Next Article in Journal
A Spatial Transcriptome Reveals Changes in Tumor and Tumor Microenvironment in Oral Cancer with Acquired Resistance to Immunotherapy
Next Article in Special Issue
Biological Rhythms, Chrono-Nutrition, and Gut Microbiota: Epigenomics Insights for Precision Nutrition and Metabolic Health
Previous Article in Journal
Antimicrobial Potential of Different Isolates of Chaetomium globosum Combined with Liquid Chromatography Tandem Mass Spectrometry Chemical Profiling
Previous Article in Special Issue
Nicotinic Acid-Mediated Modulation of Metastasis-Associated Protein 1 Methylation and Inflammation in Brain Arteriovenous Malformation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transgenerational Epigenetic DNA Methylation Editing and Human Disease

by
Joshua D. Tompkins
Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
Biomolecules 2023, 13(12), 1684; https://doi.org/10.3390/biom13121684
Submission received: 1 November 2023 / Revised: 18 November 2023 / Accepted: 20 November 2023 / Published: 22 November 2023
(This article belongs to the Special Issue DNA Methylation in Human Diseases)

Abstract

:
During gestation, maternal (F0), embryonic (F1), and migrating primordial germ cell (F2) genomes can be simultaneously exposed to environmental influences. Accumulating evidence suggests that operating epi- or above the genetic DNA sequence, covalent DNA methylation (DNAme) can be recorded onto DNA in response to environmental insults, some sites which escape normal germline erasure. These appear to intrinsically regulate future disease propensity, even transgenerationally. Thus, an organism’s genome can undergo epigenetic adjustment based on environmental influences experienced by prior generations. During the earliest stages of mammalian development, the three-dimensional presentation of the genome is dramatically changed, and DNAme is removed genome wide. Why, then, do some pathological DNAme patterns appear to be heritable? Are these correctable? In the following sections, I review concepts of transgenerational epigenetics and recent work towards programming transgenerational DNAme. A framework for editing heritable DNAme and challenges are discussed, and ethics in human research is introduced.

1. The Epigenome and DNA Methylation

The epigenome functions to regulate DNA interactions and provides a basis for the storage and extraction of information across the genome. Radiating outward from a single cell, each of the trillions of derivative cells will adopt a unique epigenome, similar by cell and tissue type, responsive, and reinforced by an array of epigenetic regulators. Major epigenetic factors include covalent DNA modifications, such as DNA methylation (DNAme) or quadruplex structures, a multitude of histone modifications and chromatin remodelers, which facilitate DNA accessibility, as well as short- and long-noncoding RNAs that mediate DNA-protein interactions [1,2,3,4,5]. There are some cells which maintain epigenome plasticity over their lifetime (e.g., multipotent stem cells), while others exhibit terminal post-mitotic configurations (e.g., cardiomyocytes) [6,7,8,9,10,11,12]. Primordial germ cells (PGCs) and cells of the pre-implantation blastocyst stage of development will uniquely undergo a remarkable form of epigenetic remodeling, in which genome presentation is reset, and the genome evolves between parent and child [11,12,13,14,15]. During these stages, DNAme is globally removed. Yet, the decoration is again re-established during gametogenesis and blastocyst stages of development (Figure 1).
It was Conrad Hal Waddington who first defined epigenetics as the “branch of biology that studies the causal interactions between genes and their products which bring the phenotype into being” (p. 5, [16]). Scientists would later ascribe an array of epigenetic modifications and editors that progressively restrict developmental potential in much the same way Waddington viewed epigenetic forces must exist, most famously illustrated by his epigenetic landscape. Indeed, the cellular potential is restricted over developmental time, much like a ball driven by gravity transversing a series of narrowing valleys finds an ultimate pathway (Figure 2). An image has been generated with the help of DALLE-3 artificial intelligence (AI) to conceptually illustrate this, with certain updates for current developmental biology. A ball of embryonic stem cells, perched at the edge of a cliff overseeing valleys of fate, having rolled slightly downward from the totipotent zygote, would fall stochastically forward in developmental time, dividing and spilling ultimately trillions of cells into the valleys below. The fog represents DNAme accumulating over developmental time and the corresponding removal of developmental potency. A somatic cell will ultimately become clouded in its identity. If we imagine a rare group of cells, uniquely protected from commitment and finding a rare pathway across the highest ridgelines, this would be akin to primordial germ cell migration and the maintenance of epigenomic potential between generations, as discussed in the following sections. Extrinsic forces may recontour these ridges and valleys, some of which are required in normal balance (e.g., maternal hormones), and some of which may be pathological (e.g., gestational diabetes).
Though Waddington did not refer to contemporary features we now understand to be epigenetic, key epigenetic factors, including methylated DNA, were, in fact, discovered prior to his work. Cytosine methylated DNA was originally synthesized by Treat Johnson in 1904, and he would use the corresponding crystal picrate to later identify 5-methylcytosine (5mC) in bacteria. He would write, “the discovery of this compound increases the number of pyrimidines functioning in life” [17,18]. However, eukaryotic detection and epigenetic implications would take several additional decades of work. Observations of asymmetric DNAme across cell types and species, and the identification of DNA methylating and demethylating systems, represent pivotal moments in epigenetic discovery. “The amounts [of 5mC] in which it occurs, however, varying with the source but constant from a given source, suggest that it is an essential constituent of certain DNAs and no accident of enzyme action”, Wyatt 1951, (p. 583, [19]). DNAme has since become the most extensively studied epigenetic mark, owing to its early discovery, demonstrated requirements for mammalian development, and long-term stability in archived DNA samples [20]. References for additional historical insights on epigenetic concepts and DNAme discovery are provided [2,20,21].

2. DNA Methylation in Development, Cell Memory, and Disease

Epigenome configurations change in response to developmental cues, often drastically, and coordinate the differentiation and function of cells as they integrate multiple complex organ systems. DNAme is required for cellular differentiation [11,12,22,23]. The enzymatic reaction occurs at the C-5 position of cytosine, predominately in CpG contexts, which are often functionally clustered together in CpG-dense regions termed CpG “islands” (CGIs) [20]. These are speckled across an evolutionarily CG-depleted genome. Three enzymes are known to form 5mC. DNMT3A and 3B’s de novo methyltransferase activity is reinforced by maintenance enzyme DNMT1, which positively associates with hemimethylated DNA and faithfully copies DNAme patterns onto the nascent strands during DNA replication [24]. On the other hand, three enzymes, TET1, 2, and 3, actively demethylate DNA via 5mC oxidation to 5-hydroxymethylC and potential removal via subsequent base excision repair or passively through DNA replication (Figure 3) [25]. For the most part, genome-wide DNAme patterns are stably maintained; however, during cellular differentiation, DNAme patterns are dynamically altered [9,10,26]. Here, DNAme represents a conserved system for transcriptional regulation and cell memory, which protects the genome from transposable elements and enables the specification of unique cellular identities [6,9,11,12,27,28,29,30,31]. For example, developmental transcription factors become gene body hypermethylated upon activation and retain residual DNAme signatures, and DNAme directs Polycomb repressive activity in chromatin organization during the earliest stages of development [9,13,32]. Such activities are also found at developmentally poised promoters and enhancer networks, which may be particularly dynamic prior to the specification of cellular fate [33,34,35].
There are times when pathological DNAme patterns are recorded from environmental influences or by mistakes through faulty DNA methyl editing machinery. For example, long-term type I diabetes (TID) complications are associated with a prior period of poor glycemic control, both micro- and macrovascular complications, including diabetic kidney disease, retinopathy, atherosclerosis, and vascular disease. These complications are predicted by a DNAme “metabolic memory”, detectable by specific CpG-DNAme changes decades before complications clinically present [6,7]. Several CpGs in combination can explain 68–97% of HbA1C association with the risk of complications development, with sites enriched genome wide for enhancers in blood and hematopoietic stem and progenitor cells (HSPCs), as well as open chromatin regions in myeloid progenitors [6]. Metabolic memory is predictive of elevated inflammation and vascular disease and may largely exist because stable DNAme changes encode a functional epigenetic history of extrinsic hyperglycemia in HSPCs, which is maintained by DNMT1 activity, and which drives functional defects in differentiated immune cells as well [6]. Therefore, DNAme signatures provide unique insights into developmental and environmental influences recorded on DNA as an epigenetic memory, even over vast stretches of time [9,36,37,38]. Given the essential role DNAme plays in development, it is not surprising that many cancers also routinely hijack DNAme networks for growth and metastatic behavior. CpG Island (CGI) hyperMethylation Phenotypes (CIMPs) and the reactivation of developmental gene networks are hallmarks of numerous cancers, frequently involving mutation or inactivation of DNMT or TET enzymes [29,39,40,41,42]. Other examples include promoter-CGI hypermethylation of the tumor suppressor MutL Homolog 1 (MLH1) in colorectal, esophageal, and thymic epithelial tumors [43,44,45,46,47] and promoter TP53 DNAme in several cancers and in stroke patients [48,49,50]. Specific DNAme changes are also observed in atherosclerosis and cardiovascular disease (e.g., ABCG1), metabolic disorders (e.g., ANKRD26, IGF2), Alzheimer’s disease, and other nervous system disorders (e.g., IGF2) [36,37,51,52,53,54,55,56,57,58,59,60]. DNAme is highly predictive of biological age, and several pathological states accelerate DNAme age [6,9,10,37,38,61,62,63,64].

3. The Erasure of DNA Methylation from the Germline

Given the lifetime accumulation of environmental and age-associated epigenetic changes, mechanisms must exist to remove pathological epigenetic information where it exists in the germline. The timing of the first wave of demethylation is unique to male and female germ cells, and the second occurs shortly after male and female genomes have integrated. Germ line erasure overview. The mammalian germ line undergoes unique migratory, replicative, and global DNAme changes, which rearrange and epigenetically reconfigure the genome for its eventual passage onto the next generation. In mice, PGCs emerge from the proximal epiblast, numbering approximately 40 by 7.25 days post coitum (dpc) [65]. Migration across the posterior primitive streak to embryonic endoderm follows, with eventual migration to the genital ridge and gonad at 10 dpc [65,66]. It is during the migration and colonization of the genital ridges when global erasure of DNAme occurs, from ~70% to 4% at 13.5 dpc PGCs [15,66,67]. The X chromosome is reactivated, and imprinting sites are also generally demethylated [15,66,67]. Demethylation occurs in two phases, the first by passive demethylation from 8.5 dpc to 9.5 dpc, and active demethylation by TET1/TET2 from 9.5 dpc to 13.5 dpc, with peak 5mC oxidation activity at 11.5 dpc [12]. In males, acquisition of sex-specific DNAme occurs at 13.5 dpc forward, but female PGC DNAme patterns emerge after birth [12,66]. Specialized recombination will follow, allowing the separation of the diploid genome to haploid states as germ cells mature through meiosis [68,69,70,71]. Pre-implantation stages of DNA demethylation overview: Prior to fertilization, the male genome is at its highest level of CpG DNAme (~90%), and the female genome is near 40% DNAme [72]. The pronucleus enters the oocyte and begins to decrease DNAme even prior to the first round of division. An active process, TET3 is predominantly involved with the rapid conversion of 5mC to 5hMC and other derivatives, which persist until they are depleted by cell division; thus, demethylation is both active and passive [12]. The maternal genome is protected from high TET3 activity by maternal factor STELLA, and demethylation proceeds in a passive fashion [73]. Despite the virtual loss of all 5mC by the 16-cell stage of development, imprinting regions often clustered together escape this activity [14,74]. This is reinforced by low levels of site-specific DNMT1 activity and requires ZFP-TRIM28 heterochromatin-inducing activity to reinforce the marks [14,74,75]. Transposable element control relies heavily on histone H3 lysine 9 trimethylation (H3K9me3), with TRIM28/SETB1 controlling this deposition, along with other Krüppel associated box (KRAB) domain-containing zinc fingers, which promote eventual stable silencing by DNAme through recruitment of DNMT activity [12,74,76]. Additional insights may be gleaned by examining the behavior of imprinting sites and transposable elements during reversible cultures of naïve and primed pluripotent embryonic stem cells (ESCs), with the naïve state being hypomethylated relative to primed, more developmentally forward ESCs [32,77]. Given the natural occurrence of rare imprinting regions that naturally escape global demethylation events and augment the acquisition of global DNAme in PGC and pre-implantation blastocyst stages of development, accumulating evidence suggests some DNAme patterns are transgenerationally heritable.

4. Transgenerational DNA Methylation

It is now widely understood that genotoxic or epimutation events in embryonic development can influence lifelong propensities to develop wide-ranging human pathophysiological conditions. This is especially true during embryonic development spanning PGC migration and development, where both first-generation (F1) embryos and second-generation (F2) PGCs may be exposed to the same toxin or its effects. Thus, when considering a gestating mother, only the detection of the epimutation that drives a phenotypic change in the F3 generation, without continued exposure to the original insult, can be considered transgenerational (Figure 4). If, however, exposure of the postnatal mother or father (F0) results in F1 germline exposure, then the first unexposed transgenerational effect is F2 [78]. In either case, continued transmission of the epigenetic effect suggests transgenerational maintenance.
There is now considerable evidence for transgenerational epigenetic phenomena. Well demonstrated in plants, major environmental changes such as drought produce distinct epigenetic changes that can alter flowering rapidly and for generations [79]. For example, increased Lcyc promoter DNAme induces radial from bilateral symmetry, which is heritable for >100 generations [80]. Transgenerational epigenetic phenomena have since been observed in insects [81,82], zebrafish [83,84], birds [85,86], and mammals, including humans [87,88,89]. In mice, early demonstrations involved gestational exposure to the fungicide vinclozolin, which results in germline DNAme changes and sperm and fertility defects through F4 generations [90]. Since this landmark study, several additional environmental pollutants have been implicated in transgenerational disease. Major examples include pesticides containing DEET [91], mercury [83], tributyltin [92], insecticides [93,94], cigarettes [95], and plastic components including Bisphenol A (BPA) and phthalates, among others [94,96,97,98]. This also includes traumatic stress and famine [99,100]. These have been summarized in multiple reviews [94,101,102,103]. Effects can be wide-ranging and include reduced sperm counts and fecundity, kidney disease, obesity, immune dysregulation, cardiovascular disease, and cancer [45,97,102,104,105].
Malnourished mothers give rise to offspring with increased metabolic and cardiovascular disease incidence [106,107]. This so-called “thrifty phenotype” is increased obesity and poor adipose mobilization in offspring subjected to poor fetal nutrition [107]. This extends to adult F2 offspring of gestationally exposed F1 fathers, who had higher weights and body mass index (BMI) relative to unexposed F1 fathers from the 1944–1945 Dutch famine [88]. Several other studies have linked transgenerational effects from both overnutrition and malnutrition to metabolic syndromes and increased adiposity [108,109,110]. Interestingly, these effects are also observed with prenatal exposure to glyphosate [93], plastics [96], dioxins, and dichlorodiphenyltrichloroethane (DDT) [111]. Among dioxins, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the so-called “Agent Orange”, was widely utilized as a herbicide in the Vietnam War. It is considered the deadliest. At 0.1% lethal levels of oral F0 exposure, the unexposed F3 generation has significantly elevated levels of kidney disease. Depending on the study, dozens to hundreds of differentially methylated regions (DMRs) were induced and observed in the F3 generation [112]. Differential DNAme was noted at the progesterone receptor and insulin-like growth factor (Igf2), which may also extend to the imprinting control region [112,113,114]. Given the role Igf2 differential methylation may play in Alzheimer’s and major psychosis events, transgenerational DNAme at this locus may provide an epigenetic basis for potential neurological conditions associated with ancestral exposures [51,52,113]. For DDT, the transgenerational effects appear to be mediated through specific changes in sperm DNAme, with many DMRs that occur at genes known to promote obesity. F3 offspring were obese, with kidney disease in both males and females [111]. Interestingly, low-density regions termed ‘CpG deserts’, which contain <15 CpGs/100 bp but near clusters of CpGs, were identified to be associated with environmentally induced differential DNAme in sperm, which appears common to transgenerational DMRs [115,116]. These DMRs escape DNAme erasure during embryonic development, similar to imprinting sites [117].

5. Concerns for Global Health

Differential methylation effects on sperm DNA have been demonstrated to be toxin-specific [116]; therefore, unique environmental exposures, whether chemical, dietary, or otherwise, will likely manifest in specific human disease conditions or kinetics. However, the human reproductive period spans decades, lifespan decades longer, and in many instances, we are currently witnessing the health span effects on F3 individuals, observing the epigenetically heritable consequences of war, trauma, unregulated industrial expansion, and pollution in real-time. We are likely only beginning to understand the transmission of epigenetic information across generations, and certain profound effects may already be here. From 1973 to 2018, sperm counts have declined a staggering 62.3% from 101.2 million/mL to 49.0 million/mL, the rate only accelerating to 2.64%/year each year since 2000 [118,119]. It is unclear where the bottom of this decline is, but counts below 20 million/mL are considered low, with expected delays to conception. At trends of 2.64% declines/year (documented through 2018 [118]), this would occur in approximately the year 2053.
The link between reduced male fertility exposure to pollution, chemicals, and endocrine-disrupting agents is quite clear, and many of these established factors are now under transgenerational scrutiny [97,101,102,104,105,118]. However, it is likely a combination of one’s individual exposure, as well as intergenerational and transgenerational effects mediated through ancestral exposure, which are collectively driving a reduction in sperm counts. The extent to which inappropriate epigenomic changes are interactive, cumulative, and/or allowable for overall human reproductive viability remains to be determined. Murine studies suggest a single acute exposure to certain environmental toxins is sufficient. However, the ubiquitous presence of environmental toxins in our lives today, coupled with the biological magnification of environmental toxins within our food chain, feeding pathological information directly into the germline of exposed early embryos and PGCs, provides a basis by which humans are concentrating pathological epigenetic signatures within the genomes of unborn children, animals, insects, and plants, and potentially generations of offspring. This extends beyond fertility- to the potential inheritance of predisposing epigenetic events to cancer, diabetes, cardiovascular disease, diseases of the central nervous system, and the overall reduction of human life expectancy.
Thus, strategies must continue to be globally implemented and expanded to limit human exposure to genotoxic and epigenotoxic chemicals and pollutants, and events such as war, trauma, disease, and famine. Otherwise, the effects may persist for countless years, well beyond the generation of initial exposure, and we may increasingly rely on high throughput screening approaches and in vitro fertilization to alleviate the inheritance of epigenetic events that reduce human health and lifespan. In cases where pathological epigenetic information exists, retention may exist for several generations, potentially hundreds of years when considering human life expectancy; thus, epigenetic editing strategies that remove sites of epigenetic dismay may be increasingly needed.

6. Locus-Specific DNA Methylation Editing

Researchers have, for some time, fused epigenetic effector domains to homing proteins [120,121,122]. The CRISPR/Cas revolution facilitated the genome-wide examination of such approaches, in which nuclease deficient or enzymatically “dead”, CRISPR-associated protein 9 (dCas9) guided to DNA by CRISPR RNAs (crRNAs) can be fused to select epigenetic modifiers for locus-specific epigenetic editing (Figure 5). Typical Cas9 inactivation involves point mutations at each of the nuclease domains (D10A and H840A), and several orthogonal dCas versions now exist with unique sizes and guide RNA-DNA binding rules [123,124,125,126]. Functional effector groups include the catalytic domains of DNMT3a, histone acetyltransferase p300, and histone demethylase LSD1, among others [127,128,129]. dCas9-DNMT3a methylates CpG’s in a 25–35 bp peak but inhibits DNAme directly beneath the dCas9 footprint, and crRNA targeting is variable across entire CGIs [127,130,131,132]. Larger effector domain fusions or presentation strategies, such as KRAB, may enhance epigenetic editing activity and facilitate larger DNAme edits that are more compatible with CGIs [133,134,135]. For example, “CRISPR-off” uses a dCas enzyme fused to Znf10-Krab, Dnmt3A, and Dnmt3L protein domains, and depending on configuration, it can effectively induce long-term target gene suppression through promoter targeting [133]. “CRISPR-on”, by comparison, utilizes transactivator domains fused to MS2 coat protein, including VP64, p65-AD, and Rta, and recruits these domains via two MS2 stem loop sequences embedded within the crRNA [133,136,137]. Yet, most DNAme editing studies to date have been conducted with simple-to-transfect immortalized cell lines (i.e., 293T cells), and delivery of ever larger dCas fusions or scaffolds within single cells alongside dozens of crRNAs simultaneously is generally not compatible with efficient in vivo viral delivery systems [138]. Smaller dCas orthologs, such as from Staphylococcus aureus, have been used in adeno-associated virus (AAV)-mediated targeting of murine liver or mRNA-based delivery in HSPCs [139,140]. dCas9-SunTag has been used in transgenic mice for gene activation in the liver and midbrain [141,142]. Other transgenic versions include Rosa26:LSL-dCas9-p300 for gene activation and Rosa26:LSL-dCas9-KRAB for gene repression, with effects examined in the liver and the brain [138]. Immune responses to bacterial Cas proteins remain a concern for the long-term, repeat genome, and epigenome editing strategies [143,144]. For example, in mice with pre-existing exposure and immunity to SaCas9, liver genome editing by AAV delivery could occur, but the effect was accompanied by cytotoxic T-cell responses, hepatocyte death, and complete elimination of gene-edited cells [144]. Despite progress in locus-specific gene activation and repression of these systems, there are few reports on using these systems for DNAme editing during early developmental windows.

7. DNAme Editing in Germ Cells and Early Embryos

During both iPSC formation and rapidly upon somatic cell nuclear transfer (SCNT), the epigenome adopts a plastic, hypomethylated state. However, there are sites resistant to pluripotent or totipotent reprogramming; residual epigenetic signatures can reflect the original somatic cell of origin and be resistant to proper X-chromosome inactivation [145,146,147,148]. Thus, some forms of global correction have been explored, such as histone deacetylase inactivation with Trichostatin A, which also decreased DNAme and increased cloning efficiency [148,149,150,151,152]. Direct locus-specific epigenetic manipulation in early development has only been recently described. In oocytes, dCas9-TET1 or, separately, dCas9-DNTM3a were introduced as mRNAs by microinjection and used to edit premeiotic oocytes prior to ovulation. This resulted in DNAme editing at the intracisternal A-particle (IAP) repetitive promoter, which regulates the Agouti gene [153]. Coat color changes were noted in accordance with IAP promoter DNAme levels. The H19 ICR was also targeted in a bimaternal mouse model with DNAme editing designed to mimic paternal imprinting states, resulting in recovering developmental competency [153]. Similarly, DNAme editing by zygote microinjection with dCas9-fused to an engineered prokaryotic DNA methyltransferase MQ1, targeting the H19/Igf2 paternally imprinted locus, was reported to increase DNAme modestly at 2/5 crRNA test sites in offspring, with inhibition of DNAme beneath the dCas9 footprint [154]. The lack of additional targets, functional weight gain with H19 DNAme increases, or heritability testing suggests a lack of transgenerational effects with this approach, or at this location [154,155]. Likely due to known global erasure events in DNAme and/or the difficulty in delivering efficient epigenetic editing systems to germ cells and zygotes, there are no other reports on germ cell or early embryonic designer DNAme in mammals using dCas9 based editing. Next-generation epigenetic editing systems may need to incorporate factors involved in DNAme deposition in germ cells and transitions from naïve to primed pluripotency during blastocyst stages of development. For example, P-element-inducing wimpy testis (PIWI)-related protein, MIWI2, is critical for retrotransposon silencing via PIWI-interacting RNAs. Accordingly, a zinc finger-MIWI2 fusion induced DNAme and suppression of type A LINE-1 gene and rescued otherwise inhibited spermatogenesis in MILI-null mice [156]. Dnmt3l (and Dnmt3C in mice) is most highly expressed in germ cells and ESCs, where it exerts effects on imprinting control regions and transposable elements via DNAme. Many studies have indicated that Dnmt3l enhances Dnmt3A and 3B activity, though this may be region or site-specific and can be antagonistic depending on Dnmt3l interactions with PRC2 at bivalent promoters [157,158]. Additional updates to early developmental epigenetic editing systems may include guide RNA modifications or the recruitment of stage-specific co-repressor, and activator complexes involved in imprinting or repetitive element maintenance.

8. Transgenerational DNA Methylation Editing in Mammals

Reminiscent of repetitive element silencing, Takahashi et al., recently identified that CG-free DNA insertion into CGIs triggered CGI-specific and CGI-wide de novo DNAme in both human PSCs and mouse PSCs [8,58] (Figure 6). Remarkably, mouse blastocyst/mPSC chimera offspring retained DNAme edits, which, despite the subsequent removal of the inserted DNA in mPSCs, was transgenerationally inherited [58]. Epigenetic obesity and hypercholesterolemia were mediated by targeted Ankrd26 and Ldlr CGI promoter DNAme gains, of which designer DNAme patterns were not reduced until the F4 and F6 generations, respectively [58]. The re-occurrence of the methylation response, generation-to-generation, despite transient erasure during PGC development, suggests that induced transgenerational reacquisition of patterned DNAme is dependent on specific histone marks that re-adopt stable DNAme silencing upon transitions through later stages of embryonic development. H3K9me3 was observed to be increased in regions showing transgenerational behavior at target CGIs where DNAme was reacquired, despite transient erasure through germ cell development. Much like retrotransposon regulation depending on H3K9me3 deposition to induce de novo DNAme and stably silence elements, CGI Methylation Responses (CIMRs) may occur due to specific changes to CGI architecture that resemble viral insertion events. This may occur in response to the expected removal of CGI binding factors when the CGI is initially interrupted, which otherwise protects the CGI from DNAme editing machinery and premature differentiation-associated acquisition of DNAme. These observations may also be related to “bivalent domain” resolution during development, in which many developmental enhancers and promoters contain both activating and repressive marks, poised for rapid and coordinated expression response in differentiation [159,160]. When synthetic CGIs are introduced randomly into mESCs, they adopt a bivalent unmethylated state. Yet, when CGIs are integrated at high AT density, all CpGs become simultaneously DNAme, and bivalency is resolved. Furthermore, low-density deserts and CpG clusters, which appear to escape transgenerational DNAme erasure during responses to environmental toxins, appear to behave similarly to these effects [115,116]. Overall, programmable transgenerational DNAme editing is possible, but whether these approaches can be generalized genome-wide remains to be determined. We have similarly observed CG-free insertion by synthetic CpG-free ssDNA insertion in both human and mouse embryonic stem cells to induce CGI-wide, stable, and globally specific DNAme [8]. Functionally retained through in vitro differentiation, engineered DNAme is retained post-CG-free DNA removal and through multilineage differentiation. Furthermore, designer MLH1 promoter DNAme was observed to skew thymic epithelial cell differentiation and sensitize multiple lineages to cisplatin [8]. By transcription factor binding enrichment analysis, we identified KLF6 as having a putative role in regulating CIMR responses. With regards to developmental CIMR timing, blastocysts did not exhibit targeted DNAme of the Ldlr CGI, but epiblasts did; thus, acquired DNAme at these interrupted CGIs occurs after implantation, upon re-establishment of global DNAme. In hESCs, we have observed CIMR DNAme acquisition to be restricted to the primed state of pluripotency and for reacquisition to occur when hESC transition from naïve to primed states using defined cultures ([8,15] unpublished observations). Collectively, this suggests that CG-free-DNA insertion into CGI or CG-dense areas of the genome, in germ cells or zygotes, will trigger locus- or region-specific DNAme as embryos transition through primed stages of development. In humans, this reflects pre-implantation blastocyst stages, and, in mice, this reflects the post-implantation epiblast stage. Regardless of the induction technique, the tracking of induced DNAme through inheritance, both with and without the continued presence of the original DNAme-inducing insert DNA, is essential for establishing transgenerational behavior. Whereas programmable transgenerational epigenetic transmission (PTET) involves designer epigenetic configurations that escape normal germline and pre-implantation stages of erasure and which influence the phenotype of subsequent generations, programmable transgenerational epigenetic reacquisition (PTER) involves epigenetic configurations that appear to be erased during PGC development and/or pre-implantation stages of development, but which are faithfully retriggered in each new developing embryo without the continued influence of epigenetic editing systems.
Mechanistically, transgenerational maintenance of programmed DNAme can be envisioned through the protection of acquired DNAme via germ line demethylation waves, in much the way STELLA, PGC7, or ZFP57 bind and protect imprinted genes. Alternatively, this may occur through the reacquisition of acquired transgenerational DNAme patterns with each ensuing generation, triggered iteratively through primed stages of pluripotency in each generation, in much the way CIMRs or specific forms of repetitive elements behave [8,58,161,162]. Demethylation strategies may be global, such as 5-aza-2′deoxycytidine and TSA, which non-specifically remove DNAme and have been used to improve SCNT cloning outcomes [151,152]; however, pathological sites of transgenerational DNAme could prove rare and highly specific, and thus warrant refinement of locus-specific demethylating strategies described above (see Locus-specific DNA methylation editing). Specifically, the use of a developmentally restricted effector or epigenetic editing domain presentation which naturally operates in germ cells or early embryonic development may be particularly helpful for removing stable sites of transgenerational memory.
In closing, inducible early embryo DNAme editing, whether by “dead” homing protein-based or programmed de novo DNAme induction events, enables inheritance testing of tailored DNAme into all derivatives of the three germ layers and through the specification of germ cells. To facilitate continued discovery and corrective strategies for these DNAme conditions, readers are provided with a framework for inducing and testing specific sites of transgenerational DNAme inheritance (Figure 7).

9. Future Concepts and Ethics

Transgenerational DNAme induction is relevant to numerous human diseases with many conditions affecting F3 generation offspring living today. Several human disease states are predicted by relatively few specific DNAme changes, and thus, it would seem even rarer that similar DNAme sites would escape normal germline erasure. However, evidence points to these exact events. The inheritance of novel sites of intrinsic epigenetic regulation may simultaneously drive germ cell selection and, when maintained into embryonic development, skew certain cell behaviors in much the way an altered epigenetic landscape changes differentiation potential (Figure 2). There is some natural variability in sperm, egg, and early embryo DNA methylation profiles; these are important drivers for stochastic events in germ cell selection and embryo specification, respectively [153,163,164,165]. However, some sites of differential DNAme are clearly disruptive to fertility. This includes H19 imprinting centers and several male age-associated CpGs and DMRs [166,167,168]. DNAme epigenetic-age testing may be additionally informative when assessing reproductive fitness and applied to in vitro fertilization screening protocols. It is possible that certain levels of epigenomic rejuvenation through the expression of naïve pluripotency, germ-cell-specific transcription factors, or small molecule mimics may enable a reduction in sperm or egg biological age and improve rates of conception.
The rapid decline in male sperm counts is troubling. Evidence indicates environmental pollution, trauma, and malnutrition as sources of epigenetic change, leading to declining germ cell function. Thus, germline or early embryo epigenetic editing may increasingly be warranted to remove disease-predisposing transgenerational information. Strategies are outlined in Figure 7. For some cases, single CGI or CG edits may be needed, requiring single-cell, single-DNAme-molecule read technologies for high-confidence selection of edited cells [153]. In the future, we may consider site-specific editing at dozens to thousands of sites simultaneously for activation, silencing, poising, and enhancing, all in coordination with the timing of extrinsic developmental signals. This form of systems-level deterministic epigenome manipulation will require significant development to safely alleviate transgenerational disease. The human lifetime is long, and perhaps even capable of extension through epigenomic rejuvenation, but accurately assessing lifespan extension can be challenging [169,170]. Epigenetic clocks that accurately predict biological age, aging rates, and disease propensity will, thus, play an important role in understanding whether certain toxins or events instill transgenerational DNAme, driving aging and multigenerational disease [61,63,171]. One may envision similar clocks applied to assessing transgenerational age or disease acceleration through the examination of specific sites or combinations of sites that persist across generations.
This review presents an overview of the literature that has implicated transgenerational DNAme in the reacquisition of certain pathophysiological conditions across generations. Strategies have also been presented for editing germline and early embryo transgenerational DNAme. Indeed, a world where the residual epigenetic effects of trauma, famine, and pollution have been erased from our germ line may very well be more peaceful and filled with longer life. This, of course, depends on the extent to which epigenetic changes can be deterministic. However, despite our technological leaps, mammalian and human development, especially post-implantation stages of development, still harbors an incredible mystery. For example, transgenerational DNAme may ostensibly arise from co-segregation with a mutation in a neighboring gene, which removes transcription termination [172,173]. When transcription was mutationally extended, de novo DNAme was also extended as it does across actively transcribed regions, and this induced promoter DNAme of the adjacent gene [172]. Thus, primary or secondary effects of mutation or DNAme are not always initially clear; even pinpointed DNAme edits early in life may have unexpected consequences among the trillions of derivative cells much further in life. Therefore, much like gene editing, developmental human epi-gene editing beyond accepted in vitro culture limits should also be under similar moratorium considerations [174].

10. Summary

Transgenerational epigenetic influences exist across plant and animal species, drive evolution, and may accelerate adaptive responses to detrimental environmental events. However, these processes occur during developmental windows, which are highly sensitive to environmentally, nutritionally, or hormonally induced changes to the genome and epigenome, of which gestational windows may simultaneously affect both F1 and F2 generations. Here, certain epigenetic insults or epigenetic edits may be transgenerationally durable. Recent advances in early embryo editing have enabled transgenerational disease modeling mammals for the first time. In a future where evidence for human transgenerational disease mounts, technologies may be needed that stably correct generations of otherwise reoccurring epigenetic disease.

Funding

Funding support for this article was provided by Riggs family donations to the Diabetes Metabolism Research Institute at City of Hope.

Acknowledgments

I would like to greatly acknowledge the long-time mentorship and dear friendship of Arthur Riggs. A pioneer in epigenetic concepts, Riggs was inspirational to many of the concepts presented in this work, and his presence is greatly missed. Additionally, thank you to Sung Hee Kil, City of Hope for proofreading and editing this work.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

5mC5-methylcytosine
CpGcytosine-phosphate-guanine
DNAmeDNA methylation
PGCprimordial germ cell
ESCembryonic stem cell
DMRdifferentially methylated region
DNA methylationThe covalent addition of methyl groups to DNA. Though this predominately occurs at 5′-CpG dinucleotides, non-CG methylation is noteworthy in pluripotency and various neural lineages
EpigeneticsThe cellular and organismal heritability of internal factors, including the modifications to them and by them, those recorded from environmental influences and in developmental history, whether physically local to the cell, signaled across an organism, or accumulated from sources larger in nature (for example, hormones, pollution, viruses, diet, and lifestyle), that influence the expression of chromosomally associated genetic information, establish stable cellular states over differentiation (or unstable states in aging and pathology), and enable the physical, biochemical, behavioral, cognitive, and social nature of an organism to emerge and function, without altering the primary DNA sequence [20]
EpimutationAn alteration to an epigenetic factor, such as DNA methylation, which may occur in response to an environmental insult or via incorrect deposition of the epigenetic feature
Programmable Transgenerational Epigenetic Reacquisition (PTER)The intentional, locus- or region-specific induction of specific epigenetic configurations that fail to escape germline or pre-implantation erasure, but which are retriggered with each ensuing generation without the continued presence of epigenetic editing systems
Programmable Transgenerational Epigenetic Transmission (PTET)The intentional, locus- or region-specific induction of specific epigenetic configurations that escape germline erasure and influence the phenotype of subsequent generations without the continued influence of epigenetic editing systems
Transgenerational Epigenetic InheritanceThe transmission of epigenetic information across generations that escapes germline erasure

References

  1. Tompkins, J.D.; Riggs, A.D. An Epigenetic Perspective on the Failing Heart and Pluripotent-Derived-Cardiomyocytes for Cell Replacement Therapy. Front. Biol. 2015, 10, 11–27. [Google Scholar] [CrossRef]
  2. Deans, C.; Maggert, K.A. What Do You Mean, “Epigenetic”? Genetics 2015, 199, 887–896. [Google Scholar] [CrossRef] [PubMed]
  3. Mukherjee, A.K.; Sharma, S.; Chowdhury, S. Non-duplex G-Quadruplex Structures Emerge as Mediators of Epigenetic Modifications. Trends Genet. 2019, 35, 129–144. [Google Scholar] [CrossRef]
  4. Zyner, K.G.; Simeone, A.; Flynn, S.M.; Doyle, C.; Marsico, G.; Adhikari, S.; Portella, G.; Tannahill, D.; Balasubramanian, S. G-Quadruplex DNA Structures in Human Stem Cells and Differentiation. Nat. Commun. 2022, 13, 142. [Google Scholar] [CrossRef]
  5. Richardson, A.E.; Zentz, Z.A.; Chambers, A.E.; Sandwith, S.N.; Reisinger, M.A.; Saunders, D.W.; Smaldino, P.J. G-Quadruplex Helicase Dhx36/G4r1 Engages Nuclear Lamina Proteins in Quiescent Breast Cancer Cells. ACS Omega 2020, 5, 24916–24926. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, Z.; Miao, F.; Braffett, B.H.; Lachin, J.M.; Zhang, L.; Wu, X.; Roshandel, D.; Carless, M.; Li, X.A.; Tompkins, J.D.; et al. DNA Methylation Mediates Development of Hba1c-Associated Complications in Type 1 Diabetes. Nat. Metab. 2020, 2, 744–762. [Google Scholar] [CrossRef] [PubMed]
  7. Chen, Z.; Miao, F.; Paterson, A.D.; Lachin, J.M.; Zhang, L.; Schones, D.E.; Wu, X.; Wang, J.; Tompkins, J.D.; Genuth, S.; et al. Epigenomic Profiling Reveals an Association between Persistence of DNA Methylation and Metabolic Memory in the Dcct/Edic Type 1 Diabetes Cohort. Proc. Natl. Acad. Sci. USA 2016, 113, E3002–E3011. [Google Scholar] [CrossRef]
  8. Tompkins, J. Engineering Pluripotent Cpg Island DNA Methylation by Cpg Island Methylation Response (Cimr) to Synthetic Cpg-Free Ssdna Insertion. Cell Rep. Methods 2023, 3, 100465. [Google Scholar] [CrossRef]
  9. Tompkins, J.D.; Jung, M.; Chen, C.Y.; Lin, Z.; Ye, J.; Godatha, S.; Riggs, A.D. Mapping Human Pluripotent-to-Cardiomyocyte Differentiation: Methylomes, Transcriptomes, and Exon DNA Methylation “Memories”. EBioMedicine 2016, 4, 74–85. [Google Scholar] [CrossRef]
  10. Tompkins, J.D.; Hall, C.; Chen, V.C.-Y.; Li, A.X.; Wu, X.; Hsu, D.; Couture, L.A.; Riggs, A.D. Epigenetic Stability, Adaptability, and Reversibility in Human Embryonic Stem Cells. Proc. Natl. Acad. Sci. USA 2012, 109, 12544–12549. [Google Scholar] [CrossRef]
  11. Smith, Z.D.; Meissner, A. DNA Methylation: Roles in Mammalian Development. Nat. Rev. Genet. 2013, 14, 204–220. [Google Scholar] [CrossRef]
  12. Yang, Z.; Chen, T. DNA Methylation Reprogramming during Mammalian Development. Genes 2019, 10, 257. [Google Scholar]
  13. McLaughlin, K.; Flyamer, I.M.; Thomson, J.P.; Mjoseng, H.K.; Shukla, R.; Williamson, I.; Grimes, G.R.; Illingworth, R.S.; Adams, I.R.; Pennings, S.; et al. DNA Methylation Directs Polycomb-Dependent 3D Genome Re-Organization in Naive Pluripotency. Cell Rep. 2019, 29, 1974–1985.e6. [Google Scholar] [CrossRef] [PubMed]
  14. SanMiguel, J.M.; Bartolomei, M.S. DNA Methylation Dynamics of Genomic Imprinting in Mouse Development. Biol. Reprod. 2018, 99, 252–262. [Google Scholar] [CrossRef] [PubMed]
  15. Seisenberger, S.; Andrews, S.; Krueger, F.; Arand, J.; Walter, J.; Santos, F.; Popp, C.; Thienpont, B.; Dean, W.; Reik, W. The Dynamics of Genome-wide DNA Methylation Reprogramming in Mouse Primordial Germ Cells. Mol. Cell 2012, 48, 849–862. [Google Scholar] [CrossRef]
  16. Waddington, C.H. Towards a Theoretical Biology. Nature 1968, 218, 525–527. [Google Scholar] [CrossRef] [PubMed]
  17. Wheeler, T.B.; Johnson, H.L. 5-Methylcytosine. Am. Chem. J. 1904, 31, 591–606. [Google Scholar]
  18. Johnson, T.B.; Coghill, R.D. The Discovery of 5-Methyl-Cytosine in Tuberculinic Acid, the Nucleic Acid of the Tubercle Bacillus. J. Am. Chem. Soc. 1925, 11, 2838–2844. [Google Scholar] [CrossRef]
  19. Wyatt, G.R. Recognition and Estimation of 5-Methylcytosine in Nucleic Acids. Biochem. J. 1951, 48, 581–584. [Google Scholar] [CrossRef]
  20. Tompkins, J.D. Discovering DNA Methylation, the History and Future of the Writing on DNA. J. Hist. Biol. 2022, 55, 865–887. [Google Scholar] [CrossRef]
  21. Deichmann, U. Epigenetics: The Origins and Evolution of a Fashionable Topic. Dev. Biol. 2016, 416, 249–254. [Google Scholar] [CrossRef] [PubMed]
  22. Liao, J.; Karnik, R.; Gu, H.; Ziller, M.J.; Clement, K.; Tsankov, A.M.; Akopian, V.; Gifford, C.A.; Donaghey, J.; Galonska, C.; et al. Targeted Disruption of Dnmt1, Dnmt3a and Dnmt3b in Human Embryonic Stem Cells. Nat. Genet. 2015, 47, 469–478. [Google Scholar] [CrossRef] [PubMed]
  23. Tsumura, A.; Hayakawa, T.; Kumaki, Y.; Takebayashi, S.; Sakaue, M.; Matsuoka, C.; Shimotohno, K.; Ishikawa, F.; Li, E.; Ueda, H.R.; et al. Maintenance of Self-Renewal Ability of Mouse Embryonic Stem Cells in the Absence of DNA Methyltransferases Dnmt1, Dnmt3a and Dnmt3b. Genes Cells 2006, 11, 805–814. [Google Scholar] [CrossRef]
  24. Goll, M.G.; Bestor, T.H. Eukaryotic Cytosine Methyltransferases. Annu. Rev. Biochem. 2005, 74, 481–514. [Google Scholar] [CrossRef]
  25. Kohli, R.M.; Zhang, Y. Tet Enzymes, Tdg and the Dynamics of DNA Demethylation. Nature 2013, 502, 472–479. [Google Scholar] [CrossRef]
  26. Warden, C.D.; Lee, H.; Tompkins, J.D.; Li, X.; Wang, C.; Riggs, A.D.; Yu, H.; Jove, R.; Yuan, Y.-C. Cohcap: An Integrative Genomic Pipeline for Single-Nucleotide Resolution DNA Methylation Analysis. Nucleic Acids Res. 2013, 41, e117. [Google Scholar] [CrossRef] [PubMed]
  27. Lyko, F.; Foret, S.; Kucharski, R.; Wolf, S.; Falckenhayn, C.; Maleszka, R. The Honey Bee Epigenomes: Differential Methylation of Brain DNA in Queens and Workers. PLoS Biol. 2010, 8, e1000506. [Google Scholar] [CrossRef]
  28. Illingworth, R.S.; Gruenewald-Schneider, U.; Webb, S.; Kerr, A.R.W.; James, K.D.; Turner, D.J.; Smith, C.; Harrison, D.J.; Andrews, R.; Bird, A.P. Orphan CpG Islands Identify Numerous Conserved Promoters in the Mammalian Genome. PLoS Genet. 2010, 6, e1001134. [Google Scholar] [CrossRef] [PubMed]
  29. Robertson, K.D. DNA Methylation and Human Disease. Nat. Rev. Genet. 2005, 6, 597–610. [Google Scholar] [CrossRef]
  30. Cohen, N.M.; Kenigsberg, E.; Tanay, A. Primate CpG Islands Are Maintained by Heterogeneous Evolutionary Regimes Involving Minimal Selection. Cell 2011, 145, 773–786. [Google Scholar] [CrossRef]
  31. Branciamore, S.; Chen, Z.-X.; Riggs, A.D.; Rodin, S.N. Cpg Island Clusters and Pro-Epigenetic Selection for Cpgs in Protein-Coding Exons of Hox and Other Transcription Factors. Proc. Natl. Acad. Sci. USA 2010, 107, 15485–15490. [Google Scholar] [CrossRef] [PubMed]
  32. Walter, M.; Teissandier, A.; Pérez-Palacios, R.; Bourc’His, D. An Epigenetic Switch Ensures Transposon Repression Upon Dynamic Loss of DNA Methylation in Embryonic Stem Cells. eLife 2016, 5, e11418. [Google Scholar] [CrossRef]
  33. Song, Y.; Berg, P.R.v.D.; Markoulaki, S.; Soldner, F.; Dall’agnese, A.; Henninger, J.E.; Drotar, J.; Rosenau, N.; Cohen, M.A.; Young, R.A.; et al. Dynamic Enhancer DNA Methylation as Basis for Transcriptional and Cellular Heterogeneity of ESCs. Mol. Cell 2019, 75, 905–920.e6. [Google Scholar] [CrossRef] [PubMed]
  34. Sharifi-Zarchi, A.; Gerovska, D.; Adachi, K.; Totonchi, M.; Pezeshk, H.; Taft, R.J.; Araúzo-Bravo, M.J. DNA Methylation Regulates Discrimination of Enhancers from Promoters through a H3k4me1-H3k4me3 Seesaw Mechanism. BMC Genom. 2017, 18, 964. [Google Scholar] [CrossRef] [PubMed]
  35. Bestor, T.H.; Edwards, J.R.; Boulard, M. Notes on the Role of Dynamic DNA Methylation in Mammalian Development. Proc. Natl. Acad. Sci. USA 2015, 112, 6796–6799. [Google Scholar] [CrossRef]
  36. Belsky, D.W.; Caspi, A.; Corcoran, D.L.; Sugden, K.; Poulton, R.; Arseneault, L.; Moffitt, T.E. Dunedinpace, a DNA Methylation Biomarker of the Pace of Aging. eLife 2022, 11, e73420. [Google Scholar] [CrossRef]
  37. Kim, M.; Long, T.I.; Arakawa, K.; Wang, R.; Yu, M.C.; Laird, P.W. DNA Methylation as a Biomarker for Cardiovascular Disease Risk. PLoS ONE 2010, 5, e9692. [Google Scholar] [CrossRef]
  38. Salameh, Y.; Bejaoui, Y.; El Hajj, N. DNA Methylation Biomarkers in Aging and Age-Related Diseases. Front. Genet. 2020, 11, 171. [Google Scholar] [CrossRef]
  39. Sasaki, M.; Knobbe, C.B.; Munger, J.C.; Lind, E.F.; Brenner, D.; Brüstle, A.; Mak, T.W. Idh1(R132h) Mutation Increases Murine Haematopoietic Progenitors and Alters Epigenetics. Nature 2012, 488, 656–659. [Google Scholar] [CrossRef]
  40. Thompson, M.J.; Rubbi, L.; Dawson, D.W.; Donahue, T.R.; Pellegrini, M. Pancreatic Cancer Patient Survival Correlates with DNA Methylation of Pancreas Development Genes. PLoS ONE 2015, 10, e0128814. [Google Scholar] [CrossRef]
  41. Paweł, K.; Maria Małgorzata, S. Cpg Island Methylator Phenotype-a Hope for the Future or a Road to Nowhere? Int. J. Mol. Sci. 2022, 23, 830. [Google Scholar] [CrossRef]
  42. Couronné, L.; Bastard, C.; Bernard, O.A. Tet2 and Dnmt3a Mutations in Human T-Cell Lymphoma. N. Engl. J. Med. 2012, 366, 95–96. [Google Scholar] [CrossRef] [PubMed]
  43. Tokairin, Y.; Kakinuma, S.; Arai, M.; Nishimura, M.; Okamoto, M.; Ito, E.; Akashi, M.; Miki, Y.; Kawano, T.; Iwai, T.; et al. Accelerated Growth of Intestinal Tumours after Radiation Exposure in Mlh1-Knockout Mice: Evaluation of the Late Effect of Radiation on a Mouse Model of Hnpcc. Int. J. Exp. Pathol. 2006, 87, 89–99. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, C.; Yin, B.; Wei, Q.; Li, D.; Hu, J.; Yu, F.; Lu, Q. Aberrant DNA Methylation in Thymic Epithelial Tumors. Cancer Investig. 2009, 27, 582–591. [Google Scholar] [CrossRef]
  45. Crépin, M.; Dieu, M.-C.; Lejeune, S.; Escande, F.; Boidin, D.; Porchet, N.; Morin, G.; Manouvrier, S.; Mathieu, M.; Buisine, M.-P. Evidence of constitutional MLH1 epimutation associated to transgenerational inheritance of cancer susceptibility. Hum. Mutat. 2012, 33, 180–188. [Google Scholar] [CrossRef]
  46. Tompkins, J.D.; Wu, X.; Her, C. Muts Homologue Hmsh5: Role in Cisplatin-Induced DNA Damage Response. Mol. Cancer 2012, 11, 10. [Google Scholar] [CrossRef]
  47. Xu, K.; Wu, X.; Tompkins, J.D.; Her, C. Assessment of Anti-Recombination and Double-Strand Break-Induced Gene Conversion in Human Cells by a Chromosomal Reporter. J. Biol. Chem. 2012, 287, 29543–29553. [Google Scholar] [CrossRef] [PubMed]
  48. Wei, Y.; Sun, Z.; Wang, Y.; Xie, Z.; Xu, S.; Xu, Y.; Zhou, X.; Bi, J.; Zhu, Z. Methylation in the Tp53 Promoter Is Associated with Ischemic Stroke. Mol. Med. Rep. 2019, 20, 1404–1410. [Google Scholar] [CrossRef]
  49. Poosari, A.; Nutravong, T.; Namwat, W.; Wasenang, W.; Sa-Ngiamwibool, P.; Ungareewittaya, P. The Relationship between P16(Ink4a) and Tp53 Promoter Methylation and the Risk and Prognosis in Patients with Oesophageal Cancer in Thailand. Sci. Rep. 2022, 12, 10337. [Google Scholar] [CrossRef]
  50. Awasthi, S.; Tompkins, J.; Singhal, J.; Riggs, A.D.; Yadav, S.; Wu, X.; Singh, S.; Warden, C.; Liu, Z.; Wang, J.; et al. Rlip Depletion Prevents Spontaneous Neoplasia in Tp53 Null Mice. Proc. Natl. Acad. Sci. USA 2018, 115, 3918–3923. [Google Scholar] [CrossRef]
  51. Fertan, E.; Gendron, W.H.; Wong, A.A.; Hanson, G.M.; Brown, R.E.; Weaver, I.C. Noncanonical Regulation of Imprinted Gene Igf2 by Amyloid-Beta 1–42 in Alzheimer’s Disease. Sci. Rep. 2023, 13, 2043. [Google Scholar] [CrossRef] [PubMed]
  52. Pai, S.; Li, P.; Killinger, B.; Marshall, L.; Jia, P.; Liao, J.; Petronis, A.; Szabó, P.E.; Labrie, V. Differential Methylation of Enhancer at Igf2 Is Associated with Abnormal Dopamine Synthesis in Major Psychosis. Nat. Commun. 2019, 10, 2046. [Google Scholar] [CrossRef] [PubMed]
  53. Serra-Juhé, C.; Cuscó, I.; Homs, A.; Flores, R.; Torán, N.; Pérez-Jurado, L.A. DNA Methylation Abnormalities in Congenital Heart Disease. Epigenetics 2015, 10, 167–177. [Google Scholar] [CrossRef] [PubMed]
  54. Lang, A.-L.; Eulalio, T.; Fox, E.; Yakabi, K.; Bukhari, S.A.; Kawas, C.H.; Corrada, M.M.; Montgomery, S.B.; Heppner, F.L.; Capper, D.; et al. Methylation Differences in Alzheimer’s Disease Neuropathologic Change in the Aged Human Brain. Acta Neuropathol. Commun. 2022, 10, 174. [Google Scholar] [CrossRef] [PubMed]
  55. Desiderio, A.; Longo, M.; Parrillo, L.; Campitelli, M.; Cacace, G.; de Simone, S.; Spinelli, R.; Zatterale, F.; Cabaro, S.; Dolce, P.; et al. Epigenetic Silencing of the Ankrd26 Gene Correlates to the Pro-Inflammatory Profile and Increased Cardio-Metabolic Risk Factors in Human Obesity. Clin. Epigenet. 2019, 11, 181. [Google Scholar] [CrossRef] [PubMed]
  56. Nishioka, M.; Bundo, M.; Kasai, K.; Iwamoto, K. DNA Methylation in Schizophrenia: Progress and Challenges of Epigenetic Studies. Genome Med. 2012, 4, 96–109. [Google Scholar] [CrossRef]
  57. Jiang, X.-Y.; Feng, Y.-L.; Ye, L.-T.; Li, X.-H.; Feng, J.; Zhang, M.-Z.; Shelat, H.S.; Wassler, M.; Li, Y.; Geng, Y.-J.; et al. Inhibition of Gata4 and Tbx5 by Nicotine-Mediated DNA Methylation in Myocardial Differentiation. Stem Cell Rep. 2017, 8, 290–304. [Google Scholar] [CrossRef]
  58. Takahashi, Y.; Valencia, M.M.; Yu, Y.; Ouchi, Y.; Takahashi, K.; Shokhirev, M.N.; Lande, K.; Williams, A.E.; Fresia, C.; Kurita, M.; et al. Transgenerational Inheritance of Acquired Epigenetic Signatures at Cpg Islands in Mice. Cell 2023, 186, 715–731.e19. [Google Scholar] [CrossRef]
  59. Ghaznavi, H.; Mahmoodi, K.; Soltanpour, M.S. A Preliminary Study of the Association between the Abca1 Gene Promoter DNA Methylation and Coronary Artery Disease Risk. Mol. Biol. Res. Commun. 2018, 7, 59–65. [Google Scholar] [CrossRef]
  60. Wojcik, A.J.; Skaflen, M.D.; Srinivasan, S.; Hedrick, C.C. A Critical Role for Abcg1 in Macrophage Inflammation and Lung Homeostasis. J. Immunol. 2008, 180, 4273–4282. [Google Scholar] [CrossRef]
  61. Horvath, S. DNA Methylation Age of Human Tissues and Cell Types. Genome Biol. 2013, 14, 3156. [Google Scholar] [CrossRef]
  62. Horvath, S.; Raj, K. DNA Methylation-Based Biomarkers and the Epigenetic Clock Theory of Ageing. Nat. Rev. Genet. 2018, 19, 371–384. [Google Scholar] [CrossRef] [PubMed]
  63. Bell, C.G.; Lowe, R.; Adams, P.D.; Baccarelli, A.A.; Beck, S.; Bell, J.T.; Christensen, B.C.; Gladyshev, V.N.; Heijmans, B.T.; Horvath, S.; et al. DNA Methylation Aging Clocks: Challenges and Recommendations. Genome Biol. 2019, 20, 249. [Google Scholar] [CrossRef] [PubMed]
  64. Lu, A.T.; Fei, Z.; Haghani, A.; Robeck, T.R.; Zoller, J.A.; Li, C.Z.; Lowe, R.; Yan, Q.; Zhang, J.; Vu, H.; et al. Universal DNA Methylation Age across Mammalian Tissues. Nat. Aging 2023, 3, 1144–1166. [Google Scholar] [CrossRef]
  65. Lawson, K.A.; Hage, W.J. Clonal Analysis of the Origin of Primordial Germ Cells in the Mouse. Ciba Found Symp. 1994, 182, 68–84; discussion 84–91. [Google Scholar]
  66. Kumar, D.L.; DeFalco, T. Of Mice and Men: In Vivo and In Vitro Studies of Primordial Germ Cell Specification. Semin. Reprod. Med. 2017, 35, 139–146. [Google Scholar] [CrossRef] [PubMed]
  67. Kobayashi, H.; Sakurai, T.; Miura, F.; Imai, M.; Mochiduki, K.; Yanagisawa, E.; Sakashita, A.; Wakai, T.; Suzuki, Y.; Ito, T.; et al. High-Resolution DNA Methylome Analysis of Primordial Germ Cells Identifies Gender-Specific Reprogramming in Mice. Genome Res. 2013, 23, 616–627. [Google Scholar] [CrossRef]
  68. Yi, W.; Lee, T.-H.; Tompkins, J.D.; Zhu, F.; Wu, X.; Her, C. Physical and Functional Interaction between Hmsh5 and C-Abl. Cancer Res. 2006, 66, 151–158. [Google Scholar] [CrossRef]
  69. Her, C.; Zhao, N.; Wu, X.; Tompkins, J.D. Muts Homologues Hmsh4 and Hmsh5: Diverse Functional Implications in Humans. Front. Biosci. 2007, 12, 905–911. [Google Scholar] [CrossRef]
  70. Wu, X.; Xu, Y.; Feng, K.; Tompkins, J.D.; Her, C. Muts Homologue Hmsh5: Recombinational Dsb Repair and Non-Synonymous Polymorphic Variants. PLoS ONE 2013, 8, e73284. [Google Scholar] [CrossRef]
  71. Pei, Z.; Deng, K.; Xu, C.; Zhang, S. The Molecular Regulatory Mechanisms of Meiotic Arrest and Resumption in Oocyte Development and Maturation. Reprod. Biol. Endocrinol. 2023, 21, 90. [Google Scholar] [CrossRef]
  72. Kobayashi, H.; Sakurai, T.; Imai, M.; Takahashi, N.; Fukuda, A.; Yayoi, O.; Sato, S.; Nakabayashi, K.; Hata, K.; Sotomaru, Y.; et al. Contribution of Intragenic DNA Methylation in Mouse Gametic DNA Methylomes to Establish Oocyte-Specific Heritable Marks. PLoS Genet. 2012, 8, e1002440. [Google Scholar] [CrossRef] [PubMed]
  73. Nakamura, T.; Liu, Y.-J.; Nakashima, H.; Umehara, H.; Inoue, K.; Matoba, S.; Tachibana, M.; Ogura, A.; Shinkai, Y.; Nakano, T. Pgc7 Binds Histone H3k9me2 to Protect against Conversion of 5mc to 5hmc in Early Embryos. Nature 2012, 486, 415–419. [Google Scholar] [CrossRef] [PubMed]
  74. Quenneville, S.; Gaetano, V.; Andrea, C.; Adamandia, K.; Johan, J.; Sandra, O.; Ilaria, B.; Paolo, V.P.; Giovanna, G.; Andrea, R.; et al. In Embryonic Stem Cells, Zfp57/Kap1 Recognize a Methylated Hexanucleotide to Affect Chromatin and DNA Methylation of Imprinting Control Regions. Mol. Cell 2011, 44, 361–372. [Google Scholar] [CrossRef] [PubMed]
  75. Hirasawa, R.; Chiba, H.; Kaneda, M.; Tajima, S.; Li, E.; Jaenisch, R.; Sasaki, H. Maternal and Zygotic Dnmt1 Are Necessary and Sufficient for the Maintenance of DNA Methylation Imprints During Preimplantation Development. Genes Dev. 2008, 22, 1607–1616. [Google Scholar] [CrossRef]
  76. Li, X.; Ito, M.; Zhou, F.; Youngson, N.; Zuo, X.; Leder, P.; Ferguson-Smith, A.C. A Maternal-Zygotic Effect Gene, Zfp57, Maintains Both Maternal and Paternal Imprints. Dev. Cell 2008, 15, 547–557. [Google Scholar] [CrossRef]
  77. Pastor, W.A.; Chen, D.; Liu, W.; Kim, R.; Sahakyan, A.; Lukianchikov, A.; Plath, K.; Jacobsen, S.E.; Clark, A.T. Naive Human Pluripotent Cells Feature a Methylation Landscape Devoid of Blastocyst or Germline Memory. Cell Stem Cell 2016, 18, 323–329. [Google Scholar] [CrossRef]
  78. Skinner, M.K. What Is an Epigenetic Transgenerational Phenotype? F3 or F2. Reprod. Toxicol. 2008, 25, 2–6. [Google Scholar] [CrossRef] [PubMed]
  79. Zheng, X.; Chen, L.; Li, M.; Lou, Q.; Xia, H.; Wang, P.; Li, T.; Liu, H.; Luo, L. Transgenerational Variations in DNA Methylation Induced by Drought Stress in Two Rice Varieties with Distinguished Difference to Drought Resistance. PLoS ONE 2013, 8, e80253. [Google Scholar] [CrossRef]
  80. Cubas, P.; Vincent, C.; Coen, E. An Epigenetic Mutation Responsible for Natural Variation in Floral Symmetry. Nature 1999, 401, 157–161. [Google Scholar] [CrossRef]
  81. Buescher, J.L.; Musselman, L.P.; Wilson, C.A.; Lang, T.; Keleher, M.; Baranski, T.J.; Duncan, J.G. Evidence for Transgenerational Metabolic Programming in Drosophila. Dis. Models Mech. 2013, 6, 1123–1132. [Google Scholar] [CrossRef]
  82. Xia, B.; de Belle, J.S. Transgenerational Programming of Longevity and Reproduction by Post-Eclosion Dietary Manipulation in Drosophila. Aging 2016, 8, 1115–1134. [Google Scholar] [CrossRef]
  83. Carvan, M.J., 3rd; Kalluvila, T.A.; Klingler, R.H.; Larson, J.K.; Pickens, M.; Mora-Zamorano, F.X.; Connaughton, V.P.; Sadler-Riggleman, I.; Beck, D.; Skinner, M.K. Mercury-Induced Epigenetic Transgenerational Inheritance of Abnormal Neurobehavior Is Correlated with Sperm Epimutations in Zebrafish. PLoS ONE 2017, 12, e0176155. [Google Scholar] [CrossRef]
  84. Baker, T.R.; Peterson, R.E.; Heideman, W. Using Zebrafish as a Model System for Studying the Transgenerational Effects of Dioxin. Toxicol. Sci. 2014, 138, 403–411. [Google Scholar] [CrossRef] [PubMed]
  85. Leroux, S.; Gourichon, D.; Leterrier, C.; Labrune, Y.; Coustham, V.; Rivière, S.; Zerjal, T.; Coville, J.L.; Morisson, M.; Minvielle, F.; et al. Embryonic Environment and Transgenerational Effects in Quail. Genet. Sel. Evol. 2017, 49, 14. [Google Scholar] [CrossRef] [PubMed]
  86. Brun, J.M.; Bernadet, M.D.; Cornuez, A.; Leroux, S.; Bodin, L.; Basso, B.; Davail, S.; Jaglin, M.; Lessire, M.; Martin, X.; et al. Influence of Grand-Mother Diet on Offspring Performances through the Male Line in Muscovy Duck. BMC Genet. 2015, 16, 145. [Google Scholar] [CrossRef]
  87. Buchwald, U.; Teupser, D.; Kuehnel, F.; Grohmann, J.; Schmieder, N.; Beindorff, N.; Schlumbohm, C.; Fuhrmann, H.; Einspanier, A. Prenatal Stress Programs Lipid Metabolism Enhancing Cardiovascular Risk in the Female F1, F2, and F3 Generation in the Primate Model Common Marmoset (Callithrix jacchus). J. Med. Primatol. 2012, 41, 231–240. [Google Scholar] [CrossRef]
  88. Veenendaal, M.; Painter, R.; de Rooij, S.; Bossuyt, P.; van der Post, J.; Gluckman, P.; Hanson, M.; Roseboom, T. Transgenerational Effects of Prenatal Exposure to the 1944-45 Dutch Famine. BJOG Int. J. Obstet. Gynaecol. 2013, 120, 548–554. [Google Scholar] [CrossRef]
  89. Bygren, L.O.; Tinghög, P.; Carstensen, J.; Edvinsson, S.; Kaati, G.; E Pembrey, M.; Sjöström, M. Change in Paternal Grandmothers’ Early Food Supply Influenced Cardiovascular Mortality of the Female Grandchildren. BMC Genet. 2014, 15, 12. [Google Scholar] [CrossRef] [PubMed]
  90. Anway, M.D.; Cupp, A.S.; Uzumcu, M.; Skinner, M.K. Epigenetic Transgenerational Actions of Endocrine Disruptors and Male Fertility. Science 2005, 308, 1466–1469. [Google Scholar] [CrossRef]
  91. Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Pesticide and Insect Repellent Mixture (Permethrin and Deet) Induces Epigenetic Transgenerational Inheritance of Disease and Sperm Epimutations. Reprod. Toxicol. 2012, 34, 708–719. [Google Scholar] [CrossRef]
  92. Chamorro-García, R.; Sahu, M.; Abbey, R.J.; Laude, J.; Pham, N.; Blumberg, B. Transgenerational Inheritance of Increased Fat Depot Size, Stem Cell Reprogramming, and Hepatic Steatosis Elicited by Prenatal Exposure to the Obesogen Tributyltin in Mice. Environ. Health Perspect. 2013, 121, 359–366. [Google Scholar] [CrossRef] [PubMed]
  93. Kubsad, D.; Nilsson, E.E.; King, S.E.; Sadler-Riggleman, I.; Beck, D.; Skinner, M.K. Assessment of Glyphosate Induced Epigenetic Transgenerational Inheritance of Pathologies and Sperm Epimutations: Generational Toxicology. Sci. Rep. 2019, 9, 6372. [Google Scholar] [CrossRef] [PubMed]
  94. Van Cauwenbergh, O.; Di Serafino, A.; Tytgat, J.; Soubry, A. Transgenerational Epigenetic Effects from Male Exposure to Endocrine-Disrupting Compounds: A Systematic Review on Research in Mammals. Clin. Epigenet. 2020, 12, 65. [Google Scholar] [CrossRef] [PubMed]
  95. Liu, J.; Yu, C.; Doherty, T.M.; Akbari, O.; Allard, P.; Rehan, V.K. Perinatal Nicotine Exposure-Induced Transgenerational Asthma: Effects of Reexposure in F1 Gestation. FASEB J. 2020, 34, 11444–11459. [Google Scholar] [CrossRef]
  96. Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Plastics Derived Endocrine Disruptors (BPA, DEHP and DBP) Induce Epigenetic Transgenerational Inheritance of Obesity, Reproductive Disease and Sperm Epimutations. PLoS ONE 2013, 8, e55387. [Google Scholar] [CrossRef]
  97. Wolstenholme, J.T.; Edwards, M.; Shetty, S.R.J.; Gatewood, J.D.; Taylor, J.A.; Rissman, E.F.; Connelly, J.J. Gestational Exposure to Bisphenol A Produces Transgenerational Changes in Behaviors and Gene Expression. Endocrinology 2012, 153, 3828–3838. [Google Scholar] [CrossRef]
  98. Thorson, J.L.M.; Beck, D.; Ben Maamar, M.; Nilsson, E.E.; Skinner, M.K. Ancestral Plastics Exposure Induces Transgenerational Disease-Specific Sperm Epigenome-Wide Association Biomarkers. Environ. Epigenet. 2021, 7, dvaa023. [Google Scholar] [CrossRef]
  99. van Steenwyk, G.; Roszkowski, M.; Manuella, F.; Franklin, T.B.; Mansuy, I.M. Transgenerational Inheritance of Behavioral and Metabolic Effects of Paternal Exposure to Traumatic Stress in Early Postnatal Life: Evidence in the 4th Generation. Environ. Epigenet. 2018, 4, dvy023. [Google Scholar] [CrossRef]
  100. Jawaid, A.; Roszkowski, M.; Mansuy, I.M. Transgenerational Epigenetics of Traumatic Stress. Prog. Mol. Biol. Transl. Sci. 2018, 158, 273–298. [Google Scholar]
  101. Nilsson, E.E.; Ben Maamar, M.; Skinner, M.K. Role of Epigenetic Transgenerational Inheritance in Generational Toxicology. Environ. Epigenet. 2022, 8, dvac001. [Google Scholar] [CrossRef] [PubMed]
  102. King, S.E.; Skinner, M.K. Epigenetic Transgenerational Inheritance of Obesity Susceptibility. Trends Endocrinol. Metab. 2020, 31, 478–494. [Google Scholar] [CrossRef]
  103. Bošković, A.; Rando, O.J. Transgenerational Epigenetic Inheritance. Annu. Rev. Genet. 2018, 52, 21–41. [Google Scholar] [CrossRef] [PubMed]
  104. Skinner, M.K.; Ben Maamar, M.; Sadler-Riggleman, I.; Beck, D.; Nilsson, E.; McBirney, M.; Klukovich, R.; Xie, Y.; Tang, C.; Yan, W. Alterations in Sperm DNA Methylation, Non-Coding Rna and Histone Retention Associate with Ddt-Induced Epigenetic Transgenerational Inheritance of Disease. Epigenet. Chromatin 2018, 11, 8. [Google Scholar] [CrossRef]
  105. Northstone, K.; Golding, J.; Smith, G.D.; Miller, L.L.; Pembrey, M. Prepubertal Start of Father’s Smoking and Increased Body Fat in His Sons: Further Characterisation of Paternal Transgenerational Responses. Eur. J. Hum. Genet. 2014, 22, 1382–1386. [Google Scholar] [CrossRef] [PubMed]
  106. Vaag, A.A.; Grunnet, L.G.; Arora, G.P.; Brøns, C. The Thrifty Phenotype Hypothesis Revisited. Diabetologia 2012, 55, 2085–2088. [Google Scholar] [CrossRef]
  107. Hales, C.N.; Barker, D.J. The Thrifty Phenotype Hypothesis. Br. Med. Bull. 2001, 60, 5–20. [Google Scholar] [CrossRef]
  108. Fullston, T.; Ohlsson Teague, E.M.; Palmer, N.O.; DeBlasio, M.J.; Mitchell, M.; Corbett, M.; Print, C.G.; Owens, J.A.; Lane, M. Paternal Obesity Initiates Metabolic Disturbances in Two Generations of Mice with Incomplete Penetrance to the F2 Generation and Alters the Transcriptional Profile of Testis and Sperm Microrna Content. FASEB J. 2013, 27, 4226–4243. [Google Scholar] [CrossRef]
  109. Chambers, T.J.G.; Morgan, M.D.; Heger, A.H.; Sharpe, R.M.; Drake, A.J. High-Fat Diet Disrupts Metabolism in Two Generations of Rats in a Parent-of-Origin Specific Manner. Sci. Rep. 2016, 6, 31857. [Google Scholar] [CrossRef]
  110. Dunn, G.A.; Bale, T.L. Maternal High-Fat Diet Effects on Third-Generation Female Body Size via the Paternal Lineage. Endocrinology 2011, 152, 2228–2236. [Google Scholar] [CrossRef]
  111. Skinner, M.K.; Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Haque, M.; Nilsson, E.E. Ancestral Dichlorodiphenyltrichloroethane (Ddt) Exposure Promotes Epigenetic Transgenerational Inheritance of Obesity. BMC Med. 2013, 11, 228. [Google Scholar] [CrossRef]
  112. Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Dioxin (TCDD) Induces Epigenetic Transgenerational Inheritance of Adult Onset Disease and Sperm Epimutations. PLoS ONE 2012, 7, e46249. [Google Scholar] [CrossRef] [PubMed]
  113. Ma, J.; Chen, X.; Liu, Y.; Xie, Q.; Sun, Y.; Chen, J.; Leng, L.; Yan, H.; Zhao, B.; Tang, N. Ancestral Tcdd Exposure Promotes Epigenetic Transgenerational Inheritance of Imprinted Gene Igf2: Methylation Status and Dnmts. Toxicol. Appl. Pharmacol. 2015, 289, 193–202. [Google Scholar] [CrossRef]
  114. Ding, T.; Mokshagundam, S.; Rinaudo, P.F.; Osteen, K.G.; Bruner-Tran, K.L. Paternal Developmental Toxicant Exposure Is Associated with Epigenetic Modulation of Sperm and Placental Pgr and Igf2 in a Mouse Model. Biol. Reprod. 2018, 99, 864–876. [Google Scholar] [CrossRef] [PubMed]
  115. Guerrero-Bosagna, C.; Settles, M.; Lucker, B.; Skinner, M.K. Epigenetic Transgenerational Actions of Vinclozolin on Promoter Regions of the Sperm Epigenome. PLoS ONE 2010, 5, e13100. [Google Scholar] [CrossRef] [PubMed]
  116. Manikkam, M.; Guerrero-Bosagna, C.; Tracey, R.; Haque, M.; Skinner, M.K. Transgenerational Actions of Environmental Compounds on Reproductive Disease and Identification of Epigenetic Biomarkers of Ancestral Exposures. PLoS ONE 2012, 7, e31901. [Google Scholar] [CrossRef]
  117. Ben Maamar, M.; Wang, Y.; E Nilsson, E.; Beck, D.; Yan, W.; Skinner, M.K. Transgenerational Sperm Dmrs Escape DNA Methylation Erasure during Embryonic Development And epigenetic Inheritance. Environ. Epigenet. 2023, 9, dvad003. [Google Scholar] [CrossRef]
  118. Levine, H.; Jørgensen, N.; Martino-Andrade, A.; Mendiola, J.; Weksler-Derri, D.; Jolles, M.; Pinotti, R.; Swan, S.H. Temporal Trends in Sperm Count: A Systematic Review and Meta-Regression Analysis of Samples Collected Globally in the 20th and 21st Centuries. Hum. Reprod. Update 2022, 29, 157–176. [Google Scholar] [CrossRef]
  119. Kumar, N.; Singh, A.K. Trends of Male Factor Infertility, an Important Cause of Infertility: A Review of Literature. J. Hum. Reprod. Sci. 2015, 8, 191–196. [Google Scholar] [CrossRef]
  120. Santillan, D.A.; Theisler, C.M.; Ryan, A.S.; Popovic, R.; Stuart, T.; Zhou, M.M.; Alkan, S.; Zeleznik-Le, N.J. Bromodomain and Histone Acetyltransferase Domain Specificities Control Mixed Lineage Leukemia Phenotype. Cancer Res. 2006, 66, 10032–10039. [Google Scholar] [CrossRef]
  121. Chaikind, B.; Kilambi, K.P.; Gray, J.J.; Ostermeier, M. Targeted DNA Methylation Using an Artificially Bisected M. Hhai Fused to Zinc Fingers. PLoS ONE 2012, 7, e44852. [Google Scholar] [CrossRef] [PubMed]
  122. Bernstein, D.L.; Le Lay, J.E.; Ruano, E.G.; Kaestner, K.H. Tale-Mediated Epigenetic Suppression of Cdkn2a Increases Replication in Human Fibroblasts. J. Clin. Investig. 2015, 125, 1998–2006. [Google Scholar] [CrossRef]
  123. Qi, L.S.; Larson, M.H.; Gilbert, L.A.; Doudna, J.A.; Weissman, J.S.; Arkin, A.P.; Lim, W.A. Repurposing Crispr as an Rna-Guided Platform for Sequence-Specific Control of Gene Expression. Cell 2013, 152, 1173–1183. [Google Scholar] [CrossRef] [PubMed]
  124. Xu, X.; Qi, L.S. A CRISPR–dCas Toolbox for Genetic Engineering and Synthetic Biology. J. Mol. Biol. 2019, 431, 34–47. [Google Scholar] [CrossRef]
  125. Adli, M. The Crispr Tool Kit for Genome Editing and Beyond. Nat. Commun. 2018, 9, 1911. [Google Scholar] [CrossRef]
  126. Brezgin, S.; Kostyusheva, A.; Kostyushev, D.; Chulanov, V. Dead Cas Systems: Types, Principles, and Applications. Int. J. Mol. Sci. 2019, 20, 6041. [Google Scholar] [CrossRef]
  127. Vojta, A.; Dobrinić, P.; Tadić, V.; Bočkor, L.; Korać, P.; Julg, B.; Klasić, M.; Zoldoš, V. Repurposing the Crispr-Cas9 System for Targeted DNA Methylation. Nucleic Acids Res. 2016, 44, 5615–5628. [Google Scholar] [CrossRef] [PubMed]
  128. Kearns, N.A.; Pham, H.; Tabak, B.; Genga, R.M.; Silverstein, N.J.; Garber, M.; Maehr, R. Functional Annotation of Native Enhancers with a Cas9-Histone Demethylase Fusion. Nat. Methods 2015, 12, 401–403. [Google Scholar] [CrossRef]
  129. Hilton, I.B.; D’Ippolito, A.M.; Vockley, C.M.; I Thakore, P.; E Crawford, G.; E Reddy, T.; A Gersbach, C. Epigenome Editing by a Crispr-Cas9-Based Acetyltransferase Activates Genes from Promoters and Enhancers. Nat. Biotechnol. 2015, 33, 510–517. [Google Scholar] [CrossRef]
  130. Stepper, P.; Kungulovski, G.; Jurkowska, R.Z.; Chandra, T.; Krueger, F.; Reinhardt, R.; Reik, W.; Jeltsch, A.; Jurkowski, T.P. Efficient Targeted DNA Methylation with Chimeric Dcas9-Dnmt3a-Dnmt3l Methyltransferase. Nucleic Acids Res. 2017, 45, 1703–1713. [Google Scholar] [CrossRef]
  131. McDonald, J.I.; Celik, H.; Rois, L.E.; Fishberger, G.; Fowler, T.; Rees, R.; Kramer, A.; Martens, A.; Edwards, J.R.; Challen, G.A. Reprogrammable Crispr/Cas9-Based System for Inducing Site-Specific DNA Methylation. Biol. Open 2016, 5, 866–874. [Google Scholar] [CrossRef] [PubMed]
  132. Yeo, N.C.; Chavez, A.; Lance-Byrne, A.; Chan, Y.; Menn, D.; Milanova, D.; Kuo, C.-C.; Guo, X.; Sharma, S.; Tung, A.; et al. An Enhanced Crispr Repressor for Targeted Mammalian Gene Regulation. Nat. Methods 2018, 15, 611–616. [Google Scholar] [CrossRef] [PubMed]
  133. Nuñez, J.K.; Chen, J.; Pommier, G.C.; Cogan, J.Z.; Replogle, J.M.; Adriaens, C.; Ramadoss, G.N.; Shi, Q.; Hung, K.L.; Samelson, A.J.; et al. Genome-Wide Programmable Transcriptional Memory by Crispr-Based Epigenome Editing. Cell 2021, 184, 2503–2519.e17. [Google Scholar] [CrossRef]
  134. Amabile, A.; Migliara, A.; Capasso, P.; Biffi, M.; Cittaro, D.; Naldini, L.; Lombardo, A. Inheritable Silencing of Endogenous Genes by Hit-and-Run Targeted Epigenetic Editing. Cell 2016, 167, 219–232.e14. [Google Scholar] [CrossRef] [PubMed]
  135. Pflueger, C.; Tan, D.; Swain, T.; Nguyen, T.; Pflueger, J.; Nefzger, C.; Polo, J.M.; Ford, E.; Lister, R. A Modular Dcas9-Suntag Dnmt3a Epigenome Editing System Overcomes Pervasive Off-Target Activity of Direct Fusion Dcas9-Dnmt3a Constructs. Genome Res. 2018, 28, 1193–1206. [Google Scholar] [CrossRef]
  136. Chavez, A.; Scheiman, J.; Vora, S.; Pruitt, B.W.; Tuttle, M.; Iyer, E.P.R.; Lin, S.; Kiani, S.; Guzman, C.D.; Wiegand, D.J.; et al. Highly Efficient Cas9-Mediated Transcriptional Programming. Nat. Methods 2015, 12, 326–328. [Google Scholar] [CrossRef]
  137. Konermann, S.; Brigham, M.D.; Trevino, A.E.; Joung, J.; Abudayyeh, O.O.; Barcena, C.; Hsu, P.D.; Habib, N.; Gootenberg, J.S.; Nishimasu, H.; et al. Genome-Scale Transcriptional Activation by an Engineered Crispr-Cas9 Complex. Nature 2015, 517, 583–588. [Google Scholar] [CrossRef] [PubMed]
  138. Gemberling, M.P.; Siklenka, K.; Rodriguez, E.; Tonn-Eisinger, K.R.; Barrera, A.; Liu, F.; Kantor, A.; Li, L.; Cigliola, V.; Hazlett, M.F.; et al. Transgenic Mice for In Vivo Epigenome Editing with Crispr-Based Systems. Nat. Methods 2021, 18, 965–974. [Google Scholar] [CrossRef] [PubMed]
  139. Thakore, P.I.; Kwon, J.B.; Nelson, C.E.; Rouse, D.C.; Gemberling, M.P.; Oliver, M.L.; Gersbach, C.A. Rna-Guided Transcriptional Silencing In Vivo with S. aureus Crispr-Cas9 Repressors. Nat. Commun. 2018, 9, 1674. [Google Scholar] [CrossRef]
  140. Saunderson, E.A.; Encabo, H.H.; Devis, J.; Rouault-Pierre, K.; Piganeau, M.; Bell, C.G.; Gribben, J.G.; Bonnet, D.; Ficz, G. Crispr/Dcas9 DNA Methylation Editing Is Heritable during Human Hematopoiesis and Shapes Immune Progeny. Proc. Natl. Acad. Sci. USA 2023, 120, e2300224120. [Google Scholar] [CrossRef]
  141. Zhou, H.; Liu, J.; Zhou, C.; Gao, N.; Rao, Z.; Li, H.; Hu, X.; Li, C.; Yao, X.; Shen, X.; et al. In Vivo Simultaneous Transcriptional Activation of Multiple Genes in the Brain Using Crispr-Dcas9-Activator Transgenic Mice. Nat. Neurosci. 2018, 21, 440–446. [Google Scholar] [CrossRef] [PubMed]
  142. Wangensteen, K.J.; Wang, Y.J.; Dou, Z.; Wang, A.W.; Mosleh-Shirazi, E.; Horlbeck, M.A.; Gilbert, L.A.; Weissman, J.S.; Berger, S.L.; Kaestner, K.H. Combinatorial Genetics in Liver Repopulation and Carcinogenesis with a In Vivo Crispr Activation Platform. Hepatology 2018, 68, 663–676. [Google Scholar] [CrossRef] [PubMed]
  143. Gough, V.; Gersbach, C.A. Immunity to Cas9 as an Obstacle to Persistent Genome Editing. Mol. Ther. 2020, 28, 1389–1391. [Google Scholar] [CrossRef] [PubMed]
  144. Li, A.; Tanner, M.R.; Lee, C.M.; Hurley, A.E.; De Giorgi, M.; Jarrett, K.E.; Davis, T.H.; Doerfler, A.M.; Bao, G.; Beeton, C.; et al. Aav-Crispr Gene Editing Is Negated by Pre-Existing Immunity to Cas9. Mol. Ther. 2020, 28, 1432–1441. [Google Scholar] [CrossRef]
  145. Kim, K.; Doi, A.; Wen, B.; Ng, K.; Zhao, R.; Cahan, P.; Kim, J.; Aryee, M.J.; Ji, H.; Ehrlich, L.I.; et al. Epigenetic Memory in Induced Pluripotent Stem Cells. Nature 2010, 467, 285–290. [Google Scholar] [CrossRef] [PubMed]
  146. Matoba, S.; Zhang, Y. Somatic Cell Nuclear Transfer Reprogramming: Mechanisms and Applications. Cell Stem Cell 2018, 23, 471–485. [Google Scholar] [CrossRef]
  147. Dean, W.; Santos, F.; Stojkovic, M.; Zakhartchenko, V.; Walter, J.; Wolf, E.; Reik, W. Conservation of Methylation Reprogramming in Mammalian Development: Aberrant Reprogramming in Cloned Embryos. Proc. Natl. Acad. Sci. USA 2001, 98, 13734–13738. [Google Scholar] [CrossRef]
  148. Kishigami, S.; Bui, H.T.; Wakayama, S.; Tokunaga, K.; Van Thuan, N.; Hikichi, T.; Mizutani, E.; Ohta, H.; Suetsugu, R.; Sata, T.; et al. Successful Mouse Cloning of an Outbred Strain by Trichostatin a Treatment after Somatic Nuclear Transfer. J. Reprod. Dev. 2007, 53, 165–170. [Google Scholar] [CrossRef]
  149. Akagi, S.; Matsukawa, K.; Mizutani, E.; Fukunari, K.; Kaneda, M.; Watanabe, S.; Takahashi, S. Treatment with a Histone Deacetylase Inhibitor after Nuclear Transfer Improves the Preimplantation Development of Cloned Bovine Embryos. J. Reprod. Dev. 2011, 57, 120–126. [Google Scholar] [CrossRef]
  150. Wee, G.; Shim, J.J.; Koo, D.B.; Chae, J.I.; Lee, K.K.; Han, Y.M. Epigenetic Alteration of the Donor Cells Does Not Recapitulate the Reprogramming of DNA Methylation in Cloned Embryos. Reproduction 2007, 134, 781–787. [Google Scholar] [CrossRef]
  151. Ding, X.; Wang, Y.; Zhang, D.; Guo, Z.; Zhang, Y. Increased Pre-Implantation Development of Cloned Bovine Embryos Treated with 5-Aza-2’-Deoxycytidine and Trichostatin A. Theriogenology 2008, 70, 622–630. [Google Scholar] [CrossRef] [PubMed]
  152. Simmet, K.; Wolf, E.; Zakhartchenko, V. Manipulating the Epigenome in Nuclear Transfer Cloning: Where, When and How. Int. J. Mol. Sci. 2020, 22, 236. [Google Scholar] [CrossRef]
  153. Wei, Y.; Lang, J.; Zhang, Q.; Yang, C.-R.; Zhao, Z.-A.; Zhang, Y.; Du, Y.; Sun, Y. DNA Methylation Analysis and Editing in Single Mammalian Oocytes. Proc. Natl. Acad. Sci. USA 2019, 116, 9883–9892. [Google Scholar] [CrossRef] [PubMed]
  154. Lei, Y.; Zhang, X.; Su, J.; Jeong, M.; Gundry, M.C.; Huang, Y.-H.; Zhou, Y.; Li, W.; Goodell, M.A. Targeted DNA Methylation in Vivo Using an Engineered Dcas9-Mq1 Fusion Protein. Nat. Commun. 2017, 8, 16026. [Google Scholar] [CrossRef] [PubMed]
  155. Singh, P.; Lee, D.-H.; Szabó, P.E. More Than Insulator: Multiple Roles of Ctcf at the H19-Igf2 Imprinted Domain. Front. Genet. 2012, 3, 214. [Google Scholar] [CrossRef]
  156. Kojima-Kita, K.; Kuramochi-Miyagawa, S.; Nagamori, I.; Ogonuki, N.; Ogura, A.; Hasuwa, H.; Akazawa, T.; Inoue, N.; Nakano, T. Miwi2 as an Effector of DNA Methylation and Gene Silencing in Embryonic Male Germ Cells. Cell Rep. 2016, 16, 2819–2828. [Google Scholar] [CrossRef] [PubMed]
  157. Das, P.P.; Hendrix, D.A.; Apostolou, E.; Buchner, A.H.; Canver, M.C.; Beyaz, S.; Ljuboja, D.; Kuintzle, R.; Kim, W.; Karnik, R.; et al. Prc2 Is Required to Maintain Expression of the Maternal Gtl2-Rian-Mirg Locus by Preventing De novo DNA Methylation in Mouse Embryonic Stem Cells. Cell Rep. 2015, 12, 1456–1470. [Google Scholar] [CrossRef]
  158. Neri, F.; Krepelova, A.; Incarnato, D.; Maldotti, M.; Parlato, C.; Galvagni, F.; Matarese, F.; Stunnenberg, H.G.; Oliviero, S. Dnmt3l Antagonizes DNA Methylation at Bivalent Promoters and Favors DNA Methylation at Gene Bodies in Escs. Cell 2013, 155, 121–134. [Google Scholar] [CrossRef]
  159. Kumar, D.; Cinghu, S.; Oldfield, A.J.; Yang, P.; Jothi, R. Decoding the Function of Bivalent Chromatin in Development and Cancer. Genome Res. 2021, 31, 2170–2184. [Google Scholar] [CrossRef]
  160. Voigt, P.; Tee, W.-W.; Reinberg, D. A Double Take on Bivalent Promoters. Minerva Anestesiol. 2013, 27, 1318–1338. [Google Scholar] [CrossRef]
  161. Voon, H.P.J.; Gibbons, R.J. Maintaining Memory of Silencing at Imprinted Differentially Methylated Regions. Cell. Mol. Life Sci. 2016, 73, 1871–1879. [Google Scholar] [CrossRef] [PubMed]
  162. Nakamura, T.; Arai, Y.; Umehara, H.; Masuhara, M.; Kimura, T.; Taniguchi, H.; Sekimoto, T.; Ikawa, M.; Yoneda, Y.; Okabe, M.; et al. Pgc7/Stella Protects against DNA Demethylation in Early Embryogenesis. Nat. Cell Biol. 2007, 9, 64–71. [Google Scholar] [CrossRef]
  163. Castillo-Fernandez, J.; Herrera-Puerta, E.; Demond, H.; Clark, S.J.; Hanna, C.W.; Hemberger, M.; Kelsey, G. Increased Transcriptome Variation and Localised DNA Methylation Changes in Oocytes from Aged Mice Revealed by Parallel Single-Cell Analysis. Aging Cell 2020, 19, e13278. [Google Scholar] [CrossRef]
  164. Zhao, J.; Lu, P.; Wan, C.; Huang, Y.; Cui, M.; Yang, X.; Hu, Y.; Zheng, Y.; Dong, J.; Wang, M.; et al. Cell-Fate Transition and Determination Analysis of Mouse Male Germ Cells Throughout Development. Nat. Commun. 2021, 12, 6839. [Google Scholar] [CrossRef] [PubMed]
  165. Ivanova, E.; Canovas, S.; Garcia-Martínez, S.; Romar, R.; Lopes, J.S.; Rizos, D.; Coy, P. DNA Methylation Changes during Preimplantation Development Reveal Inter-Species Differences and Reprogramming Events at Imprinted Genes. Clin. Epigenet. 2020, 12, 64. [Google Scholar] [CrossRef] [PubMed]
  166. Oluwayiose, O.A.; Wu, H.; Saddiki, H.; Whitcomb, B.W.; Balzer, L.B.; Brandon, N.; Suvorov, A.; Tayyab, R.; Sites, C.K.; Hill, L.; et al. Sperm DNA Methylation Mediates the Association of Male Age on Reproductive Outcomes among Couples Undergoing Infertility Treatment. Sci. Rep. 2021, 11, 3216. [Google Scholar] [CrossRef] [PubMed]
  167. Cui, X.; Jing, X.; Wu, X.; Yan, M.; Li, Q.; Shen, Y.; Wang, Z. DNA Methylation in Spermatogenesis and Male Infertility. Exp. Ther. Med. 2016, 12, 1973–1979. [Google Scholar] [CrossRef]
  168. Ben Maamar, M.; Beck, D.; Nilsson, E.; McCarrey, J.R.; Skinner, M.K. Developmental Alterations in DNA Methylation during Gametogenesis from Primordial Germ Cells to Sperm. iScience 2022, 25, 103786. [Google Scholar] [CrossRef]
  169. Alle, Q.; Le Borgne, E.; Bensadoun, P.; Lemey, C.; Béchir, N.; Gabanou, M.; Estermann, F.; Bertrand-Gaday, C.; Pessemesse, L.; Toupet, K.; et al. A Single Short Reprogramming Early in Life Initiates and Propagates an Epigenetically Related Mechanism Improving Fitness and Promoting an Increased Healthy Lifespan. Aging Cell 2022, 21, e13714. [Google Scholar] [CrossRef] [PubMed]
  170. Ocampo, A.; Reddy, P.; Martinez-Redondo, P.; Platero-Luengo, A.; Hatanaka, F.; Hishida, T.; Li, M.; Lam, D.; Kurita, M.; Beyret, E.; et al. In vivo Amelioration of Age-Associated Hallmarks by Partial Reprogramming. Cell 2016, 167, 1719–1733.e12. [Google Scholar] [CrossRef]
  171. Hannum, G.; Guinney, J.; Zhao, L.; Zhang, L.; Hughes, G.; Sadda, S.; Klotzle, B.; Bibikova, M.; Fan, J.-B.; Gao, Y.; et al. Genome-Wide Methylation Profiles Reveal Quantitative Views of Human Aging Rates. Mol. Cell 2013, 49, 359–367. [Google Scholar] [CrossRef] [PubMed]
  172. Guéant, J.L.; Chéry, C.; Oussalah, A.; Nadaf, J.; Coelho, D.; Josse, T.; Flayac, J.; Robert, A.; Koscinski, I.; Gastin, I.; et al. Aprdx1 Mutant Allele Causes a Mmachc Secondary Epimutation in Cblc Patients. Nat. Commun. 2018, 9, 67. [Google Scholar] [CrossRef]
  173. Horsthemke, B. A Critical View on Transgenerational Epigenetic Inheritance in Humans. Nat. Commun. 2018, 9, 2973. [Google Scholar] [CrossRef] [PubMed]
  174. Doudna, J. Crispr’s Unwanted Anniversary. Science 2019, 366, 777. [Google Scholar] [CrossRef] [PubMed]
Figure 1. An overview of DNAme through developmental time. Massive demethylation waves occur in primordial germ cells and pre-implantation stages of development, followed by stage-specific DNAme reacquisition.
Figure 1. An overview of DNAme through developmental time. Massive demethylation waves occur in primordial germ cells and pre-implantation stages of development, followed by stage-specific DNAme reacquisition.
Biomolecules 13 01684 g001
Figure 2. An epigenetic landscape of developmental potential generated with the help of AI DALL-E3 (OpenAI, San Francisco, CA, USA). The ball of pluripotent stem cells represents early developmental potential, where fate is intrinsically regulated by the epigenetic landscape that awaits as it rolls forward in developmental time. DNAme can be visualized as a looming fog, which accumulates over developmental time and clouds cellular potential, as cells transverse narrowing valleys. Auxiliary forces visualized by a growing storm of clouds may erode and re-contour this potential. Migrating PGCs are illustrated moving along a rare path to a distal peak and escaping the downward forces of somatic cell development, transversing upward onto the next generation’s highest developmental potential. Next-generation ESCs are displayed as well as another developing blastocyst. This feat becomes more difficult when cells have accumulated toxic transgenerational DNAme information.
Figure 2. An epigenetic landscape of developmental potential generated with the help of AI DALL-E3 (OpenAI, San Francisco, CA, USA). The ball of pluripotent stem cells represents early developmental potential, where fate is intrinsically regulated by the epigenetic landscape that awaits as it rolls forward in developmental time. DNAme can be visualized as a looming fog, which accumulates over developmental time and clouds cellular potential, as cells transverse narrowing valleys. Auxiliary forces visualized by a growing storm of clouds may erode and re-contour this potential. Migrating PGCs are illustrated moving along a rare path to a distal peak and escaping the downward forces of somatic cell development, transversing upward onto the next generation’s highest developmental potential. Next-generation ESCs are displayed as well as another developing blastocyst. This feat becomes more difficult when cells have accumulated toxic transgenerational DNAme information.
Biomolecules 13 01684 g002
Figure 3. An overview of cytosine DNAme and demethylation is provided. Briefly, C becomes methylated at C5 with DNMT activity and co-factor S-adenyl-methionine (SAM). 5mC can be actively oxidized to 5-hydroxymethylcytosine (5hmc), then to 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) intermediates, which can be removed either actively by Thymine DNA glycosylase (TDG) and base excision repair (BER), or passively through DNA replication.
Figure 3. An overview of cytosine DNAme and demethylation is provided. Briefly, C becomes methylated at C5 with DNMT activity and co-factor S-adenyl-methionine (SAM). 5mC can be actively oxidized to 5-hydroxymethylcytosine (5hmc), then to 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) intermediates, which can be removed either actively by Thymine DNA glycosylase (TDG) and base excision repair (BER), or passively through DNA replication.
Biomolecules 13 01684 g003
Figure 4. An overview of transgenerational inheritance. Potential sources of environmental exposure that may directly affect the gestating mother, developing F1 offspring, or migrating F2 germ cells within F1 offspring are displayed. The effects may also be indirect, such as F0 extreme trauma, which might alter hormone signaling and augment developmental signaling within the fetus and F2 PGCs. Phenotypic influence and epigenetic inheritance to the >F3 generation, without continued exposure to the original F0 event, is considered transgenerational.
Figure 4. An overview of transgenerational inheritance. Potential sources of environmental exposure that may directly affect the gestating mother, developing F1 offspring, or migrating F2 germ cells within F1 offspring are displayed. The effects may also be indirect, such as F0 extreme trauma, which might alter hormone signaling and augment developmental signaling within the fetus and F2 PGCs. Phenotypic influence and epigenetic inheritance to the >F3 generation, without continued exposure to the original F0 event, is considered transgenerational.
Biomolecules 13 01684 g004
Figure 5. Overview of dCas-based DNAme editing. Briefly, enzymatically “dead” Cas enzymes are positioned adjacent to target CGs or CGIs for effector domain recruitment. Primary concerns for a typical experiment are highlighted in blue text, with additional potential modifications that may enhance editing capacity. Transfection and genome homing efficiency are critical factors for successful editing, as are effector domain selection, the timing of dCas, orthogonal system deployment, and the avoidance of immune responses for repetitive editing strategies.
Figure 5. Overview of dCas-based DNAme editing. Briefly, enzymatically “dead” Cas enzymes are positioned adjacent to target CGs or CGIs for effector domain recruitment. Primary concerns for a typical experiment are highlighted in blue text, with additional potential modifications that may enhance editing capacity. Transfection and genome homing efficiency are critical factors for successful editing, as are effector domain selection, the timing of dCas, orthogonal system deployment, and the avoidance of immune responses for repetitive editing strategies.
Biomolecules 13 01684 g005
Figure 6. An overview of CpG Island Methylation Responses (CIMRs) that induce CGI-wide DNAme. Briefly, CG-free DNA is inserted into CG-dense CGI sequences (either double- or single-stranded DNA may be integrated). Uniquely, in primed pluripotency, the flanking CGI CpGs become spontaneously methylated. For most instances of promoter DNAme, this results in stable long-term gene silencing. DNAme is retained after CG-free insert removal and enables transgenerational testing of a single-engineered CGI by tracking DNAme in subsequent generations of offspring when tested in vivo.
Figure 6. An overview of CpG Island Methylation Responses (CIMRs) that induce CGI-wide DNAme. Briefly, CG-free DNA is inserted into CG-dense CGI sequences (either double- or single-stranded DNA may be integrated). Uniquely, in primed pluripotency, the flanking CGI CpGs become spontaneously methylated. For most instances of promoter DNAme, this results in stable long-term gene silencing. DNAme is retained after CG-free insert removal and enables transgenerational testing of a single-engineered CGI by tracking DNAme in subsequent generations of offspring when tested in vivo.
Biomolecules 13 01684 g006
Figure 7. A basic framework for determining whether programmed DNAme is transgenerational has been provided. Major considerations for editing are provided as follows. (I) Characterize the source of transgenerational detriment, epigenetic penetrance, and the expected generational durability. Which DNAme sites are changed relative to normal, age, and sex-matched controls without exposure? What proportion of individuals with a particular heritable DNAme exhibit the negative epiphenotype? How many generations of transgenerational inheritance may be expected? For strongly modeled candidates or those supported by long-term epidemiological studies on transgenerational inheritance, epigenetic editing for mimicry can be used to test the suspected site for programmed transgenerational effects. Closed circles represent methylated candidate CG sites, which can be found in clusters or individually. (II) Determine the targeting strategy to mimic transgenerational DNAme. Targeting of PGCs, gametes, or early embryonic windows is preferred for inheritance into entire organisms, and for targeting developmental windows, which precede next-generation PGC migration and development for inheritance testing. Editing strategies are generally protein-homing-based and may also include certain adaptations to the enzymatically dead Cas systems, such as crRNA modifications and effector domain scaffolds. Specific domains or small RNA co-factors active during key developmental windows may enhance these activities towards transgenerational outcomes, especially those building histone marks, such as H3K9me3, which promote eventual DNAme deposition during later stages of development. Demethylation strategies generally rely on the recruitment of active demethylating enzymes or activating domains, such as VP64, to override DNAme. Engineered DNAme sites may also be protected by binding certain factors, such as STELLA, to prevent first-generation removal and allow for durability testing in subsequent generations. Insertions of CpG-free DNA or mutations that drive changes in transcription, for example, may induce local CGI DNAme spreading epimutations. Retained after the original insult, these naturally aid in transgenerational testing. (III) Monitor DNAme for transgenerational inheritance. Programmable Transgenerational Epigenetic Reacquisition (PTER) is the intentional, locus-, or region-specific induction of specific epigenetic configurations, which fail to escape germline or pre-implantation erasure but that are retriggered with each ensuing generation without the continued presence of epigenetic editing systems. This is unique from Programmable Transgenerational Epigenetic Transmission (PTET), which involves protection from germline and pre-implantation stages of epigenetic erasure. Either form of inheritance is of interest to human biology, but these remain important mechanistic clarifications, which can be further defined by stage-specific isolation of developing PGCs and gametes, developing blastocysts, and the examination of multigenerational inheritance in somatic tissues of offspring. Pedigree analyses can aid in establishing transgenerational transmission, penetrance, and durability. For in vitro modeling, which may include human pluripotent stem cells, the reversible cycling of cells between primed and naïve states enables reversible global DNAme switching and the testing of DNAme between additional generations of development. Sites or regions that wane in DNAme over repeat cycling, especially with DNAme-inducing DNA or mutations removed, are less likely to maintain transgenerational activity in vivo. For human early embryo studies modeled in vitro, the 14-day rule would be sufficient for understanding DNAme inheritance through primed stages of pluripotency and up to gastrulation. Beyond this, in vitro mimics of specific human somatic lineages remain state-of-the-art. Given the difference between humans and mice in repetitive element and imprinting regulation, human studies are preferred for human biology; however, preclinical modeling remains essential to understanding and testing transgenerational DNAme correction.
Figure 7. A basic framework for determining whether programmed DNAme is transgenerational has been provided. Major considerations for editing are provided as follows. (I) Characterize the source of transgenerational detriment, epigenetic penetrance, and the expected generational durability. Which DNAme sites are changed relative to normal, age, and sex-matched controls without exposure? What proportion of individuals with a particular heritable DNAme exhibit the negative epiphenotype? How many generations of transgenerational inheritance may be expected? For strongly modeled candidates or those supported by long-term epidemiological studies on transgenerational inheritance, epigenetic editing for mimicry can be used to test the suspected site for programmed transgenerational effects. Closed circles represent methylated candidate CG sites, which can be found in clusters or individually. (II) Determine the targeting strategy to mimic transgenerational DNAme. Targeting of PGCs, gametes, or early embryonic windows is preferred for inheritance into entire organisms, and for targeting developmental windows, which precede next-generation PGC migration and development for inheritance testing. Editing strategies are generally protein-homing-based and may also include certain adaptations to the enzymatically dead Cas systems, such as crRNA modifications and effector domain scaffolds. Specific domains or small RNA co-factors active during key developmental windows may enhance these activities towards transgenerational outcomes, especially those building histone marks, such as H3K9me3, which promote eventual DNAme deposition during later stages of development. Demethylation strategies generally rely on the recruitment of active demethylating enzymes or activating domains, such as VP64, to override DNAme. Engineered DNAme sites may also be protected by binding certain factors, such as STELLA, to prevent first-generation removal and allow for durability testing in subsequent generations. Insertions of CpG-free DNA or mutations that drive changes in transcription, for example, may induce local CGI DNAme spreading epimutations. Retained after the original insult, these naturally aid in transgenerational testing. (III) Monitor DNAme for transgenerational inheritance. Programmable Transgenerational Epigenetic Reacquisition (PTER) is the intentional, locus-, or region-specific induction of specific epigenetic configurations, which fail to escape germline or pre-implantation erasure but that are retriggered with each ensuing generation without the continued presence of epigenetic editing systems. This is unique from Programmable Transgenerational Epigenetic Transmission (PTET), which involves protection from germline and pre-implantation stages of epigenetic erasure. Either form of inheritance is of interest to human biology, but these remain important mechanistic clarifications, which can be further defined by stage-specific isolation of developing PGCs and gametes, developing blastocysts, and the examination of multigenerational inheritance in somatic tissues of offspring. Pedigree analyses can aid in establishing transgenerational transmission, penetrance, and durability. For in vitro modeling, which may include human pluripotent stem cells, the reversible cycling of cells between primed and naïve states enables reversible global DNAme switching and the testing of DNAme between additional generations of development. Sites or regions that wane in DNAme over repeat cycling, especially with DNAme-inducing DNA or mutations removed, are less likely to maintain transgenerational activity in vivo. For human early embryo studies modeled in vitro, the 14-day rule would be sufficient for understanding DNAme inheritance through primed stages of pluripotency and up to gastrulation. Beyond this, in vitro mimics of specific human somatic lineages remain state-of-the-art. Given the difference between humans and mice in repetitive element and imprinting regulation, human studies are preferred for human biology; however, preclinical modeling remains essential to understanding and testing transgenerational DNAme correction.
Biomolecules 13 01684 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tompkins, J.D. Transgenerational Epigenetic DNA Methylation Editing and Human Disease. Biomolecules 2023, 13, 1684. https://doi.org/10.3390/biom13121684

AMA Style

Tompkins JD. Transgenerational Epigenetic DNA Methylation Editing and Human Disease. Biomolecules. 2023; 13(12):1684. https://doi.org/10.3390/biom13121684

Chicago/Turabian Style

Tompkins, Joshua D. 2023. "Transgenerational Epigenetic DNA Methylation Editing and Human Disease" Biomolecules 13, no. 12: 1684. https://doi.org/10.3390/biom13121684

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop