Next Article in Journal
Nuclear Receptor-Mediated Metabolic Reprogramming and the Impact on HR+ Breast Cancer
Previous Article in Journal
Therapeutic Potential of Innate Lymphoid Cells for Multiple Myeloma Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cis-Acting Factors Causing Secondary Epimutations: Impact on the Risk for Cancer and Other Diseases

by
Miguel Ruiz de la Cruz
1,2,
Aldo Hugo de la Cruz Montoya
3,
Ernesto Arturo Rojas Jiménez
1,3,
Héctor Martínez Gregorio
1,3,
Clara Estela Díaz Velásquez
1,
Jimena Paredes de la Vega
1,3,
Fidel de la Cruz Hernández-Hernández
2 and
Felipe Vaca Paniagua
1,3,4,*
1
Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico
2
Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
3
Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico
4
Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(19), 4807; https://doi.org/10.3390/cancers13194807
Submission received: 7 July 2021 / Revised: 9 August 2021 / Accepted: 15 August 2021 / Published: 26 September 2021
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)

Abstract

:

Simple Summary

Epigenetic mechanisms contribute to the regulation of gene expression. However, when they fail, they result in diseases such as cancer. Among these effects is aberrant DNA methylation caused by inherited mutations in cis of the affected gene, referred to as constitutional secondary epimutations. Little is known about this phenomenon, in which hypermethylation promotes transcriptional silencing of tumor suppressor genes in patients with inherited cancers that do not have pathogenic variants in the coding region of cancer susceptibility genes. Here we discuss these hereditary alterations and their effect during the early stages of tumorigenesis, as well as their contribution to disease historically and from a molecular perspective.

Abstract

Epigenetics affects gene expression and contributes to disease development by alterations known as epimutations. Hypermethylation that results in transcriptional silencing of tumor suppressor genes has been described in patients with hereditary cancers and without pathogenic variants in the coding region of cancer susceptibility genes. Although somatic promoter hypermethylation of these genes can occur in later stages of the carcinogenic process, constitutional methylation can be a crucial event during the first steps of tumorigenesis, accelerating tumor development. Primary epimutations originate independently of changes in the DNA sequence, while secondary epimutations are a consequence of a mutation in a cis or trans-acting factor. Secondary epimutations have a genetic basis in cis of the promoter regions of genes involved in familial cancers. This highlights epimutations as a novel carcinogenic mechanism whose contribution to human diseases is underestimated by the scarcity of the variants described. In this review, we provide an overview of secondary epimutations and present evidence of their impact on cancer. We propose the necessity for genetic screening of loci associated with secondary epimutations in familial cancer as part of prevention programs to improve molecular diagnosis, secondary prevention, and reduce the mortality of these diseases.

Graphical Abstract

1. Introduction

Cancer is caused by genetic, metabolic, inflammatory, and epigenetic factors [1]. The abnormal proliferation, the major characteristic of cancer, begins with the progressive accumulation of multiple mutations that provide evolutionary adaptations to the tumor cells. Although epigenetic mechanisms were initially overlooked, it is now known that they contribute importantly to cancer development by their capacity to alter the gene expression independently of mutations [2] (Figure 1). These epigenetic mechanisms can be classified into three main molecular groups: non-coding RNAs (ncRNAs), histone post-translational modifications, and DNA/RNA methylation [3,4].
Deregulation of ncRNAs is one of the main regulatory epigenetic mechanisms altered during carcinogenesis and cancer progression [4]. The three most studied classes of ncRNAs are miRNAs, lncRNAs, and circRNAs, which are involved in complex interactions during cancer initiation and promotion [5]. Potent tumorigenic effects of miRNAs have been well described in different malignancies and polymorphisms in these genes may cause cancer susceptibility [6,7]. LncRNAs have been shown to aberrantly promote metastatic pathways and specific mutations in these genes have a key cancer role [8]. In addition, circRNAs have oncogenic activity in organ-specific cancers, neurodegenerative pathogenic conditions, diabetes, and cardio-vascular disease [9]. For a more detailed discussion on the oncogenic role of ncRNAs we refer the reader to the comprehensive reviews published elsewhere [10,11,12].
Histone post-translational modifications alter chromatin architecture and their spatial distribution. The mechanisms that cause these changes operate at the level of both nuclear topology and histone modifications driven by the activity of the Polycomb (PcG) and Trithorax (TrxG) chromatin-modifying complexes [3,13]. Nuclear topology importantly influences the mutational composition in cancer cells, as evidenced by mutations acquired by smoking and UV light enriched at the nuclear periphery [14,15]. PcG and TrxG complexes regulate modifications on histone amino tails through repression signals (polycomb) and gene activation marks (trithorax), changes that collectively have an important role in cellular processes such as apoptosis and senescence [16,17]. Notably, there are more than 60 different histone chemical marks, including methylation, acetylation, phosphorylation, citrullination, sumoylation, adenosine diphosphate ribosylation, deamination, and crotonylation, which regulate the chromatin structure and locus accessibility, improve gene expression in euchromatin regions or mediate heterochromatin gene repression [18].
DNA methylation is the only epigenetic mechanism that directly affects the chemical structure of DNA regulating gene expression through stable silencing. Technical molecular advances for the detection of DNA methylation have given access to a wide range of studies in the fields of embryology, organism development, and disease, by global 5meC quantification [19], locus-specific DNA methylation approaches (sodium bisulfite modification strategies [20,21,22] and methylation-sensitive restriction enzyme sequencing [23,24]), and genome-wide DNA methylation [25,26,27], providing a detailed profile on genome-wide DNA methylation. No technique excels in all aspects. The number and characteristics of the sample as well as the desired precision, coverage, and resolution, define the choice of the appropriate technique. Initial methylation analyzes used qualitative approaches such as MSP, MeDIP-PCR, and microarrays, and focused only on the presence or absence of methylation in a specific locus or CpG islands. Over time, novel methylation techniques such as pyrosequencing and next generation sequencing have been coupled with an initial bisulfite conversion treatment. This has allowed to analyze, at a whole genome level, a wide range of DNA methylation patterns and provides quantitative values on an absolute scale of β distribution from 0 to 1, or 0 to 100%. For more details on methods for DNA methylation analysis, the reader is referred to the review of Laird (2010) [28]. These technological advances in DNA methylation analysis contributed to making this mark the most studied epigenetic modification in humans.
DNA methylation is associated with repressive histone modifications and the interaction between these epigenetic variations is crucial to regulate the function of the genome by changing the architecture of chromatin [29]. In this regard, methylation involves a post-replicative chemical modification through the transfer of a methyl group, mainly in CpG dinucleotides in mammals, which comprise approximately 3–6% of the total cytosines of human DNA [30], although methylation in N6-adenosine has also been described [31]. The enzymes that catalyze methylation in CpG dinucleotides are DNA methyltransferases (DNMTs). Methylation is stable through cell division, can be added de novo, and is also reversible [30,32,33]. There are three active DNMTs in mammals responsible for methylation, of which DNMT1 participates in the maintenance of the methylation patterns during cell division. Conversely, DNMT3A and DNMT3B carry out the de novo methylation in response to molecular stimuli. An additional participant in the DNA methylation process is DNMT3L, a regulatory protein that does not have methyltransferase activity, but stabilizes the DNMT3A/B-DNA complex to increase de novo methylation [34,35]. DNA methylation plays an extremely important role throughout the body, as demonstrated by its involvement in crucial biological processes including regulation of gene expression [36], early development [37], protection against intragenomic parasites [38], genomic imprinting [39], and the inactivation of chromosome X [40]. Consequently, alterations in this regulatory mechanism lead to an aberrant increase (hypermethylation) or decrease (hypomethylation) in basal levels that have been associated with diseases such as cardiovascular problems [41,42], mental disorders [43,44], and different types of cancer [45,46,47].
When these mechanisms cause aberrant disruptions in DNA activity, they are called epimutations [48]. Epimutations are epigenetic alterations that result in transcriptional repression or activation of a gene that is normally active or repressed, respectively [49,50]. They occur when a paternal or maternal allele has >50% aberrant methylated or demethylated CpG sites, showing not successful methylation imprinting [51]. Epimutations are grouped into two types [52]: (i) primary epimutations, caused by adaptive aberrant molecular mechanisms that do not involve the alteration of the DNA sequence, and (ii) secondary epimutations, acquired as a consequence of DNA mutations in cis or trans-acting factors [48].
In this article, we review the relationship between epimutations that alter gene expression and disease. We especially focus on secondary constitutional epimutations caused as a result of inherited DNA cis-alterations and describe their experimental evidence over time.

2. Origin and Consequences of Epimutations

Mutational events that contribute to carcinogenesis may occur during gametogenesis and embryonic development (Figure 2). Somatic mutations are alterations in the DNA sequence in any cell of the body that take place after fertilization. Hence, they are not passed onto the offspring, but tissues derived from the mutated cell are affected (Figure 2A) [53]. On the other hand, germline mutations are DNA changes present in gametes that can be inherited to the offspring affecting all cells throughout the body and half of the offspring’s gametes (Figure 2B) [54].
On a different level, epimutations are epigenetic alterations that can occur at specific stages in normal cells, for example, when a tumor suppressor gene is methylated during the early stages of embryonic development (Figure 2C). Recently, it has been reported that epimutations could act as initial events of tumorigenesis and together with somatic and germline mutations lead to malignant transformation and progression [55].
Epimutations studies are based on genomic imprinting, a methylation state that establishes parent-specific allele expression. A pathological phenotype could be expressed if imprinting is deficient even in one allele. The genomic-specific phenotypes of imprinting are regulated by imprinting centers (ICs). These ICs are short sequences in cis of an imprinted gene that regulate parent-specific gene expression bidirectionally over long distances in which only one allele is methylated and transcribed, allowing for imprinting regulation and allele-specific expression. Therefore, genetic alterations in the sequences of ICs can lead to constitutional epimutations [52]. Constitutional epimutations are monoallelic and can be found in tissues from the three germ layers: endoderm, ectoderm, and mesoderm [56].
During early human development, the epigenetic modifications throughout the genome occurs dynamically. At this stage, the reprogramming of methylation has functions in the imprinting, the control of gene expression, and the establishment of the cell lineage [57]. Several studies have confirmed that during embryonic development the epigenetic marks of the parents (F0) are erased through an active demethylation mechanism [58,59]. Later, during fertilization, the differentially methylated germ regions (gDMR) are established by de novo methylation, including ICRs. During this stage, constitutional epimutations are more likely to be generated. In the zygote, there is a selective demethylation phenomenon of active demethylation of the paternal genome followed by passive demethylation of the maternal genome within the preimplantation embryo [60]. It has been proposed that this selective demethylation results in the specificity of imprinted genes. However, when it fails, it could result in the impairment of methylation in the imprinted loci, generating epimutations throughout the soma. Therefore, the partial “erasure” of a constitutional epimutation in a proportion of cells in the pre-implanted embryo could result in somatic mosaicism of the epimutation in the adult [61,62]. In addition, a de novo methylation was generated in the inner cell mass (ICM) of the blastocyst and has an important role in early lineage establishment. During this stage of the early development, constitutional epimutations could be generated [58].
In the 1990s, epimutations in tumor suppressor genes such as RB [63], VHL [64], MLH1 [65], and BRCA1 [66] were identified in sporadic cancers. However, over time, the research about this phenomenon in hereditary cancers has been strengthened. We discuss the role of epimutations in inherited cancers in the following section.

3. Aberrant Methylation as the Initial Step of Carcinogenesis in Hereditary Cancers

Epimutations are one of the early molecular alterations in tumorigenesis and may be a direct cause of carcinogenesis [67]. The causality in cancer is associated with common and uncommon genetic variants [58]. However, these variants only explain a portion of the individuals with the phenotype. On the other hand, epigenetics has been established as an alternative mechanism to explain causality in cancer [68].
In 1971, Knudson postulated the “two-hit hypothesis” to explain how two mutations or “hits” were required in both RB1 alleles to develop retinoblastoma [69]. Nowadays, it is used to explain the pathogenesis of tumors in autosomal dominant cancer syndromes [70]. Knudson’s hypothesis requires that both alleles of a tumor-suppressing gene must be impaired to cause cancer. The first hit frequently comes from germline mutations, the second hit may result from somatic mutations such as SNV or aberrant methylation of the second allele [71] (Figure 3). Interestingly, constitutional epimutations can either act as the first hit in concordance with the Knudson’s model to develop cancer [72,73], or as the second hit in some tumors associated with familial cancer syndromes caused by heterozygous germline mutations [74]; contributing in this way to the carcinogenic process. Accordingly, germline epimutations that initiate during embryonic development could contribute to the “first hit” in tumorigenesis in cancer susceptibility genes. Some alterations favor transcriptional silencing and act as the first hit. Some examples of these mechanisms include the direct impairment of DNA binding of transcription factors (TFs) to the promoter region [75], the backward movement of methyl-CpG (MBD) binding domain proteins in methylated DNA, which compete directly with TFs through their greater affinity for the 5-mC site in the DNA sequence [76], and the synergistic binding effect of transcriptional repression factors (TFRs), which bind with affinity to DNA when an SNV is present. This phenomenon will be addressed in the Section 5 [77].
One of the first pieces of evidence that associated the two-hit hypothesis and epimutations was a study conducted on the MLH1 gene. A seminal study by Gazzoli et al. described an epimutation that caused MLH1 promoter hypermethylation. Interestingly, this epimutation was detected in one allele (from peripheral blood DNA) in a patient with colorectal cancer, and the second unmethylated allele was inactivated due to loss of heterozygosity in the tumor tissue. This biallelic inactivation resulted in the complete loss of MLH1 expression in the tumor, confirmed by immunohistochemistry. These findings suggested a new mechanism of germline inactivation in a cancer susceptibility gene [78]. A second work confirmed this aberrant mechanism in MLH1 in two individuals who did not have a germline mutation in DNA repair genes but presented clinical criteria for hereditary colorectal cancer; concluding that the germline epimutation in MLH1 leads to somatic mosaicism consistent with epigenetic states that resemble those of polygenic or complex traits [79].

4. Primary Epimutations and Environmental Factors

The epigenetic architecture of cells and tissues is actively shaped by their environment and molecular context. The molecular factors that influence the epigenetic landscape of cells are stress, proliferative and cell cycle regulatory signals, specific extracellular molecules, nutrition, among others [80].
Primary or “true” epimutations are aberrant adaptive changes in response to molecular stimuli that occur without any change either in the DNA sequence or in ICs, but as a result of genetic, environmental, and stochastic factors that lead to transcriptional repression (Figure 4(AI)) [52]. One of the best-characterized examples of a primary epimutation occurs in patients with Wilms tumors, where the maternal allele of the imprinting control regions (ICR) upstream of the H19 gene is aberrantly methylated, leading to loss of the imprinting (LOI) [81].
The environmental factors that have been linked to the modification of the methylation patterns include exposure to chemical agents such as heavy metals [82], aerosols in the air [83], cigarette smoke [84], endocrine-disrupting substances, or non-genotoxic carcinogens [85]. All these factors can generate changes in gene expression, activation of signaling pathways, and alterations in the epigenome and transcriptome [86].
The nutritional status is an environmental factor that is associated with primary epimutations that affect the methylation pattern [71], as well as the risk of cancer [87,88]. Dietary components rich in folates, choline, and vitamin B12 are known to influence the methylation status by increasing the levels of SAM (S-adenosyl-methionine), exhibiting a direct link between diet and the epigenetic state [89,90]. SAM serves as an enzymatic substrate in which DNA methyltransferases obtain the methyl groups to bind to DNA [91]. The impact of dietary folate on changes in DNA methylation has been observed in breast, prostate, stomach, colon, and thyroid cancers [92], as well as in animal models [93]. Thus, when evaluating the effects of the absence of folates in a rat model supplemented with vitamin B12 deficiency, DNA hypomethylation was found in colon tissue, which indicates that the restricted diet in the absence of B12 greatly reduces DNA methylation [94].
Also, primary epimutations have been linked to aging and may contribute to the late onset of cancer [95]. For example, during the aging process, DNA hypermethylation is facilitated competitively due to the destabilization of the repressive polycomb complex [96].

5. Secondary Epimutations

Secondary epimutations are driven by cis or trans-acting genetic alterations (Figure 4(AII)) [52]. Secondary epimutations that operate through the action of factors in trans include DNA mutations in genes that encode enzymes involved in DNA methylation, histone acetylation or deacetylation, histone methylation, chromatin remodeling, and epigenetic mark recognition [97]. However, secondary epimutations of cis action include deletions, insertions, substitutions, and changes in the length of tandem repeated sequences known as copy number variations (CNV) [98], all capable of affecting ICs [49]. Notably, these alterations can experience the following types of inheritance patterns: (i) autosomal dominant transmission of epimutations caused by genetic cis-acting alterations [55]; (ii) “Null” inheritance due to sporadic de novo epimutations with concomitant germ-cell erasure of epimutations and re-establishment of the somatic aberrant state in the next generation [99,100] and (iii) non-Mendelian transmission or incomplete penetrance [99]. This translates into a complex and heterogeneous landscape in the inheritance mechanisms of epimutations, even when the same gene is affected (Figure 4B). These findings were discovered on the MLH1 gene and probably additional loci may show these inheritance patterns.
In addition to secondary epimutations caused by cis-acting factors, secondary epimutations caused by mutations in trans-acting factors include alterations in the coding region of DNA methyltransferases, methyl-CpG binding proteins, histone-modifying enzymes, and the chromatin remodeling complex, among others (Figure 4(AII)). These protein complexes are essential for epigenetic maintenance and its deregulation generates a critical panorama for diseases and a potential early death of the patient [52]. One example is the case of de novo DNA methyltransferase (DNMT3A) mutations, which cause the autosomal recessive ICF syndrome (immunodeficiency, centromere instability, and facial anomalies). Patients with this syndrome present microsatellite instability associated with severe DNA hypomethylation, resulting in death at a young age from recurrent serious infections. Other diseases caused by secondary epimutations are mental retardation syndromes with X-linked–α-thalassemia (ATRX) and Rett syndrome (RTT). The former is a disorder characterized by mental retardation, facial dysmorphia, genital abnormalities, alpha thalassemia, and it is caused by a reduced expression of the α-globin genes that encode a member of the SWI/SNF complexes, which plays an important role in the system of chromatin remodeling and DNA repair [101]. The RTT is a rare genetic neurological disorder that affects brain development and causes severe mental and physical disability caused by mutations in the MECP2 gene, which encodes the methyl-CpG2 binding protein. MECP2 recruits the corepressor complex SIN3A containing histone deacetylase that favors the repressive chromatin state, leading to transcriptional inactivation [52,102].

6. Cis-Acting Factors Causing Secondary Epimutations: Historical Evidence

Constitutional secondary epimutations with cis effect have been described in several loci, but particularly in tumor suppressor genes including FMR1, SNURF/SNRPN, HBA2, H19, LIT1, MSH2, DAPK1, MLH1, CDH1, MGMT, MMACHC, and BRCA1. During the 1990s, the first reports were focused on promoter cis-acting epimutations. CGG repeat expansions of more than 200 trinucleotide repeats, located at the 5’UTR end of fragile X mental retardation gene 1 (FMR1) causes a neurodevelopmental disorder called the fragile X syndrome (FXS) (Figure 5A) [103,104]. To better understand the role of these repeated CGG expansions, subsequent studies evaluated cytosine methylation patterns within a 220 bp region on the CpG island of the human FMR1 gene in peripheral blood, finding hypermethylation in this region [104]. These initial studies demonstrated that FMR1 is also methylated and inactivated after the expansion of CGG repeats in fragile X syndrome. Methylation variability was most pronounced in a consensus binding sequence for the transcription factor α-PAL, a sequence that may play a role in regulating FMR1 expression. This evidence suggests that the maintenance of cytosine methylation is a dynamic process and could generate a fragile X-male methylation mosaicism [105].
As mentioned before, epimutations are involved in genomic imprinting. Several genetic alterations have been found in ICs such as microdeletions that result in secondary epimutations in both Prader-Willi (PWS) and Beckwith-Wiedemann syndromes (BWS) [67]. One of the genes of importance for PWS is SNURF (SNRPN upstream reading frame protein), which is found within a critical region of chromosome 15. The transcripts of this gene are initiated in an IC and have paternal imprinting [106,107]. These transcripts can be bicistronic and encode SNRPN (small nuclear ribonucleoprotein polypeptide N) from a downstream open reading frame. These regions were initially mapped at 100 Kb (including exon 1) [108], detecting its differential methylation at the CpG island of SNRPN [109,110,111,112] and intron 7 [109,113,114,115]. However, it has been reported that deletions in the paternal allele in the PWS lead to an epigenetic state that resembles the maternal imprinting [116,117]. PWS clinical characteristics include muscular hypotonia, hypogonadism, obesity, short stature, and mild to moderate mental retardation. One study of 51 patients with PWS showed that seven patients (14%) presented a paternally inherited deletion in the IC region affecting 4.3 Kb around exon 1 of SNURF-SNRPN [116,117]. This alteration influenced the maintenance of paternal imprinting during early embryogenesis, eventually leading to PWS (Figure 5B). These microdeletions have been named Prader-Willy Syndrome Smallest Region of deletion Overlap (PWS-SRO) for their association with the PW syndrome.
One interesting study of a patient with a family history of α-thalassemia showed no mutations in the exonic sequence of the HBA2 gene. However, the promoter of one allele was found to be hypermethylated and transcriptionally silenced in peripheral blood lymphocytes, with the absence of methylation in sperm. This constitutional secondary epimutation was caused by an 18 kb deletion downstream of HBA2, which eliminates some genes including LUC7L; that is antisense transcribed in the opposite direction to the HBA2 gene [118,119]. This deletion eliminates three exons of the LUC7L gene, including the stop codon. Its antisense transcription extends across the CpG island of the HBA2 promoter, causing its hypermethylation and the complete allelic silencing of the HBA2 gene (Figure 5C) [120].
Figure 5. Chronological reports of secondary epimutations causing promoter methylation in cancer and other diseases [55,77,105,116,120,121,122,123,124,125,126,127]. Red color indicates the cis-acting factor that results in secondary epimutation. The details of each disease and their pathogenic secondary epimutations are described in the text. Green arrows indicate the site +1 (ATG) of the gene and red diagonals indicate transcriptional repression of the gene. The methylation region position is described relative to the transcription start site. Methods: MSP, Methylation-Specific-PCR; PSQ, Pyrosequencing. COBRA, Combined Bisulfite Restriction Analysis; PCR-BS, PCR-Bisulfite sequencing; MLPA, Multiplex Ligation-Dependent Probe Amplification; ARMS-PCR, amplification refractory mutation system-based PCR.
Figure 5. Chronological reports of secondary epimutations causing promoter methylation in cancer and other diseases [55,77,105,116,120,121,122,123,124,125,126,127]. Red color indicates the cis-acting factor that results in secondary epimutation. The details of each disease and their pathogenic secondary epimutations are described in the text. Green arrows indicate the site +1 (ATG) of the gene and red diagonals indicate transcriptional repression of the gene. The methylation region position is described relative to the transcription start site. Methods: MSP, Methylation-Specific-PCR; PSQ, Pyrosequencing. COBRA, Combined Bisulfite Restriction Analysis; PCR-BS, PCR-Bisulfite sequencing; MLPA, Multiplex Ligation-Dependent Probe Amplification; ARMS-PCR, amplification refractory mutation system-based PCR.
Cancers 13 04807 g005
Another disease associated with aberrant genomic imprinting is the Beckwith-Wiedemann syndrome (BWS); a congenital overgrowth disorder, whose most frequent clinical characteristics are microglossia (97%), abdominal wall defect (80%), birth weight, or postnatal growth above 90th percentile (88%), ear folds/pits (76%), nephromegaly (59%) and hypoglycemia (63%) [128,129,130]. Furthermore, 5–10% of patients with BWS have been reported at high risk for developing malignant tumors, including Wilms tumor, hepatoblastomas, rhabdomyosarcomas, and adrenocortical carcinomas [131,132]. Alterations in the 11p15.5 locus cause BWS [128]. This genomic region has two imprinted domains: IC1 that regulates the expression of insulin-like growth factor 2 (IGF2) and H19; and IC2 controls cyclin-dependent kinase inhibitor 1C and KCNQ1 with antisense transcript 1 (CDKN1C/KCNQ1OT1). Epimutations of IC1 with hypermethylation of the maternal allele in the H19 DMR (differentially methylated region of the H19 gene) account for 5% of BWS cases (Figure 5D). This epimutation results in the biallelic expression of IGF2 and silencing of H19 expression [128]. These findings were strongly complemented by the investigations of Sparago et al., identifying two families that showed a microdeletion in IC1: IGF2/H19. Transmission of the deletions in the maternal allele resulted in hypermethylation of IC1: H19, biallelic expression of IGF2, silencing of H19, and BWS [121,133].
Secondary epimutations in the IC2 region with loss of methylation (LOM) in KvDMR1 (differentially methylated region of the KCNQ1OT1 gene) represent less than 2% of BWS cases (Figure 5E). This epimutation in the maternal allele results in the biallelic expression of KCNQ1OT1 (also known as LIT1) and the silencing of CDKN1C (also known as P57Kip2) in both alleles [128,134,135,136]. Subsequently, it was demonstrated by Niemitz et al., that aberrant maternal expression of LIT1 is conditioned by a microdeletion in IC2: KvDMR1 spanning approximately 250 kb and including almost all of the LIT1 coding region [122].
The presence of secondary epimutations in Chronic Lymphocytic Leukemia (CLL) has been widely documented. CLL is characterized by uncontrolled proliferation of lymphocytes (lymphocytosis) whose complications may lie in splenomegaly, anemia, and hypogammaglobulinemia, leading to recurrent infections. Between 5–10% of CLL cases are familial [137] and the silencing of death-associated protein kinase 1 (DAPK1) has been strongly associated with an epimutation mechanism. DAPK1 was identified as a familial tumor suppressor gene and it has shown aberrant promoter hypermethylation in CLL (Figure 5F). This secondary epimutation was associated with an SNV (c.-6531A>G) approximately 65 Kb upstream of the DAPK1 gene and has a dominant inheritance pattern [62]. Additionally, functional assays revealed a decrease of transcriptional activity directly associated with a higher binding affinity of the homeobox transcriptional repressor (HOXB7) in the DAPK1 promoter in the pathogenic allele [77].
Evidence linking constitutional epimutations to cancer predisposition has been well documented in some conditions like Lynch syndrome [81,138]. This syndrome has a dominant inheritance pattern and is characterized by an elevated risk to develop colorectal, endometrial, and other types of cancer [139]. It is caused by germline mutations in DNA mismatch repair (MMR) genes, including MLH1, MSH2, or MSH6 causing microsatellite instability (MSI) [140] in 2–3% of all colorectal cancers [141,142]. The majority of cases with high MSI have somatic hypermethylation of the MLH1 promoter [143]. MSI in patients with Lynch syndrome or spontaneous cancer with MLH1 promoter hypermethylation indicates that epimutations as well as somatic mutations affecting MLH1 are early onset in the carcinogenesis of colorectal cancer. Patients with clinical suspicion of Lynch syndrome, but without a germline mutation in an MMR gene or a familiar history of cancer, typically show aberrant methylation of the MLH1 promoter in normal and tumor tissues, suggesting that it is a causative agent for cancer development in these people [72]. Dominant transmission of an MLH1 epimutation with somatic mosaicism and transcriptional repression linked to a particular genetic haplotype was reported in a family affected by colorectal cancer [55,144,145]. Epimutation was erased in sperm, but was reestablished in somatic cells of the next generation, consistent with an inherited genetic effect causing the secondary hypermethylation phenotype. The affected haplotype consisted of a single nucleotide substitution, c.-27C>A, located near the transcription start site (Figure 5I). Importantly, functional assays to test this variant showed a significantly reduced transcriptional activity and is likely to be the cause of this secondary epimutation [55,146].
Furthermore, one of the most important studies over time is one of the constitutional epimutations in the MSH2 gene and their role in colorectal cancer development. It was first described in 2006 by Chan et al. [147], where a three-generation Chinese family with hereditary nonpolyposis colorectal cancer (HNPCC) exhibited germline allele-specific hypermethylation in the MSH2 promoter region without having mutations in DNA mismatch repair genes (MMR); this was key to explain the etiology of Lynch syndrome in the family. Three family members who were carriers of germline epimutations developed early-onset endometrial or colorectal cancer, all with evidence of MSI and allelic loss of MSH2 [147,148]. In the following three years, Ligtenberg et al. showed that this secondary epimutation of the MSH2 promoter region was a consequence of a 5 Kb deletion at the 3’ end of a neighboring EPCAM gene upstream of MSH2, located at 17 Kb (Figure 5G). This EPCAM deletion includes the polyadenylation signal that results in a continuous transcription, leading to an EPCAM-MSH2 fusion transcript and transcriptional repression of MSH2 [123,149]. Gastric cancer (GC) studies have shown great advances in the field of epimutations. The hereditary familial form of GC is known as diffuse hereditary gastric cancer (HDGC), has an autosomal dominant pattern, and is responsible for 1% of all types of GC. Despite its low frequency, HDGC is a public health problem due to its severity and late diagnosis [150,151]. In a study of proband patients with HDGC disease with the absence of germline mutations, hypermethylation of the CDH1 gene promoter (Cadherin-1) in peripheral blood DNA was found in a single patient. Aberrant methylation was conditioned by a specific allele with rs16260 (C.-161C>A) SNP (Figure 5H).
However, it was not possible to identify the epimutation in any other member of the family besides the proband because they had died due to the disease, which made it impossible to clarify the pattern of the inheritable epimutation of gastric cancer [124].
The gene MGMT, which codes for the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) has also been studied at the epigenetic level. The MGMT enzyme eliminates the transitional mutation of O6 methylguanine and participates in therapy resistance to alkylating agents [152]. The activity of MGMT is regulated by its promoter and its hypermethylation leads to gene silencing in cancer [153,154]. Epimutations in MGMT have been linked to various types of cancer, such as colorectal cancer, lung cancer, and brain gliomas [125,153,154,155,156,157]. An epimutation in the promoter region of the MGMT gene is one of the most important prognostic factors for patients with glioblastoma and it can predict the response to treatment with alkylating agents such as temozolomide [155]. However, it has been determined that the SNP rs16906252 (C.-56C>T) found in one of the parental alleles, is a determinant key in the acquisition of the MGMT epimutation in glioblastoma (Figure 5J). Additionally, it is known that temozolomide-treated patients with the rs16906252 T genotype have better survival probabilities, regardless of tumor methylation status [125,158].
Hereditary secondary epimutations in non-oncologic syndromes have also been reported. That is the case of the Methyl-Malonic Aciduria and Homocystinuria type cblC (MMACHC) gene, which was recently studied by Guéant et al., in 2018. This research group identified a cause of the cblC class (cobalamin, cblC), an autosomal recessive disease characterized by inborn errors in vitamin B12 metabolism, named “epi-cblC”. The cblC disorder presents both neurological and systemic metabolic abnormalities [159]. Patients with epi-cblC are heterozygotes for a genetic mutation and have a secondary epimutation at the MMACHC locus, which is flanked by the CCDC163P and PRDX1 genes and oriented in the opposite direction. Secondary epimutation was triggered by mutations in the neighboring antisense gene PRDX1 that produces an aberrant antisense transcript leading to loss of the stop codon (Figure 5K). Interestingly, this epimutation was found in three generations with PRDX1 mutations in cis (c.515-1G>T, c.515-2A>T) that strengthen the antisense transcription of MMACHC and possibly triggering the activation methylation mark H3K36me3. Furthermore, this work demonstrated that the silencing of PRDX1 transcription leads to partial epiallele hypomethylation and restoration of the expression of MMACHC [126].
Secondary epimutations have also been evaluated in breast and ovarian cancer, where people who carry germline BRCA1 and BRCA2 mutations are at high risk of developing this syndrome. In sporadic breast and ovarian tumors, epimutations of the BRCA1 promoter are well-known players of somatic carcinogenesis [160,161]. However, recently, Evans et al. (2018) identified a dominant inherited 5’UTR variant associated with epigenetic silencing of BRCA1 due to promoter hypermethylation in two families affected by breast and ovarian cancer. The clinical condition of the two families was consistent with hereditary breast and/or ovarian cancer (HBOC) but they lacked pathogenic BRCA1 or BRCA2 variants. The secondary epimutation was BRCA1 promoter hypermethylation of 10 CpG sites in these families. Hypermethylation of the BRCA1 promoter was detected in 2 of 49 independent families in the United Kingdom. The proband women were affected by high-grade breast cancer, and the hypermethylation of the BRCA1 promoter was detected in blood, oral mucosa, and hair follicle tissues. Moreover, RNA sequencing revealed the allelic loss of BRCA1 expression in both families and its cosegregation with the heterozygous variant c.-107A>T at the 5’UTR end of the BRCA1 gene (Figure 5L) [127]. Laner et al. conducted a study to detect the presence of this same variant using a competitive allele-specific PCR assay (KASP) or direct Sanger sequencing in 3297 German patients with HBOC criteria without pathogenic germline variants. In their results, they did not detect any individual carrying the variant. Therefore, they concluded that in BRCA1, the variant c.-107A>T is not common in the population of southeast Germany [162].
Research efforts are also currently directed at the improvement of epigenetic anomalies based on population studies, called “epigenetic epidemiology”. These studies are progressing in great strides by including next-generation technologies and covering a large number of patients to identify these “epivariants” or “epitypes” that condition the phenotype of the disease. It is the case of the robust study by Garg et al., 2020, on the methylation profiles of 23,116 individuals using the Illumina 450k matrix. They identified more than 4000 unique autosomal patterned epivariants potentially leading to promoter epimutations in more than 300 human disease-related tumor suppressor genes. This work suggests that epivariants may underlie a fraction of human disease that would be missed with purely genetic sequence-based approaches. This study provides a broad overview of rare epigenetic changes in the human genome, giving insight into the underlying origins and consequences of epivariations, potentially altering the methylation pattern related to human diseases [163]. In this light, it is important to distinguish the constitutional secondary epimutations that cause diseases from the common epigenetic variants, usually influenced by ethnicity and caused by non-pathogenic natural adaptive mechanisms in humans [164].

7. Conclusions

The pathogenic role of constitutional inherited secondary epimutations has substantial evidence. The cis pathogenic effect on epimutations is wide and affects imprinted genes, cancer-related genes, and other hereditary syndromes. However, as it is a rare mechanism, the pathogenic allele frequencies of these alterations among patients and the general population are still unknown and underestimated due to the absence of epigenetic monitoring of patients. Therefore, there is still a long way to go for epimutations to be included in diagnostic procedures and incorporated into the molecular detection strategies of hereditary cancers. Even in the case of the MLH1 and MSH2 genes where secondary epimutations have an established pathogenic role in disease, epigenetic screening has not yet been implemented as a routine test.
Therefore, it is key to thoroughly describe, determine the population frequencies, and understand the inheritance mechanisms of secondary epimutations, an effort that will undoubtedly contribute to the diagnosis, prevention, and management of heredo-familial diseases.

Author Contributions

Conceptualization and design, F.V.P. and M.R.d.l.C.; Formal analysis and investigation of the literature, M.R.d.l.C., A.H.d.l.C.M., E.A.R.J., H.M.G., C.E.D.V., J.P.d.l.V., F.d.l.C.H.-H., F.V.P.; Writing—original draft preparation, M.R.d.l.C., A.H.d.l.C.M., E.A.R.J., H.M.G., C.E.D.V., J.P.d.l.V., F.d.l.C.H.-H., F.V.P.; Writing—review and editing, F.V.P. and M.R.d.l.C.; Visualization and figures, M.R.d.l.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  2. Allis, C.D.; Jenuwein, T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet. 2016, 17, 487–500. [Google Scholar] [CrossRef]
  3. Baylin, S.B.; Jones, P.A. Epigenetic determinants of cancer. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef] [Green Version]
  4. Lu, Y.; Chan, Y.T.; Tan, H.Y.; Li, S.; Wang, N.; Feng, Y. Epigenetic regulation in human cancer: The potential role of epi-drug in cancer therapy. Mol. Cancer 2020, 19, 79. [Google Scholar] [CrossRef]
  5. Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2017, 18, 5–18. [Google Scholar] [CrossRef]
  6. Hayes, J.; Peruzzi, P.P.; Lawler, S. MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol. Med. 2014, 20, 460–469. [Google Scholar] [CrossRef]
  7. Ding, H.X.; Lv, Z.; Yuan, Y.; Xu, Q. MiRNA Polymorphisms and Cancer Prognosis: A Systematic Review and Meta-Analysis. Front. Oncol. 2018, 8, 596. [Google Scholar] [CrossRef]
  8. Carlevaro-Fita, J.; Lanzos, A.; Feuerbach, L.; Hong, C.; Mas-Ponte, D.; Pedersen, J.S.; Drivers, P.; Functional Interpretation, G.; Johnson, R.; Consortium, P. Cancer LncRNA Census reveals evidence for deep functional conservation of long noncoding RNAs in tumorigenesis. Commun. Biol. 2020, 3, 56. [Google Scholar] [CrossRef]
  9. Verduci, L.; Tarcitano, E.; Strano, S.; Yarden, Y.; Blandino, G. CircRNAs: Role in human diseases and potential use as biomarkers. Cell Death. Dis. 2021, 12, 468. [Google Scholar] [CrossRef]
  10. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef] [Green Version]
  11. Huarte, M. The emerging role of lncRNAs in cancer. Nat. Med. 2015, 21, 1253–1261. [Google Scholar] [CrossRef]
  12. Kristensen, L.S.; Hansen, T.B.; Veno, M.T.; Kjems, J. Circular RNAs in cancer: Opportunities and challenges in the field. Oncogene 2018, 37, 555–565. [Google Scholar] [CrossRef] [Green Version]
  13. Mir, M.; Bickmore, W.; Furlong, E.E.M.; Narlikar, G. Chromatin topology, condensates and gene regulation: Shifting paradigms or just a phase? Development 2019, 146. [Google Scholar] [CrossRef] [Green Version]
  14. Schuster-Böckler, B.; Lehner, B. Chromatin organization is a major influence on regional mutation rates in human cancer cells. Nature 2012, 488, 504–507. [Google Scholar] [CrossRef]
  15. Smith, K.S.; Liu, L.L.; Ganesan, S.; Michor, F.; De, S. Nuclear topology modulates the mutational landscapes of cancer genomes. Nat. Struct. Mol. Biol. 2017, 24, 1000–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Entrevan, M.; Schuettengruber, B.; Cavalli, G. Regulation of Genome Architecture and Function by Polycomb Proteins. Trends Cell Biol. 2016, 26, 511–525. [Google Scholar] [CrossRef]
  17. Ito, T.; Teo, Y.V.; Evans, S.A.; Neretti, N.; Sedivy, J.M. Regulation of Cellular Senescence by Polycomb Chromatin Modifiers through Distinct DNA Damage- and Histone Methylation-Dependent Pathways. Cell Rep. 2018, 22, 3480–3492. [Google Scholar] [CrossRef] [Green Version]
  18. Mills, A.A. Throwing the cancer switch: Reciprocal roles of polycomb and trithorax proteins. Nat. Rev. Cancer 2010, 10, 669–682. [Google Scholar] [CrossRef]
  19. Soares, J.; Pinto, A.E.; Cunha, C.V.; Andre, S.; Barao, I.; Sousa, J.M.; Cravo, M. Global DNA hypomethylation in breast carcinoma: Correlation with prognostic factors and tumor progression. Cancer 1999, 85, 112–118. [Google Scholar] [CrossRef]
  20. Herman, J.G.; Graff, J.R.; Myohanen, S.; Nelkin, B.D.; Baylin, S.B. Methylation-specific PCR: A novel PCR assay for methylation status of CpG islands. Proc. Natl. Acad. Sci. USA 1996, 93, 9821–9826. [Google Scholar] [CrossRef] [Green Version]
  21. Clark, S.J.; Statham, A.; Stirzaker, C.; Molloy, P.L.; Frommer, M. DNA methylation: Bisulphite modification and analysis. Nat. Protoc. 2006, 1, 2353–2364. [Google Scholar] [CrossRef] [PubMed]
  22. Eads, C.A.; Danenberg, K.D.; Kawakami, K.; Saltz, L.B.; Blake, C.; Shibata, D.; Danenberg, P.V.; Laird, P.W. MethyLight: A high-throughput assay to measure DNA methylation. Nucleic Acids Res. 2000, 28, E32. [Google Scholar] [CrossRef] [Green Version]
  23. Khulan, B.; Thompson, R.F.; Ye, K.; Fazzari, M.J.; Suzuki, M.; Stasiek, E.; Figueroa, M.E.; Glass, J.L.; Chen, Q.; Montagna, C.; et al. Comparative isoschizomer profiling of cytosine methylation: The HELP assay. Genome Res. 2006, 16, 1046–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hatada, I.; Fukasawa, M.; Kimura, M.; Morita, S.; Yamada, K.; Yoshikawa, T.; Yamanaka, S.; Endo, C.; Sakurada, A.; Sato, M.; et al. Genome-wide profiling of promoter methylation in human. Oncogene 2006, 25, 3059–3064. [Google Scholar] [CrossRef] [Green Version]
  25. Sandoval, J.; Heyn, H.; Moran, S.; Serra-Musach, J.; Pujana, M.A.; Bibikova, M.; Esteller, M. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics 2011, 6, 692–702. [Google Scholar] [CrossRef]
  26. Meissner, A.; Gnirke, A.; Bell, G.W.; Ramsahoye, B.; Lander, E.S.; Jaenisch, R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 2005, 33, 5868–5877. [Google Scholar] [CrossRef] [Green Version]
  27. Lu, H.; Yuan, Z.; Tan, T.; Wang, J.; Zhang, J.; Luo, H.J.; Xia, Y.; Ji, W.; Gao, F. Improved tagmentation-based whole-genome bisulfite sequencing for input DNA from less than 100 mammalian cells. Epigenomics 2015, 7, 47–56. [Google Scholar] [CrossRef]
  28. Laird, P.W. Principles and challenges of genomewide DNA methylation analysis. Nat. Rev. Genet. 2010, 11, 191–203. [Google Scholar] [CrossRef]
  29. Kulis, M.; Esteller, M. DNA methylation and cancer. Adv. Genet. 2010, 70, 27–56. [Google Scholar] [CrossRef]
  30. Silverman, B.R.; Shi, J. Alterations of epigenetic regulators in pancreatic cancer and their clinical implications. Int. J. Mol. Sci. 2016, 17, 2138. [Google Scholar] [CrossRef] [Green Version]
  31. Ji, P.; Wang, X.; Xie, N.; Li, Y. N6-Methyladenosine in RNA and DNA: An Epitranscriptomic and Epigenetic Player Implicated in Determination of Stem Cell Fate. Stem Cells Int. 2018, 2018, 3256524. [Google Scholar] [CrossRef]
  32. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002, 16, 6–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bock, C. Analysing and interpreting DNA methylation data. Nat. Rev. Genet. 2012, 13, 705–719. [Google Scholar] [CrossRef]
  34. Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef] [Green Version]
  35. Yokochi, T.; Robertson, K.D. Preferential methylation of unmethylated DNA by mammalian de novo DNA methyltransferase Dnmt3a. J. Biol. Chem. 2002, 277, 11735–11745. [Google Scholar] [CrossRef] [Green Version]
  36. Laird, P.W. The power and the promise of DNA methylation markers. Nat. Rev. Cancer 2003, 3, 253–266. [Google Scholar] [CrossRef]
  37. Tang, W.W.C.; Dietmann, S.; Irie, N.; Leitch, H.G.; Floros, V.I.; Bradshaw, C.R.; Hackett, J.A.; Chinnery, P.F.; Surani, M.A. A unique gene regulatory network resets the human germline epigenome for development. Cell 2015, 161, 1453–1467. [Google Scholar] [CrossRef] [Green Version]
  38. Walsh, C.P.; Chaillet, J.R.; Bestor, T.H. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat. Genet. 1998, 20, 116–117. [Google Scholar] [CrossRef]
  39. Hark, A.T.; Schoenherr, C.J.; Katz, D.J.; Ingram, R.S.; Levorse, J.M.; Tilghman, S.M. CTCF mediates methylation-sensitive enhancer-blocking activity at the H19/Igf2 locus. Nature 2000, 405, 486–489. [Google Scholar] [CrossRef]
  40. Cotton, A.M.; Price, E.M.; Jones, M.J.; Balaton, B.P.; Kobor, M.S.; Brown, C.J. Landscape of DNA methylation on the X chromosome reflects CpG density, functional chromatin state and X-chromosome inactivation. Hum. Mol. Genet. 2015, 24, 1528–1539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Westerman, K.; Sebastiani, P.; Jacques, P.; Liu, S.; Demeo, D.; Ordovás, J.M. DNA methylation modules associate with incident cardiovascular disease and cumulative risk factor exposure. Clin. Epigenetics 2019, 11, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhong, J.; Agha, G.; Baccarelli, A.A. The Role of DNA Methylation in Cardiovascular Risk and Disease. Methodol. Asp. Study Des. Data Anal. Epidemiol. Stud. 2016, 118, 119–131. [Google Scholar] [CrossRef] [Green Version]
  43. Capper, D.; Jones, D.T.W.; Sill, M.; Hovestadt, V.; Schrimpf, D.; Sturm, D.; Koelsche, C.; Sahm, F.; Chavez, L.; Reuss, D.E.; et al. DNA methylation-based classification of central nervous system tumours. Nature 2018, 555, 469–474. [Google Scholar] [CrossRef] [PubMed]
  44. Venkataramani, V.; Tanev, D.I.; Strahle, C.; Studier-Fischer, A.; Fankhauser, L.; Kessler, T.; Körber, C.; Kardorff, M.; Ratliff, M.; Xie, R.; et al. Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature 2019, 573, 532–538. [Google Scholar] [CrossRef]
  45. Choong, M.K.; Tsafnat, G. Genetic and epigenetic biomarkers of colorectal cancer. Clin. Gastroenterol. Hepatol. 2012, 10, 9–15. [Google Scholar] [CrossRef]
  46. Flavahan, W.A.; Gaskell, E.; Bernstein, B.E. Epigenetic plasticity and the hallmarks of cancer. Science 2017, 357. [Google Scholar] [CrossRef] [Green Version]
  47. Perry, A.S.; Watson, R.W.G.; Lawler, M.; Hollywood, D. The epigenome as a therapeutic target in prostate cancer. Nat. Rev. Urol. 2010, 7, 668–680. [Google Scholar] [CrossRef] [PubMed]
  48. Oey, H.; Whitelaw, E. On the meaning of the word ‘epimutation’. Trends Genet. 2014, 30, 519–520. [Google Scholar] [CrossRef] [PubMed]
  49. Hesson, L.B.; Hitchins, M.P.; Ward, R.L. Epimutations and cancer predisposition: Importance and mechanisms. Curr. Opin. Genet. Dev. 2010, 20, 290–298. [Google Scholar] [CrossRef] [PubMed]
  50. Holliday, R. The inheritance of epigenetic defects. Science 1987, 238, 163–170. [Google Scholar] [CrossRef]
  51. Haertle, L.; Maierhofer, A.; Bock, J.; Lehnen, H.; Bottcher, Y.; Bluher, M.; Schorsch, M.; Potabattula, R.; El Hajj, N.; Appenzeller, S.; et al. Hypermethylation of the non-imprinted maternal MEG3 and paternal MEST alleles is highly variable among normal individuals. PLoS ONE 2017, 12, e0184030. [Google Scholar] [CrossRef] [Green Version]
  52. Horsthemke, B. Epimutations in human disease. Curr. Top. Microbiol. Immunol. 2006, 310, 45–59. [Google Scholar] [CrossRef]
  53. Watson, I.R.; Takahashi, K.; Futreal, P.A.; Chin, L. Emerging patterns of somatic mutations in cancer. Nat. Rev. Genet. 2013, 14, 703–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mohrenweiser, H.W.; Jones, I.M. Review of the molecular characteristics of gene mutations of the germline and somatic cells of the human. Mutat. Res. 1990, 231, 87–108. [Google Scholar] [CrossRef]
  55. Hitchins, M.P.; Rapkins, R.W.; Kwok, C.T.; Srivastava, S.; Wong, J.J.; Khachigian, L.M.; Polly, P.; Goldblatt, J.; Ward, R.L. Dominantly inherited constitutional epigenetic silencing of MLH1 in a cancer-affected family is linked to a single nucleotide variant within the 5’UTR. Cancer Cell 2011, 20, 200–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Sloane, M.A.; Ward, R.L.; Hesson, L.B. Defining the criteria for identifying constitutional epimutations. Clin. Epigenetics 2016, 8, 39. [Google Scholar] [CrossRef] [Green Version]
  57. Santos, F.; Hendrich, B.; Reik, W.; Dean, W. Dynamic reprogramming of DNA methylation in the early mouse embryo. Dev. Biol. 2002, 241, 172–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Hitchins, M.P. Constitutional epimutation as a mechanism for cancer causality and heritability? Nat. Rev. Cancer 2015, 15, 625–634. [Google Scholar] [CrossRef] [PubMed]
  59. Hitchins, M.P.; Ward, R.L. Erasure of MLH1 methylation in spermatozoa-implications for epigenetic inheritance. Nat. Genet. 2007, 39, 1289. [Google Scholar] [CrossRef] [PubMed]
  60. Smallwood, S.A.; Tomizawa, S.; Krueger, F.; Ruf, N.; Carli, N.; Segonds-Pichon, A.; Sato, S.; Hata, K.; Andrews, S.R.; Kelsey, G. Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nat. Genet. 2011, 43, 811–814. [Google Scholar] [CrossRef] [Green Version]
  61. Lonning, P.E.; Eikesdal, H.P.; Loes, I.M.; Knappskog, S. Constitutional Mosaic Epimutations-a hidden cause of cancer? Cell Stress 2019, 3, 118–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Cropley, J.E.; Martin, D.I.; Suter, C.M. Germline epimutation in humans. Pharmacogenomics 2008, 9, 1861–1868. [Google Scholar] [CrossRef] [PubMed]
  63. Sakai, T.; Toguchida, J.; Ohtani, N.; Yandell, D.W.; Rapaport, J.M.; Dryja, T.P. Allele-specific hypermethylation of the retinoblastoma tumor-suppressor gene. Am. J. Hum. Genet. 1991, 48, 880–888. [Google Scholar] [PubMed]
  64. Herman, J.G.; Latif, F.; Weng, Y.; Lerman, M.I.; Zbar, B.; Liu, S.; Samid, D.; Duan, D.S.; Gnarra, J.R.; Linehan, W.M.; et al. Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc. Natl. Acad. Sci. USA 1994, 91, 9700–9704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kane, M.F.; Loda, M.; Gaida, G.M.; Lipman, J.; Mishra, R.; Goldman, H.; Jessup, J.M.; Kolodner, R. Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res. 1997, 57, 808–811. [Google Scholar] [PubMed]
  66. Dobrovic, A.; Simpfendorfer, D. Methylation of the BRCA1 gene in sporadic breast cancer. Cancer Res. 1997, 57, 3347–3350. [Google Scholar]
  67. Banno, K.; Kisu, I.; Yanokura, M.; Tsuji, K.; Masuda, K.; Ueki, A.; Kobayashi, Y.; Yamagami, W.; Nomura, H.; Tominaga, E.; et al. Epimutation and cancer: A new carcinogenic mechanism of Lynch syndrome (Review). Int. J. Oncol. 2012, 41, 793–797. [Google Scholar] [CrossRef] [Green Version]
  68. Petronis, A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 2010, 465, 721–727. [Google Scholar] [CrossRef]
  69. Knudson, A.G., Jr. Mutation and cancer: Statistical study of retinoblastoma. Proc. Natl. Acad. Sci. USA 1971, 68, 820–823. [Google Scholar] [CrossRef] [Green Version]
  70. Tucker, T.; Friedman, J.M. Pathogenesis of hereditary tumors: Beyond the "two-hit" hypothesis. Clin. Genet. 2002, 62, 345–357. [Google Scholar] [CrossRef]
  71. Mossman, D.; Scott, R.J. Epimutations, inheritance and causes of aberrant DNA methylation in cancer. Hered. Cancer Clin. Pr. 2006, 4, 75–80. [Google Scholar] [CrossRef] [Green Version]
  72. Hitchins, M.P. The role of epigenetics in Lynch syndrome. Fam. Cancer 2013, 12, 189–205. [Google Scholar] [CrossRef]
  73. Gelli, E.; Pinto, A.M.; Somma, S.; Imperatore, V.; Cannone, M.G.; Hadjistilianou, T.; De Francesco, S.; Galimberti, D.; Currò, A.; Bruttini, M.; et al. Evidence of predisposing epimutation in retinoblastoma. Hum. Mutat. 2019, 40, 201–206. [Google Scholar] [CrossRef]
  74. Esteller, M.; Fraga, M.F.; Guo, M.; Garcia-Foncillas, J.; Hedenfalk, I.; Godwin, A.K.; Trojan, J.; Vaurs-Barrière, C.; Bignon, Y.J.; Ramus, S.; et al. DNA methylation patterns in hereditary human cancers mimic sporadic tumorigenesis. Hum. Mol. Genet. 2001, 10, 3001–3007. [Google Scholar] [CrossRef] [Green Version]
  75. Zhang, X.Y.; Asiedu, C.K.; Supakar, P.C.; Khan, R.; Ehrlich, K.C.; Ehrlich, M. Binding sites in mammalian genes and viral gene regulatory regions recognized by methylated DNA-binding protein. Nucleic Acids Res. 1990, 18, 6253–6260. [Google Scholar] [CrossRef] [Green Version]
  76. Zhu, H.; Wang, G.; Qian, J. Transcription factors as readers and effectors of DNA methylation. Nat. Rev. Genet. 2016, 17, 551–565. [Google Scholar] [CrossRef] [PubMed]
  77. Raval, A.; Tanner, S.M.; Byrd, J.C.; Angerman, E.B.; Perko, J.D.; Chen, S.S.; Hackanson, B.; Grever, M.R.; Lucas, D.M.; Matkovic, J.J.; et al. Downregulation of death-associated protein kinase 1 (DAPK1) in chronic lymphocytic leukemia. Cell 2007, 129, 879–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Gazzoli, I.; Loda, M.; Garber, J.; Syngal, S.; Kolodner, R.D. A hereditary nonpolyposis colorectal carcinoma case associated with hypermethylation of the MLH1 gene in normal tissue and loss of heterozygosity of the unmethylated allele in the resulting microsatellite instability-high tumor. Cancer Res. 2002, 62, 3925–3928. [Google Scholar] [PubMed]
  79. Suter, C.M.; Martin, D.I.; Ward, R.L. Germline epimutation of MLH1 in individuals with multiple cancers. Nat. Genet. 2004, 36, 497–501. [Google Scholar] [CrossRef]
  80. Tiffon, C. The Impact of Nutrition and Environmental Epigenetics on Human Health and Disease. Int. J. Mol. Sci. 2018, 19, 3425. [Google Scholar] [CrossRef] [Green Version]
  81. Moulton, T.; Crenshaw, T.; Hao, Y.; Moosikasuwan, J.; Lin, N.; Dembitzer, F.; Hensle, T.; Weiss, L.; McMorrow, L.; Loew, T.; et al. Epigenetic lesions at the H19 locus in Wilms’ tumour patients. Nat. Genet. 1994, 7, 440–447. [Google Scholar] [CrossRef] [PubMed]
  82. Argos, M. Arsenic Exposure and Epigenetic Alterations: Recent Findings Based on the Illumina 450K DNA Methylation Array. Curr. Environ. Health Rep. 2015, 2, 137–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Philibert, R.A.; Beach, S.R.; Brody, G.H. Demethylation of the aryl hydrocarbon receptor repressor as a biomarker for nascent smokers. Epigenetics 2012, 7, 1331–1338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zeilinger, S.; Kuhnel, B.; Klopp, N.; Baurecht, H.; Kleinschmidt, A.; Gieger, C.; Weidinger, S.; Lattka, E.; Adamski, J.; Peters, A.; et al. Tobacco smoking leads to extensive genome-wide changes in DNA methylation. PLoS ONE 2013, 8, e63812. [Google Scholar] [CrossRef] [PubMed]
  85. Thomson, J.P.; Ottaviano, R.; Unterberger, E.B.; Lempiainen, H.; Muller, A.; Terranova, R.; Illingworth, R.S.; Webb, S.; Kerr, A.R.; Lyall, M.J.; et al. Loss of Tet1-Associated 5-Hydroxymethylcytosine Is Concomitant with Aberrant Promoter Hypermethylation in Liver Cancer. Cancer Res. 2016, 76, 3097–3108. [Google Scholar] [CrossRef] [Green Version]
  86. Meehan, R.R.; Thomson, J.P.; Lentini, A.; Nestor, C.E.; Pennings, S. DNA methylation as a genomic marker of exposure to chemical and environmental agents. Curr. Opin. Chem. Biol. 2018, 45, 48–56. [Google Scholar] [CrossRef]
  87. Liu, L.; Wylie, R.C.; Andrews, L.G.; Tollefsbol, T.O. Aging, cancer and nutrition: The DNA methylation connection. Mech. Ageing Dev. 2003, 124, 989–998. [Google Scholar] [CrossRef]
  88. McCabe, D.C.; Caudill, M.A. DNA methylation, genomic silencing, and links to nutrition and cancer. Nutr. Rev. 2005, 63, 183–195. [Google Scholar] [CrossRef]
  89. Zeisel, S. Choline, Other Methyl-Donors and Epigenetics. Nutrients 2017, 9, 445. [Google Scholar] [CrossRef]
  90. Brunaud, L.; Alberto, J.M.; Ayav, A.; Gerard, P.; Namour, F.; Antunes, L.; Braun, M.; Bronowicki, J.P.; Bresler, L.; Gueant, J.L. Effects of vitamin B12 and folate deficiencies on DNA methylation and carcinogenesis in rat liver. Clin. Chem. Lab. Med. 2003, 41, 1012–1019. [Google Scholar] [CrossRef]
  91. Wu, J.C.; Santi, D.V. On the mechanism and inhibition of DNA cytosine methyltransferases. Prog. Clin. Biol. Res. 1985, 198, 119–129. [Google Scholar]
  92. Ross, S.A. Diet and DNA methylation interactions in cancer prevention. Ann. N. Y. Acad. Sci. 2003, 983, 197–207. [Google Scholar] [CrossRef] [PubMed]
  93. Garfinkel, M.D.; Ruden, D.M. Chromatin effects in nutrition, cancer, and obesity. Nutrition 2004, 20, 56–62. [Google Scholar] [CrossRef]
  94. Kim, Y.I. Folate and DNA methylation: A mechanistic link between folate deficiency and colorectal cancer? Cancer Epidemiol. Prev. Biomark. 2004, 13, 511–519. [Google Scholar]
  95. Morgan, A.E.; Davies, T.J.; Mc Auley, M.T. The role of DNA methylation in ageing and cancer. Proc. Nutr. Soc. 2018, 77, 412–422. [Google Scholar] [CrossRef] [PubMed]
  96. Jung, M.; Pfeifer, G.P. Aging and DNA methylation. BMC Biol. 2015, 13, 7. [Google Scholar] [CrossRef] [Green Version]
  97. Esteller, M. Epigenetics provides a new generation of oncogenes and tumour-suppressor genes. Br. J. Cancer 2006, 94, 179–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Conrad, D.F.; Pinto, D.; Redon, R.; Feuk, L.; Gokcumen, O.; Zhang, Y.; Aerts, J.; Andrews, T.D.; Barnes, C.; Campbell, P.; et al. Origins and functional impact of copy number variation in the human genome. Nature 2010, 464, 704–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Hitchins, M.P.; Wong, J.J.; Suthers, G.; Suter, C.M.; Martin, D.I.; Hawkins, N.J.; Ward, R.L. Inheritance of a cancer-associated MLH1 germ-line epimutation. N. Engl. J. Med. 2007, 356, 697–705. [Google Scholar] [CrossRef] [Green Version]
  100. Pineda, M.; Mur, P.; Iniesta, M.D.; Borras, E.; Campos, O.; Vargas, G.; Iglesias, S.; Fernandez, A.; Gruber, S.B.; Lazaro, C.; et al. MLH1 methylation screening is effective in identifying epimutation carriers. Eur. J. Hum. Genet. 2012, 20, 1256–1264. [Google Scholar] [CrossRef] [Green Version]
  101. Gibbons, R.J.; McDowell, T.L.; Raman, S.; O’Rourke, D.M.; Garrick, D.; Ayyub, H.; Higgs, D.R. Mutations in ATRX, encoding a SWI/SNF-like protein, cause diverse changes in the pattern of DNA methylation. Nat. Genet. 2000, 24, 368–371. [Google Scholar] [CrossRef] [Green Version]
  102. Amir, R.E.; Van den Veyver, I.B.; Wan, M.; Tran, C.Q.; Francke, U.; Zoghbi, H.Y. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 1999, 23, 185–188. [Google Scholar] [CrossRef]
  103. Fu, Y.H.; Kuhl, D.P.; Pizzuti, A.; Pieretti, M.; Sutcliffe, J.S.; Richards, S.; Verkerk, A.J.; Holden, J.J.; Fenwick, R.G., Jr.; Warren, S.T.; et al. Variation of the CGG repeat at the fragile X site results in genetic instability: Resolution of the Sherman paradox. Cell 1991, 67, 1047–1058. [Google Scholar] [CrossRef]
  104. Verkerk, A.J.; Pieretti, M.; Sutcliffe, J.S.; Fu, Y.H.; Kuhl, D.P.; Pizzuti, A.; Reiner, O.; Richards, S.; Victoria, M.F.; Zhang, F.P.; et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell 1991, 65, 905–914. [Google Scholar] [CrossRef]
  105. Stoger, R.; Kajimura, T.M.; Brown, W.T.; Laird, C.D. Epigenetic variation illustrated by DNA methylation patterns of the fragile-X gene FMR1. Hum. Mol. Genet. 1997, 6, 1791–1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Bielinska, B.; Blaydes, S.M.; Buiting, K.; Yang, T.; Krajewska-Walasek, M.; Horsthemke, B.; Brannan, C.I. De novo deletions of SNRPN exon 1 in early human and mouse embryos result in a paternal to maternal imprint switch. Nat. Genet. 2000, 25, 74–78. [Google Scholar] [CrossRef] [PubMed]
  107. Perk, J.; Makedonski, K.; Lande, L.; Cedar, H.; Razin, A.; Shemer, R. The imprinting mechanism of the Prader-Willi/Angelman regional control center. EMBO J. 2002, 21, 5807–5814. [Google Scholar] [CrossRef] [Green Version]
  108. Dittrich, B.; Buiting, K.; Korn, B.; Rickard, S.; Buxton, J.; Saitoh, S.; Nicholls, R.D.; Poustka, A.; Winterpacht, A.; Zabel, B.; et al. Imprint switching on human chromosome 15 may involve alternative transcripts of the SNRPN gene. Nat. Genet. 1996, 14, 163–170. [Google Scholar] [CrossRef]
  109. Glenn, C.C.; Saitoh, S.; Jong, M.T.; Filbrandt, M.M.; Surti, U.; Driscoll, D.J.; Nicholls, R.D. Gene structure, DNA methylation, and imprinted expression of the human SNRPN gene. Am. J. Hum. Genet. 1996, 58, 335–346. [Google Scholar]
  110. Sutcliffe, J.S.; Nakao, M.; Christian, S.; Orstavik, K.H.; Tommerup, N.; Ledbetter, D.H.; Beaudet, A.L. Deletions of a differentially methylated CpG island at the SNRPN gene define a putative imprinting control region. Nat. Genet. 1994, 8, 52–58. [Google Scholar] [CrossRef] [PubMed]
  111. Buiting, K.; Saitoh, S.; Gross, S.; Dittrich, B.; Schwartz, S.; Nicholls, R.D.; Horsthemke, B. Inherited microdeletions in the Angelman and Prader-Willi syndromes define an imprinting centre on human chromosome 15. Nat. Genet. 1995, 9, 395–400. [Google Scholar] [CrossRef]
  112. Dittrich, B.; Buiting, K.; Gross, S.; Horsthemke, B. Characterization of a methylation imprint in the Prader-Willi syndrome chromosome region. Hum. Mol. Genet. 1993, 2, 1995–1999. [Google Scholar] [CrossRef]
  113. Buiting, K.; Dittrich, B.; Robinson, W.P.; Guitart, M.; Abeliovich, D.; Lerer, I.; Horsthemke, B. Detection of aberrant DNA methylation in unique Prader-Willi syndrome patients and its diagnostic implications. Hum. Mol. Genet. 1994, 3, 893–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Glenn, C.C.; Nicholls, R.D.; Robinson, W.P.; Saitoh, S.; Niikawa, N.; Schinzel, A.; Horsthemke, B.; Driscoll, D.J. Modification of 15q11-q13 DNA methylation imprints in unique Angelman and Prader-Willi patients. Hum. Mol. Genet. 1993, 2, 1377–1382. [Google Scholar] [CrossRef] [Green Version]
  115. Glenn, C.C.; Porter, K.A.; Jong, M.T.; Nicholls, R.D.; Driscoll, D.J. Functional imprinting and epigenetic modification of the human SNRPN gene. Hum. Mol. Genet. 1993, 2, 2001–2005. [Google Scholar] [CrossRef]
  116. Buiting, K.; Gross, S.; Lich, C.; Gillessen-Kaesbach, G.; el-Maarri, O.; Horsthemke, B. Epimutations in Prader-Willi and Angelman syndromes: A molecular study of 136 patients with an imprinting defect. Am. J. Hum. Genet. 2003, 72, 571–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. El-Maarri, O.; Buiting, K.; Peery, E.G.; Kroisel, P.M.; Balaban, B.; Wagner, K.; Urman, B.; Heyd, J.; Lich, C.; Brannan, C.I.; et al. Maternal methylation imprints on human chromosome 15 are established during or after fertilization. Nat. Genet. 2001, 27, 341–344. [Google Scholar] [CrossRef] [PubMed]
  118. Barbour, V.M.; Tufarelli, C.; Sharpe, J.A.; Smith, Z.E.; Ayyub, H.; Heinlein, C.A.; Sloane-Stanley, J.; Indrak, K.; Wood, W.G.; Higgs, D.R. alpha-thalassemia resulting from a negative chromosomal position effect. Blood 2000, 96, 800–807. [Google Scholar] [CrossRef] [PubMed]
  119. Hitchins, M.P. Inheritance of epigenetic aberrations (constitutional epimutations) in cancer susceptibility. Adv. Genet. 2010, 70, 201–243. [Google Scholar] [CrossRef]
  120. Tufarelli, C.; Stanley, J.A.; Garrick, D.; Sharpe, J.A.; Ayyub, H.; Wood, W.G.; Higgs, D.R. Transcription of antisense RNA leading to gene silencing and methylation as a novel cause of human genetic disease. Nat. Genet. 2003, 34, 157–165. [Google Scholar] [CrossRef]
  121. Sparago, A.; Cerrato, F.; Vernucci, M.; Ferrero, G.B.; Silengo, M.C.; Riccio, A. Microdeletions in the human H19 DMR result in loss of IGF2 imprinting and Beckwith-Wiedemann syndrome. Nat. Genet. 2004, 36, 958–960. [Google Scholar] [CrossRef] [PubMed]
  122. Niemitz, E.L.; DeBaun, M.R.; Fallon, J.; Murakami, K.; Kugoh, H.; Oshimura, M.; Feinberg, A.P. Microdeletion of LIT1 in familial Beckwith-Wiedemann syndrome. Am. J. Hum. Genet. 2004, 75, 844–849. [Google Scholar] [CrossRef] [Green Version]
  123. Ligtenberg, M.J.; Kuiper, R.P.; Chan, T.L.; Goossens, M.; Hebeda, K.M.; Voorendt, M.; Lee, T.Y.; Bodmer, D.; Hoenselaar, E.; Hendriks-Cornelissen, S.J.; et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3’ exons of TACSTD1. Nat. Genet. 2009, 41, 112–117. [Google Scholar] [CrossRef]
  124. Pinheiro, H.; Bordeira-Carrico, R.; Seixas, S.; Carvalho, J.; Senz, J.; Oliveira, P.; Inacio, P.; Gusmao, L.; Rocha, J.; Huntsman, D.; et al. Allele-specific CDH1 downregulation and hereditary diffuse gastric cancer. Hum. Mol. Genet. 2010, 19, 943–952. [Google Scholar] [CrossRef] [Green Version]
  125. Rapkins, R.W.; Wang, F.; Nguyen, H.N.; Cloughesy, T.F.; Lai, A.; Ha, W.; Nowak, A.K.; Hitchins, M.P.; McDonald, K.L. The MGMT promoter SNP rs16906252 is a risk factor for MGMT methylation in glioblastoma and is predictive of response to temozolomide. Neuro Oncol. 2015, 17, 1589–1598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Gueant, J.L.; Chery, C.; Oussalah, A.; Nadaf, J.; Coelho, D.; Josse, T.; Flayac, J.; Robert, A.; Koscinski, I.; Gastin, I.; et al. A PRDX1 mutant allele causes a MMACHC secondary epimutation in cblC patients. Nat. Commun. 2018, 9, 554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Evans, D.G.R.; van Veen, E.M.; Byers, H.J.; Wallace, A.J.; Ellingford, J.M.; Beaman, G.; Santoyo-Lopez, J.; Aitman, T.J.; Eccles, D.M.; Lalloo, F.I.; et al. A Dominantly Inherited 5’ UTR Variant Causing Methylation-Associated Silencing of BRCA1 as a Cause of Breast and Ovarian Cancer. Am. J. Hum. Genet. 2018, 103, 213–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Cooper, W.N.; Luharia, A.; Evans, G.A.; Raza, H.; Haire, A.C.; Grundy, R.; Bowdin, S.C.; Riccio, A.; Sebastio, G.; Bliek, J.; et al. Molecular subtypes and phenotypic expression of Beckwith-Wiedemann syndrome. Eur. J. Hum. Genet. 2005, 13, 1025–1032. [Google Scholar] [CrossRef]
  129. Elliott, M.; Bayly, R.; Cole, T.; Temple, I.K.; Maher, E.R. Clinical features and natural history of Beckwith-Wiedemann syndrome: Presentation of 74 new cases. Clin. Genet. 1994, 46, 168–174. [Google Scholar] [CrossRef]
  130. Elliott, M.; Maher, E.R. Beckwith-Wiedemann syndrome. J. Med. Genet. 1994, 31, 560–564. [Google Scholar] [CrossRef] [Green Version]
  131. Beckwith, J.B. National Wilms Tumor Study: An update for pathologists. Pediatr. Dev. Pathol. 1998, 1, 79–84. [Google Scholar] [CrossRef]
  132. Everman, D.B.; Shuman, C.; Dzolganovski, B.; O’Riordan M, A.; Weksberg, R.; Robin, N.H. Serum alpha-fetoprotein levels in Beckwith-Wiedemann syndrome. J. Pediatr. 2000, 137, 123–127. [Google Scholar] [CrossRef]
  133. Frevel, M.A.; Sowerby, S.J.; Petersen, G.B.; Reeve, A.E. Methylation sequencing analysis refines the region of H19 epimutation in Wilms tumor. J. Biol. Chem. 1999, 274, 29331–29340. [Google Scholar] [CrossRef] [Green Version]
  134. DeBaun, M.R.; Niemitz, E.L.; Feinberg, A.P. Association of In Vitro fertilization with Beckwith-Wiedemann syndrome and epigenetic alterations of LIT1 and H19. Am. J. Hum. Genet. 2003, 72, 156–160. [Google Scholar] [CrossRef] [Green Version]
  135. DeBaun, M.R.; Niemitz, E.L.; McNeil, D.E.; Brandenburg, S.A.; Lee, M.P.; Feinberg, A.P. Epigenetic alterations of H19 and LIT1 distinguish patients with Beckwith-Wiedemann syndrome with cancer and birth defects. Am. J. Hum. Genet. 2002, 70, 604–611. [Google Scholar] [CrossRef] [Green Version]
  136. Lee, M.P.; DeBaun, M.R.; Mitsuya, K.; Galonek, H.L.; Brandenburg, S.; Oshimura, M.; Feinberg, A.P. Loss of imprinting of a paternally expressed transcript, with antisense orientation to KVLQT1, occurs frequently in Beckwith-Wiedemann syndrome and is independent of insulin-like growth factor II imprinting. Proc. Natl. Acad. Sci. USA 1999, 96, 5203–5208. [Google Scholar] [CrossRef] [Green Version]
  137. Slager, S.L.; Kay, N.E. Familial chronic lymphocytic leukemia: What does it mean to me? Clin. Lymphoma Myeloma 2009, 9 (Suppl. 3), S194–S197. [Google Scholar] [CrossRef] [Green Version]
  138. Hitchins, M.P. Finding the needle in a haystack: Identification of cases of Lynch syndrome with MLH1 epimutation. Fam. Cancer 2016, 15, 413–422. [Google Scholar] [CrossRef]
  139. Lynch, H.T.; Snyder, C.L.; Shaw, T.G.; Heinen, C.D.; Hitchins, M.P. Milestones of Lynch syndrome: 1895–2015. Nat. Rev. Cancer 2015, 15, 181–194. [Google Scholar] [CrossRef]
  140. Marmol, I.; Sanchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [Green Version]
  141. Peltomaki, P.; Vasen, H. Mutations associated with HNPCC predisposition–Update of ICG-HNPCC/INSiGHT mutation database. Dis. Markers 2004, 20, 269–276. [Google Scholar] [CrossRef] [Green Version]
  142. Umar, A.; Boland, C.R.; Terdiman, J.P.; Syngal, S.; de la Chapelle, A.; Rüschoff, J.; Fishel, R.; Lindor, N.M.; Burgart, L.J.; Hamelin, R.; et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J. Natl. Cancer Inst. 2004, 96, 261–268. [Google Scholar] [CrossRef]
  143. Cunningham, J.M.; Christensen, E.R.; Tester, D.J.; Kim, C.Y.; Roche, P.C.; Burgart, L.J.; Thibodeau, S.N. Hypermethylation of the hMLH1 promoter in colon cancer with microsatellite instability. Cancer Res. 1998, 58, 3455–3460. [Google Scholar]
  144. Hitchins, M.P.; Owens, S.E.; Kwok, C.T.; Godsmark, G.; Algar, U.F.; Ramesar, R.S. Identification of new cases of early-onset colorectal cancer with an MLH1 epimutation in an ethnically diverse South African cohort. Clin. Genet. 2011, 80, 428–434. [Google Scholar] [CrossRef]
  145. Wong, J.J.; Hawkins, N.J.; Ward, R.L.; Hitchins, M.P. Methylation of the 3p22 region encompassing MLH1 is representative of the CpG island methylator phenotype in colorectal cancer. Mod. Pathol. 2011, 24, 396–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Hitchins, M.; Williams, R.; Cheong, K.; Halani, N.; Lin, V.A.; Packham, D.; Ku, S.; Buckle, A.; Hawkins, N.; Burn, J.; et al. MLH1 germline epimutations as a factor in hereditary nonpolyposis colorectal cancer. Gastroenterology 2005, 129, 1392–1399. [Google Scholar] [CrossRef] [PubMed]
  147. Chan, T.L.; Yuen, S.T.; Kong, C.K.; Chan, Y.W.; Chan, A.S.; Ng, W.F.; Tsui, W.Y.; Lo, M.W.; Tam, W.Y.; Li, V.S.; et al. Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer. Nat. Genet. 2006, 38, 1178–1183. [Google Scholar] [CrossRef]
  148. Chan, T.L.; Chan, Y.W.; Ho, J.W.; Chan, C.; Chan, A.S.; Chan, E.; Lam, P.W.; Tse, C.W.; Lee, K.C.; Lau, C.W.; et al. MSH2 c.1452-1455delAATG is a founder mutation and an important cause of hereditary nonpolyposis colorectal cancer in the southern Chinese population. Am. J. Hum. Genet. 2004, 74, 1035–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Niessen, R.C.; Hofstra, R.M.; Westers, H.; Ligtenberg, M.J.; Kooi, K.; Jager, P.O.; de Groote, M.L.; Dijkhuizen, T.; Olderode-Berends, M.J.; Hollema, H.; et al. Germline hypermethylation of MLH1 and EPCAM deletions are a frequent cause of Lynch syndrome. Genes. Chromosomes Cancer 2009, 48, 737–744. [Google Scholar] [CrossRef]
  150. Lynch, H.T.; Kaurah, P.; Wirtzfeld, D.; Rubinstein, W.S.; Weissman, S.; Lynch, J.F.; Grady, W.; Wiyrick, S.; Senz, J.; Huntsman, D.G. Hereditary diffuse gastric cancer: Diagnosis, genetic counseling, and prophylactic total gastrectomy. Cancer 2008, 112, 2655–2663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Pharoah, P.D.; Guilford, P.; Caldas, C.; International Gastric Cancer Linkage, C. Incidence of gastric cancer and breast cancer in CDH1 (E-cadherin) mutation carriers from hereditary diffuse gastric cancer families. Gastroenterology 2001, 121, 1348–1353. [Google Scholar] [CrossRef]
  152. Pegg, A.E. Mammalian O6-alkylguanine-DNA alkyltransferase: Regulation and importance in response to alkylating carcinogenic and therapeutic agents. Cancer Res. 1990, 50, 6119–6129. [Google Scholar]
  153. Esteller, M.; Garcia-Foncillas, J.; Andion, E.; Goodman, S.N.; Hidalgo, O.F.; Vanaclocha, V.; Baylin, S.B.; Herman, J.G. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N. Engl. J. Med. 2000, 343, 1350–1354. [Google Scholar] [CrossRef] [PubMed]
  154. Esteller, M.; Toyota, M.; Sanchez-Cespedes, M.; Capella, G.; Peinado, M.A.; Watkins, D.N.; Issa, J.P.; Sidransky, D.; Baylin, S.B.; Herman, J.G. Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is associated with G to A mutations in K-ras in colorectal tumorigenesis. Cancer Res. 2000, 60, 2368–2371. [Google Scholar] [PubMed]
  155. Christians, A.; Hartmann, C.; Benner, A.; Meyer, J.; von Deimling, A.; Weller, M.; Wick, W.; Weiler, M. Prognostic value of three different methods of MGMT promoter methylation analysis in a prospective trial on newly diagnosed glioblastoma. PLoS ONE 2012, 7, e33449. [Google Scholar] [CrossRef]
  156. Costello, J.F.; Futscher, B.W.; Kroes, R.A.; Pieper, R.O. Methylation-related chromatin structure is associated with exclusion of transcription factors from and suppressed expression of the O-6-methylguanine DNA methyltransferase gene in human glioma cell lines. Mol. Cell Biol. 1994, 14, 6515–6521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Costello, J.F.; Futscher, B.W.; Tano, K.; Graunke, D.M.; Pieper, R.O. Graded methylation in the promoter and body of the O6-methylguanine DNA methyltransferase (MGMT) gene correlates with MGMT expression in human glioma cells. J. Biol. Chem. 1994, 269, 17228–17237. [Google Scholar] [CrossRef]
  158. Qian, X.C.; Brent, T.P. Methylation hot spots in the 5’ flanking region denote silencing of the O6-methylguanine-DNA methyltransferase gene. Cancer Res. 1997, 57, 3672–3677. [Google Scholar] [PubMed]
  159. Watkins, D.; Rosenblatt, D.S. Inborn errors of cobalamin absorption and metabolism. Am. J. Med. Genet. C Semin. Med. Genet. 2011, 157C, 33–44. [Google Scholar] [CrossRef]
  160. Jönsson, G.; Staaf, J.; Vallon-Christersson, J.; Ringnér, M.; Gruvberger-Saal, S.K.; Saal, L.H.; Holm, K.; Hegardt, C.; Arason, A.; Fagerholm, R.; et al. The retinoblastoma gene undergoes rearrangements in BRCA1-deficient basal-like breast cancer. Cancer Res. 2012, 72, 4028–4036. [Google Scholar] [CrossRef] [Green Version]
  161. Baldwin, R.L.; Nemeth, E.; Tran, H.; Shvartsman, H.; Cass, I.; Narod, S.; Karlan, B.Y. BRCA1 promoter region hypermethylation in ovarian carcinoma: A population-based study. Cancer Res. 2000, 60, 5329–5333. [Google Scholar]
  162. Laner, A.; Benet-Pages, A.; Neitzel, B.; Holinski-Feder, E. Analysis of 3297 individuals suggests that the pathogenic germline 5’-UTR variant BRCA1 c.-107A > T is not common in south-east Germany. Fam. Cancer 2020, 19, 211–213. [Google Scholar] [CrossRef]
  163. Garg, P.; Jadhav, B.; Rodriguez, O.L.; Patel, N.; Martin-Trujillo, A.; Jain, M.; Metsu, S.; Olsen, H.; Paten, B.; Ritz, B.; et al. A Survey of Rare Epigenetic Variation in 23,116 Human Genomes Identifies Disease-Relevant Epivariations and CGG Expansions. Am. J. Hum. Genet. 2020, 107, 654–669. [Google Scholar] [CrossRef]
  164. Heyn, H.; Moran, S.; Hernando-Herraez, I.; Sayols, S.; Gomez, A.; Sandoval, J.; Monk, D.; Hata, K.; Marques-Bonet, T.; Wang, L.; et al. DNA methylation contributes to natural human variation. Genome Res. 2013, 23, 1363–1372. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Contribution of genetic and epigenetic factors (epimutations) to cancer development. Cancer is caused by genetic factors such as genetic variations, somatic, and germline mutations and by epigenetic factors such as non-coding RNA dysregulation, post-translational alterations in histone marks, and aberrant DNA methylation (epimutations). DNA hypermethylation is the most frequent epimutation and can be generated either from the germline or somatic cells. In germline epimutations (before fertilization), all cells in the body are affected, while in somatic mutations (after fertilization) epigenetic mosaicism can be generated.
Figure 1. Contribution of genetic and epigenetic factors (epimutations) to cancer development. Cancer is caused by genetic factors such as genetic variations, somatic, and germline mutations and by epigenetic factors such as non-coding RNA dysregulation, post-translational alterations in histone marks, and aberrant DNA methylation (epimutations). DNA hypermethylation is the most frequent epimutation and can be generated either from the germline or somatic cells. In germline epimutations (before fertilization), all cells in the body are affected, while in somatic mutations (after fertilization) epigenetic mosaicism can be generated.
Cancers 13 04807 g001
Figure 2. Early alterations during human carcinogenic development. Three important events for early carcinogenic development are described: (A) somatic mutations, (B) germline mutations, and (C) epimutations; focused on the different stages from the parental gametes, embryonic, in the human body, and within the latter detected as the presence or absence of the alteration in blood, specific tissue, and descendant gametes.
Figure 2. Early alterations during human carcinogenic development. Three important events for early carcinogenic development are described: (A) somatic mutations, (B) germline mutations, and (C) epimutations; focused on the different stages from the parental gametes, embryonic, in the human body, and within the latter detected as the presence or absence of the alteration in blood, specific tissue, and descendant gametes.
Cancers 13 04807 g002
Figure 3. Knudson’s “two-hit hypothesis” on epimutations and cancer. (A) The first hit is promoted by epimutations in the promoter region in germ cells. This prevents the binding of TFs to DNA (top). Another mechanism dependent on MBD can be recruited into methylated DNA and compete directly with TFs through their greater affinity for the 5-mC site to the DNA sequence (middle part), or that act synergistically with TFRs to transcriptionally repress a gene. In all cases, this first hit induces the inactivation of the mono-allelic expression of a gene. (B) The second hit is driven by somatic cell mutations such as deletions (LOH), point mutations, other allelic epimutations, or by haploinsufficiency mechanisms that trigger the loss-of-function of the second allele (biallelic inactivation) and induce tumorigenesis. P: Paternal, M: Maternal, TF: Transcription Factor, TFR: Transcription Factor Repressor, MBD: Methyl-CpG-binding domain (MBD), 5-mC: 5-Methylcytosine, LOH: Loss of Heterozygosity.
Figure 3. Knudson’s “two-hit hypothesis” on epimutations and cancer. (A) The first hit is promoted by epimutations in the promoter region in germ cells. This prevents the binding of TFs to DNA (top). Another mechanism dependent on MBD can be recruited into methylated DNA and compete directly with TFs through their greater affinity for the 5-mC site to the DNA sequence (middle part), or that act synergistically with TFRs to transcriptionally repress a gene. In all cases, this first hit induces the inactivation of the mono-allelic expression of a gene. (B) The second hit is driven by somatic cell mutations such as deletions (LOH), point mutations, other allelic epimutations, or by haploinsufficiency mechanisms that trigger the loss-of-function of the second allele (biallelic inactivation) and induce tumorigenesis. P: Paternal, M: Maternal, TF: Transcription Factor, TFR: Transcription Factor Repressor, MBD: Methyl-CpG-binding domain (MBD), 5-mC: 5-Methylcytosine, LOH: Loss of Heterozygosity.
Cancers 13 04807 g003
Figure 4. Constitutional epimutations in human diseases. (A) Constitutional epimutations that cause disease in humans include mechanisms that can occur without altering the DNA sequence (primary epimutations) or as a consequence of a DNA mutation in a cis- or trans-acting factor (secondary epimutations). (I) Primary epimutations result as a consequence of the action of different factors that directly modify the methylation pattern in the promoter region of a tumor suppressor gene. (II) Secondary epimutations that act in cis (2a) may be a consequence of alterations that affect the conserved parental region IC and generate hypermethylation of the promoter region. (2b) Secondary trans-acting factor epimutations occur as a result of mutations in trans elements such as CRC, DNMT’s, MBD, and PcG/TrxG complexes, which indirectly lead to an aberrant methylation state in the promoter region of the gene. In all cases, this leads to transcriptional repression of the tumor suppressor gene. (B) The transmission of the pathogenic allele occurs dominantly (in this example the paternal allele) starting from an alteration in cis (secondary epimutation in cis) in somatic cells. However, there is an erasure of the epimutation (epigenetic erasure) in the gametes, but the alteration in cis is held (in this example an SNV), which can be transmitted to the offspring while the epimutation is restored in somatic cells, being able to generate epigenetic mosaicism. IC: Imprinting Center, DNMT: DNA methyltransferase, MBD: Methyl-CpG-binding domain (MBD), 5-mC: 5 Methylcytosine, CRC: Chromatin-Remodeling Complex, PcG: Polycomb group, TrxG: Trithorax group (TrxG).
Figure 4. Constitutional epimutations in human diseases. (A) Constitutional epimutations that cause disease in humans include mechanisms that can occur without altering the DNA sequence (primary epimutations) or as a consequence of a DNA mutation in a cis- or trans-acting factor (secondary epimutations). (I) Primary epimutations result as a consequence of the action of different factors that directly modify the methylation pattern in the promoter region of a tumor suppressor gene. (II) Secondary epimutations that act in cis (2a) may be a consequence of alterations that affect the conserved parental region IC and generate hypermethylation of the promoter region. (2b) Secondary trans-acting factor epimutations occur as a result of mutations in trans elements such as CRC, DNMT’s, MBD, and PcG/TrxG complexes, which indirectly lead to an aberrant methylation state in the promoter region of the gene. In all cases, this leads to transcriptional repression of the tumor suppressor gene. (B) The transmission of the pathogenic allele occurs dominantly (in this example the paternal allele) starting from an alteration in cis (secondary epimutation in cis) in somatic cells. However, there is an erasure of the epimutation (epigenetic erasure) in the gametes, but the alteration in cis is held (in this example an SNV), which can be transmitted to the offspring while the epimutation is restored in somatic cells, being able to generate epigenetic mosaicism. IC: Imprinting Center, DNMT: DNA methyltransferase, MBD: Methyl-CpG-binding domain (MBD), 5-mC: 5 Methylcytosine, CRC: Chromatin-Remodeling Complex, PcG: Polycomb group, TrxG: Trithorax group (TrxG).
Cancers 13 04807 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ruiz de la Cruz, M.; de la Cruz Montoya, A.H.; Rojas Jiménez, E.A.; Martínez Gregorio, H.; Díaz Velásquez, C.E.; Paredes de la Vega, J.; de la Cruz Hernández-Hernández, F.; Vaca Paniagua, F. Cis-Acting Factors Causing Secondary Epimutations: Impact on the Risk for Cancer and Other Diseases. Cancers 2021, 13, 4807. https://doi.org/10.3390/cancers13194807

AMA Style

Ruiz de la Cruz M, de la Cruz Montoya AH, Rojas Jiménez EA, Martínez Gregorio H, Díaz Velásquez CE, Paredes de la Vega J, de la Cruz Hernández-Hernández F, Vaca Paniagua F. Cis-Acting Factors Causing Secondary Epimutations: Impact on the Risk for Cancer and Other Diseases. Cancers. 2021; 13(19):4807. https://doi.org/10.3390/cancers13194807

Chicago/Turabian Style

Ruiz de la Cruz, Miguel, Aldo Hugo de la Cruz Montoya, Ernesto Arturo Rojas Jiménez, Héctor Martínez Gregorio, Clara Estela Díaz Velásquez, Jimena Paredes de la Vega, Fidel de la Cruz Hernández-Hernández, and Felipe Vaca Paniagua. 2021. "Cis-Acting Factors Causing Secondary Epimutations: Impact on the Risk for Cancer and Other Diseases" Cancers 13, no. 19: 4807. https://doi.org/10.3390/cancers13194807

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