Skip to main content

Methylation as a critical epigenetic process during tumor progressions among Iranian population: an overview

Abstract

Cancer is one of the main health challenges and leading causes of deaths in the world. Various environmental and genetic risk factors are associated with tumorigenesis. Epigenetic deregulations are also important risk factors during tumor progression which are reversible transcriptional alterations without any genomic changes. Various mechanisms are involved in epigenetic regulations such as DNA methylation, chromatin modifications, and noncoding RNAs. Cancer incidence and mortality have a growing trend during last decades among Iranian population which are significantly related to the late diagnosis. Therefore, it is required to prepare efficient molecular diagnostic panels for the early detection of cancer in this population. Promoter hyper methylation is frequently observed as an inhibitory molecular mechanism in various genes associated with DNA repair, cell cycle regulation, and apoptosis during tumor progression. Since aberrant promoter methylations have critical roles in early stages of neoplastic transformations, in present review we have summarized all of the aberrant methylations which have been reported during tumor progression among Iranian cancer patients. Aberrant promoter methylations are targetable and prepare novel therapeutic options for the personalized medicine in cancer patients. This review paves the way to introduce a non-invasive methylation specific panel of diagnostic markers for the early detection of cancer among Iranians.

Background

Cancer is the main and second cause of death in developed and developing countries, respectively [1]. It is the third most common cause of death among Iranian population [2]. Gastric and breast cancers are the most common malignancies among Iranian men and women, respectively [3]. Lifestyle and environmental changes were occurred during the recent years due to the rapid industrialization in Iran [1]. Various environmental risk factors including tobacco smoking, environmental chemicals, high dietary salt intake, bacterial and viral infections, and gastro-esophageal reflux have been reported for cancer among Iranians [4,5,6]. Epigenetic involves the heritable and reversible transcriptional changes without any DNA sequence alterations which are involved in the early stages of tumor progression, embryogenesis, imprinting, and X-chromosome inactivation [7, 8]. It is regulated via different processes such as DNA methylation, chromatin modifications, and noncoding RNAs that play critical roles during tumor initiation and progression [9,10,11]. DNA methylation involves the transfer of a methyl group to the cytosine that is catalyzed by DNA methyltransferase (DNMT) [12, 13]. DNMT inhibitors are categorized into the nucleoside analogs and the non-nucleoside inhibitors [14]. The azacytidine and decitabine as nucleoside analogs are the most common DNMT inhibitors and epigenetic modulators in cancer therapy [15]. Non-nucleoside compounds such as hydralazine and procainamide inhibit the methylation through a DNA incorporation independent mechanism [14]. Curcumin belongs to the Non-nucleoside DNMT inhibitors that bind with DNMT1 catalytic domain [16]. DNA hypo methylation leads to aberrant activation of oncogenes while the hyper methylation is associated with inhibition of tumor suppressor genes. Various tumor suppressor genes such as p16, MutL homolog 1 (MLH1), and breast cancer type 1 susceptibility protein (BRCA1) which are involved in DNA repair, cell cycle, cell adhesion, and apoptosis have been shown to undergo tumor-specific silencing by hyper methylation [17,18,19]. Histone modifications through histone acetyl-transferase (HATs), histone methyltransferase (HMTs), kinases, ubiquitin ligases, and sumoligases are important regulatory processes in chromatin remodeling, gene expression, and carcinogenesis [20, 21]. Micro RNAs are also the post transcriptional regulators of more than 60% of protein-coding genes during various cellular processes that can be associated with tumorigenesis [22, 23]. Epigenetic markers are considered as emerging diagnostic and prognostic biomarkers in cancer [24, 25]. Since, aberrant DNA methylation can be tracked in body fluids; they can be suggested as efficient diagnostic and prognostic markers in primary stages of tumor progression [26,27,28,29]. It has been reported that the majority of cancer related deaths among Iranian cases are associated with late diagnosis. Therefore, it is required to determine novel diagnostic markers for the early detection of cancer in this population. In present review we have summarized all of the significant epigenetic deregulations associated with tumor progression which have been reported until now among Iranian cancer patients (Fig. 1) (Table 1).

Fig. 1
figure 1

all of the aberrant methylations involved in tumor progression among Iranian population. Blue and red colors refer to the hypo and hypermethylation, respectively

Table 1 all of the aberrant methylations which have been reported among Iranian cancer patients

DNA repair

DNA hyper methylation of tumor suppressor genes have been reported in immortalized and transformed cells [73]. The O6-methylguanine DNA methyltransferase (MGMT) is involved in methylated guanosine repair through removing alkyl group from O6-alkyl guanine [74]. CDH1 as a cell-cell adhesion factor has a critical function in regulation of cell differentiation and normal structure of epithelial cells [75,76,77]. The MGMT and CDH1 promoter methylations were assessed among a sample of Iranian OSCC patients compared with normal margins. It has been observed that there were CDH1 and MGMT promoter hyper methylations in majority of cases. Moreover, there was a significant difference in MGMT mRNA expression levels between OSCC patients and controls. It was concluded that the MGMT methylation can be used as a proper marker of poor survival among Iranian patients with advanced OSCC [30]. Similarly, there was a significant inverse association between MGMT methylation and survival among a sample of American oral cancer patients, while the frequency of MGMT hyper methylation was noticeably lower than that among Iranian patients [78]. BRCA1 is involved in DNA repair, homologous recombination, and cell cycle regulation [79]. P16 is also a regulator of G1 to S phase during cell cycle progression [80]. Histone modification and DNA methylation of MGMT, BRCA-1, and P16 were assessed in a sample of Iranian breast cancer patients. It has been shown that the promoter methylation of MGMT and BRCA-1 were higher in malignant breast tumor (MBT) compared with benign breast tumor (BBT) cases, while the P16 promoter methylation was lower in MBT patients compared with BBT. There was a significant correlation between BRCA1 hyper methylation and poor survival. Moreover, MBT cases had hypo methylation of histone H4 lysine 20 (H4K20) and hypo acetylation of histone H3 on lysine 18 (H3K18). There was a significant inverse association between H3K9ac levels and tumor size in MBT cases [31]. Similarly, the ratio of MGMT promoter methylation was significantly higher in a sample of Chinese breast cancer patients compared with controls. Moreover, there was a significant converse correlation between MGMT methylation and levels of MGMT protein expression [81]. TP53 encodes a phosphoprotein involving in regulation of apoptosis, cell cycle, DNA repair, and differentiation [82]. It has been observed that there was a significant inverse association between MGMT promoter methylation and P53 expression among a sub population of Iranian glioblastoma patients. They showed MGMT methylation in about half of the patients [32]. P53 is stabilized by posttranslational modification in the primary stages of glioblastoma progression [83]. The MGMT suppression induces p53 mutation which can further deregulate the methylation pattern of MGMT [84]. The role of MGMT promoter methylation in glioblastoma progression was also assessed among German cases and was shown that there was a correlation between MGMT promoter methylation and survival in newly diagnosed patients [85]. It has been observed that there were significantly higher levels of MGMT promoter methylation in tumors compared with controls in a sample of Iranian colorectal cancer (CRC) patients. Moreover, they observed the MGMT promoter methylation in normal margins [33]. Another group also assessed the serum MGMT methylation which showed that the majority of a sample of Iranian CRC tumors had MGMT promoter methylation which were mainly moderately differentiated and located on left colon [34]. Similarly, MGMT promoter methylation has been reported in majority of brain metastases from CRC and corresponding primary tumors in a group of Italian patients [86]. The placenta have also a characteristic of tumor cells for a successful implantation of the embryo in uterus during early pregnancy, in which it invades into the host tissues, escapes from immune response, and promotes angiogenesis. There are similar DNA methylation patterns between the tumors and placenta. The expression profile of the genes located within cancer/placenta hypomethylated blocks were assessed for CRC that showed the epigenetic regulation of NF-kB signaling during tumorigenesis and placentogenesis [87]. Human mutL homolog 1 (hMLH1) is one of the components of mismatch repair (MMR) system that is involved in the replacement of incorrectly paired nucleotides during DNA replication [88]. Therefore, the MMR aberrations can be associated with tumor progression [89, 90]. E-cadherin is a cell adhesion glycoprotein which is related to the tumor metastasis in a hyper methylated status [91]. It has been observed that there was a significant inverse association between the levels of hMLH1 mRNA expression and promoter methylation status in a sample of Iranian gastric cancer patients. Moreover, the hMLH1 hyper methylated tumors were significantly observed in advanced stage tumors. The E-cadherin promoter methylation was also significantly correlated with tumor stage and location [35].

Cell adhesion

Ras association domain family (RASSF) consists of 10 proteins that act as scaffolding agents in microtubule stability, mitotic cell division, apoptosis, cell migration, cell adhesion, inflammation, and NF-kB regulation [92]. RASSF6 and RASSF10 stabilize P53, regulate the cell cycle, inhibit tumor cell migration, and induce apoptosis [93,94,95,96,97]. Moreover, they are involved in regulation of NF-kB and WNT signaling pathways [93, 98]. Methylation status of RASSF6 and RASSF10 were assessed in a sample of Iranian Acute lymphocytic leukemia (ALL) cases. It was observed that the RASSF6 methylation was more frequent in B-Cell Acute Lymphoblastic Leukemia (B-ALL) cases compared with T-cell acute lymphoblastic leukaemia (T-ALL) cases, whereas the RASSF10 hyper methylation was more frequent in T-ALL compared with pre-B-ALL and B-ALL patients. Moreover, there was a significant correlation between RASSF6 hyper methylation and poor prognosis in pre-B-ALL patients which can be related to the NF-kB activation in the absence of RASSF6 [36]. HIC1 is a transcriptional suppressor involved in embryogenesis, P53 dependent apoptosis, cell cycle regulation, and WNT signaling regulation. It has been reported that there were significant correlations between tumor sizes more than 2 cm, lymph node involvement, and HIC1 methylation among a sub population of Iranian breast cancer patients. Moreover, there was a significant association between SASSF1A and HIC1 promoter methylation. It was concluded that the HIC1 and RASSF1A hyper methylations can be used as prognostic markers of breast cancer in this population [37]. Similarly, the RASSF1A methylation has been shown as an efficient prognostic marker in a sample of Saudi breast cancer patients [99]. Thyroid Stimulating Hormone Receptor (TSHR) is involved in growth and function of thyrocytes through stimulation of iodine uptake by NIS and iodine oxidation by thyroid peroxidase [100]. The RARb2 is a thyroid-steroid hormone receptor which is involved in embryogenesis through binding with retinoic acid [101]. It has been reported that there were higher rates of p16, TSHR, and RASSF1A hyper methylations in a sample of Iranian malignant papillary thyroid tumors compared with benign tumors [38]. TSHR methylation status was also introduced as a tumor marker for well-differentiated thyroid cancer among Turkish patients [102]. Integrin α4 binds with integrin β1 and β7 which are associated with cell adhesion to fibronectin [103]. The α4 integrin hyper methylation was observed in the majority of an Iranian prostate cancer patients group [39]. E-cadherin (CDH1) is a trans-membrane glycoprotein mainly expressed on the epithelial cells surface which is involved in Ca2+-dependent intracellular adhesion. CDH1 down regulation is associated with invasiveness and poor prognosis [104,105,106]. It has been shown that the tumor tissues had higher rates of CDH1 hyper methylation compared with normal samples in Iranian breast cancer patients. Moreover, there were significant associations between CDH1 promoter methylation, stage, grade, lymph node metastasis, and tumor size [40]. Another study on Iranian breast cancer cases also showed a significant higher ratio of CDH1 promoter hyper methylation in tumors compared with normal tissues [41]. The SPG20 is a multifunctional protein involved in intracellular EGFR traffic, cytokinesis, lipid droplet turnover, bone morphogenetic protein (BMP) signaling inhibition, and E3 ubiquitin ligases regulation [107,108,109,110,111]. It has been observed that the percentage of methylated reference (PMR) values in plasma samples of CRC patients were significantly higher than that in the healthy individuals among a sub population of Iranian subjects. The receiver-operating characteristics (ROC) curve analysis showed a sensitivity of 81.1% which was significantly higher than carcinoembryonic antigen (CEA) tumor marker (48.6%). Therefore, plasma SPG20 promoter methylation status can be an efficient noninvasive biomarker for CRC among Iranians [42].

Cell cycle

P14ARF is a cell cycle regulator that inhibits the MDM-2 mediated degradation of p53 [112,113,114]. It has been reported that there was higher ratio of p14ARF methylation in a sample of Iranian oral squamous cell carcinoma (OSCC) patients compared with controls which was also directly correlated with tumor stage [43]. Similarly, It was reported that there were significant associations between p14ARF hyper methylation, advanced stages, and lymph node involvement among Japanese OSCC patients [115]. The p53, p16INK4a, and MDM2 have critical roles during cell cycle regulation [116]. MDM2 is an oncogene that inactivates p53 during tumorigenesis [117, 118]. The G1 to S cell cycle progression is regulated by CCND1 in relationship with CDK 4/6 which is suppressed by the inhibitor of cyclin dependent kinase 4 (INK4) [119]. The INK4 family includes p19INK4D, p15INK4B, p18INK4C, and p16INK4A [112]. It has been shown that the p16INK4A inhibits G0/G1 cell cycle through suppression of CCND1–CDK4/6 complex. The p16INK4A also functions as a negative regulator of retinoblastoma protein (Rb) and regulates the cell cycle in G1/S phase in a p53-dependent pathway through CDK4 and CDK6 inhibitions [120, 121]. It has been observed that there was a significant inverse correlation between p16 hyper methylation and P53 expression in a sample of Iranian esophageal squamous cell carcinoma (ESCC) patients [44]. The serum p16 promoter hyper methylation was associated with poor prognosis among Japanese ESCC patients [122]. Another study on Iranian subjects assessed the p16 methylation status between familial and sporadic ESCC cases compared with healthy subjects. It was shown that the sporadic cases had higher ratio of p16 methylation compared with familial ESCC cases, while there was not any p16 methylation among controls [45]. Similarly, p16 methylation rate in sporadic was higher than that in familial Korean colorectal cancer patients [123]. Another group has been reported that there were direct correlations between p16 hyper methylation, tumor grade, HP infection, and smoking in a subpopulation of Iranian OSCC cases [46]. Another group has been reported that there was higher ratio of p16 and p15 methylations in tumors compared with normal margins in a sample of Iranian OSCC patients [47]. The aberrant methylation of the p15 and p16 have been also reported during OSCC progression among Japanese patients [115]. The p16 promoter hyper methylation was also involved in primary stages of sporadic breast cancer in Iranian patients [48]. Another study on Iranian gastric cancer patients showed that the P16 hyper methylation was less frequent in well-differentiated tumors and more frequent in older patients [49]. DBC2 is a tumor suppressor gene that functions through down-regulation of CCND1 [124]. It is also involved in regulation of ubiquitination, cell cycle, protein transport, apoptosis, and cytoskeleton [125,126,127,128]. It has been reported that there was significantly higher frequency of DBC2 methylation in tumor and blood samples of a group of Iranian breast cancer patients compared with normal margins [50]. Similarly, a study on Chinese breast cancer patients showed higher DBC2 methylation in breast tumors compared with normal tissues. Moreover, there was a significant correlation between DBC2 promoter methylation and lymph node metastasis [129]. The 14-3-3σ is a p53-related G2/M suppressor associated with DNA repair and apoptosis [130, 131]. It has been reported that there was higher ratio of 14-3-3σ promoter methylation in a sample of Iranian breast tumors compared with normal tissues [51]. Similarly, 14-3-3σ promoter methylation was higher in Chinese breast tumors compared with benign and normal tissues [132]. Ubiquitin-proteasome system (UPS) has a critical role in cell cycle regulation [133, 134]. The protein modification by ubiquitin is an important strategy for the elimination of abnormal proteins. UPS is also associated with pathophysiological processes during tumor progression [133]. Ubiquitination is performed by ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin-protein ligases (E3). UBE2Q2 and UBE2Q1 are members of E2 ubiquitin-conjugating enzyme family [135]. UBE2Q2 functions as an oncogene during CRC initiation and progression [136, 137]. It has been observed that there were higher levels of methylated UBE2Q1 in colorectal tumor samples compared with normal margins among a sub population of Iranian subjects [52]. Aberrant methylation of cell cycle regulators during tumor progressions among Iranian patients are illustrated in Fig. 2.

Fig. 2
figure 2

aberrant methylation of cell cycle regulators during tumor progressions among Iranian patients

Tyrosine kinase and G-protein-coupled receptors

The downstream of tyrosine kinase type 7 (DOK7) is an adaptor protein that induces the acetylcholine receptors (AChR) through muscle-specific kinase (MUSK) [138, 139]. It also inhibits proliferation and migration of cancer cells via AKT signaling pathway [140]. The vimentin is an intermediate filament that plays important roles in epithelial mesenchymal transition (EMT), immune response, and cytoskeleton structure [141,142,143,144]. CXCR4 (C-X-C chemokine receptor type 4) is a receptor involved in calcium signaling, transcription, chemotaxis, cell survival, and proliferation. The CXCR4 promoter hypo methylation has been detected in melanoma, breast, and pancreatic cancers [145, 146]. SAM pointed domain containing ETS transcription factor (SPEDF) is a tumor suppressor involved in tumor progression via p21/CIP1 regulation [147, 148]. It has been reported that there were significant DOK7, VIM, and CXCR4 hypo methylations in a sub population of Iranian breast cancer cases compared with normal subjects. Moreover, there were significant correlations between DOK7 and VIM methylations and negative ER status [53]. Another reports also showed DOK7 and VIM hyper methylations in Spanish and Australian breast cancer patients, respectively [140, 149]. The growth hormone secretagogue receptor (GHSR) belongs to the G-protein-coupled receptor (GPCR) family which acts as a receptor for ghrelin [150]. Ghrelin is associated with regulation of glucose and lipid metabolism and activates Ca2+ and P13K/AKT signaling pathways that are contributed with secretion of growth hormone in pituitary cells [151,152,153]. It has been reported that there was significant hyper methylation of GHSR in a sample of Iranian gastric cancer tissues compared with normal margins [54]. Similarly, GHSR hyper methylation was also observed in Italian colorectal cancer tissues compared with normal tissues [154]. Endothelin receptor type B (EDNRB) is a G protein coupled receptor involved in embryonic and enteric ganglia development [155,156,157]. Decreased expression of EDNRB leads to proliferation, angiogenesis, and metastasis through ET1 signaling pathway during tumor progression [158,159,160]. KISS1R is also a G-protein coupled receptor that is associated with tumor metastasis by ERK inhibition and MMP-9 reduction [161, 162]. It has been reported that there was higher frequency of EDNRB hyper methylation in a sample of Iranian colorectal cancer tissues compared with normal margins [55]. Similarly, the Chinese colorectal cancer tumors had significantly higher frequency of EDNRB promoter hyper methylation compared with normal tissues [155].

Signaling pathways

The WNT signaling pathway is involved in embryogenesis and tumor progression [163,164,165]. DNA methylation of APC, AXIN2, SFRP, and DKK as important WNT inhibitors have been reported in colorectal cancer patients [166,167,168,169]. It has been observed that there were significant correlations between APC and DDK3 aberrant promoter methylations and age and sex, respectively among a sub population of Iranian colorectal patients. The SFRP4 and WIF1 promoter methylations were significantly associated with stage and grade. Moreover, there were significant correlations between SFRP2 and SFRP5 methylations and tumor type. Univariate analysis also indicated the WIF1 promoter methylation as a prognostic factor in colorectal cancer patients [56]. Adenomatous polyposis coli (APC) is a tumor suppressor involved in regulation of cell growth through WNT signaling. In normal cells, free β-catenin is phosphorylated by Axin-APC-GSK3β complex which results in β-catenin proteasomal degradation and reduced expression of WNT signaling target genes [170]. It has been reported that there were higher rates of APC hyper methylation in tumor tissues compared with normal tissues in a sample of Iranian ESCC patients. Moreover, the hyper methylated cases had lower survival rates. There was also a direct association between APC promoter hyper methylation and grade of tumor differentiation [57]. The Chinese esophageal cancer tumors had also higher rates of APC methylation compared with controls [171]. SFRP2 is one of the negative regulators of WNT signaling pathway. It has been observed that there were higher levels of SFRP2 hyper methylation in a sample of Iranian CRC patients compared with healthy subjects [58]. Similarly, there were also high levels of SFRP1 and SFRP2 hyper methylations among a group of Hungarian CRC patients [172]. Phosphatase and tensin homolog (PTEN) is a suppressor of PI3K/AKT pathways which inhibits signal transduction from HER1, HER2, and IGFR growth factor receptors through the P13K/AKT signaling [173, 174]. It forms a nuclear complex with p53 to inhibit the p53 decomposition [175, 176]. Moreover, it induces G0-G1 cell cycle arrest by suppression of CCND1 and ERK/MAPK pathway [177]. MiR-21 promotes tumor cell growth and invasion by PTEN targeting [178,179,180]. It has been reported that there was a significant association between PTEN promoter methylation and expression in a sample of Iranian colorectal cancer patients. The levels of PTEN mRNA expressions were inversely associated with miR-21 expression. Moreover, there were converse significant associations between PTEN expression, tumor size, survival, and tumor stage [59]. Similarly, it has been observed that there was a significant correlation between PTEN promoter methylation and expression among sporadic Indian breast cancer patients [181]. Another study on Iranian sporadic breast cancer patients showed that there were correlations between PTEN hyper methylation, advanced stages, and lymph node involvement. They suggested the PTEN promoter methylation as a prognostic marker for the response to PTEN-dependent therapy [60]. Iranian Kurdish breast cancer patients also had a higher frequency of PTEN methylation compared with healthy controls. The female relatives of patients had also a significantly higher frequency of PTEN methylation compared with controls. Moreover, the PTEN methylation was higher in patients between 40-80 years old compared with patients who were between 29–39 years old which showed increased PTEN methylation in higher ages [61].

Developmental factors

Homeobox protein aristaless-like (ALX4) is a homeodomain transcription factor associated with bone, skin, and hair follicle development [182, 183]. It has been reported that there was a significant difference of ALX4 methylation status between a sample of Iranian colorectal cancer patients and controls which introduced that as an efficient marker for the early detection of colorectal cancer in this population [62]. Similarly, ALX4 methylation was observed among German patients with colorectal, esophageal, and gastric cancers [184]. Paired Box 5 (PAX5) is belonged to the PAX family of tissue-specific transcription factors associated with development and embryogenesis. Deregulation of PAX5 has been observed in various types of human tumors [185]. It is involved in neoplastic transformation through CD19 regulation which suppresses growth regulators [186]. Moreover, PAX5 is a functional tumor-suppressor in liver carcinogenesis by P53 regulation [187]. Methylation status of PAX5 was assessed in blood samples of Iranian gastric cancer patients compared with healthy blood samples. There were higher levels of PAX5 methylation in the blood samples of patients compared with controls. There were also significant correlations between the mean expression levels of PAX5, age, and promoter methylation status [63]. It has been shown that there was a significant correlation between PAX5 methylation and survival in a sample of Chinese gastric cancer patients [188]. MicroRNAs (miRNAs) are one of the main factors in gene regulation in normal and tumor tissues which function through 3’ un-translated region (3'UTR) dependent translational inhibition [189,190,191,192]. The miRNAs expressions are regulated by methylation, alkylation, and acetylation [193,194,195]. MiR-192-2 induces the apoptosis through targeting SOX4 in gastric tumor cells [196]. It has been shown that there was a significant difference of miR-129-2 methylation between a sample of Iranian gastric cancer and healthy cases [64].

Nuclear receptors

Estrogen and its receptors are involved in breast epithelial cell homeostasis through regulation of proliferation, differentiation, and apoptosis. The methylation of estrogen receptors including ER-α and ER-β play important role in primary breast cancer progression. Loss of ER-α is an important mechanism of hormone resistance in breast cancer [197,198,199,200,201]. It has been observed that there was significantly higher ER4 methylation in tumors with P53 expression among a sub population of Iranian breast cancer patients. The ER5 methylation was observed in tumors with lymph node metastasis and higher grades. ER4 and ER5 methylations in postmenopausal females were higher than that in premenopausal cases. There was also significant higher frequency of ER5 methylation in Her-2+ tumors. ER-α hyper methylation was frequently observed in invasive ductal cell carcinoma patients. Moreover, there was a direct correlation between ER5 methylation and age [65]. ER-α promoter methylation status in Egyptian breast cancer patients were assessed using MSP method which showed higher ratio of methylation in ER3 and ER5 compared with Iranian patients [202]. Another study assessed the ER-α methylation among Iranian breast cancer patients which showed methylation in majority of basal and Her2+ tumors. There was a correlation between ER-α methylation and poor prognosis in basal and Her2+ tumors. They showed that the ER-α methylation plays an important role in aggressive breast tumors in this population [66]. ER3 and ER5 methylations have been also reported in majority of a sample of Iranian ER negative breast tumors [67]. Retinoic acid receptor beta (RARB) belongs to the thyroid-steroid hormone receptors which bind with retinoic acid to mediate embryogenesis and cell differentiation. It has been reported that there was higher frequency of RARB hyper methylation in poor prognosis cases compared with good prognosis in a sample of Iranian prostate cancer patients. The p16 hyper methylation in poor prognostic cases was also higher than patients with good prognosis [68]. Similarly, RARB methylation was associated with a higher prostate cancer risk among American patients [203].

Apoptosis

The apoptotic protease activating factor 1 (APAF1) and caspase 8 (CASP8) genes are important regulators of apoptotic pathways. Extrinsic apoptosis pathway is mediated by CD95, FADD, and procaspase-8 in which the CASP8 triggers the proteolytic activation of other caspases and cleavage of cellular substrates [204,205,206,207,208,209]. Cytochrome c is released from the mitochondria following DNA damage and binds to the APAF1 in cytosol that results in CASP9, CASP3, CASP6, and CASP7 serial activations and apoptosis [210, 211]. It has been shown that there was a significant association between the levels of APAF1 methylation, tumor stage, and grade in blood samples of a subpopulation of Iranian gastric cancer patients. Moreover, the CASP8 methylation status in blood samples of patients was significantly correlated with age [69]. Chinese gastric cancer patients had also significant higher ratio of APAF1 methylation in their tumor tissues compared with normal margins [212]. Fas belongs to the tumor necrosis factor receptor (TNF-R) family that is normally expressed in lymph nodes and spleen [213]. Fas Ligand (FasL) acts as a ligand for Fas receptor that activates CASP8 through Fas-associated death domain (FADD). Subsequently, CASP8 activates CASP3 and CASP7 that mediate cell death. Moreover, it cleaves BID to generate truncated BID which enters to the mitochondria and triggers the mitochondrial apoptotic pathway [214, 215]. It has been observed that there was aberrant FAS promoter methylation in majority of a sample of Iranian oral squamous cell carcinoma patients, whereas the aberrant FADD methylation was observed in a minority of cases [70]. Ataxia telangiectasia mutated (ATM) is a serine threonine kinase which is activated by DNA double-strand break (DSB). Deregulated expression of E2F1 transcription factor up regulates ATM that leads to the apoptosis induction, cell cycle regulation, and DNA repair via phosphorylation of CHK1, CHK2, P53, and CDC25 [216, 217]. ATM promoter methylation was more frequent in meningioma and glioma patients in a sample of Iranian cases. There was a significant correlation between higher grades of brain tumors and ATM promoter methylation. There was also a significant association between ATM promoter methylation and RB expression. Moreover, there was a significant association between D1853N polymorphism and ATM promoter methylation [71]. Another report assessed the promoter methylation status of ATM among Indian breast cancer cases which showed significant higher ratio of promoter hyper methylation in tumor tissues compared with normal samples. Moreover, there were significant correlations between ATM promoter methylation, age, tumor size, and advanced tumor stages [218]. Cytotoxic T-lymphocyte-associated antigen-4 (CTLA4) is a receptor that acts as an immune check point in regulation of immune responses and is expressed on activated T-cells [219, 220]. It induces the PKB/AKT activation which up regulates the BCL-XL/BCL-2 [221]. It has been observed that there was significantly higher frequency of CTLA4 promoter methylation in a sample of Iranian gastric cancer patients compared with normal margins [72]. In contrast, a study on Qatari breast cancer patients showed significant hypo methylation of CpG islands in promoter region of CTLA-4 in tumors compared with normal margins [222]. Role of aberrant methylations in regulation of apoptosis during tumor progressions among Iranian patients are illustrated in Fig. 3.

Fig. 3
figure 3

role of aberrant methylation in regulation of apoptosis during tumor progressions among Iranian patients

Conclusions

Regarding the recent life style changes, there is a growing cancer incidence and mortality in Iran which is related to the late diagnosis. Epigenetic markers are considered as emerging diagnostic and prognostic biomarkers in cancer. Therefore, in present review we summarized all of the methylation abnormalities during tumor progressions which have been reported until now among Iranian patients. It was frequently observed that the p16 and CDH1 aberrant promoter methylations can be involved in tumor progression of ESCC, thyroid, oral, breast, gastric, and prostate cancers. The MGMT promoter hyper methylation was also frequently reported in CRC, GB, BC, and OSCC. Therefore, p16, CDH1, and MGMT methylation status can be suggested as a general methylation based panel marker for all cancers in Iranian patients. Moreover, there were various reports of PTEN and ER-α promoter hyper methylations in Iranian BC patients which introduces them as methylation based markers of BC in this population. Generally, this review paves the way to introduce a non-invasive methylation specific panel of diagnostic markers for the early detection of cancer among Iranian populations.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

DNMT:

DNA methyltransferase

HATs:

Histone acetyl-transferase

HMTs:

Histone methyltransferase

MBT:

Malignant breast tumor

BBT:

Benign breast tumor

CDH1:

E-cadherin

BMP:

Bone morphogenetic protein

PMR:

Percentage of methylated reference

ROC:

Receiver-operating characteristics

CEA:

Carcinoembryonic antigen

Rb:

Retinoblastoma protein

UPS:

Ubiquitin-proteasome system

DOK7:

Downstream of tyrosine kinase type 7

AChR:

Acetylcholine receptors

EMT:

Epithelial mesenchymal transition

SPEDF:

SAM pointed domain containing ETS transcription factor

GHSR:

Growth hormone secretagogue receptor

GPCR:

G-protein-coupled receptor

PTEN:

Phosphatase and tensin homolog

miRNAs:

MicroRNAs

3'UTR:

3’ un-translated region

RARB:

Retinoic acid receptor beta

FasL:

Fas Ligand

TNF-R:

Tumor necrosis factor receptor

ATM:

Ataxia telangiectasia mutated

DSB:

Double-strand break

CTLA4:

Cytotoxic T-lymphocyte-associated antigen-4

OSCC:

Oral squamous cell carcinoma

MGMT:

O6-methylguanine DNA methyltransferase

MLH1:

MutL homolog 1

BRCA1:

Breast cancer type 1 susceptibility protein

H4K20 :

Histone H4 lysine 20

H3K18:

Histone H3 on lysine 18

MMR:

Mismatch repair

ALL:

Acute lymphocytic leukemia

B-ALL :

B-Cell Acute Lymphoblastic Leukemia

T-ALL:

T-cell acute lymphoblastic leukaemia

TSHR:

Thyroid Stimulating Hormone Receptor

RASSF:

Ras association domain family

ESCC:

Esophageal squamous cell carcinoma

EDNRB:

Endothelin receptor type B

ALX4:

Homeobox protein aristaless-like

PAX5:

Paired Box 5

APAF1 :

Apoptotic protease activating factor 1

CASP8:

Caspase 8

CRC:

Colorectal cancer

hMLH1:

Human mutL homolog 1

References

  1. Farhood B, Geraily G, Alizadeh A. Incidence and Mortality of Various Cancers in Iran and Compare to Other Countries: A Review Article. Iran J Public Health. 2018;47(3):309–16.

    PubMed  PubMed Central  Google Scholar 

  2. Dolatkhah R, et al. Increased colorectal cancer incidence in Iran: a systematic review and meta-analysis. BMC Public Health. 2015;15:997.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Mousavi SM, et al. Cancer incidence and mortality in Iran. Ann Oncol. 2009;20(3):556–63.

    Article  CAS  PubMed  Google Scholar 

  4. Azizi MH, Bahadori M, Azizi F. History of cancer in Iran. Arch Iran Med. 2013;16(10):613–22.

    PubMed  Google Scholar 

  5. Jamaati H, et al. Risk Factors for Lung Cancer Mortality in a Referral Center. Asian Pac J Cancer Prev. 2016;17(6):2877–81.

    CAS  PubMed  Google Scholar 

  6. Malekzadeh R, Derakhshan MH, Malekzadeh Z. Gastric cancer in Iran: epidemiology and risk factors. Arch Iran Med. 2009;12(6):576–83.

    PubMed  Google Scholar 

  7. Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet. 2006;7(1):21–33.

    Article  CAS  PubMed  Google Scholar 

  8. Tsai HC, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res. 2011;21(3):502–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kanwal R, Gupta S. Epigenetics and cancer. J Appl Physiol (1985). 2010;109(2):598–605.

    Article  CAS  Google Scholar 

  10. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27–36.

    Article  CAS  PubMed  Google Scholar 

  11. Schneider R, Grosschedl R. Dynamics and interplay of nuclear architecture, genome organization, and gene expression. Genes Dev. 2007;21(23):3027–43.

    Article  CAS  PubMed  Google Scholar 

  12. Lo PK, Sukumar S. Epigenomics and breast cancer. Pharmacogenomics. 2008;9(12):1879–902.

    Article  CAS  PubMed  Google Scholar 

  13. Naghitorabi M, et al. Quantitative evaluation of DNMT3B promoter methylation in breast cancer patients using differential high resolution melting analysis. Res Pharm Sci. 2013;8(3):167–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Yang X, et al. Targeting DNA methylation for epigenetic therapy. Trends Pharmacol Sci. 2010;31(11):536–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Gnyszka A, Jastrzebski Z, Flis S. DNA methyltransferase inhibitors and their emerging role in epigenetic therapy of cancer. Anticancer Res. 2013;33(8):2989–96.

    CAS  PubMed  Google Scholar 

  16. Liu Z, et al. Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett. 2009;19(3):706–9.

    Article  PubMed  CAS  Google Scholar 

  17. Baylin SB. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005;2(Suppl 1):S4–11.

    Article  CAS  PubMed  Google Scholar 

  18. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–28.

    Article  CAS  PubMed  Google Scholar 

  19. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dillon N. Gene regulation and large-scale chromatin organization in the nucleus. Chromosome Res. 2006;14(1):117–26.

    Article  CAS  PubMed  Google Scholar 

  21. Fischle W, Wang Y, Allis CD. Histone and chromatin cross-talk. Curr Opin Cell Biol. 2003;15(2):172–83.

    Article  CAS  PubMed  Google Scholar 

  22. Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 2010;79:351–79.

    Article  CAS  PubMed  Google Scholar 

  23. Mitra SA, Mitra AP, Triche TJ. A central role for long non-coding RNA in cancer. Front Genet. 2012;3:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Paluszczak J, Baer-Dubowska W. Epigenetic diagnostics of cancer--the application of DNA methylation markers. J Appl Genet. 2006;47(4):365–75.

    Article  PubMed  Google Scholar 

  25. Qureshi IA, Mehler MF. Developing epigenetic diagnostics and therapeutics for brain disorders. Trends Mol Med. 2013;19(12):732–41.

    Article  CAS  PubMed  Google Scholar 

  26. Diaz-Lagares A, et al. A Novel Epigenetic Signature for Early Diagnosis in Lung Cancer. Clin Cancer Res. 2016;22(13):3361–71.

    Article  CAS  PubMed  Google Scholar 

  27. Haggarty SJ. Epigenetic diagnostics for neuropsychiatric disorders: Above the genome. Neurology. 2015;84(16):1618–9.

    Article  PubMed  Google Scholar 

  28. Jakubowski JL, Labrie V. Epigenetic Biomarkers for Parkinson's Disease: From Diagnostics to Therapeutics. J Parkinsons Dis. 2017;7(1):1–12.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Van Neste L, et al. The epigenetic promise for prostate cancer diagnosis. Prostate. 2012;72(11):1248–61.

    Article  PubMed  CAS  Google Scholar 

  30. Kordi-Tamandani DM, et al. Promoter hypermethylation and expression profile of MGMT and CDH1 genes in oral cavity cancer. Arch Oral Biol. 2010;55(10):809–14.

    Article  CAS  PubMed  Google Scholar 

  31. Paydar P, et al. Epigenetic modulation of BRCA-1 and MGMT genes, and histones H4 and H3 are associated with breast tumors. J Cell Biochem. 2019;120(8):13726–36.

    Article  CAS  PubMed  Google Scholar 

  32. Shamsara J, et al. Association between MGMT promoter hypermethylation and p53 mutation in glioblastoma. Cancer Invest. 2009;27(8):825–9.

    Article  CAS  PubMed  Google Scholar 

  33. Farzanehfar M, et al. Evaluation of methylation of MGMT (O(6)-methylguanine-DNA methyltransferase) gene promoter in sporadic colorectal cancer. DNA Cell Biol. 2013;32(7):371–7.

    Article  CAS  PubMed  Google Scholar 

  34. Alizadeh Naini M, et al. O6-Methyguanine-DNA Methyl Transferase (MGMT) Promoter Methylation in Serum DNA of Iranian Patients with Colorectal Cancer. Asian Pac J Cancer Prev. 2018;19(5):1223–7.

    PubMed  Google Scholar 

  35. Moghbeli M, et al. Role of hMLH1 and E-cadherin promoter methylation in gastric cancer progression. J Gastrointest Cancer. 2014;45(1):40–7.

    Article  CAS  PubMed  Google Scholar 

  36. Younesian S, et al. DNA hypermethylation of tumor suppressor genes RASSF6 and RASSF10 as independent prognostic factors in adult acute lymphoblastic leukemia. Leuk Res. 2017;61:33–8.

    Article  CAS  PubMed  Google Scholar 

  37. Rasti M, et al. Association between HIC1 and RASSF1A promoter hypermethylation with MTHFD1 G1958A polymorphism and clinicopathological features of breast cancer in Iranian patients. Iran Biomed J. 2009;13(4):199–206.

    CAS  PubMed  Google Scholar 

  38. Mohammadi-asl J, et al. Qualitative and quantitative promoter hypermethylation patterns of the P16, TSHR, RASSF1A and RARbeta2 genes in papillary thyroid carcinoma. Med Oncol. 2011;28(4):1123–8.

    Article  CAS  PubMed  Google Scholar 

  39. Mostafavi-Pour Z, et al. Methylation of Integrin alpha4 and E-Cadherin Genes in Human Prostate Cancer. Pathol Oncol Res. 2015;21(4):921–7.

    Article  CAS  PubMed  Google Scholar 

  40. Naghitorabi M, et al. Quantitation of CDH1 promoter methylation in formalin-fixed paraffin-embedded tissues of breast cancer patients using differential high resolution melting analysis. Adv Biomed Res. 2016;5:91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Shargh SA, et al. Downregulation of E-cadherin expression in breast cancer by promoter hypermethylation and its relation with progression and prognosis of tumor. Med Oncol. 2014;31(11):250.

    Article  PubMed  CAS  Google Scholar 

  42. Rezvani N, et al. Detection of SPG20 gene promoter-methylated DNA, as a novel epigenetic biomarker, in plasma for colorectal cancer diagnosis using the MethyLight method. Oncol Lett. 2017;13(5):3277–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kordi-Tamandani DM, et al. Analysis of methylation patterns and expression profiles of p14ARF gene in patients with oral squamous cell carcinoma. Int J Biol Markers. 2010;25(2):99–103.

    Article  CAS  PubMed  Google Scholar 

  44. Taghavi N, et al. p16INK4a hypermethylation and p53, p16 and MDM2 protein expression in esophageal squamous cell carcinoma. BMC Cancer. 2010;10:138.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Abbaszadegan MR, et al. Aberrant p16 methylation, a possible epigenetic risk factor in familial esophageal squamous cell carcinoma. Int J Gastrointest Cancer. 2005;36(1):47–54.

    Article  CAS  PubMed  Google Scholar 

  46. Allameh A, Moazeni-Roodi A, Harirchi I, Ravanshad M, Motiee-Langroudi M, Garajei A, Hamidavi A, Mesbah-Namin SA. Promoter DNA methylation and mRNA expression level of p16 gene in oral squamous cell carcinoma: correlation with clinicopathological characteristics. Pathol Oncol Res. 2019;25(4):1535–43.

  47. Kordi-Tamandani DM, et al. Analysis of p15INK4b and p16INK4a gene methylation in patients with oral squamous cell carcinoma. Biochem Genet. 2012;50(5-6):448–53.

    Article  CAS  PubMed  Google Scholar 

  48. Vallian S, et al. Methylation status of p16 INK4A tumor suppressor gene in Iranian patients with sporadic breast cancer. J Cancer Res Clin Oncol. 2009;135(8):991–6.

    Article  CAS  PubMed  Google Scholar 

  49. Abbaszadegan MR, et al. p16 promoter hypermethylation: a useful serum marker for early detection of gastric cancer. World J Gastroenterol. 2008;14(13):2055–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hajikhan Mirzaei M, et al. Evaluation of Methylation Status in the 5'UTR Promoter Region of the DBC2 Gene as a Biomarker in Sporadic Breast Cancer. Cell J. 2012;14(1):19–24.

    PubMed  PubMed Central  Google Scholar 

  51. Gheibi A, et al. Study of promoter methylation pattern of 14-3-3 sigma gene in normal and cancerous tissue of breast: A potential biomarker for detection of breast cancer in patients. Adv Biomed Res. 2012;1:80.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Mokarram P, et al. Promoter Methylation Status of Two Novel Human Genes, UBE2Q1 and UBE2Q2, in Colorectal Cancer: a New Finding in Iranian Patients. Asian Pac J Cancer Prev. 2015;16(18):8247–52.

    Article  PubMed  Google Scholar 

  53. Shirkavand A, Boroujeni ZN, Aleyasin SA. Examination of methylation changes of VIM, CXCR4, DOK7, and SPDEF genes in peripheral blood DNA in breast cancer patients. Indian J Cancer. 2018;55(4):366–71.

    Article  PubMed  Google Scholar 

  54. Amini M, Foroughi K, Talebi F, Aghagolzade Haji H, Kamali F, Jandaghi P, Hoheisel JD, Manoochehri M. GHSR DNA hypermethylation is a new epigenetic biomarker for gastric adenocarcinoma and beyond. J Cell Physiol. 2019;234(9):15320–9.

  55. Mousavi Ardehaie R, et al. Aberrant methylated EDNRB can act as a potential diagnostic biomarker in sporadic colorectal cancer while KISS1 is controversial. Bioengineered. 2017;8(5):555–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Samaei NM, et al. Promoter methylation analysis of WNT/beta-catenin pathway regulators and its association with expression of DNMT1 enzyme in colorectal cancer. J Biomed Sci. 2014;21:73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Zare M, et al. Qualitative analysis of Adenomatous Polyposis Coli promoter: hypermethylation, engagement and effects on survival of patients with esophageal cancer in a high risk region of the world, a potential molecular marker. BMC Cancer. 2009;9:24.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Babaei H, Mohammadi M, Salehi R. DNA methylation analysis of secreted frizzled-related protein 2 gene for the early detection of colorectal cancer in fecal DNA. Niger Med J. 2016;57(4):242–5.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Yazdani Y, et al. The prognostic effect of PTEN expression status in colorectal cancer development and evaluation of factors affecting it: miR-21 and promoter methylation. J Biomed Sci. 2016;23:9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Sadeq V, Isar N, Manoochehr T. Association of sporadic breast cancer with PTEN/MMAC1/TEP1 promoter hypermethylation. Med Oncol. 2011;28(2):420–3.

    Article  CAS  PubMed  Google Scholar 

  61. Yari K, Payandeh M, Rahimi Z. Association of the hypermethylation status of PTEN tumor suppressor gene with the risk of breast cancer among Kurdish population from Western Iran. Tumour Biol. 2016;37(6):8145–52.

    Article  CAS  PubMed  Google Scholar 

  62. Salehi R, et al. Methylation pattern of ALX4 gene promoter as a potential biomarker for blood-based early detection of colorectal cancer. Adv Biomed Res. 2015;4:252.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Haghverdi MK, Moslemi E. Expression Rate and PAX5 Gene Methylation in the Blood of People Suffering from Gastric Cancer. Open Access Maced J Med Sci. 2018;6(9):1571–6.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Alizadeh N, Asadi M, Shanehbandi D, Zafari V, Shomali N, Asvadi T, Sepehri B. Evaluation of the methylation of MIR129-2 gene in gastric cancer. J Gastrointest Cancer. 2020;51(1):267–70.

  65. Ramezani F, et al. CpG island methylation profile of estrogen receptor alpha in Iranian females with triple negative or non-triple negative breast cancer: new marker of poor prognosis. Asian Pac J Cancer Prev. 2012;13(2):451–7.

    Article  PubMed  Google Scholar 

  66. Izadi P, et al. Association of poor prognosis subtypes of breast cancer with estrogen receptor alpha methylation in Iranian women. Asian Pac J Cancer Prev. 2012;13(8):4113–7.

    Article  PubMed  Google Scholar 

  67. Izadi P, et al. Promoter hypermethylation of estrogen receptor alpha gene is correlated to estrogen receptor negativity in Iranian patients with sporadic breast cancer. Cell J. 2012;14(2):102–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Ameri A, et al. Prognostic Value of Promoter Hypermethylation of Retinoic Acid Receptor Beta (RARB) and CDKN2 (p16/MTS1) in Prostate Cancer. Chin J Cancer Res. 2011;23(4):306–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Azarkhazin F, Tehrani GA. Detecting promoter methylation pattern of apoptotic genes Apaf1 and Caspase8 in gastric carcinoma patients undergoing chemotherapy. J Gastrointest Oncol. 2018;9(2):295–302.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Saberi E, et al. Analysis of methylation and mRNA expression status of FADD and FAS genes in patients with oral squamous cell carcinoma. Med Oral Patol Oral Cir Bucal. 2014;19(6):e562–8.

    PubMed  PubMed Central  Google Scholar 

  71. Mehdipour P, et al. Linking ATM Promoter Methylation to Cell Cycle Protein Expression in Brain Tumor Patients: Cellular Molecular Triangle Correlation in ATM Territory. Mol Neurobiol. 2015;52(1):293–302.

    Article  CAS  PubMed  Google Scholar 

  72. Kordi-Tamandani DM, et al. Analysis of promoter methylation, polymorphism and expression profile of cytotoxic T-lymphocyte-associated antigen-4 in patients with gastric cancer. J Gastrointestin Liver Dis. 2014;23(3):249–53.

    Article  PubMed  Google Scholar 

  73. Antequera F, Boyes J, Bird A. High levels of de novo methylation and altered chromatin structure at CpG islands in cell lines. Cell. 1990;62(3):503–14.

    Article  CAS  PubMed  Google Scholar 

  74. Burger PC, Green SB. Patient age, histologic features, and length of survival in patients with glioblastoma multiforme. Cancer. 1987;59(9):1617–25.

    Article  CAS  PubMed  Google Scholar 

  75. Handschuh G, et al. Tumour-associated E-cadherin mutations alter cellular morphology, decrease cellular adhesion and increase cellular motility. Oncogene. 1999;18(30):4301–12.

    Article  CAS  PubMed  Google Scholar 

  76. Mayer B, et al. E-cadherin expression in primary and metastatic gastric cancer: down-regulation correlates with cellular dedifferentiation and glandular disintegration. Cancer Res. 1993;53(7):1690–5.

    CAS  PubMed  Google Scholar 

  77. Moriyama N, et al. E-cadherin is essential for gastric epithelial restitution in vitro: a study using the normal rat gastric mucosal cell line RGM1. J Lab Clin Med. 2001;138(4):236–42.

    Article  CAS  PubMed  Google Scholar 

  78. Taioli E, et al. Recurrence in oral and pharyngeal cancer is associated with quantitative MGMT promoter methylation. BMC Cancer. 2009;9:354.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Zhang L, Long X. Association of BRCA1 promoter methylation with sporadic breast cancers: Evidence from 40 studies. Sci Rep. 2015;5:17869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Xing XB, et al. The Prognostic Value of p16 Hypermethylation in Cancer: A Meta-Analysis. PLoS One. 2013;8(6):e66587.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. An N, et al. Association Between MGMT Promoter Methylation and Breast Cancer: a Meta-Analysis. Cell Physiol Biochem. 2017;42(6):2430–40.

    Article  CAS  PubMed  Google Scholar 

  82. Bouchet BP, et al. p53 as a target for anti-cancer drug development. Crit Rev Oncol Hematol. 2006;58(3):190–207.

    Article  PubMed  Google Scholar 

  83. Lacroix M, Toillon RA, Leclercq G. p53 and breast cancer, an update. Endocr Relat Cancer. 2006;13(2):293–325.

    Article  CAS  PubMed  Google Scholar 

  84. Tovy A, et al. p53 is essential for DNA methylation homeostasis in naive embryonic stem cells, and its loss promotes clonal heterogeneity. Genes Dev. 2017;31(10):959–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. von Rosenstiel C, Wiestler B, Haller B, Schmidt-Graf F, Gempt J, Bettstetter M, Rihani L, Wu W, Meyer B, Schlegel J, Liesche-Starnecker F. Correlation of the quantitative level of MGMT promoter methylation and overall survival in primary diagnosed glioblastomas using the quantitative MethyQESD method. J Clin Pathol. 2020;73(2):112–5.

  86. De Maglio G, et al. MGMT promoter methylation status in brain metastases from colorectal cancer and corresponding primary tumors. Future Oncol. 2015;11(8):1201–9.

    Article  PubMed  CAS  Google Scholar 

  87. Nordor AV, et al. The early pregnancy placenta foreshadows DNA methylation alterations of solid tumors. Epigenetics. 2017;12(9):793–803.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Xiao XQ, et al. Polymorphisms of mismatch repair gene hMLH1 and hMSH2 and risk of gastric cancer in a Chinese population. Oncol Lett. 2012;3(3):591–8.

    Article  CAS  PubMed  Google Scholar 

  89. Moghbeli M, et al. High frequency of microsatellite instability in sporadic colorectal cancer patients in Iran. Genet Mol Res. 2011;10(4):3520–9.

    Article  CAS  PubMed  Google Scholar 

  90. Li GM. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008;18(1):85–98.

    Article  CAS  PubMed  Google Scholar 

  91. Wijnhoven BP, Dinjens WN, Pignatelli M. E-cadherin-catenin cell-cell adhesion complex and human cancer. Br J Surg. 2000;87(8):992–1005.

    Article  CAS  PubMed  Google Scholar 

  92. Schagdarsurengin U, et al. Frequent epigenetic inactivation of RASSF10 in thyroid cancer. Epigenetics. 2009;4(8):571–6.

    Article  CAS  PubMed  Google Scholar 

  93. Iwasa H, Jiang X, Hata Y. RASSF6; the Putative Tumor Suppressor of the RASSF Family. Cancers (Basel). 2015;7(4):2415–26.

    Article  CAS  Google Scholar 

  94. Jin Y, et al. RASSF10 suppresses hepatocellular carcinoma growth by activating P53 signaling and methylation of RASSF10 is a docetaxel resistant marker. Genes Cancer. 2015;6(5-6):231–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Liu W, et al. Ras-association domain family 10 acts as a novel tumor suppressor through modulating MMP2 in hepatocarcinoma. Oncogenesis. 2016;5(6):e237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mezzanotte JJ, et al. RASSF6 exhibits promoter hypermethylation in metastatic melanoma and inhibits invasion in melanoma cells. Epigenetics. 2014;9(11):1496–503.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Volodko N, et al. RASSF tumor suppressor gene family: biological functions and regulation. FEBS Lett. 2014;588(16):2671–84.

    Article  CAS  PubMed  Google Scholar 

  98. Wang F, et al. RASSF10 is an epigenetically inactivated tumor suppressor and independent prognostic factor in hepatocellular carcinoma. Oncotarget. 2016;7(4):4279–97.

    Article  PubMed  Google Scholar 

  99. Buhmeida A, et al. RASSF1A methylation is predictive of poor prognosis in female breast cancer in a background of overall low methylation frequency. Anticancer Res. 2011;31(9):2975–81.

    CAS  PubMed  Google Scholar 

  100. Kopp P. The TSH receptor and its role in thyroid disease. Cell Mol Life Sci. 2001;58(9):1301–22.

    Article  CAS  PubMed  Google Scholar 

  101. Soprano DR, Qin P, Soprano KJ. Retinoic acid receptors and cancers. Annu Rev Nutr. 2004;24:201–21.

    Article  CAS  PubMed  Google Scholar 

  102. Kartal K, et al. Methylation status of TSHr in well-differentiated thyroid cancer by using cytologic material. BMC Cancer. 2015;15:824.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Jia Y, et al. Integrin fibronectin receptors in matrix metalloproteinase-1-dependent invasion by breast cancer and mammary epithelial cells. Cancer Res. 2004;64(23):8674–81.

    Article  CAS  PubMed  Google Scholar 

  104. Corn PG, et al. E-cadherin expression is silenced by 5' CpG island methylation in acute leukemia. Clin Cancer Res. 2000;6(11):4243–8.

    CAS  PubMed  Google Scholar 

  105. Caldeira JR, et al. CDH1 promoter hypermethylation and E-cadherin protein expression in infiltrating breast cancer. BMC Cancer. 2006;6:48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Sunami E, et al. Estrogen receptor and HER2/neu status affect epigenetic differences of tumor-related genes in primary breast tumors. Breast Cancer Res. 2008;10(3):R46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Bakowska JC, et al. Troyer syndrome protein spartin is mono-ubiquitinated and functions in EGF receptor trafficking. Mol Biol Cell. 2007;18(5):1683–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Eastman SW, Yassaee M, Bieniasz PD. A role for ubiquitin ligases and Spartin/SPG20 in lipid droplet turnover. J Cell Biol. 2009;184(6):881–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Hooper C, et al. Spartin activates atrophin-1-interacting protein 4 (AIP4) E3 ubiquitin ligase and promotes ubiquitination of adipophilin on lipid droplets. BMC Biol. 2010;8:72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Lind GE, et al. SPG20, a novel biomarker for early detection of colorectal cancer, encodes a regulator of cytokinesis. Oncogene. 2011;30(37):3967–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Tsang HT, et al. The hereditary spastic paraplegia proteins NIPA1, spastin and spartin are inhibitors of mammalian BMP signalling. Hum Mol Genet. 2009;18(20):3805–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature. 1993;366(6456):704–7.

    Article  CAS  PubMed  Google Scholar 

  113. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9(10):1149–63.

    Article  CAS  PubMed  Google Scholar 

  114. Weinberg RA. The retinoblastoma protein and cell cycle control. Cell. 1995;81(3):323–30.

    Article  CAS  PubMed  Google Scholar 

  115. Shintani S, et al. Inactivation of the p14(ARF), p15(INK4B) and p16(INK4A) genes is a frequent event in human oral squamous cell carcinomas. Oral Oncol. 2001;37(6):498–504.

    Article  CAS  PubMed  Google Scholar 

  116. Tsuda H, et al. Relationship between HPV typing and abnormality of G1 cell cycle regulators in cervical neoplasm. Gynecol Oncol. 2003;91(3):476–85.

    Article  CAS  PubMed  Google Scholar 

  117. Dong M, et al. Clinicopathological significance of p53 and mdm2 protein expression in human pancreatic cancer. World J Gastroenterol. 2005;11(14):2162–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Saito H, et al. The expression of murine double minute 2 is a favorable prognostic marker in esophageal squamous cell carcinoma without p53 protein accumulation. Ann Surg Oncol. 2002;9(5):450–6.

    Article  PubMed  Google Scholar 

  119. Rocco JW, Sidransky D. p16(MTS-1/CDKN2/INK4a) in cancer progression. Exp Cell Res. 2001;264(1):42–55.

    Article  CAS  PubMed  Google Scholar 

  120. Cordon-Cardo C. Mutations of cell cycle regulators. Biological and clinical implications for human neoplasia. Am J Pathol. 1995;147(3):545–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Jang SJ. Cell cycle regulators as prognostic predictor of colorectal cancers. Korean J Gastroenterol. 2004;44(6):346–9.

    PubMed  Google Scholar 

  122. Fujiwara S, et al. Hypermethylation of p16 gene promoter correlates with loss of p16 expression that results in poorer prognosis in esophageal squamous cell carcinomas. Dis Esophagus. 2008;21(2):125–31.

    Article  CAS  PubMed  Google Scholar 

  123. Kim HC, et al. CpG island methylation in familial colorectal cancer patients not fulfilling the Amsterdam criteria. J Korean Med Sci. 2008;23(2):270–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Yoshihara T, Collado D, Hamaguchi M. Cyclin D1 down-regulation is essential for DBC2's tumor suppressor function. Biochem Biophys Res Commun. 2007;358(4):1076–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Bi Y, et al. Expressions of Fas, CTLA-4 and RhoBTB2 genes in breast carcinoma and their relationship with clinicopathological factors. Zhonghua Zhong Liu Za Zhi. 2008;30(10):749–53.

    CAS  PubMed  Google Scholar 

  126. Chang FK, et al. DBC2 is essential for transporting vesicular stomatitis virus glycoprotein. J Mol Biol. 2006;364(3):302–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Hamaguchi M, et al. DBC2, a candidate for a tumor suppressor gene involved in breast cancer. Proc Natl Acad Sci U S A. 2002;99(21):13647–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Siripurapu V, et al. DBC2 significantly influences cell-cycle, apoptosis, cytoskeleton and membrane-trafficking pathways. J Mol Biol. 2005;346(1):83–9.

    Article  CAS  PubMed  Google Scholar 

  129. Han L, et al. Decreased expression of the DBC2 gene and its clinicopathological significance in breast cancer: correlation with aberrant DNA methylation. Biotechnol Lett. 2013;35(8):1175–81.

    Article  CAS  PubMed  Google Scholar 

  130. Pulukuri SM, Rao JS. CpG island promoter methylation and silencing of 14-3-3sigma gene expression in LNCaP and Tramp-C1 prostate cancer cell lines is associated with methyl-CpG-binding protein MBD2. Oncogene. 2006;25(33):4559–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Chan TA, et al. 14-3-3Sigma is required to prevent mitotic catastrophe after DNA damage. Nature. 1999;401(6753):616–20.

    Article  CAS  PubMed  Google Scholar 

  132. Ye M, et al. Detection of 14-3-3 sigma (sigma) promoter methylation as a noninvasive biomarker using blood samples for breast cancer diagnosis. Oncotarget. 2017;8(6):9230–42.

    Article  PubMed  Google Scholar 

  133. Zhu S, et al. PKC?-dependent activation of the ubiquitin proteasome system is responsible for high glucose-induced human breast cancer MCF-7 cell proliferation, migration and invasion. Asian Pac J Cancer Prev. 2013;14(10):5687–92.

    Article  PubMed  Google Scholar 

  134. Fahmidehkar MA, et al. Induction of cell proliferation, clonogenicity and cell accumulation in S phase as a consequence of human UBE2Q1 overexpression. Oncol Lett. 2016;12(3):2169–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. van Wijk SJ, Timmers HT. The family of ubiquitin-conjugating enzymes (E2s): deciding between life and death of proteins. FASEB J. 2010;24(4):981–93.

    Article  CAS  PubMed  Google Scholar 

  136. Chang R, et al. Upregulated expression of ubiquitin-conjugating enzyme E2Q1 (UBE2Q1) is associated with enhanced cell proliferation and poor prognosis in human hapatocellular carcinoma. J Mol Histol. 2015;46(1):45–56.

    Article  CAS  PubMed  Google Scholar 

  137. Seghatoleslam A, et al. Expression of UBE2Q2, a putative member of the ubiquitin-conjugating enzyme family in pediatric acute lymphoblastic leukemia. Arch Iran Med. 2012;15(6):352–5.

    CAS  PubMed  Google Scholar 

  138. Bergamin E, et al. The cytoplasmic adaptor protein Dok7 activates the receptor tyrosine kinase MuSK via dimerization. Mol Cell. 2010;39(1):100–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Cossins J, et al. The spectrum of mutations that underlie the neuromuscular junction synaptopathy in DOK7 congenital myasthenic syndrome. Hum Mol Genet. 2012;21(17):3765–75.

    Article  CAS  PubMed  Google Scholar 

  140. Heyn H, et al. DNA methylation profiling in breast cancer discordant identical twins identifies DOK7 as novel epigenetic biomarker. Carcinogenesis. 2013;34(1):102–8.

    Article  CAS  PubMed  Google Scholar 

  141. Ivaska J. Vimentin: Central hub in EMT induction? Small GTPases. 2011;2(1):51–3.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Challa AA, Stefanovic B. A novel role of vimentin filaments: binding and stabilization of collagen mRNAs. Mol Cell Biol. 2011;31(18):3773–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Katz E, et al. An in vitro model that recapitulates the epithelial to mesenchymal transition (EMT) in human breast cancer. PLoS One. 2011;6(2):e17083.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Mor-Vaknin N, et al. Vimentin is secreted by activated macrophages. Nat Cell Biol. 2003;5(1):59–63.

    Article  CAS  PubMed  Google Scholar 

  145. Mori T, et al. Epigenetic up-regulation of C-C chemokine receptor 7 and C-X-C chemokine receptor 4 expression in melanoma cells. Cancer Res. 2005;65(5):1800–7.

    Article  CAS  PubMed  Google Scholar 

  146. Sato N, et al. The chemokine receptor CXCR4 is regulated by DNA methylation in pancreatic cancer. Cancer Biol Ther. 2005;4(1):70–6.

    Article  CAS  PubMed  Google Scholar 

  147. Schaefer JS, et al. Transcriptional regulation of p21/CIP1 cell cycle inhibitor by PDEF controls cell proliferation and mammary tumor progression. J Biol Chem. 2010;285(15):11258–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Sood AK, et al. Expression characteristics of prostate-derived Ets factor support a role in breast and prostate cancer progression. Hum Pathol. 2007;38(11):1628–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Le AV, Szaumkessel M, Tan TZ, Thiery JP, Thompson EW, Dobrovic A. DNA methylation profiling of breast cancer cell lines along the epithelial mesenchymal spectrum—Implications for the choice of circulating tumour DNA methylation markers. Int J Mol Sci. 2018;19(9):2553.

  150. Wang W, Tao YX. Ghrelin Receptor Mutations and Human Obesity. Prog Mol Biol Transl Sci. 2016;140:131–50.

    Article  CAS  PubMed  Google Scholar 

  151. Lin TC, Hsiao M. Ghrelin and cancer progression. Biochim Biophys Acta Rev Cancer. 2017;1868(1):51–7.

    Article  CAS  PubMed  Google Scholar 

  152. Sun Y, et al. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proc Natl Acad Sci U S A. 2004;101(13):4679–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Yin Y, Li Y, Zhang W. The growth hormone secretagogue receptor: its intracellular signaling and regulation. Int J Mol Sci. 2014;15(3):4837–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Coppede F, et al. Investigation of GHSR and GHRL methylation in colorectal cancer. Epigenomics. 2018;10(12):1525–39.

    Article  CAS  PubMed  Google Scholar 

  155. Chen C, et al. Hypermethylation of EDNRB promoter contributes to the risk of colorectal cancer. Diagn Pathol. 2013;8:199.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Druckenbrod NR, et al. Targeting of endothelin receptor-B to the neural crest. Genesis. 2008;46(8):396–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Sanchez-Mejias A, et al. Contribution of RET, NTRK3 and EDN3 to the expression of Hirschsprung disease in a multiplex family. J Med Genet. 2009;46(12):862–4.

    Article  CAS  PubMed  Google Scholar 

  158. Lahav R, Heffner G, Patterson PH. An endothelin receptor B antagonist inhibits growth and induces cell death in human melanoma cells in vitro and in vivo. Proc Natl Acad Sci U S A. 1999;96(20):11496–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Mallat A, et al. Growth inhibitory properties of endothelin-1 in human hepatic myofibroblastic Ito cells. An endothelin B receptor-mediated pathway. J Clin Invest. 1995;96(1):42–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Zhao BJ, et al. Identification of aberrant promoter methylation of EDNRB gene in esophageal squamous cell carcinoma. Dis Esophagus. 2009;22(1):55–61.

    Article  PubMed  Google Scholar 

  161. Chen S, et al. Overexpression of KiSS-1 reduces colorectal cancer cell invasion by downregulating MMP-9 via blocking PI3K/Akt/NF-kappaB signal pathway. Int J Oncol. 2016;48(4):1391–8.

    Article  CAS  PubMed  Google Scholar 

  162. Ji K, et al. Implication of metastasis suppressor gene, Kiss-1 and its receptor Kiss-1R in colorectal cancer. BMC Cancer. 2014;14:723.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  163. Abbaszadegan MR, et al. WNT and NOTCH signaling pathways as activators for epidermal growth factor receptor in esophageal squamous cell carcinoma. Cell Mol Biol Lett. 2018;23:42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Moghbeli M, et al. Role of Msi1 and MAML1 in Regulation of Notch Signaling Pathway in Patients with Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer. 2015;46(4):365–9.

    Article  CAS  PubMed  Google Scholar 

  165. Moghbeli M, et al. Correlation Between Meis1 and Msi1 in Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer. 2016;47(3):273–7.

    Article  CAS  PubMed  Google Scholar 

  166. Arnold CN, et al. APC promoter hypermethylation contributes to the loss of APC expression in colorectal cancers with allelic loss on 5q. Cancer Biol Ther. 2004;3(10):960–4.

    Article  CAS  PubMed  Google Scholar 

  167. Koinuma K, et al. Epigenetic silencing of AXIN2 in colorectal carcinoma with microsatellite instability. Oncogene. 2006;25(1):139–46.

    Article  CAS  PubMed  Google Scholar 

  168. Qi J, et al. Hypermethylation and expression regulation of secreted frizzled-related protein genes in colorectal tumor. World J Gastroenterol. 2006;12(44):7113–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Yu J, et al. Promoter methylation of the Wnt/beta-catenin signaling antagonist Dkk-3 is associated with poor survival in gastric cancer. Cancer. 2009;115(1):49–60.

    Article  CAS  PubMed  Google Scholar 

  170. Rocheleau CE, et al. Wnt signaling and an APC-related gene specify endoderm in early C. elegans embryos. Cell. 1997;90(4):707–16.

    Article  CAS  PubMed  Google Scholar 

  171. Wang B, et al. Early diagnostic potential of APC hypermethylation in esophageal cancer. Cancer Manag Res. 2018;10:181–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Bartak BK, et al. Colorectal adenoma and cancer detection based on altered methylation pattern of SFRP1, SFRP2, SDC2, and PRIMA1 in plasma samples. Epigenetics. 2017;12(9):751–63.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Salmena L, Carracedo A, Pandolfi PP. Tenets of PTEN tumor suppression. Cell. 2008;133(3):403–14.

    Article  CAS  PubMed  Google Scholar 

  174. Waniczek D, et al. PTEN expression profiles in colorectal adenocarcinoma and its precancerous lesions. Pol J Pathol. 2013;64(1):15–20.

    Article  PubMed  Google Scholar 

  175. Freeman DJ, et al. PTEN tumor suppressor regulates p53 protein levels and activity through phosphatase-dependent and -independent mechanisms. Cancer Cell. 2003;3(2):117–30.

    Article  CAS  PubMed  Google Scholar 

  176. Su JD, et al. PTEN and phosphatidylinositol 3'-kinase inhibitors up-regulate p53 and block tumor-induced angiogenesis: evidence for an effect on the tumor and endothelial compartment. Cancer Res. 2003;63(13):3585–92.

    CAS  PubMed  Google Scholar 

  177. Planchon SM, Waite KA, Eng C. The nuclear affairs of PTEN. J Cell Sci. 2008;121(Pt 3):249–53.

    Article  CAS  PubMed  Google Scholar 

  178. Meng F, et al. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007;133(2):647–58.

    Article  CAS  PubMed  Google Scholar 

  179. Zhang BG, et al. microRNA-21 promotes tumor proliferation and invasion in gastric cancer by targeting PTEN. Oncol Rep. 2012;27(4):1019–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Zhang JG, et al. MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin Chim Acta. 2010;411(11-12):846–52.

    Article  CAS  PubMed  Google Scholar 

  181. Siddiqui S, et al. A study on promoter methylation of PTEN in sporadic breast cancer patients from North India. Breast Cancer. 2016;23(6):922–31.

    Article  PubMed  Google Scholar 

  182. Mavrogiannis LA, et al. Haploinsufficiency of the human homeobox gene ALX4 causes skull ossification defects. Nat Genet. 2001;27(1):17–8.

    Article  CAS  PubMed  Google Scholar 

  183. Antonopoulou I, et al. Alx4 and Msx2 play phenotypically similar and additive roles in skull vault differentiation. J Anat. 2004;204(6):487–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Ebert MP, et al. Aristaless-like homeobox-4 gene methylation is a potential marker for colorectal adenocarcinomas. Gastroenterology. 2006;131(5):1418–30.

    Article  CAS  PubMed  Google Scholar 

  185. Balasenthil S, et al. Identification of Pax5 as a target of MTA1 in B-cell lymphomas. Cancer Res. 2007;67(15):7132–8.

    Article  CAS  PubMed  Google Scholar 

  186. Palmisano WA, et al. Aberrant promoter methylation of the transcription factor genes PAX5 alpha and beta in human cancers. Cancer Res. 2003;63(15):4620–5.

    CAS  PubMed  Google Scholar 

  187. Liu C, et al. Meta-analysis of association studies of CYP1A1 genetic polymorphisms with digestive tract cancer susceptibility in Chinese. Asian Pac J Cancer Prev. 2014;15(11):4689–95.

    Article  PubMed  Google Scholar 

  188. Deng J, et al. Applicability of the methylated CpG sites of paired box 5 (PAX5) promoter for prediction the prognosis of gastric cancer. Oncotarget. 2014;5(17):7420–30.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Shirafkan N, et al. MicroRNAs as novel biomarkers for colorectal cancer: New outlooks. Biomed Pharmacother. 2018;97:1319–30.

    Article  CAS  PubMed  Google Scholar 

  190. Park HS, et al. High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod Pathol. 2014;27(9):1212–22.

    Article  CAS  PubMed  Google Scholar 

  191. Mayank, Jaitak V. Drug target strategies in breast cancer treatment: recent developments. Anticancer Agents Med Chem. 2014;14(10):1414–27.

    Article  CAS  PubMed  Google Scholar 

  192. Asadi M, et al. Expression Level of miR-34a in Tumor Tissue from Patients with Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer. 2019;50(2):304–7.

    Article  CAS  PubMed  Google Scholar 

  193. Shrestha S, et al. A systematic review of microRNA expression profiling studies in human gastric cancer. Cancer Med. 2014;3(4):878–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Farazi TA, et al. MicroRNAs in human cancer. Adv Exp Med Biol. 2013;774:1–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Calin GA, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A. 2004;101(9):2999–3004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Shen R, et al. Epigenetic repression of microRNA-129-2 leads to overexpression of SOX4 in gastric cancer. Biochem Biophys Res Commun. 2010;394(4):1047–52.

    Article  CAS  PubMed  Google Scholar 

  197. Riggins RB, et al. Pathways to tamoxifen resistance. Cancer Lett. 2007;256(1):1–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Pinzone JJ, et al. Molecular and cellular determinants of estrogen receptor alpha expression. Mol Cell Biol. 2004;24(11):4605–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Le Romancer M, et al. Methylation, a key step for nongenomic estrogen signaling in breast tumors. Steroids. 2010;75(8-9):560–4.

    Article  PubMed  CAS  Google Scholar 

  200. Harris L, et al. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol. 2007;25(33):5287–312.

    Article  CAS  PubMed  Google Scholar 

  201. Ascenzi P, Bocedi A, Marino M. Structure-function relationship of estrogen receptor alpha and beta: impact on human health. Mol Aspects Med. 2006;27(4):299–402.

    Article  CAS  PubMed  Google Scholar 

  202. Hagrass HA, Pasha HF, Ali AM. Estrogen receptor alpha (ERalpha) promoter methylation status in tumor and serum DNA in Egyptian breast cancer patients. Gene. 2014;552(1):81–6.

    Article  CAS  PubMed  Google Scholar 

  203. Tang D, et al. Methylation of the RARB gene increases prostate cancer risk in black Americans. J Urol. 2013;190(1):317–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Hsu H, et al. TNF-dependent recruitment of the protein kinase RIP to the TNF receptor-1 signaling complex. Immunity. 1996;4(4):387–96.

    Article  CAS  PubMed  Google Scholar 

  205. Hsu H, et al. TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell. 1996;84(2):299–308.

    Article  CAS  PubMed  Google Scholar 

  206. Kischkel FC, et al. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995;14(22):5579–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Kluck RM, et al. The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science. 1997;275(5303):1132–6.

    Article  CAS  PubMed  Google Scholar 

  208. Srinivasula SM, et al. Molecular ordering of the Fas-apoptotic pathway: the Fas/APO-1 protease Mch5 is a CrmA-inhibitable protease that activates multiple Ced-3/ICE-like cysteine proteases. Proc Natl Acad Sci U S A. 1996;93(25):14486–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Yang J, et al. Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science. 1997;275(5303):1129–32.

    Article  CAS  PubMed  Google Scholar 

  210. Zou H, et al. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell. 1997;90(3):405–13.

    Article  CAS  PubMed  Google Scholar 

  211. Li P, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91(4):479–89.

    Article  CAS  PubMed  Google Scholar 

  212. Wang HL, et al. Rationales for expression and altered expression of apoptotic protease activating factor-1 gene in gastric cancer. World J Gastroenterol. 2007;13(38):5060–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Yan MD, et al. Identification and characterization of a novel gene Saf transcribed from the opposite strand of Fas. Hum Mol Genet. 2005;14(11):1465–74.

    Article  CAS  PubMed  Google Scholar 

  214. Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell. 2004;116(2):205–19.

    Article  CAS  PubMed  Google Scholar 

  215. Nagata S. Apoptosis by death factor. Cell. 1997;88(3):355–65.

    Article  CAS  PubMed  Google Scholar 

  216. Oliver AW, Knapp S, Pearl LH. Activation segment exchange: a common mechanism of kinase autophosphorylation? Trends Biochem Sci. 2007;32(8):351–6.

    Article  CAS  PubMed  Google Scholar 

  217. Berkovich E, Ginsberg D. ATM is a target for positive regulation by E2F-1. Oncogene. 2003;22(2):161–7.

    Article  CAS  PubMed  Google Scholar 

  218. Begam N, Jamil K, Raju SG. Promoter Hypermethylation of the ATM Gene as a Novel Biomarker for Breast Cancer. Asian Pac J Cancer Prev. 2017;18(11):3003–9.

    PubMed  PubMed Central  Google Scholar 

  219. Kwon ED, et al. Elimination of residual metastatic prostate cancer after surgery and adjunctive cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade immunotherapy. Proc Natl Acad Sci U S A. 1999;96(26):15074–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Lafage-Pochitaloff M, et al. Human CD28 and CTLA-4 Ig superfamily genes are located on chromosome 2 at bands q33-q34. Immunogenetics. 1990;31(3):198–201.

    Article  CAS  PubMed  Google Scholar 

  221. Schneider H, et al. CTLA-4 activation of phosphatidylinositol 3-kinase (PI 3-K) and protein kinase B (PKB/AKT) sustains T-cell anergy without cell death. PLoS One. 2008;3(12):e3842.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  222. Sasidharan Nair V, et al. DNA methylation and repressive H3K9 and H3K27 trimethylation in the promoter regions of PD-1, CTLA-4, TIM-3, LAG-3, TIGIT, and PD-L1 genes in human primary breast cancer. Clin Epigenetics. 2018;10:78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

Not applicable

Author information

Authors and Affiliations

Authors

Contributions

IA, ARB, MRA and MM were involved in search strategy and drafting. MM revised, edited, and supervised the project. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Meysam Moghbeli.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Akhlaghipour, I., Bina, A.R., Abbaszadegan, M.R. et al. Methylation as a critical epigenetic process during tumor progressions among Iranian population: an overview. Genes and Environ 43, 14 (2021). https://doi.org/10.1186/s41021-021-00187-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41021-021-00187-1

Keywords