Abstract
Aberrant epigenetic modifications, particularly DNA methylation, play a critical role in the pathogenesis and progression of human diseases. The current review aims to reveal the role of aberrant DNA methylation in the pathogenesis and progression of diseases and to discuss the original data obtained from international research laboratories on this topic. In the review, we mainly summarize the studies exploring the role of aberrant DNA methylation as diagnostic and prognostic biomarkers in a broad range of human diseases, including monogenic epigenetics, autoimmunity, metabolic disorders, hematologic neoplasms, and solid tumors. The last section provides a general overview of the possibility of the DNA methylation machinery from the perspective of pharmaceutic approaches. In conclusion, the study of DNA methylation machinery is a phenomenal intersection that each of its ways can reveal the mysteries of various diseases, introduce new diagnostic and prognostic biomarkers, and propose a new patient-tailored therapeutic approach for diseases.
Keywords: DNA methylation, Monogenic epigenetic diseases, Autoimmune diseases, Metabolic disorders, Cancer
Graphical abstract
1. Introduction
The last two decades were the milestone in the era of the molecular biology of diseases as the footprints of epigenetic modifications became evident in the progression of a wide range of human diseases. Despite the common belief, the advent of whole-genome sequencing methods has demonstrated that the frequency of genetic alterations is notably low in many diseases. All genetic predispositions do not necessarily lead to disease development because the incidence of some diseases is entirely different in genetically identical monozygotic twins [1,2]. These new observations have raised a question: What can explain this genotype-phenotype discrepancy? Soon after, the answer showed itself in the cover of epigenetic alterations __a group of heritable changes in the genome that adapt themselves to the needs of cells according to environmental variations. In plain words, epigenetics transmits environmental signals to the genome without any alterations in the primary nucleotide sequence of DNA [3,4]. Epigenetic alterations include DNA methylation, histone modifications, and non-coding RNAs that are fundamental in the early stages of development as they can readily regulate the expression of genes, parental imprinting, and X chromosome inactivation. Moreover, these alterations have a solid foot in maintaining the stability and integrity of the genome [5].
Among different modifications, DNA methylation is one of the most popular epigenetic modifications that may recruit to change the genome signature and thus alter gene expression. DNA methylation, or what is now known as the DNA methylation machinery, was first explained by Halliday and Pugh in 1975 [6] as a process in which a group of enzymes adds a methyl group (-CH3) from S-adenosyl methionine (SAM) to the fifth carbon position of cytosine in a CpG dinucleotide [7]. They described DNA methylation as a gene expression regulatory mechanism. However, it was not long before the importance of this modification in vital biological processes such as embryogenesis [8] and aging [9] became clear. What we know now about DNA methylation is more complicated than what has been described previously; recently, it seems that advances in studying DNA methylation alteration not only might help uncover the understanding of many biological events but also may revolutionize the conventional concept of disease pathogenesis. In the present review, we aim to discuss the fundamental functions of DNA methylation in the epigenetic regulation of genes and also update the recent findings of the DNA methylation alterations in the pathogenesis/progression of different human diseases, ranging from autoimmune to metabolic disorders as well as various human malignancies.
2. Methods
To perform the literature review, our initial search consisted of an AND/OR combination of DNA methylation, DNA methyltransferases, Methyl-CpG-binding domain proteins, DNA-demethylating enzymes, diagnosis, prognosis, minimal residual disease, response, pathogenesis, survival, Imprinting disorders (Silver–Russell syndrome; Beckwith–Wiedemann syndrome; Prader–Willi syndrome; Angelman syndrome), Single-gene disorders (Immunodeficiency, centromeric instability, and facial anomalies syndrome; Hereditary sensory autonomic neuropathy 1E with dementia and hearing loss; Autosomal dominant cerebellar ataxia with deafness and narcolepsy, …), Autoimmune diseases (Systemic lupus erythematosus; Rheumatoid arthritis; Systemic sclerosis; Multiple sclerosis), Metabolic disorders (Type 2 diabetic Mellitus; Obesity), Hematological malignancies (Myeloproliferative neoplasms; myelodysplastic syndrome; Acute myeloid leukemia; T cell lymphoma; Adult T acute lymphoblastic leukemia), Solid tumors (Bladder cancer; Breast cancer; Colorectal cancer; Cervix cancer; Cancer of unknown primary; Glioblastomas; Gastric cancer; Hepatocellular carcinoma; Head and neck squamous cell carcinoma; Lung cancer; Non-small-cell lung cancer; Ovarian cancer; Prostate cancer; Pancreatic cancer; Small cell lung cancer), DNA methylation-based drugs (DNMT inhibitors; IDH1/2 inhibitors, …). We further extended our search domain by investigating scientific articles of electronic resources (Google Scholar, PubMed, Science Direct, Wiley, Scopus, and Springer) by the abbreviated form of the names. Ultimately, we investigated the results and selected the most relevant publications for review (Fig. 1).
Fig. 1.
The flowchart of the literature search process.
3. DNA methylation machinery
DNA methylation is a triphasic process in which DNA methylation groups are established (de novo DNA methylation), preserved, and removed. The equilibrium between methyltransferases, which are known as “writers”, and DNA demethylases (erasers) keeps the integrity of the process. DNA methylation machinery also needs a group of proteins known as “readers” to translate annotation into functional information [10].
3.1. DNA methyltransferases; writers
The family of DNA methyltransferases (DNMTs) is responsible for the methylation of the fifth carbon of cytosines (5 mC) in CpG dinucleotides [7]. Apart from their role in the establishment phase, DNMTs also have a hand in the maintenance of methyl groups in the DNA methylation process. There are five DNMTs in mammals, DNMT1, DNMT2, DNMT3A, DNMT3B, and DNMT3L. DNMT3A and DNMT3B, which mostly take part in de novo methylation, contain two chromatin reading domains, a Pro-Trip-Trip-Pro (PWWP) domain, and ATRX- DNMT3- DNMT3L (ADD) and a highly conserved DNMT (MTase) domain in the carboxy terminus [11]. Whereas, DNMT1 and UHRF1 hold a respectable share in copying the methylation group of template strands in daughter strands after replication, a process that is observed in the maintenance phase. DNMT1, a maintenance DNMT, contains an MTase domain in the carboxy terminus and a large N-terminal regulatory region consisting of a replication foci targeting sequence (RFTS) domain, a CXXC zinc finger domain, and a pair of bromo adjacent homology (BAH) domains [12]. Either participating in the establishment or maintenance phase, DNMTs repress gene expression through direct or indirect mechanisms; prevent the binding of transcription factors (TFs) to the promotor of genes, or recruit Methyl-CpG-binding domain (MBD) proteins, which can either inhibit the binding of TFs or recruit transcriptional repression complex [10]. While the function of DNMT1, DNMT3A, and DNMT3B is canonically related to transferring methyl groups to CpG sites, DNMT2 and DNMT3L seem to have non-canonical functions. DNMT3L forms a heterotetramer with DNMT3A to increase its catalytic activity [13]. DNMT2, on the other hand, participates in methylating the cytosine of RNAs, especially transfer RNAs (tRNAs) [14].
3.2. Methyl-CpG-binding domain proteins (MBDs); readers
To repress the expression of genes, methylated CpG sites, eventually in the form of methylated CpG islands, should be translated into the function [15]. For this purpose, the methylation status of the CpGs should first be read by a group of proteins known as readers which are the MBD family (including MECP2 and MBD1-4) [15], the Kaiso and Kaiso-like proteins [16], as well as the SET and ring finger-associated (SRA) domain family [17]. Among them, MECP2, MBD1, and MBD2 seem to have a fundamental role in gene silencing. These proteins bind to methyl groups in the CpG island via their MBD domains and recruit co-repressor complexes such as histone-modifying enzymes or chromatin remodeling complexes via their transcriptional repression domains (TRD) to prevent transcription of a specific gene [15]. Other MBD proteins such as MBD3, MBD4, MBD5/6, SETDB1/2, and BAZ2A/B have also been identified as important elements in the methylation machinery, although they might have different structures [18].
In addition to MBDs, other reader proteins such as ZBTB4, ZBTB38, and ZBTB33/Kaiso recognize methylated groups in the CpG dinucleotides and interact through their zinc-finger (ZnF) domains. Unlike ZBTB4 and ZBTB38 which only need one methylated CpG as a landing site, the binding of Kaiso would not occur unless at least two methyl groups are presented on the CpG dinucleotides [16]. ZFP57 is another ZnF protein that protects the DNA methylation imprints during early embryogenesis against DNA demethylation enzymes via binding to methylated hexanucleotide (TGCCGC) in imprinting control regions (ICRs) [19]. Another reader in the DNA methylation machinery, UHRF1, is a multi-domain protein consisting of an N-terminal ubiquitin-like (UBL) domain, a tandem Tudor domain (TTD), a plant homeodomain (PHD), a SET- and RING-associated (SRA) domain, and a C-terminal RING finger domain. This protein is more involved in the maintenance of DNA methylation than silencing gene expression and is an essential agent for recruiting DNMT1 to replication foci [20].
3.3. DNA-demethylating enzymes; erasers
It was in acute myeloid leukemia (AML) molecular investigations that a group of novel proteins named ten-eleven translocation (TET) was identified as a result of chromosomal rearrangement t(10; 11)(q22; q23). It firstly claimed that this newly identified gene was responsible for encoding the histone H3 lysine 4 (H3K4) methyltransferases [21]; however, very soon, it became evident that the TET family could also carry out active DNA demethylation [22]. TET proteins showed the ability to bind to unmethylated CpGs and maintain them in hypomethylated status [23]. Moreover, they can use alpha-ketoglutarate (α-KG) and oxygen as substrate and Fe (II) as a cofactor to oxidize 5mCs to create 5-hydroxymethylcytosine (5hmC), which consequently converts into formyl-(5-fc) and carboxyl-(5caC) derivatives. After the formation of such derivatives within the DNA, DNA repair proteins such as thymine glycosylase (TDG) come into action to exit them by engaging the base excision repair (BER) pathway [24]. Thus far, three members of the TET family have been identified. TET1 erases imprinting marks in primordial germ cells [25], TET2 regulates hematopoiesis [26], and TET3 is a paramount mediator of preimplantation development via removing methyl groups in the zygote [27]. To provide a well-conceptualized perspective of the DNA methylation machinery, we summarized the mechanisms of DNA methylation and demethylation in Fig. 2.
Fig. 2.
A schematic representation of mechanisms of DNA methylation and demethylation. DNMT3A and DNMT3B establish de novo DNA methylation, whereas DNMT1 and UHRF1 add a methylation group on daughter strands after replication during the maintenance phase. DNMTs repress gene expression by inhibiting transcription factors (TFs) binding to the gene promoters or recruiting methyl-CpG-binding domain (MBD) proteins. In active demethylation, TETs oxidize 5mCs to 5-hydroxymethylcytosine (5hmC), which consequently converts into formyl-(5-fc) and carboxyl-(5caC) derivatives. Then thymine glycosylase (TDG) removes 5-fc (not shown) and 5caC during the base excision repair (BER) pathway. In the tricarboxylic acid (TCA) cycle, isocitrate dehydrogenases (IDHs) convert isocitrate to α-KG, an essential cofactor for dioxygenases, such as TET proteins. FH: fumarate hydrogenase; SAM: s-adenosyl methionine; SDH: succinate dehydrogenase.
4. Dysregulation in the DNA methylation machinery; a missing piece in the puzzle of human diseases
The footprint of DNA methylation dysregulation in the pathogenesis of a wide range of human diseases emphasizes that methylation machinery should be positioned at the center of attention in many diseases. The results of new sequencing methods have revealed the high incidence of mutation in genes encoding DNMTs and TET in several human diseases. For example, mutations in genes encoding DNMT1, DNMT3A, and DNMT3B, have been identified in different diseases, such as ICF syndrome. In addition, MECP2 mutations have been well-detected in many neurological disorders, such as Rett syndrome. The following part of this review aims to update recent advances about the role of DNA methylation abnormalities in different human diseases.
4.1. DNA methylation in monogenic epigenetic diseases
4.1.1. DNA methylation in imprinting disorders
Imprinting disorders were the first human diseases in which the footprint of DNA methylation was identified. In the human genome, more than a hundred imprinted genes have been recognized, and their expressions can be mediated in mono-allelic manners. This happens due to the induction of de novo methylation in one germ cell that inhibits the expression of one copy of the gene in the offspring [28]. Due to the importance of imprinting genes in human development, it is not surprising that any dysregulation in the expression of these genes leads to life-threatening congenital disorders [29]. Imprinting disorders can result from four specific causes: epimutations, uniparental disomy (UPD), deletions, and mutations in the imprinting control regions (ICRs). These molecular alterations result in the expression of the inactive allele or aberrant silencing of the active allele [30].
Epimutation can be caused by alternations of DNA methylation in the ICRs or disruptions of histone modifications among the imprinted loci. The best evidence of the mechanism through which imprinting abnormalities cause a disorder can be detected on chromosome 11p15.5, where the insulin-like growth factor 2 (IGF2)/H19 is placed. Of note, the adjusted intracellular levels of IGF2 and H19 are of such importance that the excessive production of each protein is ceased through the imprinting process. Silver–Russell syndrome (SRS) and Beckwith–Wiedemann syndrome (BWS) are two imprinting disorders that arise from defective methylation of ICR1 in the H19/IGF2 cluster. In SRS, the mistaken hypomethylation of parental ICR1 leads to the downregulation of IGF2 [31]. On the other hand, both paternal and maternal ICR1 display hypermethylation form in BWS, an event that leads to unnecessary up-regulation of IGF2 in neonates [32]. Within H19/IGF2 cluster, there is another imprinted gene named CDKN1C/KCNQ1 cluster. The regulation of this cluster is in the hands of ICR2 (KvDMR1). In half of BWS cases, the maternal ICR2 appears in the hypomethylated form, which in synergism with hypomethylated paternal ICR2 profoundly reduces the intracellular amount of CDKN1C in patients [33].
Induction of paternal and maternal deletion in 15q11.2-q13 causes two other imprinting disorders named Prader–Willi syndrome (PWS) and Angelman syndrome (AS), respectively [34,35]. The methylation of PWS and AS ICRs controls the expression of small nuclear ribonucleoprotein polypeptide N (SNRPN) and SNURF in the SNURF/SNRPN cluster within chromosome 15q11.2–q13, which in turn, determines the expression of ubiquitin-protein ligase E3A (UBE3A) [36] __a protein that is responsible for the function of neurons and the typical development of synapses. In normal conditions, only AS/PWS ICRs of the maternal allele are methylated, which allows the expression of UBE3A in the brain. AS is caused by the loss of maternal UBE3A expression in neuronal cells [37], whereas PWS is caused by the loss of paternal expression of SNRPN, snoRNAs, and other genes [38].
Apart from inherited abnormalities in the imprinting process which eventually lead to the development of several imprinting disorders, evidence suggested that some assisted reproductive technologies (ARTs) that are widely used for the solving of infertility problems in recent decades can cause dysregulation in the imprinting process. The incidence of BWS, AS, and SRS in ART-born children has shed light on the detrimental impact of these technologies on CpG methylation in the imprinting genes [39]. Nevertheless, it should not be forgotten that these techniques are entirely new, and still, many questions remain unanswered in this field due to the differences in study design and the small size of cohorts.
4.1.2. Single-gene disorders of the DNA methylation machinery
Immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome is an atypical immunoglobulin deficiency disorder with life-threatening infections in which DNA methylation plays a crucial role in its pathogenesis [40]. According to genetic mutations, ICF is discussed in four groups ICF1, ICF2, ICF3, and ICF4 [[41], [42], [43]]. The molecular hallmarks of ICF typically include instability of the juxtacentromeric heterochromatin regions of chromosomes 1, 9, and 16 enriched with specific hypomethylation of pericentromeric satellite repeats II and III [44]. A wealth of evidence declared that DNMT3B mutations, heavily methylated α-satellites [45], and the hypomethylation of subtelomeric repeats [46] that is detectable in 50 % of patients with ICF can be used to distinguish ICF1 from other types of ICF syndrome. Notably, patients with ICF1 uniquely demonstrate promoter hypomethylation of germline genes [47] and loss of methylation at X-linked genes [48], in agreement with a function for DNMT3B in establishing DNA methylation in early development. The aberrancies in other components of the DNA methylation machinery include genes encoding ZBTB24, CDCA7, and HELLS detected in ICF2, ICF3, and ICF4, respectively. Moreover, it has been indicated that these types of ICFs __in contrast to ICF1__ display hypomethylated α-satellite [49] without any aberrations of DNA methylation in subtelomeric repeats [46]. This data suggests that ZBTB14, CDCA7, and HELLS may have a role in recruiting a DNMT other than DNMT3B to these sequences, although their associations with DNA methylation are not fully understood.
Recent studies have reported that the absence of ZBTB24, CDCA7, or HELLS leads to a decrease in DNA methylation at satellite repeats in the human embryonic kidney (HEK) 293T cells [50] and mouse embryonic fibroblasts (MEFs) [43]. In the mouse embryonic stem cells (mESCs)-based ZBTB24 or CDCA7 knockout model, fibroblasts and T cells derived from ICF2 patients, it has been shown that ZBTB24, a C2H2 zinc finger (ZF) transcription factor, directly binds to a sequence in the CDCA7 promotor and regulates its expression [51,52]. Moreover, the studies indicated that CDCA7 is an essential agent for the recruitment of HELLS to chromatin and can stimulate HELLS nucleosome remodeling activity, facilitating the access of the DNA methylation machinery to genomic regions [53]. Aberrant DNA methylation patterns in ICF3 patients could be due to a defect in recruiting HELLS to chromatin [53]. Therefore, it seems that ZBTB24, CDCA7, and HELLS can serve as guides for the DNA methylation machinery.
However, it remains elusive how reduced DNA methylation can cause the phenotypes of ICF patients. Perhaps the simplest explanation is that DNA methylation alterations associated with gene expression changes play a role in the pathogenesis of ICF syndrome. For example, aberrant DNA methylation associated with dysregulation of the clustered protocadherins (PCDH) may be related to the neural defects observed in all patients with ICF [49]. A recent study has reported that CDCA7 and HELLS have a role in the classical non-homologous end joining (c-NHEJ) pathway and the deficiency of CDCA7 or HELLS in HEK293T cells triggers the C-NHEJ defect and delay in Ku80 accumulation at DNA damage sites. Defects in the C-NHEJ pathway are associated with the phenotypes of ICF patients, including abnormal chromosome configuration, instability of satellite repeats, reduced replication rates, and apoptosis [50]. Since the C-NHEJ pathway is required for both V(D)J- and class switch-recombination (CSR) [54,55], perhaps impaired IgE class switch induction in a non-ICF1 patient could be explained by a defect in the C-NHEJ pathway [56]. Finally, how aberrant DNA methylation can lead to immunodeficiency in ICF patients is unclear. Due to embryonic lethality and/or lack of immunoglobulin deficiency, mouse modeling for ICF patients is poorly informed [51,57]. There is only a limited study of data on the pericentromeric SAT1 hypomethylation that activates the innate immune system by derepressing SAT1-associated transcripts during embryonic development in the ZBTB24-mutant zebrafish. Such animal models may help identify the mechanisms involved in the pathogenesis of ICF syndrome in humans [58].
Apart from ICF, defects in the DNA methylation machinery have been detected in other autosomal-dominant progressive cognitive and behavioral deterioration. Hereditary sensory autonomic neuropathy 1E with dementia and hearing loss (HSNA1E; OMIM 614116) [59], and autosomal dominant cerebellar ataxia with deafness and narcolepsy (ADCA-DN; OMIM 604121) [60] all share a similarity in the presence of heterozygous mutations in DNMT1. Interestingly, the mutations of HSNA1E mostly affected the RFTS domain of DNMT1 leading to cytoplasmic aggregation of this enzyme. In response to cellular stress and aging, it is believed that these aggregates induce toxicity, an event that leads to the degeneration of the central and peripheral nervous systems [61]. DNMT1 mutation in HSNA1E and ADAC-DN, however, occur in the N-terminal and C-terminal of the RFTS domain, respectively [61]. Tatton-Brown–Rahman syndrome is another genetic disorder in which heterozygous haploinsufficiency mutations in DNMT3A have been reported [62]. Individuals with microcephalic dwarfism also showed to have either missense or gain-of-function mutations in the PWWP domain of DNMT3A [63]. The investigation of DNA methylation alterations in single-gene disorders has a long way to go; however, the identification of such mutations suggests the auxiliary impact of the DNA methylation machinery in the pathogenesis of these diseases. Table 1 summarizes DNA methylation aberrancy in monogenic epigenetic diseases.
Table 1.
DNA methylation in monogenic epigenetic diseases.
| Diseases [OMIM] | Locus | Molecular defect (frequency) | Major features | Ref |
|---|---|---|---|---|
| Imprinting disordersa | ||||
| SRS [180860] | 11p15 | H19/IGF2: paternal ICR1 LoM (50 %) | Intrauterine and postnatal growth restriction, dwarfism, triangular face, congenital hemihypertrophy (asymmetric body), low birth weight, and feeding difficulties | [31,64] |
| 7 | Maternal UPD7 (10 %) | |||
| 12q14 | Maternal UPD (11p15) (rare) | |||
| 8q12 | 11p15 CNVs (<1 %) | |||
| CDKN1C, IGF2, HMGA2, PLAG1 point mutations (rare) | ||||
| BWS [130650] | 11p15 | CDKN1C/KCNQ1: maternal ICR2 LoM (50–60 %) | Lateralized overgrowth, macroglossia, placental overgrowth, exomphalos, neonatal hypoglycemia, hyperinsulinism, adrenal cortex cytomegaly, pancreatic adenomatosis, and a predisposition to embryonal tumors (primarily Wilms tumor, nephroblastomatosis) | [32,33,65] |
| Paternal UPD (11p15) (20–25 %) | ||||
| H19/IGF2: maternal ICR1 GoM (5–10 %) | ||||
| 11p15 CNVs (2–4 %) | ||||
| CDKN1C point mutations on maternal allele (5 % sporadic; 50 % in families) | ||||
| PWS [176270] | 15q11.2–q13 | 15q11–q13 Paternal deletion (70–75 %) | Developmental delay, Hypotonia, short stature, delayed cognitive development, behavioral complications, pediatric obesity, and hyperphagia | [34,38] |
| Maternal UPD15 (25–30 %) | ||||
| SNURF/SNRPN: paternal PWS/ICR GoM (∼1 %) | ||||
| AS [105830] | 15q1.21–q13 | 15q11–q13 maternal deletion (75 %) | Developmental delay, microcephaly, severe intellectual disability, absent or limited speech, ataxia, seizures, scoliosis, unmotivated laughing, hyperactivity, happy facial appearance, and autistic traits | [35,37] |
| UBE3A point mutations on maternal allele (5–10 %) | ||||
| Paternal UPD15 (1–2 %) | ||||
| SNURF/SNRPN: maternal PWS/ICR LoM (3 %) | ||||
| Single-gene disorders | ||||
| ICF [602900] | 20q11.21 | AR mutations of the DNMT3B in ICF1 (50 %); ZBTB24 in ICF2; CDCA7 in ICF3; HELLS in ICF4 (50 %) | Immunodeficiency, centromeric instability, failure to thrive, mental retardation, and facial anomalies (hypertelorism, epicanthic folds, a flat nasal bridge, and low-set ears) | [[41], [42], [43]] |
| HSAN1E [614116] | 19p13.2 | DNMT1: AD mutations in the amino-terminal and middle part of the RFTS domain | Hearing and sensory loss, reduction in cognitive function, ataxia, brain atrophy, and dementia | [59] |
| ADAC-DN [604121] | 19p13.2 | DNMT1: AD mutations in the carboxy terminus of the RFTS domain | Cerebellar ataxia, deafness, narcolepsy, progressive cognitive and behavioral deterioration | [60] |
| TBRS [602729] | 2p23.3 | DNMT3A: Heterozygous DNMT3A haploinsufficiency mutations | Macrocephalic overgrowth, distinctive facial appearance, and moderate intellectual disability, and leading to AML in approximately 25 % of patients | [62] |
| MD | 2p23.3 | DNMT3A: GoF mutations in the PWWP domain | Intrauterine and postnatal growth restriction, a profound yet proportionate reduction in body size and head size | [63] |
| c-RTT [312750] | Xq28 | MECP2: LoF mutation (over 90 % of the case) | A severe neurological disorder with intellectual, language, motor impairments, and autistic traits | [66] |
| MDS | Xq28 | MECP2: GoF mutation (MECP2 duplication) | Infantile hypotonia, poor speech development, severe to profound mental retardation, progressive spasticity, recurrent infections, epilepsy, autistic features | [67] |
| TET3 deficiency | 2p13.1 | TET3: Reduction in TET3 catalytic activity | Intellectual disability and/or global developmental delay, hypotonia, autistic traits, movement disorders, growth abnormalities, and facial dysmorphism | [68] |
| Other monogenic epigenetic diseases | ||||
| FXS [300624] | Xq27.3 | The aberrant repeat of CGG trinucleotide in the 5´ UTR of the FMR1 gene and silencing of the FMR1 gene (<1 % point mutation or deletion of FMR1 gene) | Intellectual disability, autistic phenotypes, elongated face, protruding ears, larger testes, stereotypic movements | [66] |
Imprinting disorders can result from specific four causes; epimutations, uniparental disomy (UPD), deletions, and mutations in the imprinting control regions (ICRs). AS: angelman syndrome; ADAC-DN: autosomal-dominant cerebellar ataxia, deafness and narcolepsy; AR: autosomal recessive; AD: autosomal-dominant; AML: acute myeloid leukemia; BWS: beckwith–wiedemann syndrome; CNVs: copy number variation; CDKN1C: cyclin dependent kinase inhibitor 1C; CDCA7: cell division cycle associated 7; MD: microcephalic dwarfism; DNMT: DNA (cytosine-5)-methyltransferase; FXS: fragile X syndrome; FMR1: fragile X mental retardation 1; GoM: gain of methylation; GoF: gain of-function; HSAN1E: hereditary sensory autonomic neuropathy 1E; HMGA2: high mobility group A2; HELLS: helicase lymphoid specific; ICF: immunodeficiency, centromeric instability and facial anomalies syndrome; IGF2: insulin-like growth factor 2; ICR: imprinting control region; KCNQ1: potassium voltage-gated channel subfamily Q member 1; LoM: loss of methylation; LoF: loss of function; MECP2: methyl CpG binding protein 2; MDS: MECP2 duplication syndrome; PWS: prader–willi syndrome; PLAG1: pleomorphic adenoma gene 1; PWWP: ProTrpTrpPro; c-RTT: classical rett Syndrome; RFTS: replication foci targeting sequence; SRS: silver–russell syndrome; SNURF: SNRPN upstream reading frame; SNRPN: small nuclear ribonucleoprotein polypeptide N; TBRS: tatton-brown–rahman syndrome; TET3: tet methylcytosine dioxygenase 3; UPD: uniparental disomy; UBE3A: ubiquitin-protein ligase E3A; UTR: untranslated region; ZBTB24: zinc finger and BTB domain containing 24.
4.2. DNA methylation in autoimmune diseases
4.2.1. Systemic lupus erythematosus (SLE)
SLE is a chronic autoimmune disease characterized by autoantibody production and immune complex deposition. External triggers (e.g., UV radiation) cause the apoptosis of cells. Inadequate clearance of the nuclei of these cells increases the burden of nuclear antigens and may lead to impaired ability to tolerate autoreactive B and T lymphocytes in susceptible individuals. In response to the self-antigens, activated plasmacytoid dendritic cells (pDCs) release type I interferons (IFNs), which increase antigen presentation by dendritic cells (DCs), thereby activating T cells. B cells are activated by exposure to self-antigens and interaction with T cells, producing autoantibodies. Eventually, the generation of immune complexes triggers increased inflammation and organ damage (Fig. 3).
Fig. 3.
A schematic representation of DNA methylation alterations involved in the pathogenesis of SLE. DNA methylation alterations in SLE immune cells (especially T helper cells) may contribute to more inflammation and autoimmunity by increased expression of genes involved in the interaction of T helper cells with B cells and DCs. BCR: B-cell receptor; CAMP: cathelicidin antimicrobial peptide; DCs: dendritic cells; FCR: Fc receptor; HMGB1: high mobility group box 1 protein; ICAM-1: intercellular adhesion molecule 1; IFNs: type I interferons; ITGB2: integrin subunit beta 2; MHC: major histocompatibility complex; pDCs: plasmacytoid dendritic cells; TCR: T-cell receptor; TLR: toll-like receptor.
At the primary stages of the disease and along with the formation of SLE, decreased expression of DNMT1 together with induction of DNA hypomethylation in CD4+ T cells increases the expression of inflammatory cytokines; an event which in turn leads to unnecessary stimulation of B cells [69]. As the disease progressed, DNA hypomethylation induced polarization of T helper 2 (Th2) cells by elevating the expression of IL-4, IL-5, and IL-13. Noteworthy, the shift of Th1/Th2 equilibrium in favor of Th2 reinforces the vicious cycle of auto-reactive antibody production [70]. These findings have been confirmed from another perspective, as well. The exposure of healthy mice to several DNA methylation inhibitors such as procainamide and hydralazine or injection of azacitidine (5-AC)-treated healthy CD4+ T cells to the mice were all coupled with the development of SLE [71]. These make a consensus on the importance of CD4+ T cells' methylation status in SLE pathogenesis. It has been also declared that the hypomethylation at two CpG sites within IFI44L can distinguish SLE patients from healthy individuals [72]. Besides, increased levels of 5hmC were associated with elevated expression of TET2 and TET3 in CD4+ T cells from patients with SLE [73]. The evidence supporting the higher frequency of demethylation of the CD40L gene, which is located on chromosome X in females, also answers the question of why the incidence of SLE is higher in females [74]. As a straightforward interpretation of these events, DNA methylation modifications not only seem to contribute to SLE formation but also can guarantee disease progression.
4.2.2. Rheumatoid arthritis (RA)
RA is a chronic autoimmune disease characterized by inflammation of the synovium associated with the destruction of joint cartilage and bone erosion. Stressors cause post-translational modifications (especially citrullination) in mucosal sites. The citrullinated proteins trigger the immune response and autoantibody formation in susceptible individuals. Activated immune cells enter joints, respond to the self-proteins, and mainly trigger tissue injury by cytokine secretion and perhaps by antibody-dependent effector mechanisms. The pro-inflammatory environment in RA joints strongly activates fibroblast-like synoviocytes (FLSs) and leads to expanded synovial sublining. FLSs produce inflammatory cytokines, chemokines, and pro-angiogenic factors (such as IL-6, CXCL10, and VEGF) that promote and maintain joint inflammation. These cells also produce matrix metalloproteinases (MMPs) which lead to cartilage destruction. FLSs help bone erosion by facilitating osteoclastogenesis and interfering with bone repair (Fig. 4).
Fig. 4.
A schematic representation of DNA methylation alterations involved in the pathogenesis of RA. DNA methylation alterations contribute to RA pathogenesis by affecting the FLSs and immune cells. DNA methylation in RA FLSs transforms a normal FLS into an apoptotic-resistant and aggressive phenotype that promotes inflammation and destruction of joints. On the other hand, DNA methylation in immune cells (especially regulatory T cells) may remove the immune brake and promote inflammation. . CTLA4: cytotoxic T-Lymphocyte Associated Protein 4; FLSs: fibroblast-like synoviocytes.
RA is one of the most important autoimmune diseases with genetic predispositions in the HLA gene [75]. However, it was in 2013 that the results of a study comparing the blood samples of RA patients and healthy counterparts revealed the presence of two methylation sites within the MHC region; opening a new chapter in the pathogenesis of the disease: this time from the perspective of DNA methylation regulation [76]. It should be noted that both RA and SLE may share similar methylation patterns in genes encoding CD40L [77] and FOXP3 [78,79]. The hypermethylation of CTLA4 in RA patients may also play an essential role in the auto-reactivity of immune responses against host antigens [80]. Noteworthy, the correlation between the amount of DNA methylation and the response rate to methotrexate has also been reported; as the methylation levels are higher in RA patients, they may display lower responses to methotrexate [81].
4.2.3. Systemic sclerosis (SSc)
SSc is a multifaceted disease characterized by initial vascular injury and uncontrolled inflammatory reaction that leads to fibrosis in the skin and internal organs. Following early vascular injury, active endothelial cells produce endothelin-1 and chemokines, increasing the expression of adhesion molecules and ultimately leading to the recruitment of inflammatory cells. Activated platelets, dendritic cells, macrophages, T cells, and B cells secrete profibrotic growth factors and cytokines that in turn lead to the activation and differentiation of resident fibroblasts to myofibroblasts, which secrete the excessive extracellular matrix (ECM).
DNA methylation analysis has demonstrated that the cross-talk between CD4+ T cells and fibroblasts in SSc patients can control the disease; while induction of DNA hypermethylation in fibroblasts is coupled with up-regulation of some DNA methylation regulators [[82], [83], [84], [85], [86]], DNA hypomethylation in CD4+ T cells of SSc patients is responsible for orchestrating the pro-inflammatory responses [[87], [88], [89], [90], [91]]. Also, studying DNA methylation profiles may be an effective method to distinguish two subtypes of SSc: limited cutaneous SSc (lcSSc) and diffuse cutaneous SSc (dcSSc). Thus far, 153 and 266 differentially methylation sites have been identified for lcSSc and dcSSc, respectively [92]. A schematic representation of the role of DNA methylation alterations in the pathogenesis of SSc is demonstrated in Fig. 5.
Fig. 5.
A schematic representation of DNA methylation alterations involved in the pathogenesis of SSc. DNA methylation alterations contribute to SSc pathogenesis by affecting the endothelial cells, immune cells, and fibroblasts, resulting in vascular dysfunction, autoimmunity, inflammation, and fibrosis. ROS: reactive oxygen species.
4.2.4. Multiple sclerosis (MS)
MS is a chronic, inflammatory, autoimmune, demyelinating, and neurodegenerative disease of the CNS. DNA methylation signatures can be used as a marker to predict the risk of MS development. For example, patients with evidence of hypermethylation at the genes of Alu and LINE1 may develop a severe form of the disease [93]. Or, the methylation of vitamin D receptor (VDR) and IL2RA can also increase the risk of MS development [94,95]. The correlation between the incidence of methylation in HLADRB1 in CD4+ T cells and the severity of the disease has also been reported [96]. Moreover, the reduction in 5hmC in peripheral blood mononuclear cells (PBMCs) is associated with the downregulation of TET2 expression [97]. Fig. 6 provides a schematic representation of the role of DNA methylation alterations in the pathogenesis of MS. Table 2 summarizes the results of various studies that highlighted the role of DNA methylation markers in autoimmune diseases.
Fig. 6.
A schematic representation of DNA methylation alterations involved in the pathogenesis of MS. DNA methylation alterations contribute to MS pathogenesis by affecting the immune and neurological compartments. In the immune compartment, DNA methylation may increase leukocyte-mediated inflammation through hypermethylation of the PTPN6 gene (also known as SHP-1), which encodes an anti-inflammatory tyrosine phosphatase. In the various regions of the brain, specific hypomethylated and hypermethylated genes increase antigen presentation and myelin destabilization, while reducing the survival of nerve and oligodendrocyte cells.
Table 2.
Potential DNA methylation markers in autoimmune diseases.
| Diseases | Cell/tissue type | Main findings | Ref |
|---|---|---|---|
| SLE | CD4+ T cells from SLE patients and HCs | Disease activity was correlated with hypomethylation of genes involved in T cell activation and differentiation, and hypermethylation in genes involved in the inhibitory TGF-β signaling pathway. | [70] |
| CD4+ T cells, CD19+ B cells, and CD14+ monocytes from SLE patients and HCs | DMCpGs: 166 in B-cells, 97 in monocytes, and 1033 in T-cells; significant hypomethylation of IFN-regulated genes was detected in monocytes, T cells, and B cells of SLE patients relative to HCs. | [98] | |
| CD4+ T cells, monocytes, granulocytes, and B-cells from discordant MZ and DZ twins for SLE | Significant hypomethylation of IFN-regulated genes was detected in all cell types, which was more pronounced in twins with disease flare within the past two years. | [99] | |
| PBMNs from AA SLE patients and EA SLE patients and HCs | 85 % of 41 DMCpGs were hypomethylated in SLE patients relative to HCs. The number of hypomethylated CpG sites relevant to IFN-related genes was higher in AA SLE patients and SLE patients with SLEDAI>6 compared with EA SLE patients and SLE patients with SLEDAI≤6. | [100] | |
| Naïve CD4+ T cells from HCs and SLE patients with and without a history of renal involvement | 191 DMCpGs (121 DAGs) were only identified in SLE patients with a history of renal involvement. A hypomethylated CpG site in the gene CHST12 had a sensitivity of 85.7 % and a specificity of 71.4 % to detect renal involvement in SLE patients. | [101] | |
| PMNCs from SLE patients and HCs | The methylation levels of IL-10 and IL-1R2 genes significantly reduce in SLE relative to HCs and correlate with higher disease activity. | [102] | |
| WB from SLE, RA, pSS patients, and HCs | Significant hypomethylation of two CpG sites within the IFI44L promoter can distinguish SLE patients from RA patients, pSS patients, and HCs, and was correlated with high disease activity and renal damage in SLE. | [72] | |
| PB from HCs, SLE, and DLE patients | The promoter hypomethylation of IFI44L and higher serum levels of IFN-a1 were detected in SLE patients relative to DLE patients and HCs, which may potentially serve as biomarkers to distinguish SLE from DLE. | [103] | |
| T cells from SLE patients and HCs | Promoter hypomethylation and increased expression of CD70 were detected in T cells of patients with SLE compared to HCs and may lead to overstimulation of B cells. | [104] | |
| T cells from active SLE patients, inactive SLE patients, and HCs | Hypomethylation of specific sequences flanking the ITGAL promoter increases the expression of ITGAL (also known as CD11a) in T cells from patients with active lupus compared to patients with inactive lupus and HCs. | [105] | |
| RA | CD14+ monocytes, CD19+ B cells, CD4+ naive T cells, and CD4+ memory T cells from RA patients and HCs | Promoter hypomethylation of CYP2E1 and DUSP22 genes were correlated with active and erosive disease, respectively. | [106] |
| WB from pre-treatment RA patients | Five differentially methylated CpG sites at diagnosis time can distinguish RA patients with a good response from RA patients without response to 3 months of TNFi therapy. | [107] | |
| T lymphocytes from pre-treatment RA patients | The methylation levels of two CpG sites at diagnosis time can distinguish RA patients with a good response from RA patients with a poor response to 6 months of DMARDs therapy. | [108] | |
| WB from early RA patients | A higher baseline global leukocyte DNA methylation was associated with a decreased clinical response to MTX therapy. | [81] | |
| WB DNA at baseline and following 4 weeks of MTX therapy from RA patients | The methylation status of four CpGs after 4 weeks of MTX initiation was associated with disease improvement after 6 months of therapy. | [109] | |
| Synovial tissue from RA patients and OA patients | Promoter hypermethylation and the decreased protein expression of the TNFRSF25 gene (also known as DR3), a member of the apoptosis-inducing Fas gene family, may be associated with resistance to apoptosis in RA synovial cells. | [110] | |
| FLSs from synovial tissues of patients with RA and OA | Hypomethylation downstream of the TSS of the IL-6R gene increases the expression of IL-6R in RASFs compared with OASFs and may be associated with an increase in the risk of joint destruction in RA. | [111] | |
| Synovial tissue from patients with OA and RA | Hypermethylation of two CpGs in an intronic enhancer region in the PTPN11 gene (also known as SHP2) increases the expression of PTPN11 in RA FLSs compared with OA FLSs and promotes RA FLSs aggressiveness and joint inflammation. | [112] | |
| FLSs from HCs and patients with RA in very early, resolving, established stages | Differentially methylation in the promoter of genes involved in pathways related to integrin, cadherin, Wnt signaling, the actin cytoskeleton, and components of the antigen presentation was found in FLSs from patients with very early, resolving, or established RA compared with HCs. | [113] | |
| SSc | Fibroblasts from biopsies of lesional skin and PB from patients with SSc and HCs | Promoter hypermethylation and the decreased expression of DKK1 and SFRP1 genes in fibroblasts and PMNCs of patients with SSc may lead to aberrant activation of the Wnt signaling pathway. | [82] |
| Dermal fibroblasts of SSc patients and HCs | Promoter hypermethylation and the decreased expression of the FLI1 gene, a potent repressor of the type I collagen gene, were detected in dermal fibroblasts of SSc patients compared with HCs. | [83] | |
| Dermal fibroblasts from SSc patients and HCs | miR-135b, which reduces collagen induction in fibroblasts, was downregulated in dermal fibroblasts from SSc patients compared with HCs. Its downregulation may result from methylation and elevated levels of MECP2. | [84] | |
| Dermal fibroblasts from SSc patients and HCs | Promoter hypermethylation and the decreased expression of the TIPARP gene (also known as PARP-1) were detected in the dermal fibroblasts from SSc patients compared with HCs. TIPARP downregulation may contribute to hyperactive TGF-β signaling and continuous fibroblast activation in SSc. | [85] | |
| Dermal fibroblasts from SSc patients and HCs | The level of MECP2 protein was elevated in fibroblasts from SSc patients compared with HCs. Increased binding of MECP2 to SFRP-1 promoter leads to transcriptional silencing of SFRP-1 and increased pro-fibrotic Wnt signaling. |
[86] | |
| CD4+ T cells from SSc patients and HCs | The global DNA methylation levels and DNMT1, MBD3, and MBD4 expression were reduced in CD4+ T cells from SSc patients compared with HCs. | [87] | |
| CD4+ T cells from SSc patients and HCs | Decreased methylation in regulatory elements and increased expression of the ITGAL gene, encoding CD11a, was detected in CD4+ T cells from SSc patients compared with HCs and may be associated with disease activity. | [88] | |
| CD4+ and CD8+ T cells from SSc patients and HCs | Hypomethylation and increased expression of type I IFN-associated genes were observed in CD4+ and CD8+ T cells from SSc patients compared with HCs. | [89] | |
| CD4+ T cells from SSc patients and HCs | Decreased methylation of the promoter region and increased expression of the TNFSF7 gene, encoding CD70, was detected in CD4+ T cells from SSc patients compared with HCs. | [90] | |
| CD4+ T cells from SSc patients and HCs | Decreased methylation of the CD40L regulatory sequence on the inactive X chromosome leads to increased expression of the CD40L in CD4+ T cells from female patients with SSc compared with female HCs. | [91] | |
| CD4+ T cells from SSc patients and HCs | Increased methylation of regulatory elements and the decreased expression of the FOXP3 were detected in CD4+ T cells from SSc patients compared with HCs. | [114] | |
| MVECs of skin biopsies from SSc patients and HCs | Promoter hypermethylation and the decreased expression of the BMPR2 gene, a member of the TGF-β superfamily of proteins that coordinate cell proliferation, differentiation, and survival, may contribute to inducing apoptosis in MVECs of SSc patients. | [115] | |
| Fibrotic lung tissue from SSc patients and control tissue from histologically normal areas of a peripheral lung removed at lung cancer resection as HCs | Hypermethylation and the decreased expression of the TCIM (also known as c8orf4) gene were detected in fibrotic lung fibroblasts from SSc patients compared with HCs fibroblasts. The aberrant expression of c8or4 decreases expression of anti-fibrotic PTGS2 (also known as COX2) and its product PGE2 in SSc. | [116] | |
| WB from twin pairs discordant for SSc | Methylation of 153 and 266 distinct CpG sites was correlated with lcSSc and dcSSc, respectively. | [92] | |
| Fibroblasts from dcSSc patients, lcSSc patients, and HCs | 2710 and 1021 CpG sites were differentially methylated in dcSSc and lcSSc, respectively. The majority of these CpG sites were hypomethylated in both dcSSc (61 %) and lcSSc (90 %). Hypomethylated genes such as ADAM12, COL23A1, COL4A2, ITGA9, MYO1E, PAX9, RUNX2, and RUNX3 were overexpressed in fibroblasts of both lcSSc and dcSSc patients compared with HCs fibroblasts and may contribute to fibrosis events in SSc. | [117] | |
| MS | WB from MS patients and HCs | Alu and LINE-1 were hypermethylated in MS patients compared to HCs. The methylation levels of these genes were associated with the degree of EDSS score in MS patients. | [93] |
| T cells from RRMS patients and HCs | Increased DNA methylation levels in the alternative promoter of the VDR gene lead to increased expression of the VDR in T cells of RRMS patients compared to HCs. | [94] | |
| PBMNs from MS patients and HCs | The decrease in methylation associated with an increase in IL-2RA expression was found in T cells from MS patients relative to HCs. | [95] | |
| Leukocytes of MS patients and HCs | Promoter hypermethylation and associated silencing of the PTPN6 gene (also known as SHP-1), a negative regulator of pro-inflammatory signaling, may increase leukocyte-mediated inflammation in MS patients compared to HCs. | [118] | |
| cfDNA of plasma or serum from MS patients and HCs | The increased levels of fully unmethylated cfDNA fragments from the MBP3 or WM1 loci can distinguish genomic debris from oligodendrocyte cell death in MS patients from normal cellular debris in HCs. | [119] | |
| Cortical white matter from the brains of MS patients and HCs | The PADI2 gene, which regulates the production of citrullinated MBP, showed decreased promoter methylation associated with the increased protein in the cortical white matter of MS patients compared to HCs. The aberrant expression of the PADI2 enzyme leads to myelin destabilization due to increased citrullinated MBP. | [120] | |
| NAWM from post-mortem brain FC specimens of MS patients and HCs | Hypermethylation and the decreased expression of oligodendrocyte survival regulatory genes, such as BCL2L2 and NDRG1, were found in NAWM of MS–affected brains compared to HC brains. While the proteolytic genes LGMN and CTSZ were hypomethylated, their increased expression may increase microglial-mediated antigen presentation. | [121] | |
| Post-mortem hippocampus from myelinated and demyelinated MS patients | 144 DMPs were detected in the demyelinated MS hippocampus compared to the myelinated MS hippocampus. The AKNA and SFRP1 genes showed hypomethylation and increased expression, while WDR81, NHLH2, and PLCH1 genes showed hypermethylation and associated silencing. Increased expression of AKNA, a gene involved in the regulation of CD40 and CD40 ligand interaction that induces the expression of TNF-α, as well as decreased expression of WDR81, a gene involved in nerve survival, may lead to neuronal damage. | [122] |
ADAM12: ADAM metallopeptidase domain 12; AKNA: AT-hook transcription factor; BMPR2: bone morphogenetic protein receptor 2; BCL2L2: bcl-2-like protein 2; CHST12: carbohydrate sulfotransferase 12; CYP2E1: cytochrome p450 family 2 subfamily E member 1; CD40L: CD40 ligand; COL23A1: collagen type XXIII alpha 1 chain; COL4A2: collagen type IV alpha 2 chain; cfDNA: circulating cell-free DNA; CTSZ: cathepsin Z; DZ: dizygotic; DLE: discoid lupus erythematosus; DMCpGs: differentially methylated CpGs; DAGs: DMR-associated genes; DUSP22: dual specificity phosphatase 22; DMARDs: disease-modifying antirheumatic drugs; DKK1: dickkopf WNT signaling pathway inhibitor 1; dcSSc: diffuse cutaneous SSc; DMPs: differentially methylated positions; EA: European-American; EDSS: expanded disability status scale; FLSs: fibroblast-like synoviocytes; FLI1: friend leukemia integration 1 transcription factor; FOXP3: forkhead box P3; FC: frontal cortex; HCs: healthy controls; IFN: interferon; IL-10: interleukin 10; IL-1R2: interleukin 1 receptor type 2; IFI44L: interferon induced protein 44 like; ITGAL: integrin, alpha L; IL-6R: interleukin-6 receptor; IL-2RA: interleukin 2 receptor subunit alpha; ITGA9: integrin subunit alpha 9; lcSSc: limited cutaneous SSc; LINE-1: long interspersed nuclear element 1; LGMN: legumain; MS: multiple sclerosis; MZ: monozygotic; MTX: methotrexate; MECP2: methyl-CpG binding protein 2; MBD: methyl-CpG binding domain protein; MVECs: microvascular endothelial cells; MYO1E: myosin IE; MBP: myelin basic protein; NAWM: normal-appearing white matter; NDRG1: N-myc downstream regulated 1; NHLH2: nescient helix-loop-helix 2; PB: peripheral blood; PBMNs: peripheral blood mononuclear cells; pSS: primary Sjögren's syndrome; PTPN11: protein tyrosine phosphatase non-receptor type 11; PTPN6: protein tyrosine phosphatase non-receptor type 6; PAX9: paired box 9; PADI2: peptidyl arginine deiminase 2; PTGS2: prostaglandin-endoperoxide synthase 2; PLCH1: phospholipase C Eta 1; PGE2: prostaglandin E2; OA: osteoarthritis; OASFs: osteoarthritis synovial fibroblasts; RA: rheumatoid arthritis; RASFs: rheumatoid arthritis synovial fibroblasts; RUNX: runt-related transcription factor; RRMS: relapsing-remitting MS; SLE: systemic lupus erythematosus; SSc: systemic sclerosis; SLEDAI: systemic lupus erythematosus disease activity index; TNFRSF25: TNF receptor superfamily member 25; TCIM: transcriptional and immune response regulator; SFRP1: secreted frizzled related protein 1; TSS: transcription start site; TGF-β: transforming growth factor β; TNFSF7: tumor necrosis factor ligand superfamily member 7; TNF-α: tumor necrosis factor-α; TNFi: tumor necrosis factor inhibitor; TIPARP: TCDD inducible poly(ADP-ribose) polymerase; VDR: vitamin D receptor; WB: whole-blood; WM1: white matter 1; WDR81: WD repeat domain 81.
4.3. DNA methylation in metabolic disorders
4.3.1. Type 2 diabetic Mellitus
The signs of DNA methylation patterns in 1649 CpG sites of about 843 genes involved in the pathogenesis of Type 2 diabetic (T2D), such as those encoding insulin and PDX1 in the pancreatic islets, clearly suggest that epigenetic alterations could impair insulin release. Overexpression of CDKN1A, PDE7B, and SEPTIN9 in clonal β-cells of pancreatic islets due to induction of DNA hypomethylation not only halts the proliferation of these cells but also reduces their sensitivity to glucose fluctuations, leading to the lack of insulin production [123]. Notably, DNA methylation alterations in T2D patients are not restricted to the genes expressed in pancreatic tissue, as the Illumina sequencing arrays identified methylation sites in more than thousands of genes expressed in other tissues, such as human adipose tissue, liver, and skeletal muscle [[124], [125], [126], [127]]. Identifying these sites sheds light on the mechanisms by which we can control the development of T2D with some common behaviors such as physical exercise or energy-dense diets with high-fat content. In the skeletal muscle of patients with T2D, for instance, it has been reported that the methylation pattern of the gene encoding PPARGC1A is altered in response to exercise [128]. The overexpression of PPARGC1A, a primary regulator of liver gluconeogenesis, can alleviate the symptoms in T2D patients [129]. The existence of such non-pancreatic methylation sites can connect T2D to other inflammatory difficulties. Indeed, alteration in the methylation of these sites upregulates the expression of pro-inflammatory mediators within the target cell, which may increase the risk of cardiovascular and reproductive diseases or the incidence of cancer [130].
4.3.2. Obesity
The genome-wide DNA methylation analyses showed the presence of several methylation sites in obese patients, especially in the gene encoding HIF3A [131,132]. Over 1000 CpG DNA methylation signatures have also been identified in FTO, CCL18, MTCH2, SPP1, and IRS genes in subcutaneous adipose tissue (SAT). Interestingly, the methylation of these CpG sites has a tight correlation with the higher body mass index (BMI) [132]. Some of these methylation sites were similar to those previously described in T2D; highlighting that these metabolic diseases might influence each other [132]. Kuehnen et al. proposed that the hypermethylation in intron 2/exon 3 of POMC may be associated with childhood obesity [133]. Also, such methylation sites can be used as biomarkers to predict the risk of future T2D development in obese patients [134]. Table 3 summarizes the results of various studies highlighting the role of DNA methylation markers in metabolic disorders.
Table 3.
Potential DNA methylation markers in metabolic disorders.
| Diseases | Cell/tissue type | Main findings | Ref |
|---|---|---|---|
| T2D | Pancreatic islets from non-diabetics and diabetics | 1649 CpG sites annotated to 843 genes, including INS, PDX1, ADCY5, FTO, IRS1, KCNQ1, and TCF7L2, were differently methylated in islets from T2D patients relative to HCs. 102 of these genes, including CDKN1A, PDE7B, SEPTIN9, and EXOC3L2 exhibited differential expression in T2D islets. | [123] |
| Muscle and SAT from MZ pairs discordant for T2D | Hypermethylation in promoter regions of known T2D-related genes, such as PPARGC1A in muscle and HNF4A in adipose tissue, were detected in the MZ twins discordant for T2D. | [124] | |
| SAT from MZ pairs discordant for T2D and independent case-control cohorts | 15,627 CpG sites annotated to 7046 genes, including PPARG, KCNQ1, TCF7L2, and IRS1, were differently methylated in adipose tissue from T2D patients relative to HCs, and 1410 of these sites did also exhibit differential DNA methylation in the MZ twins discordant for T2D. | [125] | |
| Liver biopsies from T2D patients and nondiabetic subjects | 251 CpG sites were differently methylated in the liver from T2D patients relative to HCs, and 94 % of the CPG sites displayed decreased DNA methylation that may be explained by reduced circulating folate levels in the human diabetic liver. | [126] | |
| Liver biopsies from obese women with T2D and obese women with normal glucose levels | The hypomethylation associated with overexpression of PDGFA in the liver of obese patients with T2D was correlated with increased insulin resistance, hyperinsulinemia, and increased T2D risk. | [127] | |
| WB and skeletal muscle from lean healthy, obese non-diabetic, and T2D volunteers before and after exercise | In patients with T2D, the decreased methylation levels of PPARGC1A (also known as PGC-1α) promoter after exercise may be used potentially as a prognostic marker for monitoring lifestyle intervention. | [128] | |
| Adipose tissue from healthy men, with a previous low level of physical activity, before and after exercise intervention | 17,975 CpG sites annotated to 7663 genes, 18 obesity, and 21 T2D candidate genes (such as TCF7L2 and KCNQ1) showed altered levels of DNA methylation in adipose tissue after the exercise intervention. | [135] | |
| Blood DNA of cases that were all non-diabetic at baseline | DNA methylation at the ABCG1 locus was associated with a 9 % increased risk, while DNA methylation at the PHOSPHO1 locus was associated with a 15 % decreased risk of future T2D. | [136] | |
| PB leukocytes from T2D cases and HCs | The promoter region of 1091 genes hypermethylated in the blood DNA of T2D patients compared to HCs. Promoter hypermethylation and associated silencing of the NT5C2 gene were strongly correlated with increased susceptibility to T2D. |
[137] | |
| Obesity | Adipose tissue and WB samples | Increased methylation at the HIF3A locus in adipose tissue and blood cells was associated with increased BMI. | [131] |
| Adipose tissue and WB samples | DNA methylation and expression of 2825 genes (e.g., FTO, MTCH2, ITIH5, CCL18, IRS1, and SPP1) were associated with BMI. | [132] | |
| PB of obese children and normal-weight individuals | Hypermethylation in intron 2/exon 3 of POMC might have an association with childhood obesity. | [133] | |
| WB from obese cases and normal weight controls | 187 CpG sites were significantly associated with BMI. 62 of 187 methylation markers were associated with the incidence of T2D and can be used as a biomarker to predict the risk of future T2D development in obese patients. | [134] | |
| CD4+ T cells of participants | Four CpG sites annotated to CPT1A, PHGDH, CD38, and LINC00263 were associated with BMI and WC. The methylation level of a CpG site near the TSS of CD38 was positively associated with BMI and WC, while a CpG site in the first intron of CPT1A was inversely associated with BMI and WC. | [138] | |
| WB and CD4+ T cells | 83 CpG sites were significantly associated with BMI, and a CpG site in the intron of SREBF1 gene was strongly associated with BMI, adiposity-related traits, and coronary artery disease. | [139] | |
| PB leukocytes and neutrophils of obese and lean individuals | 54 CpG sites were significantly associated with obesity, and the decreased DNA methylation level and increased expressions of SOCS3, CISH, PIM3, and KLF4 genes were associated with obesity. | [140] |
ADCY5: adenylate cyclase 5; ABCG1: ATP binding cassette subfamily G member 1; BMI: body mass index; CDKN1A: cyclin dependent kinase inhibitor 1A; CCL18: C–C motif chemokine ligand 18; CISH: cytokine inducible SH2 containing protein; EXOC3L2: exocyst complex component 3 like 2; FTO: FTO alpha-ketoglutarate dependent dioxygenase; HCs: healthy controls; HNF4A: hepatocyte nuclear factor 4 alpha; HIF3A: hypoxia inducible factor 3 subunit; alpha; IRS: insulin receptor substrate; INS: insulin; ITIH5: inter-alpha-trypsin inhibitor heavy chain 5; KCNQ1: potassium voltage-gated channel subfamily Q member 1; KLF4: kruppel like factor 4; LINC00263: long intergenic non-coding RNA 00263; MZ: monozygotic; MTCH2: mitochondrial carrier 2; NT5C2: 5′-nucleotidase, cytosolic II; PB: peripheral blood; PDX1: pancreatic and duodenal homeobox 1; PDE7B: phosphodiesterase 7B; PPARGC1A: PPARG coactivator 1 alpha; PPARG: peroxisome proliferator activated receptor gamma; PDGFA: platelet derived growth factor subunit A; PHOSPHO1: phosphoethanolamine/phosphocholine phosphatase 1; PHGDH: phosphoglycerate dehydrogenase; PIM3: pim-3 proto-oncogene, serine/threonine kinase; SAT: subcutaneous adipose tissue; SEPTIN9: septin 9; SPP1: secreted phosphoprotein 1; SREBF1: sterol regulatory element binding transcription factor 1; SOCS3: suppressor of cytokine signaling 3; TSS: transcription start site; T2D: type 2 diabete; TCF7L2: transcription factor 7 like 2; WB: whole-blood; WC: waist circumference.
4.4. DNA methylation in cancer
DNA methylation alterations are one of the principal mechanisms that cancer cells recruit to prolong their survival, especially in the presence of anti-cancer signaling. DNA methylation can diminish the expression of a wide range of tumor suppressor proteins, foremost TP53, which is known as a guardian of the genome. Moreover, loss of CpG methylation can make the genome unstable and make it more vulnerable to genetic abnormalities [141]. As epigenetic alteration is an age-related phenomenon, it has been widely believed that changes in DNA methylation patterns happen during the aging of normal cells, making them transform into malignant counterparts. The confirmation that the degree of DNA methylation of one tissue differs in different cancer patients based on age [142].
4.4.1. DNA methylation in hematological malignancies
Hematologic malignancy was the first cancer in which the influence of DNA methylation alteration in the control of cell differentiation, proliferation, and survival has been described. Interestingly, the signs of abnormalities in DNA methylation machinery are evident in all types of hematologic malignancies from acute leukemia to chronic neoplasms such as myeloproliferative neoplasms (MPNs) and myelodysplastic syndrome (MDS). For example, the mutation in DNMT3A with the ability to reduce the catalytic activity of this DNA methyltransferase is a common feature in early colons of AML, MDS, T cell lymphoma, and adult T acute lymphoblastic leukemia (ALL) [[143], [144], [145], [146], [147], [148], [149], [150], [151]]. Notably, the importance of DNMT3A mutation is to the degree that endows it the prognostic value in hematologic malignancies [144,151]. Interestingly, more than half of DNMT3A mutations in AML patients occur at amino acid R882 in the catalytic domain of the enzyme. It seems that R882 mutations have a dominant-negative effect on the formation of wild-type DNMT3A tetramers, leading to reduced catalytic activity and consequent focal hypomethylation compared to the hypermethylation pattern observed with wild-type DNMT3A R882 in AML [145]. DNMT3A mutations are generally present in the ancestral or founder preleukemic clone, and they may create an environment in which additional mutations trigger malignant clones.
Co-occurring with DNMT3A mutations, TET2 mutations have also been reported in AML, MDS, T cell lymphoma, and MPNs [[147], [148], [149],[152], [153], [154], [155]]. The collaboration between TET2 and DNMT3A mutations could block the differentiation of hematopoietic stem cells (HSCs) and increase their self-renewal capacity by altering the methylation pattern of several genes [156]. TET2 LoF mutations and isocitrate dehydrogenases (IDH) GoF mutations are mutually exclusive in AML, suggesting that lesions in these genes may be biologically redundant [157]. DNMT1 and DNMT3B are other DNA methylation writers whose expressions are manipulated in hematologic malignancies. In chronic myeloid leukemia (CML), DNMT1, DNMT3A, and DNMT3B overexpression contribute to the progression of the chronic phase into the acute phase [158]. In agreement, the up-regulation of these enzymes in AML can support the survival and proliferation of leukemic cells [158].
In addition to the mutation in DNA methylation machinery, the aberrant DNA methylation in the promoter of the tumor suppressor genes (TSGs) plays a critical role in the pathogenesis and progression of blood malignancies. Recent studies have reported that there is a good correlation between the DNA methylation status of the gene prompters and the residual disease in patients with neuroblastoma, natural killer cell lymphoma, diffuse large B cell lymphoma (DLBCL), and ALL. Residual methylation of gene promoters might be potential biomarkers for monitoring minimal residual disease (MRD) in patients with blood malignancies [159].
4.4.2. DNA methylation in solid tumors
The incidence rate of mutations in DNA methylation machinery in solid tumors is lower compared to hematologic malignancies. For example, as DNMT3A mutations have been detected in the majority of cases of hematologic malignancies, this abnormality is only detectable in less than 2 % of solid tumors based on the last reports of the Cancer Genome Atlas (TCGA) [160,161]. Aberrant alternative splicing in the DNMT3B gene, which gives rise to DNMT3B4, is mainly detectable in hepatocellular carcinoma. The non-functional DNMT3B4 __which lacks DNMT motifs__ induces hypomethylation in pericentromeric satellite regions, increasing the sensitivity of the genome to additional mutations [162]. Mutations within MBD domains of DNA methylation readers are other abnormalities of the DNA methylation machinery in different solid tumors. Notably, the function of mutant MBD proteins is distinct in each type of cancer. In this vein, MBD2 is the best example. MBD2 overexpression in prostate cancer is associated with aberrant hypermethylation of CpG islands of a tumor suppressor protein named GSTP1 [163]. In colorectal cancer, MBD2 recognizes methylated sites in promotors of p16 and p14 and makes an anchor for the binding of histone deacetylases (HDACs) to promote gene silencing [164]. Therefore, in cancer cells, MBD2 is not only a reader of DNA methylation machinery but also can promote a rewriter, as it can make a complex with several DNMTs to change the expression of a broad group of tumor suppressor genes [163]. Another MBD protein whose mutation seems to play a fundamental role in tumorigenesis is MBD4, a methylation reader that reduces the chance of DNA repair in malignant cells. The incidence of MBD4 mutations in cancer with microsatellite instability, such as colorectal and gastric cancer, is higher [165,166]. The frequency of mutations in other genes encoding the components of DNA methylation machinery is very infrequent in solid tumors.
Although occurring on a rare scale, DNA methylation-based biomarkers have found solid feet in the early diagnosis of many solid tumors due to easy detection, long stability, presence in various body fluids, and non-invasiveness detection, and, more importantly, cell-type specificity. These alterations can also be used as prognostic factors. Of particular interest, analysis of the DNA methylation pattern can be useable for establishing the type of those cancers with unknown sources or carcinoma of unknown primary (CUP) [167]. Thus far, two tests have succeeded to received FDA approval for analyzing the alteration of DNA methylation: Cologuard, also known as NDRG4 or BMP3, is developed to analyze DNA methylation patterns in stool samples [168], and Epi proColon (SEPTIN9) analyzes the DNA methylation in blood samples of patients [169,170]. Therefore, studying DNA methylation alterations seem to be fertile ground for revolutionizing the molecular landscape of solid tumors. To provide a better overview, we summarized the aberrations of the DNA methylation machinery and its outcomes in different cancer types spanning from hematologic malignancies to various solid tumors in Table 4. The promising applications of DNA methylation markers in solid tumors are also represented in Table 5.
Table 4.
Aberrations of the DNA methylation machinery in human cancers.
| Cancer type | Aberrations | Outcome of aberrations | Ref | |
|---|---|---|---|---|
| DNA methyltransferases; writers | ||||
| DNMT1 | CRC | Rare inactivating mutations | – | [171] |
| PA | Overexpression | Correlated with aggressive disease and high-methylation tumors. | [172] | |
| GC | Overexpression | Regional DNA hypermethylation and gene silencing; Correlated to the development of poorly differentiated GC. | [173] | |
| LC | Overexpression | Correlated with aberrant TP53/SP1 pathway and poor prognosis. | [174] | |
| CML | Overexpression | Increased expression during progression to the acute phase. | [158] | |
| AML | Overexpression | Regional DNA hypermethylation and gene silencing. | [158] | |
| DNMT3A | PA | Overexpression | Correlated with aggressive disease and high-methylation tumors. | [172] |
| VSCC | Overexpression | Correlated with an increased risk of LVR. | [175] | |
| GC | Allelic loss | Regional DNA hypomethylation. | [176] | |
| LC | Allelic loss | Regional DNA hypomethylation. | [176] | |
| CML | Overexpression | Increased expression during progression to the acute phase. | [158] | |
| AML | Overexpression | Regional DNA hypermethylation and gene silencing. | [158] | |
| AML | Inactivating mutation | DNA hypomethylation by DNMT3A R882 mutation correlated with a poor prognosis. | [[143], [144], [145]] | |
| MDS | Inactivating mutation | Causes regional DNA hypomethylation, and is correlated with worse OS and more rapid progression to AML. | [146] | |
| TCL | Inactivating mutation | Regional DNA hypomethylation. | [[147], [148], [149], [150]] | |
| T-ALL | Inactivating mutation | Causes regional DNA hypomethylation, and correlated with poor prognosis in adult T-ALL. | [151] | |
| NHL | Allelic loss | Regional DNA hypomethylation. | [176] | |
| DNMT3B | HCC | Overexpression | Increased expression of DNMT3B/OCT4 by the IL6/STAT3 pathway triggers sorafenib resistance and early recurrence. | [177] |
| HCC | Overexpression of DNMT3B4 | Correlated with DNA hypomethylation in pericentromeric satellite regions and genome instability. | [162] | |
| EC | Overexpression | Regional DNA hypermethylation. | [178] | |
| CML | Overexpression | Increased expression during progression to the acute phase. | [158] | |
| AML | Overexpression | Regional DNA hypermethylation and gene silencing. | [158] | |
| Methyl-CpG binding proteins (MBDs); Reader | ||||
| MECP2 | BrC | Overexpression | Correlated with higher levels of ER in both neoplastic and non-neoplastic tissues. | [179] |
| MBD1 | LC | SNPs (rs125555, rs140689) | Correlated with increased LC risk. | [180] |
| PC | Overexpression | Promotes PC invasion and metastasis by epigenetic down-regulation of E-cadherin. | [181] | |
| MBD2 | PrC | Overexpression | Associated with aberrant hypermethylation of the GSTP1. | [163] |
| MBD3 | EC | Deletion | Frequent deletion of a small segment of chromosome 19 containing MBD3. | [182] |
| MBD4 | CRC and GC | Frameshift mutations | Correlated with microsatellite instability. | [165,166] |
| HCC | Downregulated | Significantly correlated with poorer tumor differentiation and involvement of portal vein. | [183] | |
| DNA-demethylating enzymes; Erasers | ||||
| TET1 | AML | Rare LOF mutations | – | [184] |
| TET2 | HM | Frequent LOF mutations | Causes promoter DNA hypermethylation and gene silencing, and is correlated with poor prognosis in AML and lymphomas (AITL and PTCL-NOS). | [[147], [148], [149],[152], [153], [154], [155]] |
| Metabolic enzymes (enzymes of the tricarboxylic acid (TCA) cycle) | ||||
| IDH1 | LGG, CS, CCCs, TC, and HM | Frequent oncogenic mutation* | IDH1 mutant produces 2-HG, which inhibits demethylases and triggers hypermethylation of DNA and histones. | [157,[185], [186], [187], [188], [189]] |
| IDH2 | LGG, CS, and HM | Frequent oncogenic mutation** | IDH2 Mutant produces 2-HG, which inhibits demethylases and triggers hypermethylation of DNA and histones. | [149], [157], [[185], [186], [187], [188]] |
| FH | UF and RC | Inactivating mutations | Accumulation of fumarate inhibits demethylases and triggers hypermethylation of DNA and histones. | [190] |
| SDH | HPGL/PCC and GIST | Inactivating mutations | Accumulation of succinate inhibits demethylases and triggers hypermethylation of DNA and histones. | [191,192] |
*R132H being the most common; ** mutations at R172 and R140 being most common; AML: acute myeloid leukemia; AITL: angioimmunoblastic T-cell lymphoma; BrC: breast cancer; CRC: colorectal cancer; CML: chronic myelogenous leukemia; CMML: chronic myelomonocytic leukemia; CS: chondrosarcoma; CCCs: cholangiocarcinomas; DNMT: DNA (cytosine-5)-methyltransferase; EC: endometrial cancer; FH: fumarate Hydratase; GC: gastric cancer; GIST: gastrointestinal stromal tumors; GSTP1: glutathione S-transferase pi 1; HCC: hepatocellular carcinoma; HM: hematological malignancies; 2-HG: 2-hydroxyglutarate; IDH: isocitrate dehydrogenase; IL6/STAT3: interleukin-6/signal transducer and activator of transcription 3; LC: lung cancer; LGG: low-grade gliomas; LOF: loss-of-function; LVR: local vulvar recurrence; MECP2: methyl CpG binding protein 2; MBD: methyl-CpG binding domain; MDS: myelodysplastic syndrome; MPNs: myeloproliferative neoplasms; NHL: non-hodgkin lymphoma; OS: overall survival; OCT4: octamer-binding transcription factor 4; ER: oestrogen receptor; PA: pituitary adenoma; PC: pancreatic cancer; PrC: prostate cancer; HPGL/PCC: paraganglioma/phaeochromocytoma syndrome; PTCL-NOS: peripheral T-cell lymphoma, not otherwise specified; RC: renal cancer; SNPs: single nucleotide polymorphisms; SDH: succinate dehydrogenase; TET: tet methylcytosine dioxygenase; TCL: T-cell lymphoma; T-ALL: T-cell acute lymphoblastic leukemia; TC: thyroid cancer; UF: uterine fibroid; VSCC: vulvar squamous cell carcinoma.
Table 5.
Potential DNA methylation markers in human cancers.
| Sample type | Biomarkers | Indication | Status | Ref | |
|---|---|---|---|---|---|
| Markers for diagnosis/detection | |||||
| BC | Voided urine (Patients diagnosed with hematuria) | TWIST1, ONECUT2, and OTX1 methylation | Diagnosis (NIBC detection) | CE-IVD device (AssureMDX) | [193] |
| BC | Urine samples | Methylation levels of TRNA-Cys, SIM2, and NKX1-1 and 2 internal control loci | Diagnosis | Bladder CARE™ (CE-marked IVD) test | [194] |
| BC | Urine samples | ZNF671, OTX1, and IRF8 methylation | Diagnosis | Preclinical | [195] |
| BrC | Plasma cfDNA | Differentially methylated regions (DMRs) of SPAG6, LINC10606 and TBCD/ZNF750 genes | Diagnosis (early detection of triple-negative breast cancer) | Preclinical | [196] |
| BrC | PBMCs | Four selected PBMCs DNA methylation markers | Diagnosis | Preclinical | [197] |
| Small BrC | Tissue samples | GCM2, ITPRIPL1, CACNA1E, and DLGAP2 methylation | Diagnosis | Preclinical | [198] |
| CRC | Stool (Patients who are candidates for screening) | BMP3, and NDRG4 methylation | Early detection of CRC | FDA-approved (ColoGuard) | [168] |
| CRC | Stool (Patients unwilling or unable to undergo a more invasive exam) | VIM methylation | Early detection of CRC | CLIA level (ColoSure) | [199] |
| CRC | Plasma (Patients, 50 years or older, with average risk for CRC, who decline other CRC screening) | SEPTIN9 methylation | Early detection of CRC | FDA approved (Epi proColon); CLIA level (ColoVantage) | [169,170,200] |
| CRC | Blood samples | MYO1G methylation | Diagnosis (Monitoring disease) | Preclinical | [201] |
| CRC | Stool | Methylated COL4A1, COL4A2, TLX2, and ITGA4 | Diagnosis | Preclinical | [202] |
| CRC | Plasma samples | Methylated SFRP1, SFRP2, SDC2, and PRIMA1 | Diagnosis | Preclinical | [203] |
| CC | A cervical smear of women who are HPV+ with abnormal cytology findings | ASTN1, DLX1, ITGA4, RXFP3, SOX17, and ZNF671 methylation | Triage of unclear CC screening tests | CE-IVD (GynTect®) | [204] |
| CC | Cervical scrape and vaginal samples of HPV+ women | FAM19A4, and hsa-mir124-2 methylation | Triage of unclear CC screening tests | CE-IVD (QIAsure) | [205] |
| CC | Liquid-based cytology (LBC) samples | Methylation detection of the promoter region of EPB41L3 and five late regions of HPV types 16, 18, 31, and 33 by pyrosequencing | Predicts incident high-grade CIN and ICC among high-risk HPV+ women | Preclinical (S5 DNA methylation classifier test) | [206] |
| CC | Cervical brush samples | PCDHGB7 hypermethylation-based Cervical cancer Methylation (CerMe) detection | CerMe detection efficiently differentiated CIN grade 2 or worse (CIN2+) from CIN grade 1 or normal (CIN1−) women | Preclinical | [207] |
| CC | Histological cervical specimens | ASTN1, DLX1, ITGA4, RXFP3, SOX17, and ZNF671 | ZNF671 methylation has diagnostic value for the diagnosis of CIN2+ and CIN3+. | Preclinical (GynTect® assay) | [208] |
| GC | Circulating cell-free DNA | 5-hydroxymethylcytosine of FBXL7, PDE3A, TPO, SNTG2, and STXBP5 | Diagnosis | Preclinical | [209] |
| GC | Plasma cfDNA | Methylated RNF180, and SEPTIN9 | Diagnosis | Preclinical | [210] |
| HCC | Plasma cfDNA | SEPTIN9 methylation | Detection of HCC in patients with cirrhosis | CE-IVD device (HCCBloodTest) | [211] |
| HCC | Plasma cfDNA | A single F12 gene CpG site | Diagnosis | Preclinical | |
| HPV-HNC | Saliva samples | HPV late genes and long control region (LCR) | Higher levels of DNA methylation in HPV late genes is a potential supplementary biomarker for salivary HPV detection-based HPV-HNC screening. | Preclinical | [212] |
| LC | Plasma (Increased risk patients defined by life history, symptoms, and radiological findings in the lung) | SHOX2 and PTGER4 methylation | Detection of LC in patients at increased risk for the disease | CE-IVD device (Epi proLung) | [213] |
| LC | Plasma cfDNA | Circulating tumor DNA methylation model by Next-generation sequencing (NGS) | Accurate diagnosis of early stage lung cancer from pulmonary nodule patients | Clinical trial (NCT03651986); PulmoSeek model |
[214] |
| LC | Plasma cfDNA | Circulating tumor DNA methylation model by Next-generation sequencing (NGS) | Early diagnosis of lung cancer from pulmonary nodule patients | Clinical trials (NCT03181490, NCT03651986); PulmoSeek plus model (CE Marked) | [215] |
|
NSCLC SCLC |
Plasma cfDNA | Six-marker panel methylation-based plasma test | Early detection | Clinical trial (NCT02373917); Lung EpiCheck (Nucleix) | [216] |
| LC | Broncho-exfoliated cells (BEC) | SHOX2 and RASSF1A methylation | Diagnosis | Preclinical (LungMe® kit) | [217] |
| LC | Bronchial washing fluid | PCDHGA12 and CDO1 methylation | Diagnosis | Preclinical | [218] |
| OC | Plasma cfDNA | ZNF154, C2CD4D, and WNT6 | Ovarian cancer screening in high-risk populations | Preclinical | [219] |
| EOC | CfDNA liquid biopsy | A cfDNA Methylation Liquid Biopsy test (OvaPrint) | Diagnosis (Risk assessment of High-grade serous ovarian carcinoma in symptomatic women) | Preclinical | [220] |
| PCa | Prostate biopsy from men with established risk factors | GSTP1, APC, and RASSF1 methylation | Detection of occult PCa in men with previous histopathologically negative biopsies who were considered to be at risk for harboring cancer | CE-IVD device (ConfirmMDX) | [221] |
| PCa | Urine extracellular vesicles (EVs) DNA | RASSF1 methylation | Diagnosis | Preclinical | [222] |
| PCa | Plasma cfDNA | A targeted DNA methylation sequencing panel | Detection of Neuroendocrine PCa | Preclinical | [223] |
| PCa | Prostate tissue | GSTP1, APC, TNFRFS10c, RASSF1A, and RUNX3 methylation | Diagnosis (APC was associated with an increased risk of biochemical recurrence.) | Preclinical | [224] |
| Markers for prognosis | |||||
| BC | PB samples | DNA methylation clocks | Age acceleration was associated with worse OS | Preclinical | [225] |
| CRC | Plasma samples | High methylated SEPTIN9 levels | Shorter OS and PFS | Preclinical | [226] |
| CRC | Primary tumor biopsy samples | Hypermethylation of BMP2 | Worse OS | Preclinical | [227] |
| CRC | Primary tumor biopsy samples | Methylation level of POU3F3, SYN2, and TMEM178A | Significantly related to poorer survival in the high-risk tumor mutation burden (TMB) samples | Preclinical | [228] |
| CRC | Circulating tumour DNA (ctDNA) methylation haplotype patterns | ctDNA methylation haplotype patterns using the ColonES assay | CRC patients with high preoperative ctDNA methylation levels have worse relapse-free survival (RFS) than patients with low ctDNA methylation levels | Preclinical | [229] |
| CRC | Tumor biopsy samples | Hypermethylation of 3 site (cg02604524, cg07628404, and cg27364741) | Predicts poor survival | Preclinical | [230] |
| CC | Plasma samples | SEPTIN9 methylation | Predicts pelvic nodal metastasis | Preclinical | [231] |
| GBM | Tomur samples | MGMT methylation | Higher PFS and OS | Phase II and III clinical trials | [232] |
| GBM | Tissue samples | Hypermethylation of specific LTBR CpG sites | Predicts better OS | Preclinical | [233] |
| HCC | Tissue samples | Methylation level of cg01798157 in the BTG2 gene | Shorter OS | Preclinical | [234] |
| HCC | Liquid biopsy | SEPTIN9 methylation | Poor OS | Preclinical | [235] |
| OC | Primary OC samples with tumor protein 53 (TP53) wild-type | Five aberrant methylated differentially expressed genes (DEGs) | Predict the risk of death | Preclinical | [236] |
| PC | Pancreatic cancer tissues and adjacent non-cancer tissues | CHFR promoter methylation | Significantly associated with lymph node metastasis | Preclinical | [237] |
| PCa | Biopsy tissues | Methylation levels of ADM, AEN, CCND1, CDC2, EPHX3, RARB, and VCAM1 | Predicts risk of metastasis in untreated localized PCa patients | Preclinical | [238] |
| PCa | Biopsy tissues | Hypermethylation of CCND2 and GSTP1 | Associated with an increased risk of tumor progression | Preclinical | [239] |
| Markers for spatial localization | |||||
| CUP | Fresh frozen or FFPE tumor biopsy | The algorithm established from Illumina 450K/EPIC methylation arrays | Predict cancer tissue of origin to enable the direction of tumor type-specific therapy | CE-IVD device (EPICUP) | [167] |
| Multiple types of tumors | Plasma cfDNA (from cancer and non-cancer participants) and FFPE tumor biopsies | A panel of >100,000 informative methylation regions (whole-genome bisulfite sequencing) | Multi-cancer early detection, as well as TOO prediction | Clinical trial | [240] |
| Multiple types of tumors | Plasma (cfDNA) and primary tumor biopsy samples | A panel of 42,374 CpG clusters was established using TCGA tumor data and healthy plasma data (CancerLOCATOR) | Non-invasive cancer diagnosis and TOO prediction | Preclinical | [241] |
| Multiple types of tumors | Tumor tissue, adjacent normal tissue, and normal WB samples | Selected 3139 CpG sites that were hypermethylated in more than 10 % of tumors within any of the 33 cancer types (using HM27/HM450K array) | Diagnosis and prediction of the cell of origin (Classification of tumors using DNA hypermethylation, mRNA–miRNA expression levels, and reverse-phase protein data) | Preclinical | [242] |
| Markers for drug response | |||||
| BC | Pretreatment cfDNA | The levels of MIR145 core promoter | Reduced pretreatment cfDNA methylation of MIR145 core promoter predicts response of therapy | Preclinical | [243] |
| BC | Plasma cfDNA | Methylation-based response score (mR-score) | Predicts response to neoadjuvant chemotherapy | Preclinical | [244] |
| BrC | FFPE tissue samples from LN+, ER+ and HER2- HRBC patients | Higher methylation levels of PITX2 | Predicts resistance to anthracycline-based chemotherapy | CE-IVD (Therascreen®PITX2 RGQ) | [245] |
| BrC, OC | Primary tumor biopsy samples | BRCA1 methylation | Predicts sensitivity to PARP inhibitor | Phase II clinical trial | [246] |
| EOC | Tissue sample | DDR2 hypomethylation | Predicts resistance to chemotherapy | Preclinical | [247] |
| GBM | Primary tumor biopsy and PB samples | MGMT methylation | Predicts sensitivity to temozolomide | CE-IVD Therascreen MGMT Pyro kit | [248] |
| GBM | Primary and relapsed samples | Mean higher global DNA methylation | Associated with better radiotherapy response and less aggressive phenotype | Preclinical | [249] |
| CRC | Primary tumor biopsy samples | SRBC methylation | Predicts resistance to oxaliplatin | Preclinical | [250] |
| CRC | Primary tumor biopsy samples | DERL3 methylation | Predicts sensitivity to glycolysis inhibitors | Preclinical | [251] |
| CRC | FFPE tissue samples | LINE-1 hypomethylation | Predicts sensitivity to 5-FU | Preclinical | [252] |
| CRC | Tissue samples | TFAP2E methylation | Predicts resistance to chemotherapy | Preclinical | [253] |
| mCRC | Metastatic tumor biopsy samples | CHFR methylation | Predicts sensitivity to Irinotecan | Preclinical | [254] |
| mCRC | Plasma cfDNA | NPY methylation | Predicts respose to therapy | Preclinical | [255] |
| GC, CC | Primary tumor biopsy samples | TP53TG1 methylation | Predicts resistance to DNA-damaging drugs | Preclinical | [256] |
| OC, NSCLC | Primary tumor biopsy samples | SLFN11methylation | Predicts resistance to platinum drugs | Preclinical | [257] |
| NSCLC | Tumor biopsy samples | Unmethylated FOXP1 | Predicts sensitivity to PD-1 blockade | Preclinical | [258] |
| Markers for monitoring residual disease or predicting cancer recurrence | |||||
| BC | Voided urine | Hypermethylated SOX1 and IRAK3; hypomethylated LINE1 | Predicts bladder cancer recurrence | CLIA LDT (Bladder CARE™) | [259] |
| NMIBC | Voided urine | Score over 15 methylation markers | Monitoring for tumor recurrence in patients previously diagnosed with bladder cancer | CE-IVD (BladderEpiCheck®) | [260,261] |
| CRC | Plasma | IKZF1 and BCAT1 methylation | Early detection of CRC recurrence, and detection of residual disease post-surgical resection | CLIA LDT (COLVERA™) | [262] |
| CRC | Plasma cfDNA | SEPTIN9 methylation | Serial detecting of mSEPTIN9 could be a potential biomarker for minimal residual disease (MRD) in CRC patients. | Preclinical | [263] |
| CRC | Tumor biopsy samples | Eight-CpG DNA methylation signature | Predicts disease recurrence in patients with stage II CRC | Preclinical | [264] |
| HNSCC | Plasma cfDNA at the time of post-surgically (days 2–30) from patients with HNSCC | SEPTIN9 methylation | SEPT9 ccfDNA methylation could be a potential biomarker for detecting minimal residual disease (MRD) in HNSCC patients | Preclinical | [265] |
ASTN1: astrotactin 1; APC: adenomatous polyposis coli; BC: bladder cancer; BrC: breast cancer; BMP3: bone morphogenetic protein 3; BRCA1: breast cancer 1; BCAT1: branched chain amino acid transaminase 1; CRC: colorectal cancer; CC: cervix cancer; CIN: cervical intraepithelial neoplasia; CUP: cancer of unknown primary; cfDNA: circulating cell-free DNA; CHFR: checkpoint with forkhead and ring finger domains; CE-IVD: european conformity–in vitro diagnostic medical device; CLIA: clinical laboratory improvement amendments; DLX1: distal-less homeobox 1; DERL3: derlin 3; ER+: estrogen receptor-positive; FFPE: formalin-fixed paraffin embedded; FAM19A4: family with sequence similarity 19 (chemokine (C–C motif)-like) member A4 (also known as TAFA4); FOXP1: forkhead box P1; FDA: US food and drug administration; 5-FU: 5-fluorouracil; GBM: glioblastomas; GC: gastric cancer; GSTP1: glutathione S-transferase pi 1; HCC: hepatocellular carcinoma; HPV-HNC: HPV-driven HNC; HRBC: high-risk breast cancer; HPV+: human papillomavirus positive; HER2-: human epidermal growth factor receptor 2-negative; HM27/HM450K array: humanmethylation27/humanmethylation 450 beadchip array; HNSCC: head and neck squamous cell carcinoma; ITGA4: integrin subunit alpha 4; IRAK3: interleukin 1 receptor associated kinase 3; IKZF1: IKAROS family zinc finger 1; ICC: invasive cervical cancer; LC: lung cancer; LN+: lymph node-positive; LINE-1: long interspersed nuclear element 1; mCRC: metastatic CRC; MGMT: O6-methylguanine–DNA methyltransferase; NSCLC: non-small-cell lung cancer; NMIBC: Non-muscle-invasive bladder cancer; NDRG4: NDRG family member 4; NIBC: noninvasive bladder cancer; OC: ovarian cancer; ONECUT2: one cut homeobox 2; OTX1: orthodenticle homeobox 1; PCa: prostate cancer; PC: pancreatic cancer; PB: peripheral blood; PTGER4: prostaglandin E receptor 4; PITX2: paired like homeodomain 2; PD-1: programmed cell death protein 1 (also known as PDCD1); RXFP3: relaxin family peptide receptor 3; RASSF1: ras association domain family member 1; PARP: poly (adenosine diphosphate) ribose polymerase; SCLC: small cell lung cancer; SEPTIN9: septin 9; SOX17: SRY-box transcription factor 17; SHOX2: short stature homeobox 2; SRBC: also known as CD2; SLFN11: schlafen 11; SOX1: SRY-box transcription factor 1; TWIST1: twist family BHLH transcription factor 1; TFAP2E: transcription factor AP-2 epsilon; TP53TG1: TP53 target 1; TOO: tissue of origin; VIM: vimentin WB: whole blood; ZNF671: zinc finger protein 671.
5. The DNA methylation machinery in the perspective of pharmaceutic approaches
The development of technologies, including the Infinium humanmethylation450K (HM450K), 850k EPIC array, whole-genome bisulfite sequencing (WGBS), methylated DNA immunoprecipitation (MeDIP), and chromatin immunoprecipitation sequencing (ChIP-Seq) all together opened the eyes of researchers to the value of DNA methylation alterations in the better understanding of diseases with different types and origins. These molecular efforts soon translated into pharmaceutical attempts to develop several DNA methylation inhibitors for treating various diseases, especially cancers [266]. However, most of these efforts failed to move into the clinics due to the high toxicity, cell cycle dependency, and need for continuous administration [266,267]. Moreover, most of these agents cause random DNA hypomethylation in genes with devastating outcomes in some cases [268]. There are always some expectations, such as what happened for two nucleoside DNMT inhibitors named 5-AC and decitabine (DAC), which gained European Union approval for the treatment of newly diagnosed or secondary AML who are ineligible for transplantation [266]. Of course, it should not be forgotten that neither 5-AC nor DAC exerts favorable anti-cancer effects on solid tumors due to the slower pace of cell cycle progression [269]. Nevertheless, efforts are ongoing to identify suitable non-nucleoside DNMT inhibitors to overcome these limitations. The promising results of a recent study have introduced a first-in-class, potent, non-nucleoside, reversible, selective inhibitor of DNMT1, GSK3685032, with improved tolerability and efficacy in AML models. Hence, due to the higher in vivo tolerability compared with DAC, GSK3685032 induces robust loss of DNA methylation associated with transcriptional activation, resulting in tumor regression and improved survival in mouse models of AML [270].
Other DNA methylation-based agents that have received FDA approval are enasidenib (AG-221) and ivosidenib (AG-120), which target IDH2 and IDH1, respectively [271,272]. Both agents can significantly prevent the production of 2-Hydroxyglutarate (2-HG) from mutant IDH1/IDH2 and thereby reactivate TET2 in malignant cells [271,272]. But, despite efficacy, the lower incidence of IDH1/IDH2 mutations in human cancers practically reduces the usefulness of the agents. Glioblastoma is the only cancer with a higher rate of IDH1/IDH2 mutations; however, in this type of cancer, the blood-brain barrier (BBB) prevents the penetration of enasidenib and ivosidenib into the malignant cells. Recently, the results of phase 1 clinical study declared the success of Vorasidenib (AGG-881), a dual IDH1/IDH2 inhibitor, in penetrating BBB of glioma patients, suggesting perhaps this agent may bring a ray of hope for glioma patients with IDH1/IDH2 mutations [273]. Table 6 provides further information about drugs that target DNA methylation.
Table 6.
DNA methylation-targeting drugs that are approved or in clinical trials.
| Drug | Diseases | Phase | Clinical intervention | Ref/NCT |
|---|---|---|---|---|
| DNA methyltransferase (DNMT) inhibitors | ||||
| DAC | AML and MDS | EMA and FDA approval | There is a significant improvement in survival or PFS compared to supportive care or conventional therapy in AML and MDS. | [266] |
| Solid tumors | Phase I/II | Although single-agent efficacy in solid tumors was not convincing, combination therapy has resulted in promising responses. | [274,275] | |
| 5-AC | AML, CMML, MDS | EMA and FDA approval | There is a significant improvement in survival or PFS compared to supportive care or conventional therapy in AML, CMML, and MDS. | [266] |
| Solid tumors | Phase I/II | Although single-agent efficacy in solid tumors was not convincing, combination therapy has resulted in promising responses. | [274,276] | |
| ASTX727 | CMML and MDS | Phase I/II | The safety and efficacy of ASTX727 were consistent with those from IV DAC in CMML and MDS patients. | [277] |
| Patients with MPNST | Phase II | 25 patients were enrolled to evaluate whether ASTX727 could be an effective treatment for MPNST patients with a PRC2 mutation. | NCT04872543 | |
| SCCHN | Phase I/II | 25 patients were enrolled to evaluate the safety and efficacy of oral ASTX727 and MEDI4736 combination in recurrent and/or metastatic SCCHN who have progressed during or after Immunotherapy. | NCT03019003 | |
| CC-486 | AML, MDS | Phase I/II | CC-486 maintenance therapy after alloHSCT was correlated with a relatively low rate of relapse (21 %) during treatment. | [278] |
| AML | Phase III | 360 patients were enrolled to evaluate the safety and efficacy of oral ABT-199 and oral or injectable 5-AC vs best supportive care as maintenance therapy in patients with AML. | NCT04102020 | |
| R/R iBCL | Phase I | 24 patients were enrolled to evaluate the side effects of CC-486 and how well it works in combination with GA-101 and CC-5013 in R/R iBCL. | NCT04578600 | |
| 75 years or older patients with newly diagnosed DLBCL | Phase III | 422 patients were enrolled to evaluate the side effects and activity of CC-486 in combination with the standard drug therapy (R-miniCHOP) vs R-miniCHOP alone in patients 75 years or older with newly diagnosed DLBCL. | NCT04799275 | |
| Advanced microsatellite stable CRC; Platinum-resistant OC; ER+/HER2- BrC | Phase II | The combination therapy of CC-486 and MEDI4736 did not show robust PD or clinical activity in advanced solid tumors. | [279] NCT02811497 | |
| Patients with mM | Phase I | 71 patients were enrolled to evaluate the efficacy and safety of CC-486 in combination with MK-3475 in patients with melanoma. | NCT02816021 | |
| SGI-110 | AML | Phase III | 302 participants were enrolled to evaluate the safety and efficacy of SGI-110 vs choice treatment in adults with previously treated AML. | NCT02920008 |
| mCRC | Phase I/II | The study is investigating if SGI-110 can reverse chemoresistance to IRI in 18 mCRC patients that have developed resistance to IRI. | NCT01896856 | |
| mCRC | Phase I | SGI-110 + GVAX was tolerable but demonstrated no significant clinical activity in mCRC patients | [280] | |
| Platinum-resistant, recurrent OC | Phase II | Although this trial did not demonstrate superiority for PFS of SGI-110 and carboplatin (G + C) versus treatment of choice, the 6-month PFS increased in G + C treated patients. | [281] NCT01696032 | |
| R/R GCT | Phase I | 14 patients were enrolled to evaluate the efficacy and safety of SGI-110 in combination with cisplatin in subjects with R/R GCT. | NCT02429466 | |
| Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) inhibitors | ||||
| AG-221 | R/R AML patients with IDH2 mutation | FDA approval | There is a significant increase in ORR (40 %) and CRR (19 %) in R/R AML patients with an IDH2 mutation. | [271] |
| AG-120 | R/R AML with IDH1-mutated | FDA approval | There is a significant increase in ORR (41.6) and CRR (21.6 %) in R/R AML patients with IDH1 mutation | [272] |
| AGG-881 | LGG with IDH1 or IDH2 mutation | Phase I | Vorasidenib showed good brain penetrance and a decrease in 2-HG levels by approximately 93 % in LGG patients. | [273] |
| R/R Grade 2 glioma with IDH1 or IDH2 mutation. | Phase III | 340 patients were enrolled to evaluate the efficacy of AGG-881 compared to placebo in patients with residual or relapsed Grade 2 glioma with IDH1 or IDH2 mutation. | NCT04164901 | |
| FT-2102 | AML or MDS with an IDH1 mutation | Phase I/II | 500 patients were enrolled to evaluate the safety, efficacy, PK, and PD of FT-2102 as a single agent or in combination with 5-AC or Ara-C. | NCT02719574 |
| LY3410738 | AML, CMML, MDS, and MPNs patients with IDH1 or IDH2 mutation | Phase I | 220 patients were enrolled to evaluate the safety and clinical activity of oral LY3410738 in patients with advanced HMs with IDH1 or IDH2 mutations. | NCT04603001 |
| LY3410738 | Advanced solid tumors with IDH1 mutations | Phase I | 180 patients were enrolled to evaluate the safety and efficacy of oral LY3410738 in advanced solid tumors with IDH1 R132-mutation. | NCT04521686 |
| PEPIDH1M | Recurrent Grade II Glioma with IDH1 mutation | Phase I | 24 patients were enrolled to evaluate the safety of the PEPIDH1M vaccine in combination with standard chemotherapy (TMZ). | NCT02193347 |
DAC (decitabine; also known as 5-aza-2-deoxycytidine, dacogen; is a DNMT inhibitor); 5-AC (azacitidine; also known as vidaza, azadine, onureg; is a DNMT inhibitor); ASTX727 (oral cedazuridine/decitabine; is an oral DNMT inhibitor); CC-486 (oral azacitidine; is an oral DNMT inhibitor); SGI-110 (guadecitabine; is an DNMT inhibitor); AG-221 (enasidenib; is an IDH2 inhibitor); AG-120 (ivosidenib; is an IDH1 inhibitor); AGG-881 (vorasidenib; is a dual IDH1/IDH2 inhibitor); FT-2102 (olutasidenib; is an oral IDH1 inhibitor); LY3410738 (is an oral IDH1/IDH2 inhibitor); PEPIDH1M (is an IDH1 peptide vaccine); MEDI4736 (durvalumab; is a check point inhibitor); ABT-199 (venetoclax; is a B-cell lymphoma 2 (BCL2) inhibitor); GA-101 (obinutuzumab; is an anti-CD20 monoclonal antibody R7159); CC-5013 (lenalidomide; is a TNF-α secretion inhibitor); MK-3475 (pembrolizumab; is an anti–PD-1 monoclonal antibody); IRI (irinotecan, is a topoisomerase I inhibitor); GVAX (is an allogeneic colon cancer cell vaccine); Carboplatin (is an alkylating antineoplastic agent); Cisplatin (is a an cytotoxic chemotherapy drug); Ara-C (cytosine arabinoside; also known as cytarabine; is a an cytotoxic chemotherapy drug): TMZ (temozolomide; is an alkylating antineoplastic agent); AML: acute myeloid leukemia; Allo-HSCT: allogeneic hematopoietic stem cell transplantation; BrC: breast cancer; CMML: chronic myelomonocytic leukemia; CRR: complete remission rate; DLBCL: diffuse large B-cell lymphoma; ER+: estrogen receptor positive; EMA: european medicines agency; FDA: US food and drug administration; HER2-: human epidermal growth factor receptor 2-negative; 2-HG: 2-hydroxyglutarate; HMs: hematologic malignancies; IDH: isocitrate dehydrogenase; IV: intravenous; LGG: low grade glioma MDS: myelodysplastic syndrome; MPNST: malignant peripheral nerve sheath tumors; mM: metastatic melanoma; mCRC: metastatic colorectal cancer; MPNs: myeloproliferative neoplasms; OC: ovarian cancer; ORR: overall response rate; PFS: progression-free survival; PRC2: polycomb repressive complex 2; PD: pharmacodynamic; PK: pharmacokinetic; R/R iBCL: relapsed/refractory indolent B-Cell lymphoma; R/R GCT: relapsed/refractory germ cell tumor; R/R AML: relapsed or refractory AML; RR: relapsed or refractory; R-miniCHOP: reduced dose rituximab-cyclophosphamide, doxorubicin, vincristine, and prednisone; SCCHN: squamous cell carcinoma of the head and neck.
6. Conclusion and future perspectives
Despite numerous studies trying to shed light on the importance of DNA methylation machinery in the development of several diseases, the translation of this knowledge into a clinical perspective is in a nascent stage. It may be due to the complexity of DNA methylation alterations that make it impossible to find out whether these alterations are a cause or consequence of disease [76,134,282]. A non-causal DNA methylation marker could be useable for diagnosis, prediction of treatment response, and monitoring residual disease, but it would not be useable as a potential therapeutic target for mitigating the disease phenotype. Another problem in studying DNA methylation alterations is the tissue- or cell-specific feature of DNA methylation and that it changes over time. Furthermore, most cell populations contain heterogeneous cellular compositions that have different methylation patterns. This challenge is most important in diseases such as cancer, which have a mixed population of normal and tumor cells, or in immune diseases, where the composition of blood cells varies widely among patients. To address this issue, two strategies have been developed to decode DNA methylation patterns according to the cell origin. While one of these strategies can resolute DNA modifications (5 mC or 5hmC) in the single-cell scale [283], the other can generalize these modifications to variable cellular composition by comparison with a reference epigenome or by using reference-free algorithms [284].
Despite all the mentioned challenges, increasing reports reveal a strong association between DNA methylation profiles and clinical outcomes in various diseases, making DNA methylation profiles valuable as biomarkers for diagnosis, prognosis, or response to therapy. Detection of DNA methylation changes in non-invasive liquid biopsy samples, such as blood cells, plasma cell-free DNA, saliva, stool, or urine, is potentially used as a useful diagnostic and prognostic biomarker in clinical practice. However, the sensitivity and reproducibility of liquid biopsy-based techniques still need to be improved, especially when working with samples with suboptimal DNA quality or low cell numbers.
Nevertheless, DNA methylation alterations are well-recognized drivers that initiate and prolong human disease. Fortunately, the fact that DNA methylation alterations are reversible converts them into valuable therapeutic targets. Current FDA-approved DNA methylation-based drugs are used to treat cancer alone or in combination with other therapies, but this approach appears to affect DNA methylation signatures at many genomic sites indiscriminately. The CRISPR technology can direct the enzymatic DNA methylation machinery to specific sites in the genome, making characteristic changes in the DNA methylation pattern to control gene expression. CRISPR, sequencing, and cloning technologies can all come to give value to DNA methylation alterations in the new era of molecular biology. In conclusion, the study of DNA methylation alterations is a phenomenal intersection that each of its ways can reveal the mysteries of various diseases, introduce new diagnostic and prognostic biomarkers, and propose a new patient-tailored therapeutic approach for diseases.
Funding statement
The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.
Data availability statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Ethics statement
Not applicable.
CRediT authorship contribution statement
Samareh Younesian: Writing – review & editing, Writing – original draft, Investigation, Conceptualization. Mohammad Hossein Mohammadi: Writing – review & editing. Ommolbanin Younesian: Writing – review & editing, Writing – original draft. Majid Momeny: Writing – review & editing. Seyed H. Ghaffari: Writing – review & editing. Davood Bashash: Writing – review & editing, Supervision, Project administration, Conceptualization.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgments
The authors would like to express their gratitude to Shahid Beheshti University of Medical Sciences (Tehran, Iran) for supporting this study.
Abbreviations
- 2-HG
2-Hydroxyglutarate
- 5-AC
5-azacitidine
- 5-carboxyl
5-caC 5-carboxyl
- 5-fc
5-formyl
- 5hmC
5-hydroxymethylcytosine
- 5 mC
5-methylcytosine
- ADAC-DN
Autosomal dominant cerebellar ataxia with deafness and narcolepsy
- ADD
ATRX- DNMT3- DNMT3L
- ALL
Acute lymphoblastic leukemia
- AML
Acute myeloid leukemia
- ARTs
Assisted reproductive technologies
- AS
Angelman syndrome
- Aβ
Amyloid beta
- BAH
Bromo adjacent homology
- BBB
Blood-brain barrier
- BER
Base excision repair
- BMI
Body mass index
- BWS
Beckwith–Wiedemann syndrome
- ChIP-Seq
Chromatin immunoprecipitation sequencing
- CML
Chronic myeloid leukemia
- c-NHEJ
Classical non-homologous end joining
- CREB1
cAMP-responsive element-binding protein 1
- CSR
Class switch-recombination
- CUP
Carcinoma of unknown primary
- DAC
Decitabine
- dcSSc
Diffuse cutaneous SSc
- DNMTs
DNA methyltransferases
- GoF
Gain-of-function
- H3K4
Histone H3 lysine 4
- HDACs
Histone deacetylases
- HEK
Human embryonic kidney
- HM450K
Infinium humanmethylation450K
- HSCs
Hematopoietic stem cells
- HSNA1E
Hereditary sensory autonomic neuropathy 1E
- ICF
Immunodeficiency, centromeric instability, and facial anomalies syndrome
- ICRs
Imprinting control regions
- IDHs
Isocitrate dehydrogenases
- IGF2
Insulin-like growth factor 2
- lcSSc
Limited cutaneous SSc
- LoF
Loss-of-function
- MBD
Methyl-CpG-binding domain
- MDS
Myelodysplastic syndrome
- MeDIP
Methylated DNA immunoprecipitation
- MEFs
Mouse embryonic fibroblasts
- mESCs
Mouse embryonic stem cells
- MPNs
Myeloproliferative neoplasms
- MS
Multiple sclerosis
- MTase
Highly conserved DNMT
- MRD
Minimal residual disease
- PB
Peripheral blood
- PBMCs
Peripheral blood mononuclear cells
- PHD
Plant homeodomain
- PTMs
Post-translation modifications
- PWS
Prader–Willi syndrome
- PWWP
Pro-Trip-Trip-Pro
- RA
Rheumatoid arthritis
- RFTS
Replication foci targeting sequence
- SAH
S-adenosyl homocysteine
- SAM
S-adenosyl methionine
- SAT
Subcutaneous adipose tissue
- SLE
Systemic lupus erythematosus
- SNP
Single-nucleotide polymorphism
- SNRPN
Small nuclear ribonucleoprotein polypeptide N
- SRA
SET- and RING-associated
- SRS
Silver–Russell syndrome
- SSc
Systemic sclerosis
- T2D
Type 2 diabetic
- TCGA
The Cancer Genome Atlas
- TDG
Thymine glycosylase
- TET
Ten-eleven translocation
- TFs
Transcription factors
- Th2
T helper 2
- TRD
Transcriptional repression domain
- tRNAs
Transfer RNAs
- TTD
Tandem Tudor domain
- UBL
Ubiquitin-like
- UPD
Uniparental disomy
- WGBS
Whole-genome bisulfite sequencing
- ZnF
Zinc-finger
- α-KG
Alpha-ketoglutarate
References
- 1.Dennis C. Epigenetics and disease: altered states. Nature. 2003;421(6924):686–688. doi: 10.1038/421686a. [DOI] [PubMed] [Google Scholar]
- 2.Kaminsky Z.A., et al. DNA methylation profiles in monozygotic and dizygotic twins. Nat. Genet. 2009;41(2):240–245. doi: 10.1038/ng.286. [DOI] [PubMed] [Google Scholar]
- 3.Jaenisch R., Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat. Genet. 2003;33(3):245–254. doi: 10.1038/ng1089. [DOI] [PubMed] [Google Scholar]
- 4.Meaney M.J. Epigenetics and the biological definition of gene× environment interactions. Child Dev. 2010;81(1):41–79. doi: 10.1111/j.1467-8624.2009.01381.x. [DOI] [PubMed] [Google Scholar]
- 5.Esteller M. Epigenetics in evolution and disease. Lancet. 2008;372:S90–S96. [Google Scholar]
- 6.Holliday R., Pugh J.E. DNA modification mechanisms and gene activity during development. Science. 1975;187(4173):226–232. [PubMed] [Google Scholar]
- 7.Jones P.A., Takai D. The role of DNA methylation in mammalian epigenetics. Science. 2001;293(5532):1068–1070. doi: 10.1126/science.1063852. [DOI] [PubMed] [Google Scholar]
- 8.Straussman R., et al. Developmental programming of CpG island methylation profiles in the human genome. Nat. Struct. Mol. Biol. 2009;16(5):564. doi: 10.1038/nsmb.1594. [DOI] [PubMed] [Google Scholar]
- 9.Unnikrishnan A., et al. The role of DNA methylation in epigenetics of aging. Pharmacology & therapeutics. 2019;195:172–185. doi: 10.1016/j.pharmthera.2018.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Moore L.D., Le T., Fan G. DNA methylation and its basic function. Neuropsychopharmacology. 2013;38(1):23–38. doi: 10.1038/npp.2012.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Okano M., et al. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99(3):247–257. doi: 10.1016/s0092-8674(00)81656-6. [DOI] [PubMed] [Google Scholar]
- 12.Arita K., et al. Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature. 2008;455(7214):818–821. doi: 10.1038/nature07249. [DOI] [PubMed] [Google Scholar]
- 13.Jia D., et al. Structure of Dnmt3a bound to Dnmt3L suggests a model for de novo DNA methylation. Nature. 2007;449(7159):248–251. doi: 10.1038/nature06146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bohnsack K.E., Höbartner C., Bohnsack M.T. Eukaryotic 5-methylcytosine (m5C) RNA methyltransferases: mechanisms, cellular functions, and links to disease. Genes. 2019;10(2):102. doi: 10.3390/genes10020102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ballestar E., Wolffe A.P. Methyl‐CpG‐binding proteins: targeting specific gene repression. Eur. J. Biochem. 2001;268(1):1–6. doi: 10.1046/j.1432-1327.2001.01869.x. [DOI] [PubMed] [Google Scholar]
- 16.Filion G.J., et al. A family of human zinc finger proteins that bind methylated DNA and repress transcription. Molecular and cellular biology. 2006;26(1):169–181. doi: 10.1128/MCB.26.1.169-181.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Unoki M., Nishidate T., Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23(46):7601–7610. doi: 10.1038/sj.onc.1208053. [DOI] [PubMed] [Google Scholar]
- 18.Roloff T.C., Ropers H.H., Nuber U.A. Comparative study of methyl-CpG-binding domain proteins. BMC Genom. 2003;4(1):1–9. doi: 10.1186/1471-2164-4-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Quenneville S., 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(3):361–372. doi: 10.1016/j.molcel.2011.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Avvakumov G.V., et al. Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1. Nature. 2008;455(7214):822–825. doi: 10.1038/nature07273. [DOI] [PubMed] [Google Scholar]
- 21.Lorsbach R., et al. TET1, a member of a novel protein family, is fused to MLL in acute myeloid leukemia containing the t (10; 11)(q22; q23) Leukemia. 2003;17(3):637–641. doi: 10.1038/sj.leu.2402834. [DOI] [PubMed] [Google Scholar]
- 22.Tahiliani M., et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science. 2009;324(5929):930–935. doi: 10.1126/science.1170116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhang H., et al. TET1 is a DNA-binding protein that modulates DNA methylation and gene transcription via hydroxylation of 5-methylcytosine. Cell Res. 2010;20(12):1390–1393. doi: 10.1038/cr.2010.156. [DOI] [PubMed] [Google Scholar]
- 24.Kohli R.M., Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature. 2013;502(7472):472–479. doi: 10.1038/nature12750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yamaguchi S., et al. Role of Tet1 in erasure of genomic imprinting. Nature. 2013;504(7480):460–464. doi: 10.1038/nature12805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Solary E., et al. The Ten-Eleven Translocation-2 (TET2) gene in hematopoiesis and hematopoietic diseases. Leukemia. 2014;28(3):485–496. doi: 10.1038/leu.2013.337. [DOI] [PubMed] [Google Scholar]
- 27.Gu T.-P., et al. The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature. 2011;477(7366):606–610. doi: 10.1038/nature10443. [DOI] [PubMed] [Google Scholar]
- 28.Barlow D.P., Bartolomei M.S. Genomic imprinting in mammals. Cold Spring Harbor Perspect. Biol. 2014;6(2):a018382. doi: 10.1101/cshperspect.a018382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tucci V., et al. Genomic imprinting and physiological processes in mammals. Cell. 2019;176(5):952–965. doi: 10.1016/j.cell.2019.01.043. [DOI] [PubMed] [Google Scholar]
- 30.Elhamamsy A.R. Role of DNA methylation in imprinting disorders: an updated review. J. Assist. Reprod. Genet. 2017;34(5):549–562. doi: 10.1007/s10815-017-0895-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gicquel C., et al. Epimutation of the telomeric imprinting center region on chromosome 11p15 in Silver-Russell syndrome. Nat. Genet. 2005;37(9):1003–1007. doi: 10.1038/ng1629. [DOI] [PubMed] [Google Scholar]
- 32.Sparago A., et al. Microdeletions in the human H19 DMR result in loss of IGF2 imprinting and Beckwith-Wiedemann syndrome. Nat. Genet. 2004;36(9):958–960. doi: 10.1038/ng1410. [DOI] [PubMed] [Google Scholar]
- 33.Gaston V., et al. Analysis of the methylation status of the KCNQ1OT and H19 genes in leukocyte DNA for the diagnosis and prognosis of Beckwith–Wiedemann syndrome. European journal of human genetics. 2001;9(6):409–418. doi: 10.1038/sj.ejhg.5200649. [DOI] [PubMed] [Google Scholar]
- 34.Ledbetter D.H., et al. Deletions of chromosome 15 as a cause of the Prader–Willi syndrome. N. Engl. J. Med. 1981;304(6):325–329. doi: 10.1056/NEJM198102053040604. [DOI] [PubMed] [Google Scholar]
- 35.Magenis R.E., et al. Is angelman syndrome an alternate result of del (15)(qllql3)? Am. J. Med. Genet. 1987;28(4):829–838. doi: 10.1002/ajmg.1320280407. [DOI] [PubMed] [Google Scholar]
- 36.Runte M., et al. The IC-SNURF–SNRPN transcript serves as a host for multiple small nucleolar RNA species and as an antisense RNA for UBE3A. Hum. Mol. Genet. 2001;10(23):2687–2700. doi: 10.1093/hmg/10.23.2687. [DOI] [PubMed] [Google Scholar]
- 37.Buiting K., et al. Clinical utility gene card for: angelman Syndrome. Eur. J. Hum. Genet. 2015;23(2) doi: 10.1038/ejhg.2014.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cassidy S.B., et al. Prader-Willi syndrome. Genet. Med. 2012;14(1):10–26. doi: 10.1038/gim.0b013e31822bead0. [DOI] [PubMed] [Google Scholar]
- 39.Canovas S., et al. DNA methylation in embryo development: epigenetic impact of ART (assisted reproductive technologies) Bioessays. 2017;39(11) doi: 10.1002/bies.201700106. [DOI] [PubMed] [Google Scholar]
- 40.Jeanpierre M., et al. An embryonic-like methylation pattern of classical satellite DNA is observed in ICF syndrome. Hum. Mol. Genet. 1993;2(6):731–735. doi: 10.1093/hmg/2.6.731. [DOI] [PubMed] [Google Scholar]
- 41.Xu G.-L., et al. Chromosome instability and immunodeficiency syndrome caused by mutations in a DNA methyltransferase gene. Nature. 1999;402(6758):187–191. doi: 10.1038/46052. [DOI] [PubMed] [Google Scholar]
- 42.De Greef J.C., et al. Mutations in ZBTB24 are associated with immunodeficiency, centromeric instability, and facial anomalies syndrome type 2. Am. J. Hum. Genet. 2011;88(6):796–804. doi: 10.1016/j.ajhg.2011.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Thijssen P.E., et al. Mutations in CDCA7 and HELLS cause immunodeficiency–centromeric instability–facial anomalies syndrome. Nat. Commun. 2015;6(1):1–8. doi: 10.1038/ncomms8870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hagleitner M.M., et al. Clinical spectrum of immunodeficiency, centromeric instability and facial dysmorphism (ICF syndrome) J. Med. Genet. 2008;45(2):93–99. doi: 10.1136/jmg.2007.053397. [DOI] [PubMed] [Google Scholar]
- 45.Jiang Y., et al. DNMT3B mutations and DNA methylation defect define two types of ICF syndrome. Hum. Mutat. 2005;25(1):56–63. doi: 10.1002/humu.20113. [DOI] [PubMed] [Google Scholar]
- 46.Toubiana S., et al. Subtelomeric methylation distinguishes between subtypes of Immunodeficiency, Centromeric instability and Facial anomalies syndrome. Hum. Mol. Genet. 2018;27(20):3568–3581. doi: 10.1093/hmg/ddy265. [DOI] [PubMed] [Google Scholar]
- 47.Velasco G., et al. Germline genes hypomethylation and expression define a molecular signature in peripheral blood of ICF patients: implications for diagnosis and etiology. Orphanet J. Rare Dis. 2014;9:56. doi: 10.1186/1750-1172-9-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hansen R.S., et al. Escape from gene silencing in ICF syndrome: evidence for advanced replication time as a major determinant. Hum. Mol. Genet. 2000;9(18):2575–2587. doi: 10.1093/hmg/9.18.2575. [DOI] [PubMed] [Google Scholar]
- 49.Velasco G., et al. Comparative methylome analysis of ICF patients identifies heterochromatin loci that require ZBTB24, CDCA7 and HELLS for their methylated state. Hum. Mol. Genet. 2018;27(14):2409–2424. doi: 10.1093/hmg/ddy130. [DOI] [PubMed] [Google Scholar]
- 50.Unoki M., et al. CDCA7 and HELLS mutations undermine nonhomologous end joining in centromeric instability syndrome. J. Clin. Invest. 2019;129(1):78–92. doi: 10.1172/JCI99751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wu H., et al. Converging disease genes in ICF syndrome: ZBTB24 controls expression of CDCA7 in mammals. Hum. Mol. Genet. 2016;25(18):4041–4051. doi: 10.1093/hmg/ddw243. [DOI] [PubMed] [Google Scholar]
- 52.Hardikar S., et al. The ZBTB24-CDCA7 axis regulates HELLS enrichment at centromeric satellite repeats to facilitate DNA methylation. Protein & Cell. 2020;11(3):214–218. doi: 10.1007/s13238-019-00682-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jenness C., et al. HELLS and CDCA7 comprise a bipartite nucleosome remodeling complex defective in ICF syndrome. Proc Natl Acad Sci U S A. 2018;115(5):E876–e885. doi: 10.1073/pnas.1717509115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Casellas R., et al. Ku80 is required for immunoglobulin isotype switching. Embo j. 1998;17(8):2404–2411. doi: 10.1093/emboj/17.8.2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Björkman A., et al. DNA-PKcs is involved in ig class switch recombination in human B cells. J. Immunol. 2015;195(12):5608–5615. doi: 10.4049/jimmunol.1501633. [DOI] [PubMed] [Google Scholar]
- 56.Blanco-Betancourt C.E., et al. Defective B-cell-negative selection and terminal differentiation in the ICF syndrome. Blood. 2004;103(7):2683–2690. doi: 10.1182/blood-2003-08-2632. [DOI] [PubMed] [Google Scholar]
- 57.Ueda Y., et al. Roles for Dnmt3b in mammalian development: a mouse model for the ICF syndrome. Development. 2006;133(6):1183–1192. doi: 10.1242/dev.02293. [DOI] [PubMed] [Google Scholar]
- 58.Rajshekar S., et al. Pericentromeric hypomethylation elicits an interferon response in an animal model of ICF syndrome. Elife. 2018;7 doi: 10.7554/eLife.39658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Klein C.J., et al. Mutations in DNMT1 cause hereditary sensory neuropathy with dementia and hearing loss. Nat. Genet. 2011;43(6):595–600. doi: 10.1038/ng.830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Winkelmann J., et al. Mutations in DNMT1 cause autosomal dominant cerebellar ataxia, deafness and narcolepsy. Hum. Mol. Genet. 2012;21(10):2205–2210. doi: 10.1093/hmg/dds035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Baets J., et al. Defects of mutant DNMT1 are linked to a spectrum of neurological disorders. Brain. 2015;138(4):845–861. doi: 10.1093/brain/awv010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Tatton-Brown K., et al. Mutations in the DNA methyltransferase gene DNMT3A cause an overgrowth syndrome with intellectual disability. Nat. Genet. 2014;46(4):385–388. doi: 10.1038/ng.2917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Heyn P., et al. Gain-of-function DNMT3A mutations cause microcephalic dwarfism and hypermethylation of Polycomb-regulated regions. Nat. Genet. 2019;51(1):96–105. doi: 10.1038/s41588-018-0274-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Binder G., et al. Novel mutation points to a hot spot in CDKN1C causing Silver–Russell syndrome. Clin. Epigenet. 2020;12(1):1–7. doi: 10.1186/s13148-020-00945-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Eggermann T., et al. Clinical utility gene card for: beckwith–Wiedemann syndrome. Eur. J. Hum. Genet. 2014;22(3):435. doi: 10.1038/ejhg.2013.132. 435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Younesian S., et al. The DNA methylation in neurological diseases. Cells. 2022;11(21) doi: 10.3390/cells11213439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ramocki M.B., Tavyev Y.J., Peters S.U. The MECP2 duplication syndrome. Am. J. Med. Genet. 2010;152A(5):1079–1088. doi: 10.1002/ajmg.a.33184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Beck D.B., et al. Delineation of a human mendelian disorder of the DNA demethylation machinery: TET3 deficiency. Am. J. Hum. Genet. 2020;106(2):234–245. doi: 10.1016/j.ajhg.2019.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gorelik G., et al. Impaired T cell protein kinase C delta activation decreases ERK pathway signaling in idiopathic and hydralazine-induced lupus. J. Immunol. 2007;179(8):5553–5563. doi: 10.4049/jimmunol.179.8.5553. [DOI] [PubMed] [Google Scholar]
- 70.Coit P., et al. Epigenetic reprogramming in naive CD4+ T cells favoring T cell activation and non-Th1 effector T cell immune response as an early event in lupus flares. Arthritis Rheumatol. 2016;68(9):2200–2209. doi: 10.1002/art.39720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Quddus J., et al. Treating activated CD4+ T cells with either of two distinct DNA methyltransferase inhibitors, 5-azacytidine or procainamide, is sufficient to cause a lupus-like disease in syngeneic mice. J. Clin. Invest. 1993;92(1):38–53. doi: 10.1172/JCI116576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhao M., et al. IFI44L promoter methylation as a blood biomarker for systemic lupus erythematosus. Ann. Rheum. Dis. 2016;75(11):1998–2006. doi: 10.1136/annrheumdis-2015-208410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhao M., et al. Increased 5-hydroxymethylcytosine in CD4(+) T cells in systemic lupus erythematosus. J. Autoimmun. 2016;69:64–73. doi: 10.1016/j.jaut.2016.03.001. [DOI] [PubMed] [Google Scholar]
- 74.Hewagama A., et al. Overexpression of X-linked genes in T cells from women with lupus. J. Autoimmun. 2013;41:60–71. doi: 10.1016/j.jaut.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.McInnes I.B., Schett G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011;365(23):2205–2219. doi: 10.1056/NEJMra1004965. [DOI] [PubMed] [Google Scholar]
- 76.Liu Y., et al. Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat. Biotechnol. 2013;31(2):142–147. doi: 10.1038/nbt.2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liao J., et al. CD40L demethylation in CD4(+) T cells from women with rheumatoid arthritis. Clin Immunol. 2012;145(1):13–18. doi: 10.1016/j.clim.2012.07.006. [DOI] [PubMed] [Google Scholar]
- 78.Kennedy A., et al. A novel upstream enhancer of FOXP3, sensitive to methylation-induced silencing, exhibits dysregulated methylation in rheumatoid arthritis Treg cells. Eur. J. Immunol. 2014;44(10):2968–2978. doi: 10.1002/eji.201444453. [DOI] [PubMed] [Google Scholar]
- 79.Ngalamika O., et al. Peripheral whole blood FOXP3 TSDR methylation: a potential marker in severity assessment of autoimmune diseases and chronic infections. Immunol. Invest. 2015;44(2):126–136. doi: 10.3109/08820139.2014.938165. [DOI] [PubMed] [Google Scholar]
- 80.Cribbs A.P., et al. Treg cell function in rheumatoid arthritis is compromised by ctla-4 promoter methylation resulting in a failure to activate the indoleamine 2,3-dioxygenase pathway. Arthritis Rheumatol. 2014;66(9):2344–2354. doi: 10.1002/art.38715. [DOI] [PubMed] [Google Scholar]
- 81.Gosselt H.R., et al. Higher baseline global leukocyte DNA methylation is associated with MTX non-response in early RA patients. Arthritis Res. Ther. 2019;21(1):157. doi: 10.1186/s13075-019-1936-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dees C., et al. The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis. Ann. Rheum. Dis. 2014;73(6):1232–1239. doi: 10.1136/annrheumdis-2012-203194. [DOI] [PubMed] [Google Scholar]
- 83.Wang Y., Fan P.S., Kahaleh B. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis Rheum. 2006;54(7):2271–2279. doi: 10.1002/art.21948. [DOI] [PubMed] [Google Scholar]
- 84.O'Reilly S., et al. IL-13 mediates collagen deposition via STAT6 and microRNA-135b: a role for epigenetics. Sci. Rep. 2016;6 doi: 10.1038/srep25066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhang Y., et al. Poly(ADP-ribose) polymerase-1 regulates fibroblast activation in systemic sclerosis. Ann. Rheum. Dis. 2018;77(5):744–751. doi: 10.1136/annrheumdis-2017-212265. [DOI] [PubMed] [Google Scholar]
- 86.Henderson J., et al. Methyl cap binding protein 2: a key epigenetic protein in systemic sclerosis. Rheumatology. 2019;58(3):527–535. doi: 10.1093/rheumatology/key327. [DOI] [PubMed] [Google Scholar]
- 87.Lei W., et al. Abnormal DNA methylation in CD4+ T cells from patients with systemic lupus erythematosus, systemic sclerosis, and dermatomyositis. Scand. J. Rheumatol. 2009;38(5):369–374. doi: 10.1080/03009740902758875. [DOI] [PubMed] [Google Scholar]
- 88.Wang Y., et al. Hypomethylation and overexpression of ITGAL (CD11a) in CD4(+) T cells in systemic sclerosis. Clin. Epigenet. 2014;6(1):25. doi: 10.1186/1868-7083-6-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ding W., et al. Genome-wide DNA methylation analysis in systemic sclerosis reveals hypomethylation of IFN-associated genes in CD4(+) and CD8(+) T cells. J. Invest. Dermatol. 2018;138(5):1069–1077. doi: 10.1016/j.jid.2017.12.003. [DOI] [PubMed] [Google Scholar]
- 90.Jiang H., et al. Demethylation of TNFSF7 contributes to CD70 overexpression in CD4+ T cells from patients with systemic sclerosis. Clin Immunol. 2012;143(1):39–44. doi: 10.1016/j.clim.2012.01.005. [DOI] [PubMed] [Google Scholar]
- 91.Lian X., et al. DNA demethylation of CD40l in CD4+ T cells from women with systemic sclerosis: a possible explanation for female susceptibility. Arthritis Rheum. 2012;64(7):2338–2345. doi: 10.1002/art.34376. [DOI] [PubMed] [Google Scholar]
- 92.Ramos P.S., et al. Integrative analysis of DNA methylation in discordant twins unveils distinct architectures of systemic sclerosis subsets. Clin. Epigenet. 2019;11(1):58. doi: 10.1186/s13148-019-0652-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Neven K.Y., et al. Repetitive element hypermethylation in multiple sclerosis patients. BMC Genet. 2016;17(1):84. doi: 10.1186/s12863-016-0395-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ayuso T., et al. Vitamin D receptor gene is epigenetically altered and transcriptionally up-regulated in multiple sclerosis. PLoS One. 2017;12(3) doi: 10.1371/journal.pone.0174726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Field J., et al. Interleukin-2 receptor-α proximal promoter hypomethylation is associated with multiple sclerosis. Gene Immun. 2017;18(2):59–66. doi: 10.1038/gene.2016.50. [DOI] [PubMed] [Google Scholar]
- 96.Kular L., et al. DNA methylation as a mediator of HLA-DRB1* 15: 01 and a protective variant in multiple sclerosis. Nat. Commun. 2018;9(1):1–15. doi: 10.1038/s41467-018-04732-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Calabrese R., et al. TET2 gene expression and 5-hydroxymethylcytosine level in multiple sclerosis peripheral blood cells. Biochim. Biophys. Acta. 2014;1842(7):1130–1136. doi: 10.1016/j.bbadis.2014.04.010. [DOI] [PubMed] [Google Scholar]
- 98.Absher D.M., et al. Genome-wide DNA methylation analysis of systemic lupus erythematosus reveals persistent hypomethylation of interferon genes and compositional changes to CD4+ T-cell populations. PLoS Genet. 2013;9(8) doi: 10.1371/journal.pgen.1003678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ulff‐Møller C.J., et al. Twin DNA methylation profiling reveals flare‐dependent interferon signature and B cell promoter hypermethylation in systemic lupus erythematosus. Arthritis Rheumatol. 2018;70(6):878–890. doi: 10.1002/art.40422. [DOI] [PubMed] [Google Scholar]
- 100.Joseph S., et al. Epigenome-wide association study of peripheral blood mononuclear cells in systemic lupus erythematosus: identifying DNA methylation signatures associated with interferon-related genes based on ethnicity and SLEDAI. J. Autoimmun. 2019;96:147–157. doi: 10.1016/j.jaut.2018.09.007. [DOI] [PubMed] [Google Scholar]
- 101.Coit P., et al. Renal involvement in lupus is characterized by unique DNA methylation changes in naïve CD4+ T cells. J. Autoimmun. 2015;61:29–35. doi: 10.1016/j.jaut.2015.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lin S.Y., et al. A whole genome methylation analysis of systemic lupus erythematosus: hypomethylation of the IL10 and IL1R2 promoters is associated with disease activity. Gene Immun. 2012;13(3):214–220. doi: 10.1038/gene.2011.74. [DOI] [PubMed] [Google Scholar]
- 103.Zhang B., et al. Difference of IFI44L methylation and serum IFN-a1 level among patients with discoid and systemic lupus erythematosus and healthy individuals. J Transl Autoimmun. 2021;4 doi: 10.1016/j.jtauto.2021.100092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Oelke K., et al. Overexpression of CD70 and overstimulation of IgG synthesis by lupus T cells and T cells treated with DNA methylation inhibitors. Arthritis Rheum. 2004;50(6):1850–1860. doi: 10.1002/art.20255. [DOI] [PubMed] [Google Scholar]
- 105.Lu Q., et al. Demethylation of ITGAL (CD11a) regulatory sequences in systemic lupus erythematosus. Arthritis Rheum. 2002;46(5):1282–1291. doi: 10.1002/art.10234. [DOI] [PubMed] [Google Scholar]
- 106.Mok A., et al. Hypomethylation of CYP2E1 and DUSP22 promoters associated with disease activity and erosive disease among rheumatoid arthritis patients. Arthritis Rheumatol. 2018;70(4):528–536. doi: 10.1002/art.40408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Plant D., et al. Differential methylation as a biomarker of response to etanercept in patients with rheumatoid arthritis. Arthritis Rheumatol. 2016;68(6):1353–1360. doi: 10.1002/art.39590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Glossop J.R., et al. DNA methylation at diagnosis is associated with response to disease-modifying drugs in early rheumatoid arthritis. Epigenomics. 2017;9(4):419–428. doi: 10.2217/epi-2016-0042. [DOI] [PubMed] [Google Scholar]
- 109.Nair N., et al. Differential DNA methylation correlates with response to methotrexate in rheumatoid arthritis. Rheumatology. 2020;59(6):1364–1371. doi: 10.1093/rheumatology/kez411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Takami N., et al. Hypermethylated promoter region of DR3, the death receptor 3 gene, in rheumatoid arthritis synovial cells. Arthritis Rheum. 2006;54(3):779–787. doi: 10.1002/art.21637. [DOI] [PubMed] [Google Scholar]
- 111.de la Rica L., et al. Identification of novel markers in rheumatoid arthritis through integrated analysis of DNA methylation and microRNA expression. J. Autoimmun. 2013;41:6–16. doi: 10.1016/j.jaut.2012.12.005. [DOI] [PubMed] [Google Scholar]
- 112.Maeshima K., et al. Abnormal PTPN11 enhancer methylation promotes rheumatoid arthritis fibroblast-like synoviocyte aggressiveness and joint inflammation. JCI Insight. 2016;1(7) doi: 10.1172/jci.insight.86580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Karouzakis E., et al. Analysis of early changes in DNA methylation in synovial fibroblasts of RA patients before diagnosis. Sci. Rep. 2018;8(1):7370. doi: 10.1038/s41598-018-24240-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wang Y.Y., et al. DNA hypermethylation of the forkhead box protein 3 (FOXP3) promoter in CD4+ T cells of patients with systemic sclerosis. Br. J. Dermatol. 2014;171(1):39–47. doi: 10.1111/bjd.12913. [DOI] [PubMed] [Google Scholar]
- 115.Wang Y., Kahaleh B. Epigenetic repression of bone morphogenetic protein receptor II expression in scleroderma. J. Cell Mol. Med. 2013;17(10):1291–1299. doi: 10.1111/jcmm.12105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Evans I.C., et al. Epigenetic regulation of cyclooxygenase-2 by methylation of c8orf4 in pulmonary fibrosis. Clin. Sci. (Lond.) 2016;130(8):575–586. doi: 10.1042/CS20150697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Altorok N., et al. Genome-wide DNA methylation analysis in dermal fibroblasts from patients with diffuse and limited systemic sclerosis reveals common and subset-specific DNA methylation aberrancies. Ann. Rheum. Dis. 2015;74(8):1612–1620. doi: 10.1136/annrheumdis-2014-205303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kumagai C., et al. Increased promoter methylation of the immune regulatory gene SHP-1 in leukocytes of multiple sclerosis subjects. J. Neuroimmunol. 2012;246(1–2):51–57. doi: 10.1016/j.jneuroim.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lehmann-Werman R., et al. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci U S A. 2016;113(13):E1826–E1834. doi: 10.1073/pnas.1519286113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Mastronardi F.G., et al. Peptidyl argininedeiminase 2 CpG island in multiple sclerosis white matter is hypomethylated. J. Neurosci. Res. 2007;85(9):2006–2016. doi: 10.1002/jnr.21329. [DOI] [PubMed] [Google Scholar]
- 121.Huynh J.L., et al. Epigenome-wide differences in pathology-free regions of multiple sclerosis-affected brains. Nat. Neurosci. 2014;17(1):121–130. doi: 10.1038/nn.3588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Chomyk A.M., et al. DNA methylation in demyelinated multiple sclerosis hippocampus. Sci. Rep. 2017;7(1):8696. doi: 10.1038/s41598-017-08623-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Dayeh T., et al. Genome-wide DNA methylation analysis of human pancreatic islets from type 2 diabetic and non-diabetic donors identifies candidate genes that influence insulin secretion. PLoS Genet. 2014;10(3) doi: 10.1371/journal.pgen.1004160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ribel-Madsen R., et al. Genome-wide analysis of DNA methylation differences in muscle and fat from monozygotic twins discordant for type 2 diabetes. PLoS One. 2012;7(12) doi: 10.1371/journal.pone.0051302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Nilsson E., et al. Altered DNA methylation and differential expression of genes influencing metabolism and inflammation in adipose tissue from subjects with type 2 diabetes. Diabetes. 2014;63(9):2962–2976. doi: 10.2337/db13-1459. [DOI] [PubMed] [Google Scholar]
- 126.Nilsson E., et al. Epigenetic alterations in human liver from subjects with type 2 diabetes in parallel with reduced folate levels. J. Clin. Endocrinol. Metab. 2015;100(11):E1491–E1501. doi: 10.1210/jc.2015-3204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Abderrahmani A., et al. Increased hepatic PDGF-AA signaling mediates liver insulin resistance in obesity-associated type 2 diabetes. Diabetes. 2018;67(7):1310–1321. doi: 10.2337/db17-1539. [DOI] [PubMed] [Google Scholar]
- 128.Barrès R., et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metabol. 2012;15(3):405–411. doi: 10.1016/j.cmet.2012.01.001. [DOI] [PubMed] [Google Scholar]
- 129.Soyal S., et al. PGC-1 alpha: a potent transcriptional cofactor involved in the pathogenesis of type 2 diabetes. Diabetologia. 2006;49(7):1477–1488. doi: 10.1007/s00125-006-0268-6. [DOI] [PubMed] [Google Scholar]
- 130.Jacobsen S.C., et al. Effects of short-term high-fat overfeeding on genome-wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia. 2012;55(12):3341–3349. doi: 10.1007/s00125-012-2717-8. [DOI] [PubMed] [Google Scholar]
- 131.Dick K.J., et al. DNA methylation and body-mass index: a genome-wide analysis. Lancet. 2014;383(9933):1990–1998. doi: 10.1016/S0140-6736(13)62674-4. [DOI] [PubMed] [Google Scholar]
- 132.Rönn T., et al. Impact of age, BMI and HbA1c levels on the genome-wide DNA methylation and mRNA expression patterns in human adipose tissue and identification of epigenetic biomarkers in blood. Hum. Mol. Genet. 2015;24(13):3792–3813. doi: 10.1093/hmg/ddv124. [DOI] [PubMed] [Google Scholar]
- 133.Kuehnen P., et al. An Alu element-associated hypermethylation variant of the POMC gene is associated with childhood obesity. PLoS Genet. 2012;8(3) doi: 10.1371/journal.pgen.1002543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wahl S., et al. Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity. Nature. 2017;541(7635):81–86. doi: 10.1038/nature20784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Rönn T., et al. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS Genet. 2013;9(6) doi: 10.1371/journal.pgen.1003572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Dayeh T., et al. DNA methylation of loci within ABCG1 and PHOSPHO1 in blood DNA is associated with future type 2 diabetes risk. Epigenetics. 2016;11(7):482–488. doi: 10.1080/15592294.2016.1178418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chen Y.T., et al. NT5C2 methylation regulatory interplay between DNMT1 and insulin receptor in type 2 diabetes. Sci. Rep. 2020;10(1) doi: 10.1038/s41598-020-71336-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Aslibekyan S., et al. Epigenome-wide study identifies novel methylation loci associated with body mass index and waist circumference. Obesity. 2015;23(7):1493–1501. doi: 10.1002/oby.21111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mendelson M.M., et al. Association of body mass index with DNA methylation and gene expression in blood cells and relations to cardiometabolic disease: a Mendelian randomization approach. PLoS Med. 2017;14(1) doi: 10.1371/journal.pmed.1002215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Wang X., et al. An epigenome-wide study of obesity in African American youth and young adults: novel findings, replication in neutrophils, and relationship with gene expression. Clin. Epigenet. 2018;10:3. doi: 10.1186/s13148-017-0435-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Berdasco M., Esteller M. Aberrant epigenetic landscape in cancer: how cellular identity goes awry. Dev. Cell. 2010;19(5):698–711. doi: 10.1016/j.devcel.2010.10.005. [DOI] [PubMed] [Google Scholar]
- 142.Klutstein M., et al. Contribution of epigenetic mechanisms to variation in cancer risk among tissues. Proc Natl Acad Sci U S A. 2017;114(9):2230–2234. doi: 10.1073/pnas.1616556114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ley T.J., et al. DNMT3A mutations in acute myeloid leukemia. N. Engl. J. Med. 2010;363(25):2424–2433. doi: 10.1056/NEJMoa1005143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ribeiro A.F., et al. Mutant DNMT3A: a marker of poor prognosis in acute myeloid leukemia. Blood. 2012;119(24):5824–5831. doi: 10.1182/blood-2011-07-367961. [DOI] [PubMed] [Google Scholar]
- 145.Russler-Germain D.A., et al. The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell. 2014;25(4):442–454. doi: 10.1016/j.ccr.2014.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Walter M.J., et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia. 2011;25(7):1153–1158. doi: 10.1038/leu.2011.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Couronné L., Bastard C., Bernard O.A. TET2 and DNMT3A mutations in human T-cell lymphoma. N. Engl. J. Med. 2012;366(1):95–96. doi: 10.1056/NEJMc1111708. [DOI] [PubMed] [Google Scholar]
- 148.Odejide O., et al. A targeted mutational landscape of angioimmunoblastic T-cell lymphoma. Blood. 2014;123(9):1293–1296. doi: 10.1182/blood-2013-10-531509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Palomero T., et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nat. Genet. 2014;46(2):166–170. doi: 10.1038/ng.2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Choi J., et al. Genomic landscape of cutaneous T cell lymphoma. Nat. Genet. 2015;47(9):1011–1019. doi: 10.1038/ng.3356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Grossmann V., et al. The molecular profile of adult T-cell acute lymphoblastic leukemia: mutations in RUNX1 and DNMT3A are associated with poor prognosis in T-ALL. Genes Chromosomes Cancer. 2013;52(4):410–422. doi: 10.1002/gcc.22039. [DOI] [PubMed] [Google Scholar]
- 152.Lemonnier F., et al. Recurrent TET2 mutations in peripheral T-cell lymphomas correlate with TFH-like features and adverse clinical parameters. Blood. 2012;120(7):1466–1469. doi: 10.1182/blood-2012-02-408542. [DOI] [PubMed] [Google Scholar]
- 153.Abdel-Wahab O., et al. Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood. 2009;114(1):144–147. doi: 10.1182/blood-2009-03-210039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Langemeijer S.M., et al. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat. Genet. 2009;41(7):838–842. doi: 10.1038/ng.391. [DOI] [PubMed] [Google Scholar]
- 155.Tefferi A., et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia. 2009;23(5):905–911. doi: 10.1038/leu.2009.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Zhang X., et al. DNMT3A and TET2 compete and cooperate to repress lineage-specific transcription factors in hematopoietic stem cells. Nat. Genet. 2016;48(9):1014–1023. doi: 10.1038/ng.3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Marcucci G., et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J. Clin. Oncol. 2010;28(14):2348–2355. doi: 10.1200/JCO.2009.27.3730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Mizuno S., et al. Expression of DNA methyltransferases DNMT1, 3A, and 3B in normal hematopoiesis and in acute and chronic myelogenous leukemia. Blood. 2001;97(5):1172–1179. doi: 10.1182/blood.v97.5.1172. [DOI] [PubMed] [Google Scholar]
- 159.Younesian S., et al. Residual methylation of tumor suppressor gene promoters, RASSF6 and RASSF10, as novel biomarkers for minimal residual disease detection in adult acute lymphoblastic leukemia. Ann. Hematol. 2019;98(12):2719–2727. doi: 10.1007/s00277-019-03775-y. [DOI] [PubMed] [Google Scholar]
- 160.Cerami E., et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404. doi: 10.1158/2159-8290.CD-12-0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Gao J., et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013;6(269):pl1. doi: 10.1126/scisignal.2004088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Saito Y., et al. Overexpression of a splice variant of DNA methyltransferase 3b, DNMT3b4, associated with DNA hypomethylation on pericentromeric satellite regions during human hepatocarcinogenesis. Proc Natl Acad Sci U S A. 2002;99(15):10060–10065. doi: 10.1073/pnas.152121799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Stirzaker C., et al. Methyl-CpG-binding protein MBD2 plays a key role in maintenance and spread of DNA methylation at CpG islands and shores in cancer. Oncogene. 2017;36(10):1328–1338. doi: 10.1038/onc.2016.297. [DOI] [PubMed] [Google Scholar]
- 164.Magdinier F., Wolffe A.P. Selective association of the methyl-CpG binding protein MBD2 with the silent p14/p16 locus in human neoplasia. Proc Natl Acad Sci U S A. 2001;98(9):4990–4995. doi: 10.1073/pnas.101617298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Bader S., Walker M., Harrison D. Most microsatellite unstable sporadic colorectal carcinomas carry MBD4 mutations. Br. J. Cancer. 2000;83(12):1646–1649. doi: 10.1054/bjoc.2000.1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Yamada T., et al. Frameshift mutations in the MBD4/MED1 gene in primary gastric cancer with high-frequency microsatellite instability. Cancer Lett. 2002;181(1):115–120. doi: 10.1016/s0304-3835(02)00043-5. [DOI] [PubMed] [Google Scholar]
- 167.Moran S., et al. Epigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysis. Lancet Oncol. 2016;17(10):1386–1395. doi: 10.1016/S1470-2045(16)30297-2. [DOI] [PubMed] [Google Scholar]
- 168.Imperiale T.F., et al. Multitarget stool DNA testing for colorectal-cancer screening. N. Engl. J. Med. 2014;370(14):1287–1297. doi: 10.1056/NEJMoa1311194. [DOI] [PubMed] [Google Scholar]
- 169.Warren J.D., et al. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011;9:133. doi: 10.1186/1741-7015-9-133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Lamb Y.N., Dhillon S. Epi proColon(®) 2.0 ce: a blood-based screening test for colorectal cancer. Mol. Diagn. Ther. 2017;21(2):225–232. doi: 10.1007/s40291-017-0259-y. [DOI] [PubMed] [Google Scholar]
- 171.Kanai Y., et al. Mutation of the DNA methyltransferase (DNMT) 1 gene in human colorectal cancers. Cancer Lett. 2003;192(1):75–82. doi: 10.1016/s0304-3835(02)00689-4. [DOI] [PubMed] [Google Scholar]
- 172.Ma H.S., et al. Overexpression of DNA (Cytosine-5)-Methyltransferase 1 (DNMT1) and DNA (Cytosine-5)-Methyltransferase 3A (DNMT3A) is associated with aggressive behavior and hypermethylation of tumor suppressor genes in human pituitary adenomas. Med Sci Monit. 2018;24:4841–4850. doi: 10.12659/MSM.910608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Etoh T., et al. Increased DNA methyltransferase 1 (DNMT1) protein expression correlates significantly with poorer tumor differentiation and frequent DNA hypermethylation of multiple CpG islands in gastric cancers. Am. J. Pathol. 2004;164(2):689–699. doi: 10.1016/S0002-9440(10)63156-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Lin R.K., et al. Dysregulation of p53/Sp1 control leads to DNA methyltransferase-1 overexpression in lung cancer. Cancer Res. 2010;70(14):5807–5817. doi: 10.1158/0008-5472.CAN-09-4161. [DOI] [PubMed] [Google Scholar]
- 175.Leonard S., et al. Over-expression of DNMT3A predicts the risk of recurrent vulvar squamous cell carcinomas. Gynecol. Oncol. 2016;143(2):414–420. doi: 10.1016/j.ygyno.2016.09.001. [DOI] [PubMed] [Google Scholar]
- 176.Kim M.S., et al. Mutational analysis of DNMT3A gene in acute leukemias and common solid cancers. Apmis. 2013;121(2):85–94. doi: 10.1111/j.1600-0463.2012.02940.x. [DOI] [PubMed] [Google Scholar]
- 177.Lai S.-C., et al. DNMT3b/OCT4 expression confers sorafenib resistance and poor prognosis of hepatocellular carcinoma through IL-6/STAT3 regulation. J. Exp. Clin. Cancer Res. 2019;38(1):1–18. doi: 10.1186/s13046-019-1442-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Jin F., et al. Up-regulation of DNA methyltransferase 3B expression in endometrial cancers. Gynecol. Oncol. 2005;96(2):531–538. doi: 10.1016/j.ygyno.2004.10.039. [DOI] [PubMed] [Google Scholar]
- 179.Müller H.M., et al. MeCP2 and MBD2 expression in human neoplastic and non-neoplastic breast tissue and its association with oestrogen receptor status. Br. J. Cancer. 2003;89(10):1934–1939. doi: 10.1038/sj.bjc.6601392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Liu H., et al. Methyl-CpG binding domain 1 gene polymorphisms and lung cancer risk in a Chinese population. Biomarkers. 2008;13(6):607–617. doi: 10.1080/13547500802168031. [DOI] [PubMed] [Google Scholar]
- 181.Xu J., et al. Up-regulation of MBD1 promotes pancreatic cancer cell epithelial-mesenchymal transition and invasion by epigenetic down-regulation of E-cadherin. Curr. Mol. Med. 2013;13(3):387–400. [PubMed] [Google Scholar]
- 182.Zhao S., et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A. 2013;110(8):2916–2921. doi: 10.1073/pnas.1222577110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Saito Y., et al. Expression of mRNA for DNA methyltransferases and methyl-CpG-binding proteins and DNA methylation status on CpG islands and pericentromeric satellite regions during human hepatocarcinogenesis. Hepatology. 2001;33(3):561–568. doi: 10.1053/jhep.2001.22507. [DOI] [PubMed] [Google Scholar]
- 184.Dolnik A., et al. Commonly altered genomic regions in acute myeloid leukemia are enriched for somatic mutations involved in chromatin remodeling and splicing. Blood. 2012;120(18):e83–e92. doi: 10.1182/blood-2011-12-401471. [DOI] [PubMed] [Google Scholar]
- 185.Kosmider O., et al. Mutations of IDH1 and IDH2 genes in early and accelerated phases of myelodysplastic syndromes and MDS/myeloproliferative neoplasms. Leukemia. 2010;24(5):1094–1096. doi: 10.1038/leu.2010.52. [DOI] [PubMed] [Google Scholar]
- 186.Green A., Beer P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N. Engl. J. Med. 2010;362(4):369–370. doi: 10.1056/NEJMc0910063. [DOI] [PubMed] [Google Scholar]
- 187.Yan H., et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 2009;360(8):765–773. doi: 10.1056/NEJMoa0808710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Amary M.F., et al. IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J. Pathol. 2011;224(3):334–343. doi: 10.1002/path.2913. [DOI] [PubMed] [Google Scholar]
- 189.Hemerly J.P., Bastos A.U., Cerutti J.M. Identification of several novel non-p.R132 IDH1 variants in thyroid carcinomas. Eur. J. Endocrinol. 2010;163(5):747–755. doi: 10.1530/EJE-10-0473. [DOI] [PubMed] [Google Scholar]
- 190.Tomlinson I.P., et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat. Genet. 2002;30(4):406–410. doi: 10.1038/ng849. [DOI] [PubMed] [Google Scholar]
- 191.Bardella C., Pollard P.J., Tomlinson I. SDH mutations in cancer. Biochim. Biophys. Acta. 2011;1807(11):1432–1443. doi: 10.1016/j.bbabio.2011.07.003. [DOI] [PubMed] [Google Scholar]
- 192.Killian J.K., et al. Succinate dehydrogenase mutation underlies global epigenomic divergence in gastrointestinal stromal tumor. Cancer Discov. 2013;3(6):648–657. doi: 10.1158/2159-8290.CD-13-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.van Kessel K.E., et al. Evaluation of an epigenetic profile for the detection of bladder cancer in patients with hematuria. J. Urol. 2016;195(3):601–607. doi: 10.1016/j.juro.2015.08.085. [DOI] [PubMed] [Google Scholar]
- 194.Ghoreifi A., et al. A urine-based DNA methylation marker test to detect upper tract urothelial carcinoma: a prospective cohort study. J. Urol. 2023;209(5):854–862. doi: 10.1097/JU.0000000000003188. [DOI] [PubMed] [Google Scholar]
- 195.Jiang Y.H., et al. Hypermethylation loci of ZNF671, IRF8, and OTX1 as potential urine-based predictive biomarkers for bladder cancer. Diagnostics. 2024;14(5) doi: 10.3390/diagnostics14050468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Manoochehri M., et al. DNA methylation biomarkers for noninvasive detection of triple-negative breast cancer using liquid biopsy. Int. J. Cancer. 2023;152(5):1025–1035. doi: 10.1002/ijc.34337. [DOI] [PubMed] [Google Scholar]
- 197.Wang T., et al. A multiplex blood-based assay targeting DNA methylation in PBMCs enables early detection of breast cancer. Nat. Commun. 2023;14(1):4724. doi: 10.1038/s41467-023-40389-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Krasnyi A.M., et al. Methylation profile of small breast cancer tumors evaluated by modified MS-HRM. Int. J. Mol. Sci. 2023;24(16) doi: 10.3390/ijms241612660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Ned R.M., Melillo S., Marrone M. Fecal DNA testing for colorectal cancer screening: the ColoSure™ test. PLoS Curr. 2011;3:Rrn1220. doi: 10.1371/currents.RRN1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Chang L., et al. External quality assessment for detection of colorectal cancer by Septin9 DNA methylation in clinical laboratories. Clin. Chim. Acta. 2024;552 doi: 10.1016/j.cca.2023.117663. [DOI] [PubMed] [Google Scholar]
- 201.Lin W.H., et al. Circulating tumor DNA methylation marker MYO1-G for diagnosis and monitoring of colorectal cancer. Clin. Epigenet. 2021;13(1):232. doi: 10.1186/s13148-021-01216-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Liu X., et al. High-yield methylation markers for stool-based detection of colorectal cancer. Dig. Dis. Sci. 2020;65(6):1710–1719. doi: 10.1007/s10620-019-05908-9. [DOI] [PubMed] [Google Scholar]
- 203.Barták B.K., et al. Colorectal adenoma and cancer detection based on altered methylation pattern of SFRP1, SFRP2, SDC2, and PRIMA1 in plasma samples. Epigenetics. 2017;12(9):751–763. doi: 10.1080/15592294.2017.1356957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Schmitz M., et al. Performance of a methylation specific real-time PCR assay as a triage test for HPV-positive women. Clin. Epigenet. 2017;9(1):1–8. doi: 10.1186/s13148-017-0419-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.De Strooper L.M.A., et al. Validation of the FAM19A4/mir124-2 DNA methylation test for both lavage- and brush-based self-samples to detect cervical (pre)cancer in HPV-positive women. Gynecol. Oncol. 2016;141(2):341–347. doi: 10.1016/j.ygyno.2016.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Gilham C., et al. Long-term prediction by DNA methylation of high-grade cervical intraepithelial neoplasia: results of the ARTISTIC cohort. Int. J. Cancer. 2024;155(1):81–92. doi: 10.1002/ijc.34913. [DOI] [PubMed] [Google Scholar]
- 207.Cao D., et al. PCDHGB7 hypermethylation-based Cervical cancer Methylation (CerMe) detection for the triage of high-risk human papillomavirus-positive women: a prospective cohort study. BMC Med. 2024;22(1):55. doi: 10.1186/s12916-024-03267-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Shi L., et al. Promoter hypermethylation analysis of host genes in cervical intraepithelial neoplasia and cervical cancers on histological cervical specimens. BMC Cancer. 2023;23(1):168. doi: 10.1186/s12885-023-10628-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Fu Y., et al., Genome-wide 5-hydroxymethylcytosines in circulating cell-free DNA as noninvasive diagnostic markers for gastric cancer, Gastric Cancer. 2024. Epub ahead of print. [DOI] [PubMed]
- 210.Nie Y., et al. Combining methylated SEPTIN9 and RNF180 plasma markers for diagnosis and early detection of gastric cancer. Cancer Commun. 2023;43(11):1275–1279. doi: 10.1002/cac2.12478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Oussalah A., et al. Plasma mSEPT9: a novel circulating cell-free DNA-based epigenetic biomarker to diagnose hepatocellular carcinoma. EBioMedicine. 2018;30:138–147. doi: 10.1016/j.ebiom.2018.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Ekanayake Weeramange C., et al. Human papillomavirus (HPV) DNA methylation changes in HPV-associated head and neck cancer. Carcinogenesis. 2024;45(3):140–148. doi: 10.1093/carcin/bgae001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Weiss G., et al. Validation of the SHOX2/PTGER4 DNA methylation marker panel for plasma-based discrimination between patients with malignant and nonmalignant lung disease. J. Thorac. Oncol. 2017;12(1):77–84. doi: 10.1016/j.jtho.2016.08.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Liang W., et al. Accurate diagnosis of pulmonary nodules using a noninvasive DNA methylation test. J. Clin. Invest. 2021;131(10) doi: 10.1172/JCI145973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.He J., et al. Accurate classification of pulmonary nodules by a combined model of clinical, imaging, and cell-free DNA methylation biomarkers: a model development and external validation study. Lancet Digit Health. 2023;5(10):e647–e656. doi: 10.1016/S2589-7500(23)00125-5. [DOI] [PubMed] [Google Scholar]
- 216.Gaga M., et al. Validation of Lung EpiCheck, a novel methylation-based blood assay, for the detection of lung cancer in European and Chinese high-risk individuals. Eur. Respir. J. 2021;57(1) doi: 10.1183/13993003.02682-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Liu J., et al. Evaluating the comprehensive diagnosis efficiency of lung cancer, including measurement of SHOX2 and RASSF1A gene methylation. BMC Cancer. 2024;24(1):282. doi: 10.1186/s12885-024-12022-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Park S.J., et al. Combination analysis of PCDHGA12 and CDO1 DNA methylation in bronchial washing fluid for lung cancer diagnosis. J. Kor. Med. Sci. 2024;39(2):e28. doi: 10.3346/jkms.2024.39.e28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Herzog C., et al. Plasma cell-free DNA methylation analysis for ovarian cancer detection: analysis of samples from a case-control study and an ovarian cancer screening trial. Int. J. Cancer. 2024;154(4):679–691. doi: 10.1002/ijc.34757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Buckley D.N., et al. OvaPrint-A cell-free DNA methylation liquid biopsy for the risk assessment of high-grade serous ovarian cancer. Clin. Cancer Res. 2023;29(24):5196–5206. doi: 10.1158/1078-0432.CCR-23-1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Wojno K.J., et al. Reduced rate of repeated prostate biopsies observed in ConfirmMDx clinical utility field study. Am Health Drug Benefits. 2014;7(3):129–134. [PMC free article] [PubMed] [Google Scholar]
- 222.Ding T., et al. Influential factors on urine EV DNA methylation detection and its diagnostic potential in prostate cancer. Front. Genet. 2024;15 doi: 10.3389/fgene.2024.1338468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Franceschini G.M., et al. Noninvasive detection of neuroendocrine prostate cancer through targeted cell-free DNA methylation. Cancer Discov. 2024;14(3):424–445. doi: 10.1158/2159-8290.CD-23-0754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Eismann L., et al. Methylation status of various gene loci in localized prostate cancer: novel biomarkers for diagnostics and biochemical recurrence. Urol. Oncol.: Seminars and Original Investigations. 2023;41(7):325.e1–325.e8. doi: 10.1016/j.urolonc.2023.04.009. [DOI] [PubMed] [Google Scholar]
- 225.Chen J.Q., et al. Genome-scale methylation analysis identifies immune profiles and age acceleration associations with bladder cancer outcomes. Cancer Epidemiol. Biomarkers Prev. 2023;32(10):1328–1337. doi: 10.1158/1055-9965.EPI-23-0331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Yang X., et al. Clinical value of preoperative methylated septin 9 in Chinese colorectal cancer patients. World J. Gastroenterol. 2019;25(17):2099–2109. doi: 10.3748/wjg.v25.i17.2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Miura T., et al. Methylation of bone morphogenetic protein 2 is associated with poor prognosis in colorectal cancer. Oncol. Lett. 2020;19(1):229–238. doi: 10.3892/ol.2019.11091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Huang H., et al. DNA methylation-based patterns for early diagnostic prediction and prognostic evaluation in colorectal cancer patients with high tumor mutation burden. Front. Oncol. 2022;12 doi: 10.3389/fonc.2022.1030335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Mo S., et al. Early detection and prognosis prediction for colorectal cancer by circulating tumour DNA methylation haplotypes: a multicentre cohort study. EClinicalMedicine. 2023;55 doi: 10.1016/j.eclinm.2022.101717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Ye Z., et al. Optimized screening of DNA methylation sites combined with gene expression analysis to identify diagnostic markers of colorectal cancer. BMC Cancer. 2023;23(1):617. doi: 10.1186/s12885-023-10922-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Bu Q., et al. Septin9 DNA methylation as a promising biomarker for cervical cancer. J Obstet Gynaecol. 2023;43(1) doi: 10.1080/01443615.2022.2151356. [DOI] [PubMed] [Google Scholar]
- 232.Kessler T., et al. Prognostic markers of DNA methylation and next-generation sequencing in progressive glioblastoma from the EORTC-26101 trial. Clin. Cancer Res. 2023;29(19):3892–3900. doi: 10.1158/1078-0432.CCR-23-0926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Tang Q., et al. Comprehensive analysis reveals that LTBR is a immune-related biomarker for glioma. Comput. Biol. Med. 2024;174 doi: 10.1016/j.compbiomed.2024.108457. [DOI] [PubMed] [Google Scholar]
- 234.Ma J., et al. Prognostic effect of DNA methylation of BTG2 gene in Chinese hepatocellular carcinoma. Heliyon. 2024;10(7) doi: 10.1016/j.heliyon.2024.e28580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Saeki I., et al. Methylated SEPT9 assay-based liquid biopsy as a biomarker in molecular targeted agent-treated hepatocellular carcinoma. Hepatology International. 2023;17(5):1289–1299. doi: 10.1007/s12072-023-10488-y. [DOI] [PubMed] [Google Scholar]
- 236.Zhou Z., Jin H., Xu J. A gene signature driven by abnormally methylated DEGs was developed for TP53 wild-type ovarian cancer samples by integrative omics analysis of DNA methylation and gene expression data. Ann. Transl. Med. 2023;11(1):20. doi: 10.21037/atm-22-5764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Maekawa H., et al. Analysis of the methylation of CpG islands in the CDO1, TAC1 and CHFR genes in pancreatic ductal cancer. Oncol. Lett. 2020;19(3):2197–2204. doi: 10.3892/ol.2020.11340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Habeshian T.S., et al. DNA methylation markers for risk of metastasis in a cohort of men with localized prostate cancer. Epigenetics. 2024;19(1) doi: 10.1080/15592294.2024.2308920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Zhang W., et al. Gene methylation status in focus of advanced prostate cancer diagnostics and improved individual outcomes. Transl. Androl. Urol. 2023;12(12):1813–1826. doi: 10.21037/tau-23-405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Liu M.C., et al. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 2020;31(6):745–759. doi: 10.1016/j.annonc.2020.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Kang S., et al. CancerLocator: non-invasive cancer diagnosis and tissue-of-origin prediction using methylation profiles of cell-free DNA. Genome Biol. 2017;18(1):53. doi: 10.1186/s13059-017-1191-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Hoadley K.A., et al. Cell-of-Origin patterns dominate the molecular classification of 10,000 tumors from 33 types of cancer. Cell. 2018;173(2):291–304.e6. doi: 10.1016/j.cell.2018.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Pilala K.M., et al. MIR145 core promoter methylation in pretreatment cell-free DNA: a liquid biopsy tool for muscle-invasive bladder cancer treatment outcome. JCO Precis Oncol. 2024;8 doi: 10.1200/PO.23.00414. [DOI] [PubMed] [Google Scholar]
- 244.Lu Y.-T., et al. Cell-free DNA methylation as a predictive biomarker of response to neoadjuvant chemotherapy for patients with muscle-invasive bladder cancer in SWOG S1314. European Urology Oncology. 2023;6(5):516–524. doi: 10.1016/j.euo.2023.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Schricker G., et al. Clinical performance of an analytically validated assay in comparison to microarray technology to assess PITX2 DNA-methylation in breast cancer. Sci. Rep. 2018;8(1):1–10. doi: 10.1038/s41598-018-34919-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Veeck J., et al. BRCA1 CpG island hypermethylation predicts sensitivity to poly(adenosine diphosphate)-ribose polymerase inhibitors. J. Clin. Oncol. 2010;28(29):e563–e564. doi: 10.1200/JCO.2010.30.1010. author reply e565-e566. [DOI] [PubMed] [Google Scholar]
- 247.Duan C., et al. DNA methylation characteristics associated with chemotherapy resistance in epithelial ovarian cancer. Heliyon. 2024;10(5) doi: 10.1016/j.heliyon.2024.e27212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Quillien V., et al. Clinical validation of the CE-IVD marked Therascreen MGMT kit in a cohort of glioblastoma patients. Cancer Biomarkers. 2017;20(4):435–441. doi: 10.3233/CBM-170191. [DOI] [PubMed] [Google Scholar]
- 249.Eckhardt A., et al. Mean global DNA methylation serves as independent prognostic marker in IDH-wildtype glioblastoma. Neuro Oncol. 2023;26(3):503–513. doi: 10.1093/neuonc/noad197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Moutinho C., et al. Epigenetic inactivation of the BRCA1 interactor SRBC and resistance to oxaliplatin in colorectal cancer. J Natl Cancer Inst. 2014;106(1):djt322. doi: 10.1093/jnci/djt322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Lopez-Serra P., et al. A DERL3-associated defect in the degradation of SLC2A1 mediates the Warburg effect. Nat. Commun. 2014;5:3608. doi: 10.1038/ncomms4608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Kawakami K., et al. Long interspersed nuclear element-1 hypomethylation is a potential biomarker for the prediction of response to oral fluoropyrimidines in microsatellite stable and CpG island methylator phenotype-negative colorectal cancer. Cancer Sci. 2011;102(1):166–174. doi: 10.1111/j.1349-7006.2010.01776.x. [DOI] [PubMed] [Google Scholar]
- 253.Ebert M.P., et al. TFAP2E-DKK4 and chemoresistance in colorectal cancer. N. Engl. J. Med. 2012;366(1):44–53. doi: 10.1056/NEJMoa1009473. [DOI] [PubMed] [Google Scholar]
- 254.Cha Y., et al. Association of CHFR promoter methylation with treatment outcomes of irinotecan-based chemotherapy in metastatic colorectal cancer. Neoplasia. 2019;21(1):146–155. doi: 10.1016/j.neo.2018.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Janssens K., et al. NPY methylated ctDNA is a promising biomarker for treatment response monitoring in metastatic colorectal cancer. Clin. Cancer Res. 2023;29(9):1741–1750. doi: 10.1158/1078-0432.CCR-22-1500. [DOI] [PubMed] [Google Scholar]
- 256.Diaz-Lagares A., et al. Epigenetic inactivation of the p53-induced long noncoding RNA TP53 target 1 in human cancer. Proc Natl Acad Sci U S A. 2016;113(47):E7535–e7544. doi: 10.1073/pnas.1608585113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Nogales V., et al. Epigenetic inactivation of the putative DNA/RNA helicase SLFN11 in human cancer confers resistance to platinum drugs. Oncotarget. 2016;7(3):3084–3097. doi: 10.18632/oncotarget.6413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Duruisseaux M., et al. Epigenetic prediction of response to anti-PD-1 treatment in non-small-cell lung cancer: a multicentre, retrospective analysis. Lancet Respir. Med. 2018;6(10):771–781. doi: 10.1016/S2213-2600(18)30284-4. [DOI] [PubMed] [Google Scholar]
- 259.Su S.F., et al. A panel of three markers hyper- and hypomethylated in urine sediments accurately predicts bladder cancer recurrence. Clin. Cancer Res. 2014;20(7):1978–1989. doi: 10.1158/1078-0432.CCR-13-2637. [DOI] [PubMed] [Google Scholar]
- 260.Witjes J.A., et al. Performance of the bladder EpiCheck™ methylation test for patients under surveillance for non-muscle-invasive bladder cancer: results of a multicenter, prospective, blinded clinical trial. Eur Urol Oncol. 2018;1(4):307–313. doi: 10.1016/j.euo.2018.06.011. [DOI] [PubMed] [Google Scholar]
- 261.Pierconti F., et al. DNA methylation analysis in urinary samples: a useful method to predict the risk of neoplastic recurrence in patients with urothelial carcinoma of the bladder in the high-risk group. Cancer Cytopathol. 2023;131(3):158–164. doi: 10.1002/cncy.22657. [DOI] [PubMed] [Google Scholar]
- 262.Young G.P., et al. A cross-sectional study comparing a blood test for methylated BCAT1 and IKZF1 tumor-derived DNA with CEA for detection of recurrent colorectal cancer. Cancer Med. 2016;5(10):2763–2772. doi: 10.1002/cam4.868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Jiang H., et al. Role of blood mSEPT9 in evaluating tumor burden and disease monitoring in colorectal cancer patients. J. Clin. Lab. Anal. 2021;35(11) doi: 10.1002/jcla.24030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Li M., et al. A DNA methylation signature for the prediction of tumour recurrence in stage II colorectal cancer. Br. J. Cancer. 2023;128(9):1681–1689. doi: 10.1038/s41416-023-02155-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Dietrich D., et al. Circulating cell-free SEPT9 DNA methylation in blood is a biomarker for minimal residual disease detection in head and neck squamous cell carcinoma patients. Clin Chem. 2023 Sep 1;69(9):1050–1061. doi: 10.1093/clinchem/hvad084. [DOI] [PubMed] [Google Scholar]
- 266.Derissen E.J., Beijnen J.H., Schellens J.H. Concise drug review: azacitidine and decitabine. Oncol. 2013;18(5):619–624. doi: 10.1634/theoncologist.2012-0465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Tsai H.C., et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell. 2012;21(3):430–446. doi: 10.1016/j.ccr.2011.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Cheishvili D., Boureau L., Szyf M. DNA demethylation and invasive cancer: implications for therapeutics. Br. J. Pharmacol. 2015;172(11):2705–2715. doi: 10.1111/bph.12885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Issa J.P., Kantarjian H.M. Targeting DNA methylation. Clin. Cancer Res. 2009;15(12):3938–3946. doi: 10.1158/1078-0432.CCR-08-2783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Pappalardi M.B., et al. Discovery of a first-in-class reversible DNMT1-selective inhibitor with improved tolerability and efficacy in acute myeloid leukemia. Nat Cancer. 2021;2(10):1002–1017. [PMC free article] [PubMed] [Google Scholar]
- 271.Stein E.M., et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood. 2019;133(7):676–687. doi: 10.1182/blood-2018-08-869008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.DiNardo C.D., et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N. Engl. J. Med. 2018;378(25):2386–2398. doi: 10.1056/NEJMoa1716984. [DOI] [PubMed] [Google Scholar]
- 273.Mellinghoff I.K., et al. PL3.1 A phase 1, open-label, perioperative study of ivosidenib (AG-120) and vorasidenib (AG-881) in recurrent, IDH1-mutant, low-grade glioma: results from cohort 1. Neuro Oncol. 2019;21(Suppl 3):iii2. [Google Scholar]
- 274.Linnekamp J.F., et al. Clinical and biological effects of demethylating agents on solid tumours - a systematic review. Cancer Treat Rev. 2017;54:10–23. doi: 10.1016/j.ctrv.2017.01.004. [DOI] [PubMed] [Google Scholar]
- 275.Zhang Y., et al. The safety, efficacy, and treatment outcomes of a combination of low-dose decitabine treatment in patients with recurrent ovarian cancer. OncoImmunology. 2017;6(9) doi: 10.1080/2162402X.2017.1323619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Juergens R.A., et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1(7):598–607. doi: 10.1158/2159-8290.CD-11-0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Garcia-Manero G., et al. Oral cedazuridine/decitabine for MDS and CMML: a phase 2 pharmacokinetic/pharmacodynamic randomized crossover study. Blood. 2020;136(6):674–683. doi: 10.1182/blood.2019004143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.de Lima M., et al. CC-486 maintenance after stem cell transplantation in patients with acute myeloid leukemia or myelodysplastic syndromes. Biol. Blood Marrow Transplant. 2018;24(10):2017–2024. doi: 10.1016/j.bbmt.2018.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Taylor K., et al. An open-label, phase II multicohort study of an oral hypomethylating agent CC-486 and durvalumab in advanced solid tumors. Journal for immunotherapy of cancer. 2020;8(2) doi: 10.1136/jitc-2020-000883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Bever K.M., et al. A feasibility study of combined epigenetic and vaccine therapy in advanced colorectal cancer with pharmacodynamic endpoint. Clin. Epigenet. 2021;13(1):1–9. doi: 10.1186/s13148-021-01014-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Oza A.M., et al. A randomized phase II trial of epigenetic priming with guadecitabine and carboplatin in platinum-resistant, recurrent ovarian cancer. Clin. Cancer Res. 2020;26(5):1009–1016. doi: 10.1158/1078-0432.CCR-19-1638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Juvinao-Quintero D.L., et al. DNA methylation and type 2 diabetes: the use of mendelian randomization to assess causality. Curr Genet Med Rep. 2019;7(4):191–207. doi: 10.1007/s40142-019-00176-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Kelsey G., Stegle O., Reik W. Single-cell epigenomics: recording the past and predicting the future. Science. 2017;358(6359):69–75. doi: 10.1126/science.aan6826. [DOI] [PubMed] [Google Scholar]
- 284.Zou J., et al. Epigenome-wide association studies without the need for cell-type composition. Nat. Methods. 2014;11(3):309–311. doi: 10.1038/nmeth.2815. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.







