Recent findings on epigenetic gene abnormalities involved in uterine cancer (Review)

  • Authors:
    • Megumi Yanokura
    • Kouji Banno
    • Yusuke Kobayashi
    • Hiroyuki Nomura
    • Shigenori Hayashi
    • Eiichiro Tominaga
    • Daisuke Aoki
  • View Affiliations

  • Published online on: September 20, 2017     https://doi.org/10.3892/mco.2017.1428
  • Pages: 733-737
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Selective aberrant genetic effects that do not depend on abnormal DNA sequences are referred to as epigenetic abnormalities and are involved in carcinogenesis. In uterine cancer, various genes involved in apoptosis, cell cycle, DNA repair, cell proliferation and cell adhesion are abnormally methylated, resulting in gene silencing. Reversal of such epigenetic abnormalities in cancer cells is a potential strategy for cancer therapy, and studies on epigenetic abnormalities and treatment methods in uterine cancer are in progress. These include the evaluation of 5‑hydroxymethylcytosine, which is present in cancer tissues at lower levels compared with those in normal tissues, as a prognostic marker in cervical cancer; combination therapy with 5‑azacytidine and cisplatin; combination treatment focusing on tumor necrosis factor‑related apoptosis‑inducing ligand in cervical cancer; studies focusing on DNA mismatch repair in endometrial cancer; and use of a demethylating agent to reactivate tumor suppressor genes and inhibit tumor proliferation. Detection of epigenetic changes using biomarkers may be used for histological classification, evaluation of disease progression and identification of compounds that are able to modulate epigenetic changes and may be useful for uterine cancer treatment.

Introduction

Cervical and endometrial cancer rank fourth and fifth, respectively, in cancer prevalence by organ among women worldwide (1). Studies of oncogenic pathways have shown early and frequent DNA methylation, with the extent of this change considered to be related to severity. In both cervical and endometrial cancer, specific genes involved in various pathways are known to be methylated. In cervical cancer, these genes include tumor protein p73, fragile histidine triad (FHIT), death-associated protein kinase 1 and PRDI-BF and RIZ domain containing 14 in the apoptotic pathway; cyclin A1 (CCNA1) and double C2-like domain β in the cell cycle; adenomatous polyposis coli (APC) and secreted frizzled-related protein (SFRP) in the Wnt/β-catenin pathway; Fanconi anemia, complementation group F, O-6-methylguanine-DNA methyltransferase, human MutL homolog 1 (hMLH1) and CCNA1 in DNA repair; FHIT, retinoic acid receptor-β and myelin and lymphocyte in the cell growth pathway; and CXC chemokine receptor 4 and cell adhesion molecule 1, which are involved in cell adhesion (221). In endometrial cancer, the following genes are methylated: APC, caspase-8, checkpoint with forkhead-associated and ring finger, E-cadherin, hMLH1, p73, progesterone receptor, phosphatase and tensin homologue deleted on chromosome 10, Ras association domain family 1 isoform A and thrombospondin 2 (2247). The roles of these methylated genes are listed in Table I for cervical cancer and in Table II for endometrial cancer. Gene silencing caused by methylation may promote cancer progression, and studies on the clinical significance of gene silencing are ongoing.

Table I.

Methylated genes in cervical cancer.

Table I.

Methylated genes in cervical cancer.

Gene nameFunction(Refs.)
TP73Apoptosis(4)
FHITApoptosis(3,5,7,15,16)
DAPK1Apoptosis(3)
PRDM14Apoptosis(6)
CCNA1Cell cycle, DNA repair(8,9)
DOC2BCell cycle(10)
APCWnt/β catenin pathway(11)
SFRPWnt/β catenin pathway(12)
FANCFDNA repair(13)
MGMTDNA repair(3,14,15)
hMLH1DNA repair(15)
FHITCell growth(3,5,7,15,16)
RARCell growth(15,17)
MALCell growth(17)
CXCR4Cell-cell adhesion(19)
CADM1Cell-cell adhesion(20)

Table II.

Methylated genes in endometrial cancer.

Table II.

Methylated genes in endometrial cancer.

Gene nameFunction(Refs.)
hMLH1DNA repair(2226)
MGMTDNA repair(27)
APCWnt/β catenin pathway(25,28)
CDHWnt/β catenin pathway(29,30)
E-cadherinWnt/β catenin pathway(25,2932)
PTENAkt pathway(33)
RASSF1AApoptosis(30,34)
CASP8Apoptosis(35)
GSTP1Apoptosis(30,36)
P73Apoptosis(33)
PAR-4Apoptosis(37)
SOX2Apoptosis(38)
CHFRCell cycle checkpoint(39)
P14Cell cycle checkpoint(40)
P16Cell cycle checkpoint(40,41)
ARReceptor(42)
PRReceptor(43)
RARβReceptor(44)
TESTIN Epithelial-to-mesenchymal transition(45)
GATA4Transcription factor(46)
TIMP3Proteasome system(47)

Diagnosis of cervical cancer and epigenetic abnormalities

A recent study demonstrated that combined detection of methylated chromosome 3 open reading frame 18, junctional adhesion molecule 3 (JAM3) and ankyrin repeat domain 18C (ANKRD18CP) provided good diagnostic outcomes (48). The sensitivity for lesions of CIN2 or higher was 74% in this screening, which is comparable with the 79% sensitivity of the high-risk human papillomavirus (HPV) DNA test. The specificity of this screening was 76%, exceeding the value of 42% for the high-risk HPV DNA test (P<0.05). Combined detection using JAM3, glial cell line-derived neurotrophic factor family receptor α 1 and ANKRD18CP has 73% sensitivity and 77% specificity, and that using JAM3/ANKRD18CP has 72% sensitivity and 79% specificity, which are also considered as relatively good diagnostic outcomes. Higher specificity is preferable for the prevention of unnecessary follow-up. Furthermore, if HPV vaccination is promoted, the incidence of HPV-negative cervical cancer may increase and the HPV test may become less significant. Therefore, an alternative screening method with a higher specificity for HPV is needed and the methylated markers found in the abovementioned studies are candidates for this screening.

5-Hydroxymethylcytosine (5hmC) has been shown to be a prognostic marker for squamous cell carcinoma of the cervix (49). 5hmC is a cytosine modification found in various biological species that is present at high levels in human and murine nerve tissues. 5hmC is a novel DNA modification marker in mammalian genomes and is involved in DNA demethylation in epigenetic regulation. Tet protein hydroxylates 5-methylcytosine (5mC) to form 5hmC, and this oxidation pathway is involved in the activation or reduction of gene expression (50). The expression levels of 5hmC, 5mC and TET1, TET2 and TET3 were determined in squamous cell carcinoma tissues from the uterine cervix in 140 patients and in normal uterine cervix tissues in 40 healthy volunteers. In addition, the prognostic values of 5hmC, 5mC and TET2 were assessed for therapeutic outcomes in squamous cell carcinoma of the cervix. The results demonstrated that 5hmC is significantly decreased and 5mC is significantly increased in squamous cell carcinoma of the cervix compared with normal cervical tissues. Furthermore, the expression of TET2, but not that of TET1 or TET3, was decreased in squamous cell carcinoma of the cervix. A reduced level of 5hmC was associated with a poor outcome for patients with squamous cell carcinoma of the cervix. 5hmC expression was an independent prognostic factor for event-free and overall survival in these patients, and is likely to be useful as a prognostic marker for cervical cancer in clinical practice.

Targeting of epigenetic abnormalities in cervical cancer therapy

A recent study demonstrated the efficacy of cisplatin combined with 5-azacytidine, which acts as a demethylating agent (51). Chemotherapy with cisplatin inhibited cervical cancer cells, but the same dose of cisplatin with added 5-azacytidine exerted a more potent inhibitory effect. Combination of another demethylating agent, 5-aza-2′-deoxycytidine, with taxol or cisplatin, also inhibited growth of drug-resistant cervical cancer cells (52), with taxol exerting a particularly strong effect. The standard of care for cervical cancer is a combination of paclitaxel and cisplatin. Therefore, potentiating the effects of these drugs and eliminating chemotherapy resistance are important clinical advances.

Combination therapy including tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is also effective (53). TRAIL is a cytokine in the tumor necrosis factor (TNF) family that induces apoptosis via its receptor in various human tumor cells, but not in normal cells. TNFRSF10C (DcR1) and TNFRSF10D (DcR2) are decoy receptors and multi-drug resistance (MDR) genes, and have been identified as epigenetically inactivated genes. TNFRSF10C methylation is found in precancerous lesions, which suggests that this is an early event in the cervical neoplastic process. Cervical cancer cells containing inactivated DcR1 and DcR2 exhibited increased TRAIL-induced apoptosis through activation of extrinsic apoptosis pathways (53). Therefore, patients without expression of decoy receptors are predicted to exhibit an enhanced response to combined therapy with TRAIL. This suggests that a strategy aimed at downregulating decoy receptors may be a feasible approach to treatment using TRAIL.

Diagnosis of endometrial cancer and epigenetic abnormalities

Endometrial cancer induced by epigenetic abnormalities is frequently caused by breakdown of the DNA mismatch repair (MMR) system. Under normal conditions, this system perceives DNA insertions and deletions, induces expression of repair enzymes, deletes affected DNA regions and resynthesizes damaged genes via DNA polymerases (54). If the MMR system is dysfunctional, DNA replication mistakes are not repaired and microsatellites (repeats of 1–5 bases) are likely to increase or decrease. This condition is referred to as microsatellite instability (MSI). MMR genes may be abnormally methylated and cancer develops when MSI occurs in regions of tumor suppressor genes. Numerous microsatellites are detectable by PCR and are found in 20–40% of cases of endometrial cancer (22,5557).

Genes involved in the MMR system include hMLH1, MutS protein homolog 2 (hMSH2) and hMSH6, and postmeiotic segregation increased 2 (hPMS2) is frequently abnormally methylated (56). hMLH1 methylation has been found in 40.1% of cases of endometrial cancer and in 14.3% of cases of atypical endometrial hyperplasia, a precancerous endometrial lesion; therefore, abnormal methylation of hMLH1 is considered to be significantly involved in carcinogenesis (43).

A recent study of cullin-5 (CUL5) demonstrated that this gene is involved in breast and cervical cancer and hepatocellular carcinoma. CUL5 was significantly downregulated in serous endometrial adenocarcinoma with a poor prognosis, compared with its level in endometrial carcinoma with a good prognosis (58). The action of CUL5 involves non-coding RNAs (ncRNAs), which are RNAs that do not include genetic information for protein synthesis. These include microRNAs (miRNAs) of 18–25 nucleotides that lead to mature miRNAs that bind to the 3′-untranslated region (UTR) of target genes and inhibit gene function (46). Two highly-conserved miR-182-binding regions are present in the 3′-UTR of CUL5, and CUL5 targets miR-182 in endometrial cancer. CUL5 downregulation causes miR-182 upregulation and progression of endometrial cancer (58). Zhou et al demonstrated that miR-30c overexpression inhibited metastasis-associated gene-1 (MTA1) (59). miR-30c is decreased in endometrial, ovarian, breast and gastric cancer (6063) and it is involved in carcinogenesis through its association with MTA1.

Targeting of epigenetic abnormalities in endometrial cancer therapy

Unlike genetic changes, epigenetic alterations are not irreversible, and it may be possible to use molecular-targeted drugs to induce transcription of tumor suppressor genes via demethylation (64). The effects of demethylating drugs on tumor suppression include inhibition of growth of cervical cancer cells in vivo by demethylation of APC by hydralazine (65) and 5-azacytidine-induced expression of APC and hMLH1 in endometrial cancer in vitro (22). With regard to treatment of epigenetic changes by miRNAs, Tsuruta et al demonstrated in vivo and in vitro that exogenous miR-152 targeting of DNA methyltransferase 1 (DNMT1) in endometrial cancer cells without expression of miR-152 inhibited tumor proliferation (66). Zhao et al recently reported similar results for miR-126 targeting of insulin receptor substrate 1 (67). These results suggest that cancer therapy may be established by delivery of dsRNA.

DNMTs and histone deacetylases (HDACs) are key enzymes mediating epigenetic regulation of gene expression. The majority of events involving DNA overexpression and histone deacetylation in promoter regions are associated with transcriptional downregulation or silencing, and epigenetic silencing of tumor suppressor genes plays an important role in malignant transformation (68). DNMT inhibitors induce DNA demethylation and HDAC inhibitors cause histone acetylation, resulting in reactivation of silenced genes and functional and morphological changes in cancer cells. DNMT inhibitor-mediated demethylation of the cadherin (CDH)1 promoter results in upregulation of E-cadherin in endometrial cancer cells. A combination of DNMT and HDAC inhibitors upregulated CDH1 and downregulated B-cell lymphoma 2 at the mRNA level, inducing cell cycle arrest and apoptosis (69). This combination has a synergistic effect and is likely to become a new treatment for endometrial cancer.

Conclusion

In uterine cancer, epigenetic and genetic changes are intertwined in a complex manner, resulting in cancer onset. Epigenetic changes cause phenomena including inhibition of apoptosis, DNA repair inhibition, overgrowth and enhanced cancer growth. Genes involved in epigenetic changes are inhibited by methylation, and reactivation of these genes may inhibit the growth of cancer cells. Epigenetic information may also be useful in screening for uterine cancer with high sensitivity and specificity and for development of novel molecular-targeted drugs, leading to improved treatment outcomes in uterine and other cancers. Therefore, further studies on cancer epigenetics are essential for improvement of cancer therapy.

References

1 

Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: GLOBOCAN 2012 v1.1Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 (Internet). Lyon, France: International Agency for Research on Cancer; 2014 http://globocan.iarc.frAccessed. August 10–2016

2 

Bhat S, Kabekkodu SP, Noronha A and Satyamoorthy K: Biological implications and therapeutic significance of DNA methylation regulated genes in cervical cancer. Biochimie. 121:298–311. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Banzai C, Nishino K, Quan J, Yoshihara K, Sekine M, Yahata T and Tanaka K: Gynecological Cancer Registry of Niigata: Promoter methylation of DAPK1, FHIT, MGMT, and CDKN2A genes in cervical carcinoma. Int J Clin Oncol. 19:127–132. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Liu SS, Leung RC, Chan KY, Chiu PM, Cheung AN, Tam KF, Ng TY, Wong LC and Ngan HY: p73 expression is associated with the cellular radiosensitivity in cervical cancer after radiotherapy. Clin Cancer Res. 10:3309–3316. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Roz L, Gramegna M, Ishii H, Croce CM and Sozzi G: Restoration of fragile histidine triad (FHIT) expression induces apoptosis and suppresses tumorigenicity in lung and cervical cancer cell lines. Proc Natl Acad Sci USA. 99:3615–3320. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Snellenberg S, Cillessen SA, Van Criekinge W, Bosch L, Meijer CJ, Snijders PJ and Steenbergen RD: Methylation-mediated repression of PRDM14 contributes to apoptosis evasion in HPV-positive cancers. Carcinogenesis. 35:2611–2618. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wu Q, Shi H, Suo Z and Nesland JM: 5′-CpG island methylation of the FHIT gene is associated with reduced protein expression and higher clinical stage in cervical carcinomas. Ultrastruct Pathol. 27:417–422. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Kitkumthorn N, Yanatatsanajit P, Kiatpongsan S, Phokaew C, Triratanachat S, Trivijitsilp P, Termrungruanglert W, Tresukosol D, Niruthisard S and Mutirangura A: Cyclin A1 promoter hypermethylation in human papillomavirus-associated cervical cancer. BMC Cancer. 6:552006. View Article : Google Scholar : PubMed/NCBI

9 

Chujan S, Kitkumthorn N, Siriangkul S and Mutirangura A: CCNA1 promoter methylation: A potential marker for grading Papanicolaou smear cervical squamous intraepithelial lesions. Asian Pac J Cancer Prev. 15:7971–7975. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Kabekkodu SP, Bhat S, Radhakrishnan R, Aithal A, Mascarenhas R, Pandey D, Rai L, Kushtagi P, Mundyat GP and Satyamoorthy K: DNA promoter methylation-dependent transcription of the double C2-like domain β (DOC2B) gene regulates tumor growth in human cervical cancer. J Biol Chem. 289:10637–10649. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Aoki K and Taketo MM: Adenomatous polyposis coli (APC): A multi-functional tumor suppressor gene. J Cell Sci. 120:3327–3335. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Lin YW, Chung MT, Lai HC, De Yan M, Shih YL, Chang CC and Yu MH: Methylation analysis of SFRP genes family in cervical adenocarcinoma. J Cancer Res Clin Oncol. 135:1665–1674. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Narayan G, Arias-Pulido H, Nandula SV, Basso K, Sugirtharaj DD, Vargas H, Mansukhani M, Villella J, Meyer L, Schneider A, et al: Promoter hypermethylation of FANCF: Disruption of Fanconi Anemia-BRCA pathway in cervical cancer. Cancer Res. 64:2994–2997. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Henken FE, Wilting SM, Overmeer RM, van Rietschoten JG, Nygren AO, Errami A, Schouten JP, Meijer CJ, Snijders PJ and Steenbergen RD: Sequential gene promoter methylation during HPV-induced cervical carcinogenesis. Br J Cancer. 97:1457–1464. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Virmani AK, Muller C, Rathi A, Zoechbauer-Mueller S, Mathis M and Gazdar AF: Aberrant methylation during cervical carcinogenesis. Clin Cancer Res. 7:584–589. 2001.PubMed/NCBI

16 

Bai LX, Wang JT, Ding L, Jiang SW, Kang HJ, Gao CF, Chen X, Chen C and Zhou Q: Folate deficiency and FHIT hypermethylation and HPV 16 infection promote cervical cancerization. Asian Pac J Cancer Prev. 15:9313–9317. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Ivanova T, Petrenko A, Gritsko T, Vinokourova S, Eshilev E, Kobzeva V, Kisseljov F and Kisseljova N: Methylation and silencing of the retinoic acid receptor-beta 2 gene in cervical cancer. BMC Cancer. 2:42002. View Article : Google Scholar : PubMed/NCBI

18 

De Strooper LM, van Zummeren M, Steenbergen RD, Bleeker MC, Hesselink AT, Wisman GB, Snijders PJ, Heideman DA and Meijer CJ: CADM1, MAL and miR124-2 methylation analysis in cervical scrapes to detect cervical and endometrial cancer. J Clin Pathol. 67:1067–1071. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Kim JH, Choi YD, Lee JS, Lee JH, Nam JH and Choi C: Assessment of DNA methylation for the detection of cervical neoplasia in liquid-based cytology specimens. Gynecol Oncol. 116:99–104. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Yadav SS, Prasad SB, Das M, Kumari S, Pandey LK, Singh S, Pradhan S and Narayan G: Epigenetic silencing of CXCR4 promotes loss of cell adhesion in cervical cancer. Biomed Res Int. 2014:5814032014. View Article : Google Scholar : PubMed/NCBI

21 

Overmeer RM, Henken FE, Snijders PJ, Claassen-Kramer D, Berkhof J, Helmerhorst TJ, Heideman DA, Wilting SM, Murakami Y, Ito A, et al: Association between dense CADM1 promoter methylation and reduced protein expression in high-grade CIN and cervical SCC. J Pathol. 215:388–397. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Bischoff J, Ignatov A, Semczuk A, Schwarzenau C, Ignatov T, Krebs T, Küster D, Przadka-Rabaniuk D, Roessner A, Costa SD and Schneider-Stock R: hMLH1 promoter hypermethylation and MSI status in human endometrial carcinomas with and without metastases. Clin Exp Metastasis. 29:889–900. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Guida M, Sanguedolce F, Bufo P, Di Spiezio Sardo A, Bifulco G, Nappi C and Pannone G: Aberrant DNA hypermethylation of hMLH-1 and CDKN2A/p16 genes in benign, premalignant and malignant endometrial lesions. Eur J Gynaecol Oncol. 30:267–270. 2009.PubMed/NCBI

24 

Pallarés J, Velasco A, Eritja N, Santacana M, Dolcet X, Cuatrecasas M, Palomar-Asenjo V, Catasús L, Prat J and Matias-Guiu X: Promoter hypermethylation and reduced expression of RASSF1A are frequent molecular alterations of endometrial carcinoma. Mod Pathol. 21:691–699. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Banno K, Yanokura M, Susumu N, Kawaguchi M, Hirao N, Hirasawa A, Tsukazaki K and Aoki D: Relationship of the aberrant DNA hypermethylation of cancer-related genes with carcinogenesis of endometrial cancer. Oncol Rep. 16:1189–1196. 2006.PubMed/NCBI

26 

Jo H, Kim JW, Kang GH, Park NH, Song YS, Kang SB and Lee HP: Association of promoter hypermethylation of the RASSF1A gene with prognostic parameters in endometrial cancer. Oncol Res. 16:205–209. 2006.PubMed/NCBI

27 

Lee KE: Immunohistochemical assessment of O(6)-methylguanine-DNA methyltransferase (MGMT) and Its relationship with p53 expression in endometrial cancers. J Cancer Prev. 18:351–354. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Ignatov A, Bischoff J, Ignatov T, Schwarzenau C, Krebs T, Kuester D, Costa SD, Roessner A, Semczuk A and Schneider-Stock R: APC promoter hypermethylation is an early event in endometrial tumorigenesis. Cancer Sci. 101:321–327. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Suehiro Y, Okada T, Okada T, Anno K, Okayama N, Ueno K, Hiura M, Nakamura M, Kondo T, Oga A, et al: Aneuploidy predicts outcome in patients with endometrial carcinoma and is related to lack of CDH13 hypermethylation. Clin Cancer Res. 14:3354–3361. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Fiolka R, Zubor P, Janusicova V, Visnovsky J, Mendelova A, Kajo K, Lasabova Z, Plank L and Danko J: Promoter hypermethylation of the tumor-suppressor genes RASSF1A, GSTP1 and CDH1 in endometrial cancer. Oncol Rep. 30:2878–2886. 2013.PubMed/NCBI

31 

Park JH, Lee BI, Song ES, Whang SO, Lee WY and Cho SJ: Hypermethylation of E-cadherin in endometrial carcinoma. J Gynecol Oncol. 19:241–245. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Saito T, Nishimura M, Yamasaki H and Kudo R: Hypermethylation in promoter region of E-cadherin gene is associated with tumor dedifferention and myometrial invasion in endometrial carcinoma. Cancer. 97:1002–1009. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Yang HJ, Liu VW, Wang Y, Tsang PC and Ngan HY: Differential DNA methylation profiles in gynecological cancers and correlation with clinico-pathological data. BMC Cancer. 6:2122006. View Article : Google Scholar : PubMed/NCBI

34 

Kang S, Lee JM, Jeon ES, Lee S, Kim H, Kim HS, Seo SS, Park SY, Sidransky D and Dong SM: RASSF1A hypermethylation and its inverse correlation with BRAF and/or KRAS mutations in MSI-associated endometrial carcinoma. Int J Cancer. 119:1316–1321. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Sánchez-Vega F, Gotea V, Petrykowska HM, Margolin G, Krivak TC, DeLoia JA, Bell DW and Elnitski L: Recurrent patterns of DNA methylation in the ZNF154, CASP8 and VHL promoters across a wide spectrum of human solid epithelial tumors and cancer cell lines. Epigenetics. 8:1355–1372. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Chan QK, Khoo US, Chan KY, Ngan HY, Li SS, Chiu PM, Man LS, Ip PP, Xue WC and Cheung AN: Promoter methylation and differential expression of pi-class glutathione S-transferase in endometrial carcinoma. J Mol Diagn. 7:8–16. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Moreno-Bueno G, Fernandez-Marcos PJ, Collado M, Tendero MJ, Rodriguez-Pinilla SM, Garcia-Cao I, Hardisson D, Diaz-Meco MT, Moscat J, Serrano M and Palacios J: Inactivation of the candidate tumor suppressor par-4 in endometrial cancer. Cancer Res. 67:1927–1934. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Wong OG, Huo Z, Siu MK, Zhang H, Jiang L, Wong ES and Cheung AN: Hypermethylation of SOX2 promoter in endometrial carcinogenesis. Obstet Gynecol Int. 2010:pii:6825042010. View Article : Google Scholar

39 

Muraki Y, Banno K, Yanokura M, Kobayashi Y, Kawaguchi M, Nomura H, Hirasawa A, Susumu N and Aoki D: Epigenetic DNA hypermethylation: Clinical applications in endometrial cancer (Review). Oncol Rep. 22:967–972. 2009.PubMed/NCBI

40 

Zhang QY, Yi DQ, Zhou L, Zhang DH and Zhou TM: Status and significance of CpG island methylator phenotype in endometrial cancer. Gynecol Obstet Invest. 72:183–191. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Semczuk A, Boltze C, Marzec B, Szczygielska A, Roessner A and Schneider-Stock R: p16INK4A alterations are accompanied by aberrant protein immunostaining in endometrial carcinomas. J Cancer Res Clin Oncol. 129:589–596. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Sasaki M, Oh BR, Dharia A, Fujimoto S and Dahiya R: Inactivation of the human androgen receptor gene is associated with CpG hypermethylation in uterine endometrial cancer. Mol Carcinog. 29:59–66. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Sasaki M, Dharia A, Oh BR, Tanaka Y, Fujimoto S and Dahiya R: Progesterone receptor B gene inactivation and CpG hypermethylation in human uterine endometrial cancer. Cancer Res. 61:97–102. 2001.PubMed/NCBI

44 

Li R, Saito T, Tanaka R, Satohisa S, Adachi K, Horie M, Akashi Y and Kudo R: Hypermethylation in promoter region of retinoic acid receptor-beta gene and immunohistochemical findings on retinoic acid receptors in carcinogenesis of endometrium. Cancer Lett. 219:33–40. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Dong R, Pu H, Wang Y, Yu J, Lian K and Mao C: TESTIN was commonly hypermethylated and involved in the epithelial-mesenchymal transition of endometrial cancer. APMIS. 123:394–400. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Chmelarova M, Kos S, Dvorakova E, Spacek J, Laco J, Ruszova E, Hrochova K and Palicka V: Importance of promoter methylation of GATA4 and TP53 genes in endometrioid carcinoma of endometrium. Clin Chem Lab Med. 52:1229–1234. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Catasus L, Pons C, Muñoz J, Espinosa I and Prat J: Promoter hypermethylation contributes to TIMP3 down-regulation in high stage endometrioid endometrial carcinomas. Histopathology. 62:632–641. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Boers A, Wang R, van Leeuwen RW, Klip HG, de Bock GH, Hollema H, van Criekinge W, de Meyer T, Denil S, van der Zee AGJ, et al: Discovery of new methylation markers to improve screening for cervical intraepithelial neoplasia grade 2/3. Clin Epigenetics. 8:292016. View Article : Google Scholar : PubMed/NCBI

49 

Zhang LY, Han CS, Li PL and Zhang XC: 5-Hydroxyme-thylcytosine expression is associated with poor survival in cervical squamous cell carcinoma. Jpn J Clin Oncol. 46:427–434. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Iyer LM, Tahiliani M, Rao A and Aravind L: Prediction of novel families of enzymes involved in oxidative and other complex modifications of bases in nucleic acids. Cell Cycle. 8:1698–1710. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Sood S and Srinivasan R: Alterations in gene promoter methylation and transcript expression induced by cisplatin in comparison to 5-azacytidine in HeLa and SiHa cervical cancer cell lines. Mol Cell Biochem. 404:181–191. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Chen CC, Lee KD, Pai MY, Chu PY, Hsu CC, Chiu CC, Chen LT, Chang JY, Hsiao SH and Leu YW: Changes in DNA methylation are associated with the development of drug resistance in cervical cancer cells. Cancer Cell Int. 15:982015. View Article : Google Scholar : PubMed/NCBI

53 

Narayan G, Xie D, Ishdorj G, Scotto L, Mansukhani M, Pothuri B, Wright JD, Kaufmann AM, Schneider A, Arias-Pulido H and Murty VV: Epigenetic inactivation of TRAIL decoy receptors at 8p12-21.3 commonly deleted region confers sensitivity to Apo2L/trail-Cisplatin combination therapy in cervical cancer. Genes Chromosomes Cancer. 55:177–189. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Jascur T and Boland CR: Structure and function of the components of the human DNA mismatch repair system. Int J Cancer. 119:2030–2035. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Eto T, Zhao Y, Maruyama A, Miyashita K, Yasui A, Nakao S, Taguchi K, Shimokawa M, Oda S and Saito T: Modal variety of microsatellite instability in human endometrial carcinomas. J Cancer Res Clin Oncol. 142:353–363. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Bilbao C, Lara PC, Ramírez R, Henríquez-Hernández LA, Rodríguez G, Falcón O, León L, Perucho M, Díaz-Chico BN and Díaz-Chico JC: Microsatellite instability predicts clinical outcome in radiation-treated endometrioid endometrial cancer. Int J Radiat Oncol Biol Phys. 76:9–13. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Hampel H, Frankel W, Panescu J, Lockman J, Sotamaa K, Fix D, Comeras I, La Jeunesse J, Nakagawa H, Westman JA, et al: Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients. Cancer Res. 66:7810–7817. 2006. View Article : Google Scholar : PubMed/NCBI

58 

Devor EJ, Schickling BM, Reyes HD, Warrier A, Lindsay B, Goodheart MJ, Santillan DA and Leslie KK: Cullin-5, a ubiquitin ligase scaffold protein, is significantly underexpressed in endometrial adenocarcinomas and is a target of miR-182. Oncol Rep. 35:2461–2465. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Zhou H, Xu X, Xun Q, Yu D, Ling J, Guo F, Yan Y, Shi J and Hu Y: microRNA-30c negatively regulates endometrial cancer cells by targeting metastasis-associated gene-1. Oncol Reports. 27:807–812. 2012.

60 

Boren T, Xiong Y, Hakam A, Wenham R, Apte S, Wei Z, Kamath S, Chen DT, Dressman H and Lancaster JM: MicroRNAs and their target messenger RNAs associated with endometrial carcinogenesis. Gynecol Oncol. 110:206–215. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Lee H, Park CS, Deftereos G, Morihara J, Stern JE, Hawes SE, Swisher E, Kiviat NB and Feng Q: MicroRNA expression in ovarian carcinoma and its correlation with clinicopathological features. World J Surg Oncol. 10:1742012. View Article : Google Scholar : PubMed/NCBI

62 

Li S, Yang C, Zhai L, Zhang W, Yu J, Gu F, Lang R, Fan Y, Gong M, Zhang X and Fu L: Deep sequencing reveals small RNA characterization of invasive micropapillary carcinomas of the breast. Breast Cancer Res Treat. 136:77–87. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, Rossi S, Alder H, Liu CG, Oue N, Yasui W, et al: Relation between microRNA expression and progression and prognosis of gastric cancer: A microRNA expression analysis. Lancet Oncol. 11:136–146. 2010. View Article : Google Scholar : PubMed/NCBI

64 

Duenas-Gonzalez A, Medina-Franco JL, Chavez-Blanco A, Dominguez-Gomez G and Fernández-de Gortari E: Developmental DNA methyltransferase inhibitors in the treatment of gynecologic cancers. Expert Opin Pharmacother. 17:323–338. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Song Y and Zhang C: Hydralazine inhibits human cervical cancer cell growth in vitro in association with APC demethylation and re-expression. Cancer Chemother Pharmacol. 63:605–613. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Tsuruta T, Kozaki K, Uesugi A, Furuta M, Hirasawa A, Imoto I, Susumu N, Aoki D and Inazawa J: miR-152 is a tumor suppressor microRNA that is silenced by DNA hypermethylation in endometrial cancer. Cancer Res. 71:6450–6462. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Zhao X, Zhu D, Lu C, Yan D, Li L and Chen Z: MicroRNA-126 inhibits the migration and invasion of endometrial cancer cells by targeting insulin receptor substrate 1. Oncol Lett. 11:1207–1212. 2016.PubMed/NCBI

68 

Ren J, Zhang J, Cai H, Li Y, Zhang Y, Zhang X, Zhao D, Li Z, Ma H, Wang J, et al: HDAC as a therapeutic target for treatment of endometrial cancers. Curr Pharm Des. 20:1847–1856. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Yi TZ, Li J, Han X, Guo J, Qu Q, Guo L, Sun HD and Tan WH: DNMT inhibitors and HDAC inhibitors regulate E-cadherin and bcl-2 expression in endometrial carcinoma in vitro and in vivo. Chemotherapy. 58:19–29. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 7 Issue 5

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yanokura M, Banno K, Kobayashi Y, Nomura H, Hayashi S, Tominaga E and Aoki D: Recent findings on epigenetic gene abnormalities involved in uterine cancer (Review). Mol Clin Oncol 7: 733-737, 2017
APA
Yanokura, M., Banno, K., Kobayashi, Y., Nomura , H., Hayashi, S., Tominaga, E., & Aoki, D. (2017). Recent findings on epigenetic gene abnormalities involved in uterine cancer (Review). Molecular and Clinical Oncology, 7, 733-737. https://doi.org/10.3892/mco.2017.1428
MLA
Yanokura, M., Banno, K., Kobayashi, Y., Nomura , H., Hayashi, S., Tominaga, E., Aoki, D."Recent findings on epigenetic gene abnormalities involved in uterine cancer (Review)". Molecular and Clinical Oncology 7.5 (2017): 733-737.
Chicago
Yanokura, M., Banno, K., Kobayashi, Y., Nomura , H., Hayashi, S., Tominaga, E., Aoki, D."Recent findings on epigenetic gene abnormalities involved in uterine cancer (Review)". Molecular and Clinical Oncology 7, no. 5 (2017): 733-737. https://doi.org/10.3892/mco.2017.1428