Open Access

Research progress of DNA methylation in colorectal cancer (Review)

  • Authors:
    • Yuxin Wang
    • Chengcheng Wang
    • Ruiqi Zhong
    • Liang Wang
    • Lei Sun
  • View Affiliations

  • Published online on: July 3, 2024     https://doi.org/10.3892/mmr.2024.13278
  • Article Number: 154
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA methylation is one of the earliest and most significant epigenetic mechanisms discovered. DNA methylation refers, in general, to the addition of a methyl group to a specific base in the DNA sequence under the catalysis of DNA methyltransferase, with S‑adenosine methionine as the methyl donor, via covalent bonding and chemical modifications. DNA methylation is an important factor in inducing cancer. There are different types of DNA methylation, and methylation at different sites plays different roles. It is well known that the progression of colorectal cancer (CRC) is affected by the methylation of key genes. The present review did not only discuss the potential relationship between DNA methylation and CRC but also discussed how DNA methylation affects the development of CRC by affecting key genes. Furthermore, the clinical significance of DNA methylation in CRC was highlighted, including that of the therapeutic targets and biomarkers of methylation; and the importance of DNA methylation inhibitors was discussed as a novel strategy for treatment of CRC. The present review did not only focus upon the latest research findings, but earlier reviews were also cited as references to older literature.

Introduction

Colorectal cancer (CRC) is one of the common malignant tumors of the digestive tract, and its incidence is second only to that of gastric and esophageal cancers. CRC ranks fifth in men and sixth in women as a major cause of tumor-related deaths in China. In the past 20 years, the morbidity of CRC has been on the rise, and the onset age has been increasing. In the western developed countries, CRC is the second most common malignancy after lung cancer. The incidence rate varies by up to 60 times in different countries. The sites most prone are the rectum and the junction between the rectum and sigmoid colon, accounting for 60% of the cancerous region. The incidence of CRC is related to various factors, such as eating habits, genetics and colitis. The disease is sporadic in most cases and usually indicates the joint effect of genes and environment, as only 20–25% of patients demonstrated a family history. Only 5–6% of CRC cases are attributed to genetic conditions and are referred to as the CRC genetic syndrome (1). In recent years, the morbidity of colon cancer in western developed countries has demonstrated a declining trend, especially in the 50–74 age group, which is closely associated with precancer prevention and treatment (2). One of the reasons for the high incidence rate of colon cancer in China is the lack of effective early detection methods. When clinically diagnosed, most nodules present as advanced colon cancer. CRC is a serious health risk, ≥1,000,000 new cases are diagnosed every year globally. This disease occurs as a result of a multi-step process that causes the accumulation of genetic and epigenetic changes in colonic mucosal cells, which mainly affect onco-, tumor suppressor, and DNA repair genes, all of which take part in the key pathways of CRC initiation and progression (3,4). Among them, p53, the representative tumor suppressor gene, remains under study, and a number of scientists are still discussing the therapeutic strategy of targeting p53 (4). Epigenetic processes-DNA methylation, histone tail modification and chromatin remodeling, as well as the mechanisms mediated by noncoding RNA molecules-are used to describe the mechanisms that can modify the expression levels of selected genes without necessarily changing their DNA sequence. Epigenetic modifications are usually environmentally induced and are tissue-specific phenomena. They can have similar effects on pathogenic mutations or functional polymorphisms because they can silence, increase, or reduce the expression of selected genes in different tissues. This is particularly relevant to cancer-related genes, such as tumor suppressor or DNA repair genes (5,6).

With the progress of epigenetics, the association between DNA methylation and tumors has become a research hotspot. A series of studies have revealed that there are some specific abnormal methylations in the process of tumorigenesis, which can be used as a molecular index for tumor diagnosis. Genome-wide methylation analysis has now characterized novel genes in patients with synchronous CRC, which has the clinical potential to improve the diagnosis and management of patients with CRC (7).

The present study review focused on how DNA methylation affects key genes, which in turn affect CRC development, and describes its clinical applications, including the detection of CRC with methylation biomarkers, the prognostic analysis of methylated CRC cells, and the relationship between DNA methylation and targeted CRC therapy. Finally, it was highlighted that DNA methylation is one of the important therapeutic targets for CRC, and that its inhibitors provide new ideas for the clinic therapy of CRC. In summary, compared with other reviews, this review introduces the relationship between DNA methylation and CRC more comprehensively, from mechanism and key gene targets to clinical research, and provides support for the treatment of CRC with DNA methylation as a breakthrough (Fig. 1).

DNA methylation

Introduction

DNA methylation is one of the different ways of gene regulation in mammals, and it has been a research hot spot in epigenetics in recent years. DNA methylation is a chemical modification of DNA that can regulate genetic expression without changing the DNA sequence. In a broad sense, DNA methylation refers to the chemical modification process wherein a methyl group is added to a specific base of the DNA sequence via covalent bonding, with S-adenosylmethionine (SAM) as the methyl donor, under the catalysis of DNA methyltransferase (DNMT). Although there are numerous variations in methylation modification, the bases of the modified sites are usually adenine n-6, cytosine n-4, guanine N-7 and cytosine C-5. DNA methylation involved in general research mainly refers to the methylation process of the fifth carbon atom of cytosine in cytosine guanine dinucleotide (CpG) dinucleotide, and the final product of this process is 5-methyl-cytosine.

After fertilization in mammals, entire methylation of DNA molecules inherited from their parents is cleared by specific enzymes. After the embryo is implanted into the uterus, a new round of methylation begins, and the genome of the fertilized egg is methylated again under the action of DNA methylase. The stability of DNA methylation is maintained by DNMT1.

In mammals, DNMT1 and DNMT3 are the most essential DNA methyltransferases. DNMT1 is involved in the maintenance of DNA methylation and is related to the extension of DNA methylation. DNMT3 includes DNMT3a and DNMT3b, which are involved in de novo methylation. Other roles and applications of DNMT family remain under investigation (8,9).

DNA demethylation is regulated by internal fragments of genes and their binding factors. There are two hypotheses that can explain the molecular mechanism underlying DNA demethylation. One hypothesis associated with DNA semi retention replication is passive demethylation. If DNA methylation does not occur after semi-retention replication, the DNA would be in a semi-methylated state. If the semi-retention replication of semi-methylated DNA occurs again and the DNA methylation activity is still inhibited, it indicates that 50% of the cells are in a semi-methylated state. The second hypothesis is independent of the semi-retention replication and is an active process, that is to say, DNA demethylation is catalyzed by DNA demethylase. DNA demethylation is the removal of methylated bases under the action of DNA glycosidase, which is equivalent to the repair of damaged DNA by glycosidases and base free nucleases.

DNA methylation affects gene expression in tumors

Numerous genes, especially the promoter region of the housekeeping gene, usually have some regions rich in di-nuclear glycoside ‘CG’, called ‘CpG islands’. They are usually found in the promoter and exon regions of genes. Some regions are rich in CpG dinucleotides, with a length of 300–3,000 base pairs. Methylation of specific CpG dinucleotides in the promoter region is involved in transcription regulation. Scientific studies have revealed that DNA methylation modification demonstrates a stronger inertia in vitro (10).

Several studies revealed that the methylation status of tumor cells in the promoter regions of tumor suppressor genes and repair genes is enhanced, resulting in the inhibition of the expression of corresponding tumor suppressor gene (11,12). Moreover, abnormal methylation is common in chronic inflammation (13), which in turn contributes directly to tumors and cancers to a large extent.

In 2015, a study on the CRC subtype alliance summarized four common molecular subtypes (CMS) of CRC: CMS1, CMS2, CMS3 and CMS4. Among them, CMS1 is highly mutated, and all microsatellite-unstable CRCs are CMS1 (14). These kinds of tumors have high methylation status and BRAF mutation rates and a poor prognosis with the other types. However, the other CMSs demonstrate more chromosomal instability (CIN) in CRC typing. Although CMS3 demonstrates moderate hypermethylation levels, the KRAS gene is highly mutated. CMS3 CRC has a high mortality rate once it recurs. In the present study, the classic chromosome instability (CIN) and microsatellite instability (MSI) pathways have been used and other pathways for the pathogenesis of CRC as examples to elucidate the development process of CRC.

CIN pathway

Studies have revealed that mutations of the tumor suppressor genes APC regulator of Wnt signaling pathway (APC) and TP53, and activation mutations of KRAS and PIK3CA are related events in CIN pathway-induced tumors. Interestingly, the earliest event in colorectal tumors appears to be mutation inactivation of APC (15), which causes the Wnt signaling pathway to be activated, a feature common to almost all tumors (16).

After the mutation of APC, the mutation of KRAS also occurs (17): When normal, the KRAS image molecular switch can control the pathway that regulates cell proliferation; however, when abnormal, it causes continuous cell proliferation and prevents self-destruction. The KRAS image molecular switch participates in intracellular signal transduction. When the KRAS gene is mutated, it is permanently activated and cannot produce normal RAS protein, leading to intracellular signal transduction error, rampant cell proliferation and canceration (18).

Most sporadic CRC follow the CIN pathway, whereas 85% of sporadic CRC have cyclooxygenase 2 (COX-2) expression (19). This suggests that COX-2 is also an important factor in inducing tumors. During the development of colorectal tumors, COX-2 may convert free arachidonic acid into prostaglandins and regulate the proliferation of CRC cells; and it can be used as an alternative approach for the treatment of CRC (20).

MSI pathway

Regionalized hypermethylation is a common feature of CRC with MSI phenotype, including CpG island methylation phenotype (CIMP). In a study by Samowitz et al (21), APC mutations were observed in both MSI and CIMP high cancers, and there was a significant inverse correlation trend between APC mutations and CIMP. However, the mutation frequency of APC and TP53 that was aforementioned is lower in the MSI pathway than in the CIN pathway (22).

Carcinogenesis of the MSI pathway includes the mutation of TGF-βR2. The TGF-βR2 gene encodes a protein that inhibits the proliferation of colonic epithelial cells. The mutated expression product of the gene no longer sends a signal to prevent proliferation, which leads to abnormal proliferation of colonic epithelial cells. Other mutated genes lead to cell cycle arrest (CASP5 and FAS) and abnormal DNA repair (MBD4, BLM and CHK1) (2327).

Other pathways

The serrated pathway is an alternative multistep mechanism of carcinogenesis. It has been revealed that serrated colorectal lesions rarely harbor truncated APC mutations, most are BRAF mutations, and KRAS mutations remain rare. Regarding the serrated pathway, there was evidence of Fusobacterium nucleatum in 56% of CRCS, which was associated with CIMP-H status and large tumors (28).

In addition, IBD-CRC may represent a distinct tumorigenic pathway. Typical epithelial tumor subtypes associated with Wnt signaling are completely absent in IBD-CRCs, and different mechanisms of Wnt pathway dysregulation bias IBD-CRC toward mesenchymal tumor subtypes (29) (Table I).

Table I.

DNA methylation effects gene expression in tumors.

Table I.

DNA methylation effects gene expression in tumors.

GenesRolesMethylation sites
APCAPC affects cell growth and division, and also plays an important role in DNA repair, gene silencing and cell differentiation.15 CpG sites within the APC 1A promoter
TP53TP53 plays an important role in cell cycle regulation, DNA damage repair, apoptosis and anti-aging.C-terminal, K305
KRASKRAS controls the pathway that regulates cell growth.G13D, G12C, G12V, G12S, G12D
MTHFRMTHFR is involved in folate metabolism. 5,10-methylenetetrahydrofolate is converted to 5-methylenetetrahydrofolate with biological functions. It is essential for maintaining the normal methylation status in the human body.C677T, A1298C
ADHFE1ADHFE1 is responsible for the oxidation of 4-hydroxybutyrate. Energy metabolism and detoxification processes may be involved.405K

[i] APC, regulator of WNT signaling pathway; CpG, cytosine guanine dinucleotide; MTHFR, methylenetetrahydrofolate reductase; ADHFE1, alcohol dehydrogenase iron containing 1.

DNA methylation changes in several key genes in CRC

DNA methylation refers to the addition of methyl groups to the bases of DNA molecules by SAM under the action of DNMT. The covalent bond between cytosine 5 carbon atom and a base in CpG is the most common mode of modification. DNA methylation modification demonstrates a stronger inertia in vitro than in living systems. For example, sodium bisulfite can transform unmethylated cytosine into uracil, but cannot change the cytosine in methylated CpG; while in vivo, it leads to a reduction in gene expression. Therefore, hypermethylation status indicates inactivation/inhibition/silencing of gene expression (30); however, the hypomethylation state indicates the activation of gene expression. Early studies revealed that tumor cells are extensively hypomethylated at the genome-wide level, resulting in the activation of proto-oncogenes and an increase in genomic instability (31). It was recently discovered that tumor cells in the promoter regions of tumor suppressor and repair genes are hypermethylated, which causes the inhibition of the expression of corresponding tumor suppressor genes; additionally, it was revealed that the hypermethylated tumor cells mainly occur in CpG islands in the promoter region (32), although the corresponding cells are mostly in the non-methylated state.

Hypermethylation

Hypermethylation of the tumor suppressor gene CpG island is one of the mechanisms underlying CRC formation. So far, it has been revealed that numerous cancerous genes experience hypermethylation, and only a few protective mechanisms, such as active transcription, active demethylation, replication timing, and prevention of the acquisition of local chromatin structure of deoxyribonucleic acid methyltransferase, can prevent hypermethylation of CpG islands.

Recently, hypermethylation of ADHFE1, CNN1 and NR3C1 has been revealed to play important roles in signal transduction, cell cycle regulation, angiogenesis, as well as CRC (3335). Suzuki et al (36) reported that the incidence of hypermethylation and downregulation of the SFRP gene (a negative regulator of Wnt signaling pathway) in normal colonic mucosae of patients with CRC is higher than in the mucosae that of patients without CRC.

A meta-analysis revealed a relationship between ITGA4 promoter methylation status and malignancy, that is, IITGA4 hypermethylation was more frequent in tumor samples than in non-tumor samples. ITGA4 methylation analysis enables a reliable method for screening CRC in tissue samples. This has important implications for the detection of early CRC (37).

Hypermethylation of the promoter may contribute to the epigenetic silencing of ADAMTS14 in CRC. ADAMTS14 protein expression was higher in the anterior part of the invasive tumor than in the tumor center or other regions of the tumor. Its high expression is associated with poor prognosis in patients with CRC, suggesting that ADAMTS14 may be a promising indicator for evaluating the prognosis of CRC (38).

Hypomethylation

Hypomethylation, including CIN and MSI, is considered to promote tumorigenesis by activating proto-oncogenes, such as CMYC and HRAS, or causing genomic instability.

There are two common low functional polymorphic variants of methylenetetrahydrofolate reductase (MTHFR): T variant at nucleotide 677 (MTHFR C677T) and C variant at nucleotide 1298 (MTHFR A1298C). The first variant, C677T, is related to a reduced risk of CRC, and the cancer risk associated with MTHFR polymorphism may be regulated by folate intake. With sufficient folate intake, individuals carrying the variant MTHFR genotype may have a reduced risk of cancer. When folate intake is low, DNA methylation and DNA synthesis/repair in polymorphic individuals may be impaired, resulting in an increased risk of cancer. In animal studies, folate deficiency has been revealed to lead to exon-specific hypomethylation of p53 gene and increased DNA methyltransferase activity (39). However, moderate folate deficiency did not cause DNA methylation (40). It was discovered that MTHFR C677T polymorphism affected DNA methylation status through interaction with folate status. Another study examined the relationship between plasma folate status and colorectal adenoma, by assessing the effect of modification on the polymorphism of its gene (C677T). Compared with subjects with normal CC folate metabolism or diminished CT folate metabolism, subjects with poor TT folate metabolism had a low risk of developing colorectal adenoma at higher plasma folate levels (adjusted odds ratio, 0.58; 95% confidence interval, 0.21 to 1.61) and increased risk when their folate levels were low (adjusted odds ratio, 2.13; 95% confidence interval, 0.82 to 5.54) (41). This indicates that the risk of CRC is reduced when folate intake is low or metabolism is abnormal, that is, when the degree of DNA methylation is low. This also provides a breakthrough for the clinical treatment of CRC by controlling folate intake or using DNA methylation inhibitors.

The present study revealed upregulation and hypomethylation of HER3 gene expression in CRC cases. High expression and hypomethylation of HER3 may play an important role in the occurrence and development of CRC. CpG hypomethylation may be associated with the early stages of tumorigenesis. The discovery of this biomarker provides a powerful approach to improve current diagnostic and therapeutic measures (42).

Key genes

KRAS is a mouse sarcoma virus oncogene. There are three genes in the RAS gene family associated with human tumors: HRAS, KRAS and NRAS, located on chromosomes 11, 12, and 1, respectively. KRAS encodes a 21-kDa RAS protein, also known as the p21 gene, and has the greatest impact on human cancer. It is like a molecular switch: When normal, it can control the pathways regulating cell proliferation, and when abnormalities occur, it leads to continuous cell proliferation and prevents cell self-destruction. It is involved in intracellular signal transmission. When KRAS is mutated, the gene is permanently activated and cannot produce normal RAS protein, leading to an intracellular signal transduction disorder, that is, the inability to control cell proliferation and thus, cancer (43).

CRC is characterized by a series of mutation events involving APC, KRAS and TP53. KRAS is the most significant oncogenic mutation in CRC, occurring in 30–40% of patients with CRC (44). KRAS cooperates with the Wnt/β-catenin pathway to promote CRC and confers resistance to anti-EGFR antibodies (45).

The SLC25A22 process is required for the survival of CRC cells expressing activated KRAS, which are then rapidly incorporated into the tricarboxylic cycle (glutamine hydrolysis). In the absence of glutamine, cells can proliferate simply via incubation with succinate. The cells with mutated KRAS have a lower ratio of α-ketoglutarate to succinic acid, leading to reduced hypermethylation of 5-hydroxymethyl cytosine (a marker of DNA demethylation) and CpG sites. Numerous hypermethylated genes are located in the pro-cadherin gene cluster in the Wnt signaling pathway and chromosome 5q31. In CRC cells without KRAS mutations or with KRAS mutations and SLC25A22 knockout, the pro-cadherin gene expressed is not methylated at these sites (46).

In summary, in CRC cells expressing activated KRAS, SLC25A22 promotes the accumulation of succinic acid, resulting in increased DNA methylation, activation of Wnt signaling for increased expression of β-catenin, LGR5, proliferation, stem cell properties and resistance to 5-fluorouracil. Strategies that block this pathway may be used to treat CRC in the future (Fig. 2).

Clinical significance of DNA methylation in CRC

Using methylation markers to diagnose CRC

DNA methylation is a chemical modification that can alter genetic properties without altering the DNA sequence. The incidence of DNA methylation increases with aging, indicating that methylation may be related to aging and carcinogenesis (47). In the precursors of CRC (colorectal polyps) studies, similar abnormal methylation accumulation has been discovered in DNA sequences of patients, indicating that methylation biomarker detection in CRC could be a major trend in the future. Furthermore, it is considered that it is also possible to trace the origin of tumors through specific methylation of different types of tumors, which is helpful in determining the medication and other treatment strategies for patients. Interestingly, similar methylation also occurs in the colonic mucosae of healthy people, which means that the use of methylation biomarkers may predict the possibility of colon cancer (48). As aforementioned, it is considered by the authors that some of these genes, such as BRAF and KRAS, may provide a key breakthrough in methylation bioassay research.

Furthermore, MSI and CIN are two important types of CRC (49). MSI cancer is caused by the mutation of DNA mismatch repair (MMR) gene and the high methylation of promoter region. MMR is a protein that is produced by correcting the wrong base pair during the process of DNA replication. Therefore, detecting MMR status can predict the incidence of CRC to a certain extent.

In the study by Hinoue et al (50), CRC cases were divided into CIMP-high, CIMP-low, and non-CIMP types. Among them, cancer-related genes in the CIMP-high subgroup demonstrated hypermethylation and the BRAFV600 gene was mutated, while MLHL demonstrated hypermethylation. The CIMP-low subgroup is rich in KRAS mutation, which is characterized by DNA hypermethylation of CIMP-h-related marker subgroup.

Primarily, the study of Jensen et al (51) emphasized the potential utility of using the sensitivity and specificity of DNA methylation markers in blood samples to detect early tumors. Trimethoxy is a minimally invasive method for detecting early CRC, derived from the evaluation of three tumor specific DNA methylation markers in the blood. According to a study (52), the sensitivity of CRC cells to trimethoxy reached 78%, and the specificity was close to 100%. This method undoubtedly lights the way for the early prediction of CRC. Recently, according to Li et al (52), SEPT9, SDC2 and ALX4 methylation status can cover multiple molecular pathways of tumor formation, and combined detection will be hopeful to further improve the sensitivity of CRC detection.

Furthermore, a new combination of plasma DNA methylation-based biomarkers has now been developed and validated using clinical samples from multiple medical centers. It is expected to provide an alternative and cost-effective strategy for early detection of targeted gastrointestinal cancers (53).

Effect of DNA methylation on prognosis

Since the high status of MSI has been detected in numerous types of CRC, some scholars have proposed MSI status as a major marker of prognostic analysis (54). However, a meta-analysis has found that MSI-H is not a robust prognostic marker in stage I and stage IV CRC without immunotherapy (55). Interestingly, Popat et al (56) proposed MSI status as a prospective condition for patient management. Consequently, their research revealed that patients with MSI administered with fluorouracil exhibited better survival rates; however, explaining why MSI patients had an improved prognosis and the mechanism of Fu was a great challenge for later researchers. A previous study also posited that gene mutations such as those in the tumor suppressor gene TP53 and KRAS are rare in MSI tumors, and that these gene mutations could be associated with poor prognoses (57).

SMAD4 is a tumor suppressor and a component of the transforming growth factor (TGF)-β signaling pathway, which is associated with cell proliferation, differentiation, migration, and apoptosis. Methylation has been reported to lead to activation of the DPC4/SMAD4 gene, which plays an important role in the development of CRC. The study also pointed out that DPC4/SMAD4 is an important factor in prognostic analysis (58).

SDS2 methylation can be used as a potential biomarker to evaluate preoperative and postoperative fecal DNA in patients with CRC and can be used to determine whether methylated SDC2 in stool DNA returns to normal after surgical resection of CRC (59).

Relationship between DNA methylation and targeted therapy

CRC is characterized by hypermethylation and overall hypomethylation of gene promoters. However, DNA methylation is a double-edged sword. It cannot only affect the development of tumor but is also a favorable treatment site. In the present study, several well-studied methylation targeted therapy sites were summarized.

Cyclin-dependent kinase inhibitor p16INK4a

Methylation of p16INK4a can be detected in ~30% of patients with CRC (60), and the mutation of p16INK4a leads to BRAF mutation, which has been proved to be a direct cause of tumorigenesis in the intestinal tract of mice (61). Therefore, p16INK4a can inhibit tumorigenesis, and targeted demethylation could potentially be a novel treatment method.

RASSF1

Hypermethylation of the RASSF1 promoter can be detected in 80% of CRCs. Interestingly, it has been revealed that knockout of the RASSF1 gene in mice increases the risk of CRC, which suggests that RASSF1 may be a tumor suppressor gene (62).

Cell division cycle 7 (Cdc7)

Studies have identified somatic mutations of Cdc7 in CRC, and clinical studies are ongoing. These findings emphasize the potential of Cdc7 in targeted therapy (63).

DNA methylation is an important therapeutic target for CRC

Preclinical study: Effects of DNA methylation inhibitors on colorectal tumor cells

At the time of the formation of colon cancer, hypermethylation occurs at multiple gene sites on the DNA, resulting in the inactivation of important genes such as tumor suppressors. Therefore, DNA methyltransferase inhibitors can inhibit methylation at specific sites, correct wrong methylation modifications, and directly alter gene expression.

DNA methylation is a physiological process that regulates gene expression. The formation and maintenance of DNA methylation are realized under the action of DNMT. DNMT has three families, namely DNMT1, whose activity is accelerated during DNA semi-retention replication, and DNMT2 and DNMT3, which include DNMT3a, DNMT3b and DNMT3l. The human body possesses DNMT1, DNMT3a and DNMT3b.

DNMT3a and DNMT3b play important roles in catalytic de novo methylation during embryonic development. It has also been suggested that DNMT3a and DNMT3b may correct the errors left by DNMT1, as they are involved in maintaining DNA methylation patterns. The three DNMTs maintain DNA methylation and exhibit low expression in normal tissues and high expression in tumor tissues (64). Gradually, selecting DNMT as the target has become a new direction for drug research and development.

Herbal drugs

In recent years, Traditional Chinese Medicine (TCM) therapy has become increasingly popular. Since ancient times, there have been TCM compounds used to treat CRC, such as Wumei Wan and Sini Tang (65,66). Among them, numerous natural compounds have been revealed to treat CRC by acting on their methylated genes. Ginseng is one such case. It enhances apoptosis by regulating apoptosis-related genes in CRC cells and down-regulates the expression of DNMTs and reduces the global methylation level in CRC cells (67). Studies that have revealed promising results indicate the role of vitamins as DNA methylation modifiers, but studies with improved study designs are necessary (68).

A clinical study has revealed that 0.5–1 g of resveratrol administered orally daily produces anticancer effects in the human GI tract while being well tolerated in patients with cancer. It actually inhibits cancer spread by regulating epigenetic changes in tumor cells, through DNA methylation (69).

Curcumin from turmeric induces demethylation of specific CpG sites in CRC cells, but it does not induce global DNA methylation changes. Curcumin-induced methylation changes occur in a gene- and cell line-specific manner, and have a direct impact on the transcription of various genes involved in important biological processes (70). A number of experiments have revealed that some natural compounds can treat CRC through DNA methylation, proving that DNA methylation can be used as a target to treat CRC.

Inhibitor alone

There are two classes of DNA methyltransferase inhibitors: Nucleoside and non-nucleoside, wherein the former includes 5-azacytosine and its derivatives, and the latter includes polyphenol epigallocatechin-3-gallate and rgl08 (71).

Although a variety of compounds can inhibit DNA methylation in mammalian cells, such as genistein (GE) and curcumin (72,73), the only widely tested DNA methylase inhibitors are ZCyd (5-azacytidine), DZCyd (5,6-dihydro-5-azacytidine, also known as DHAC) and ZdCyd (decitabine). These three compounds can inhibit DNA methylation only when incorporated into DNA. As aforementioned, ZdCyt is a more effective DNA methylation inhibitor than ZCyd because it binds only with DNA. Additionally, ZCyd is synthesized as a more stable analogue of ZCyd (74), which is at least one order of magnitude weaker than ZdCyt in blocking methylation in vivo (75). This is attributed to the limited incorporation of ZdCyt in DNA because it is inefficiently phosphorylated by cytidine kinase. Data, however, revealed that CRC can be treated with ZCyd.

ZNF671 has been discovered to be an important cancer inhibitor in a variety of tumors. High methylation level of ZNF671 gene promoter region was discovered in CRC, which was negatively correlated with ZN671 expression. It functions as a tumor suppressor in CRC through inactivation of Notch signaling. This suggests that ZNF671 can be used as a candidate target for the treatment of CRC (76).

Clinical research

Changes in DNA methylation may even be an important mediator in the transformation of metastatic cancer: Dramatic and reproducible changes in methylation have been described in several important longitudinal studies. Such studies have been completed for CRC (77), prostate (78) and breast cancers (79). E-cadherin (CDH1) is widely considered to be the target of DNA methylation for breast cancer metastasis (80). It is worth noting that DNA methylation of the metastasis suppressor gene is observed in circulating tumor cells from various tumor types (81). This increases the possibility that DNA methylation may also be a useful prognostic marker for metastasis. It is noteworthy that a pan-cancer meta-analysis identified the methylation site associated with metastasis of breast cancer and CRC (82). Therefore, the potential of methylated DNA as a marker for prognosis, diagnosis and metastasis is an area to be further explored.

Histologically, normal colonic mucosal tissues can be distinguished from colonic tissue with precancerous lesions at the molecular level. Therefore, the change in DNA methylation can be used as a marker of colon cancer. However, the reversibility of DNA methylation also provides a theoretical basis for the treatment of CRC from the perspective of epigenetics, and research revealed that the analyses of the DNA methylation results of specific genes are expected to provide clinicians with useful information such as early diagnosis and disease stages. Although these DNA methylation inhibitors have a potent demethylation effect, they can be accompanied by strong cytotoxicity and lack of specificity in the selection of action sites. Therefore, more studies are needed to solve these problems to facilitate the widespread use of DNA methylation inhibitors in the clinic (Table II).

Table II.

DNA methylation is an important therapeutic target for CRC.

Table II.

DNA methylation is an important therapeutic target for CRC.

A, DNA methyltransferase inhibitors

DrugsMechanismsClinical applications(Refs.)
ZCyd (5-azacytidine, 5-AzaC)It downregulates the expression of DNMTs in CRC cells.AML, MDS, CML, CLL, pancreatic cancer, breast cancer, CRC. It is often used in combination with other drugs.(74,75)
DZCyd (5,6-dihydro-5-azacytidine, DHAC)DZCyt functions as a transition-state inhibitor by forming a tight, non-covalent complex with DNA C5-MTases.Leukemia, breast cancer, bronchial carcinoma, pancreatic cancer, CRC.(88)
ZdCyd (decitabine)It induces CpG demethylation.AML, MDS, CML, pancreatic cancer, breast cancer, CRC.(74,75)
RG108It binds to the SAM binding site on DNMT1 with its indole moiety.Hearing loss, lung adenocarcinoma, epilepsy, CRC.(89,90)
ZebularineIt inhibits DNA methyltransferases.Diabetes mellitus, liver cancer, stroke, breast cancer, lung cancer, CRC.(91)

B, Herbal drugs

GinsengIt has anti-tumor ability by down-regulating DNMTs to regulate cell apoptosis and reversing the methylation status of transcriptionally silenced genes in CRC.Diabetes mellitus, depression, epilepsy, liver cancer, CRC.(67)
CurcuminIt inhibits colon cancer cell growth by up-regulating DLEC1 and reducing CpG methylation.Lung cancer, bowel cancer, stomach cancer, breast cancer, leukemia, CRC.(92)
EGCGIt directly inhibits DNMT1.RA, Alzheimer's disease, breast cancer, CRC, lung cancer, prostate cancer, oral cancer.(93)

[i] CRC, colorectal cancer; DNMTs, DNA methyltransferases; AML, acute myeloid leukemia; MDS, myelodysplastic syndromes; CML, chronic myelogenous leukemia; CLL, chronic lymphocytic leukemia; CpG, cytosine guanine dinucleotide; SAM, S-adenosyl methionine; DLEC1, cilia and flagella associated protein; EGCG, epigallocatechin gallate; RA, rheumatoid arthritis.

Conclusions and outlook

The research prospect of DNA methylation and CRC is promising. It is considered by the authors that DNA methylation in genome sequences can be an important marker to predict the incidence and molecular typing of CRC and the prognosis of patients with cancer. Further experiments are required to make improved use of the DNA methylation spectrum. DNA methylation is expected to be an important site for CRC screening. High sensitivity is its biggest advantage, which helps to reduce the misdiagnosis rate. Therefore, it is important to clarify the methylation markers corresponding to different types of CRC through further in-depth research. Using methylation biomarkers to identify cancer cells should be a trend in the future. Reducing the damage caused by invasive examination of the body is also a major research direction.

Studies have revealed that CRC is a highly heterogeneous disease. For example, MLH1 gene hypermethylation is only discovered in a small proportion of sporadic CRC cases (83). This enables the individualized treatment of patients. However, each patient has considerable differences in drug sensitivity, effect of treatment and mortality. However, since epigenetics is reversible, that is, DNA methylation is reversible, it offers new strategies for CRC treatment. DNMT inhibitors are expected to become targeted drugs for the treatment of CRC, which can be used to target patients with hypermethylated CRC subtypes. For example, azacytidine, decitabine, temozolomide and other specific drugs can reverse the methylation state of DNA to achieve the purpose of targeted treatment of CRC; however, more clinical studies are required to demonstrate the sensitivity and effectiveness of these drugs on various CRC subtypes. Furthermore, the side effects of these drugs also have a significant impact on the prognosis. Clinically, according to the different symptoms of patients, a more appropriate treatment plan can be formulated, and symptomatic drugs can be used to improve the treatment effect. The best possible strategy would be to adapt the drugs to local conditions, reduce side effects, and improve tumor prognosis. The identification of metastatic potential signatures is complicated by the heterogeneity among tumor cells, and there is still a long way from this.

Immunotherapy is an emerging field in the treatment of CRC, and DNA methylation is likely to be related to the effect of immunotherapy. It has been reported that m6 methylation-mediated intercellular communication in the tumor microenvironment plays an antitumor immunomodulatory role (84); and ALKBH5, an RNA N6-methyladenosine eraser, can be used as a target to promote CRC immunotherapy (85). A recent study revealed that upregulated THBS2 expression in CRC cells inhibits antitumor immunity through HIF1A/lactic acid/GPR132 pathway. In addition, THBS2 expression is also related to PFI, immune cell infiltration and immune regulation (86). Moreover, some mouse experiments have revealed that the combination of epigenetics and immunotherapy is more effective than the use of blockers alone, which further demonstrates the feasibility of methylation immunotherapy (87). Scientists also continue to actively explore the role of methylation with immune escape and immunotherapy resistance to find new immunotherapy strategies. This helps to improve the cure rate of patients.

DNA methylation has a broad application prospect in the field of CRC research. Through in-depth study of the mechanism and regulatory network of methylation in CRC, it is expected to provide new breakthrough points for early diagnosis, individualized treatment and immunotherapy of CRC, thereby improving the prognosis and survival rate of patients.

According to the present study, DNA methylation can be used as a biomarker for early diagnosis of CRC. By detecting the methylation status of DNA in feces, minimal residual disease can be detected, which is helpful for early detection of the disease. In addition, DNA methylation also plays an important role in the prognosis of CRC. The survival rate and quality of life of the patients were tested by detecting the methylation status. With the development of sequencing technology and bioinformatics, the study of DNA methylation in CRC will be more in-depth. In conclusion, DNA methylation still has great potential in the study of CRC and provides more possibilities for the treatment of CRC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Dalian Medical University Affiliated Second Hospital Subspecialty Fund (grant no. 84671291).

Availability of data and materials

Not applicable.

Authors' contributions

LW and LS conceptualized, wrote, reviewed and edited the manuscript. YW, CW and RZ prepared and wrote the original draft. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

CMS

common molecular subtypes

CIMP

CpG island methylation phenotype

SAM

s-adenosylmethionine

DNMT

DNA methyltransferase

CIN

chromosomal instability

MSI

microsatellite instability

COX-2

cyclooxygenase 2

TCM

Traditional Chinese Medicine

References

1 

Migliore L, Migheli F, Spisni R and Coppedè F: Genetics, cytogenetics, and epigenetics of colorectal cancer. J Biomed Biotechnol. 2011:7923622011.PubMed/NCBI

2 

Araghi M, Soerjomataram I, Bardot A, Ferlay J, Cabasag CJ, Morrison DS, De P, Tervonen H, Walsh PM, Bucher O, et al: Changes in colorectal cancer incidence in seven high-income countries: A population-based study. Lancet Gastroenterol Hepatol. 4:511–518. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Reilly NM, Novara L, Di Nicolantonio F and Bardelli A: Exploiting DNA repair defects in colorectal cancer. Mol Oncol. 13:681–700. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Liebl MC and Hofmann TG: The role of p53 signaling in colorectal cancer. Cancers (Basel). 13:21252021. View Article : Google Scholar : PubMed/NCBI

5 

Bose S, Saha S, Goswami H, Shanmugam G and Sarkar K: Involvement of CCCTC-binding factor in epigenetic regulation of cancer. Mol Biol Rep. 50:10383–10398. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Jamai D, Gargouri R, Selmi B and Khabir A: ERCC1 and MGMT methylation as a predictive marker of relapse and FOLFOX response in colorectal cancer patients from South Tunisia. Genes (Basel). 14:14672023. View Article : Google Scholar : PubMed/NCBI

7 

Okada Y, Peng F, Perea J, Corchete L, Bujanda L, Li W and Goel A: Genome-wide methylation profiling identifies a novel gene signature for patients with synchronous colorectal cancer. Br J Cancer. 128:112–120. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Okano M, Bell DW, Haber DA and Li E: DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 99:247–257. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Zhai P, Zhang H, Li Q, Yang M, Guo Y and Xing C: DNMT1-mediated NR3C1 DNA methylation enables transcription activation of connexin40 and augments angiogenesis during colorectal cancer progression. Gene. 892:1478872024. View Article : Google Scholar : PubMed/NCBI

10 

Christman JK, Sheikhnejad G, Dizik M, Abileah S and Wainfan E: Reversibility of changes in nucleic acid methylation and gene expression induced in rat liver by severe dietary methyl deficiency. Carcinogenesis. 14:551–557. 1993. View Article : Google Scholar : PubMed/NCBI

11 

Goll MG, Kirpekar F, Maggert KA, Yoder JA, Hsieh CL, Zhang X, Golic KG, Jacobsen SE and Bestor TH: Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science. 311:395–398. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Smith ZD and Meissner A: DNA methylation: Roles in mammalian development. Nat Rev Genet. 14:204–220. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Cho HY, Wang X, Campbell MR, Panduri V, Coviello S, Caballero MT, Bennett BD, Kleeberger SR, Polack FP, Ofman G and Bell DA: Prospective epigenome and transcriptome analyses of cord and peripheral blood from preterm infants at risk of bronchopulmonary dysplasia. Sci Rep. 13:122622023. View Article : Google Scholar : PubMed/NCBI

14 

Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, et al: The consensus molecular subtypes of colorectal cancer. Nat Med. 21:1350–1356. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B and Kinzler KW: APC mutations occur early during colorectal tumorigenesis. Nature. 359:235–237. 1992. View Article : Google Scholar : PubMed/NCBI

16 

Cancer Genome Atlas Network, . Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Lin CY, Shen MY, Chen WTL and Yang CA: Evaluation of the prognostic value of low-frequency KRAS mutation detection in circulating tumor DNA of patients with metastatic colorectal cancer. J Pers Med. 13:10512023. View Article : Google Scholar : PubMed/NCBI

18 

Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B and Velculescu VE: Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature. 418:9342002. View Article : Google Scholar : PubMed/NCBI

19 

Tsujii M, Kawano S and DuBois RN: Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci USA. 94:3336–3340. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Hidalgo-Estévez AM, Stamatakis K, Jiménez-Martínez M, López-Pérez R and Fresno M: Cyclooxygenase 2-regulated genes an alternative avenue to the development of new therapeutic drugs for colorectal cancer. Front Pharmacol. 11:5332020. View Article : Google Scholar : PubMed/NCBI

21 

Samowitz WS, Slattery ML, Sweeney C, Herrick J, Wolff RK and Albertsen H: APC mutations and other genetic and epigenetic changes in colon cancer. Mol Cancer Res. 5:165–170. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Pruitt K and Der CJ: Ras and Rho regulation of the cell cycle and oncogenesis. Cancer Lett. 171:1–10. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Tozaki Y, Aoki H, Kato R, Toriuchi K, Arame S, Inoue Y, Hayashi H, Kubota E, Kataoka H and Aoyama M: The combination of ATM and Chk1 inhibitors induces synthetic lethality in colorectal cancer cells. Cancers (Basel). 15:7352023. View Article : Google Scholar : PubMed/NCBI

24 

Lai WL, Lee SC, Chang KF, Huang XF, Li CY, Lee CJ, Wu CY, Hsu HJ and Tsai NM: Juniperus communis extract induces cell cycle arrest and apoptosis of colorectal adenocarcinoma in vitro and in vivo. Braz J Med Biol Res. 54:e108912021. View Article : Google Scholar : PubMed/NCBI

25 

Zhou Y, Dai W, Wang H, Pan H and Wang Q: Long non-coding RNA CASP5 promotes the malignant phenotypes of human glioblastoma multiforme. Biochem Biophys Res Commun. 500:966–972. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Gönenc II, Wolff A, Schmidt J, Zibat A, Müller C, Cyganek L, Argyriou L, Räschle M, Yigit G and Wollnik B: Single-cell transcription profiles in Bloom syndrome patients link BLM deficiency with altered condensin complex expression signatures. Hum Mol Genet. 31:2185–2193. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Bader S, Walker M, Hendrich B, Bird A, Bird C, Hooper M and Wyllie A: Somatic frameshift mutations in the MBD4 gene of sporadic colon cancers with mismatch repair deficiency. Oncogene. 18:8044–8047. 1999. View Article : Google Scholar : PubMed/NCBI

28 

De Palma FDE, D'Argenio V, Pol J, Kroemer G, Maiuri MC and Salvatore F: The molecular hallmarks of the serrated pathway in colorectal cancer. Cancers (Basel). 11:10172019. View Article : Google Scholar : PubMed/NCBI

29 

Rajamäki K, Taira A, Katainen R, Välimäki N, Kuosmanen A, Plaketti RM, Seppälä TT, Ahtiainen M, Wirta EV, Vartiainen E, et al: Genetic and epigenetic characteristics of inflammatory bowel disease-associated colorectal cancer. Gastroenterology. 161:592–607. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Pajares MJ, Palanca-Ballester C, Urtasun R, Alemany-Cosme E, Lahoz A and Sandoval J: Methods for analysis of specific DNA methylation status. Methods. 187:3–12. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H and Jaenisch R: Induction of tumors in mice by genomic hypomethylation. Science. 300:489–492. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Takeuchi C, Yamashita S, Liu YY, Takeshima H, Sasaki A, Fukuda M, Hashimoto T, Naka T, Ishizu K, Sekine S, et al: Precancerous nature of intestinal metaplasia with increased chance of conversion and accelerated DNA methylation. Gut. 73:255–267. 2024. View Article : Google Scholar : PubMed/NCBI

33 

Oshima M, Murai N, Kargman S, Arguello M, Luk P, Kwong E, Taketo MM and Evans JF: Chemoprevention of intestinal polyposis in the Apcdelta716 mouse by rofecoxib, a specific cyclooxygenase-2 inhibitor. Cancer Res. 61:1733–1740. 2001.PubMed/NCBI

34 

Xu X, Nie J, Lu L, Du C, Meng F and Song D: LINC00337 promotes tumor angiogenesis in colorectal cancer by recruiting DNMT1, which suppresses the expression of CNN1. Cancer Gene Ther. 28:1285–1297. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Hu YH, Ma S, Zhang XN, Zhang ZY, Zhu HF, Ji YH, Li J, Qian XL and Wang YX: Hypermethylation of ADHFE1 promotes the proliferation of colorectal cancer cell via modulating cell cycle progression. Onco Targets Ther. 12:8105–8115. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Suzuki H, Watkins DN, Jair KW, Schuebel KE, Markowitz SD, Chen WD, Pretlow TP, Yang B, Akiyama Y, Van Engeland M, et al: Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet. 36:417–422. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Jafarpour S, Yazdi M, Nedaeinia R, Vatandoost N, Ferns GA and Salehi R: Status of integrin subunit alpha 4 promoter DNA methylation in colorectal cancer and other malignant tumors: A systematic review and meta-analysis. Res Pharm Sci. 18:231–243. 2023. View Article : Google Scholar : PubMed/NCBI

38 

Wang Y, Zhou J, Zhang J, Cao H, Han F, Zhang H and Xu E: The expression of ADAMTS14 is regulated by promoter DNA methylation and is associated with poor prognosis in colorectal cancer. Exp Cell Res. 410:1129532022. View Article : Google Scholar : PubMed/NCBI

39 

Kim YI, Pogribny IP, Basnakian AG, Miller JW, Selhub J, James SJ and Mason JB: Folate deficiency in rats induces DNA strand breaks and hypomethylation within the p53 tumor suppressor gene. Am J Clin Nutr. 65:46–52. 1997. View Article : Google Scholar : PubMed/NCBI

40 

Kim YI, Christman JK, Fleet JC, Cravo ML, Salomon RN, Smith D, Ordovas J, Selhub J and Mason JB: Moderate folate deficiency does not cause global hypomethylation of hepatic and colonic DNA or c-myc-specific hypomethylation of colonic DNA in rats. Am J Clin Nutr. 61:1083–1090. 1995. View Article : Google Scholar : PubMed/NCBI

41 

Marugame T, Tsuji E, Kiyohara C, Eguchi H, Oda T, Shinchi K and Kono S: Relation of plasma folate and methylenetetrahydrofolate reductase C677T polymorphism to colorectal adenomas. Int J Epidemiol. 32:64–66. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Othman R, Mohtarrudin N, Ahmad Zubir NM, Seow HF, Ngan KW and Osman M: HER3 overexpression and hypomethylation in colorectal adenocarcinoma. Malays J Pathol. 44:67–74. 2022.PubMed/NCBI

43 

Timar J and Kashofer K: Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev. 39:1029–1038. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Santini D, Loupakis F, Vincenzi B, Floriani I, Stasi I, Canestrari E, Rulli E, Maltese PE, Andreoni F, Masi G, et al: High concordance of KRAS status between primary colorectal tumors and related metastatic sites: Implications for clinical practice. Oncologist. 13:1270–1275. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Lièvre A, Bachet JB, Boige V, Cayre A, Le Corre D, Buc E, Ychou M, Bouché O, Landi B, Louvet C, et al: KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 26:374–379. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Wong CC, Xu J, Bian X, Wu JL, Kang W, Qian Y, Li W, Chen H, Gou H, Liu D, et al: In colorectal cancer cells with mutant KRAS, SLC25A22-mediated glutaminolysis reduces DNA demethylation to increase wnt signaling, stemness, and drug resistance. Gastroenterology. 159:2163–2180.e6. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Mangelinck A and Mann C: DNA methylation and histone variants in aging and cancer. Int Rev Cell Mol Biol. 364:1–110. 2021. View Article : Google Scholar : PubMed/NCBI

48 

Sakai E, Nakajima A and Kaneda A: Accumulation of aberrant DNA methylation during colorectal cancer development. World J Gastroenterol. 20:978–987. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Kasprzak A: Prognostic biomarkers of cell proliferation in colorectal cancer (CRC): From immunohistochemistry to molecular biology techniques. Cancers (Basel). 15:45702023. View Article : Google Scholar : PubMed/NCBI

50 

Hinoue T, Weisenberger DJ, Lange CPE, Shen H, Byun HM, Van Den Berg D, Malik S, Pan F, Noushmehr H, van Dijk CM, et al: Genome-scale analysis of aberrant DNA methylation in colorectal cancer. Genome Res. 22:271–282. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Jensen SØ, Øgaard N, Ørntoft MW, Rasmussen MH, Bramsen JB, Kristensen H, Mouritzen P, Madsen MR, Madsen AH, Sunesen KG, et al: Novel DNA methylation biomarkers show high sensitivity and specificity for blood-based detection of colorectal cancer-a clinical biomarker discovery and validation study. Clin Epigenetics. 11:1582019. View Article : Google Scholar : PubMed/NCBI

52 

Li Y, Li B, Jiang R, Liao L, Zheng C, Yuan J, Zeng L, Hu K, Zhang Y, Mei W, et al: A novel screening method of DNA methylation biomarkers helps to improve the detection of colorectal cancer and precancerous lesions. Cancer Med. 12:20626–20638. 2023. View Article : Google Scholar : PubMed/NCBI

53 

Dai Y, Li H, Wu Q, Wang J, Wang K, Fei S, Pei B, Song L, Chen G, Ma Y, et al: A sensitive and robust plasma-based DNA methylation panel for early detection of target gastrointestinal cancers. Neoplasia. 46:1009412023. View Article : Google Scholar : PubMed/NCBI

54 

Benatti P, Gafà R, Barana D, Marino M, Scarselli A, Pedroni M, Maestri I, Guerzoni L, Roncucci L, Menigatti M, et al: Microsatellite instability and colorectal cancer prognosis. Clin Cancer Res. 11:8332–8340. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Toh JWT, Phan K, Reza F, Chapuis P and Spring KJ: Rate of dissemination and prognosis in early and advanced stage colorectal cancer based on microsatellite instability status: Systematic review and meta-analysis. Int J Colorectal Dis. 36:1573–1596. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Popat S, Hubner R and Houlston RS: Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol. 23:609–618. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Klump B, Nehls O, Okech T, Hsieh CJ, Gaco V, Gittinger FS, Sarbia M, Borchard F, Greschniok A, Gruenagel HH, et al: Molecular lesions in colorectal cancer: Impact on prognosis? Original data and review of the literature. Int J Colorectal Dis. 19:23–42. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Koyama M, Ito M, Nagai H, Emi M and Moriyama Y: Inactivation of both alleles of the DPC4/SMAD4 gene in advanced colorectal cancers: Identification of seven novel somatic mutations in tumors from Japanese patients. Mutat Res. 406:71–77. 1999.PubMed/NCBI

59 

Song JH, Oh TJ, An S, Lee KH, Kim JY and Kim JS: Comparative detection of syndecan-2 methylation in preoperative and postoperative stool DNA in patients with colorectal cancer. World J Gastrointest Surg. 15:2032–2041. 2023. View Article : Google Scholar : PubMed/NCBI

60 

Shima K, Nosho K, Baba Y, Cantor M, Meyerhardt JA, Giovannucci EL, Fuchs CS and Ogino S: Prognostic significance of CDKN2A (p16) promoter methylation and loss of expression in 902 colorectal cancers: Cohort study and literature review. Int J Cancer. 128:1080–1094. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Carragher LAS, Snell KR, Giblett SM, Aldridge VS, Patel B, Cook SJ, Winton DJ, Marais R and Pritchard CA: V600EBraf induces gastrointestinal crypt senescence and promotes tumour progression through enhanced CpG methylation of p16INK4a. EMBO Mol Med. 2:458–471. 2010. View Article : Google Scholar : PubMed/NCBI

62 

van der Weyden L, Arends MJ, Dovey OM, Harrison HL, Lefebvre G, Conte N, Gergely FV, Bradley A and Adams DJ: Loss of rassf1a cooperates with Apc(Min) to accelerate intestinal tumourigenesis. Oncogene. 27:4503–4508. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Melling N, Muth J, Simon R, Bokemeyer C, Terracciano L, Sauter G, Izbicki JR and Marx AH: Cdc7 overexpression is an independent prognostic marker and a potential therapeutic target in colorectal cancer. Diagn Pathol. 10:1252015. View Article : Google Scholar : PubMed/NCBI

64 

el-Deiry WS, Nelkin BD, Celano P, Yen RW, Falco JP, Hamilton SR and Baylin SB: High expression of the DNA methyltransferase gene characterizes human neoplastic cells and progression stages of colon cancer. Proc Natl Acad Sci USA. 88:3470–3474. 1991. View Article : Google Scholar : PubMed/NCBI

65 

Lu ZH, Ding Y, Wang YJ, Chen C, Yao XR, Yuan XM, Bu F, Bao H, Dong YW, Zhou Q, et al: Early administration of Wumei Wan inhibit myeloid-derived suppressor cells via PI3K/Akt pathway and amino acids metabolism to prevent colitis-associated colorectal cancer. J Ethnopharmacol. 333:1182602024.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

66 

Chen J, Zheng X, Xu G, Wang B, Hu L, Mao J, Lu X, Cai Y, Chai K and Chen W: Sini decoction inhibits tumor progression and enhances the anti-tumor immune response in a murine model of colon cancer. Comb Chem High Throughput Screen. 26:2517–2526. 2023. View Article : Google Scholar : PubMed/NCBI

67 

Okuno K, Pratama MY, Li J, Tokunaga M, Wang X, Kinugasa Y and Goel A: Ginseng mediates its anticancer activity by inhibiting the expression of DNMTs and reactivating methylation-silenced genes in colorectal cancer. Carcinogenesis. 44:394–403. 2023. View Article : Google Scholar : PubMed/NCBI

68 

Boughanem H, Kompella P, Tinahones FJ and Macias-Gonzalez M: An overview of vitamins as epidrugs for colorectal cancer prevention. Nutr Rev. 81:455–479. 2023. View Article : Google Scholar : PubMed/NCBI

69 

Brockmueller A, Sajeev A, Koklesova L, Samuel SM, Kubatka P, Büsselberg D, Kunnumakkara AB and Shakibaei M: Resveratrol as sensitizer in colorectal cancer plasticity. Cancer Metastasis Rev. 43:55–85. 2024. View Article : Google Scholar : PubMed/NCBI

70 

Link A, Balaguer F, Shen Y, Lozano JJ, Leung HC, Boland CR and Goel A: Curcumin modulates DNA methylation in colorectal cancer cells. PLoS One. 8:e577092013. View Article : Google Scholar : PubMed/NCBI

71 

Lopez M, Gilbert J, Contreras J, Halby L and Arimondo PB: Inhibitors of DNA methylation. Adv Exp Med Biol. 1389:471–513. 2022. View Article : Google Scholar : PubMed/NCBI

72 

Fabianowska-Majewska K, Kaufman-Szymczyk A, Szymanska-Kolba A, Jakubik J, Majewski G and Lubecka K: Curcumin from turmeric rhizome: A potential modulator of DNA methylation machinery in breast cancer inhibition. Nutrients. 13:3322021. View Article : Google Scholar : PubMed/NCBI

73 

Sharma M and Tollefsbol TO: Combinatorial epigenetic mechanisms of sulforaphane, genistein and sodium butyrate in breast cancer inhibition. Exp Cell Res. 416:1131602022. View Article : Google Scholar : PubMed/NCBI

74 

Futterman B, Derr J, Beisler JA, Abbasi MM and Voytek P: Studies on the cytostatic action, phosphorylation and deamination of 5-azacytidine and 5,6-dihydro-5-azacytidine in HeLa cells. Biochem Pharmacol. 27:907–909. 1978. View Article : Google Scholar : PubMed/NCBI

75 

Ghanim V, Herrmann H, Heller G, Peter B, Hadzijusufovic E, Blatt K, Schuch K, Cerny-Reiterer S, Mirkina I, Karlic H, et al: 5-Azacytidine and decitabine exert proapoptotic effects on neoplastic mast cells: role of FAS-demethylation and FAS re-expression, and synergism with FAS-ligand. Blood. 119:4242–4252. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Wang Y, Chen FR, Wei CC, Sun LL, Liu CY, Yang LB and Guo XY: Zinc finger protein 671 has a cancer-inhibiting function in colorectal carcinoma via the deactivation of Notch signaling. Toxicol Appl Pharmacol. 458:1163262023. View Article : Google Scholar : PubMed/NCBI

77 

Bhullar DS, Barriuso J, Mullamitha S, Saunders MP, O'Dwyer ST and Aziz O: Biomarker concordance between primary colorectal cancer and its metastases. EBioMedicine. 40:363–374. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Guo H, Vuille JA, Wittner BS, Lachtara EM, Hou Y, Lin M, Zhao T, Raman AT, Russell HC, Reeves BA, et al: DNA hypomethylation silences anti-tumor immune genes in early prostate cancer and CTCs. Cell. 186:2765–2782.e28. 2023. View Article : Google Scholar : PubMed/NCBI

79 

Gkountela S, Castro-Giner F, Szczerba BM, Vetter M, Landin J, Scherrer R, Krol I, Scheidmann MC, Beisel C, Stirnimann CU, et al: Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding. Cell. 176:98–112.e14. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Padmanaban V, Krol I, Suhail Y, Szczerba BM, Aceto N, Bader JS and Ewald AJ: E-cadherin is required for metastasis in multiple models of breast cancer. Nature. 573:439–444. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Warton K and Samimi G: Methylation of cell-free circulating DNA in the diagnosis of cancer. Front Mol Biosci. 2:132015. View Article : Google Scholar : PubMed/NCBI

82 

Zhou X, Yu L, Wang L, Xiao J, Sun J, Zhou Y, Xu X, Xu W, Spiliopoulou A, Timofeeva M, et al: Alcohol consumption, blood DNA methylation and breast cancer: A Mendelian randomisation study. Eur J Epidemiol. 37:701–712. 2022. View Article : Google Scholar : PubMed/NCBI

83 

Gaiani F, Marchesi F, Negri F, Greco L, Malesci A, de'Angelis GL and Laghi L: Heterogeneity of colorectal cancer progression: Molecular gas and brakes. Int J Mol Sci. 22:52462021. View Article : Google Scholar : PubMed/NCBI

84 

Bao Y, Zhai J, Chen H, Wong CC, Liang C, Ding Y, Huang D, Gou H, Chen D, Pan Y, et al: Targeting m6A reader YTHDF1 augments antitumour immunity and boosts anti-PD-1 efficacy in colorectal cancer. Gut. 72:1497–1509. 2023. View Article : Google Scholar : PubMed/NCBI

85 

Zhai J, Chen H, Wong CC, Peng Y, Gou H, Zhang J, Pan Y, Chen D, Lin Y, Wang S, et al: ALKBH5 drives immune suppression via targeting AXIN2 to promote colorectal cancer and is a target for boosting immunotherapy. Gastroenterology. 165:445–462. 2023. View Article : Google Scholar : PubMed/NCBI

86 

Liu Y, Jiang C, Xu C and Gu L: Systematic analysis of integrated bioinformatics to identify upregulated THBS2 expression in colorectal cancer cells inhibiting tumour immunity through the HIF1A/Lactic Acid/GPR132 pathway. Cancer Cell Int. 23:2532023. View Article : Google Scholar : PubMed/NCBI

87 

Yang L, Chen X, Lee C, Shi J, Lawrence EB, Zhang L, Li Y, Gao N, Jung SY, Creighton CJ, et al: Functional characterization of age-dependent p16 epimutation reveals biological drivers and therapeutic targets for colorectal cancer. J Exp Clin Cancer Res. 42:1132023. View Article : Google Scholar : PubMed/NCBI

88 

Sheikhnejad G, Brank A, Christman JK, Goddard A, Alvarez E, Ford H Jr, Marquez VE, Marasco CJ, Sufrin JR, O'Gara M and Cheng X: Mechanism of inhibition of DNA (cytosine C5)-methyltransferases by oligodeoxyribonucleotides containing 5,6-dihydro-5-azacytosine. J Mol Biol. 285:2021–2034. 1999. View Article : Google Scholar : PubMed/NCBI

89 

Zheng Z, Zeng S, Liu C, Li W, Zhao L, Cai C, Nie G and He Y: The DNA methylation inhibitor RG108 protects against noise-induced hearing loss. Cell Biol Toxicol. 37:751–771. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Ou Y, Zhang Q, Tang Y, Lu Z, Lu X, Zhou X and Liu C: DNA methylation enzyme inhibitor RG108 suppresses the radioresistance of esophageal cancer. Oncol Rep. 39:993–1002. 2018.PubMed/NCBI

91 

Tanaka S, Hosokawa M, Matsumura J, Matsubara E, Kobori A, Ueda K and Iwakawa S: Effects of zebularine on invasion activity and intracellular expression level of let-7b in colorectal cancer cells. Biol Pharm Bull. 40:1320–1325. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Guo Y, Shu L, Zhang C, Su ZY and Kong ANT: Curcumin inhibits anchorage-independent growth of HT29 human colon cancer cells by targeting epigenetic restoration of the tumor suppressor gene DLEC1. Biochem Pharmacol. 94:69–78. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Singh BN, Shankar S and Srivastava RK: Green tea catechin, epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and clinical applications. Biochem Pharmacol. 82:1807–1821. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2024
Volume 30 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Wang C, Zhong R, Wang L and Sun L: Research progress of DNA methylation in colorectal cancer (Review). Mol Med Rep 30: 154, 2024
APA
Wang, Y., Wang, C., Zhong, R., Wang, L., & Sun, L. (2024). Research progress of DNA methylation in colorectal cancer (Review). Molecular Medicine Reports, 30, 154. https://doi.org/10.3892/mmr.2024.13278
MLA
Wang, Y., Wang, C., Zhong, R., Wang, L., Sun, L."Research progress of DNA methylation in colorectal cancer (Review)". Molecular Medicine Reports 30.3 (2024): 154.
Chicago
Wang, Y., Wang, C., Zhong, R., Wang, L., Sun, L."Research progress of DNA methylation in colorectal cancer (Review)". Molecular Medicine Reports 30, no. 3 (2024): 154. https://doi.org/10.3892/mmr.2024.13278