Frequent promoter methylation of HOXD10 in endometrial carcinoma and its pathological significance

  • Authors:
    • Fan Yang
    • Dongchen Liu
    • Yupeng Deng
    • Jun Wang
    • Shuyu Mei
    • Shuang Ge
    • Hailing Li
    • Cuijuan Zhang
    • Tingguo Zhang
  • View Affiliations

  • Published online on: March 19, 2020     https://doi.org/10.3892/ol.2020.11467
  • Pages: 3602-3608
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Homeobox D 10 (HOXD10) is important in cell differentiation and morphogenesis and serves as a tumor suppressor gene (TSG) in a number of malignancies. The present study investigated its promoter methylation status and association with the clinicopathological features of endometrial cancer (EC), and measured HOXD10 protein expression levels. EC samples (n=62), including 50 endometroid adenocarcinoma (EA) and 12 mucinous endometrial carcinoma samples (EC) and 70 non‑cancerous samples were collected. All samples were evaluated for the methylation status of several TSGs, including HOXD10, using methylation‑specific PCR. HOXD10 expression level was evaluated using immunohistochemistry. 5‑Aza‑2‑deoxycytidine treatment was performed in the EC cell line Ishikawa to observe the change in HOXD10 expression levels. HOXD10 promoter methylation was more frequent in cancer samples (P<0.001). Downregulation of HOXD10 in EC samples was confirmed at the protein level using immunohistochemistry (P<0.001) and immunohistochemical staining was negatively associated with methylation status (P<0.05). Less HOXD10 protein was expressed in MEC compared with EA samples (P<0.001). The HOXD10 promoter was hypermethylated in both EA and MEC, causing decreased HOXD10 protein expression levels in EC cells. HOXD10 expression levels were partially reversed by 5‑Aza‑2‑deoxycytidine treatment. The results of the present study demonstrated that epigenetic silencing of HOXD10 putatively contributed to the tumorigenesis of EA. Although there was no significant difference in HOXD10 methylation between EA and MEC, HOXD10 protein expression levels differed between these two diseases, indicating that it may be a useful protein biomarker for distinguishing between these two lesions.

Introduction

Endometrial carcinoma (EC) is one of the most common gynecological malignancies in developed countries, accounting for 20–30% of female malignancies, with a prevalence that is increasing annually (1,2). Early diagnosis and treatment has allowed a possible 5-year survival rate of 90% (3,4). Mucinous endometrial carcinoma (MEC) is an independent and infrequent pathological pattern of EC and accounts for 1–9% of all uterine carcinomas (5). Previous studies describing its pathomorphism and diagnostic criteria have suggested that MEC and endometroid adenocarcinoma (EA) differ with respect to patient age and lymphatic metastasis (69). MEC is more frequent in older women, and is more likely to involve lymphatic metastasis (69). Compared with EA, MEC samples exhibit decreased paired box 2 expression levels, low TP53 mutation rates and increased CD10, estrogen receptor (ER), progesterone receptor (PR), K-ras, p16, c-MET, epidermal growth factor receptor (EGFR), PTEN and PD-L1 expression levels and sporadic promoter hypermethylation of the mutL homolog 1 gene (1015).

Like many malignancies, endometrial cancer is a complex disease driven by genetic, epigenetic and environmental factors (1618). DNA methylation at the 5-C of a cytosine ring of a CpG island is one of the most important epigenetic alterations in cancer initiation (19). Studies have demonstrated that aberrant DNA methylation is associated with malignant formation (2023). Tumor suppressor genes (TSG) may be hypermethylated, which leads to decreased expression levels and alteration of cell growth, differentiation, proliferation and apoptosis, resulting in the development of tumors (2023). Therefore, assaying DNA methylation status may be a method for early diagnosis (24,25).

Hypermethylation of promoters of a number of TSGs, such as EGF-containing fibulin extracellular matrix protein 1, glutathione S-transferase P1, suppressor of cytokine signaling 3, 3OST2, basic helix-loop-helix family member E22/cysteine deoxygenase 1/CUGBP Elva-like family member 4, SHP1 and transmembrane protein with EGF-like and two follistatin-like domains 2, have been studied in certain EC tissues and cell lines (such as Ishikawa and KLE) and the methylation frequency is high (2631). The present study investigated five TSGs (HOXD10, SHH, ZNF545, PCDH17 and MEIS1) whose promoters are reported to be hypermethylated in other malignancies but have not been evaluated for methylation in EC (3240). For example, HOXD10 is hypermethylated and lowly expressed in colon adenocarcinoma cells, which downregulates the RHOC/AKT/MAPK pathway to enhance apoptosis and restrict the proliferation, migration and invasion of colon adenocarcinoma (32,33). Methylation of the Shh promoter and reduced expression of SHH is frequent in basal cell carcinoma (34,35). ZNF545 functions as a tumor suppressor in colorectal cancer and is frequently inactivated by promoter methylation (36,37). Hypermethylation of PCDH17 is correlated to poor prognosis in acute lymphoblastic leukemia (38,39). MEIS1 genes are frequently hypermethylated in different types of leukemia (40). The present study measured the methylation statuses of these genes, and then selected the most hypermethylated genes and measured protein expression levels in tumor and control samples. The present study aimed to analyze the methylation status of the TSG in EC and to elucidate the association between methylation, expression levels and clinicopathological characteristics. In order to further validate the association between the methylation status of a gene and its expression level, the present study detected the expression level of HOXD10 before and after DNA methylation transferase inhibitor treatment.

Materials and methods

Patients and samples

A total of 132 well-conserved paraffin-embedded tissue blocks were obtained from the Department of Pathology, Shandong University Qilu Hospital (Shandong, China), which were initially taken between 2006 and 2015, including endometrial adenocarcinoma (n=50), mucinous endometrial carcinoma (n=12), simple hyperplasia (n=22) and complex hyperplasia (n=48). Patient data, such as age, tumor differentiation, depth of myometrial invasion and lymph node metastasis, were collected. Case diagnoses were made according to World Health Organization Classification of Tumors of Female Reproductive Organs (4th edition) (41). The present study was approved by the Ethical Research Committee at Shandong University (approval no. mecsdums 2012032).

Cell lines and culture

The human endometrial carcinoma cell line Ishikawa was obtained from the European Collection of Cell Cultures (Sigma-Aldrich; Merck KGaA) and maintained at 37°C in Dulbecco's modified Eagle's medium/Ham's F-12 medium (DMEM/F12; Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.). Cell lines were treated at 37°C for 72 h with 5-Aza-2′-deoxycytidine (5-Aza-CdR; Sigma-Aldrich; Merck KGaA) at a concentration of 1, 2, 5 µM as a demethylation treatment. Media and 5-Aza-CdR were replaced every 24 h.

Methylation-specific PCR (MSP)

DNA samples were extracted from paraffin-embedded tissue blocks using a genomic DNA purification kit (Qiagen GmbH), and treated to convert unmethylated cytosine to uracil using a cpGenome DNA modification kit (InterGen Co.). Primer sequences, conditions and product length for MSP are presented in Table SI. MSP reactions were performed in a 10 µl system, using 1 µl (20 ng) template DNA for each reaction. H2O was used as a negative control.

Immunohistochemistry

A two-step method was used according to the manufacturer's instructions regarding primary antibody and secondary antibody described below. Sections (5-µm thick) were pre-treated using sodium citrate buffer (pH 6.0; 0.01 mol/l; Beijing Solarbio Science & Technology Co., Ltd.) at 98°C for 5 min to retrieve cell antigens and blocked with goat serum (1:10; cat. no. ZLI-9021; OriGene Technologies, Inc.) at room temperature for 20 min. Sections were incubated overnight at 4°C with primary antibody (anti-HOXD10; 1:150; cat. no. ab172865; Abcam). Colonic carcinoma tissue sections (5-µm thick) were used as the positive control. The primary antibody was replaced with phosphate-buffered saline and was used as a negative control. The sections were incubated with horseradish peroxidase-labeled secondary antibody (1:100; Universal PV9000 kit; OriGene Technologies) for 30 min at 37°C and visualized using DAB (1:20; Beijing Solarbio Science & Technology Co., Ltd.). The sections were stained with hematoxylin for 2 min at room temperature. Optical microscope (magnification, ×100 and ×400; Carl Zeiss AG) was used for visualization.

A semi-quantitative scoring system was used to obtain a staining score for HOXD10 expression levels. A total of five high power fields from each section were randomly selected, and scores were assigned according to intensity and percentage of stained cells. Scores were averaged to yield a final score. Intensity scores were classified according to staining intensity: Negative staining (staining intensity score, 0); positive yellow staining (staining intensity score, 1); positive brown staining (staining intensity score ≤2); and positive dark brown staining (staining intensity score, 3). Percentage score was classified according to percentage of stained cells: No staining (score =0); positive staining 0–10% (score, 1); positive staining 11–50% (score, 2); positive staining 50–80% (score, 3); 80–100% (score, 4). A two-score average was used as the score of the fields. Final scores of 0–3 were considered negative, while scores >4 were considered positive.

Western blot analysis

Ishikawa cells were harvested and subjected to protein extraction with RIPA lysis buffer (Beijing Solarbio Science & Technology Co., Ltd.). The concentration of protein was measured using the BCA method (BCA Protein Assay kit; cat. no. PC0020; Beijing Solarbio Science & Technology Co., Ltd.). Equal amounts of protein (50 µg) were separated by 10% SDS-PAGE gel and transferred to PVDF membranes (EMD Millipore). After blocking with 5% skim milk (Beijing Solarbio Science & Technology Co., Ltd.) at room temperature for 1 h, the membranes were incubated overnight at 4°C with primary antibodies as follows: Anti-HOXD10 (1:100; cat. no. ab172865; Abcam) and anti-β-actin (1:1,000; cat. no. 4970T; Cell Signaling Technology, Inc.). The immune complexes were incubated with horseradish peroxidase-conjugated secondary antibody. The blots were developed using chemiluminescence (EMD Millipore) with the Las-4000 Imaging system (Fujifilm).

Statistical analysis

SPSS software (version 22.0; IBM, Corp.) was used for statistical analysis. A Pearson χ2 test was used to compare gene methylation status with clinical data among patients with cancer. P<0.05 was considered to indicate a statistically significant difference.

Results

HOXD10 promoter hypermethylation in EC

HOXD10 promoter methylation measured by MSP was greater in cancer samples compared with non-cancerous samples (Fig. 1 and Table I). Hypermethylation of SHH, ZNF545, PCDH17 and MEIS1 in EA and MEC occurred, but a similar methylation frequency was observed in non-cancerous lesions (Fig. S1).

Table I.

MSP data for HOXD10 in primary endometrial lesions and clinical pathological correlations.

Table I.

MSP data for HOXD10 in primary endometrial lesions and clinical pathological correlations.

HOXD10

CategoryMethylatedUnmethylatedP-value
Simple hyperplasia4180.4750
Complex and atypical hyperplasia741
Endometroid adenocarcinoma35150.5380
Mucinous endometrial84
Total endometrial carcinoma4319<0.0001
Total control group1159

[i] HOX, homeobox.

Decreased expression levels of HOXD10 in EC

HOXD10 expression levels were measured in cancer and control samples. Downregulation of HOXD10 in cancer samples was confirmed at the protein level using immunohistochemistry. HOXD10 expression levels were decreased in cancer cases (EA and MEC) compared with the controls (37/62 vs. 14/70; P<0.01). Compared with EA, MEC samples did not express HOXD10 (P<0.001; Table II). In EC, positive staining for HOXD10 occurred in the cytoplasm. For simple and complex hyperplasia, positive staining was evident in the nucleus and cytoplasm (Fig. 2). Staining of colonic carcinoma tissues was provided as positive control (Fig. S2).

Table II.

Immunohistochemistry data of HOXD10 in primary endometrial lesions.

Table II.

Immunohistochemistry data of HOXD10 in primary endometrial lesions.

HOXD10

CategoryPositiveNegativeP-value
EA2525   0.0010
MEC  012
Cancer samples2537<0.0001
Control samples5614

[i] HOX, homeobox; EA, endometroid adenocarcinoma; MEC, mucinous endometrial carcinoma.

Association between HOXD10 methylation, gene expression levels and clinicopathological features in EC

The present study compared methylation of the HOXD10 gene and its expression levels in cancer and control samples. Staining intensity of HOXD10 was negatively associated with promoter methylation (P<0.05). The majority of samples with promoter methylation lacked HOXD10 protein expression levels, indicating a significant association between promoter hypermethylation and transcriptional silencing (P<0.05).

There was no significant difference in HOXD10 methylation between EA and MEC samples (P>0.05). No association between HOXD10 methylation and clinical characteristics (patient age, tumor differentiation, tumor size, depth of myometrial invasion and lymph node metastases) was observed in EA or MEC cases (Table III).

Table III.

Promoter methylation status of HOXD10 in primary endometrial lesions and associations with clinical features.

Table III.

Promoter methylation status of HOXD10 in primary endometrial lesions and associations with clinical features.

HODX10

Clinical featureMethylatedUnmethylatedP-value
Age, years50.56±16.1753.32±16.630.700
Differentiation, n
  Well2213
  Moderate12  30.630
  Poor  9  3
Depth of myometrial invasion, n
  <1/238160.652
  ≥1/2  5  3
Lymph node metastasis, n
  Yes38190.149
  No  5  0
5-Aza-CdR treatment reverses the expression levels of HOXD10 in Ishikawa cells

In order to further validate the association between the methylation status of the HOXD10 gene and its expression level, the present study detected the expression level of HOXD10 in the Ishikawa cell line both before and after DNA methylation transferase inhibitor treatment. As presented in Fig. 3, HOXD10 was weakly expressed in Ishikawa cell lines and the expression level of HOXD10 was increased after 72 h treatment with 5-Aza-CdR (P<0.01). These results suggested that DNA methylation may be associated with the promoter methylation status of HOXD10.

Discussion

DNA promoter methylation is considered to be a promising diagnostic biomarker for cancer. For certain malignancies, aberrant methylation occurs at the promoter of TSG, which plays a crucial role in the regulation of tumor growth and cell differentiation, proliferation and apoptosis. This causes low gene expression levels, leading to alterations in tumor growth and cell differentiation, proliferation and apoptosis, finally resulting in malignancies that may occur prior to tumor formation. Therefore, DNA methylation may be used as a marker in early cancer screening (24,4244). Hypermethylation of specific TSG, as well as low mRNA and protein expression levels, has been reported in a number of human malignancies, including EC (28,45,46).

HOXD10 is one of the ANTP homeobox genes located on chromosome 2 (2p31). This gene cluster consists of 39 genes and multiple transcripts and can be divided into four groups (HOX A, B, C and D) (47). HOX regulates stem cell differentiation and embryonic development (48). Abnormal HOX gene expression levels are responsible for certain malignancies, such as breast, thyroid and ovarian cancer (49). The potential use of homeobox genes as diagnostic and prognostic biomarkers has been described in the literature (50,51).

HOXD10 acts as a TSG in a number of malignancies, such as breast cancer (52), gastric carcinogenesis (53) and cholangiocellular carcinoma (54). It inhibits tumorigenesis (55) and angiogenesis and inhibits vascular EGFR, matrix metalloproteinase 14 and uPAR (56). In certain tumor cell lines, upregulation of HOXD10 decreases invasiveness, migration and survival of cancer cells (40,34,44). For example, HOXD10 acts as a tumor-suppressive factor that suppresses proliferation and invasion, promotes G1/S progression arrest and apoptosis via inhibition of the RHOC/AKT/MAPK pathway in human cholangiocellular carcinoma (40). Low or absent HOXD10 expression levels promoted invasion and migration in colorectal cancer (54,57) and inhibited benign transformation of breast tumors (58).

The present study demonstrated that hypermethylation of the HOXD10 promoter occurred more frequently in EC and thus may be a diagnostic tool for EC compared with non-cancerous tissue. In EA and MEC samples, HOXD10 expression levels were negatively associated with methylation status. Aberrant methylation of the HOXD10 promoter may decrease HOXD10 protein expression levels.

DNA methylation transferase inhibitor 5-Aza-CdR treatment partly reverted the expression level of HOXD10 in the Ishikawa cell line. These findings suggest that aberrant DNA methylation may be one of the mechanisms underlying HOXD10 downregulation in EC.

In pathological and clinical diagnostic practice, MEC is typically mucinous (5), but better biomarkers to clearly identify MEC are lacking. MEC often co-occurs with EA and lacks clear boundaries, which complicates diagnosis (7). Previous studies have suggested that in MEC, immunohistochemistry has demonstrated diffuse positivity for ER/PR and vimentin, high p16 and low Ki-67 labeling index, has proven useful for the differential diagnosis of EA (12,51). In the present study, immunohistochemistry data revealed a lack of HOXD10 expression levels in MEC, so this may be a reliable diagnostic biomarker.

In the present study, MSP data indicated no differences in methylation of the HOXD10 promoter between EA and MEC (EA samples vs. MEC; 70 vs. 66.6%; P>0.05). Dysregulation of HOXD10 may have mechanisms other than promoter methylation. The precise mechanism of dysregulation of HOXD10 in cancer is not clear. In hepatocellular carcinoma, and colon, gastric and papillary thyroid cancer, it has been demonstrated that promoter methylation of HOXD10 can lead to loss of HOXD10 expression levels (32,33,53,59). MicroRNA (miR) can affect HOXD10 expression levels at the post-transcriptional level in breast, colorectal, ovarian, gastric and non-small cell lung cancer, as well as hepatocellular carcinoma, glioma and hemangioma. miR-10b, miR-224, miR-23a, miR-501, miR-92b-3p and miR-376b reportedly accelerate cancer progression by directly targeting HOXD10 within the 3′UTR (6065). Other suggestions of mechanisms of HOXD10 dysregulation include the possible role of long non-coding RNAs (lncRNA), such as HOX transcript antisense RNA (HOTAIR). lncRNA HOTAIR expression is dysregulated in breast cancer and can inhibit HOXD10 expression levels (66). No evidence of HOXD10 mutations has been identified in cancer. Further studies are required to understand the pathology underlying MEC.

In summary, as decreased protein expression levels caused by hypermethylation of TSG may result in tumor progression, the present study demonstrated that promoter hypermethylation-mediated silencing of HOXD10 is a frequent event in EC and thus this may be used to diagnose cancers and guide epigenetic treatment for EA and MEC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant nos. 81272277 and 81372810).

Availability of data and materials

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

Authors' contributions

TZ and CZ designed and supervised experiments. SM, FY and YD wrote the manuscript. SG, JW and HL collected research data. FY and DL performed the MSP and immunohistochemistry. YD and SM conducted the cell line experiments. DL performed clinical evaluations and statistical analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Ethics approval was obtained from the Committee of Ethical Research at Shandong University (approval no. mecsdums2012032). Written informed consent was obtained from participants.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EA

endometroid adenocarcinoma

EC

endometrial carcinoma

MEC

mucinous endometrial carcinoma

ER

estrogen receptor

PR

progesterone receptor

PCDHs

protocadherins

ZNF

zinc finger protein

TSG

tumor suppressor gene

MSP

methylation-specific PCR

5-aza-CdR

5-aza-2′-deoxycytidine

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Morice P, Leary A, Creutzberg C, Abu-Rustum N and Darai E: Endometrial cancer. Lancet. 387:1094–1108. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Eiriksson L, Cuartero J, Steed H, Pearcey R, Capstick V, Schepansky A, Faught W and Dundas G: Assessment of outcomes in surgically staged I/II endometrial adenocarcinoma patients treated with postoperative vaginal vault radiotherapy only. Int J Gynecol Cancer. 20:1356–1362. 2010.PubMed/NCBI

5 

Gungorduk K, Ozdemir A, Ertas IE, Selcuk I, Solmaz U, Ozgu E, Mat E, Gokcu M, Karadeniz T, Akbay S, et al: Is mucinous adenocarcinoma of the endometrium a risk factor for lymph node involvement? A multicenter case-control study. Int J Clin Oncol. 20:782–789. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Jalloul RJ, Elshaikh MA, Ali-Fehmi R, Haley MM, Yoon J, Mahan M and Munkarah AR: Mucinous adenocarcinoma of the endometrium: Case series and review of the literature. Int J Gynecol Cancer. 22:812–818. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Rauh-Hain JA, Vargas RJ, Clemmer J, Clark RM, Bradford LS, Growdon WB, Goodman A, Boruta DM II, Schorge JO and del Carmen MG: Mucinous adenocarcinoma of the endometrium compared with endometrioid endometrial cancer: A SEER analysis. Am J Clin Oncol. 39:43–48. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Musa F, Huang M, Adams B, Pirog E and Holcomb K: Mucinous histology is a risk factor for nodal metastases in endometrial cancer. Gynecol Oncol. 125:541–545. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Galic V, Schiavone MB, Herzog TJ, Holcomb K, Lewin SN, Lu YS, Neugut AI, Hershman DL and Wright JD: Prognostic significance of mucinous differentiation of endometrioid adenocarcinoma of the endometrium. Cancer Invest. 31:500–504. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Jackson CL, Hang S, Hansen K, He M, Sung CJ, Quddus MR, Xiong M, Wang Y, Patel NR, Lawrence WD and Xiong J: Endometrial adenocarcinomas with significant mucinous differentiation: A characterization of intratumoral heterogeneity of KRAS mutations in mucinous and endometrioid histologic components. Int J Gynecol Cancer. 28:241–247. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Sloan EA, Moskaluk CA and Mills AM: Mucinous differentiation with tumor infiltrating lymphocytes is a feature of sporadically methylated endometrial carcinomas. Int J Gynecol Pathol. 36:205–216. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Lax SF: Pathology of endometrial carcinoma. Adv Exp Med Biol. 943:75–96. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Fadare O, Roma AA, Mhawech-Fauceglia P, Parkash V and Rabban JT: The diagnosis of mucinous lesions in endometrial samplings by gynaecological pathologists: An analysis of diagnostic reproducibility. Pathology. 50:276–285. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Jones NL, Xiu J, Chatterjee-Paer S, Buckley de Meritens A, Burke WM, Tergas AI, Wright JD and Hou JY: Distinct molecular landscapes between endometrioid and nonendometrioid uterine carcinomas. Int J Cancer. 140:1396–1404. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Lee S, Sahasrabuddhe VV, Mendoza-Cervantes D, Zhao R and Duggan MA: Tissue-based immunohistochemical biomarker expression in malignant glandular lesions of the uterine cervix: A systematic review. Int J Gynecol Pathol. 37:128–140. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Stampoliou A, Arapantoni-Dadioti P and Pavlakis K: Epigenetic mechanisms in endometrial cancer. J BUON. 21:301–306. 2016.PubMed/NCBI

17 

Zhou JY, Zhang L, Wei LH and Wang JL: Endometrial carcinoma-related genetic factors: Application to research and clinical practice in China. BJOG. 123 (Suppl 3):S90–S96. 2016. View Article : Google Scholar

18 

Banno K, Yanokura M, Iida M, Masuda K and Aoki D: Carcinogenic mechanisms of endometrial cancer: Involvement of genetics and epigenetics. J Obstet Gynaecol Res. 40:1957–1967. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Men C, Chai H, Song X, Li Y, Du H and Ren Q: Identification of DNA methylation associated gene signatures in endometrial cancer via integrated analysis of DNA methylation and gene expression systematically. J Gynecol Oncol. 28:e832017. View Article : Google Scholar : PubMed/NCBI

20 

Caplakova V, Babusikova E, Blahovcova E, Balharek T, Zelieskova M and Hatok J: DNA methylation machinery in the endometrium and endometrial cancer. Anticancer Res. 36:4407–4420. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Romanek-Piva K, Gałczyński K, Adamiak-Godlewska A, Rechberger T and Postawski K: DNA methylation and DNA methyltransferase (DNMT1) activity pattern in endometrial carcinoma. Ginekol Pol. 87:6–10. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Fialkova V, Vidomanova E, Balharek T, Marcinek J, Kudela E, Hanysova S, Visnovsky J, Dobrota D and Hatok J: DNA methylation as mechanism of apoptotic resistance development in endometrial cancer patients. Gen Physiol Biophys. 36:521–529. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Bartosch C, Lopes JM and Jeronimo C: Epigenetics in endometrial carcinogenesis-part 1: DNA methylation. Epigenomics. 9:737–755. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Lai HC, Wang YC, Yu MH, Huang RL, Yuan CC, Chen KJ, Wu CC, Chiang KJ and Chao TK: DNA methylation as a biomarker for the detection of hidden carcinoma in endometrial atypical hyperplasia. Gynecol Oncol. 135:552–559. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Pan Y, Liu G, Zhou F, Su B and Li Y: DNA methylation profiles in cancer diagnosis and therapeutics. Clin Exp Med. 18:1–14. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Yang T, Qiu H, Bao W, Li B, Lu C, Du G, Luo X, Wang L and Wan X: Epigenetic inactivation of EFEMP1 is associated with tumor suppressive function in endometrial carcinoma. PLoS One. 8:e674582013. View Article : Google Scholar : PubMed/NCBI

27 

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. View Article : Google Scholar : PubMed/NCBI

28 

Sheng Y, Wang H, Liu D and Zhang C, Deng Y, Yang F, Zhang T and Zhang C: Methylation of tumor suppressor gene CDH13 and SHP1 promoters and their epigenetic regulation by the UHRF1/PRMT5 complex in endometrial carcinoma. Gynecol Oncol. 140:145–151. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Chen YC, Tsao CM, Kuo CC, Yu MH, Lin YW, Yang CY, Li HJ, Yan MD, Wang TJ, Chou YC and Su HY: Quantitative DNA methylation analysis of selected genes in endometrial carcinogenesis. Taiwan J Obstet Gynecol. 54:572–579. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Huang RL, Su PH, Liao YP, Wu TI, Hsu YT, Lin WY, Wang HC, Weng YC, Ou YC, Huang TH and Lai HC: Integrated epigenomics analysis reveals a DNA methylation panel for endometrial cancer detection using cervical scrapings. Clin Cancer Res. 23:263–272. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Chen H, Zhang C, Sheng Y, Yao S, Liu Z, Zhang C and Zhang T: Frequent SOCS3 and 3OST2 promoter methylation and their epigenetic regulation in endometrial carcinoma. Am J Cancer Res. 5:180–190. 2015.PubMed/NCBI

32 

Guo Y, Peng Y, Gao D, Zhang M, Yang W, Linghu E, Herman JG, Fuks F, Dong G and Guo M: Silencing HOXD10 by promoter region hypermethylation activates ERK signaling in hepatocellular carcinoma. Clin Epigenetics. 9:1162017. View Article : Google Scholar : PubMed/NCBI

33 

Yuan YH, Wang HY, Lai Y, Zhong W, Liang WL, Yan FD, Yu Z, Chen JK and Lin Y: Epigenetic inactivation of HOXD10 is associated with human colon cancer via inhibiting the RHOC/AKT/MAPK signaling pathway. Cell Commun Signal. 17:92019. View Article : Google Scholar : PubMed/NCBI

34 

Wang LH, Choi YL, Hua XY, Shin YK, Song YJ, Youn SJ, Yun HY, Park SM, Kim WJ, Kim HJ, et al: Increased expression of sonic hedgehog and altered methylation of its promoter region in gastric cancer and its related lesions. Mod Pathol. 19:675–683. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Brinkhuizen T, van den Hurk K, Winnepenninckx VJ, de Hoon JP, van Marion AM, Veeck J, van Engeland M and van Steensel MA: Epigenetic changes in basal cell carcinoma affect SHH and WNT signaling components. PLoS One. 7:e517102012. View Article : Google Scholar : PubMed/NCBI

36 

Fan Y, Wang Y, Fu S, Liu D and Lin S: Methylation-regulated ZNF545 inhibits growth of the p53-mutant KYSE150 cell line by inducing p21 and Bax. Exp Ther Med. 18:1563–1570. 2019.PubMed/NCBI

37 

Xiang S, Xiang T, Xiao Q, Li Y, Shao B and Luo T: Zinc-finger protein 545 is inactivated due to promoter methylation and functions as a tumor suppressor through the Wnt/β-catenin, PI3K/AKT and MAPK/ERK signaling pathways in colorectal cancer. Int J Oncol. 51:801–811. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Lin YL, Wang YP, Li HZ and Zhang X: Aberrant promoter methylation of PCDH17 (Protocadherin 17) in serum and its clinical significance in renal cell carcinoma. Med Sci Monit. 23:3318–3323. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Uyen TN, Sakashita K, Al-Kzayer LF, Nakazawa Y, Kurata T and Koike K: Aberrant methylation of protocadherin 17 and its prognostic value in pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer. 64:2017. View Article : Google Scholar : PubMed/NCBI

40 

Musialik E, Bujko M, Kober P, Grygorowicz MA, Libura M, Przestrzelska M, Juszczyński P, Borg K, Florek I, Jakóbczyk M and Siedlecki JA: Promoter DNA methylation and expression levels of HOXA4, HOXA5 and MEIS1 in acute myeloid leukemia. Mol Med Rep. 11:3948–3954. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Kurman RJ, Carcangiu ML, Herrington CS and Young RH: WHO classification of tumours of female reproductive organs. International Agency for Research on Cancer. 307:2014.

42 

Akhavan-Niaki H and Samadani AA: DNA methylation and cancer development: Molecular mechanism. Cell Biochem Biophys. 67:501–513. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Ding WJ, Yang Y, Chen ZX, Wang YY, Dong WL, Cen JN, Qi XF, Jiang F and Chen SN: Methylation level of Rap1GAP and the clinical significance in MDS. Oncol Lett. 16:7287–7294. 2018.PubMed/NCBI

44 

Kim DS, Lee WK and Park JY: Promoter methylation of Wrap53α, an antisense transcript of p53, is associated with the poor prognosis of patients with non-small cell lung cancer. Oncol Lett. 16:5823–5828. 2018.PubMed/NCBI

45 

Jones A, Teschendorff AE, Li Q, Hayward JD, Kannan A, Mould T, West J, Zikan M, Cibula D, Fiegl H, et al: Role of DNA methylation and epigenetic silencing of HAND2 in endometrial cancer development. PLoS Med. 10:e10015512013. View Article : Google Scholar : PubMed/NCBI

46 

Meršaková S, Holubeková V, Grendár M, Višňovský J, Ňachajová M, Kalman M, Kúdela E, Žúbor P, Bielik T, Lasabová Z and Danko J: Methylation of CADM1 and MAL together with HPV status in cytological cervical specimens serves an important role in the progression of cervical intraepithelial neoplasia. Oncol Lett. 16:7166–7174. 2018.PubMed/NCBI

47 

Abbasi AA: Diversification of four human HOX gene clusters by step-wise evolution rather than ancient whole-genome duplications. Dev Genes Evol. 225:353–357. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Seifert A, Werheid DF, Knapp SM and Tobiasch E: Role of Hox genes in stem cell differentiation. World J Stem Cells. 7:583–595. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Bhatlekar S, Fields JZ and Boman BM: HOX genes and their role in the development of human cancers. J Mol Med (Berl). 92:811–823. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Javed S and Langley SE: Importance of HOX genes in normal prostate gland formation, prostate cancer development and its early detection. BJU Int. 113:535–540. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Chekmareva M, Ellenson LH and Pirog EC: Immunohistochemical differences between mucinous and microglandular adenocarcinomas of the endometrium and benign endocervical epithelium. Int J Gynecol Pathol. 27:547–554. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Vardhini NV, Rao PJ, Murthy PB and Sudhakar G: HOXD10 expression in human breast cancer. Tumour Biol. 35:10855–10860. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Wang L, Chen S, Xue M, Zhong J, Wang X, Gan L, Lam EK, Liu X, Zhang J, Zhou T, et al: Homeobox D10 gene, a candidate tumor suppressor, is downregulated through promoter hypermethylation and associated with gastric carcinogenesis. Mol Med. 18:389–400. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Yang H, Zhou J, Mi J, Ma K, Fan Y, Ning J, Wang C, Wei X, Zhao H and Li E: HOXD10 acts as a tumor-suppressive factor via inhibition of the RHOC/AKT/MAPK pathway in human cholangiocellular carcinoma. Oncol Rep. 34:1681–1691. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Joo MK, Park JJ and Chun HJ: Impact of homeobox genes in gastrointestinal cancer. World J Gastroenterol. 22:8247–8256. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Myers C, Charboneau A, Cheung I, Hanks D and Boudreau N: Sustained expression of homeobox d10 inhibits angiogenesis. Am J Pathol. 161:2099–2109. 2002. View Article : Google Scholar : PubMed/NCBI

57 

Wang Y, Li Z, Zhao X, Zuo X and Peng Z: miR-10b promotes invasion by targeting HOXD10 in colorectal cancer. Oncol Lett. 12:488–494. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Carrio M, Arderiu G, Myers C and Boudreau NJ: Homeobox D10 induces phenotypic reversion of breast tumor cells in a three-dimensional culture model. Cancer Res. 65:7177–7185. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Cao YM, Gu J, Zhang YS, Wei WJ, Qu N, Wen D, Liao T, Shi RL, Zhang L, Ji QH, et al: Aberrant hypermethylation of the HOXD10 gene in papillary thyroid cancer with BRAFV600E mutation. Oncol Rep. 39:338–348. 2018.PubMed/NCBI

60 

Ma L, Teruya-Feldstein J and Weinberg RA: Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 449:682–688. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Li S, Zhang J, Zhao Y, Wang F, Chen Y and Fei X: miR-224 enhances invasion and metastasis by targeting HOXD10 in non-small cell lung cancer cells. Oncol Lett. 15:7069–7075. 2018.PubMed/NCBI

62 

Yachi K, Tsuda M, Kohsaka S, Wang L, Oda Y, Tanikawa S, Ohba Y and Tanaka S: miR-23a promotes invasion of glioblastoma via HOXD10-regulated glial-mesenchymal transition. Signal Transduct Target Ther. 3:332018. View Article : Google Scholar : PubMed/NCBI

63 

Zeng Z, Liu S, Cai J, Li Z, Wu H, Chen H and Huang Y: miR-501 promotes hemangioma progression by targeting HOXD10. Am J Transl Res. 11:2439–2446. 2019.PubMed/NCBI

64 

Li C, Huo B, Wang Y and Cheng C: Downregulation of microRNA-92b-3p suppresses proliferation, migration, and invasion of gastric cancer SGC-7901 cells by targeting Homeobox D10. J Cell Biochem. 120:17405–17412. 2019. View Article : Google Scholar : PubMed/NCBI

65 

An N, Luo X, Zhang M and Yu R: MicroRNA-376b promotes breast cancer metastasis by targeting Hoxd10 directly. Exp Ther Med. 13:79–84. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2020
Volume 19 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang F, Liu D, Deng Y, Wang J, Mei S, Ge S, Li H, Zhang C and Zhang T: Frequent promoter methylation of HOXD10 in endometrial carcinoma and its pathological significance. Oncol Lett 19: 3602-3608, 2020
APA
Yang, F., Liu, D., Deng, Y., Wang, J., Mei, S., Ge, S. ... Zhang, T. (2020). Frequent promoter methylation of HOXD10 in endometrial carcinoma and its pathological significance. Oncology Letters, 19, 3602-3608. https://doi.org/10.3892/ol.2020.11467
MLA
Yang, F., Liu, D., Deng, Y., Wang, J., Mei, S., Ge, S., Li, H., Zhang, C., Zhang, T."Frequent promoter methylation of HOXD10 in endometrial carcinoma and its pathological significance". Oncology Letters 19.5 (2020): 3602-3608.
Chicago
Yang, F., Liu, D., Deng, Y., Wang, J., Mei, S., Ge, S., Li, H., Zhang, C., Zhang, T."Frequent promoter methylation of HOXD10 in endometrial carcinoma and its pathological significance". Oncology Letters 19, no. 5 (2020): 3602-3608. https://doi.org/10.3892/ol.2020.11467