Open Access

Increased expression of EHMT2 associated with H3K9me2 level contributes to the poor prognosis of gastric cancer

  • Authors:
    • Ping Chen
    • Qi Qian
    • Zhiyuan Zhu
    • Xiaohui Shen
    • Shenling Yu
    • Zhenghong Yu
    • Rui Sun
    • Yiping Li
    • Didi Guo
    • Hong Fan
  • View Affiliations

  • Published online on: June 5, 2020     https://doi.org/10.3892/ol.2020.11694
  • Pages: 1734-1742
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Di‑methylated lysine 9 of histone H3 (H3K9me2), regulated by histone methyltransferases, is involved in the epigenetic regulation of tumor‑associated genes. The present study aimed to evaluate whether the H3K9me2 methylation level is associated with the expression level of euchromatic histone lysine methyltransferase 2 (EHMT2) in the prognosis of gastric cancer (GC). H3K9me2 methylation level and EHMT2 expression level were detected by immunohistochemistry in 118 GC samples. The clinicopathological significance of H3K9me2 and EHMT2 in patients with GC was assessed using a paired Student's t‑test, χ2 test, Kaplan‑Meier analysis with a log‑rank test and Cox's proportional hazard analysis. Strong positive immunostaining of H3K9me2 and EHMT2 was observed in cancerous tissues compared with adjacent non‑cancerous tissues. Positive immunostaining of EHMT2 and H3K9me2 was associated with lymph node metastasis, pathological grade and tumor‑node‑metastasis stage. H3K9me2 expression level was increased in tumor tissue and associated with worse specific‑disease and disease‑free survival time. In addition, EHMT2 protein expression levels were associated with the expression levels of H3K9me2. Low expression levels of H3K9me2 and EHMT2 predicted a better prognosis of patients with GC. The survival time of patients with a high expression of H3K9me2 and/or EHMT2 was significantly shorter compared with that of the patients with a low expression of H3K9me2 and/or EHMT2. In conclusion, an overexpression pattern of H3K9me2 and/or EHMT2 may be associated with clinicopathological features of GC and may be predictor markers of progression and prognosis in patients with GC, in addition to putative therapeutic targets.

Introduction

Gastric cancer (GC) is one of the most severe tumor types with a high mortality rate (1,2) and poor prognosis (3,4). The global pattern of histone modifications may serve as a predictor of the risk of recurrence of human cancer (5,6). Histone modification, as a notable component of epigenetics, occurs in a diverse range of biological processes. Aberrant post-translational modification of histone tails by methylation is closely associated with tumor development, progression, prognosis and recurrence (7). For example, di-methylation of lysine 9 of histone H3 (H3K9me2) is correlated with gene repression and serves a well-established function in heterochromatin formation and gene transcription regulation in human cancer (8). Among well-studied histone methylations, the methylation pattern of H3K9 is associated with gene regulation including repression (9). Euchromatic histone lysine methyltransferase 2 (EHMT2; also known as G9a), which is a lysine methyltransferase that contributes to the epigenetic silencing of tumor suppressor genes, is required for H3K9me2 (10). EHMT2 may catalyze a modification at histone 3 lysine 9 including H3K9me1 and H3K9me2; H3K9me1 is associated with gene activation, whereas H3K9me2 is predominant in silenced genes (11). EHMT2-dependent H3K9me2 is associated with gene silencing and functions primarily through the recruitment of H3K9me2-binding proteins that prevent transcriptional activation (12). EHMT2 has been reported to be overexpressed in pancreatic (13), breast (14,15), lung (16,17), hepatocellular (18), colorectal carcinoma (19) and GC (20).

The abnormal expression level of EHMT2 and H3K9me2 has been identified in multiple types of cancer, including hematologic malignancies (21). However, the clinical significance of EHMT2, H3K9me2 and their interactions in solid tumor types, including in GC, remains unclear. A previous study has revealed that H3K9me2 may contribute to DNA methylation via DNA (cytosine-5-) methyltransferase 3 αb to repress E-cadherin in the epithelial-mesenchymal transition-associated metastasis of GC (22). Additionally, the hypoxic silencing of tumor suppressor Runt-related transcription factor 3 may also be mediated by upregulated EHMT2 and histone deacetylase 1 in GC cells (20). Increased EHMT2 levels in GC tissues may also promote tumor invasion and metastasis, and are associated with an with advanced stage and shorter overall survival time in a SET domain-independent manner (23). Previously, accumulating evidence has indicated that investigation into the clinical importance of EHMT2 levels and H3K9me2 methylation patterns may be of help for the diagnosis and treatment of GC (2426).

The aim of the present study was to evaluate the methylation pattern of H3K9me2 and EHMT2 expression levels in GC and adjacent healthy tissues, and to reveal the association between the increased EHMT2 expression and H3K9me2 methylation levels.

Materials and methods

Clinical cases with GC

A total of 118 archived paraffin-embedded GC specimen blocks were selected retrospectively from the Department of Pathology of Yancheng Hospital (Jiangsu, China). The specimens were collected from patients (82 men and 36 women) with GC who underwent surgery between March 2010 and December 2011. Medical records, including clinicopathological parameters and follow-up data, were also obtained. The inclusion criteria were as follows: i) No other serious or fatal diseases and ii) if the patients died during the follow-up period, the cause of mortality should be the secondary change, including tumor progression, cachexia, recurrence and metastasis, and follow-up data were complete. The American Joint Committee on Cancer staging system (8th edition) (27) was used for pathological tumor-node-metastasis (pTNM) staging. The mean and median follow-up period was 38.1 months and 41.0 months (range, 1–60 months), respectively. The present study was ethically approved and supervised by the Committee for Ethical Review of Research of Yancheng Hospital.

Immunohistochemical staining

The selected paraffin blocks were sliced into 4-µm tissue sections, heated for 60 min at 65°C and cooled down. The sections were subsequently deparaffinized, rehydrated and placed with ethylene diamine tetraacetic acid antigen retrieval solution (pH 8.0) to retrieve the antigens, followed by incubation with 3% H2O2 for 25 min at room temperature (RT). Following blocking with 3% BSA for 30 min at RT, the sections were treated with primary antibodies against H3K9me2 (1:200; cat. no. ab1220; Abcam) and EHMT2 (1:800; cat. no. ab185050; Abcam) in a wet box at 4°C overnight. The sections were further incubated with horseradish peroxidase-conjugated secondary antibodies (cat. no. K5007; Dako; Agilent Technologies GmbH) for 50 min at RT. The staining was visualized using freshly prepared 3,3′-diaminobenzidine reagent (cat. no. G1211; Wuhan Servicebio Technology Co., Ltd.). The chromogenic reaction was stopped when the nuclei appeared brown/yellow under the microscope; subsequently, the nuclei were counterstained with hematoxylin staining solution at RT. Following dehydration and washing, the slides were mounted with coverslips, and the staining was evaluated under an Axiocam 105 light microscope (magnification, ×100 and ×400; Carl Zeiss AG). Cells treated with PBS instead of the primary antibody were used as a negative control.

Evaluation of immunostained epithelial tissues

The staining was blindly examined by two associate professors from the Department of Pathology, Medical School, Southeast University (Nanjing, China). The analyzed areas were tumor cells in the cancerous tissues and epithelial cells in the adjacent healthy epithelial tissues. The scoring system was described in previous studies (28,29). In brief, the final score of immunostaining was calculated as the sum product of the scale of staining intensity and the scale of staining area. The scale of intensity was as follows: 0=negative; 1=weak staining; 2=moderate staining and 3=intensive staining; and the scale of staining area was as follows: 0=0-5% positive cells; 1=6-25% positive cells; 2=26-50% positive cells; 3=51-75% positive cells and 4=76-100% positive cells. The expression patterns were classified into two groups: Low (score >8) and high (score >8) scoring group. The patients were divided into three groups according to H3K9me2 and EHMT2 expression: i) Low expression group for patients with low expression levels of H3K9me2 and EHMT2; ii) high expression group for patients with high expression levels of H3K9me2 and EHMT2 and iii) other group for patients with high H3K9me2 expression and low EHMT2 expression, and vice versa.

Statistical analysis

Statistical analysis was performed using SPSS software (v.19; IBM Corp.). Data are presented as the mean ± standard error of the mean. The histone modification levels of cancerous and adjacent healthy tissues were assessed using a paired Student's t-test. The associations between clinicopathological variables and the immunostaining scores of histone methylation were analyzed using a χ2 test or Fischer's exact test. Kaplan-Meier (K-M) analysis with a log-rank test was used to determine the contribution of the clinicopathological features and the immunostaining expression patterns to the patients' survival time. Multivariate analysis using Cox's proportional hazard regression was used to examine the clinical value of the levels of the studied protein and the clinicopathological parameters of the patients. Statistical analyses were performed using SPSS Statistics v.17 (SPSS Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Clinicopathological profiles of the patients

In the present study, the tissue samples from 82 male and 36 female patients with GC were studied; the median age was 59.7 years (age range, 33–83 years), and the clinicopathological variables are summarized in Table I. The samples comprised 16 well-differentiated cases, 62 poorly differentiated cases and 40 moderately differentiated cases. The pTNM staging was as follows: 30 cases were stage I (25.4%), 33 were stage II (28%), 47 were stage III (39.8%) and 8 were IV (6.8%). Among the 118 patients, the 5-year survival rate following gastrectomy was 36.4%.

Table I.

Clinicopathological characteristics of patients with gastric carcinoma (n=118).

Table I.

Clinicopathological characteristics of patients with gastric carcinoma (n=118).

CharacteristicsPatient, nPercentage, %
Sex
  Male8269.5
  Female3630.5
Age, years
  ≤605950.0
  >605950.0
Differentiation degree
  Well-differentiated1613.6
  Moderately differentiated4033.9
  Poorly differentiated6252.5
Lymph node metastasis
  Negative5143.2
  Positive6756.8
Distal metastasis
  Negative11093.2
  Positive86.8
pTNM
  I3025.4
  II3328.0
  III4739.8
  IV86.8
Survival time, months following operation
  ≤57563.6
  >54336.4

[i] pTNM, pathological tumor-node-metastasis.

H3K9me2 and EHMT2 expression patterns are associated with clinicopathological characteristics in patients with GC

The expression of histone methylation marker H3K9me2 and histone methyltransferase EHMT2 were evaluated in the surgical samples from 118 patients with GC. H3K9me2 was localized in the nuclei of epithelial cells, whereas EHMT2 mainly labelled the nucleus with partial staining in the cytoplasm (Fig. 1). Example tissues exhibiting low and high expression areas are presented in Fig. 2. The assessment of the positive immunoreaction in the epithelial cells of cancerous and non-cancerous tissues demonstrated that cancerous tissues exhibited significantly stronger immunostaining compared with adjacent healthy tissues (P<0.001; Table II).

Table II.

Differential expression of histone methylation between gastric cancer and peri-cancer tissues.

Table II.

Differential expression of histone methylation between gastric cancer and peri-cancer tissues.

Histone methylationCancerPeri-cancerP-value
H3K9me2   7.557±0.206   7.311±0.236 <0.001a
EHMT26.765±0.2164.319±0.228 <0.001a

a P<0.001. H3K9me2, di-methylated lysine 9 of histone H3; EHMT2, euchromatic histone-lysine N-methyltransferase 2.

To clarify the association between patient clinicopathological characteristics, EHMT2 expression and H3K9me2 methylation, the data were analyzed using a χ2 test; the results demonstrated that EHMT2 overexpression and H3K9me2 methylation levels were significantly associated with the degree of differentiation (P=0.025 and P=0.031, respectively), lymph node involvement (P=0.021 and P=0.021, respectively) and pTNM stage (P=0.036 and P=0.022, respectively), but not sex, age or distal metastasis (Table III; Fig. 3). The results of distal metastasis may have been affected by the low ratio of positive cases (6.8%) in the studied samples.

Table III.

Association between H3K9me2 or EHMT2 expression patterns and clinicopathological characteristics of patients with gastric carcinoma (n=118).

Table III.

Association between H3K9me2 or EHMT2 expression patterns and clinicopathological characteristics of patients with gastric carcinoma (n=118).

H3K9me2 expressionEHMT2 expression


CharacteristicsLowHighχ2P-valueLowHighχ2P-value
Sex 0.7980.372 0.0310.861
  Male5725 5131
  Female2214 2313
Age, years 0.9570.328 0.1450.703
  ≤603722 3821
  >604217 3623
Differentiation degree 4.6610.031a 5.0270.025a
  Well + moderately differentiated4313 4115
  Poorly differentiated3626 3329
Lymph node metastasis 5.3520.021a 5.3460.021a
#x00A0; Negative4011 3813
#x00A0; Positive3928 3631
Distal metastasis 1.1140.291 0.5540.457
#x00A0; Negative7535 6842
#x00A0; Positive44 62
pTNM 5.2170.022a 4.3920.036a
#x00A0; I+II4815 4518
#x00A0; III+IV3124 2926

a P<0.05. H3K9me2, di-methylated lysine 9 of histone H3; EHMT2, euchromatic histone-lysine N-methyltransferase 2; pTNM, pathological tumor-node-metastasis.

EHMT2 overexpression was significantly associated with H3K9me2 methylation level (P=0.040; data not shown). The results indicated that EHMT2 and H3K9me2 expression patterns exhibited notable consistency. As presented in Fig. 3, H3K9me2 exhibited similar ratios of cases of each stage of pTNM to EHMT2 when analyzed based on the expression level, which further confirmed their positive association.

Effect of H3K9me2 and EHMT2 on the survival of patients with GC

To understand the impact of H3K9me2 and EHMT2 expression patterns on the survival of patients with GC, overall survival time based on H3K9me2 and EHMT expression levels was further analyzed by K-M analysis with a log-rank test. Patients with low expression levels of H3K9me2 exhibited a significantly longer survival time compared with those in the high expression group (P<0.05). Overexpression of EHMT2 presented the same trend (P<0.05; Fig. 4; Table S1). Mean progression-free survival time of patients in the low and high H3K9me2 expression groups were 45.521±2.195 and 34.061±3.861 months, respectively, and in the low and high EHMT2 expression level groups were 41.761±2.399 and 32.243±3.364 months, respectively (Table S1). The median survival period exhibited similar tendencies. Univariate K-M survival curves demonstrated that tumor differentiation degree, lymph node involvement, distal metastasis and pTNM stage were significantly associated with a shorter survival time of patients with GC (P<0.001; Table S2). However, the sex and age of patients exhibited no association with patient survival time in the present study.

Combination of EHMT2 overexpression and H3K9me2 level is an effective prognostic marker for patients with GC

To identify the independent risk factors for the prognosis of patients with GC, multivariate Cox's regression analysis was conducted to evaluate the clinicopathological features of patients with GC and the expression levels of H3K9me2 and EHMT2. The results demonstrated that the expression patterns of H3K9me2 and EHMT2, differentiation degree, lymph node involvement, distal metastasis and pTNM stage exhibited independent significant prognostic effects on patient survival time (P<0.05; Table IV). To present the significant factors more accurately, the patients were regrouped by combining the expression patterns of H3K9me2 and EHMT2. The expression patterns were classified into two groups: Low (score ≤8) and high (score >8) scoring groups. The patients were allocated into three groups: i) Low expression group for patients with low expression levels of H3K9me2 and EHMT2; ii) high expression group for patients with high expression levels of H3K9me2 and EHMT2 and iii) other group for patients with high H3K9me2 expression and low EHMT2 expression, and vice versa. The survival analysis revealed that the high expression group exhibited the shortest survival time (31.467±5.671 months), the low expression group exhibited the longest survival period (46.000±2.711 months) and the other group exhibited an intermediate survival duration (37.943±2.830 months; Table S1). Therefore, the combination of H3K9me2 and EHMT2 expression patterns may be used as a more accurate indicator for the overall survival time of patients with GC compared with either H3K9me2 or EHMT2 alone (P=0.036; Table S1). In addition, multivariate Cox analysis revealed that the combined expression patterns of H3K9me2 and EHMT2, differentiation degree, lymph node involvement, distal metastasis and pTNM stage significantly predicted the survival time of patients with GC (P<0.05; Table IV).

Table IV.

Multivariate Cox analysis of overall survival based on individual and combined groups of the histone signatures.

Table IV.

Multivariate Cox analysis of overall survival based on individual and combined groups of the histone signatures.

A, Individual groups of histone signatures

CharacteristicsHR95% CIP-value
Sex1.0880.633–1.8680.939
Age0.7730.462–1.2930.539
Differentiation degree0.5140.299–0.8820.016a
Lymph node metastasis0.2300.113–0.467 <0.001c
Distal metastasis0.4020.185–0.8720.021a
pTNM0.1100.044–0.271 <0.001c
H3K9me21.0500.637–1.7290.042a
EHMT21.0040.599–1.6830.045a

B, Combined groups of histone signatures

CharacteristicsHR95% CIP-value

Differentiation degree0.5310.313–0.9010.019a
Lymph node metastasis0.2340.116–0.474 <0.001c
Distal metastasis0.3880.183–0.8220.013a
pTNM0.1060.043–0.258 <0.001c
Low expression group0.009b
High expression group1.1780.709–1.9570.018a
Other groups1.4340.715–2.8760.020a

a P<0.05

b P<0.01

c P<0.001. HR, hazard ratio; CI, confidence interval; pTNM, pathological tumor-node-metastasis; H3K9me2, di-methylated lysine 9 of histone H3; EHMT2, euchromatic histone-lysine N-methyltransferase 2.

Discussion

Alterations of epigenetic regulation genes, including DNA methylation, histone modifications, chromatin remodeling and non-coding RNA regulation, have been detected in early carcinogenesis and cancer progression (3034). A number of them have been proposed as biomarkers for cancer detection and tumor prognosis (3537). A number of diverse factors, including DNA damage, DNA and histone methylation, in addition to environmental influence, are associated with GC-associated mortality in China (17). Previously, increasing evidence has revealed that aberrant epigenetic regulation serves an important function during tumorigenesis. Histone methylations and their corresponding catalytic enzymes were the focus of a previous study; among various best-studied histone methylations, H3K9 methylation is associated with gene repression (9). Epigenetic alterations of tumor genes are present during carcinogenesis and may provide novel biomarkers for diagnosis (3840).

EHMT2 was first identified as a gene located in the major histocompatibility complex locus in mice and human leukocyte antigen locus in humans (41). Previous studies have indicated that EHMT2 serves an important function during the carcinogenesis and development of a tumor (13,14,17,42,43). EHMT2 is also a crucial factor in a variety of biological progresses, including behavior plasticity, lymphocyte development, stem cell differentiation and tumor cell growth (44). The clinical importance of EHMT2 expression in numerous cancer tissues has been studied (16); however, the expression pattern of EHMT2 and its significance in GC is largely unclear. Although the crucial functions of H3K9me2 and EHMT2 have been elucidated in several tumor types (45,46), the associations between H3K9me2 methylation pattern and EHMT2 expression level in GC are unknown. EHMT2 is significantly upregulated in numerous different tumor types compared with matched normal controls, and knocking down EHMT2 or the pharmacological inhibition of its activity suppresses tumor cell growth and invasion, indicating that EHMT2 may be an oncogenic and metastatic factor (47,48). In addition, high expression levels of EHMT2 are correlated with a poor overall survival in patients with lung adenocarcinoma (16).

EHMT2 is a main histone lysine methylation enzyme which catalyzes the modification at histone 3 lysine 9, including H3K9me1 and H3K9me2. The patterns of H3K9me1 or H3K9me2 are different during development, as there are either more mono- or dimethylations at H3K9 during the maturation of the auditory system (49). During the development of the zebrafish retina, EHMT2 expression and H3K9me2 markers have been noted to be closely associated (50).

H3K9 methylation is a crucial event in reprogramming to pluripotency (51). Numerous studies have demonstrated that the global level of H3K9me2 is associated with the prognosis of prostate and kidney cancer (52,53). These results indicate that there is an association between EHMT2 expression and H3K9me2 markers.

The focus of the present study was the function of EHMT2 and H3K9me2 in the processes associated with a poor prognosis of GC. H3K9me2 and EHMT2 expression exhibited strong immunostaining in GC tumor tissues compared with adjacent healthy tissues. There was an association between the levels of H3K9me2 and EHMT2. The results also demonstrated that EHMT2 expression was associated with the differentiation degree and lymph node metastasis. These results were consistent with a previous study (23), which demonstrated that increased EHMT2 expression in GC tissues correlated with an advanced stage and promoted tumor invasion and metastasis. Depletion of EHMT2 may be of therapeutic value by inhibiting cell proliferation and inducing apoptosis in GC (54). High expression levels of H3K9me2 or EHMT2 were significantly associated with a worse overall survival time in patients with GC. Patients with combined lower levels of H3K9me2 and EHMT2 exhibited a better survival rate and prognosis compared with those with combined higher levels of H3K9me2 and EHMT2, in addition to the other group. Furthermore, the results of the present study suggested that the development of GC is associated with pathological grade, lymph node metastasis and TNM stage.

In conclusion, EHMT2 expression level and H3K9me2 methylation level may be associated with the development risk and prognosis of GC. Patients with increased EHMT2 and H3K9me2 levels exhibited worse overall survival and a poorer prognosis compared with other patients. Overexpression of EHMT2 and H3K9me2 levels may be predictor markers of progression and prognosis in patients with GC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant nos. 81672414 and 81472548).

Availability of data and materials

The analyzed data sets generated during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

PC conceived the study and was a major contributor in writing the manuscript. QQ and ZZ contributed to the analysis and interpretation of the data. XS and SY were involved in drafting the manuscript and the re-analysis of the immunohistochemistry scores. ZY acquired the data and performed statistical analysis. RS also acquired the data. YL performed the immunostaining. DG made contributions to the experimental studies. HF designed the research protocols and gave final approval of the version to be published. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was reviewed and ethically approved by the Committee for Ethical Review of Research of Yancheng Hospital (Yancheng, China), and written informed consent was obtained from the patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Norollahi SE, Alipour M, Rashidy-Pour A, Samadani AA and Larijani LV: Regulatory fluctuation of WNT16 gene expression is associated with human gastric adenocarcinoma. J Gastrointest Cancer. 50:42–47. 2017. View Article : Google Scholar

3 

Nashimoto A, Akazawa K, Isobe Y, Miyashiro I, Katai H, Kodera Y, Tsujitani S, Seto Y, Furukawa H, Oda I, et al: Gastric cancer treated in 2002 in Japan: 2009 annual report of the JGCA nationwide registry. Gastric Cancer. 16:1–27. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Theuer CP, Kurosaki T, Ziogas A, Butler J and Anton-Culver H: Asian patients with gastric carcinoma in the United States exhibit unique clinical features and superior overall and cancer specific survival rates. Cancer. 89:1883–1892. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J, Schotta G, Bonaldi T, Haydon C, Ropero S, Petrie K, et al: Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet. 37:391–400. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, et al: The genomic landscapes of human breast and colorectal cancers. Science. 318:1108–1113. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Benard A, Goossens-Beumer IJ, van Hoesel AQ, de Graaf W, Horati H, Putter H, Zeestraten EC, van de Velde CJ and Kuppen PJ: Histone trimethylation at H3K4, H3K9 and H4K20 correlates with patient survival and tumor recurrence in early-stage colon cancer. BMC Cancer. 14:5312014. View Article : Google Scholar : PubMed/NCBI

8 

Tachibana M, Ueda J, Fukuda M, Takeda N, Ohta T, Iwanari H, Sakihama T, Kodama T, Hamakubo T and Shinkai Y: Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9. Genes Dev. 19:815–826. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Dawson MA and Kouzarides T: Cancer epigenetics: From mechanism to therapy. Cell. 150:12–27. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, Fukuda M, Takeda N, Niida H, Kato H, et al: G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 16:1779–1791. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I and Zhao K: High-resolution profiling of histone methylations in the human genome. Cell. 129:823–837. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Scheer S and Zaph C: The Lysine Methyltransferase G9a in Immune Cell Differentiation and Function. Front Immunol. 8:4292017. View Article : Google Scholar : PubMed/NCBI

13 

Tian YF, Wang HC, Luo CW, Hung WC, Lin YH, Chen TY, Li CF, Lin CY and Pan MR: Preprogramming therapeutic response of PI3K/mTOR dual inhibitor via the regulation of EHMT2 and p27 in pancreatic cancer. Am J Cancer Res. 8:1812–1822. 2018.PubMed/NCBI

14 

Casciello F, Al-Ejeh F, Kelly G, Brennan DJ, Ngiow SF, Young A, Stoll T, Windloch K, Hill MM, Smyth MJ, Gannon F and Lee JS: G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis. Proc Natl Acad Sci U S A. 114:7077–7082. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Kim K, Son MY, Jung CR, Kim DS and Cho HS: EHMT2 is a metastasis regulator in breast cancer. Biochem Biophys Res Commun. 496:758–762. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Huang T, Zhang P, Li W, Zhao T, Zhang Z, Chen S, Yang Y, Feng Y, Li F, Shirley Liu X, Zhang L, Jiang G and Zhang F: G9A promotes tumor cell growth and invasion by silencing CASP1 in non-small-cell lung cancer cells. Cell Death Dis. 8:e27262017. View Article : Google Scholar : PubMed/NCBI

17 

Wang L, Dong X, Ren Y, Luo J, Liu P, Su D and Yang X: Targeting EHMT2 reverses EGFR-TKI resistance in NSCLC by epigenetically regulating the PTEN/AKT signaling pathway. Cell Death Dis. 9:1292018. View Article : Google Scholar : PubMed/NCBI

18 

Qin J, Li Q, Zeng Z, Wu P, Jiang Y, Luo T, Ji X, Zhang Q, Hao Y and Chen L: Increased expression of G9A contributes to carcinogenesis and indicates poor prognosis in hepatocellular carcinoma. Oncol Lett. 15:9757–9765. 2018.PubMed/NCBI

19 

Qin J, Zeng Z, Luo T, Li Q, Hao Y and Chen L: Clinicopathological significance of G9A expression in colorectal carcinoma. Oncol Lett. 15:8611–8619. 2018.PubMed/NCBI

20 

Lee SH, Kim J, Kim WH and Lee YM: Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene. 28:184–194. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Renneville A, Van Galen P, Canver MC, McConkey M, Krill-Burger JM, Dorfman DM, Holson EB, Bernstein BE, Orkin SH, Bauer DE, et al: EHMT1 and EHMT2 inhibition induces fetal hemoglobin expression. Blood. 126:1930–1939. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Cui H, Hu Y, Guo D, Zhang A, Gu Y, Zhang S, Zhao C, Gong P, Shen X, Li Y, et al: DNA methyltransferase 3A isoform b contributes to repressing E-cadherin through cooperation of DNA methylation and H3K27/H3K9 methylation in EMT-related metastasis of gastric cancer. Oncogene. 37:4358–4371. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Hu L, Zang MD, Wang HX, Zhang BG, Wang ZQ, Fan ZY, Wu H, Li JF, Su LP, Yan M, et al: G9A promotes gastric cancer metastasis by upregulating ITGB3 in a SET domain-independent manner. Cell Death Dis. 9:2782018. View Article : Google Scholar : PubMed/NCBI

24 

Zhang C, Wei S, Hu J and Xiong Z: Upregulated expression of G9a is correlated with poor prognosis of gastric cancer patients. Medicine (Baltimore). 98:e182122019. View Article : Google Scholar : PubMed/NCBI

25 

Li Y, Guo D, Sun R, Chen P, Qian Q and Fan H: Methylation patterns of Lys9 and Lys27 on histone H3 correlate with patient outcome in gastric cancer. Dig Dis Sci. 64:439–446. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Lee KH, Park JW, Sung HS, Choi YJ, Kim WH, Lee HS, Chung HJ, Shin HW, Cho CH, Kim TY, et al: PHF2 histone demethylase acts as a tumor suppressor in association with p53 in cancer. Oncogene. 34:2897–2909. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Ji X, Bu ZD, Yan Y, Li ZY, Wu AW, Zhang LH, Zhang J, Wu XJ, Zong XL, Li SX, Shan F, et al: The 8th edition of the American Joint Committee on Cancer tumor-node-metastasis staging system for gastric cancer is superior to the 7th edition: results from a Chinese mono-institutional study of 1663 patients. Gastric Cancer. 21:643–652. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Axiotis CA, Monteagudo C, Merino MJ, LaPorte N and Neumann RD: Immunohistochemical detection of P-glycoprotein in endometrial adenocarcinoma. Am J Pathol. 138:799–806. 1991.PubMed/NCBI

29 

Fisher KE, Cohen C, Siddiqui MT, Palma JF, Lipford EH III and Longshore JW: Accurate detection of BRAF p.V600E mutations in challenging melanoma specimens requires stringent immunohistochemistry scoring criteria or sensitive molecular assays. Hum Pathol. 45:2281–2293. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Sina AA, Carrascosa LG, Liang Z, Grewal YS, Wardiana A, Shiddiky MJA, Gardiner RA, Samaratunga H, Gandhi MK, Scott RJ, et al: Epigenetically reprogrammed methylation landscape drives the DNA self-assembly and serves as a universal cancer biomarker. Nat Commun. 9:49152018. View Article : Google Scholar : PubMed/NCBI

31 

Noberini R, Osti D, Miccolo C, Richichi C, Lupia M, Corleone G, Hong SP, Colombo P, Pollo B, Fornasari L, et al: Extensive and systematic rewiring of histone post-translational modifications in cancer model systems. Nucleic Acids Res. 46:3817–3832. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Wu Q, Lian JB, Stein JL, Stein GS, Nickerson JA and Imbalzano AN: The BRG1 ATPase of human SWI/SNF chromatin remodeling enzymes as a driver of cancer. Epigenomics. 9:919–931. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Kumar R, Li DQ, Müller S and Knapp S: Epigenomic regulation of oncogenesis by chromatin remodeling. Oncogene. 35:4423–4436. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Ma Y, Yang Y, Wang F, Moyer MP, Wei Q, Zhang P, Yang Z, Liu W, Zhang H, Chen N, et al: Long non-coding RNA CCAL regulates colorectal cancer progression by activating Wnt/β-catenin signalling pathway via suppression of activator protein 2α. Gut. 65:1494–1504. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Tahara T and Arisawa T: DNA methylation as a molecular biomarker in gastric cancer. Epigenomics. 7:475–486. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Lomberk G, Blum Y, Nicolle R, Nair A, Gaonkar KS, Marisa L, Mathison A, Sun Z, Yan H, Elarouci N, et al: Distinct epigenetic landscapes underlie the pathobiology of pancreatic cancer subtypes. Nat Commun. 9:19782018. View Article : Google Scholar : PubMed/NCBI

37 

Tvardovskiy A, Schwämmle V, Kempf SJ, Rogowska-Wrzesinska A and Jensen ON: Accumulation of histone variant H3.3 with age is associated with profound changes in the histone methylation landscape. Nucleic Acids Res. 45:9272–9289. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Okugawa Y, Grady WM and Goel A: Epigenetic alterations in colorectal cancer: Emerging biomarkers. Gastroenterology. 149:1204–1225 e12. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Costa-Pinheiro P, Montezuma D, Henrique R and Jerónimo C: Diagnostic and prognostic epigenetic biomarkers in cancer. Epigenomics. 7:1003–1015. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Gu L, Frommel SC, Oakes CC, Simon R, Grupp K, Gerig CY, Bär D, Robinson MD, Baer C, Weiss M, et al ICGC Project on Early Onset Prostate Cancer, : BAZ2A (TIP5) is involved in epigenetic alterations in prostate cancer and its overexpression predicts disease recurrence. Nat Genet. 47:22–30. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Brown SE, Campbell RD and Sanderson CM: Novel NG36/G9a gene products encoded within the human and mouse MHC class III regions. Mamm Genome. 12:916–924. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Wang YF, Zhang J, Su Y, Shen YY, Jiang DX, Hou YY, Geng MY, Ding J and Chen Y: G9a regulates breast cancer growth by modulating iron homeostasis through the repression of ferroxidase hephaestin. Nat Commun. 8:2742017. View Article : Google Scholar : PubMed/NCBI

43 

Wei L, Chiu DK, Tsang FH, Law CT, Cheng CL, Au SL, Lee JM, Wong CC, Ng IO and Wong CM: Histone methyltransferase G9a promotes liver cancer development by epigenetic silencing of tumor suppressor gene RARRES3. J Hepatol. 67:758–769. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Mayr C, Helm K, Jakab M, Ritter M, Shrestha R, Makaju R, Wagner A, Pichler M, Beyreis M, Staettner S, et al: The histone methyltransferase G9a: A new therapeutic target in biliary tract cancer. Hum Pathol. 72:117–126. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Casciello F, Al-Ejeh F, Kelly G, Brennan DJ, Ngiow SF, Young A, Stoll T, Windloch K, Hill MM, Smyth MJ, et al: G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis. Proc Natl Acad Sci USA. 114:7077–7082. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Salzberg AC, Harris-Becker A, Popova EY, Keasey N, Loughran TP, Claxton DF and Grigoryev SA: Genome-wide mapping of histone H3K9me2 in acute myeloid leukemia reveals large chromosomal domains associated with massive gene silencing and sites of genome instability. PLoS One. 12:e01737232017. View Article : Google Scholar : PubMed/NCBI

47 

Dong C, Wu Y, Yao J, Wang Y, Yu Y, Rychahou PG, Evers BM and Zhou BP: G9a interacts with Snail and is critical for Snail-mediated E-cadherin repression in human breast cancer. J Clin Invest. 122:1469–1486. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Liu S, Ye D, Guo W, Yu W, He Y, Hu J, Wang Y, Zhang L, Liao Y, Song H, et al: G9a is essential for EMT-mediated metastasis and maintenance of cancer stem cell-like characters in head and neck squamous cell carcinoma. Oncotarget. 6:6887–6901. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Ebbers L, Runge K and Nothwang HG: Differential patterns of histone methylase EHMT2 and its catalyzed histone modifications H3K9me1 and H3K9me2 during maturation of central auditory system. Cell Tissue Res. 365:247–264. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Olsen JB, Wong L, Deimling S, Miles A, Guo H, Li Y, Zhang Z, Greenblatt JF, Emili A and Tropepe V: G9a and ZNF644 physically associate to suppress progenitor gene expression during neurogenesis. Stem Cell Reports. 7:454–470. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Sridharan R, Gonzales-Cope M, Chronis C, Bonora G, McKee R, Huang C, Patel S, Lopez D, Mishra N, Pellegrini M, et al: Proteomic and genomic approaches reveal critical functions of H3K9 methylation and heterochromatin protein-1γ in reprogramming to pluripotency. Nat Cell Biol. 15:872–882. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Seligson DB, Horvath S, McBrian MA, Mah V, Yu H, Tze S, Wang Q, Chia D, Goodglick L and Kurdistani SK: Global levels of histone modifications predict prognosis in different cancers. Am J Pathol. 174:1619–1628. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Mosashvilli D, Kahl P, Mertens C, Holzapfel S, Rogenhofer S, Hauser S, Büttner R, Von Ruecker A, Müller SC and Ellinger J: Global histone acetylation levels: Prognostic relevance in patients with renal cell carcinoma. Cancer Sci. 101:2664–2669. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Lin X, Huang Y, Zou Y, Chen X and Ma X: Depletion of G9a gene induces cell apoptosis in human gastric carcinoma. Oncol Rep. 35:3041–3049. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 20 Issue 2

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
Chen P, Qian Q, Zhu Z, Shen X, Yu S, Yu Z, Sun R, Li Y, Guo D, Fan H, Fan H, et al: Increased expression of EHMT2 associated with H3K9me2 level contributes to the poor prognosis of gastric cancer . Oncol Lett 20: 1734-1742, 2020
APA
Chen, P., Qian, Q., Zhu, Z., Shen, X., Yu, S., Yu, Z. ... Fan, H. (2020). Increased expression of EHMT2 associated with H3K9me2 level contributes to the poor prognosis of gastric cancer . Oncology Letters, 20, 1734-1742. https://doi.org/10.3892/ol.2020.11694
MLA
Chen, P., Qian, Q., Zhu, Z., Shen, X., Yu, S., Yu, Z., Sun, R., Li, Y., Guo, D., Fan, H."Increased expression of EHMT2 associated with H3K9me2 level contributes to the poor prognosis of gastric cancer ". Oncology Letters 20.2 (2020): 1734-1742.
Chicago
Chen, P., Qian, Q., Zhu, Z., Shen, X., Yu, S., Yu, Z., Sun, R., Li, Y., Guo, D., Fan, H."Increased expression of EHMT2 associated with H3K9me2 level contributes to the poor prognosis of gastric cancer ". Oncology Letters 20, no. 2 (2020): 1734-1742. https://doi.org/10.3892/ol.2020.11694