Open Access

Clinicopathological significance of G9A expression in colorectal carcinoma

  • Authors:
    • Jian Qin
    • Zhi Zeng
    • Tao Luo
    • Qingyun Li
    • Yarong Hao
    • Lang Chen
  • View Affiliations

  • Published online on: April 10, 2018     https://doi.org/10.3892/ol.2018.8446
  • Pages: 8611-8619
  • Copyright: © Qin 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

G9A, the primary histone methyltransferase (HMTase) for histone H3 lysine 9, is upregulated in numerous types of cancer and is critical for tumor cell proliferation. The present study aimed to investigate the G9A expression level in colorectal carcinoma (CRC) to evaluate the clinical significance of G9A in CRC. First, the present study detected the expression of G9A protein in 100 pairs of CRC specimens by immunohistochemistry staining and analyzed the correlations between G9A expression and pathological tumor features. It was found that G9A expression was increased markedly in CRC tumor specimens and the high expression was associated with tumor distant metastasis. Oncomine database analysis demonstrated an elevated expression level of G9A in various types of CRC. In total, 6 public available data sets from the Gene Expression Omnibus (GEO) were used and Gene set enrichment analysis (GSEA) was conducted. The results of the bioinformatics analysis demonstrated that high G9A expression was associated with American Joint Committee on Cancer staging, tumor differentiation, tumor relapse of CRC, and may serve a role in CRC cell proliferation. These findings suggested that G9A was overexpressed in CRC and involved in the tumorigenesis and distant metastasis of CRC. The expression level may also serve as a potential indicator for tumor recurrence in CRC. The present findings aided in the understanding of the crucial role of G9A in tumorigenesis and also offered novel ideas for CRC therapy.

Introduction

Aberrant epigenetic regulations have been found in various types of cancer and numerous types of human disease. The cancer epigenome is characterized by global changes in DNA methylation and altered histone modification patterns (1). The global pattern of histone modifications may serve as a predictor for the risk of recurrence of human cancers (2,3). It seems that epigenetics have reached the mainstream in pathogenesis research of numerous types of human disease, particularly in cancer. Colorectal cancer (CRC) is the third most commonly diagnosed cancer in the world (4). Cumulative evidence has proved that the disruption of epigenetic regulation may drive the initiation and progression of CRC, such as histone modifications (57). Emerging evidences reveal that altered expression of histone methyltransferases (HMTs) and histone demethylases (HDMs) may be involved in cancer progression of CRC, such as Enhancer of zeste homolog 2 (EZH2) and lysine demethylase 4C (5,8,9). The functions of HMTs and HDMs in the pathogenesis of CRC require further investigation.

G9A is the primary HMT for mono- and dimethylation of H3K9 in vivo (10). Among various best-studied histone methylations, H3K9 methylation is thought to be associated with gene repression (11). Recently, G9A has been reported to perform critical roles in a number of biological progresses, such as behavior plasticity, lymphocyte development, stem cells differentiation and tumor cell growth (1217). It has been found that the expression level of G9A is increased in numerous types of cancer as compared with their corresponding normal tissues, such as melanoma, lung cancer, neuroblastoma, leukemia and hepatocellular carcinoma (HCC) (1719). It has been demonstrated that decreasing G9A expression level or inhibiting its activity reduces cellular proliferation and induces autophagy related cell death in colon cancer cells, breast cancer cells and neuroblastoma cells (17,20,21). A recent study also reported that G9A suppression induces DNA damage in colorectal cancer cells (21).

Although G9A has been reported to be crucial in numerous types of cancer, the function in colorectal cancer progression remains unknown. In the present study, the expression profile of G9A in CRC was examined to explore the function in CRC progression. The immunohistochemistry analysis of 100 pairs of tumor specimens revealed that G9A protein expression was markedly increased in CRC and the high expression may be related to distant metastasis. A significant elevated mRNA expression level of G9A in various colorectal carcinomas compared with normal colon and rectum tissues by Oncomine database analysis was also identified. Furthermore, the present study investigated the association between G9A expression level and the clinicopathological features of CRC using 6 publicly available datasets from Gene Expression Omnibus (GEO), and found that G9A expression was associated with American Joint Committee on Cancer (AJCC) staging, tumor differentiation, and tumor relapse. The present findings provide novel evidence to further understand the crucial role of G9A in tumorigenesis, and also offers significant ideas for CRC therapy.

Materials and methods

Patients

In total, 100 patients were diagnosed with CRC (53 with colon cancer and 47 with rectal cancer; Table I), which was classified with the 7th edition of the International Union against Cancer TNM staging system (classified into T1, T2, T3 and T4 based on the size and the extension of the primary tumor; classified into N0, N1 and N2 based on the degree of spread to regional lymph nodes; classified into M0 and M1 based on the presence of distant metastasis), the Dukes' staging system (classified into A, B, C and D) and histological grading (classified into well, moderately, or poorly differentiated) (2224). All patients underwent surgical resection of tumors at the Renmin Hospital of Wuhan University in Wuhan, China. Ethical approval for the study was granted by the Renmin Hospital's ethics committee. Informed written consent was obtained from all participants involved in the study. The key clinical characteristics of the patients are summarized in Table I. Normal specimens were obtained from adjacent, grossly normal-appearing tissue taken at least 10 cm away from the cancer. None of the patients included in this study had chemotherapy or radiotherapy prior to surgery. There was no follow-up information available for these patients.

Table I.

Association between G9A protein expression and clinicopathological features of CRC.

Table I.

Association between G9A protein expression and clinicopathological features of CRC.

G9A expression, n (%)

Clinicopathological featuresCase size, nLowHigh P-valuea
Diagnosis age (years) 0.028
  ≤604922 (44.90)27 (55.10)
  >605134 (66.67)17 (33.33)
Sex 1.000
  Male5028 (56.00)22 (44.00)
  Female5028 (56.00)22 (44.00)
Size (diameter) 0.349
  <5 cm4724 (51.06)23 (48.94)
  ≥5 cm5332 (60.38)21 (39.62)
Depth of invasion 0.106
  T1138 (61.54)5 (38.46)
  T24224 (57.14)18 (42.86)
  T33522 (62.86)13 (37.14)
  T4102 (20.00)8 (80.00)
Nodal metastasis 0.799
  N05733 (57.89)24 (42.11)
  N12312 (52.17)11 (47.83)
  N2189 (50.00)9 (50.00)
Distant metastasis 0.043
  M08953 (59.55)36 (40.45)
  M1113 (27.27)8 (72.73)
TNM stage 0.106
  I138 (61.54)5 (38.46)
  II4224 (57.14)18 (42.86)0.035b
  III3522 (62.86)13 (37.14)0.020b
  IV102 (20.00)8 (80.00)
Dukes stage 0.209
  A149 (64.29)5 (35.71)
  B4123 (56.10)18 (43.90)
  C3421 (61.76)13 (38.24)
  D113 (27.27)8 (72.73)0.049c
Differentiation 0.631
  Well2013 (65.00)7 (35.00)
  Moderately5730 (52.63)27 (47.37)
  Poorly2313 (56.52)10 (43.48)

a Chi-square test was used to statistically analyze the association between title category and G9A protein expression (all unmarked P-values)

b Compared with TNM stage IV

c Compared with Dukes stage C.

Immunohistochemistry

Immunostaining was performed as described previously (25). Briefly, deparaffinized sections were treated with 3% H2O2 and subjected to antigen retrieval by citric acid (pH 6.0). Subsequent to overnight incubation with primary antibody of G9A (1:100; cat. no. ab133482; Abcam, Cambridge, UK) at 4°C, the sections were incubated for 15 min at room temperature with horseradish peroxidase-labeled polymer conjugated with secondary antibody (MaxVision™ HRP-Polymer anti-Rabbit IHC Kit; Maixin-Bio, Fuzhou, China) and incubated for 1 min with diaminobenzidine. The sections were then lightly counterstained with hematoxylin. The sections without primary antibody served as negative controls. The positive brown staining was visualized and then photographed using a light microscope at ×200 or ×400 magnification (BX51, Olympus Corporation, Tokyo, Japan).

Evaluation of immunohistochemical staining

The immunohistochemical staining results of all sections were evaluated by two independent observers (Dr Zhi Zeng and Dr Jian Qin, the co-authors), who were unaware of the disease outcome. Expression levels were ascertained according to the two observers' evaluations. As G9A is mainly expressed in the cell nucleus, the percentage of nucleus staining-positive cells were graded as 0 (<10%), 1 (≥10%, and <25%), 2 (≥25%, and <50%), 3 (≥50%, and <75%), 4 (≥75%) (Fig. 1A-F). For analysis the G9A protein expression levels were divided into two groups: Low expression level group (score value ≤2) and high expression level group (score value ≥3).

Oncomine database analysis

The Oncomine database (https://www.oncomine.org) (26), which is a cancer microarray database and web-based data-mining platform, was interrogated to validate the expression status of G9A mRNA in various types of CRC. Filter indexes were set in Oncomine based on research interests. Primary filters were set as differential analysis (cancer vs. normal) and cancer type (colorectal cancer). Dataset filters were set as data type (mRNA). Datasets were ordered by overexpression with P-value. Datasets were set by P-value (1E-6) and fold change (1.5+). The datasets were then sorted to analyze the expression of G9A mRNA associated with different cancer types vs. normal tissue.

Publicly available data analysis

The data sets of the patients with CRC and the corresponding clinical data were downloaded from the publicly available Gene Expression Omnibus (GEO) datasets (http://www.ncbi.nlm.nih.gov/gds/; National Center for Biotechnology Information, Bethesda, USA). A total of 6 independent data sets from GSE38832 (27) (n=122), GSE37892 (28) (n=130), GSE28722 (29) (n=125), GSE17536 (30,31) (n=177), GSE18088 (32) (n=53), and GSE33113 (33,34) (n=96) were utilized to analyze the expression level of G9A in CRC. The patients were divided into two groups according to their G9A expression level (top 50%, high vs. bottom 50%, low). For GSE38832, GSE37892, GSE17536, GSE18088 and GSE33113, Log2 intensity of probe 202326 were used to represent the expression level of G9A. For GSE28722 and GSE6988, Log2 intensity of probe 10023819278 and AA434117 were used to represent the expression level of G9A, respectively. The significance was defined as P<0.05.

Gene set enrichment analysis (GSEA)

JAVA program for GSEA (http://www.broadinstitute.org/gsea) (35,36) was utilized to analyze the potential genes influenced by G9A high expression. CRC patient gene profiling data (GSE37892 and GSE18088) was obtained from the GEO site. The patients were divided into two groups according to their G9A expression level (top 50%, high vs. bottom 50%, low) and GSEA was carried out to assess the effects of G9A expression level on various biological gene sets. MsigDB c5 (GO gene sets, 1,454 gene sets) was used. Gene sets with a false discovery rate value (FDR) of <0.25 and normal values of P<0.05 subsequent to performing 1,000 permutations were regarded as having significant enrichment.

Statistical analysis

Experimental data were analyzed with SPSS 13.0 statistical software (SPSS, Inc., Chicago, IL, USA). The χ2 and Fisher's exact tests were used to analyze the statistical significance of the relationship between G9A expression and the clinicopathological features. For univariate recurrence analysis, recurrence curves were obtained with the Kaplan-Meier method and compared using the log-rank test. P<0.05 was considered to represent a statistically significant difference.

Results

G9A expression was increased in CRC tumor tissues

To investigate the role of G9A in colorectal cancer, we examined G9A protein expression level in 100 pairs of colorectal cancer tissues and the corresponding non-cancerous tissues by immunohistochemistry. G9A positively staining cells exhibited brown particles that localized in nuclei (Fig. 1A-G). Results showed that G9A protein expression was significantly increased in tumor samples (44%, n=100) in comparison to that in the adjacent non-cancer tissue samples (8.54%, n=82) (Fig. 1H). The difference was significant (P<0.05). In Fig. 1G, the represented specimen was from the junction between tumoral and non-tumoral tissue. As illustrated in Fig. 1G, G9A protein is strongly stained in the tumoral region, whereas the non-tumoral region is weakly stained. The present results indicate that G9A expression in CRC tumor samples is significantly higher than that in adjacent noncancerous tissue samples (Fig. 1H; P<0.05).

Association between G9A expression and clinicopathological features of CRC

The present study analyzed the association of G9A protein expression level with patient age, gender, size, TNM stages (primary tumor status (T1-T4), nodal metastasis (N0-N2), distant metastasis), Dukes stages (A-D), and histological grade (well, moderately, or poorly differentiated) in the CRC samples (Table I). There were significant differences in the G9A protein expression levels between later the TNM stage and the other TNM stages (P=0.035, P=0.020; IV compared with II and III, respectively; Table I, Fig. 2A). In addition, expression of G9A in tumor with distant metastasis was increased compared with that without distant metastasis (P=0.043, Table I, Fig. 2B). It also demonstrated a weak association with Dukes Stage that G9A expression in Dukes stage D tumor was significantly increased compared with that in Dukes stage C tumor (P=0.049; Table I, Fig. 2C). There was no significant correlation of G9A expression with other clinicopathological features, such as patient age, gender, tumor size, invasion and nodal metastasis.

Expression of G9A using online analysis platform

The present study explored the mRNA expression level of G9A in CRC. Data mining and analysis of G9A mRNA expression level from the publicly available Oncomine database was performed. The threshold was set by the P-value (1E-6) and by fold change (1.5+). There were 4 data sets that met the requirements, which were the TCGA (The Cancer Genome Atlas) colorectal cancer dataset consisting of 237 clinical samples (37), the Kaiser colon dataset consisting of 105 clinical samples (38), the Gaedcke colorectal dataset consisting of 130 clinical samples (39) and the Hong colorectal dataset consisting of 82 samples (40). The results from Oncomine further confirmed the significantly higher expression level of G9A identified in various colorectal carcinomas, such as cecum adenocarcinoma, colon adenocarcinoma, colon mucinous adenocarcinoma and rectal adenocarcinoma (Fig. 3A-C). The corresponding P-values and fold changes are exhibited in Fig. 3D-F, respectively. These results indicated that G9A mRNA expression was increased in CRC tumor tissues.

Association between G9A mRNA expression and clinicopathological features of colorectal cancer

To figure out the relationships between elevated G9A expression level and the clinicopathological features of CRC, the present study chose 5 GEO datasets with corresponding clinical information for further analysis (Table II). The results demonstrated that G9A expression was associated with AJCC staging (P=0.027) and tumor cell differentiation (P=0.012) in GSE17536 (n=177), and it was also related with cancer relapse in GSE18088 (P=0.045, n=53). In GSE33113 (n=96), a marked association between G9A expression and the recurrence rate in CRC was found (P=0.0507, Fig. 4). It might suggest that CRC patients with high expression level of G9A were more likely to suffer relapse. However, cause the result based on GSE33113 was borederline signficant (P=0.0507), this result still requires further investigation to further verify, as the association in this dataset did not prove to be statistically significant. No other associations between G9A expression and clinicopathological features in any other CRC datasets we found, including metastasis, staging, location or survival rate.

Table II.

Correlation between G9A mRNA expression and the clinicopathological features of the CRC.

Table II.

Correlation between G9A mRNA expression and the clinicopathological features of the CRC.

G9 expression

DatasetsCharacteristicCase sizeHighLow P-valuea
GSE38832Ajcc staging 0.259
Probe: 202326118711
2351718
3392514
4301416
GSE28722Duke's staging 0.618
Probe: 10023819278A312
B833845
C341222
D532
Metastasis 0.916
Y331419
N924052
GSE17536Ajcc staging 0.162
Probe: 2023261247170.027b
2573225
3572928
4391821
Differentiated 0.012
Well16214
Moderately1346965
Poorly271413
Recurrence 0.313
Y362016
N1095059
GSE18088Relapse 0.045
Probe: 202326Y13103
N401822
Location 0.506
Proximal281612
Distal251213
Differentiation 0.327
Well211
Moderate352114
Low16610

{ label (or @symbol) needed for fn[@id='tfn4-ol-0-0-8446'] } Gene expression classification into either low or high groups was determined by whether the value was lower or higher than the mean of that dataset.

a Chi-square test was used to statistically analyze the association between title category and G9A protein expression (all unmarked P-values)

b AJCC stage 1 compared with stage 2. AJCC, American Joint Committee on Cancer.

G9A expression level associated with proliferation of CRC cells

The G9A expression level was found consistently increased in both public available datasets and in the present CRC tumor samples. A GSEA analysis was conducted to investigate the potential biological processes that G9A high expression may influence. The results from GSEA using GSE37892 demonstrated that gene sets differences in G9A high vs. low patients indicated that G9A regulated gene sets mainly associated with DNA replication (P=0.016, FDR=0.160, Fig. 5A). The result by analyzing GSE18088 also indicated that high expression of G9A regulated gene sets associated with mitosis, although the FDR value was weak (M phase, P=0.042, FDR=0.433; mitosis, P=0.040, FDR=0.442; Fig. 5B and C). The present study concluded that G9A may be important for proliferation of CRC cells.

Discussion

The present study investigated the clinicopathological significance of G9A expression in CRC progression. Immunohistochemistry was performed to explore G9A protein expression pattern in 100 pairs of CRC samples. Combined with the results of the bioinformatics analysis, it was demonstrated that G9A expression was increased in CRC and that it is therefore involved in the carcinogenesis of CRC.

G9A is the primary histone lysine methyltransferase of lysine 9 on histone H3. Various studies have demonstrated that G9A is critical for numerous biological progresses, such as embryo development, behavior plasticity, lymphocyte development, stem cell differentiation and tumor cell growth (13,16,17,41). It has been reported as overexpressed in numerous types of cancer, including hepatocellular carcinoma, bladder cancer, leukemia and lung cancer (17,18,42). Inhibition of G9A represses cellular proliferation by inducing autophagy related cell death in colon cancer, breast cancer and neuroblastoma cells (17,20). The present study investigated the potential functions of G9A in CRC progression.

The results of the present immunohistochemistry analysis were consistent with Oncomine datasets, demonstrating that G9A expression was significantly elevated in CRC tumor tissues. Fig. 1G exhibited clearly that G9A was strongly stained in the tumor region and weakly stained in the non-tumor region. The clinical significance of high G9A expression in CRC was further analyzed. It was found that G9A protein expression was significantly higher in TNM stage IV tumor than any other TNM stages in our 100 specimens. In addition, the G9A expression exhibited a higher expression in Dukes stage D tumor than that in stage C tumor, and also a higher expression in tumor with distant metastasis than that without distant metastasis. According to the principle of all these different types of staging methods, G9A expression levels may be important for the metastasis status of CRC. A previous study on lung cancer has stated that G9A has the ability to promote tumor invasion and metastasis in lung cancer (43). The present study provided the evidence that increased expression of G9A was associated with metastasis in CRC. In accordance, by using publicly available datasets from GEO, it was found that high G9A expression was associated with AJCC staging and differentiation of CRC in one of the datasets analyzed. However, since the results were not consistent among different datasets used, and the association between G9A expression and tumor relapse in CRC was borderline significant, the associations between G9A and tumor metastasis or tumor recurrence need to be further confirmed.

To find the potential function of G9A in CRC progression, GSEA analysis was performed. The results demonstrated that G9A was significantly associated with DNA replication and mitosis in CRC tumor cells in 2 datasets analyzed. These results strongly indicated that G9A was important for colorectal cancer cell proliferation, which was consistent with the previous research (20). It has been demonstrated that G9A inhibition represses proliferation in colon cancer HCT116 cells and numerous other types of cancer cell (20,21). The present study suggested that high expression level of G9A was critical for CRC tumorigenesis.

In the present study, the clinicopathological significance of G9A high expression in CRC was discussed. In conclusion, the results suggested that G9A expression was increased in CRC tumor samples and the high expression was important for tumorigenesis. Evidence that G9A high expression may be important for distant metastasis in CRC has been provided. The findings helped to further understand the crucial role of G9A in tumorigenesis, and also offered significant ideas for CRC therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 31300609), the Natural Science Foundation of Hubei Province in China (grant nos. 2012FFB04316 and 2013CFB255) and The Incubator Project of Renmin Hospital Wuhan University (grant no. 2013RMFH008).

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

JQ and ZZ analyzed and interpreted the patient data. ZZ, QL and YH collected the samples. TL contributed to data analysis and the manuscript drafting. JQ and LC contributed to the study design and manuscript writing.

Ethics approval and consent to participate

Ethical approval for the study was granted by the Renmin Hospital's ethics committee. Informed written consent was obtained from all participants involved in the study.

Consent for publication

Informed written consent for publication was obtained from all participants involved in the study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rodríguez-Paredes M and Esteller M: Cancer epigenetics reaches mainstream oncology. Nat Med. 17:330–339. 2011. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

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

4 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Carmona FJ and Esteller M: Moving closer to a prognostic DNA methylation signature in colon cancer. Clin Cancer Res. 17:1215–1217. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, et al: The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 41:178–186. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Jiang X, Tan J, Li J, Kivimäe S, Yang X, Zhuang L, Lee PL, Chan MT, Stanton LW, Liu ET, et al: DACT3 is an epigenetic regulator of Wnt/beta-catenin signaling in colorectal cancer and is a therapeutic target of histone modifications. Cancer Cell. 13:529–541. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Fluge Ø, Gravdal K, Carlsen E, Vonen B, Kjellevold K, Refsum S, Lilleng R, Eide TJ, Halvorsen TB, Tveit KM, et al: Expression of EZH2 and Ki-67 in colorectal cancer and associations with treatment response and prognosis. Br J Cancer. 101:1282–1289. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Yamamoto S, Tateishi K, Kudo Y, Yamamoto K, Isagawa T, Nagae G, Nakatsuka T, Asaoka Y, Ijichi H, Hirata Y, et al: Histone demethylase KDM4C regulates sphere formation by mediating the cross talk between Wnt and Notch pathways in colonic cancer cells. Carcinogenesis. 34:2380–2388. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Shinkai Y and Tachibana M: H3K9 methyltransferase G9a and the related molecule GLP. Genes Dev. 25:781–788. 2011. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Maze I, Covington HE III, Dietz DM, LaPlant Q, Renthal W, Russo SJ, Mechanic M, Mouzon E, Neve RL, Haggarty SJ, et al: Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science. 327:213–216. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Thomas LR, Miyashita H, Cobb RM, Pierce S, Tachibana M, Hobeika E, Reth M, Shinkai Y and Oltz EM: Functional analysis of histone methyltransferase g9a in B and T lymphocytes. J Immunol. 181:485–493. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Katoh K, Yamazaki R, Onishi A, Sanuki R and Furukawa T: G9a histone methyltransferase activity in retinal progenitors is essential for proper differentiation and survival of mouse retinal cells. J Neurosci. 32:17658–17670. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Ueda J, Ho JC, Lee KL, Kitajima S, Yang H, Sun W, Fukuhara N, Zaiden N, Chan SL, Tachibana M, et al: The hypoxia-inducible epigenetic regulators Jmjd1a and G9a provide a mechanistic link between angiogenesis and tumor growth. Mol Cell Biol. 34:3702–3720. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Chen X, Skutt-Kakaria K, Davison J, Ou YL, Choi E, Malik P, Loeb K, Wood B, Georges G, Torok-Storb B and Paddison PJ: G9a/GLP-dependent histone H3K9me2 patterning during human hematopoietic stem cell lineage commitment. Genes Dev. 26:2499–2511. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Ke XX, Zhang D, Zhu S, Xia Q, Xiang Z and Cui H: Inhibition of H3K9 methyltransferase G9a repressed cell proliferation and induced autophagy in neuroblastoma cells. PLoS One. 9:e1069622014. View Article : Google Scholar : PubMed/NCBI

18 

Huang J, Dorsey J, Chuikov S, Pérez-Burgos L, Zhang X, Jenuwein T, Reinberg D and Berger SL: G9a and Glp methylate lysine 373 in the tumor suppressor p53. J Biol Chem. 285:9636–9641. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Kondo Y, Shen L, Suzuki S, Kurokawa T, Masuko K, Tanaka Y, Kato H, Mizuno Y, Yokoe M, Sugauchi F, et al: Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol Res. 37:974–983. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Kim Y, Kim YS, Kim DE, Lee JS, Song JH, Kim HG, Cho DH, Jeong SY, Jin DH, Jang SJ, et al: BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production. Autophagy. 9:2126–2139. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Zhang J, He P, Xi Y, Geng M, Chen Y and Ding J: Down-regulation of G9a triggers DNA damage response and inhibits colorectal cancer cells proliferation. Oncotarget. 6:2917–2927. 2015.PubMed/NCBI

22 

Edge SB and Compton CC: The American joint committee on cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Kyriakos M: The President's cancer, the Dukes classification, and confusion. Arch Pathol Lab Med. 109:1063–1066. 1985.PubMed/NCBI

24 

Barresi V, Bonetti Reggiani L, Ieni A, Caruso RA and Tuccari G: Histological grading in colorectal cancer: New insights and perspectives. Histol Histopathol. 30:1059–1067. 2015.PubMed/NCBI

25 

Zeng Z, Wu HX, Zhan N, Huang YB, Wang ZS, Yang GF, Wang P and Fu GH: Prognostic significance of USP10 as a tumor-associated marker in gastric carcinoma. Tumour Biol. 35:3845–3853. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, Barrette T, Pandey A and Chinnaiyan AM: ONCOMINE: A cancer microarray database and integrated data-mining platform. Neoplasia. 6:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Tripathi MK, Deane NG, Zhu J, An H, Mima S, Wang X, Padmanabhan S, Shi Z, Prodduturi N, Ciombor KK, et al: Nuclear factor of activated T-cell activity is associated with metastatic capacity in colon cancer. Cancer Res. 74:6947–6957. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Laibe S, Lagarde A, Ferrari A, Monges G, Birnbaum D and Olschwang S: COL2 Project: A seven-gene signature aggregates a subgroup of stage II colon cancers with stage III. OMICS. 16:560–565. 2012.PubMed/NCBI

29 

Loboda A, Nebozhyn MV, Watters JW, Buser CA, Shaw PM, Huang PS, Van't Veer L, Tollenaar RA, Jackson DB, Agrawal D, et al: EMT is the dominant program in human colon cancer. BMC Med Genomics. 4:92011. View Article : Google Scholar : PubMed/NCBI

30 

Smith JJ, Deane NG, Wu F, Merchant NB, Zhang B, Jiang A, Lu P, Johnson JC, Schmidt C, Bailey CE, et al: Experimentally derived metastasis gene expression profile predicts recurrence and death in patients with colon cancer. Gastroenterology. 138:958–968. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Freeman TJ, Smith JJ, Chen X, Washington MK, Roland JT, Means AL, Eschrich SA, Yeatman TJ, Deane NG and Beauchamp RD: Smad4-mediated signaling inhibits intestinal neoplasia by inhibiting expression of β-catenin. Gastroenterology. 142:562–571.e2. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Gröne J, Lenze D, Jurinovic V, Hummel M, Seidel H, Leder G, Beckmann G, Sommer A, Grützmann R, Pilarsky C, et al: Molecular profiles and clinical outcome of stage UICC II colon cancer patients. Int J Colorectal Dis. 26:847–858. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Kemper K, Versloot M, Cameron K, Colak S, de Sousa e Melo F, de Jong JH, Bleackley J, Vermeulen L, Versteeg R, Koster J and Medema JP: Mutations in the Ras-Raf Axis underlie the prognostic value of CD133 in colorectal cancer. Clin Cancer Res. 18:3132–3141. 2012. View Article : Google Scholar : PubMed/NCBI

34 

de Sousa E, Melo F, Colak S, Buikhuisen J, Koster J, Cameron K, de Jong JH, Tuynman JB, Prasetyanti PR, Fessler E, van den Bergh SP, et al: Methylation of cancer-stem-cell-associated Wnt target genes predicts poor prognosis in colorectal cancer patients. Cell Stem Cell. 9:476–485. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 34:267–273. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Kaiser S, Park YK, Franklin JL, Halberg RB, Yu M, Jessen WJ, Freudenberg J, Chen X, Haigis K, Jegga AG, et al: Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer. Genome Biol. 8:R1312007. View Article : Google Scholar : PubMed/NCBI

39 

Gaedcke J, Grade M, Jung K, Camps J, Jo P, Emons G, Gehoff A, Sax U, Schirmer M, Becker H, et al: Mutated KRAS results in overexpression of DUSP4, a MAP-kinase phosphatase, and SMYD3, a histone methyltransferase, in rectal carcinomas. Genes Chromosomes Cancer. 49:1024–1034. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Hong Y, Downey T, Eu KW, Koh PK and Cheah PY: A ‘metastasis-prone’ signature for early-stage mismatch-repair proficient sporadic colorectal cancer patients and its implications for possible therapeutics. Clin Exp Metastasis. 27:83–90. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Tachibana M, Sugimoto K, Nozaki M, Ueda J, Ohta T, Ohki M, Fukuda M, Takeda N, Niida H, Kato H and Shinkai Y: 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

42 

Cho HS, Kelly JD, Hayami S, Toyokawa G, Takawa M, Yoshimatsu M, Tsunoda T, Field HI, Neal DE, Ponder BA, et al: Enhanced expression of EHMT2 is involved in the proliferation of cancer cells through negative regulation of SIAH1. Neoplasia. 13:676–684. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, Shiah SG, Chen PS, Jeng YM, Cheng TY, et al: H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 70:7830–7840. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 15 Issue 6

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
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
APA
Qin, J., Zeng, Z., Luo, T., Li, Q., Hao, Y., & Chen, L. (2018). Clinicopathological significance of G9A expression in colorectal carcinoma. Oncology Letters, 15, 8611-8619. https://doi.org/10.3892/ol.2018.8446
MLA
Qin, J., Zeng, Z., Luo, T., Li, Q., Hao, Y., Chen, L."Clinicopathological significance of G9A expression in colorectal carcinoma". Oncology Letters 15.6 (2018): 8611-8619.
Chicago
Qin, J., Zeng, Z., Luo, T., Li, Q., Hao, Y., Chen, L."Clinicopathological significance of G9A expression in colorectal carcinoma". Oncology Letters 15, no. 6 (2018): 8611-8619. https://doi.org/10.3892/ol.2018.8446