary antioxidant supplementation negated the LDIR-induced coat color shift of the offspring to brown, the increased IAP DNA methylation, and the positive adaptive health responses in the offspring exposed to 3 cGy [34]. Consequently, LDIR exposure during gestation elicits epigenetic alterations that lead to positive adaptive phenotypic changes that are mitigated with antioxidants. This indicates the hormetic effects are mediated in part by oxidative stress. These findings provide evidence that in the isogenic Avy mouse, epigenetic alterations resulting from LDIR play a role in radiation hormesis; however, they do not define the repertoire of genes whose expressions are altered by exposure to LDIR. This would require the use of next-generation sequencing technologies, like those developed by Oxford Nanopore Technologies (Oxford, UK) [36], to determine the human methylome in a radiation dose-dependent manner. This long-read sequencing technology can directly determine the DNA methylation of CpG sites, including regions that are difficult to map using short-read methods, making it particularly useful for studying differential methylation throughout the human genome [37].

Our findings bring into question the assumption that every dose of radiation is harmful, as required by the LNT radiation risk model (Figure 2). Although our important epigenetic LDIR study is not often discussed in the radiation risk literature, it makes a critical contribution to the debate that occurred over a quarter of a century ago on the importance of radiation hormesis in human risk assessment and health.

LNT and hormesis radiation dose response models.

Figure 2:
LNT and hormesis radiation dose response models.

Linear no-treshold (LNT) and hormesis radiation risk assessment models of negative biological effect (e.g., unhealthy yellow Avy mice, cancer incidence) versus radiation dose overlap at high doses where most radiation response data are collected (magenta ellipse) but vary markedly in their assessment of risk in the hormetic region at doses < 10 cGy (gray area). Exposure of Avy mice in utero to LDIR results in a decrease in the incidence of unhealthy yellow Avy offspring with a concomitant increase in the healthy brown offspring. The nadir of this epigenetically regulated hormetic effect is at 1–3 cGy [34]; redrawn from [38].

Figure 2:
LNT and hormesis radiation dose response models.

Linear no-treshold (LNT) and hormesis radiation risk assessment models of negative biological effect (e.g., unhealthy yellow Avy mice, cancer incidence) versus radiation dose overlap at high doses where most radiation response data are collected (magenta ellipse) but vary markedly in their assessment of risk in the hormetic region at doses < 10 cGy (gray area). Exposure of Avy mice in utero to LDIR results in a decrease in the incidence of unhealthy yellow Avy offspring with a concomitant increase in the healthy brown offspring. The nadir of this epigenetically regulated hormetic effect is at 1–3 cGy [34]; redrawn from [38].

Close modal

At the turn of the millennium, the argument concerning the scientific validity of radiation hormesis was quite contentious and remains so today. This conflict ultimately resulted in a debate in 1998 between John Cameron, Professor of Medical Physics at the University of Wisconsin, and John Moulder, Professor of Radiation Oncology at the Medical College of Wisconsin, on whether radiation hormesis occurs at low doses of radiation exposure [38]. Cameron presented a significant amount of experimental animal and human epidemiological data in support of radiation hormesis, which ultimately resulted in his publishing a review article on this subject where he concluded, ‘We need increased background radiation to improve our health’ [39]. Moulder argued against the phenomenon of hormesis, principally because no known biological mechanism existed to support its existence.

Our LDIR Avy mouse study is not only consistent with the existence of radiation hormesis but also provides direct evidence that the mechanism of radiation hormesis involves epigenetic modifications that alter the regulation of gene expression [34]. If ionizing radiation only caused genetic mutations, the LNT model shown in Figure 2 would be correct; however, LDIR also modifies the epigenome—the cellular programs that tell the genes when, where, and how to work. At low radiation doses, this gives rise to the radiation dose-dependent positive adaptive changes observed in our Avy LDIR study that maximize at 3 cGy and are lost near 10 cGy (Figure 2). This dose response is described best by a radiation hormesis model of radiation risk assessment, not the LNT model.

Hormesis is a biphasic dose-response phenomenon where low doses of radiation induce positive adaptive effects, while high doses lead to harmful effects [40]. Hormetic dose response relationships are present at the cell, organ, and individual level and involve bystander communications. The bystander effect also results from changes in the epigenome, rather than mutations in the genome, since it is blocked by the elimination of the DNA cytosine methyltransferases, DNMT1 and DNMT3A [15].

The role of hormesis in determining cancer risk at low doses of radiation has become of particular importance, since the increasing number of people receiving CT scans raises the possibility of increased radiation-induced malignancies in the population based upon the LNT model of radiation risk assessment [41–43]. Cancer risk could also potentially be increased from high natural background radiation [44] and nuclear power plant disasters [45]. A recent study demonstrated, however, that a preponderance of studies, using higher quality case control and cohort methodology, indicate that cumulative radiation doses up to 100 mSv do not increase cancer risk, consistent with the findings in a number of other experimental and human studies [39,44,46]. Because of the paucity of cancer response data at doses < 10 cGy and its high variability, the ability of epidemiological studies to discriminate between the LNT and hormetic dose response models is low [40].

Thus, rather than continuing to debate about the existence of radiation hormesis [38], the time has come to use next-generation DNA methylation sequencing techniques [36] to define the repertoire of gene regulatory regions and the genes and biochemical pathways they control that are altered epigenetically in a radiation dose-dependent manner. It will be interesting to see if LDIR is beneficial to human health, as postulated over two decades ago by Cameron [38], because it significantly alters epigenetically the expression of genes involved in 1) damage prevention by temporarily stimulating detoxification of molecular radical species; 2) damage repair by temporarily stimulating repair mechanisms; 3) damage removal by stimulating apoptosis; and 4) damage removal by stimulating the immune response. We presently need this information to accurately define human risk to LDIR and to identify novel ways to protect humans, as we become more reliant on nuclear power for electricity and venture deeper into space.

Genomic imprinting is an example of intergenerational epigenetic inheritance where the transmission of parental genomes to the mammalian embryo results in epigenetically mediated, parentally biased gene expression. Thus, it results in an exception to Mendel’s Laws of inheritance.

The first experimental evidence that the parental genomes of mammals are not functionally equivalent came from elegant mouse nuclear transplantation studies in the mid-1980s (Figure 3) [47–49]. They demonstrated that the abortive pregnancies resulting from diploid gynogenotes derived from two female pronuclei are markedly different from those that developed from diploid androgenotes derived from two male pronuclei. The former displayed severe placental defects, while the latter had marked embryonic growth retardation. Thus, in contrast to parthenogenesis being observed in fish, amphibians, reptiles, and birds, it does not normally occur in Therian mammals [50,51]. Interestingly, however, it has now been shown that of all the imprint control regions (ICRs) in the mouse genome, the paternally methylated germline ICRs regulating imprinting at the Igf2/H19 and Dlk1/Dios imprinted domains are the only paternal barriers for the development of parthenogenesis in mice [52].

Maternal and paternal genomes are not functionally equivalent.

Figure 3:
Maternal and paternal genomes are not functionally equivalent.

Normal mouse development occurs when one pronucleus is female and the other is male. Mouse nuclear transplantation studies show abnormal development of the trophoblasts and embryo in diploid gynogenotes derived from two female pronuclei and diploid androgenotes derived from two male pronuclei, respectively; redrawn from [47,48].

Figure 3:
Maternal and paternal genomes are not functionally equivalent.

Normal mouse development occurs when one pronucleus is female and the other is male. Mouse nuclear transplantation studies show abnormal development of the trophoblasts and embryo in diploid gynogenotes derived from two female pronuclei and diploid androgenotes derived from two male pronuclei, respectively; redrawn from [47,48].

Close modal

In 1974, the T-maternal effect (Tme) locus was reported with parent-of-origin effects on the viability of mice with small deletions in chromosome 17 [53]. Using this murine model, Denise Barlow identified in 1991 the first imprinted gene, Igf2r, which is expressed only from the maternal allele [54]. In addition to the scavenging of Igf2, Igf2r is involved in the intracellular trafficking of lysosomal enzymes, TGFβ1 activation, T cell-mediated apoptosis, and the internalization of extracellular phosphomannosyl glycoproteins [55]. Additionally, the Igf2r plays a central role in mediating the signaling pathways required for Igf2 to facilitate memory and synaptic plasticity [56]. Soon after Igf2r was identified to be imprinted, Igf2 was demonstrated to be imprinted and paternally expressed [57]. The following year, the first human gene identified to be imprinted was the maternally expressed H19 [58]. This gene was subsequently found to form a reciprocally imprinted domain at chromosome location 11p15.5 with paternally expressed IGF2 [59]. The era of genomic imprinting research had begun, and there are now approximately 143 genes experimentally demonstrated to be imprinted in mice, 120 in humans, and 23 in marsupials (https://www.geneimprint.com/site/genes-by-species).

Evolution of genomic imprinting

Some imprinted genes in eutherian mammals (e.g. human and mouse) are likewise imprinted in marsupials (e.g., wallaby and opossum) while others are uniquely imprinted only in Eutherians or marsupials [60–63] with the first NNAT being the first Eutherian-specific imprinted gene identified [64]. In contrast, genes tested for imprint status in the monotremes (e.g., echidna and platypus) and Aves (e.g., chicken) are biallelically expressed [65–68]. These findings are consistent with genomic imprinting originating in the Jurassic period approximately 150 MYA in a common ancestor to marsupials and eutherians [65,67,69,70]. The phenomenon of genomic imprinting also evolved independently in some insects [71] and flowering plants [72]. The functional haploidy resulting from a gene being imprinted enables a single genetic mutation or epigenetic modification to alter the function of an imprinted gene, making imprinted genes unique disease susceptibility loci.

Scientists have struggled to formulate a theory to explain the adaptive evolutionary advantage of imprinted genes, since their development eliminates the protection that diploidy affords against the deleterious effects of recessive mutations. A number of theories have been proposed to explain its evolution, such as the co-adaption theory, the genome defense theory, and the conflict/kinship theory (see reviews [60,73,74]). A comparison of the paternal expression of the growth factor, Igf2 [57], and the maternal expression of Igf2r [54], the receptor involved in the degradation of Igf2, highlights their opposite effects on growth. These opposite effects led to the formulation of the most prominent and actively debated theory for the evolution of genomic imprinting, the parental kinship or conflict hypothesis [75,76]. This theory maintains that genomic imprinting developed in response to viviparity and polygamy and speculates that fitness effects during placental development were the principal factors that shaped its evolution. For the first time in evolutionary history, the placenta exists as an interface in which both the paternal and maternal genomes can exert their influence on resource allocation within the intrauterine environment. Postnatal nurturing behavior, like lactation, further extends this interaction between the mother and offspring, indicating that the mammary gland may be the functional equivalent of the placenta in the postnatal stage of eutherian mammal development [77].

Consequently, imprinted genes are not only prominently expressed in the placenta and mammary gland but also half of known imprinted genes are imprinted in the brain and affect behavior [62,78]. Pioneering mouse chimeric studies demonstrated that gynogenote/WT chimeric embryos developed abnormally large brains, whereas androgenote/WT chimeras had small brains [79]. Furthermore, the gynogenote cells were not distributed randomly throughout the brain, but rather were located in the cortex, striatum, and hippocampus; whereas, the androgenote cells were enriched in the hypothalamus. These results provide evidence that genomic imprinting may have facilitated a rapid expansion of the mammalian brain and altered behavioral development over evolutionary time. These findings are also consistent with the imprinted brain theory, an extension of the conflict theory, which posits that skewed paternal and maternal expression of imprinted genes results in the behavioral conditions of autism and schizophrenia, respectively [80,81]. The identification of the human imprintome [82] and the imprintome array [83], coupled with the development of high-throughput DNA sequencers, may finally allow for this novel postulate to be experimentally tested.

According to the conflict theory, genes expressed from the paternal allele favor increased maternal investment to enhance an offspring’s own fitness at a cost to all others. In contrast, genes expressed from the maternal allele maximize reproductive fitness of the mother, ensuring availability of resources for all of her current and future progeny. This postulate explains successfully the parental allelic expression bias of a number of imprinted genes, including the paternal expression of Igf2, a growth enhancer, and the maternal expression of Igf2r, a growth inhibitor [75,84]. It likewise is consistent with the opposing growth effects of Dlk1 and Grb10 [85]. Supporting evidence for this postulate also comes from the discovery of imprinted genes in eutherian mammals and marsupials, but not in the egg-laying monotremes or birds [62,65,67,68,70], another prediction of the conflict theory [75].

Imprinting mechanisms

Genomic imprinting appears to have evolved independently at least three times. In the mealybug, imprinting is manifested as heterochromatization of the entire paternal genome selectively in males [86]. In Arabidopsis, genes are imprinted primarily in the nutrient-providing endosperm, and a number have been identified to be imprinted, including MEA and PHE1, which are maternally and paternally expressed, respectively [87].

Both classical or canonical and non-canonical genomic imprinting exist in eutherian mammals [88,89]. Canonical imprinting is regulated by ICRs that are differently methylated at CpG sites in a parental-dependent manner (Figures 4 and 5) [82,90]. The ICRs can be hundreds to thousands of bases in length [82], and since imprinted genes are frequently clustered, control one or more proximal genes (Figure 4). Canonical imprinted genes participate in metabolism, embryonic and placental growth, and brain development. In contrast, non-canonical imprinted genes are not regulated by differentially methylated ICRs, as in canonical imprinting. This form of genomic imprinting relies on histone modifications like H3K27me3. It primarily occurs in the maternal germline and affects gene expression in the developing embryo, particularly in the placenta [88,89].

Models of imprinted gene expression control in mammals.

Figure 4:
Models of imprinted gene expression control in mammals.

(A) Chromatin boundary imprinting model, and (B) noncoding RNA imprinting model; redrawn from [90]. Black box, unexpressed gene, black circle, cohesin, blue box, paternally expressed gene, black oval, methylated ICR; gray box, biallelically expressed gene; light gray oval, CTCF; magenta box, maternally expressed gene, white oval, unmethylated ICR. ICR, imprint control region.

Figure 4:
Models of imprinted gene expression control in mammals.

(A) Chromatin boundary imprinting model, and (B) noncoding RNA imprinting model; redrawn from [90]. Black box, unexpressed gene, black circle, cohesin, blue box, paternally expressed gene, black oval, methylated ICR; gray box, biallelically expressed gene; light gray oval, CTCF; magenta box, maternally expressed gene, white oval, unmethylated ICR. ICR, imprint control region.

Close modal

Candidate ICR for human IGF2R.

Figure 5:
Candidate ICR for human IGF2R.

WGBS identified a candidate gICR (gray rectangle) in intron 2 of the human IGF2R (ICR_409 and ICR_410, red rectangles) in tissues from the three embryonic germ layers (i.e., brain, kidney, and liver); the average percent methylation in these tissues is approximately 50%. The average DNA methylation in sperm and oocytes is 0% and 100%, respectively. Dots indicate hemi-methylated (green), hypomethylated (blue) and hypermethylated (yellow) CpG sites [82], https://humanicr.org/. gICR, germline imprint control regionICR, imprint control region; WGBS, whole genome bisulfite sequencing.

Figure 5:
Candidate ICR for human IGF2R.

WGBS identified a candidate gICR (gray rectangle) in intron 2 of the human IGF2R (ICR_409 and ICR_410, red rectangles) in tissues from the three embryonic germ layers (i.e., brain, kidney, and liver); the average percent methylation in these tissues is approximately 50%. The average DNA methylation in sperm and oocytes is 0% and 100%, respectively. Dots indicate hemi-methylated (green), hypomethylated (blue) and hypermethylated (yellow) CpG sites [82], https://humanicr.org/. gICR, germline imprint control regionICR, imprint control region; WGBS, whole genome bisulfite sequencing.

Close modal

Canonical imprinting mechanism in vertebrates is more complex than in insects and plants. The inherited gametic imprint regulatory marks necessarily undergo a cycle involving their establishment in the PGCs of one generation, maintenance during somatic cell divisions throughout life in the resulting individual, and erasure and re-establishment in the germ cells during embryogenesis to reflect the sex of the individual in which they reside [14,91,92]. DNA methylation is a candidate for this inherited marking system, since it can be modulated with the help of the de novo methyltransferases DNMT3A, DNMT3B, and DNMT3L in PGCs and maintained throughout life with the aid of DNMT1 [93–95]. Proteins like CTCF, coupled with its testis-specific counterpart CTCFL, non-coding RNAs, and methyl-CpG-binding domain proteins, which recruit histone deacetylases, are all involved in imprinting regulation, emphasizing the complexity of the imprinting process [92,96,97].

The ICRs that regulate canonical genomic imprinting are present in imprinted gene clusters where discrete cis-acting DNA elements that carry a heritable epigenetic mark distinguish the two parental alleles [92]. A germline imprint control region (gICR) in Eutherians contains DNA methylation at specific CpG sites, resulting in both a methylated and unmethylated allele in somatic cells. For these gICRs, the inherited methylation marks can be paternally derived from the sperm, as in the gICR located upstream of H19 in the IGF2/H19 cluster (Figure 4A) [92,98] or maternal in origin like the gICR in intron 2 of murine Igf2r (Figure 4B).

The ICRs have to be protected from the wave of DNA demethylation that occurs soon after fertilization, or the parental imprint memory of the ICRs would be lost. DPPA3 protects imprinted genes in the zygote from active demethylation by inhibiting 5mC conversion to 5hmC [99]. In rodents, Zfp57 and Zfp445 cooperate in preserving the ICRs during cleavage divisions [100]. In humans, ZFP445 appears to play a larger role in this process [100–102]; however, mutations in ZFP57 are present in individuals with transient neonatal diabetes who show a variable pattern of DNA hypomethylation at imprinted loci throughout the genome [103]. Interestingly, we predict ZFP57 (ICR_383) to potentially be imprinted while ZFP445 is not predicted to be imprinted [82].

CpG-rich sequences can also acquire gICR-dependent, parental-specific DNA methylation somatic imprint control region (sICR) marks around the time of implantation. An example is the sICR in the promoter region of mouse Igf2r that is methylated on the paternal allele (Figure 4B) [104,105]. Additionally, since the gICR imprint regulatory marks may not always be ‘read’, monoallelic expression of imprinted genes can be dependent upon cell type, developmental stage, and sex of the individual, resulting in significant cell type variation in monoallelic expression [106,107]. It has been proposed that this epigenetically mediated variation in imprinted gene expression may have played a role in mammalian speciation [108]. This would help explain why the repertoires of imprinted genes vary among Therian mammals [109,110] (https://www.geneimprint.com/site/genes-by-species).

The parent-of-origin-dependent expression of imprinted genes is mediated by at least two different imprinting mechanisms. Briefly, the chromatin boundary imprinting model holds that allele-specific modifications at ICRs affect binding of insulator proteins (e.g. CTCF), thereby mediating gene silencing. Such is the case for the IGF2/H19 imprinted domain (Figure 4A) [111]. On the paternal allele, CTCF does not bind the methylated gICR, resulting in distinct maternal/paternal patterns of cohesin-mediated chromatin remodeling around H19. This results in maternal expression of H19 and the silencing of IGF2 isolated from the downstream enhancer. In contrast, the CTCF/cohesin interactions on the paternal allele bring the downstream enhancer into range of the IGF2 promoter, activating its expression [90].

In the noncoding RNA imprinting model, the production of mouse Airn is critical for the establishment of imprinting (Figure 4B) [92]. During early development of the embryo and placenta, paternal allele silencing only requires transcriptional interference of the Airn transcript originating from the unmethylated gICR and overlapping of the Igf2r promoter; however, in late development, the paternal allele is methylated at the sICR [112]. The paternal alleles of two additional genes, Slc22a2 and Slc22a3, are also paternally silenced, but in a more tissue-dependent manner than Igf2r. DNA methylation on the maternal allele of the gICR inhibits the production of Airn from the mother’s copy of the gene. Thus, the sICR on the maternal allele remains unmethylated, permitting expression of Igf2r, Slc22a2, and Slc22a3 from only the maternal allele. The adjacent gene, Slc22a1, escapes imprinting control and is biallelically expressed.

As in the mouse genome, the human IGF2R has a gICR in intron 2 (Figure 5, ICR_409 and ICR_410) [82], and according to GTEx Project data, AIRN is significantly expressed principally in the brain (https://gtexportal.org/home/gene/AIRN). Nevertheless, IGF2R is biallelically expressed in all human tissues investigated, except for sporadic monoallelic expression in the human placenta [113]. Interestingly, AIRN is not present in other mammals that show imprinted IGF2R expression, such as the dog [114] and opossum [115]. This suggests that other epigenetic modifications are involved in controlling imprinted expression at the IGF2R locus in these mammalian species [61]. Since the IGF2R is involved in carcinogenesis [116,117], human longevity [118], and cognitive ability [56], the biological function of genomic imprinting at the IGF2R locus in human growth, aging, and brain development needs to be more thoroughly investigated.

Imprinting and disease susceptibility

George Orwell wrote in Animal Farm [119], ‘All animals are equal, but some animals are more equal than others’. The same is true of genes when it comes to disease susceptibility, and those that are ‘more equal’ are the imprinted genes. The parental allele expressed can be chosen either randomly [120] or in a parental-specific manner, as observed in genomically imprinted genes [92,121]. Genomic imprinting may be evolutionarily adaptive because of its involvement in metabolism [78] and brain development [80,122], and its potential ability to accelerate mammalian speciation [108,123]. At the same time, the presence of functionally haploid imprinted genes in the human genome can be disastrous to the health of an individual.

A number of developmental disorders in humans, such as Beckwith–Wiedemann and Silver–Russell syndromes, result not only from genomic mutations, but also from epigenetic dysregulation of imprinted genes [124]. Interestingly, Silver–Russell syndrome, a congenital disease characterized by growth retardation, is the first human disorder shown to result from epigenetically mediated imprinting defects affecting two different chromosomes [125]. Ten percent of patients present with maternal UPD of chromosome 7, while 40% to 65% show hypomethylation at the gICR upstream of H19 at chromosome location 11p15.5 (Figure 4A), resulting in reduced IGF2 expression [124]. In contrast, hypermethylation of the H19 gICR, with concomitant biallelic expression of IGF2, is associated with the overgrowth disorder, Beckwith-Wiedemann syndrome [126]. These two developmental syndromes are mirror disorders resulting from loss and gain of methylation (i.e. LOI) at the same ICR. LOIs at other ICRs that result in the formation of mirrored pathological conditions need to be identified.

GWAS and CNV studies have identified genomic regions linked to complex disorders such as autism, bipolar disorder, schizophrenia, and Tourette’s syndrome with a parent-of-origin inheritance preference, also indicating the involvement of imprinted genes in their etiology [127]. Two imprinted genes implicated in the development of autism and schizophrenia are 1) DLGAP2, a membrane-associated guanylate kinase localized at postsynaptic density in neuronal cells [109,128]; and 2) MAGI2, a multi-PDZ domain scaffolding protein that interacts with several different ligands in the brain [109,129,130], respectively. Thus, the role of imprinted genes in the etiology of all behavioral disorders needs to be systematically investigated, particularly in autism since its rapid increase in incidence implies the involvement of environmentally-induced alterations in the epigenome [131].

Exposure to famine conditions while in utero both increases the risk of developing cardiovascular disease, obesity, and diabetes [132] and also doubles the incidence of schizophrenia [133,134]. Furthermore, individuals who were prenatally exposed to famine during the 1944–1945 Dutch Hunger Winter had less DNA methylation at the H19/IGF2 domain gICR six decades later (Figure 4A) [135]. These data suggest that a major reduction in nutrition during pregnancy – particularly during the first trimester – markedly increases psychosis formation by modifying ICRs. In contrast, the calorie-rich Western diet is predicted to potentially increase the prevalence of autism [81]. Thus, a better understanding of the role of nutrition [81] and environmental factors, like endocrine-disrupting agents [131], in the genesis of autism is needed.

Determining the role of imprinted genes in abnormal behavioral formation is challenging since brain-specific DNA methylation profiles cannot be measured in living individuals. Nevertheless, given the consistency across tissues of the parentally established epigenetic marks present in gICRs (Figure 5), they should also be detectable in peripheral tissues that are amenable to analysis. This postulate is supported by a recent study in mice where hypermethylation was observed in the ICR of Grb10 in both the blood and liver of Pb-exposed male animals [136]. Thus, even in the absence of tissue-specific DNA methylation profiles, screening affected individuals for epigenetic disruptions in imprint regulatory elements should prove to be highly informative for ascertaining the role of genomic imprinting in environmentally-induced psychiatric conditions and other chronic diseases.

Cathrine Hoyo established a NEST human cohort to investigate imprinting dysregulation and disease risk [137]. This is an ongoing prospective study of women and their children. It was designed to identify early exposures associated with stable epigenetic alterations in the gICRs of infants that may alter chronic disease susceptibility later in life. Between 2005 and 2011, more than 2000 pregnant women visiting prenatal clinics at Duke or Durham Regional Hospitals were enrolled. Children from these pregnancies are still being followed every two years to collect growth trajectories, disease diagnoses, and behavioral data.

These pioneering environmental epigenomic studies initially involved the measurement of gICR DNA methylation in peripheral blood lymphocytes for only a few known imprinted genes (i.e. IGF2/H19, PLAGL1, IGF2, MEST, PEG3, MEG3, and NNAT). They demonstrated that the methylation pattern of gICRs in children was significantly altered with maternal folic acid consumption [137], maternal and paternal obesity [138], maternal depression and antidepressant drug use [139,140], intrauterine infection [141], maternal antibiotic use [142], maternal smoking [143], and heavy metal exposure to Cd [144] and Pb [145]. Susan Murphy and her colleagues further showed that the DNA methylation of 3979 CpG sites in human sperm was associated with cannabis exposure, of which 19 were also deregulated in male tobacco smokers [146]. Of these genes, APC2 and RFPL2 were recently predicted to be imprinted [82]. They also showed, with an in vitro human spermatogenesis model, that chronic cannabis exposure altered DNA methylation of the known paternally expressed imprinted genes SGCE, GRB10, and PEG3, and a candidate imprinted autism gene, HCN1 [82,147].

Nevertheless, to enhance our understanding of human behavior and disease formation, it was important to define both the complete repertoire of human imprinted genes and their regulatory elements - the human imprintome [90,148]. Following the publication of the human genome in 2001 [149,150], we first used computer machine learning algorithms or AI approaches to predict the genome-wide imprint status of human genes from sequence features [109]. Of the 102 annotated genes identified to be potentially imprinted, we have now identified ICRs for 35 of them (34%) (Supplementary Table S4, [83[82]), providing additional evidence that they are indeed imprinted. Two of these computationally identified putative imprinted genes, DLGAP2 and KCNK9, were also demonstrated experimentally to be paternally and maternally expressed, respectively. DLGAP2 is a membrane-associated protein that plays a role in synapse organization and signaling in neuronal cells, and as previously stated, is implicated in the etiology of autism [128]. Since it is imprinted, it can potentially be altered both genetically and/or epigenetically in the pathogenesis of autism. Maternally expressed KCNK9 encodes for the pH-sensitive potassium channel protein, TASK3. TASK3 is present at the plasma membrane and regulates membrane depolarization in response to acidosis via inhibition of the background potassium current. Maternal germline inactivation of the maternally expressed allele of KCNK9 results in Birk–Barel syndrome [151], and KCNK9 loss of imprinting is also prevalent in triple-negative breast cancer [152].

We recently determined the human imprintome by performing WGBS of DNA derived from tissues arising from the three germ layers (i.e., brain, liver, and kidney) and from the egg and sperm [82]. The gICRs in somatic cells will be 50% methylated because the gametes are either methylated or unmethylated at the gICR locations (Figure 5). We identified 1,488 hemi-methylated candidate ICRs. Gamete methylation approached 0% or 100% in 332 gICRs (i.e., 178 paternally and 154 maternally methylated), supporting parent-of-origin-specific methylation. This first draft of the human imprintome (https://humanicr.org/) allows for a more systematic determination of the importance of imprinting dysregulation in the formation of human diseases and behavioral disorders.

We recently used the human imprintome coupled with WGBS to determine whether aberrant DNA methylation at ICRs is associated with Alzheimer's disease (AD) [153]. This study showed that 120 candidate ICRs varied significantly between AD cases and controls. The number of ICRs with altered methylation in individuals with AD is three times higher in non-Hispanic blacks (NHBs) than in non-Hispanic whites (NHWs), suggesting a possible reason for the higher prevalence of AD in NHBs than in NHWs [154]. Interestingly, only two ICRs are common to both NHBs and NHWs, and they are proximal to the inflammasome gene, NLRP1, and a known imprinted gene, MEST/MESTIT1. These findings indicate, for the first time, that early developmental alterations in DNA methylation of regions regulating genomic imprinting may contribute to AD risk, and that this epigenetic risk differs between NHBs and NHWs.

WGBS is the most precise method to interrogate gICRs for DNA methylation changes in environmental epigenomic studies; however, it is expensive, requires high coverage, and it is computationally intensive. To address this deficiency, a custom methylation array containing 22,819 probes was developed in collaboration with TruDiagnostics (Lexington, KY) (Figure 6A) [83]. It contains 9,757 probes mapping to 1,088 out of the 1,488 candidate ICRs in the human imprintome (Figure 6B).

Infinium DNA methylation array for the human imprintome.

Figure 6:
Infinium DNA methylation array for the human imprintome.

(A) Human imprintome array for the DNA methylation ICR analysis of 24 samples. (B) Coverage of the 1488 ICRs in the human imprintome is shown for WGBS (100% ICR coverage), the human imprintome array (73% ICR coverage), EPICv1 array (10% ICR coverage) (Illumina, Inc. San Diego, CA), and EPICv2 array ((36% ICR coverage) (Illumina, Inc. San Diego, CA) [82,83]). ICR, imprint control region.

Figure 6:
Infinium DNA methylation array for the human imprintome.

(A) Human imprintome array for the DNA methylation ICR analysis of 24 samples. (B) Coverage of the 1488 ICRs in the human imprintome is shown for WGBS (100% ICR coverage), the human imprintome array (73% ICR coverage), EPICv1 array (10% ICR coverage) (Illumina, Inc. San Diego, CA), and EPICv2 array ((36% ICR coverage) (Illumina, Inc. San Diego, CA) [82,83]). ICR, imprint control region.

Close modal

This custom array should help accelerate the rapid screening for gICRs associated with a wide range of chronic diseases and exposures, thereby advancing our understanding of genomic imprinting and its relevance in development and disease formation throughout the life course.

Traditional research to determine the role of gene-environment interactions in disease risk examines the relationship among disease susceptibility, environmental exposures, and germline mutations. Such research efforts have highlighted the importance of genotype in human diseases; however, it has become clear that full understanding of the mechanisms by which disease risk is altered by environmental exposures will require epigenetic mechanisms to also be taken into account.

Summary

  • Two epigenetically regulated subsets of genes that link environmental exposures early in development to adult diseases are imprinted genes and those with metastable epialleles. The Avy mouse metastable epiallele demonstrated, for the first time, that early developmental exposures to nutritional (i.e., methyl donors and genistein), chemical (i.e., BPA), and physical agents (i.e., LDIR) alter disease susceptibility in adulthood by modifying DNA methylation. Thus, human health and disease stem not only from genetic mutations but also from changes in the epigenome.

  • Genomic imprinting evolved about 150 million years ago in a common ancestor to Therians, resulting in the formation of disease susceptibility loci since only a single mutation or epigenetic event is needed to cause loss of gene function. Consequently, deregulation of imprinted gene expression is involved in a variety of chronic human diseases and behavioral disorders.

  • The level of methylation at gICRs in the human imprintome [82] and metastable epialleles (i.e. CoRSIVs) [24] is independent of tissue type, making them ideal for DOHaD epidemiology studies where the tissue of interest cannot be obtained. The human imprintome and metastable epiallele control regions in humans have been identified [24,82]. Although a human imprintome array is available to facilitate the determination of gICRs deregulated in chronic diseases and behavioral disorders when exposed to environmental factors [83], a similar array for CoRSIVs is presently unavailable [155]. This deficiency needs to be corrected in order to more systematically determine the role of these two subsets of epigenetically regulated genes in human health and disease and risk to LDIR.

The author declares there are no competing interests associated with the manuscript.

This work was supported in part by National Institutes of Health grants R01HD098857, R01MD011746, and R01MD011746-S1.

R.L.J.: Writing—original draft.

AD

Alzheimer's disease

AI

artificial intelligence

Avy

Agouti viable yellow

BPA

Bisphenol A

CT

computed tomography

CTCF

CCCTC-binding factor

CTCFL

CCCTC-binding factor like

CoRSIV

correlated regions of systemic interindividual variation

DEN

diethylnitrosamine

DNMT1

DNA methyltransferase 1

DNMT3A

DNA methyltransferase 3 alpha

DNMT3B

DNA methyltransferase 3 beta

DOHaD

Developmental origins of health and disease

DPPA3

Developmental pluripotency-associated 3

EMB

embryo

GRB10

growth factor receptor bound protein 10

HCN1

hyperpolarization activated cyclic nucleotide gated potassium channel 1

HDACs

histone deacetylases

IAP

intracisternal A-particle

ICR

imprint control region

LDIR

Low dose ionizing radiation

MC4R

Melanocortin 4 receptor

MYA

million years ago

NHBs

non-Hispanic blacks

NHWs

non-Hispanic whites

PEG3

paternally expressed gene 3

PGCs

primordial germ cells

PS1A

Pseudoexon 1A

SGCE

Sarcoglycan epsilon

TASK3

Tandem pore domain acid-sensitive K+ channel 3

TB

Trophoblasts

TNBC

Triple negative breast cancer

Tme

T-maternal effect

WGBS

whole genome bisulfite sequencing

WT

wildtype

YS

Yolk sac

gICR

Germline imprint control region

sICR

somatic imprint control region

1
Waddington
,
C.H
. (
1940
)
Organisers and Genes
,
Cambridge, UK
,
Cambridge University Press
2
Holliday
,
R.
and
Pugh
,
J.E
. (
1975
)
DNA modification mechanisms and gene activity during development
.
Science
187
,
226
232
3
Wolffe
,
A.P.
and
Matzke
,
M.A
. (
1999
)
Epigenetics: regulation through repression
.
Science
286
,
481
486
https://doi.org/10.1126/science.286.5439.481
4
Barker
,
D.J.
and
Osmond
,
C
. (
1986
)
Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales
.
Lancet
1
,
1077
1081
https://doi.org/10.1016/s0140-6736(86)91340-1
5
Barker
,
D.J.
. (
1991
)
The intrauterine origins of cardiovascular and obstructive lung disease in adult life. the marc daniels lecture 1990
.
J. R. Coll. Physicians Lond.
25
,
129
133
https://doi.org/10.1016/S0035-8819(25)02001-X
6
Lumey
,
L.H.
,
Stein
,
A.D.
and
Susser
,
E
. (
2011
)
Prenatal famine and adult health
.
Annu. Rev. Public Health
32
,
237
262
https://doi.org/10.1146/annurev-publhealth-031210-101230
7
Susser
,
E.
and
St Clair
,
D
. (
2013
)
Prenatal famine and adult mental illness: interpreting concordant and discordant results from the Dutch and Chinese Famines
.
Soc. Sci. Med.
97
,
325
330
https://doi.org/10.1016/j.socscimed.2013.02.049
8
Torreggiani
,
M.
,
Fois
,
A.
,
Santagati
,
G.
,
De Marco
,
O.
,
Bedogni
,
S.
,
Cacciatori
,
N.
et al.
(
2025
)
Severe maternal undernutrition during pregnancy and its long-term effects on the offspring health, with a focus on kidney health
.
Pediatr. Nephrol.
40
,
1853
1862
https://doi.org/10.1007/s00467-024-06552-w
9
Pembrey
,
M.
,
Saffery
,
R.
,
Bygren
,
L.O.
,
Epigenetic
,
E.
and
Epigenetic
,
E
. (
2014
)
Human transgenerational responses to early-life experience: potential impact on development, health and biomedical research
.
J. Med. Genet.
51
,
563
572
https://doi.org/10.1136/jmedgenet-2014-102577
10
Jawaid
,
A.
,
Jehle
,
K.L.
and
Mansuy
,
I.M
. (
2021
)
Impact of parental exposure on offspring health in humans
.
Trends Genet.
37
,
373
388
https://doi.org/10.1016/j.tig.2020.10.006
11
Wadhwa
,
P.D.
,
Buss
,
C.
,
Entringer
,
S.
and
Swanson
,
J.M
. (
2009
)
Developmental origins of health and disease: brief history of the approach and current focus on epigenetic mechanisms
.
Semin. Reprod. Med.
27
,
358
368
https://doi.org/10.1055/s-0029-1237424
12
Waterland
,
R.A.
and
Jirtle
,
R.L
. (
2003
)
Transposable elements: targets for early nutritional effects on epigenetic gene regulation
.
Mol. Cell. Biol.
23
,
5293
5300
https://doi.org/10.1128/MCB.23.15.5293-5300.2003
13
Bianco-Miotto
,
T.
,
Craig
,
J.M.
,
Gasser
,
Y.P.
,
van Dijk
,
S.J.
and
Ozanne
,
S.E
. (
2017
)
Epigenetics and DOHaD: from basics to birth and beyond
.
J. Dev. Orig. Health Dis.
8
,
513
519
https://doi.org/10.1017/S2040174417000733
14
Jirtle
,
R.L.
and
Skinner
,
M.K
. (
2007
)
Environmental epigenomics and disease susceptibility
.
Nat. Rev. Genet.
8
,
253
262
https://doi.org/10.1038/nrg2045
15
Rugo
,
R.E.
,
Mutamba
,
J.T.
,
Mohan
,
K.N.
,
Yee
,
T.
,
Chaillet
,
J.R.
,
Greenberger
,
J.S.
et al.
(
2011
)
Methyltransferases mediate cell memory of a genotoxic insult
.
Oncogene
30
,
751
756
https://doi.org/10.1038/onc.2010.480
16
Gaudet
,
F.
,
Rideout
,
W.M.
3rd
,
Meissner
,
A.
,
Dausman
,
J.
,
Leonhardt
,
H.
and
Jaenisch
,
R
. (
2004
)
Dnmt1 expression in pre- and postimplantation embryogenesis and the maintenance of IAP silencing
.
Mol. Cell. Biol.
24
,
1640
1648
https://doi.org/10.1128/MCB.24.4.1640-1648.2004
17
Duhl
,
D.M.
,
Vrieling
,
H.
,
Miller
,
K.A.
,
Wolff
,
G.L.
and
Barsh
,
G.S
. (
1994
)
Neomorphic agouti mutations in obese yellow mice
.
Nat. Genet.
8
,
59
65
https://doi.org/10.1038/ng0994-59
18
Vasicek
,
T.J.
,
Zeng
,
L.
,
Guan
,
X.J.
,
Zhang
,
T.
,
Costantini
,
F.
and
Tilghman
,
S.M
. (
1997
)
Two dominant mutations in the mouse fused gene are the result of transposon insertions
.
Genetics
147
,
777
786
https://doi.org/10.1093/genetics/147.2.777
19
Druker
,
R.
,
Bruxner
,
T.J.
,
Lehrbach
,
N.J.
and
Whitelaw
,
E
. (
2004
)
Complex patterns of transcription at the insertion site of a retrotransposon in the mouse
.
Nucleic Acids Res.
32
,
5800
5808
https://doi.org/10.1093/nar/gkh914
20
Weinhouse
,
C.
,
Anderson
,
O.S.
,
Jones
,
T.R.
,
Kim
,
J.
,
Liberman
,
S.A.
,
Nahar
,
M.S.
et al.
(
2011
)
An expression microarray approach for the identification of metastable epialleles in the mouse genome
.
Epigenetics
6
,
1105
1113
https://doi.org/10.4161/epi.6.9.17103
21
Chang
,
W.J.
,
Baker
,
M.S.
,
Laritsky
,
E.
,
Gunasekara
,
C.J.
,
Maduranga
,
U.
,
Galliou
,
J.C.
et al.
(
2024
)
Systemic interindividual DNA methylation variants in cattle share major hallmarks with those in humans
.
Genome Biol.
25
, 185 https://doi.org/10.1186/s13059-024-03307-6
22
Waterland
,
R.A.
,
Kellermayer
,
R.
,
Laritsky
,
E.
,
Rayco-Solon
,
P.
,
Harris
,
R.A.
,
Travisano
,
M.
et al.
(
2010
)
Season of conception in rural gambia affects DNA methylation at putative human metastable epialleles
.
Plos Genet.
6
, e1001252 https://doi.org/10.1371/journal.pgen.1001252
23
Dominguez-Salas
,
P.
,
Moore
,
S.E.
,
Baker
,
M.S.
,
Bergen
,
A.W.
,
Cox
,
S.E.
,
Dyer
,
R.A.
et al.
(
2014
)
Maternal nutrition at conception modulates DNA methylation of human metastable epialleles
.
Nat. Commun.
5
, 3746 https://doi.org/10.1038/ncomms4746
24
Gunasekara
,
C.J.
,
Scott
,
C.A.
,
Laritsky
,
E.
,
Baker
,
M.S.
,
MacKay
,
H.
,
Duryea
,
J.D.
et al.
(
2019
)
A genomic atlas of systemic interindividual epigenetic variation in humans
.
Genome Biol.
20
, 105 https://doi.org/10.1186/s13059-019-1708-1
25
Dolinoy
,
D.C.
,
Weidman
,
J.R.
,
Waterland
,
R.A.
and
Jirtle
,
R.L
. (
2006
)
Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome
.
Environ. Health Perspect.
114
,
567
572
https://doi.org/10.1289/ehp.8700
26
Miltenberger
,
R.J.
,
Mynatt
,
R.L.
,
Wilkinson
,
J.E.
and
Woychik
,
R.P
. (
1997
)
The role of the agouti gene in the yellow obese syndrome
.
J. Nutr.
127
,
1902S
1907S
https://doi.org/10.1093/jn/127.9.1902S
27
Morgan
,
H.D.
,
Sutherland
,
H.G.
,
Martin
,
D.I.
and
Whitelaw
,
E
. (
1999
)
Epigenetic inheritance at the agouti locus in the mouse
.
Nat. Genet.
23
,
314
318
https://doi.org/10.1038/15490
28
Yen
,
T.T.
,
Gill
,
A.M.
,
Frigeri
,
L.G.
,
Barsh
,
G.S.
and
Wolff
,
G.L
. (
1994
)
Obesity, diabetes, and neoplasia in yellow A(vy)/- mice: ectopic expression of the agouti gene
.
FASEB J.
8
,
479
488
https://doi.org/10.1096/fasebj.8.8.8181666
29
Williams
,
G.
,
Harrold
,
J.A.
and
Cutler
,
D.J
. (
2000
)
The hypothalamus and the regulation of energy homeostasis: lifting the lid on a black box
.
Proc. Nutr. Soc.
59
,
385
396
https://doi.org/10.1017/s0029665100000434
30
Dolinoy
,
D.C.
,
Das
,
R.
,
Weidman
,
J.R.
and
Jirtle
,
R.L
. (
2007
)
Metastable epialleles, imprinting, and the fetal origins of adult diseases
.
Pediatr. Res.
61
,
30R
37R
https://doi.org/10.1203/pdr.0b013e31804575f7
31
Maffini
,
M.V.
,
Rubin
,
B.S.
,
Sonnenschein
,
C.
and
Soto
,
A.M
. (
2006
)
Endocrine disruptors and reproductive health: the case of bisphenol-A
.
Mol. Cell. Endocrinol.
254–255
,
179
186
https://doi.org/10.1016/j.mce.2006.04.033
32
Wu
,
S.
,
Wei
,
X.
,
Jiang
,
J.
,
Shang
,
L.
and
Hao
,
W
. (
2012
)
Effects of bisphenol a on the proliferation and cell cycle of HBL-100 cells
.
Food Chem. Toxicol.
50
,
3100
3105
https://doi.org/10.1016/j.fct.2012.06.029
33
Dolinoy
,
D.C.
,
Huang
,
D.
and
Jirtle
,
R.L
. (
2007
)
Maternal nutrient supplementation counteracts bisphenol a-induced DNA hypomethylation in early development
.
Proc. Natl. Acad. Sci. U.S.A.
104
,
13056
13061
https://doi.org/10.1073/pnas.0703739104
34
Bernal
,
A.J.
,
Dolinoy
,
D.C.
,
Huang
,
D.
,
Skaar
,
D.A.
,
Weinhouse
,
C.
and
Jirtle
,
R.L
. (
2013
)
Adaptive radiation-induced epigenetic alterations mitigated by antioxidants
.
FASEB J.
27
,
665
671
https://doi.org/10.1096/fj.12-220350
35
Calabrese
,
E.J.
and
Baldwin
,
L.A
. (
2003
)
Hormesis: the dose-response revolution
.
Annu. Rev. Pharmacol. Toxicol.
43
,
175
197
https://doi.org/10.1146/annurev.pharmtox.43.100901.140223
36
Slatko
,
B.E.
,
Gardner
,
A.F.
and
Ausubel
,
F.M
. (
2018
)
Overview of next-generation sequencing technologies
.
Curr. Protoc. Mol. Biol.
122
, e59 https://doi.org/10.1002/cpmb.59
37
Kolmogorov
,
M.
,
Billingsley
,
K.J.
,
Mastoras
,
M.
,
Meredith
,
M.
,
Monlong
,
J.
,
Lorig-Roach
,
R.
et al.
(
2023
)
Scalable nanopore sequencing of human genomes provides a comprehensive view of haplotype-resolved variation and methylation
.
Nat. Methods
20
,
1483
1492
https://doi.org/10.1038/s41592-023-01993-x
38
Cameron
,
J.R.
and
Moulder
,
J.E
. (
1998
)
Proposition: radiation hormesis should be elevated to a position of scientific respectability
.
Med. Phys. Mex. Symp. Med. Phys.
25
,
1407
1410
https://doi.org/10.1118/1.598312
39
Cameron
,
J.R
. (
2005
)
Moderate dose rate ionizing radiation increases longevity
.
Br. J. Radiol.
78
,
11
13
https://doi.org/10.1259/bjr/62063624
40
Iavicoli
,
I.
,
Fontana
,
L.
,
Santocono
,
C.
,
Guarino
,
D.
,
Laudiero
,
M.
and
Calabrese
,
E.J
. (
2023
)
The challenges of defining hormesis in epidemiological studies: the case of radiation hormesis
.
Sci. Total Environ.
902
, 166030 https://doi.org/10.1016/j.scitotenv.2023.166030
41
Hauptmann
,
M.
,
Byrnes
,
G.
,
Cardis
,
E.
,
Bernier
,
M.O.
,
Blettner
,
M.
,
Dabin
,
J.
et al.
(
2023
)
Brain cancer after radiation exposure from CT examinations of children and young adults: results from the EPI-CT cohort study
.
Lancet Oncol.
24
,
45
53
https://doi.org/10.1016/S1470-2045(22)00655-6
42
Brenner
,
D.J.
and
Hall
,
E.J
. (
2012
)
Cancer risks from CT scans: now we have data, what next?
Radiology
265
,
330
331
https://doi.org/10.1148/radiol.12121248
43
Smith-Bindman
,
R.
,
Chu
,
P.W.
,
Azman Firdaus
,
H.
,
Stewart
,
C.
,
Malekhedayat
,
M.
,
Alber
,
S.
et al.
(
2025
)
Projected lifetime cancer risks from current computed tomography imaging
.
JAMA Intern. Med.
185
,
710
719
https://doi.org/10.1001/jamainternmed.2025.0505
44
Sudheer
,
K.R.
,
Mohammad Koya
,
P.K.
,
Prakash
,
A.J.
,
Prakash
,
A.M.
,
Manoj Kumar
,
R.
,
Shyni
,
S.
et al.
(
2022
)
Evaluation of risk due to chronic low dose ionizing radiation exposure on the birth prevalence of congenital heart diseases (CHD) among the newborns from high-level natural radiation areas of Kerala coast, India
.
Genes Environ.
44
, 1 https://doi.org/10.1186/s41021-021-00231-0
45
Saenko
,
V.
and
Mitsutake
,
N
. (
2024
)
Radiation-related thyroid cancer
.
Endocr. Rev.
45
,
1
29
https://doi.org/10.1210/endrev/bnad022
46
Luckey
,
T.D
. (
2006
)
Radiation hormesis: the good, the bad, and the ugly
.
Dose Response
4
,
169
190
https://doi.org/10.2203/dose-response.06-102.Luckey
47
Barton
,
S.C.
,
Surani
,
M.A.H.
and
Norris
,
M.L
. (
1984
)
Role of paternal and maternal genomes in mouse development
.
Nature
311
,
374
376
https://doi.org/10.1038/311374a0
48
Surani
,
M.A.H.
,
Barton
,
S.C.
and
Norris
,
M.L
. (
1984
)
Development of reconstituted mouse eggs suggests imprinting of the genome during gametogenesis
.
Nature
308
,
548
550
https://doi.org/10.1038/308548a0
49
McGrath
,
J.
and
Solter
,
D
. (
1984
)
Completion of mouse embryogenesis requires both the maternal and paternal genomes
.
Cell
37
,
179
183
https://doi.org/10.1016/0092-8674(84)90313-1
50
Ramachandran
,
R.
and
McDaniel
,
C.D
. (
2018
)
Parthenogenesis in birds: a review
.
Reproduction
155
,
R245
R257
https://doi.org/10.1530/REP-17-0728
51
Fujita
,
M.K.
,
Singhal
,
S.
,
Brunes
,
T.O.
and
Maldonado
,
J.A
. (
2020
)
Evolutionary dynamics and consequences of parthenogenesis in vertebrates
.
Annu. Rev. Ecol. Evol. Syst.
51
,
191
214
https://doi.org/10.1146/annurev-ecolsys-011720-114900
52
Kawahara
,
M.
,
Wu
,
Q.
,
Takahashi
,
N.
,
Morita
,
S.
,
Yamada
,
K.
,
Ito
,
M.
et al.
(
2007
)
High-frequency generation of viable mice from engineered bi-maternal embryos
.
Nat. Biotechnol.
25
,
1045
1050
https://doi.org/10.1038/nbt1331
53
Johnson
,
D.R
. (
1974
)
Hairpin-tail: a case of post-reductional gene action in the mouse egg
.
Genetics
76
,
795
805
https://doi.org/10.1093/genetics/76.4.795
54
Barlow
,
D.P.
,
Stöger
,
R.
,
Herrmann
,
B.G.
,
Saito
,
K.
and
Schweifer
,
N
. (
1991
)
The mouse insulin-like growth factor type-2 receptor is imprinted and closely linked to the Tme locus
.
Nature
349
,
84
87
https://doi.org/10.1038/349084a0
55
Ghosh
,
P.
,
Dahms
,
N.M.
and
Kornfeld
,
S
. (
2003
)
Mannose 6-phosphate receptors: new twists in the tale
.
Nat. Rev. Mol. Cell Biol.
4
,
202
212
https://doi.org/10.1038/nrm1050
56
Chen
,
D.Y.
,
Stern
,
S.A.
,
Garcia-Osta
,
A.
,
Saunier-Rebori
,
B.
,
Pollonini
,
G.
,
Bambah-Mukku
,
D.
et al.
(
2011
)
A critical role for IGF-II in memory consolidation and enhancement
.
Nature
469
,
491
497
https://doi.org/10.1038/nature09667
57
DeChiara
,
T.M.
,
Robertson
,
E.J.
and
Efstratiadis
,
A
. (
1991
)
Parental imprinting of the mouse insulin-like growth factor II gene
.
Cell
64
,
849
859
https://doi.org/10.1016/0092-8674(91)90513-x
58
Zhang
,
Y.
and
Tycko
,
B
. (
1992
)
Monoallelic expression of the human H19 gene
.
Nat. Genet.
1
,
40
44
https://doi.org/10.1038/ng0492-40
59
Ohlsson
,
R.
,
Nyström
,
A.
,
Pfeifer-Ohlsson
,
S.
,
Töhönen
,
V.
,
Hedborg
,
F.
,
Schofield
,
P.
et al.
(
1993
)
IGF2 is parentally imprinted during human embryogenesis and in the beckwith-wiedemann syndrome
.
Nat. Genet.
4
,
94
97
https://doi.org/10.1038/ng0593-94
60
Das
,
R.
,
Hampton
,
D.D.
and
Jirtle
,
R.L
. (
2009
)
Imprinting evolution and human health
.
Mamm. Genome
20
,
563
572
https://doi.org/10.1007/s00335-009-9229-y
61
Das
,
R.
,
Anderson
,
N.
,
Koran
,
M.I.
,
Weidman
,
J.R.
,
Mikkelsen
,
T.S.
,
Kamal
,
M.
et al.
(
2012
)
Convergent and divergent evolution of genomic imprinting in the marsupial monodelphis domestica
.
BMC Genomics
13
, 394 https://doi.org/10.1186/1471-2164-13-394
62
Kaneko-Ishino
,
T.
and
Ishino
,
F
. (
2022
)
The evolutionary advantage in mammals of the complementary monoallelic expression mechanism of genomic imprinting and its emergence from a defense against the insertion into the host genome
.
Front. Genet.
13
, 832983 https://doi.org/10.3389/fgene.2022.832983
63
Newman
,
T.
,
Bond
,
D.M.
,
Ishihara
,
T.
,
Rizzoli
,
P.
,
Gouil
,
Q.
,
Hore
,
T.A.
et al.
(
2024
)
PRKACB is a novel imprinted gene in marsupials
.
Epigenetics Chromatin
17
, 29 https://doi.org/10.1186/s13072-024-00552-8
64
Evans
,
H.K.
,
Weidman
,
J.R.
,
Cowley
,
D.O.
and
Jirtle
,
R.L
. (
2005
)
Comparative phylogenetic analysis of blcap/nnat reveals eutherian-specific imprinted gene
.
Mol. Biol. Evol.
22
,
1740
1748
https://doi.org/10.1093/molbev/msi165
65
Killian
,
J.K.
,
Nolan
,
C.M.
,
Stewart
,
N.
,
Munday
,
B.L.
,
Andersen
,
N.A.
,
Nicol
,
S.
et al.
(
2001
)
Monotreme IGF2 expression and ancestral origin of genomic imprinting
.
J. Exp. Zool.
291
,
205
212
https://doi.org/10.1002/jez.1070
66
Renfree
,
M.B.
,
Hore
,
T.A.
,
Shaw
,
G.
,
Graves
,
J.A.M.
and
Pask
,
A.J
. (
2009
)
Evolution of genomic imprinting: insights from marsupials and monotremes
.
Annu. Rev. Genomics Hum. Genet.
10
,
241
262
https://doi.org/10.1146/annurev-genom-082908-150026
67
Nolan
,
C.M.
,
Killian
,
J.K.
,
Petitte
,
J.N.
and
Jirtle
,
R.L
. (
2001
)
Imprint status of M6P/IGF2R and IGF2 in chickens
.
Dev. Genes Evol.
211
,
179
183
https://doi.org/10.1007/s004270000132
68
Frésard
,
L.
,
Leroux
,
S.
,
Servin
,
B.
,
Gourichon
,
D.
,
Dehais
,
P.
,
Cristobal
,
M.S.
et al.
(
2014
)
Transcriptome-wide investigation of genomic imprinting in chicken
.
Nucleic Acids Res.
42
,
3768
3782
https://doi.org/10.1093/nar/gkt1390
69
Killian
,
J.K.
,
Byrd
,
J.C.
,
Jirtle
,
J.V.
,
Munday
,
B.L.
,
Stoskopf
,
M.K.
,
MacDonald
,
R.G.
et al.
(
2000
)
M6P/IGF2R imprinting evolution in mammals
.
Mol. Cell
5
,
707
716
https://doi.org/10.1016/s1097-2765(00)80249-x
70
O’Neill
,
M.J.
,
Ingram
,
R.S.
,
Vrana
,
P.B.
and
Tilghman
,
S.M
. (
2000
)
Allelic expression of IGF2 in marsupials and birds
.
Dev. Genes Evol.
210
,
18
20
https://doi.org/10.1007/pl00008182
71
Khosla
,
S.
,
Mendiratta
,
G.
and
Brahmachari
,
V
. (
2006
)
Genomic imprinting in the mealybugs
.
Cytogenet. Genome Res.
113
,
41
52
https://doi.org/10.1159/000090814
72
Scott
,
R.J.
and
Spielman
,
M
. (
2006
)
Genomic imprinting in plants and mammals: how life history constrains convergence
.
Cytogenet. Genome Res.
113
,
53
67
https://doi.org/10.1159/000090815
73
Schuff
,
M.
,
Strong
,
A.D.
,
Welborn
,
L.K.
and
Ziermann-Canabarro
,
J.M
. (
2024
)
Imprinting as basis for complex evolutionary novelties in eutherians
.
Biology (Basel)
13
https://doi.org/10.3390/biology13090682
74
Spencer
,
H.G.
and
Clark
,
A.G
. (
2014
)
Non-conflict theories for the evolution of genomic imprinting
.
Heredity (Edinb)
113
,
112
118
https://doi.org/10.1038/hdy.2013.129
75
Haig
,
D.
and
Graham
,
C
. (
1991
)
Genomic imprinting and the strange case of the insulin-like growth factor II receptor
.
Cell
64
,
1045
1046
https://doi.org/10.1016/0092-8674(91)90256-x
76
Moore
,
T.
and
Haig
,
D
. (
1991
)
Genomic imprinting in mammalian development: a parental tug-of-war
.
Trends Genet.
7
,
45
49
https://doi.org/10.1016/0168-9525(91)90230-N
77
Hanin
,
G.
and
Ferguson-Smith
,
A.C
. (
2020
)
The evolution of genomic imprinting: epigenetic control of mammary gland development and postnatal resource control
.
Wiley Interdiscip. Rev. Syst. Biol. Med.
12
, e1476 https://doi.org/10.1002/wsbm.1476
78
Tucci
,
V.
,
Isles
,
A.R.
,
Kelsey
,
G.
,
Ferguson-Smith
,
A.C.
and
Erice Imprinting
,
G
. (
2019
)
Genomic imprinting and physiological processes in mammals
.
Cell
176
,
952
965
https://doi.org/10.1016/j.cell.2019.01.043
79
Keverne
,
E.B.
,
Fundele
,
R.
,
Narasimha
,
M.
,
Barton
,
S.C.
and
Surani
,
M.A
. (
1996
)
Genomic imprinting and the differential roles of parental genomes in brain development
.
Brain Res. Dev. Brain Res.
92
,
91
100
https://doi.org/10.1016/0165-3806(95)00209-x
80
Badcock
,
C.
and
Crespi
,
B
. (
2008
)
Battle of the sexes may set the brain
.
Nature
454
,
1054
1055
https://doi.org/10.1038/4541054a
81
Badcock
,
C
. (
2011
)
The imprinted brain: how genes set the balance between autism and psychosis
.
Epigenomics
3
,
345
359
https://doi.org/10.2217/epi.11.19
82
Jima
,
D.D.
,
Skaar
,
D.A.
,
Planchart
,
A.
,
Motsinger-Reif
,
A.
,
Cevik
,
S.E.
,
Park
,
S.S.
et al.
(
2022
)
Genomic map of candidate human imprint control regions: the imprintome
.
Epigenetics
17
,
1920
1943
https://doi.org/10.1080/15592294.2022.2091815
83
Carreras-Gallo
,
N.
,
Dwaraka
,
V.B.
,
Jima
,
D.D.
,
Skaar
,
D.A.
,
Mendez
,
T.L.
,
Planchart
,
A.
et al.
(
2024
)
Creation and validation of the first infinium DNA methylation array for the human imprintome
.
Epigenetics Commun.
4
,
5
https://doi.org/10.1186/s43682-024-00028-6
84
Haig
,
D
. (
2004
)
Genomic imprinting and kinship: how good is the evidence?
Annu. Rev. Genet.
38
,
553
585
https://doi.org/10.1146/annurev.genet.37.110801.142741
85
Madon-Simon
,
M.
,
Cowley
,
M.
,
Garfield
,
A.S.
,
Moorwood
,
K.
,
Bauer
,
S.R.
and
Ward
,
A
. (
2014
)
Antagonistic roles in fetal development and adult physiology for the oppositely imprinted Grb10 and Dlk1 genes
.
BMC Biol.
12
, 771 https://doi.org/10.1186/s12915-014-0099-8
86
Brahmachari
,
V.
,
Kohli
,
S.
and
Gulati
,
P
. (
2018
)
In praise of mealybugs
.
J. Genet.
97
,
379
389
87
Bauer
,
M.J.
and
Fischer
,
R.L
. (
2011
)
Genome demethylation and imprinting in the endosperm
.
Curr. Opin. Plant Biol.
14
,
162
167
https://doi.org/10.1016/j.pbi.2011.02.006
88
Kobayashi
,
H
. (
2021
)
Canonical and non-canonical genomic imprinting in rodents
.
Front. Cell Dev. Biol.
9
, 713878 https://doi.org/10.3389/fcell.2021.713878
89
Inoue
,
A
. (
2023
)
Noncanonical imprinting: intergenerational epigenetic inheritance mediated by polycomb complexes
.
Curr. Opin. Genet. Dev.
78
, 102015 https://doi.org/10.1016/j.gde.2022.102015
90
Skaar
,
D.A.
,
Li
,
Y.
,
Bernal
,
A.J.
,
Hoyo
,
C.
,
Murphy
,
S.K.
and
Jirtle
,
R.L
. (
2012
)
The human imprintome: regulatory mechanisms, methods of ascertainment, and roles in disease susceptibility
.
ILAR J.
53
,
341
358
https://doi.org/10.1093/ilar.53.3-4.341
91
Murphy
,
S.K.
and
Jirtle
,
R.L
. (
2003
)
Imprinting evolution and the price of silence
.
Bioessays
25
,
577
588
https://doi.org/10.1002/bies.10277
92
Barlow
,
D.P
. (
2011
)
Genomic imprinting: a mammalian epigenetic discovery model
.
Annu. Rev. Genet.
45
,
379
403
https://doi.org/10.1146/annurev-genet-110410-132459
93
Hermann
,
A.
,
Gowher
,
H.
and
Jeltsch
,
A
. (
2004
)
Biochemistry and biology of mammalian DNA methyltransferases
.
Cell. Mol. Life Sci.
61
,
2571
2587
https://doi.org/10.1007/s00018-004-4201-1
94
Turek-Plewa
,
J.
and
Jagodziński
,
P.P
. (
2005
)
The role of mammalian DNA methyltransferases in the regulation of gene expression
.
Cell. Mol. Biol. Lett.
10
,
631
647
95
Jia
,
D.
,
Jurkowska
,
R.Z.
,
Zhang
,
X.
,
Jeltsch
,
A.
and
Cheng
,
X
. (
2007
)
Structure of Dnmt3a bound to Dnmt3L suggests a model for de novo DNA methylation
.
Nature New Biol.
449
,
248
251
https://doi.org/10.1038/nature06146
96
Klenova
,
E.M.
,
Morse
,
H.C.
III
,
Ohlsson
,
R.
and
Lobanenkov
,
V.V
. (
2002
)
The novel BORIS + CTCF gene family is uniquely involved in the epigenetics of normal biology and cancer
.
Semin. Cancer Biol.
12
,
399
414
https://doi.org/10.1016/S1044-579X(02)00060-3
97
Lewis
,
A.
and
Reik
,
W
. (
2006
)
How imprinting centres work
.
Cytogenet. Genome Res.
113
,
81
89
https://doi.org/10.1159/000090818
98
Reik
,
W.
and
Walter
,
J
. (
2001
)
Genomic imprinting: parental influence on the genome
.
Nat. Rev. Genet.
2
,
21
32
https://doi.org/10.1038/35047554
99
Voon
,
H.P.J.
and
Gibbons
,
R.J
. (
2016
)
Maintaining memory of silencing at imprinted differentially methylated regions
.
Cell. Mol. Life Sci.
73
,
1871
1879
https://doi.org/10.1007/s00018-016-2157-6
100
Juan
,
A.M.
and
Bartolomei
,
M.S
. (
2019
)
Evolving imprinting control regions: KRAB zinc fingers hold the key
.
Genes Dev.
33
,
1
3
https://doi.org/10.1101/gad.322990.118
101
Denomme
,
M.M.
and
Mann
,
M.R.W
. (
2013
)
Maternal control of genomic imprint maintenance
.
Reprod. Biomed. Online
27
,
629
636
https://doi.org/10.1016/j.rbmo.2013.06.004
102
Takahashi
,
N.
,
Coluccio
,
A.
,
Thorball
,
C.W.
,
Planet
,
E.
,
Shi
,
H.
,
Offner
,
S.
et al.
(
2019
)
ZNF445 is a primary regulator of genomic imprinting
.
Genes Dev.
33
,
49
54
https://doi.org/10.1101/gad.320069.118
103
Mackay
,
D.J.G.
,
Callaway
,
J.L.A.
,
Marks
,
S.M.
,
White
,
H.E.
,
Acerini
,
C.L.
,
Boonen
,
S.E.
et al.
(
2008
)
Hypomethylation of multiple imprinted loci in individuals with transient neonatal diabetes is associated with mutations in ZFP57
.
Nat. Genet.
40
,
949
951
https://doi.org/10.1038/ng.187
104
Stöger
,
R.
,
Kubicka
,
P.
,
Liu
,
C.G.
,
Kafri
,
T.
,
Razin
,
A.
,
Cedar
,
H.
et al.
(
1993
)
Maternal-specific methylation of the imprinted mouse Igf2r locus identifies the expressed locus as carrying the imprinting signal
.
Cell
73
,
61
71
https://doi.org/10.1016/0092-8674(93)90160-r
105
John
,
R.M.
and
Lefebvre
,
L
. (
2011
)
Developmental regulation of somatic imprints
.
Differentiation.
81
,
270
280
https://doi.org/10.1016/j.diff.2011.01.007
106
Gregg
,
C.
,
Zhang
,
J.
,
Butler
,
J.E.
,
Haig
,
D.
and
Dulac
,
C
. (
2010
)
Sex-specific parent-of-origin allelic expression in the mouse brain
.
Science
329
,
682
685
https://doi.org/10.1126/science.1190831
107
Prickett
,
A.R.
and
Oakey
,
R.J
. (
2012
)
A survey of tissue-specific genomic imprinting in mammals
.
Mol. Genet. Genomics
287
,
621
630
https://doi.org/10.1007/s00438-012-0708-6
108
Vrana
,
P.B.
,
Guan
,
X.J.
,
Ingram
,
R.S.
and
Tilghman
,
S.M
. (
1998
)
Genomic imprinting is disrupted in interspecific peromyscus hybrids
.
Nat. Genet.
20
,
362
365
https://doi.org/10.1038/3833
109
Luedi
,
P.P.
,
Dietrich
,
F.S.
,
Weidman
,
J.R.
,
Bosko
,
J.M.
,
Jirtle
,
R.L.
and
Hartemink
,
A.J
. (
2007
)
Computational and experimental identification of novel human imprinted genes
.
Genome Res.
17
,
1723
1730
https://doi.org/10.1101/gr.6584707
110
Luedi
,
P.P.
,
Hartemink
,
A.J.
and
Jirtle
,
R.L
. (
2005
)
Genome-wide prediction of imprinted murine genes
.
Genome Res.
15
,
875
884
https://doi.org/10.1101/gr.3303505
111
Schoenherr
,
C.J.
,
Levorse
,
J.M.
and
Tilghman
,
S.M
. (
2003
)
CTCF maintains differential methylation at the Igf2/H19 locus
.
Nat. Genet.
33
,
66
69
https://doi.org/10.1038/ng1057
112
Latos
,
P.A.
,
Pauler
,
F.M.
,
Koerner
,
M.V.
,
Şenergin
,
H.B.
,
Hudson
,
Q.J.
,
Stocsits
,
R.R.
et al.
(
2012
)
Airn transcriptional overlap, but not its lncRNA products, induces imprinted Igf2r silencing
.
Science
338
,
1469
1472
https://doi.org/10.1126/science.1228110
113
Monk
,
D.
,
Arnaud
,
P.
,
Apostolidou
,
S.
,
Hills
,
F.A.
,
Kelsey
,
G.
,
Stanier
,
P.
et al.
(
2006
)
Limited evolutionary conservation of imprinting in the human placenta
.
Proc. Natl. Acad. Sci. U.S.A.
103
,
6623
6628
https://doi.org/10.1073/pnas.0511031103
114
O’Sullivan
,
F.M.
,
Murphy
,
S.K.
,
Simel
,
L.R.
,
McCann
,
A.
,
Callanan
,
J.J.
and
Nolan
,
C.M
. (
2007
)
Imprinted expression of the canine IGF2R, in the absence of an anti-sense transcript or promoter methylation
.
Evol. Dev.
9
,
579
589
https://doi.org/10.1111/j.1525-142X.2007.00198.x
115
Weidman
,
J.R.
,
Dolinoy
,
D.C.
,
Maloney
,
K.A.
,
Cheng
,
J.F.
and
Jirtle
,
R.L
. (
2006
)
Imprinting of opossum Igf2r in the absence of differential methylation and air
.
Epigenetics
1
,
49
54
https://doi.org/10.4161/epi.1.1.2592
116
De Souza
,
A.T.
,
Hankins
,
G.R.
,
Washington
,
M.K.
,
Orton
,
T.C.
and
Jirtle
,
R.L
. (
1995
)
M6P/IGF2R gene is mutated in human hepatocellular carcinomas with loss of heterozygosity
.
Nat. Genet.
11
,
447
449
https://doi.org/10.1038/ng1295-447
117
Hughes
,
J.
,
Surakhy
,
M.
,
Can
,
S.
,
Ducker
,
M.
,
Davies
,
N.
,
Szele
,
F.
et al.
(
2019
)
Maternal transmission of an Igf2r domain 11: IGF2 binding mutant allele (Igf2r(I1565A) results in partial lethality, overgrowth and intestinal adenoma progression)
.
Sci. Rep.
9
, 11388 https://doi.org/10.1038/s41598-019-47827-9
118
Smith-Vikos
,
T.
,
Liu
,
Z.
,
Parsons
,
C.
,
Gorospe
,
M.
,
Ferrucci
,
L.
,
Gill
,
T.M.
et al.
(
2016
)
A serum miRNA profile of human longevity: findings from the baltimore longitudinal study of aging (BLSA)
.
Aging (Milano)
8
,
2971
2987
https://doi.org/10.18632/aging.101106
119
Orwell
,
G
. (
1945
)
Animal farm
,
London, England
,
Secker and Warburg
120
Tycko
,
B
. (
2010
)
Allele-specific DNA methylation: beyond imprinting
.
Hum. Mol. Genet.
19
,
R210
20
https://doi.org/10.1093/hmg/ddq376
121
Jirtle
,
R.
and
Weidman
,
J
. (
2007
)
Imprinted and more equal
.
Am. Sci.
95
,
143
149
https://doi.org/10.1511/2007.64.143
122
Keverne
,
E.B
. (
2001
)
Genomic imprinting and the maternal brain
.
Prog. Brain Res.
133
,
279
285
https://doi.org/10.1016/s0079-6123(01)33021-2
123
Hunter
,
P
. (
2007
)
The silence of genes. Is genomic imprinting the software of evolution or just a battleground for gender conflict?
EMBO Rep.
8
,
441
443
https://doi.org/10.1038/sj.embor.7400965
124
Ishida
,
M.
and
Moore
,
G.E
. (
2013
)
The role of imprinted genes in humans
.
Mol. Aspects Med.
34
,
826
840
https://doi.org/10.1016/j.mam.2012.06.009
125
Eggermann
,
T.
,
Eggermann
,
K.
and
Schönherr
,
N
. (
2008
)
Growth retardation versus overgrowth: silver-russell syndrome is genetically opposite to beckwith-wiedemann syndrome
.
Trends Genet.
24
,
195
204
https://doi.org/10.1016/j.tig.2008.01.003
126
Riccio
,
A.
,
Sparago
,
A.
,
Verde
,
G.
,
De Crescenzo
,
A.
,
Citro
,
V.
,
Cubellis
,
M.V.
et al.
(
2009
)
Inherited and sporadic epimutations at the IGF2-H19 locus in beckwith-wiedemann syndrome and wilms’ tumor
.
Endocr. Dev.
14
,
1
9
https://doi.org/10.1159/000207461
127
Morison
,
I.M.
,
Ramsay
,
J.P.
and
Spencer
,
H.G
. (
2005
)
A census of mammalian imprinting
.
Trends Genet.
21
,
457
465
https://doi.org/10.1016/j.tig.2005.06.008
128
Pinto
,
D.
,
Pagnamenta
,
A.T.
,
Klei
,
L.
,
Anney
,
R.
,
Merico
,
D.
,
Regan
,
R.
et al.
(
2010
)
Functional impact of global rare copy number variation in autism spectrum disorders
.
Nature
466
,
368
372
https://doi.org/10.1038/nature09146
129
Walsh
,
T.
,
McClellan
,
J.M.
,
McCarthy
,
S.E.
,
Addington
,
A.M.
,
Pierce
,
S.B.
,
Cooper
,
G.M.
et al.
(
2008
)
Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia
.
Science
320
,
539
543
https://doi.org/10.1126/science.1155174
130
Karlsson
,
R.
,
Graae
,
L.
,
Lekman
,
M.
,
Wang
,
D.
,
Favis
,
R.
,
Axelsson
,
T.
et al.
(
2012
)
MAGI1 copy number variation in bipolar affective disorder and schizophrenia
.
Biol. Psychiatry Cogn. Neurosci. Neuroimaging
71
,
922
930
https://doi.org/10.1016/j.biopsych.2012.01.020
131
Escher
,
J.
,
Yan
,
W.
,
Rissman
,
E.F.
,
Wang
,
H.L.V.
,
Hernandez
,
A.
and
Corces
,
V.G
. (
2022
)
Beyond genes: germline disruption in the etiology of autism spectrum disorders
.
J. Autism Dev. Disord.
52
,
4608
4624
https://doi.org/10.1007/s10803-021-05304-1
132
McMillen
,
I.C.
and
Robinson
,
J.S
. (
2005
)
Developmental origins of the metabolic syndrome: prediction, plasticity, and programming
.
Physiol. Rev.
85
,
571
633
https://doi.org/10.1152/physrev.00053.2003
133
Susser
,
E.
,
Neugebauer
,
R.
,
Hoek
,
H.W.
,
Brown
,
A.S.
,
Lin
,
S.
and
Labovitz
,
D
. (
1996
)
Schizophrenia after prenatal famine
.
Arch. Gen. Psychiatry
53
,
25
31
https://doi.org/10.1001/archpsyc.1996.01830010027005
134
St Clair
,
D.
,
Xu
,
M.
,
Wang
,
P.
,
Yu
,
Y.
,
Fang
,
Y.
,
Zhang
,
F.
et al.
(
2005
)
Rates of adult schizophrenia following prenatal exposure to the Chinese famine of 1959-1961
.
JAMA
294
,
557
562
https://doi.org/10.1001/jama.294.5.557
135
Heijmans
,
B.T.
,
Tobi
,
E.W.
,
Stein
,
A.D.
,
Putter
,
H.
,
Blauw
,
G.J.
,
Susser
,
E.S.
et al.
(
2008
)
Persistent epigenetic differences associated with prenatal exposure to famine in humans
.
Proc. Natl. Acad. Sci. U.S.A.
105
,
17046
17049
https://doi.org/10.1073/pnas.0806560105
136
Morgan
,
R.K.
,
Wang
,
K.
,
Svoboda
,
L.K.
,
Rygiel
,
C.A.
,
Lalancette
,
C.
,
Cavalcante
,
R.
et al.
(
2024
)
Effects of developmental lead and phthalate exposures on dna methylation in adult mouse blood, brain, and liver: a focus on genomic imprinting by tissue and sex
.
Environ. Health Perspect.
132
,
67003
, 067003 https://doi.org/10.1289/EHP14074
137
Hoyo
,
C.
,
Murtha
,
A.P.
,
Schildkraut
,
J.M.
,
Forman
,
M.R.
,
Calingaert
,
B.
,
Demark-Wahnefried
,
W.
et al.
(
2011
)
Folic acid supplementation before and during pregnancy in the newborn epigenetics STudy (NEST)
.
BMC Public Health
11
, 46 https://doi.org/10.1186/1471-2458-11-46
138
Soubry
,
A.
,
Schildkraut
,
J.M.
,
Murtha
,
A.
,
Wang
,
F.
,
Huang
,
Z.
,
Bernal
,
A.
et al.
(
2013
)
Paternal obesity is associated with IGF2 hypomethylation in newborns: results from a newborn epigenetics Study (NEST) cohort
.
BMC Med.
11
, 29 https://doi.org/10.1186/1741-7015-11-29
139
Soubry
,
A.
,
Murphy
,
S.
,
Huang
,
Z.
,
Murtha
,
A.
,
Schildkraut
,
J.
,
Jirtle
,
R.
et al.
(
2011
)
The effects of depression and use of antidepressive medicines during pregnancy on the methylation status of the IGF2 imprinted control regions in the offspring
.
Clin. Epigenetics
3
, 2 https://doi.org/10.1186/1868-7083-3-2
140
Liu
,
Y.
,
Murphy
,
S.K.
,
Murtha
,
A.P.
,
Fuemmeler
,
B.F.
,
Schildkraut
,
J.
,
Huang
,
Z.
et al.
(
2012
)
Depression in pregnancy, infant birth weight and DNA methylation of imprint regulatory elements
.
Epigenetics
7
,
735
746
https://doi.org/10.4161/epi.20734
141
Liu
,
Y.
,
Hoyo
,
C.
,
Murphy
,
S.
,
Huang
,
Z.
,
Overcash
,
F.
,
Thompson
,
J.
et al.
(
2013
)
DNA methylation at imprint regulatory regions in preterm birth and infection
.
Am. J. Obstet. Gynecol.
208
,
395
https://doi.org/10.1016/j.ajog.2013.02.006
142
Vidal
,
A.C.
,
Murphy
,
S.K.
,
Murtha
,
A.P.
,
Schildkraut
,
J.M.
,
Soubry
,
A.
,
Huang
,
Z.
et al.
(
2013
)
Associations between antibiotic exposure during pregnancy, birth weight and aberrant methylation at imprinted genes among offspring
.
Int. J. Obes. (Lond)
37
,
907
913
https://doi.org/10.1038/ijo.2013.47
143
Murphy
,
S.K.
,
Adigun
,
A.
,
Huang
,
Z.
,
Overcash
,
F.
,
Wang
,
F.
,
Jirtle
,
R.L.
et al.
(
2012
)
Gender-specific methylation differences in relation to prenatal exposure to cigarette smoke
.
Gene
494
,
36
43
https://doi.org/10.1016/j.gene.2011.11.062
144
House
,
J.S.
,
Hall
,
J.
,
Park
,
S.S.
,
Planchart
,
A.
,
Money
,
E.
,
Maguire
,
R.L.
et al.
(
2019
)
Cadmium exposure and MEG3 methylation differences between Whites and African Americans in the NEST Cohort
.
Environ. Epigenet.
5
, dvz014 https://doi.org/10.1093/eep/dvz014
145
Li
,
Y.
,
Xie
,
C.
,
Murphy
,
S.K.
,
Skaar
,
D.
,
Nye
,
M.
,
Vidal
,
A.C.
et al.
(
2016
)
Lead Exposure during Early Human Development and DNA Methylation of Imprinted Gene Regulatory Elements in Adulthood
.
Environ. Health Perspect.
124
,
666
673
https://doi.org/10.1289/ehp.1408577
146
Murphy
,
S.K.
,
Itchon-Ramos
,
N.
,
Visco
,
Z.
,
Huang
,
Z.
,
Grenier
,
C.
,
Schrott
,
R.
et al.
(
2018
)
Cannabinoid exposure and altered DNA methylation in rat and human sperm
.
Epigenetics
13
,
1208
1221
https://doi.org/10.1080/15592294.2018.1554521
147
Schrott
,
R.
,
Greeson
,
K.W.
,
King
,
D.
,
Symosko Crow
,
K.M.
,
Easley
,
C.A. t.
and
Murphy
,
S.K
. (
2022
)
Cannabis alters DNA methylation at maternally imprinted and autism candidate genes in spermatogenic cells
.
Syst. Biol. Reprod. Med.
68
,
357
369
https://doi.org/10.1080/19396368.2022.2073292
148
Bland
,
J.S
. (
2021
)
A Discovery that Reframes the Whole of Global Healthcare in the 21st Century: The Importance of the Imprintome
.
Integr. Med. (Encinitas).
20
,
18
22
149
Lander
,
E.S.
,
Linton
,
L.M.
,
Birren
,
B.
,
Nusbaum
,
C.
,
Zody
,
M.C.
,
Baldwin
,
J.
et al.
(
2001
)
Initial sequencing and analysis of the human genome
.
Nature New Biol.
409
,
860
921
https://doi.org/10.1038/35057062
150
Venter
,
J.C.
,
Adams
,
M.D.
,
Myers
,
E.W.
,
Li
,
P.W.
,
Mural
,
R.J.
,
Sutton
,
G.G.
et al.
(
2001
)
The sequence of the human genome
.
Science
291
,
1304
1351
https://doi.org/10.1126/science.1058040
151
Barel
,
O.
,
Shalev
,
S.A.
,
Ofir
,
R.
,
Cohen
,
A.
,
Zlotogora
,
J.
,
Shorer
,
Z.
et al.
(
2008
)
Maternally inherited birk barel mental retardation dysmorphism syndrome caused by a mutation in the genomically imprinted potassium channel KCNK9
.
Am. J. Hum. Genet.
83
,
193
199
https://doi.org/10.1016/j.ajhg.2008.07.010
152
Skaar
,
D.A.
,
Dietze
,
E.C.
,
Alva-Ornelas
,
J.A.
,
Ann
,
D.
,
Schones
,
D.E.
,
Hyslop
,
T.
et al.
(
2021
)
Epigenetic dysregulation of KCNK9 imprinting and triple-negative breast cancer
.
Cancers (Basel)
13
, 6031 https://doi.org/10.3390/cancers13236031
153
Cevik
,
S.E.
,
Skaar
,
D.A.
,
Jima
,
D.D.
,
Liu
,
A.J.
,
Østbye
,
T.
,
Whitson
,
H.E.
et al.
(
2024
)
DNA methylation of imprint control regions associated with Alzheimer’s disease in non-hispanic blacks and non-hispanic whites
.
Clin. Epigenetics
16
, 58 https://doi.org/10.1186/s13148-024-01672-4
154
Alzheimer’s Association
. (
2022
)
2022 Alzheimer’s disease facts and figures
.
Alzheimers. Dement.
18
,
700
789
https://doi.org/10.1002/alz.12638
155
Chang
,
W.J.
,
Maduranga
,
U.
,
Gunasekara
,
C.J.
,
Yang
,
A.
,
Hirschtritt
,
M.
,
Rodriguez
,
K.
et al.
(
2025
)
Human genomic regions of systemic interindividual epigenetic variation are implicated in neurodevelopmental and metabolic disorders
.
medRxiv
2025.02.10.25322021 https://doi.org/10.1101/2025.02.10.25322021
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).