1. Introduction
Ovarian cancer (OC) is the seventh most common and one of the deadliest types of cancer in women [
1]. It most commonly occurs in postmenopausal women with an average of 65 years old [
2]. Major risk factors for OC are: Hereditary Breast and Ovarian Cancer (HBOC) Syndrome, Lynch Syndrome, postmenopausal hormonal therapy, endometriosis, IVF treatment, use of fertility drugs, late menopause and null parity [
3]. OC is difficult to detect at early stage and 60% of the cases are diagnosed at advanced stage disease [
4]. Due to its asymptomatic nature and late presentation, it is known as the “whispering” or “silent” cancer [
5].
Epithelial ovarian cancer (EOC) accounts for 90% of the OC cases. Five major histological types of EOC are recognized: high-grade serous ovarian carcinoma (HGSOC), low grade serous ovarian carcinoma (LGSOC), mucinous ovarian carcinoma (MOC), clear cell ovarian carcinoma (CCOC) and endometrioid ovarian carcinoma (EnOC) [
4]. Each EOC subtype is characterized by different gene mutations that deregulate diverse signaling pathways and this knowledge should be employed for the development of personalized treatment strategies. HGSOC, the largest subgroup (75%), is the most aggressive and chemoresistant form of EOCs, responsible for 70–80% of OC-related deaths [
6]. Approximately 15–20% of HGSOC patients have germline
BRCA1 or
BRCA2 mutations [
7]. Homologous recombination deficiency (HRD), either sporadic or germ-line, is seen in nearly half of HGSOC [
8]. HRD is a key determinant of platinum sensitivity in HGSOC and has been exploited for treatment with poly (ADP-ribose) polymerase inhibitors (PARPi). HGSOCs are also characterized by a high frequency (90%) of somatic
TP53 mutations [
9] and frequent DNA copy number alterations involving CCN1, PIK3CA and PTEN [
10].
LGSOCs are less aggressive tumors with a relatively better prognosis than HGSOCs, occur in younger women (median age of 55 years) and account less than 5% of all OCs [
4]. A small subset of patients has a prior history of a serous borderline tumour (SBT). LGSOCs have a significantly higher expression of ER and PR compared to HGSOCs and are not associated with
BRCA germline mutations [
11], showing
KRAS and
BRAF mutations instead [
10].
Mucinous, clear cell and endometrioid carcinomas are less frequently seen, with the latter two being associated with endometriosis. Numerous studies have demonstrated increased incidence of defective mismatch repair (dMMR) and an associated microsatellite instability-high (MSI-H) phenotype in non-serous ovarian cancer (typically tumors of endometrioid or clear-cell histology) [
12].
Despite the combined use of surgery and chemotherapy in the modern era, mortality rates have not improved significantly over the last decades for OC [
13]. LGSOCs are chemoresistant, with less than 5% response rate to first-line chemotherapy compared to the 80% response rate of HGSOC treated with platinum/taxane drugs [
11]. Even though HGSOCs initially respond to platinum/taxane-based therapies, the majority of the patients will eventually develop chemotherapy resistance and tumor recurrence [
14]. Thus, the detection of novel therapeutic targets that will drive personalized therapy is crucial for improving OC patients’ prognosis [
15].
Epigenetic mechanisms, that is changes in DNA expression without any alteration in DNA sequence, include DNA methylation, histone post-transcriptional modifications and microRNA expression, are considered pivotal in tumor initiation and progression and represent potential therapeutic targets [
16]. DNA methylation is a biological process in which the cytosine bases of eukaryotic DNA are converted to 5-methylcytosine. This event usually takes place in CpG dinucleotides located within the promoter region of the genes, thereby silencing transcriptional activity [
17]. Transcriptional silencing can be inherited to daughter cells following cell division [
18] and can be pharmacologically inverted [
19]. In cancer cells, the genome is generally hypomethylated [
20] compared to normal cells and this leads to genomic instability and oncogene expression [
21,
22]. However, hypermethylation is seen in specific promoters, namely those of tumor-suppressor genes, resulting in reduced expression of the respective protein [
23,
24].
DNA methylation is mediated by DNA methyltransferases (DNMT1, 2, 3A, 3B and 3L). DNMT1 is responsible for maintaining methylation patterns following replication, and DNMT3A and DNMT3B initiate
de novo methylation [
25]. DNMT3L is catalytically inactive but enables DNMT3s’ interaction with the histone code. DNMT2 is the fifth member of the family, and even though it shows structural similarity to the other DNMTs, it harbors a weak DNA methylation activity, but is, instead, capable of methylating aspartic acid tRNAs [
26]. Thus, DNMT2, also called TRDMT1, represents an RNA methyltransferase.
Alterations in the expression of DNMTs have been shown in various solid tumors and hematologic malignancies and have been associated with diverse clinicopathologic features and survival outcomes [
27], frequently, independently of other prognostic factors, underscoring their importance as potential drivers in a variety of cancer types. These data rationalized the development of DNA methyltransferase inhibitors (DNMTi) which can restore the exrpession of tumour suppressor genes [
28]. At present, two DNMTi drugs, 5- azacytidine (AZA) and decitabine, have been approved for treating patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) [
29], and are also being tested as therapeutic options in several solid cancers such as colon, ovarian, and lung carcinomas [
30]. However, many items remain unsolved considering tumor responses, suggesting the presence of alternative ways by which DNMTi may modulate cancer cells [
31].
A mounting amount of evidence suggests that DNA methylation is implicated in OC development and progression. Studies have shown that tumor suppressor gene loss in OC is mediated by DNA methylation [
32] and many epidemiological studies focus on the role of DNA methylation in OC susceptibility [
33]. DNMTs are often overexpressed in various cancer tissues and cell lines and their levels have been associated with poor survival [
34]. However, few studies have analyzed the expression of DNMTs in ovarian carcinomas, with conflicting results, and the prognostic effect of these enzymes in ovarian cancer remains to be investigated [
35,
36,
37].
In the current study, we used publicly available datasets from patients with OC (TNMplot, Gepia2., Protein Data Commons and Kaplan-Meier Plotter) to analyze the expression of DNMTs at the RNA and protein level and correlate it with clinical data. We also analyzed the expression of DNMTs in a cohort of primary epithelial ovarian carcinomas and theirs relapses using immunohistochemistry, in an attempt to provide a thorough understanding of the role of DNMTs in ovarian cancer and identify potentially effective drug targets.
4. Discussion
In the present study we systematically retrieved data regarding the mRNA and protein expression of DNMT1, 2, 3A, 3B and 3L in epithelial ovarian carcinoma samples from interactive web applications such as TNMplot, GEPIA2, Kaplan-Meier Plotter and Proteomic data commons. We performed differential gene expression analyses between tumor and normal samples and correlation of mRNA expression levels with FIGO Stage. Then, we investigated the correlation of DNMTs’ expression with patients’ prognosis. Finally, using immunohistochemistry, we evaluated DNMTs’ protein expression in 108 tissue samples from patients with primary and relapsed ovarian tumors and correlated with normal tissues and histological subtypes. Ιt should be noted that the expression of DNMT2 and DNMT3L proteins in ovarian carcinomas by immunohistochemistry has not been reported in the literature before.
Our results showed that DNMT1 expression was elevated from non-neoplastic to tumor tissue, at the mRNA and protein level, further increased with grade progression and in tumor relapses, and its phosphorylation levels were also increased in carcinomas. DNMT3A levels increased from non-neoplastic to tumor tissue at both the mRNA and protein level, with further increase with increasing the grade of the tumors, albeit its levels were lower in higher stage and in metastatic tumors. DNMT3B expression was increased at the mRNA level from non-neoplastic to tumor tissue and was decreased in the metastases compared to primary foci but no difference was noted at the protein level. DNMT3L was increased in the primary tumors compared to non-neoplastic tissue at the mRNA and protein level and was further increased in relapses. In contrast to the other DNMTs DNMT2 levels seemed to decrease from non-neoplastic to tumor tissue at the mRNA and protein level and further decrease with stage progression. To the best of our knowledge this is the first study to have addressed the differential expression of DNMTs according to the histology of OEC, and between primary and relapsed tumors at the protein and cellular/sucellular level by immunohistochemistry. This is important as we and others [
40,
41] have shown that mRNA levels do not always correlate with protein levels, which is attributed to translational regulation, differences in protein in vivo half-lives, and differences in experimental conditions [
42]. In addition, using immunohistochemistry, the specific cellular and subcellular expression of the markers was assessed, a feature that PCR and RNA sequencing techniques can not address [
43,
44].
Previous studies have shown that DNMTs expression is upregulated in various malignancies (colon, prostate, breast, liver, leukemia) [
45,
46] and that their expression is associated with aggressive pathologic characteristics [
46,
47]. Results regarding the expression of DNMTs in ovarian tumors in the literature are equivocal. This is probably a result of the different patient populations studied and the different methodologies used. In addition, as we have shown in this study and reviewed here [
48], DNMT expression depends on OEC histologic subtype.
One of the first studies of DNMT expression in ovarian tumors used cancer cell lines and showed increased expression of DNMT1, and DNMT3B, but not DNMT3A mRNA levels in ovarian cancer cell lines compared to normal ovarian surface epithelial cells [
49]. In patients samples, and similarly to our results, DNMT3A protein, was found to be elevated in ovarian cancer tissues compared to normal ovary tissues [
50]. Importantly, and in agreement with our results, protein and mRNA levels did not correlate with each other [
50]. Another study showed significantly lower expression of DNMT3A1 and DNMT2 mRNAs, the latter in agreement with our results, but higher expression of DNMT3B1/B2 mRNA isoforms in carcinomas than in low malignant potential tumors and that DNMT3B1/3B2 mRNA levels were associated with the methylation status of
CDH13, MLH-M2B, SEZ6L and
MINT31-M1B [
37]. In accordance with this, using qPCR in 63 ovarian cancer patient samples, DNMT3B1 and DNMT3B3 overexpression was seen in advanced stages and high-grade serous carcinomas [
51]. More recently, the expression of DNMT1, DNMT3A, and DNMT3B was examined by immunohistochemistry in ovarian cancers and benign tumors. The data showed higher DNMT3A and lower DNMT3B protein expression in ovarian cancers compared to that of the benign tumors whereas DNMT1 expression showed no difference [
35]. In contrast, using qRT-PCR and immunohistochemistry, DNMT1 and DNMT3B expression was shown to be elevated in carcinomas compared to non-neoplastic tissue and further enhanced in higher stage carcinomas [
52].
Taken together and in accordance with our findings, there is abundant evidence in the literature of aberrant expression of DNMTs in ovarian carcinomas compared to non-neoplastic cells or benign tumors, with further deregulation in high grade ovarian carcinomas and in metastases/relapses. Considering our comprehensive analysis of DNMTs expression across the progression of OEC
, we hypothesize that DNMT1 and DNTM3A seem to play a role in tumor initiation, DNMT3B in tumor progression and DNMT3L in tumor relapse whereas DNMT2 may have an opposite role, acting as a tumor-suppressor gene. This stage specific role of distinct DNMTs has been accounted for the progression of other neoplasms like prostate carcinomas [
46,
53]. In addition, others have hypothesized a similar carcinogenesis model in which DNMT3A and DNMT3B are specifically recruited during tumor initiation and promotion, with subsequent downregulation of their expression, whereas DNMT1 is involved in tumor progression [
54]. These findings support the notion that alterations in DNMT expression might contribute to the development and progression of high-grade ovarian carcinomas i.e., through the establishment of a CpG Island methylator phenotype in ovarian cancer.
We, also, examined the association of DNMTs expression with patient’s survival and showed that DNMT3A, DNMT3B and DNMT2 mRNA levels were correlated with bad prognosis and DNMT1 expression with favorable prognosis. The correlation of DNMTs with prognosis is ambiguous and seems to be cancer specific. Some studies have shown that DNMTs levels are associated with an ominous prognosis [
47], while others have found that the expression of these molecules, in particular DNMT3A, is associated with good prognosis [
27,
47]. Regarding ovarian carcinoma, the literature regarding the prognostic role of DNMTs expression is rather sparse. Using immunohistochemistry, Bai et al showed that expression of DNMT1 and DNMT3B were marginally associated with better overall survival [
35], the first in agreement with our results. The combined expression of both markers had a better statistical correlation [
35]. Analysis of DNMT3B isoforms expression using qPCR in 63 OEC showed that DNMT3B1 and DNMT3B3 overexpression was associated with poor prognosis [
51]. This agrees with our results, albeit we did not specifically analyze the different isoforms of the molecule. Similarly, in agreement with our findings, DNMT3A has been found to be associated with poor prognosis [
50]. Taken together, these findings show that DNMT1, DNMT3A and DNMT3B may have a prognostic role in OEC. Further studies, with a higher number of patients and a combination of techniques will be needed to validate this hypothesis.
Regarding DNMT1, an interesting finding was that the phosphorylation levels at s154 were increased in tumors compared to normal tissues. In Human Mammary Epithelial Cells it has been shown that the elevated DNMT1 protein levels are not a result of increased mRNA levels, but rather an increase in protein half-life, due to the deleted destruction domain which is required for its proper ubiquitination and degradation [
55]. It has also been suggested that phosphorylation on Ser154 by cyclin-dependent kinases may play a role in controlling DNMT1 activity and protein stability, leading to increased DNMT1 activity [
56]. Thus, our findings highlight protein phosphorylation as one of the mechanisms of DNMT1 overexpression in OEC.
DNMT3L is involved in DNMT3A function as it forms a complex with DNMT3A2 and prevents DNMT3A2 from being degraded. Restoring the DNMT3A protein level in DNMT3L-deficient embryonal stem cells (ESC) partially recovers DNA methylation [
57]. DNMT3L is expressed in ESC and downregulated in differentiated embryonal cells, thus, being associated with pluripotency [
58]. The available data regarding DNMT3L mRNA and protein expression in different cancer subtypes is very limited. Overexpressiοn of DNMT3L protein has been shown in embryonal carcinoma, but not in the other types of testicular germ cell tumors [
59]. It has, also been shown to be overexpressed in prostate carcinomas, especially high grade ones [
46]. Expression in OEC has not been studied before. Ιn our study we observed overexpression of DNMT3L protein in high grade carcinomas compared to low grade tumors and non-neoplastic epithelium and in relapses compared to primary neoplasms. Further study of the role of DNMT3L in de novo DNA methylation in ovarian tumors is warranted.
DNMT2 (TRDMT1) is actually an aspartic acid tRNA methyltransferase, with weak, if any, DNA methyltransferase activity [
26]. DNMT2 mutations have been reported in human tumors and are associated with alterations of its methylation activity [
60]. DNMT2 upregulation has been reported in GIST [
61], glioma [
62], prostate carcinoma [
46] and clear cell renal cell carcinoma [
63]. Its expression in OEC has not been studied before. In the present study we showed both cytoplasmic and nuclear expression of DNMT2/TRDMT1 as has been reported previously [
26,
46]. It is reasonable and expected that a cytosine-5 methyltransferase such as DNMT2, with activity towards both DNA and RNA, would function in both cellular compartments. Database analysis showed that DNMT2 was decreased in carcinomas compared to non-neoplastic tissue. We also noticed that both cytoplasmic and nuclear DNMT2 expression gradually decreased from borderline to high grade serous tumors, although it did not reach a statistically significant level. It has previously been reported that DNMT2/TRDMT1 is required for efficient homologous recombination and that loss of its function makes the cells sensitive to PARP inhibitors [
64]. This observation is of particular importance in ovarian cancer patients where homologous recombination deficiency (HRD) and PARP inhibitor therapy is the treatment of choice for at least half of the patients with high grade serous ovarian tumors. Analysis of DNMT2 expression in ovarian cancer could contribute to the selection of patients as candidates for PARP inhibitor therapy.
Even though a straight link between hypermethylation of specific genes that contribute to carcinogenesis and overexpression of DNMTs has not been proven, DNMTs have been correlated with tumor progression and patients’ prognosis, and therefore, they may represent potential prognostic biomarkers and therapeutic targets [
37,
65]. A recent, very promising, study showed that overexpression of DNMT1 and DNMT3B is highly linked to genome wide hypermethylation profile in prostate cancer and paved the way studying their use in clinical practice in many malignances [
66]. DNMT inhibitors (DNMTi) have shown promising results in ovarian cancer in preclinical models by enhancing IFNγ-mediated [
67] and type I interferon-mediated inflammatory responses [
68]. In addition, DNMTi seem to work synergistically with other therapies. Administration of DNMTis seems to enhance sensitivity to platinum treatment [
69], immunotherapy [
70] and PARP inhibition [
71].