Next Article in Journal
The Effect of Mycotoxins and Silymarin on Liver Lipidome of Mice with Non-Alcoholic Fatty Liver Disease
Next Article in Special Issue
CD44: A Multifunctional Mediator of Cancer Progression
Previous Article in Journal
Real-World Diagnostic Accuracy and Use of Immunohistochemical Markers in Lung Cancer Diagnostics
Previous Article in Special Issue
New Insights on Old Biomarkers Involved in Tumor Microenvironment Changes and Their Diagnostic Relevance in Non-Small Cell Lung Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Methylation Biomarkers for Colorectal Cancer Prognosis

Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile
*
Authors to whom correspondence should be addressed.
Biomolecules 2021, 11(11), 1722; https://doi.org/10.3390/biom11111722
Submission received: 15 September 2021 / Revised: 3 November 2021 / Accepted: 9 November 2021 / Published: 19 November 2021
(This article belongs to the Special Issue Biomarkers in Cancer)

Abstract

:
Colorectal cancer (CRC) comprises the third most common cancer worldwide and the second regarding number of deaths. In order to make a correct and early diagnosis to predict metastasis formation, biomarkers are an important tool. Although there are multiple signaling pathways associated with cancer progression, the most recognized are the MAPK pathway, p53 pathway, and TGF-β pathway. These pathways regulate many important functions in the cell, such as cell cycle regulation, proliferation, differentiation, and metastasis formation, among others. Changes in expression in genes belonging to these pathways are drivers of carcinogenesis. Often these expression changes are caused by mutations; however, epigenetic changes, such as DNA methylation, are increasingly acknowledged to play a role in the deregulation of oncogenic genes. This makes DNA methylation changes an interesting biomarkers in cancer. Among the newly identified biomarkers for CRC metastasis INHBB, SMOC2, BDNF, and TBRG4 are included, all of which are highly deregulated by methylation and closely associated with metastasis. The identification of such biomarkers in metastasis of CRC may allow a better treatment and early identification of cancer formation in order to perform better diagnostics and improve the life expectancy.

1. Introduction

Colorectal cancer (CRC) is the third most common cancer worldwide and the second most common regarding global cancer-related deaths [1]. Most CRC cases occur sporadically, with risk factors including environmental and food-borne mutagens as well as chronic intestinal inflammation [2,3,4]. Hereditary factors are only found in a minority of cases but are nevertheless important to consider [5,6,7]. Most cases of CRC are thought to occur through a stepwise accumulation of mutations, encompassing the repression of tumor suppressor genes and activation of oncogenes [8]. At the time of diagnosis, 20% of the patients, with CRC, are presenting metastasis. In addition, 35–45% of the patients with localized CRC show recurrence within 5 years [9]. Metastasis usually results in conventional therapies becoming inadequate, and in consequence is associated with a poor 5-years survival rate of <10% [10,11]. It is still unclear what exactly causes a carcinoma to metastasize, although recent studies have associated key genes such as APC, K-RAS, and SMAD4, among others in CRC metastasis formation [12]. Understanding the metastasis process will allow the identification of molecular biomarkers, which are important tools for early diagnosis and prognosis of CRC, as early prognosis is currently the most successful and cost-effective way to enhance survival rate of CRC patients.
In the last decades, cancer diagnosis and prognosis at early stages have improved resulting in an increase of cure rates. New screening tools have been developed, focusing on early detection of cancer [8,9]. Biomarkers are used to distinguish between normal or abnormal (cancer) status in patients and to provide relevant information about disease diagnosis, prognosis, prediction, stratification, and therapy monitoring and improving treatment in cancer patients [10]. Usually, biomarkers are referring to DNA mutations, RNA or microRNA expression changes, epigenetic changes, and changes of metabolite abundance [13,14,15]. Their usages depend on multiple factors, including patients, stages of disease, and therapy/treatment. One of the largest challenges in medical diagnosis at the moment is to understand the molecular mechanisms of metastasis formation. This will aid the discovery of new and better ways to predict and diagnostic early metastasis events. One new and interesting method uses tumor budding as a predictive biomarker for distant metastasis. However, the scoring system needs to be optimized for it to be reliably used and it requires invasive colonoscopy [16]. Many studies focus on biomarkers that can be identified in blood plasma or stool samples, as these can be obtained in a less invasive way, while still having high sensitivity and accuracy to diagnose CRC and metastasis While several biomarkers for CRC diagnosis are available, it is not that easy to diagnose or predict metastasis. Identifying epigenetic changes, such as DNA methylation changes that are specific for metastasis, may help the diagnosis and prognosis of CRC [17].
The use of biomarkers has become robust and efficient method to evaluate potential abnormal biological processes such as cancer [14]. Using DNA methylation changes as biomarkers depends on the sensitivity of the detection method to distinguish between unmethylated cytosines from methylated cytosines. A tumor sheds cells, which are degraded, and DNA fragments from these cells can be detected in the blood or stool of patients. However, as these fragments only form a tiny fraction of the total DNA found in blood and stool, both the biomarker and method need to be carefully selected. The biomarker needs to show a very high methylation change (for example 0% methylation in normal tissue and 100% methylation in tumor tissue). The change must also be very homogeneous across tumor cells and patients. The method is preferably quantitative, such as qPCR, rather than qualitative like regular PCR, to detect methylation changes, as only a small fraction of DNA will show the change. One of the most common ways to detect DNA methylation changes is using bisulfite conversion of DNA. Treating DNA with bisulfite leads to the deamination of cytosines to uracil, while methylated cytosines remain unchanged [18]. The methods to detect DNA methylation in cancer are constantly evolving, trying to solve limitations associated with DNA quantification and repeatability [19].
In this review, we describe our current understanding of the mechanisms leading to colorectal cancer and metastasis formation and discuss the role of DNA methylation in these pathways. We show how our increasing knowledge in these processes has led to the identification of biomarkers for the prognosis of CRC and metastasis. We present and discuss novel biomarkers that have the potential to change our treatment strategies for CRC in the future.

2. Search Strategy

The literature search was performed with PubMed using the following search terms: “Cancer”, “Colorectal Cancer”, “Biological Markers”, “Biomarkers”, “DNA methylation”, and “Methylation marker”. Animal studies were excluded, as well articles and reviews written in languages other than English. The search yielded a total of 1245 papers, of which 306 were found relevant. These 306 articles were further selected after full-text reading, leaving 297 articles. Based on our inclusion and exclusion criteria, another 209 articles were eliminated, resulting in 88 articles selected for this review (Figure 1). The novel biomarkers discussed in Section 6 were selected based on the following criteria: (1) multiple studies have highlighted their deregulation in CRC and other cancers, (2) studies have associated their deregulation to methylation changes, and (3) their deregulation has been linked to poor prognosis and metastasis formation. This list is a selection of interesting candidates and by no means exhaustive.

3. Insights into Colorectal Carcinogenesis

In the majority of CRC cases (83%) [20], the process of adenoma formation is initiated by a loss-of-function mutation genes such as tumor suppressor gene TP53, oncogenes KRAS and PIK3CA [13], or APC. The loss of APC function leads to activation of the WNT signaling pathway [21]. The canonical Wnt pathway is normally regulated through to the phosphorylation of β-catenin by the “destruction complex” composed of APC, glycogen synthase kinase 3β (GSK3β), Axin, and casein kinase 1 (CK1). Phosphorylation marks β-catenin for subsequent ubiquitination and degradation in the proteosome. In absence of functional APC, β-catenin is not degraded, allowing for the translocation of β-catenin to the nucleus, where it activates tumorigenic genes [22,23].
CRC formation is usually driven by the activation of three well-characterized pathways: the MAPK pathway, p53 pathway, and TGF-β pathway [9]. The MAPK pathway plays a crucial role in the regulation of cell proliferation, differentiation, and stress response [24]. There are several MAPK protein family members, which can trigger different disorders [25]. The p53 pathway acts in response to cellular stress, targeting genes involved in apoptosis, cell-cycle arrest, DNA repair, and senescence [18,19,20,21]. The p53 pathway can be activated by at least three different mechanisms [26,27]: DNA damage [28,29], oncogene expression such RAS or Myc [30], and chemotherapeutic drugs or protein-kinase inhibitors [29,31]. TGF-β corresponds to a superfamily of cytokines composed of 33 members [32], comprising TGF-β1, -β2, and -β3, as well as activins, inhibins, growth differentiation factors (GDFs), and bone morphogenic proteins (BMPs) [33,34]. Aberrant activation of TGF-β results in cell-cycle modifications [35] and is involved in immune suppression, growth inhibition, epithelial-to-mesenchymal transition (EMT), and cell migration, among others [34]. These effects are caused by the three isoforms of TGF-β through two possible mechanisms, Smad and non-Smad signaling [36]. TGF-β ligands bind to their corresponding receptors, which marks to beginning of a signaling cascade, which is initiated by type II receptors (TβRII), functioning as an activator of the type I receptor (TβRI), by phosphorylating its glycine/serine-rich GS domain. When activated, TβRI transduces the signal downstream [32,36] through Smad signaling. Smad proteins are classified into three groups: R-Smads, Co-Smad, and I-Smad. Receptor-activated Smads (R-Smads) are comprised of Smad-1-5-8 and Smad-2-3 and are phosphorylated and activated by different receptors, including TβR, ActR, and BMPR, [37]. The second group has the common mediator Smad (Co-Smad or Smad4), which binds and forms a complex with R-Smads and they translocate to the nucleus, where they bind to other transcriptional receptors to enhance their DNA affinity. The third group are Smad inhibitors (I-Smad) and include Smad-6, which inhibits BMP signaling, and Smad-7, which can inhibit both BMP and TGF-β [38]. In CRC, members of the TGF-β/Smad-4 complex are among the most important biomarkers listed in Table 1. They are involved in DNA binding and gene expression regulation [39], and include genes such as SNAIL, SLUG, TWIST, and ZEB, promoting EMT formation and genes important for DNA damage response, (e.g., ATM, MSH2, and BRCA1) [32,33].

4. From CRC to Metastasis: A Complex Journey

Metastasis is the final and most devastating stage of cancer. It is the leading cause for fatalities in cancer [75,76]. Furthermore, we still do not fully understand what causes a carcinoma to metastasize. It is, therefore, not surprising that metastasis formation is a highly interesting topic in both research and clinical studies [90]. It appears that metastasis formation is a diverse process, as researchers have found that both the affected organs and the time course differ between cancers. For example, prostate cancer metastasis and ocular melanoma metastasis are restricted mostly to bone and liver, respectively [91]. In contrast, some types of cancer disseminate readily like breast, lungs, and testicles, others progress faster when metastatic cells come from luminal cells and are accompanied with poor survival [92].
Acquisition of a mesenchymal phenotype is an important step during metastasis progression. Pathways such as WNT, TGF-β, or p53 collaborate to drive CRC progression as described before, but the location (organ) where metastasis implants needs to be taken into consideration in the metastasis formation [9]. To explain the mechanism driving a metastatic event, Stephen Paget proposed an interconnection between metastatic cells released and organ or tissue receptors providing the right environment for the cells to invade [93,94]. This theory suggests a collaboration between the released cells and receptor cells and is termed “Seed and Soil” [95]. It implies that every metastatic event occurs only when both “seed” and “soil” groups of cells are compatible to interact together. This theory was later adapted by James Ewing, suggesting that metastasis is a result of purely mechanical factors of cells circulating in the vascular network. However, this theory did not explain the behavior or dissemination in which some organs with small blood supply become hosts of metastatic events and others with considerably more blood supply do not [94]. Nowadays it is thought that it is most likely a combination of both theories that explains the behavior of metastatic cells [96].
One of the key processes in tumor metastasis is thought to be the EMT [95]. This process comprises the early differentiation between epithelial to mesenchymal cells and has multiple consequences, including the modification of normal cell-to-cell junction and apical–basal polarity to become more invasive and loss of apical–basal polarity [97]. EMT is initiated by deregulation of three principal groups of regulators that are involved in the inactivation of E-cadherin, consisting of the bHLH family of transcription factors, the zinc-finger E-box-binding homeobox family, and the Snail zinc-finger family of transcription factors [98]. Together they enhance the effects of N-cadherins, integrin-αvβ6, and upregulate genes involved in the invasion and migratory behavior [94].
WNT signaling is another essential component of metastasis. It is involved in the process through its β-catenin-dependent (canonical) and β-catenin-independent (non-canonical) pathways. In the canonical pathway, WNT is degraded in the cytoplasm and migrates to the nucleus. There it interacts with TCF/LEF transcription factors, leading to abnormal secretion of β-catenin/TCF-responsive target genes, which in turn provoke cell differentiation and apoptosis [99,100]. The β-catenin-independent pathway involves the activation of GTPases, which regulate the activity of pathways like MAPK through the release of intracellular calcium. This results in changes in cell adhesion, cell polarity, and cellular migration [101].
Another important factor in metastasis formation is the TGF-β pathway, which can work through the SMAD-dependent and non-SMAD-dependent pathways. In the SMAD dependent-pathway, the E-cadherin transcriptional repressor induces the modification of SMAD levels [102] through phosphorylation of the C-terminal region of SMAD [34]. Once the SMAD complex is activated, it is translocated to the nucleus where it binds to SMAD-binding DNA elements (SBE), which activate or block the transcription of target genes [103]. In the SMAD-independent pathway, TGF- β activates other mediators such as MAPK, extracellular signal-regulated kinase (Erk), and c-Jun amino-terminal kinase (JNK) [104], which have been implicated in invasive behavior [105], and all of which are known biomarkers related to cell differentiation and promoting cell release.
The EMT phenotype is not only important for the formation of metastasis, but also plays a critical role in the formation and progression of cancer in general. EMT is responsible for the formation of metastasis-initiating cells (MICs), which form a small group of circulating tumors cells (CTCs) [106]. CTCs are tumor cells that enter the bloodstream and are considered to be metastasis precursors displaying EMT-like behavior [107]. CTC development is an intricate process, in which all of the common deregulated mechanisms, such as the PI3K/AKT, MAPK, and ERK pathways [108], as well as macrophage participation in EMT induction [109], are involved. However, CTCs and its capability to migrate is not the final step of invasion. After circulating through the bloodstream and finding a favorable microenvironment, CTCs must colonize a host tissue through a reverse phenotypical event known as mesenchymal-to-epithelial transition (MET) [110]. MET describes the transition from mesenchymal to epithelial cells and provides CTCs with a growth advantage in the new organ [109].
So far, we have described how genomic mutations leading to the disruption of protein–protein interactions, drive carcinogenesis. However, it is important to understand that there are other factors besides point mutations and protein signaling involved in cancer progression. Changes in chromatin structure and epigenetic modification are often observed in cancer [111]. These changes include a multiple of phenotypes that are commonly used to phenotype cancers, such as chromosome instability (CIN), microsatellite instability (MSI), and the CpG island methylator phenotype (CIMP). These phenotypes can be used for prognosis and direct treatment strategies. Furthermore, hypermethylation and hypomethylation profiles have been shown to alter gene expression, therefore contributing to cancer proliferation from CTCs clusters and indicating that epigenetic changes may act as drivers of carcinogenesis and prognostic markers.

5. Chromosome Stability and DNA Methylation Profiles: Can Epigenetic Changes Drive Metastasis?

With its relevance in development and disease, DNA methylation of the human genome has become a widely studied object. The human genome consists of over 28 million CpG sites, which are on average 70% methylated [112]. DNA methylation is characterized as the covalent addition of a methyl group (-CH3) at the carbon-5 of cytosine, which results in 5-methylcytosine (5mC) [113,114]. DNA methylation is catalyzed by three DNA methyltransferases (DNMTs), consisting of DNMT1, DNMT3a, and DNMT3b [71,115]. DNMT3a and DNMT3b are de novo DNA methyltransferases and are important during development and cell differentiation [116]. DNMT1 is required for the maintenance of DNA methylation in each cell cycle and methylates hemimethylated DNA after each cell division to ensure that the newly synthesized chromosomes inherit the same methylation pattern as the original ones.
There are three major epigenetic phenotypes in colorectal cancer that can be used to categorize tumors into different subtypes with different prognosis. These include CIN, MSI, and CIMP [117]. The CIN pathway is characterized by chromosomal abnormalities [118], such as aneuploidies, deletions, insertions, and amplifications, and is associated with poor prognosis, metastasis, and therapeutic resistance [21]. It is thought to be caused by changes in chromosome segregation, telomere dysfunction, and DNA damage response, which alter the expression of genes essential for maintenance of normal cell functions, including K-RAS, APC, PI3K, and TP53, among others [119]. The MSI pathway is named after the characteristic changes (insertions or deletions) of short repetitive DNA nucleotide sequences, knows as microsatellites, which are important for heterochromatin stability [120,121]. Loss of a functional DNA mismatch repair system (MMR), through mutations of MSH2, MSH3, or MSH6, results in a hypermutable phenotype [122], with MSI tumors often harboring > 1000 mutations. CIMP was first identified by Toyota et al. [123] and described as hypermethylation of CpG surrounding the promoter regions of certain genes in some CRC patients [124,125]. Since then, many more aberrantly methylated CpG islands (CGI) have been found in CRC and added to the CIMP [103]. The phenotype is used as a prognostic marker, with CIMP-positive patients having a poorer prognosis than CIMP-negative patients [103,110]. Among the CIMP genes, there are some interesting candidates, in which aberrant methylation may have functional consequences regarding CRC development and progression [126]. These include the DNA repair enzymes MLH1 and MGMT and the tumor suppressor genes CDKN2A and THBS1 [127].
Besides these three classic phenotypes, the cancer genome commonly undergoes widespread DNA methylation changes. One commonly observed characteristic is global hypomethylation of CpG poor regions, including intergenic regions and gene bodies, which are normally hypermethylated. In contrast, CpG-rich regions, also known as CpG islands (CGIs), are usually hypomethylated. In the process of cancer progression many of these CGIs become hypermethylated. As the majority of gene promoters contain CGIs, this hypermethylation may directly affect gene expression or enforce a repressive state that is caused by other chromatin modifications or changes in transcription factor binding [46]. Indeed, DNA methylation changes often correlate with suppression or inactivation of tumor suppressor genes [128]. These DNA methylation changes are therefore interesting biomarker candidates. For example, promotor methylation at the TERT hypermethylated oncological region (THOR) correlates with the upregulation the TERT gene affecting telomere maintenance [129]. THOR has therefore been suggested as a prognostic biomarker. To increase sensitivity and specificity, gene panels have been shown to improve detection of methylation changes in blood or stool [130,131,132]. For example, analysis of promoter hypermethylation in ALX4, BMP3, NPTX2, RARB, SDC2, SEPT9, and VIM improves CRC detection in blood plasma [46]. Even so, the sensitivity of biomarker assays in blood/stool is often low, which currently still hinders their use in medical treatment.
It is important to keep in mind that CRC is a heterogeneous disease and different patients display different phenotypes associated with different prognosis and treatment options. To summarize all these changes, the international CRC Subtype Consortium was formed, which combined classifications using transcriptomics with models based on molecular and clinical features to generate four consensus molecular subtypes of CRC [133]. Together, these subtypes can categorize 87% of CRC cases. The remaining 13% display mixed features of several subtypes and possibly represent transition phenotypes or intratumor heterogeneity. The categorization of these subtypes depends heavily on epigenetic pathways, demonstrating the importance of epigenetic changes for cancer research CMS1 (MSI Immune), which is characterized by CIMP-high, MSI, BRAF mutation, and gene hypermutation. CMS2 (Canonical) cancers show high CIN and upregulation of the WNT pathway. CMS3 (Metabolic) tumors are enriched for KRAS mutations and frequently exhibit MSI, as well as low CIN and low CIMP. CMS4 (Mesenchymal) is characterized by high CIN profiles, activation of TGF-β, and upregulation of EMT. Most CMS4 tumors harbor KRAS and BRAF mutations [117,133]. Epigenetic profiles are important for the classification of CRC. As most of the aberrant hypermethylation occurs in CpG islands, many of which are overlapping promoter regions enhancers, it was hypothesized that DNA methylation changes may directly affect gene transcription and therefore have functional consequences in carcinogenesis and metastasis formation [134]. However, it is not clear how DNA methylation may inhibit gene expression [135]. One possibility is that DNA methylation may prevent transcription factor binding [136]. DNA methylation may also contribute to heterochromatin formation by recruiting histone modifiers and chromatin remodelers [135]. Independent of their function, DNA methylation changes serve as extremely stable and easily detectable markers of changes in transcriptional state. Furthermore, changes in DNA methylation occur very early in carcinogenesis, making them interesting candidates for the development of biomarkers.
One of the biggest challenges in using CRC DNA methylation biomarkers for non-invasive screening assays is their reliable detection in blood and stool samples. These types of assays require detection of methylation changes in cell-free tumor DNA among the unchanged cellular DNA of the sample. There is currently no general consensus for an adequate technology, which has been suggested to slow implementation of DNA methylation changes in clinical diagnosis [137]. Delvenne et al. assessed different methods for DNA methylation analysis, including bisulphite sequencing, MS-DBA, MS-HRM/SMART-MSP, COBRA, MS-SnuPE, Q-MSP/MethylQuant, MethyLight, mass spectrometry, pyrosequencing, and Illumina Golden Gate [138]. All of them have two major criteria in common: (i) a biological/chemical module by which can be distinguish methylated from unmethylated fragments; and (ii) an amplification step that allows for the detection of methylation changes using a variety of techniques such as DNA sequencing, mass spectroscopy, microarray, and PCR [139]. Even though these techniques are all well established, they have advantages and disadvantages when it comes to DNA methylation biomarker screening. Delvenne et al. identifies bisulfite sequencing as the gold standard regarding sensitivity, but also highlights disadvantages, such as it being a labor-intensive, high-cost method, among others [138]. The lack of appropriate methods is currently hindering the implementation of DNA methylation biomarkers in clinical diagnosis. Method development is therefore one of the key issues that should be addressed.

6. Novel Methylation Biomarkers in CRC and Metastasis Prognosis

Biomarkers are important tools for disease detection and prognosis. They are cost-effective and often less invasive than other screens, like for example in the case of CRC colonoscopy. Currently, the majority of biomarkers used in clinical assays are designed for the early detection of cancer. Among the established CRC biomarkers, there are several based on DNA methylation changes, including NDRG4, BMP3 [140], SEPT9 [141], BCAT1, and IKZF1 [142]. The variety of biomarkers already on the market demonstrates the successful translation from research studies to clinical assays. However, none of these assays cover metastasis prognosis. Recently, novel biomarkers have been described that are aimed at detection of metastasis formation.
It is well known that CGI methylation is affected in CRC, and epigenetic alterations in the pathogenesis of CRC and liver metastasis have been widely reported. When comparing DNA methylation of distinct genes in CRC tumors of different stages (I–IV), including liver metastasis, Ju et al. found distinct DNA methylation in CRC with metastasis [143]. Importantly, in stage IV, methylation profile appeared to be established before metastasis occurred, which would make it an excellent biomarker for early diagnosis of metastasis formation. Specifically, this study found progressively increased DNA methylation levels of MGMT and TIMP3 during the metastatic process. MGMT is a DNA repair gene that is frequently methylated in CRC and is correlated with G to A transition mutations in cancer-related genes, such as KRAS, TP53, and PIK3CA [143]. MGMT hypermethylation has also commonly been found in brain metastasis from CRC [144]. A study examining genome-wide DNA methylation in CRC tissue of stages I–III and in paired primary and metastatic tumors also found that methylation frequencies of MGMT and TIMP3 were progressively increased during the metastatic process in CRC. Furthermore, UPK3A was found to be methylated only in hepatic metastasis [143]. Other genes that displayed a higher incidence of methylation in liver metastases compared to local CRC are ID4 and HPP1/TPEF. The inhibitor of DNA binding 4 (ID4), a dominant negative helix-loop-helix protein, acts as a tumor suppressor gene. ID4 methylation is correlated with histopathological tumor grade and poor prognosis [145]. Methylation of the HPP1/TPEF gene was also observed in the early stage of primary CRC (77%) as well as liver metastasis (79%) [146]. Interferon regulatory factor 8 (IRF8), a transcription factor of the IRF family, acts as a tumor suppressor gene and is silenced by epigenetic mechanisms in several human tumors [147,148]. In liver metastasis from CRC, IRF8 was found hypermethylated in 83% of the cases, whereas it was detected in 43% of primary colon carcinomas [149]. Hypermethylation of other genes were associated with lymph node metastasis, such as CDKN2A/p16 and CDKN2A/p1445, DFNA546,47, HLTF23, and ESR1 [150].
Recently, genome-wide sequencing studies have helped to study DNA methylation changes in metastasis on a wider scale. A study of 10 paired samples of CRC primary tumors and metastasis, using single-cell multi-omics sequencing analysis, characterized colon cancer cell populations according to their somatic copy number alterations (SCNAs), using both transcription and methylation profiles. The samples showed low methylation levels in CGIs regions of normal tissues, high methylation levels in primary tumors, and intermediate methylation levels in metastasis [151]. Another study in 59 CRC patients (stages I/II and IV), where primary tumor, adjacent normal tissue, and metastatic tumor tissues were analyzed using targeted bisulfite sequencing, identified 23 differentially methylated regions, which were associated with a high probability of liver metastasis of CRC [152]. We recently compared five paired samples from CRC patients (normal adjacent tissue, primary tumor, and lymph node metastasis) using bisulphite sequencing and identified five hypermethylated genes in all tumor samples that were associated with poor prognosis: BDNF, FIGN, HCN4, HTRA3, and STAC2 [88]. In addition, TBRG4 was found to be hypomethylated exclusively in CRC metastasis samples [88].

6.1. Microbiome and DNA Methylation as Biomarkers of Colorectal Cancer

Even though both the gut microbiome and DNA methylation are recognized to have important roles during colon carcinogenesis, only a few studies have looked at the combined function of them in CRC development. However, in the last decade it has emerged that DNA methylation changes correlate with changes in the microbiome, indicating an intricate relationship between the two.
DNA methylation changes of genes involved in early colon carcinogenesis have been associated with Peptostreptococcus and Schwartzia, and the absence of the Flavinofractor genus. Furthermore, the Firmicutes phyla has been correlated with CDH13 methylation [153]. In addition, the commensal bacteria Bifidobacterium breve and Lactobacillus rhamnosus GG can increase global DNA methylation and decrease histone acetylation in inflammatory bowel disease, one of the risk factors for CRC; therefore, both bacteria are beneficial for the colon [154,155]. A recent study evaluated the effects of butyrate, the fermentation end product of gut microbiota and an anti-tumor reagent, on α-ketoglutarato-mediated epigenetic modification, in colorectal adenocarcinoma HT-29 and Caco-2 cells [156]. There is evidence that butyrate has a direct effect on DNA methylation by regulating key enzymes, including TET and DNMT1 [157]. In HT-29 and Caco-2 cells, it was found that butyrate suppressed proliferation, potentiated differentiation, and induced apoptosis. Moreover, butyrate upregulated acetyl-CoA and α-ketoglutarato, which was concomitant with enhanced histone acetylation and DNA demethylation in the promoter of the DNA mismatch repair (MMR) genes [156]. Butyrate has also been shown to modify DNA methylation and inhibit WNT signaling in human gastric cancer cells, a pathway also known to be activated in CRC [157].
A recent multi-omics study analyzed the microbiome, metabolome, transcriptome, and DNA methylome of paired CRC and normal adjacent tissues [157]. The investigation identified 16 genes to which promoter CGI hypermethylation was related with a downregulation of gene expression. Furthermore, an abundance of Fusobacterium was significantly associated with the downregulated expression of PI16, FCRLA, and LSP1 in cancer tissues compared to healthy tissues [157]. Interestingly, a relationship between Fusobacterium status and epigenetic features of CRC, such as CIMP status and MSI, was previously reported, where Fusobacterium enrichment was associated with specific molecular subsets of CRCs, offering support for a pathogenic role in CRC for this bacterium [158]. Fusobacterium is able to affect gene expression and/or DNA methylation directly through nucleomodulins. These are bacteria that secrete virulence proteins, which allow them to enter the nucleus of the surrounding colon cells. In tissue samples, F. nucleatum intracellular infection has been associated with CDKN2A hypermethylation [159]. There are other bacteria using the same pathway [160]. For example, the nuclear traffic of Acinetobacter baumannii Tnp induces CGI hypermethylation of E-cadherin in COS-17 cells [161]. It remains unclear though how microorganisms, such as F. nucleatum, can induce epigenetic changes mechanistically.

6.2. Inhibin Subunit Beta B (INHBB)

Inhibin subunit beta B gene (INHBB) encodes a pre-protein, which subsequently is processed to inhibin and activin [86]. Inhibin protein consists of a heterodimer formed by the two subunits α and β, which are secreted by ovarian granulosa cells and testicular Sertoli cells [162]. It belongs to TGF-β superfamily, where it is unique, being the only heterodimer in the TGF- β family. Even though the inhibin signaling pathway is not fully known, it acts through antagonizing activin (another TGF- β protein encoded by INHBB) by binding to the activin receptor ActRII [163]. This prevents a signaling cascade that results in the downregulation of intracellular phosphorylation, and finally the downregulation of SMAD signaling [164]. In many cancers, including CRC, as well as oral, endometrial, prostate, renal, and thyroid cancer, INHBB is overexpressed [163]. Interestingly, INHBB is also associated to DNA methylation changes [165,166]. In fact, INHBB is hypermethylated in CRC, and its expression was negatively correlated with methylation. Furthermore, the overexpression of INHBB is significantly and positively associated with invasion depth, distant metastasis, and CRC stage [86,167].

6.3. SPARC-Related Modular Calcium-Binding Protein 2 (SMOC2)

SPARC-related modular calcium-binding protein 2 (SMOC2) is a member of the secreted protein acidic and rich in the cysteine SPARC family of matricellular proteins [168]. SPARC family proteins have been shown to be associated with several biological processes, such as cell adhesion, migration, and tissue repair, among others [169,170]. Moreover, SPARC proteins have been related to the promotion of various tumors, such as glioma, prostate, and gastric carcinomas [171]. Initial studies demonstrated the capability of SMOC2 to promote cell cycle progression and DNA synthesis, as well as impact cell growth in hepatocellular carcinoma [172]. Even though the mechanism of how SMOC2 promotes cell deregulation is not fully understood, recent research has shown that SMOC2 interacts with Fzd6 and LRP6 to activate the Wnt/β-catenin pathway in endometrial CSCs [173]. In CRC, SMOC2 has been suggested as a prognostic marker, having tumor suppressor activity in cancer progression [89]. Finally, the hypermethylation of CpG sites in the promoter region of SMOC2 suggest an epigenetic regulation in papillary thyroid carcinomas (PTCs) [174]. Whether SMOC2 is also a good candidate for CRC metastasis prognosis needs to be determined.

6.4. Helicase-like Transcription Factor (HLTF)

Helicase-like transcription factor (HLTF) is a member of the Switch/sucrose non-fermenting family (SWI/SNF) [175] and is comprised of a DNA-binding domain, a RING helicase motif, and a HIRAN domain [176,177]. HLTF has an important function in nucleosome remodeling. It is a mediator in replication fork reversal through its HIRAN domain and can act as a putative tumor suppressor through its role in DNA damage tolerance [178], genome stability maintenance, and gene transcription [58]. It has been suggested that the loss of expression of HLTF in several cancers (colon, gastric, esophageal, cervix, lung, hepatocellular, and bladder) may be caused by promoter methylation, as the downregulation of HLTF is correlated with hypermethylation of the promoter region [86,167,179]. This association makes HLTF a candidate as a cancer methylation biomarker [180]. Importantly, HLTF and more specifically, HLTF methylation, has been related to metastasis formation. Aberrant HLTF methylation is thought to affects HTLF’s capability to suppress migration and invasion through the TFG-β/SMAD pathway [58]. Therefore, HLTF may be a new methylation biomarker for CRC metastasis prognosis.

6.5. Eye Absent Homolog 4 (EYA4)

The eye absent gene family consist of four members (EYA1, EYA2, EYA3, and EYA4) that are characterized by a highly conserved C-terminal EYA domain [181]. EYA proteins have phosphatase activity through their threonine and tyrosine residues on their different domains [53]. They are involved in different cellular functions, like cell proliferation and survival, cell migration, DNA damage repair, angiogenesis, and developmental cell polarity, among others [182]. In cancer, the EYA family has been related to the RTK/RAS/MAPK family and the TGF-β pathway, controlling cell apoptosis and proliferation [183]. Specifically, in CRC, EYA4 has been suggested as a tumor suppressor involved in the inhibition of the WNT signaling pathway. EYA4 is a possible candidate to be regulated by epigenetic mechanisms like DNA methylation. It has been proposed that the inactivation by aberrant promoter methylation affects its interaction with WNT, MAPK, and local adhesion pathways [184]. In esophageal squamous cell carcinoma (ESCC), its hypermethylated EYA4 reduces the phosphorylation of AKT, therefore inducing EMT formation [185]. EYA4 promoter hypermethylation may therefore serve as a biomarker for detection of CRC undergoing EMT.

6.6. GATA-Binding Protein 5 (GATA5)

GATA-binding proteins comprise a small family of zinc-finger transcriptions factors that bind to the GATA motif and are composed of six members ranging from GATA1 to GATA6 [186]. Through its highly conserved zinc-finger DNA-binding domain, it has been associated with various cell functions, such as gene regulation, epithelial differentiation, and organogenesis [187,188]. Moreover, studies have identified some GATA family members, specifically GATA4, GATA5, and GATA6, as tumor suppressor genes associated with several cancer diseases. In lung cancer, aberrant promoter DNA methylation leads to a loss of GATA4 and GATA5 activity as tumor suppressor genes [187]. In cholangiocarcinoma, deregulated GATA5 gene expression has been correlated to cell proliferation and metastasis. This deregulation was thought to occur through DNA methylation changes, resulting in the inhibition of Wnt/β-catenin signaling activity [55]. DNA methylation changes of GATA5 have also been observed in CRC and gastric cancer [189], marking it as a potential novel biomarker in early stages of colorectal cancer. It will be important to elucidate if it can also serve as a marker for metastasis in CRC, as was shown in other cancer types.

6.7. Brain-Derived Neurotrophic Factor (BDNF)

Brain-derived neurotrophic factor (BDNF) is a member of neurotrophin family, which includes Nerve Growth Factor (NGF), Neurotrophin 3 (NT-3), and Neurotrophin 4 (NT-4) [190]. All neurotrophins are secreted as precursors or proneurotrophin, which are cleaved in order to produce mature neurotrophin [191]. Neurotrophins like BDNF act as intracellular after binding to their receptors TrkB and p75NTR. This interaction activates the RAS signaling pathway and is associated to apoptosis resistance, high cell proliferation, and poor prognosis [192]. Activation of neurotrophins has also been associated with the promotion of cell survival and differentiation through PI3K/AKT activation [193]. Moreover, methylation status of BDNF has been linked with several adversities and diseases, like psychiatric vulnerabilities [194]. Furthermore, BDNF hypermethylation has been associated to poor prognosis and metastasis formation in CRC [88,151,152].

6.8. Transforming Growth Factor Beta Regulator 4 (TBRG4)

The FASTK protein family is involved in mitochondrial RNA regulator homeostasis [195]. It is comprised of six members named FASTK and FASTKD1 to FASTKD5 and characterized by three poorly conserved domains called FAST1, FAST2, and RAP [196]. The overexpression of FASTKD4/TBRG4 in different types of cancer is highly associated with cancer progression. In lung cancer, an increase in the expression of TBRG4 is observed compared to normal tissue [197]. In esophageal cancer, high levels of TBRG4 promote ESCC formation, by regulation of the levels of reactive oxygen species (ROS) and inhibition of the mitochondrial-dependent apoptotic signal [198]. Specifically in osteosarcoma, high levels of TBRG4 corresponded to lower survival rates, promoting cell proliferation and invasion by upregulation of TGF-β and PI3K/AKT signaling [199]. New discoveries have found that TBRG4 is hypomethylated in colorectal cancer, which highlight its capability to be a new putative biomarker in prognosis of metastasis [88]. However, there is not enough information yet, and further studies are needed to assess its potential as a metastasis biomarker.

6.9. Open Issues Regarding the Use of Methylation Biomarkers

There are many studies describing putative DNA methylation biomarkers for CRC metastasis. However, to date, none of them have made it to be used in clinical diagnosis. Some of the most promising candidates, including INHBB, SMOC2, BDNF, and TBRG4, have been tested in large patient cohorts and validated across groups and/or been identified by multiple studies. However, most of the candidate biomarkers described above currently lack large cohort studies and need to be clinically verified before they can be generally used. Some of the candidates, such as BDNF and TBRG4, are involved in important signaling pathways as PI3K/AKT and TGF-β. Studying their deregulation in more detail would not only benefit the development of novel biomarkers, but could also improve our understanding of the mechanisms involved in metastasis formation.

7. Conclusions

Cancer is a global health problem, affecting millions of people and saturating health systems worldwide. Although there is still no general cure for cancer, treatments are continuously improving and are significantly increasing the life expectancy of patients. Nevertheless, the aggressiveness of late-stage cancer and the difficulty of curing the disease highlights the importance for new strategies and methods to improve early discovery and therefore treatment success.
Novel DNA methylation biomarkers for the prognosis during the early stages of cancer, such as INHBB, SMOC2, BDNF, and TBRG4, are being tested for clinical use to improve detection methods and guide the treatment and diagnoses of cancer patients. Other interesting candidates, including HLTF, GATA5, and EYA4, still have to be tested in more detail. Furthermore, besides the clinical application, DNA methylation biomarkers may increase our understanding of the processes involved in metastasis formation.
Importantly, for common use of DNA methylation biomarkers in clinical diagnosis, detection methods need to be optimized and standardized. This will improve the sensitivity and specificity of the assays, which are currently still low. Together with the proposed biomarker validation described above, this will pave the way to establish methylation biomarkers in clinical diagnosis and prognosis.

Author Contributions

Conceptualization, A.G. and C.G.I.; investigation, A.G.; resources, C.G.I. and P.B.; writing—original draft preparation, A.G.; writing—review and editing, H.D., P.B. and C.G.I.; visualization, C.G.I.; supervision, C.G.I. and P.B.; project administration, C.G.I.; funding acquisition, C.G.I. and P.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by FONDEF Idea ID21I10027, Millennium Institute on Immunology and Immunotherapy (IMII) ICN2021_045, DIUFRO DI20-0160, DI20-0129 and DI20-0128, FONDECYT 1210440 and 3200676.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the result.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA. Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. Borgna, V.; Villegas, J.; Burzio, V.A.; Belmar, S.; Araya, M.; Jeldes, E.; Lobos-González, L.; Silva, V.; Villota, C.; Oliveira-Cruz, L.; et al. Mitochondrial ASncmtRNA-1 and ASncmtRNA-2 as Potent Targets to Inhibit tumor Growth and Metastasis in the RenCa murine Renal Adenocarcinoma Model. Oncotarget 2017, 8, 43692–43708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hanley, M.P.; Hahn, M.A.; Li, A.X.; Wu, X.; Lin, J.; Wang, J.; Choi, A.H.; Ouyang, Z.; Fong, Y.; Pfeifer, G.P.; et al. Genome-wide DNA methylation profiling reveals cancer-associated changes within early colonic neoplasia. Oncogene 2017, 36, 5035–5044. [Google Scholar] [CrossRef] [Green Version]
  4. Slattery, M.L. Diet, lifestyle, and colon cancer. Semin. Gastrointest. Dis. 2000, 11, 142–146. [Google Scholar]
  5. Aran, V.; Victorino, A.P.; Thuler, L.C.; Ferreira, C.G. Colorectal Cancer: Epidemiology, Disease Mechanisms and Interventions to Reduce Onset and Mortality. Clin. Colorectal Cancer 2016, 15, 195–203. [Google Scholar] [CrossRef] [PubMed]
  6. Burt, R.; Neklason, D.W. Genetic testing for inherited colon cancer. Gastroenterology 2005, 128, 1696–1716. [Google Scholar] [CrossRef] [PubMed]
  7. Fearon, E.R. Molecular genetics of colorectal cancer. Annu. Rev. Pathol. Mech. Dis. 2011, 6, 479–507. [Google Scholar] [CrossRef]
  8. Fearon, E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61, 759–767. [Google Scholar] [CrossRef]
  9. Tauriello, D.V.F.; Calon, A.; Lonardo, E.; Batlle, E. Determinants of metastatic competency in colorectal cancer. Mol. Oncol. 2017, 11, 97–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Manfredi, S.; Lepage, C.; Hatem, C.; Coatmeur, O.; Faivre, J.; Bouvier, A.M. Epidemiology and management of liver metastases from colorectal cancer. Ann. Surg. 2006, 244, 254–259. [Google Scholar] [CrossRef]
  11. Brenner, H.; Kloor, M.; Pox, C.P. Colorectal cancer. Lancet 2014, 383, 1490–1502. [Google Scholar] [CrossRef]
  12. Huang, D.; Sun, W.; Zhou, Y.; Li, P.; Chen, F.; Chen, H.; Xia, D.; Xu, E.; Lai, M.; Wu, Y.; et al. Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev. 2018, 37, 173–187. [Google Scholar] [CrossRef] [PubMed]
  13. Schiffman, J.D.; Fisher, P.G.; Gibbs, P. Early Detection of Cancer: Past, Present, and Future. Introduction to cancer screening and Tumor Markers for Early Cancer Detection. ASCO Educ. B 2015, 35, 57–65. [Google Scholar]
  14. Wu, L.; Qu, X. Cancer biomarker detection: Recent achievements and challenges. Chem. Soc. Rev. 2015, 44, 2963–2997. [Google Scholar] [CrossRef] [PubMed]
  15. Lech, G.; Słotwiński, R.; Słodkowski, M.; Krasnodębski, I.W. Colorectal cancer tumour markers and biomarkers: Recent therapeutic advances. World J. Gastroenterol. 2016, 22, 1745–1755. [Google Scholar] [CrossRef]
  16. Zlobec, I. Novel biomarkers for the prediction of metastasis in colorectal cancer. Expert Opin. Med. Diagn. 2013, 7, 137–146. [Google Scholar] [CrossRef]
  17. Sartor, O.; de Bono, J.S. Metastatic Prostate Cancer. N. Engl. J. Med. 2018, 378, 645–657. [Google Scholar] [CrossRef] [PubMed]
  18. Feng, L.; Lou, J. DNA Methylation Analysis. Methods Mol. Biol. 2019, 1894, 181–227. [Google Scholar]
  19. Zeng, H.; He, B.; Yi, C.; Peng, J. Liquid biopsies: DNA methylation analyses in circulating cell-free DNA. J. Genet. Genom. 2018, 45, 185–192. [Google Scholar] [CrossRef]
  20. Muzny, D.M.; Bainbridge, M.N.; Chang, K.; Dinh, H.H.; Drummond, J.A.; Fowler, G.; Kovar, C.L.; Lewis, L.R.; Morgan, M.B.; Newsham, I.F.; et al. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012, 487, 330–337. [Google Scholar] [CrossRef] [Green Version]
  21. Nguyen, L.H.; Goel, A.; Chung, D.C.; Hospital, M.G.; Unit, T.E.; Hospital, M.G.; Sammons, C.A.; Hospital, M.G.; Medical, H. Pathways of Colorectal Carcinogenesis. Gastroenterology 2020, 158, 291–302. [Google Scholar] [CrossRef] [PubMed]
  22. Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef]
  23. Cheng, X.; Xu, X.; Chen, D.; Zhao, F.; Wang, W. Therapeutic potential of targeting the Wnt/β-catenin signaling pathway in colorectal cancer. Biomed. Pharmacother. 2019, 110, 473–481. [Google Scholar] [CrossRef]
  24. Guo, Y.; Pan, W.; Liu, S.; Shen, Z.; Xu, Y.; Hu, L. ERK/MAPK signalling pathway and tumorigenesis (Review). Exp. Ther. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Rauen, K.A. The RASopathies. Annu. Rev. Genomics Hum. Genet 2013, 14, 355–369. [Google Scholar] [CrossRef] [Green Version]
  26. Joerger, A.C.; Fersht, A.R. The p53 Pathway: Origins, Inactivation in Cancer, and Emerging Therapeutic Approaches. Annu. Rev. Biochem. 2016, 85, 375–404. [Google Scholar] [CrossRef]
  27. Mantovani, F.; Collavin, L.; Del Sal, G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 2019, 26, 199–212. [Google Scholar] [CrossRef]
  28. Essmann, F.; Schulze-Osthoff, K. Translational approaches targeting the p53 pathway for anti-cancer therapy. Br. J. Pharmacol. 2012, 165, 328–344. [Google Scholar] [CrossRef] [PubMed]
  29. Vogelstein, B.; Lane, D.; Levine, A.J. Surfing the p53 network. Nature 2000, 408, 307–310. Available online: http://www.nature.com/nature/journal/v408/n6810/full/408307a0.html#B41 (accessed on 2 August 2021). [CrossRef]
  30. Levine, A.J. P53: 800 Million Years of Evolution and 40 Years of Discovery. Nat. Rev. Cancer 2020, 20, 471–480. [Google Scholar] [CrossRef] [PubMed]
  31. Agrawal, A.; Yang, J.; Murphy, R.F.; Agrawal, D.K. Regulation of the p14ARF-Mdm2-p53 pathway: An overview in breast cancer. Exp. Mol. Pathol. 2006, 81, 115–122. [Google Scholar] [CrossRef] [PubMed]
  32. Itatani, Y.; Kawada, K.; Sakai, Y. Transforming growth factor-β signaling pathway in colorectal cancer and its tumor microenvironment. Int. J. Mol. Sci. 2019, 20, 5822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Syed, V. TGF-β Signaling in Cancer. J. Cell. Biochem. 2016, 117, 1279–1287. [Google Scholar] [CrossRef]
  34. Xie, F.; Ling, L.; Van Dam, H.; Zhou, F.; Zhang, L. TGF-β signaling in cancer metastasis. Acta Biochim. Biophys. Sin. 2018, 50, 121–132. [Google Scholar] [CrossRef] [Green Version]
  35. Colak, S.; ten Dijke, P. Targeting TGF-β Signaling in Cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
  36. Tu, S.; Huang, W.; Huang, C.; Luo, Z.; Yan, X. Contextual Regulation of TGF-β Signaling in Liver Cancer. Cells 2019, 8, 1235. [Google Scholar] [CrossRef] [Green Version]
  37. Haque, S.; Morris, J.C. Transforming growth factor-β: A therapeutic target for cancer. Hum. Vaccines Immunother. 2017, 13, 1741–1750. [Google Scholar] [CrossRef]
  38. Gu, S.; Feng, X.H. TGF-β signaling in cancer. Acta Biochim. Biophys. Sin. 2018, 50, 941–949. [Google Scholar] [CrossRef] [Green Version]
  39. Zhao, M.; Mishra, L.; Deng, C.X. The role of TGF-β/SMAD4 signaling in cancer. Int. J. Biol. Sci. 2018, 14, 111–123. [Google Scholar] [CrossRef] [Green Version]
  40. Yang, J.; Han, F.; Liu, W.; Chen, H.; Hao, X.; Jiang, X.; Yin, L.; Huang, Y.; Cao, J.; Zhang, H.; et al. ALX4, an epigenetically down regulated tumor suppressor, inhibits breast cancer progression by interfering Wnt/β-catenin pathway. J. Exp. Clin. Cancer Res. 2017, 36, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Nassar, F.J.; Msheik, Z.S.; Nasr, R.R.; Temraz, S.N. Methylated circulating tumor DNA as a biomarker for colorectal cancer diagnosis, prognosis, and prediction. Clin. Epigenetics 2021, 13, 111. [Google Scholar] [CrossRef]
  42. Barchitta, M.; Maugeri, A.; Li Destri, G.; Basile, G.; Agodi, A. Epigenetic biomarkers in colorectal cancer patients receiving adjuvant or neoadjuvant therapy: A systematic review of epidemiological studies. Int. J. Mol. Sci. 2019, 20, 3842. [Google Scholar] [CrossRef] [Green Version]
  43. Liu, R.; Su, X.; Long, Y.; Zhou, D.; Zhang, X.; Ye, Z.; Ma, J.; Tang, T.; Wang, F.; He, C. A systematic review and quantitative assessment of methylation biomarkers in fecal DNA and colorectal cancer and its precursor, colorectal adenoma. Mutat. Res.—Rev. Mutat. Res. 2019, 779, 45–57. [Google Scholar] [CrossRef]
  44. Petit, J.; Carroll, G.; Gould, T.; Pockney, P.; Dun, M.; Scott, R.J. Cell-free DNA as a Diagnostic Blood-Based Biomarker for Colorectal Cancer: A Systematic Review. J. Surg. Res. 2019, 236, 184–197. [Google Scholar] [CrossRef]
  45. Vacante, M.; Borzì, A.M.; Basile, F.; Biondi, A. Biomarkers in colorectal cancer: Current clinical utility and future perspectives. World J. Clin. Cases 2018, 6, 869–881. [Google Scholar] [CrossRef]
  46. Rasmussen, S.L.; Krarup, H.B.; Sunesen, K.G.; Johansen, M.B.; Stender, M.T.; Pedersen, I.S.; Madsen, P.H.; Thorlacius-Ussing, O. Hypermethylated DNA, a circulating biomarker for colorectal cancer detection. PLoS ONE 2017, 12, e0180809. [Google Scholar] [CrossRef] [Green Version]
  47. Salehi, R.; Atapour, N.; Vatandoust, N.; Farahani, N.; Ahangari, F.; Salehi, A. Methylation pattern of ALX4 gene promoter as a potential biomarker for blood-based early detection of colorectal cancer. Adv. Biomed. Res. 2015, 4, 252. [Google Scholar] [CrossRef] [PubMed]
  48. Laugsand, E.A.; Brenne, S.S.; Skorpen, F. DNA methylation markers detected in blood, stool, urine, and tissue in colorectal cancer: A systematic review of paired samples. Int. J. Colorectal Dis. 2021, 36, 239–251. [Google Scholar] [CrossRef] [PubMed]
  49. Zhan, Y.X.; Luo, G.H. DNA methylation detection methods used in colorectal cancer. World J. Clin. Cases 2019, 7, 2916–2929. [Google Scholar] [CrossRef] [PubMed]
  50. Jedi, M.; Young, G.P.; Pedersen, S.K.; Symonds, E.L. Methylation and Gene Expression of BCAT1 and IKZF1 in Colorectal Cancer Tissues. Clin. Med. Insights Oncol. 2018, 12, 1179554918775064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Wen, J.; Liu, X.; Qi, Y.; Niu, F.; Niu, Z.; Geng, W.; Zou, Z.; Huang, R.; Wang, J.; Zou, H. BMP3 suppresses colon tumorigenesis via ActRIIB/SMAD2-dependent and TAK1/JNK signaling pathways. J. Exp. Clin. Cancer Res. 2019, 38, 428. [Google Scholar] [CrossRef] [PubMed]
  52. Jung, G.; Moreira, L.; Balaguer, F.; Goel, A.; Sammons, C.A.; Therapeutics, E. Epigenetics of colorectal cancer: Biomarker and therapeutic potential. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 111–130. [Google Scholar] [CrossRef] [PubMed]
  53. Gu, F.; Yuan, S.; Liu, L.; Zhu, P.; Yang, Y.; Pan, Z.; Zhou, W. EYA4 serves as a prognostic biomarker in hepatocellular carcinoma and suppresses tumour angiogenesis and metastasis. J. Cell. Mol. Med. 2019, 23, 4208–4216. [Google Scholar] [CrossRef] [PubMed]
  54. Ma, Z.; Williams, M.; Cheng, Y.Y.; Leung, W.K. Roles of methylated DNA biomarkers in patients with colorectal cancer. Dis. Markers 2019, 2019, 2673543. [Google Scholar] [CrossRef] [Green Version]
  55. Liu, P.; Zhou, T.F.; Qiu, B.A.; Yang, Y.X.; Zhu, Y.J.; An, Y.; Zhao, W.C.; Wu, Y.T.; Ma, P.F.; Li, J.B.; et al. Methylation-Mediated Silencing of GATA5 Gene Suppresses Cholangiocarcinoma Cell Proliferation and Metastasis. Transl. Oncol. 2018, 11, 585–592. [Google Scholar] [CrossRef]
  56. Raut, J.R.; Guan, Z.; Schrotz-King, P.; Brenner, H. Fecal DNA methylation markers for detecting stages of colorectal cancer and its precursors: A systematic review. Clin. Epigenetics 2020, 12, 122. [Google Scholar] [CrossRef]
  57. Hu, B.; Zhang, K.; Li, S.; Li, H.; Yan, Z.; Huang, L.; Wu, J.; Han, X.; Jiang, W.; Mulatibieke, T.; et al. HIC1 attenuates invasion and metastasis by inhibiting the IL-6/STAT3 signalling pathway in human pancreatic cancer. Cancer Lett. 2016, 376, 387–398. [Google Scholar] [CrossRef]
  58. Liu, L.; Liu, H.; Zhou, Y.; He, J.; Liu, Q.; Wang, J.; Zeng, M.; Yuan, D.; Tan, F.; Zhou, Y.; et al. HLTF suppresses the migration and invasion of colorectal cancer cells via TGF-β/SMAD signaling in vitro. Int. J. Oncol. 2018, 53, 2780–2788. [Google Scholar] [CrossRef] [Green Version]
  59. Wallner, M.; Herbst, A.; Behrens, A.; Crispin, A.; Stieber, P.; Göke, B.; Lamerz, R.; Kolligs, F.T. Methylation of serum DNA is an independent prognostic marker in colorectal cancer. Clin. Cancer Res. 2006, 12, 7347–7352. [Google Scholar] [CrossRef] [Green Version]
  60. Cai, Q.; Dozmorov, M.; Oh, Y. IGFBP-3/IGFBP-3 Receptor System as an Anti-Tumor and Anti-Metastatic Signaling in Cancer. Cells 2020, 9, 1261. [Google Scholar] [CrossRef]
  61. Kumar, A.; Singh, P.; Pandey, A.; Gosipatala, S.B. IGFBP3 gene promoter methylation analysis and its association with clinicopathological characteristics of colorectal carcinoma. Mol. Biol. Rep. 2020, 47, 6919–6927. [Google Scholar] [CrossRef]
  62. Lam, K.; Pan, K.; Linnekamp, J.F.; Medema, J.P.; Kandimalla, R. DNA methylation based biomarkers in colorectal cancer: A systematic review. Biochim. Biophys. Acta—Rev. Cancer 2016, 1866, 106–120. [Google Scholar] [CrossRef] [PubMed]
  63. Javierre, B.M.; Rodriguez-Ubreva, J.; Al-Shahrour, F.; Corominas, M.; Graña, O.; Ciudad, L.; Agirre, X.; Pisano, D.G.; Valencia, A.; Roman-Gomez, J.; et al. Long-range epigenetic silencing associates with deregulation of Ikaros targets in colorectal cancer cells. Mol. Cancer Res. 2011, 9, 1139–1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhang, S.; Xu, J.; Wu, S.; Wang, R.; Qu, X.; Yu, W.; Li, J.; Chen, L. IRF4 promotes cell proliferation by JNK pathway in multiple myeloma. Med. Oncol. 2013, 30, 594. [Google Scholar] [CrossRef] [PubMed]
  65. Baiula, M.; Spampinato, S.; Gentilucci, L.; Tolomelli, A. Novel Ligands Targeting α4β1 Integrin: Therapeutic Applications and Perspectives. Front. Chem. 2019, 7, 489. [Google Scholar] [CrossRef]
  66. Kerachian, M.A.; Kerachian, M. Long interspersed nucleotide element-1 (LINE-1) methylation in colorectal cancer. Clin. Chim. Acta 2019, 488, 209–214. [Google Scholar] [CrossRef]
  67. Lou, Y.T.; Chen, C.W.; Fan, Y.C.; Chang, W.C.; Lu, C.Y.; Wu, I.C.; Hsu, W.H.; Huang, C.W.; Wang, J.Y. LINE-1 methylation status correlates significantly to post-therapeutic recurrence in stage III colon cancer patients receiving FOLFOX-4 adjuvant chemotherapy. PLoS ONE 2015, 10, e0123973. [Google Scholar] [CrossRef] [PubMed]
  68. Li, Q.; Ren, B.; Gui, Q.; Zhao, J.; Wu, M.; Shen, M.; Li, D.; Li, D.; Chen, K.; Tao, M.; et al. Blocking MAPK/ERK pathway sensitizes hepatocellular carcinoma cells to temozolomide via downregulating MGMT expression. Ann. Transl. Med. 2020, 8, 1305. [Google Scholar] [CrossRef]
  69. Li, X.; Wang, Y.; Zhang, Z.; Yao, X.; Ge, J.; Zhao, Y. Correlation of MLH1 and MGMT methylation levels between peripheral blood leukocytes and colorectal tissue DNA samples in colorectal cancer patients. Oncol. Lett. 2013, 6, 1370–1376. [Google Scholar] [CrossRef] [PubMed]
  70. Liang, J.Y.; Yang, Q.F.; Zeng, Y.L.; Liu, Y.Y.; Liu, Y.T.; Gu, F.F.; Hu, Y.; Zhang, K.; Zhong, H.; Liu, L. Clinical value of MLH1-negative circulating tumor cells in lung cancer patients. Medicine 2019, 98, e15721. [Google Scholar] [CrossRef]
  71. Tse, J.W.T.; Jenkins, L.J.; Chionh, F.; Mariadason, J.M. Aberrant DNA Methylation in Colorectal Cancer: What Should We Target? Trends Cancer 2017, 3, 698–712. [Google Scholar] [CrossRef] [PubMed]
  72. Constâncio, V.; Nunes, S.P.; Henrique, R.; Jerónimo, C. DNA Methylation-Based Testing in Liquid Biopsies as Detection and Prognostic Biomarkers for the Four Major Cancer Types. Cells 2020, 9, 624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wang, D.; O’Rourke, D.; Sanchez-Garcia, J.F.; Cai, T.; Scheuenpflug, J.; Feng, Z. Development of a liquid biopsy based purely quantitative digital droplet PCR assay for detection of MLH-1promoter methylation. BMC Cancer 2020, 21. [Google Scholar] [CrossRef]
  74. Zheng, J.; Li, Y.; Zhu, S.; Li, J.; Zhao, Q.; Ji, G.; Wang, W.; Chu, D. NDRG4 stratifies the prognostic value of body mass index in colorectal cancer. Oncotarget 2016, 7, 1311–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Yu, Z.; Li, Z.; Wang, C.; Pan, T.; Chang, X.; Wang, X.; Zhou, Q.; Wu, X.; Li, J.; Zhang, J.; et al. Oncostatin M receptor, positively regulated by SP1, promotes gastric cancer growth and metastasis upon treatment with Oncostatin M. Gastric Cancer 2019, 22, 955–966. [Google Scholar] [CrossRef] [Green Version]
  76. Liang, Y.; Besch-Williford, C.; Hyder, S.M. PRIMA-1 inhibits growth of breast cancer cells by re-activating mutant p53 protein. Int. J. Oncol. 2009, 35, 1015–1023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Malpeli, G.; Innamorati, G.; Decimo, I.; Bencivenga, M.; Kamdje, A.H.N.; Perris, R.; Bassi, C. Methylation dynamics of RASSF1A and its impact on cancer. Cancers 2019, 11, 959. [Google Scholar] [CrossRef] [Green Version]
  78. Matthaios, D.; Balgkouranidou, I.; Karayiannakis, A.; Bolanaki, H.; Xenidis, N.; Amarantidis, K.; Chelis, L.; Romanidis, K.; Chatzaki, A.; Lianidou, E.; et al. Methylation status of the APC and RASSF1A promoter in cell-free circulating DNA and its prognostic role in patients with colorectal cancer. Oncol. Lett. 2016, 12, 748–756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hua, R.; Yu, J.; Yan, X.; Ni, Q.; Zhi, X.; Li, X.; Jiang, B.; Zhu, J. Syndecan-2 in colorectal cancer plays oncogenic role via epithelial-mesenchymal transition and MAPK pathway. Biomed. Pharmacother. 2020, 121, 109630. [Google Scholar] [CrossRef]
  80. Su, W.C.; Kao, W.Y.; Chang, T.K.; Tsai, H.L.; Huang, C.W.; Chen, Y.C.; Li, C.C.; Hsieh, Y.C.; Yeh, H.J.; Chang, C.C.; et al. Stool DNA test targeting methylated syndecan-2 (SDC2) as a noninvasive screening method for colorectal cancer. Biosci. Rep. 2021, 41, BSR20201930. [Google Scholar] [CrossRef] [PubMed]
  81. Zeng, Y.; Cao, Y.; Liu, L.; Zhao, J.; Zhang, T.; Xiao, L.; Jia, M.; Tian, Q.; Yu, H.; Chen, S.; et al. SEPT9_i1 regulates human breast cancer cell motility through cytoskeletal and RhoA/FAK signaling pathway regulation. Cell Death Dis. 2019, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Tham, C.; Chew, M.; Soong, R.; Lim, J.; Ang, M.; Tang, C.; Zhao, Y.; Ong, S.Y.K.; Liu, Y. Postoperative serum methylation levels of TAC1 and SEPT9 are independent predictors of recurrence and survival of patients with colorectal cancer. Cancer 2014, 120, 3131–3141. [Google Scholar] [CrossRef]
  83. Majchrzak-Celińska, A.; Słocińska, M.; Barciszewska, A.M.; Nowak, S.; Baer-Dubowska, W. Wnt pathway antagonists, SFRP1, SFRP2, SOX17, and PPP2R2B, are methylated in gliomas and SFRP1 methylation predicts shorter survival. J. Appl. Genet. 2016, 57, 189–197. [Google Scholar] [CrossRef] [Green Version]
  84. Zhao, D.; Qiao, J.; He, H.; Song, J.; Zhao, S.; Yu, J. TFPI2 suppresses breast cancer progression through inhibiting TWIST-integrin α5 pathway. Mol. Med. 2020, 26, 27. [Google Scholar] [CrossRef] [PubMed]
  85. Satelli, A.; Li, S. Vimentin as a potential molecular target in cancer therapy Or Vimentin, an overview and its potential as a molecular target for cancer therapy. Cell Mol. Life Sci. 2011, 68, 3033–3046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Yuan, J.; Xie, A.; Cao, Q.; Li, X.; Chen, J. INHBB Is a Novel Prognostic Biomarker Associated with Cancer-Promoting Pathways in Colorectal Cancer. BioMed Res. Int. 2020, 2020, 6909672. [Google Scholar] [CrossRef] [PubMed]
  87. Li, T.; Yu, Y.; Song, Y.; Li, X.; Lan, D.; Zhang, P.; Xiao, Y.; Xing, Y. Activation of BDNF/TrkB pathway promotes prostate cancer progression via induction of epithelial-mesenchymal transition and anoikis resistance. FASEB J. 2020, 34, 9087–9101. [Google Scholar] [CrossRef]
  88. Ili, C.; Buchegger, K.; Demond, H.; Castillo-Fernandez, J.; Kelsey, G.; Zanella, L.; Abanto, M.; Riquelme, I.; López, J.; Viscarra, T.; et al. Landscape of genome-wide dna methylation of colorectal cancer metastasis. Cancers 2020, 12, 2710. [Google Scholar] [CrossRef]
  89. Jang, B.G.; Kim, H.S.; Bae, J.M.; Kim, W.H.; Kim, H.U.; Kang, G.H. SMOC2, an intestinal stem cell marker, is an independent prognostic marker associated with better survival in colorectal cancers. Sci. Rep. 2020, 10, 14591. [Google Scholar] [CrossRef]
  90. Yuan, L.; Guo, F.; Wang, L.; Zou, Q. Prediction of tumor metastasis from sequencing data in the era of genome sequencing. Brief. Funct. Genom. 2019, 18, 412–418. [Google Scholar] [CrossRef] [PubMed]
  91. Nguyen, D.X.; Bos, P.D.; Massagué, J. Metastasis: From dissemination to organ-specific colonization. Nat. Rev. Cancer 2009, 9, 274–284. [Google Scholar] [CrossRef]
  92. Wan, L.; Pantel, K.; Kang, Y. Tumor metastasis: Moving new biological insights into the clinic. Nat. Med. 2013, 19, 1450–1464. [Google Scholar] [CrossRef]
  93. Fidler, I.J. The pathogenesis of cancer metastasis: The “seed and soil” hyphotesis revisited. Nat. Rev. Cancer 2003, 3, 453–458. [Google Scholar] [CrossRef] [PubMed]
  94. Akhtar, M.; Haider, A.; Rashid, S.; Al-Nabet, A.D.M.H. Paget’s “seed and Soil” Theory of Cancer Metastasis: An Idea Whose Time has Come. Adv. Anat. Pathol. 2019, 26, 69–74. [Google Scholar] [CrossRef] [PubMed]
  95. Seretis, F.; Seretis, C.; Youssef, H.; Chapman, M. Colorectal cancer: Seed and soil hypothesis revisited. Anticancer Res. 2014, 34, 2087–2094. [Google Scholar] [PubMed]
  96. Fokas, E.; Engenhart-Cabillic, R.; Daniilidis, K.; Rose, F.; An, H.X. Metastasis: The seed and soil theory gains identity. Cancer Metastasis Rev. 2007, 26, 705–715. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, Y.; Weinberg, R.A. Epithelial-to-mesenchymal transition in cancer: Complexity and opportunities. Front. Med. 2018, 12, 361–373. [Google Scholar] [CrossRef] [Green Version]
  98. Cao, H.; Xu, E.; Liu, H.; Wan, L.; Lai, M. Epithelial-mesenchymal transition in colorectal cancer metastasis: A system review. Pathol. Res. Pract. 2015, 211, 557–569. [Google Scholar] [CrossRef]
  99. Teeuwssen, M.; Fodde, R. Wnt Signaling in Ovarian Cancer Stemness, EMT, and Therapy Resistance. J. Clin. Med. 2019, 8, 1658. [Google Scholar] [CrossRef] [Green Version]
  100. Basu, S.; Cheriyamundath, S.; Ben-Ze’ev, A. Cell–cell adhesion: Linking wnt/β-catenin signaling with partial emt and stemness traits in tumorigenesis [version 1; peer review: 4 approved]. F1000Research 2018, 7, 1488. [Google Scholar] [CrossRef] [Green Version]
  101. Vincan, E.; Barker, N. The upstream components of the Wnt signalling pathway in the dynamic EMT and MET associated with colorectal cancer progression. Clin. Exp. Metastasis 2008, 25, 657–663. [Google Scholar] [CrossRef] [PubMed]
  102. Hao, Y.; Baker, D.; Dijke, P. Ten TGF-β-mediated epithelial-mesenchymal transition and cancer metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Villalba, M.; Evans, S.R.; Vidal-Vanaclocha, F.; Calvo, A. Role of TGF-β in metastatic colon cancer: It is finally time for targeted therapy. Cell Tissue Res. 2017, 370, 29–39. [Google Scholar] [CrossRef]
  104. Seoane, J.; Gomis, R.R. TGF-B Family Signaling in Tumor Suppression and Cancer Progression. Cold Spring Harb. Perspect. Biol. 2017, 9, a022277. [Google Scholar] [CrossRef] [Green Version]
  105. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanism of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
  106. Castro-Giner, F.; Aceto, N. Tracking cancer progression: From circulating tumor cells to metastasis. Genome Med. 2020, 12, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Wei, C.; Yang, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Liu, Q.; Dou, R.; Xiong, B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol. Cancer 2019, 18, 64. [Google Scholar] [CrossRef]
  108. Park, H.A.; Brown, S.R.; Kim, Y. Cellular mechanisms of circulating tumor cells during breast cancer metastasis. Int. J. Mol. Sci. 2020, 21, 5040. [Google Scholar] [CrossRef]
  109. Hamilton, G.; Rath, B. Mesenchymal-epithelial transition and circulating tumor cells in small cell lung cancer. Adv. Exp. Med. Biol. 2017, 994, 229–245. [Google Scholar] [CrossRef]
  110. Hamidi, S.; Nakaya, Y.; Nagai, H.; Alev, C.; Kasukawa, T.; Chhabra, S.; Lee, R.; Niwa, H.; Warmflash, A.; Shibata, T.; et al. Mesenchymal-epithelial transition regulates initiation of pluripotency exit before gastrulation. Development 2020, 147, dev.184960. [Google Scholar] [CrossRef]
  111. Gkountela, S.; Castro-Giner, F.; Szczerba, B.M.; Vetter, M.; Landin, J.; Scherrer, R.; Krol, I.; Scheidmann, M.C.; Beisel, C.; Stirnimann, C.U.; et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019, 176, 98–112. [Google Scholar] [CrossRef] [Green Version]
  112. Meng, H.; Cao, Y.; Qin, J.; Song, X.; Zhang, Q.; Shi, Y.; Cao, L. DNA methylation, its mediators and genome integrity. Int. J. Biol. Sci. 2015, 11, 604–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Miranda Furtado, C.L.; Dos Santos Luciano, M.C.; Da Silva Santos, R.; Furtado, G.P.; Moraes, M.O.; Pessoa, C. Epidrugs: Targeting epigenetic marks in cancer treatment. Epigenetics 2019, 14, 1164–1176. [Google Scholar] [CrossRef] [PubMed]
  114. Skvortsova, K.; Stirzaker, C.; Taberlay, P. The DNA methylation landscape in cancer. Essays Biochem. 2019, 63, 797–811. [Google Scholar] [CrossRef]
  115. Morgan, A.E.; Davies, T.J.; McAuley, M.T. The role of DNA methylation in ageing and cancer. Proc. Nutr. Soc. 2018, 77, 412–422. [Google Scholar] [CrossRef] [PubMed]
  116. Castillo-Aguilera, O.; Depreux, P.; Halby, L.; Arimondo, P.B.; Goossens, L. DNA Methylation Targeting: The DNMT/HMT Crosstalk Challenge. Biomolecules 2017, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  117. Yiu, A.J.; Yiu, C.Y. Biomarkers in Colorectal Cancer. Anticancer Res. 2016, 36, 1093–1102. [Google Scholar]
  118. Bakhoum, S.F.; Cantley, L.C. The multifaceted role of chromosomal instability in cancer and its microenvironment. Cell 2018, 174, 1347–1360. [Google Scholar] [CrossRef] [Green Version]
  119. Mármol, I.; Sánchez-de-Diego, C.; Dieste, A.P.; Cerrada, E.; Yoldi, M.J.R. Colorectal carcinoma: A general overview and future perspectives in colorectal cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [Green Version]
  120. Setaffy, L.; Langner, C. Microsatellite instability in colorectal cancer: Clinicopathological significance. Polish J. Pathol. 2015, 66, 203–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Yamamoto, H.; Imai, K. Microsatellite instability: An update. Arch. Toxicol. 2015, 89, 899–921. [Google Scholar] [CrossRef]
  122. Nojadeh, J.N.; Sharif, S.B.; Sakhinia, E. Microsatellite instability in colorectal cancer. EXCLI J. 2018, 17, 159–168. [Google Scholar] [CrossRef] [PubMed]
  123. Toyota, M.; Ahuja, N.; Ohe-Toyota, M.; Herman, J.G.; Baylin, S.B.; Issa, J.-P. CpG island methylator phenotype in colorectal cancer. Proc. Natl. Acad. Sci. USA 1999, 96, 8681–8686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Jia, M.; Gao, X.; Zhang, Y.; Hoffmeister, M.; Brenner, H. Different definitions of CpG island methylator phenotype and outcomes of colorectal cancer: A systematic review. Clin. Epigenetics 2016, 8, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Liu, J.; Tang, L.; Yi, J.; Li, G.; Lu, Y.; Xu, Y.; Zhao, S.; Mao, R.; Li, X.; Ren, L.; et al. Unique characteristics of CpG island methylator phenotype (CIMP) in a Chinese population with colorectal cancer. BMC Gastroenterol. 2019, 19, 173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Suzuki, H.; Yamamoto, E.; Maruyama, R.; Niinuma, T.; Kai, M. Biological significance of the CpG island methylator phenotype. Biochem. Biophys. Res. Commun. 2014, 455, 35–42. [Google Scholar] [CrossRef]
  127. Kim, Y.S.; Deng, G. Epigenetic Changes (Aberrant DNA Methylation) in Colorectal Neoplasia. Gut Liver 2007, 1, 1–11. [Google Scholar] [CrossRef] [Green Version]
  128. Park, J.Y. Promoter Hypermethylation in Prostate Cancer. Cancer Control 2010, 17, 245–255. [Google Scholar] [CrossRef]
  129. Lee, D.D.; Leão, R.; Komosa, M.; Gallo, M.; Zhang, C.H.; Lipman, T.; Remke, M.; Heidari, A.; Nunes, N.M.; Apolónio, J.D.; et al. DNA hypermethylation within TERT promoter upregulates TERT expression in cancer. J. Clin. Investig. 2019, 129, 223–229. [Google Scholar] [CrossRef] [Green Version]
  130. Imperiale, T.F.; Ransohoff, D.F.; Itzkowitz, S.H.; Levin, T.R.; Lavin, P.; Lidgard, G.P.; Ahlquist, D.A.; Berger, B.M. Multitarget Stool DNA Testing for Colorectal-Cancer Screening. N. Engl. J. Med. 2014, 370, 1287–1297. [Google Scholar] [CrossRef] [Green Version]
  131. Cassinotti, E.; Melson, J.; Liggett, T.; Melnikov, A.; Yi, Q.; Replogle, C.; Mobarhan, S.; Boni, L.; Segato, S.; Levenson, V. DNA methylation patterns in blood of patients with colorectal cancer and adenomatous colorectal polyps. Int. J. Cancer 2012, 131, 1153–1157. [Google Scholar] [CrossRef] [PubMed]
  132. He, Q.; Chen, H.Y.; Bai, E.Q.; Luo, Y.X.; Fu, R.J.; He, Y.S.; Jiang, J.; Wang, H.Q. Development of a multiplex MethyLight assay for the detection of multigene methylation in human colorectal cancer. Cancer Genet. Cytogenet. 2010, 202, 1–10. [Google Scholar] [CrossRef] [PubMed]
  133. 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. 2015, 21, 1350–1356. [Google Scholar] [CrossRef] [PubMed]
  134. Casalino, L.; Verde, P. Multifaceted roles of dna methylation in neoplastic transformation, from tumor suppressors to emt and metastasis. Genes 2020, 11, 922. [Google Scholar] [CrossRef]
  135. Greenberg, M.V.C.; Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 2019, 20, 590–607. [Google Scholar] [CrossRef]
  136. Yin, Y.; Morgunova, E.; Jolma, A.; Kaasinen, E.; Sahu, B.; Das, P.K.; Kivioja, T.; Dave, K.; Zhong, F.; Nitta, R.; et al. Impact of cytosine methylation on DNA bindgin specificities of human transcription factors. Science 2017, 356, 1–36. [Google Scholar] [CrossRef] [PubMed]
  137. Mikeska, T.; Bock, C.; Do, H.; Dobrovic, A. DNA methylation biomarkers in cancer: Progress towards clinical implementation. Expert Rev. Mol. Diagn. 2012, 12, 473–487. [Google Scholar] [CrossRef] [Green Version]
  138. Dehan, P.; Kustermans, G.; Guenin, S.; Horion, J.; Boniver, J.; Delvenne, P. DNA methylation and cancer diagnosis. Expert Rev. Mol. Diagn. 2009, 9, 651–657. [Google Scholar] [CrossRef] [PubMed]
  139. Levenson, V.V. DNA methylation as a universal biomarker. Expert Rev. Mol. Diagn. 2010, 10, 481–488. [Google Scholar] [CrossRef] [Green Version]
  140. Sobanski, T.; Arantes, L.M.R.B.; dos Santos, W.; Matsushita, M.; de Oliveira, M.A.; Costa, M.; de Carvalho, A.C.; Berardinelli, G.N.; Syrjänen, K.; Reis, R.M.; et al. Methylation profile of colon cancer genes in colorectal precursor lesions and tumor tissue: Perspectives for screening. Scand. J. Gastroenterol. 2021, 56, 920–928. [Google Scholar] [CrossRef] [PubMed]
  141. Warren, J.D.; Xiong, W.; Bunker, A.M.; Vaughn, C.P.; Furtado, L.V.; Roberts, W.L.; Fang, J.C.; Samowitz, W.S.; Heichman, K.A. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011, 9, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Mitchell, S.M.; Ross, J.P.; Drew, H.R.; Ho, T.; Brown, G.S.; Saunders, N.F.W.; Duesing, K.R.; Buckley, M.J.; Dunne, R.; Beetson, I.; et al. A panel of genes methylated with high frequency in colorectal cancer. BMC Cancer 2014, 14, 54. [Google Scholar] [CrossRef] [Green Version]
  143. Ju, H.X.; An, B.; Okamoto, Y.; Shinjo, K.; Kanemitsu, Y.; Komori, K.; Hirai, T.; Shimizu, Y.; Sano, T.; Sawaki, A.; et al. Distinct profiles of epigenetic evolution between colorectal cancers with and without metastasis. Am. J. Pathol. 2011, 178, 1835–1846. [Google Scholar] [CrossRef]
  144. De Maglio, G.; Casagrande, M.; Guardascione, M.; Fontanella, C.; Lutrino, S.E.; Rihawi, K.; Pisa, F.E.; Tuniz, F.; Fasola, G.; Pizzolitto, S.; et al. MGMT promoter methylation status in brain metastases from colorectal cancer and corresponding primary tumors. Future Oncol. 2015, 11, 1201–1209. [Google Scholar] [CrossRef] [PubMed]
  145. Umetani, N.; Takeuchi, H.; Fujimoto, A.; Shinozaki, M.; Bilchik, A.J.; Hoon, D.S.B. Epigenetic inactivation of ID4 in colorectal carcinomas correlates with poor differentiation and unfavorable prognosis. Clin. Cancer Res. 2004, 10, 7475–7483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Kim, M.S.; Lee, J.; Sidransky, D. DNA methylation markers in colorectal cancer. Cancer Metastasis Rev. 2010, 29, 181–206. [Google Scholar] [CrossRef]
  147. Tshuikina, M.; Jernberg-Wiklund, H.; Nilsson, K.; Öberg, F. Epigenetic silencing of the interferon regulatory factor ICSBP/IRF8 in human multiple myeloma. Exp. Hematol. 2008, 36, 1673–1681. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, Q.; Zhang, L.; Li, L.L.; Wang, Z.; Ying, J.; Fan, Y.; Xu, B.; Wang, L.; Liu, Q.; Chen, G.; et al. Interferon regulatory factor 8 functions as a tumor suppressor in renal cell carcinoma and its promoter methylation is associated with patient poor prognosis. Cancer Lett. 2014, 354, 227–234. [Google Scholar] [CrossRef] [PubMed]
  149. Kim, Y.H.; Petko, Z.; Dzieciatkowski, S.; Lin, L.; Ghiassi, M.; Stain, S.; Chapman, W.C.; Washington, M.K.; Willis, J.; Markowitz, S.D.; et al. CpG island methylation of genes accumulates during the adenoma progression step of the multistep pathogenesis of colorectal cancer. Genes Chromosom. Cancer 2006, 45, 781–789. [Google Scholar] [CrossRef]
  150. Wynter, C.V.A.; Walsh, M.D.; Higuchi, T.; Leggett, B.A.; Young, J.; Jass, J.R. Methylation patterns define two types of hyperplastic polyp associated with colorectal cancer. Gut 2004, 53, 573–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Bian, S.; Hou, Y.; Zhou, X.; Li, X.; Yong, J.; Wang, Y.; Wang, W.; Yan, J.; Hu, B.; Guo, H.; et al. Single-cell multiomics sequencing and analyses of human colorectal cancer. Science 2018, 362, 1060–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Li, W.; Guo, L.; Tang, W.; Ma, Y.; Wang, X.; Shao, Y.; Zhao, H.; Ying, J. Identification of DNA methylation biomarkers for risk of liver metastasis in early-stage colorectal cancer. Clin. Epigenetics 2021, 13, 126. [Google Scholar] [CrossRef] [PubMed]
  153. Rezasoltani, S.; Asadzadeh-Aghdaei, H.; Nazemalhosseini-Mojarad, E.; Dabiri, H.; Ghanbari, R.; Zali, M.R. Gut microbiota, epigenetic modification and colorectal cancer. Iran. J. Microbiol. 2017, 9, 55–63. [Google Scholar]
  154. Ghadimi, D.; Helwig, U.; Schrezenmeir, J.; Heller, K.J.; de Vrese, M. Epigenetic imprinting by commensal probiotics inhibits the IL-23/IL-17 axis in an in vitro model of the intestinal mucosal immune system. J. Leukoc. Biol. 2012, 92, 895–911. [Google Scholar] [CrossRef] [PubMed]
  155. Yang, T.; Owen, J.L.; Lightfoot, Y.L.; Kladde, M.P.; Mohamadzadeh, M. Microbiota impact on the epigenetic regulation of colorectal cancer. Trends Mol. Med. 2013, 19, 714–725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Sun, X.; Zhu, M.-J. Butyrate Inhibits Indices of Colorectal Carcinogenesis via Enhancing α-Ketoglutarate-Dependent DNA Demethylation of Mismatch Repair Genes. Mol. Nutr. Food Res. 2018, 62, 1700932. [Google Scholar] [CrossRef]
  157. Wang, Q.; Ye, J.; Fang, D.; Lv, L.; Wu, W.; Shi, D.; Li, Y.; Yang, L.; Bian, X.; Wu, J.; et al. Multi-omic profiling reveals associations between the gut mucosal microbiome, the metabolome, and host DNA methylation associated gene expression in patients with colorectal cancer. BMC Microbiol. 2020, 20, 83. [Google Scholar] [CrossRef]
  158. Tahara, T.; Yamamoto, E.; Suzuki, H.; Maruyama, R.; Chung, W.; Garriga, J.; Jelinek, J.; Yamano, H.-O.; Sugai, T.; An, B.; et al. Fusobacterium in Colonic Flora and Molecular Features of Colorectal Carcinoma. Cancer Res. 2014, 74, 1311–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Park, H.E.; Kim, J.H.; Cho, N.Y.; Lee, H.S.; Kang, G.H. Intratumoral Fusobacterium nucleatum abundance correlates with macrophage infiltration and CDKN2A methylation in microsatellite-unstable colorectal carcinoma. Virchows Arch. 2017, 471, 329–336. [Google Scholar] [CrossRef]
  160. Denzer, L.; Schroten, H.; Schwerk, C. From gene to protein—How bacterial virulence factors manipulate host gene expression during infection. Int. J. Mol. Sci. 2020, 21, 3730. [Google Scholar] [CrossRef]
  161. Moon, D.C.; Choi, C.H.; Lee, S.M.; Lee, J.H.; Kim, S., II; Kim, D.S.; Lee, J.C. Nuclear translocation of Acinetobacter baumannii transposase induces DNA methylation of CpG regions in the promoters of E-cadherin gene. PLoS ONE 2012, 7, e38974. [Google Scholar] [CrossRef] [Green Version]
  162. Makanji, Y.; Zhu, J.; Mishra, R.; Holmquist, C.; Wong, W.P.S.; Schwartz, N.B.; Mayo, K.E.; Woodruff, T.K. Inhibin at 90: From Discovery to Clinical Application, a Historical Review. Endocr. Rev. 2014, 35, 747–794. [Google Scholar] [CrossRef]
  163. Robertson, D.M.; Burger, H.G.; Fuller, P.J. Inhibin/activin and ovarian cancer. Endocr. Relat. Cancer 2004, 11, 35–49. [Google Scholar] [CrossRef] [Green Version]
  164. Goney, M.P.; Wilce, M.C.; Wilce, J.A.; Stocker, W.A.; Goodchild, G.M.; Chan, K.L.; Harrison, C.A.; Walton, K.L. Engineering the ovarian hormones inhibin A and inhibin B to enhance synthesis and activity. Endocrinology 2020, 161, bqaa099. [Google Scholar] [CrossRef]
  165. Mayor, R.; Casadomé, L.; Azuara, D.; Moreno, V.; Clark, S.J.; Capellà, G.; Peinado, M.A. Long-range epigenetic silencing at 2q14.2 affects most human colorectal cancers and may have application as a non-invasive biomarker of disease. Br. J. Cancer 2009, 100, 1534–1539. [Google Scholar] [CrossRef] [Green Version]
  166. Karpinski, P.; Ramsey, D.; Grzebieniak, Z.; Sasiadek, M.M.; Blin, N. The CpG island methylator phenotype correlates with long-range epigenetic silencing in colorectal cancer. Mol. Cancer Res. 2008, 6, 585–591. [Google Scholar] [CrossRef] [Green Version]
  167. Xu, Z.; Li, Y.; Cui, Y.; Guo, Y. Identifications of Candidate Genes Significantly Associated With Rectal Cancer by Integrated Bioinformatics Analysis. Technol. Cancer Res. Treat. 2020, 19, 1533033820973270. [Google Scholar] [CrossRef]
  168. Gerarduzzi, C.; Kumar, R.K.; Trivedi, P.; Ajay, A.K.; Iyer, A.; Boswell, S.; Hutchinson, J.N.; Waikar, S.S.; Vaidya, V.S. Silencing SMOC2 ameliorates kidney fibrosis by inhibiting fibroblast to myofibroblast transformation. JCI Insight 2017, 2, e90299. [Google Scholar] [CrossRef] [Green Version]
  169. Zhu, X.C.; Dong, Q.Z.; Zhang, X.F.; Deng, B.; Jia, H.L.; Ye, Q.H.; Qin, L.X.; Wu, X.Z. MicroRNA-29a suppresses cell proliferation by targeting SPARC in hepatocellular carcinoma. Int. J. Mol. Med. 2012, 30, 1321–1326. [Google Scholar] [CrossRef] [PubMed]
  170. Robert, G.; Gaggioli, C.; Bailet, O.; Chavey, C.; Abbe, P.; Aberdam, E.; Sabatié, E.; Cano, A.; Garcia De Herreros, A.; Ballotti, R.; et al. SPARC represses E-cadherin and induces mesenchymal transition during melanoma development. Cancer Res. 2006, 66, 7516–7523. [Google Scholar] [CrossRef] [Green Version]
  171. Deng, B.; Qu, L.; Li, J.; Fang, J.; Yang, S.; Cao, Z.; Mei, Z.; Sun, X. MiRNA-211 suppresses cell proliferation, migration and invasion by targeting SPARC in human hepatocellular carcinoma. Sci. Rep. 2016, 6, 26679. [Google Scholar] [CrossRef] [Green Version]
  172. Su, J.R.; Kuai, J.H.; Li, Y.Q. Smoc2 potentiates proliferation of hepatocellular carcinoma cells via promotion of cell cycle progression. World J. Gastroenterol. 2016, 22, 10053–10063. [Google Scholar] [CrossRef] [PubMed]
  173. Lu, H.; dan Ju, D.; Yang, G.D.; Zhu, L.Y.; Yang, X.M.; Li, J.; Song, W.W.; Wang, J.H.; Zhang, C.C.; Zhang, Z.G.; et al. Targeting cancer stem cell signature gene SMOC-2 Overcomes chemoresistance and inhibits cell proliferation of endometrial carcinoma. EBioMedicine 2019, 40, 276–289. [Google Scholar] [CrossRef] [Green Version]
  174. Kim, H.S.; Choi, J.H.; Lee, J.Y.; Kang, J.H.; Myung, J.K.; Kim, W.H.; Jang, B.G. Downregulation of SMOC2 expression in papillary thyroid carcinoma and its prognostic significance. Sci. Rep. 2020, 10, 4853. [Google Scholar] [CrossRef]
  175. Dhont, L.; Mascaux, C.; Belayew, A. The helicase-like transcription factor (HLTF) in cancer: Loss of function or oncomorphic conversion of a tumor suppressor? Cell. Mol. Life Sci. 2016, 73, 129–145. [Google Scholar] [CrossRef]
  176. Hishiki, A.; Hara, K.; Ikegaya, Y.; Yokoyama, H.; Shimizu, T.; Sato, M.; Hashimoto, H. Structure of a novel DNA-binding domain of Helicase-like Transcription Factor (HLTF) and its functional implication in DNA damage tolerance. J. Biol. Chem. 2015, 290, 13215–13223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Gallo, D.; Brown, G.W. Post-replication repair: Rad5/HLTF regulation, activity on undamaged templates, and relationship to cancer. Crit. Rev. Biochem. Mol. Biol. 2019, 54, 301–332. [Google Scholar] [CrossRef]
  178. Helmer, R.A.; Martinez-Zaguilan, R.; Kaur, G.; Smith, L.A.; Dufour, J.M.; Chilton, B.S. Helicase-like transcription factor-deletion from the tumor microenvironment in a cell linederived xenograft model of colorectal cancer reprogrammed the human transcriptome-Snitroso-proteome to promote inflammation and redirect metastasis. PLoS ONE 2021, 16, e0251132. [Google Scholar] [CrossRef]
  179. Unk, I.; Hajdú, I.; Blastyák, A.; Haracska, L. Role of yeast Rad5 and its human orthologs, HLTF and SHPRH in DNA damage tolerance. DNA Repair 2010, 9, 257–267. [Google Scholar] [CrossRef] [PubMed]
  180. Elserafy, M.; Abugable, A.A.; Atteya, R.; El-Khamisy, S.F. Rad5, HLTF, and SHPRH: A Fresh View of an Old Story. Trends Genet. 2018, 34, 574–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Zhou, H.; Zhang, L.; Vartuli, R.L.; Ford, H.L.; Zhao, R. The Eya Phosphatase: Its unique role in cancer. Int. J. Biochem. Cell Biol. 2018, 96, 165–170. [Google Scholar] [CrossRef]
  182. Tadjuidje, E.; Hegde, R.S. The Eyes Absent proteins in development and disease. Cell. Mol. Life Sci. 2013, 70, 1897–1913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Hou, X.; Peng, J.X.; Hao, X.Y.; Cai, J.P.; Liang, L.J.; Zhai, J.M.; Zhang, K.S.; Lai, J.M.; Yin, X.Y. DNA Methylation Profiling Identifies EYA4 Gene as a Prognostic Molecular Marker in Hepatocellular Carcinoma. Ann. Surg. Oncol. 2014, 21, 3891–3899. [Google Scholar] [CrossRef] [PubMed]
  184. Kim, S.J.; Tae, C.H.; Hong, S.N.; Min, B.H.; Chang, D.K.; Rhee, P.L.; Kim, J.J.; Kim, H.C.; Kim, D.H.; Kim, Y.H. EYA4 Acts as a New Tumor Suppressor Gene in Colorectal Cancer. Mol. Carcinog. 2015, 54, 1748–1757. [Google Scholar] [CrossRef] [PubMed]
  185. Luo, M.; Li, Y.; Shi, X.; Yang, W.; Zhou, F.; Sun, N.; He, J. Aberrant methylation of EYA4 promotes epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Cancer Sci. 2018, 109, 1811–1824. [Google Scholar] [CrossRef]
  186. Peters, I.; Eggers, H.; Atschekzei, F.; Hennenlotter, J.; Waalkes, S.; Tränkenschuh, W.; Groãhennig, A.; Merseburger, A.S.; Stenzl, A.; Kuczyk, M.A.; et al. GATA5 CpG island methylation in renal cell cancer: A potential biomarker for metastasis and disease progression. BJU Int. 2012, 110, E144–E152. [Google Scholar] [CrossRef]
  187. Guo, M.; Akiyama, Y.; House, M.G.; Hooker, C.M.; Heath, E.; Gabrielson, E.; Yang, S.C.; Han, Y.; Baylin, S.B.; Herman, J.G.; et al. Hypermethylation of the GATA genes in lung cancer. Clin. Cancer Res. 2004, 10, 7917–7924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Wen, X.Z.; Akiyama, Y.; Pan, K.F.; Liu, Z.J.; Lu, Z.M.; Zhou, J.; Gu, L.K.; Dong, C.X.; Zhu, B.D.; Ji, J.F.; et al. Methylation of GATA-4 and GATA-5 and development of sporadic gastric carcinomas. World J. Gastroenterol. 2010, 16, 1201–1208. [Google Scholar] [CrossRef]
  189. Akiyama, Y.; Watkins, N.; Suzuki, H.; Jair, K.-W.; van Engeland, M.; Esteller, M.; Sakai, H.; Ren, C.-Y.; Yuasa, Y.; Herman, J.G.; et al. GATA-4 and GATA-5 Transcription Factor Genes and Potential Downstream Antitumor Target Genes Are Epigenetically Silenced in Colorectal and Gastric Cancer. Mol. Cell. Biol. 2003, 23, 8429–8439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Hempstead, B.L. Brain-Derived Neurotrophic Factor: Three Ligands, Many Actions. Trans. Am. Clin. Climatol. Assoc. 2015, 126, 9–19. [Google Scholar]
  191. Lu, B.; Pang, P.T.; Woo, N.H. The yin and yang of neurotrophin action. Nat. Rev. Neurosci. 2005, 6, 603–614. [Google Scholar] [CrossRef] [Green Version]
  192. Tajbakhsh, A.; Mokhtari-Zaer, A.; Rezaee, M.; Afzaljavan, F.; Rivandi, M.; Hassanian, S.M.; Ferns, G.A.; Pasdar, A.; Avan, A. Therapeutic Potentials of BDNF/TrkB in Breast Cancer; Current Status and Perspectives. J. Cell. Biochem. 2017, 118, 2502–2515. [Google Scholar] [CrossRef]
  193. Mazouffre, C.; Geyl, S.; Perraud, A.; Blondy, S.; Jauberteau, M.O.; Mathonnet, M.; Verdier, M. Dual inhibition of BDNF/TrkB and autophagy: A promising therapeutic approach for colorectal cancer. J. Cell. Mol. Med. 2017, 21, 2610–2622. [Google Scholar] [CrossRef]
  194. Kundakovic, M.; Gudsnuk, K.; Herbstman, J.B.; Tang, D.; Perera, F.P.; Champagne, F.A. DNA methylation of BDNF as a biomarker of early-life adversity. Proc. Natl. Acad. Sci. USA 2015, 112, 6807–6813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Boehm, E.; Zaganelli, S.; Maundrell, K.; Jourdain, A.A.; Thore, S.; Martinou, J.C. FASTKD1 and FASTKD4 have opposite effects on expression of specific mitochondrial RNAs, depending upon their endonuclease-like RAP domain. Nucleic Acids Res. 2017, 45, 6135–6146. [Google Scholar] [CrossRef]
  196. Jourdain, A.A.; Popow, J.; De La Fuente, M.A.; Martinou, J.C.; Anderson, P.; Simarro, M. Survey and summary: The FASTK family of proteins: Emerging regulators of mitochondrial RNA biology. Nucleic Acids Res. 2017, 45, 10941–10947. [Google Scholar] [CrossRef] [Green Version]
  197. Wang, A.; Zhao, C.; Liu, X.; Su, W.; Duan, G.; Xie, Z.; Chu, S.; Gao, Y. Knockdown of TBRG4 affects tumorigenesis in human H1299 lung cancer cells by regulating DDIT3, CAV1 and RRM2. Oncol. Lett. 2018, 15, 121–128. [Google Scholar] [CrossRef] [PubMed]
  198. Wang, J.; Luo, Q.; Liu, M.; Zhang, C.; Jia, Y.; Tong, R.; Yang, L.; Fu, X. TBRG4 silencing promotes progression of squamous cell carcinoma via regulation of CAV-1 expression and ROS formation. Cell. Mol. Biol. 2020, 66, 157–164. [Google Scholar] [CrossRef] [PubMed]
  199. Huang, F.; Zhou, P.; Wang, Z.; Zhang, X.L.; Liao, F.X.; Hu, Y.; Chang, J. Knockdown of TBRG4 suppresses proliferation, invasion and promotes apoptosis of osteosarcoma cells by downregulating TGF-β1 expression and PI3K/AKT signaling pathway. Arch. Biochem. Biophys. 2020, 686, 108351. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram depicting the process of the identification, inclusion and exclusion criteria of selected studies.
Figure 1. Flow diagram depicting the process of the identification, inclusion and exclusion criteria of selected studies.
Biomolecules 11 01722 g001
Table 1. Methylation biomarkers in CRC diagnosis.
Table 1. Methylation biomarkers in CRC diagnosis.
GenePathwaysType of MarkerSamplesReferences
ALX4Tumor suppressor of Wnt/β-catenin pathway [40]DiagnosisPlasma[41,42,43,44,45,46,47]
BCAT1mTOR1 regulatorDiagnosis/PrognosisPlasma, FFPE[41,44,48,49,50]
BMP3Tumor suppressor via TAK1/JNK and ActRIIB/SMAD2-dependent gene [51]DiagnosisPlasma, Stool, FFPE[41,43,46,48,49,52]
EYA4Tumor suppressor of Wnt/β-catenin pathway [53]Diagnosis/PrognosisPlasma[41,43,44,54]
GATA4/GATA5Tumor suppressor of Wnt/β-catenin pathway [55]DiagnosisPlasma, Stool[41,43,44,49,56]
HICTumor suppressor of IL-6/STAT3 pathway [57]PrognosisPlasma[41,43,44,56]
HLTFTumor suppressor TGF- β/SMAD pathway [58]Diagnosis/Prognosis/PredictivePlasma, Stool[43,44,48,59]
IGFBP-3Involved in TGF-β independent function [60]Prognosis/PredictivePlasma, FFPE[42,45,54,61,62]
IKZF1/IKZF2Related to binding to relevant genes as PTPN6 and MEIS2 [50,63]Diagnosis/PrognosisPlasma, FFPE[41,44,48,49,50]
IRF4Promotes proliferation through JNK/Jun pathway [64]DiagnosisPlasma[41,44,49]
ITGA4Increases migration through interacting with VCAM-1 [65]DiagnosisStool, FFPE[43,44,48,56]
LINE-1Inserts into genes causing gene disruption [66]Diagnosis/PrognosisPlasma[41,42,44,67]
MGMTDNA repair system [68] DiagnosisPlasma, Stool, FFPE[41,42,43,44,45,49,69]
MLH1Member of mismatch repair (MMR) machinery [70]DiagnosisStool, FFPE[42,43,44,48,49,54,69,71,72,73]
NDRG4Inhibition of PI3K/AKT pathway [74] Diagnosis/PrognosisStool, FFPE[41,43,48,49,52,56]
OSMRActivation of STAT3/FAK/Src pathway [75]DiagnosisPlasma, Stool[41,43,48,56]
PRIMA1Activates mutant p53 pathway [76]DiagnosisPlasma[41,49,52]
RASSF1A/RASSF2AInteracts with Hippo and Wnt Pathway [77]Diagnosis/Prognosis/PredictivePlasma, Stool, FFPE[41,43,44,49,52,78]
SDC2Promotes MAPK and EMT pathway activation [79]DiagnosisPlasma, Stool[41,44,45,46,49,52,56,80]
SEPT9Activates Rho/ROCK and FAK pathway [81]Diagnosis/Prognosis/PredictivePlasma, FFPE[41,42,45,46,48,49,52,54,82]
SFRP1/SFRP2Inactivation of Wnt pathway [83]DiagnosisPlasma, Stool, FFPE[41,43,44,45,48,49,52,56]
TAC1-Diagnosis/Prognosis/PredictivePlasma[41,44,54,82]
TFPI2Involved in TWIST-integrin α5 [84]DiagnosisPlasma, Stool[43,48,49]
VIMInteracts with MAPK/ERK Pathway [85]Diagnosis/PrognosisPlasma, Stool, FFPE, Urine[41,45,46,48,49,52,56]
INHBBActivates ERK/Smad2/3 pathway [86]Diagnosis/PrognosisFFPE[86]
BDNFActivates AKT pathway [87]PrognosisFFPE[88]
SMOC2Activates Wnt/β-catenin pathway [89] PrognosisFFPE[89]
FFPE: Formalin-Fixed Paraffin-Embedded tissue from Colorectal biopsy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gutierrez, A.; Demond, H.; Brebi, P.; Ili, C.G. Novel Methylation Biomarkers for Colorectal Cancer Prognosis. Biomolecules 2021, 11, 1722. https://doi.org/10.3390/biom11111722

AMA Style

Gutierrez A, Demond H, Brebi P, Ili CG. Novel Methylation Biomarkers for Colorectal Cancer Prognosis. Biomolecules. 2021; 11(11):1722. https://doi.org/10.3390/biom11111722

Chicago/Turabian Style

Gutierrez, Alvaro, Hannah Demond, Priscilla Brebi, and Carmen Gloria Ili. 2021. "Novel Methylation Biomarkers for Colorectal Cancer Prognosis" Biomolecules 11, no. 11: 1722. https://doi.org/10.3390/biom11111722

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

Article Metrics

Back to TopTop