Open Access

DNA methylation is a common molecular alteration in colorectal cancer cells and culture method has no influence on DNA methylation

  • Authors:
    • Shibao Wang
    • Yinghui Huang
    • Xupeng Mu
    • Tianyang Qi
    • Sha Qiao
    • Zhenxia Lu
    • Hongjun Li
  • View Affiliations

  • Published online on: January 30, 2018     https://doi.org/10.3892/etm.2018.5809
  • Pages: 3173-3180
  • Copyright: © Wang 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

The present study aimed to explore whether culture method had an influence on DNA methylation in colorectal cancer (CRC). In the present study, CRC cells were cultured in two‑dimensional (2D), three‑dimensional (3D) and mouse orthotopic transplantation (Tis) cultures. Principal component analysis (PCA) was used for global visualization of the three samples. A Venn diagram was applied for intersection and union analysis for different comparisons. The methylation condition of 5'‑C‑phosphate‑G‑3' (CpG) location was determined using unsupervised clustering analysis. Scatter plots and histograms of the mean β values between 3D vs. 2D, 3D vs. Tis and Tis vs. 2D were constructed. In order to explore the biological function of the genes, gene ontology and Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway analyses were utilized. To explore the influence of culture condition on genes, quantitative methylation specific polymerase chain reaction (QMSP) was performed. The three samples connected with each other closely, as demonstrated by PCA. Venn diagram analysis indicated that some differential methylation positions were commonly shared in the three groups of samples and 16 CpG positions appeared hypermethylated in the three samples. The methylation patterns between the 3D and 2D cultures were more similar than those of 3D and Tis, and Tis and 2D. Results of gene ontology demonstrated that differentially expressed genes were involved in molecular function, cellular components and biological function. KEGG analysis indicated that genes were enriched in 13 pathways, of which four pathways were the most evident. These pathways were pathways in cancer, mitogen‑activated protein kinase signaling, axon guidance and insulin signaling. Furthermore, QMSP demonstrated that methylation of mutL homolog, phosphatase and tensin homolog, runt‑related transcription factor, Ras association family member, cadherin‑1, O‑6‑methylguanine‑DNA‑methyltransferase and P16 genes had no obvious difference in 2D, 3D and Tis culture conditions. In conclusion, the culture method had no influence on DNA methylation in CRC cells.

Introduction

Colorectal cancer (CRC), originating from the epithelial lining of the large bowel (1), is a malignant neoplasm containing colon and rectal cancer. Its morbidity rate has increased rapidly over the last 30 years in many countries, including China, Japan and Spain (2). The treatment of CRC predominantly includes surgery, chemotherapy, radiation therapy, biological therapy and the combinational therapy; however, these treatments are not always entirely effective and so CRC often has a poor prognosis (3,4). The development of CRC is a multistage and complex process, as with the majority of malignant tumors. The mechanisms involved in the development of CRC are not fully understood and have attracted the interest of many researchers (5). P16 is a tumor suppressor that has a key influence in many diseases, including CRC (6). The hypermethylation of P16 has been demonstrated to have a key role in carcinogenesis (7). The occurrence of CRC is reported to result from a position named ‘field defect’; however, this mechanism is not well known (8). It has been reported that DNA methylation may have a role in mediating the field defect (8). In CRC cells, alterations to DNA methylation has an important influence (9). However, the molecular mechanism of how DNA hypermethylated genes influence the development of cancer has not been fully elucidated.

To study the varied processes in cancer, such as the inactivation of the X chromosome and the silencing of tumor-related genes, it is necessary to understand the details of DNA methylation in 5′-C-phosphate-G-3′ (CpG) regions (10). CpG islands are often made up of gene promoters or exons (11). However, these islands are often unmethylated in normal cells, and methylation of CpG regions may affect condensed chromatin (12). CpG regions often have roles in promoting DNA replication origins, which are fundamental to chromosome organization and duplication (13). They are bimodal and enriched at CpG islands in mouse and Drosophila. (14). Research has demonstrated that there are often some molecular abnormalities in tumor tissues and adjacent tissues, despite them appearing histologically normal (15). The occurrence of these abnormalities in the adjacent tissues is named ‘field defect’ (16). Research has indicated that higher DNA methylation levels of Ras association family member (RASSF) 1A, adenomatous polyposis coli and human mutL homolog (MLH) exist in CRC than in adjacent tissues (17).

Three-dimensional (3D) cell culture systems as a tumor model in vitro is a vital tool in research of DNA methylation (18). In 3D cultured pluripotent stem cells, DNA methylation was changed and some genes were upregulated (19). However, it is not known whether the culture method affects the DNA methylation in CpG regions in CRC cells. The present study aimed to identify the relationship between DNA methylation and CRC, and the influence of culture method on DNA methylation.

Materials and methods

Cell culture

CRC cell line, DLD-1, was purchased from Renmin Hospital of Hubei University of Medicine (Shiyan, China). DLD-1 cells were routinely cultivated in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin at two-dimensional (2D), 3D and orthotopic transplantation (Tis) stages in a humidified cell incubator (5% CO2) at 37°C. 2D culture refers to DLD-1 culture in RPMI-1640 supplemented with 10% FBS in a common culture dish. 3D culture refers to the culture of DLD-1 cells in suspension in RPMI-1640 free of FBS in a culture plate containing Matrigel, which was used to distribute the signal cells. This means that DLD-1 cells were inoculated into 20 nude rats (age, 6 months; weight, 100–150 g; Animal Center of Shandong University, Jinan, China) as previously described (20) to establish a xenograft animal model. The ratio of male to female rats was 1:1. Rats were housed at an ambient temperature of 20–22°C with a relative humidity of 50±5% and a 12 h light/dark cycle. The rats were allowed free access to food and water until the next procedure was performed. Following the formation of an orthotopic colorectal tumor, after 14 days, rats were sacrificed by cervical dislocation under 1% pentobarbital sodium anesthesia (40 mg/kg; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Tumor tissue was harvested, cut into small pieces and rapidly frozen in liquid nitrogen. The present study was approved by the Institutional Animal Care and Use Committee of Jilin University (Changchun, China).

Total RNA isolation and methylation chip genome-wide detection

An RNeasy mini kit (Qiagen, Inc., Valencia, CA, USA) was used to isolate total RNA from rat tissues, according to the manufacturer's instructions. The reverse transcription reaction was carried out with TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in 15 µl containing 5 µl RNA extract, 0.15 µl 100 mM dNTPs, 1 µl Multiscribe Reverse Transcriptase (50 U/µl), 1.5 µl 10× reverse transcription buffer, 0.19 µl RNase inhibitor (20 U/µl), 1 µl of gene-specific primer and 4.16 µl of nuclease-free water. For synthesis of cDNA, the reaction mixtures were incubated at 16°C for 30 min, 42°C for 30 min, 85°C for 5 min and stored at 4°C. A total of 1.33 µl cDNA solution was amplified using 10 µl of TaqMan 2X Universal polymerase chain reaction (PCR) Master Mix with no AmpErase UNG (Applied Biosystems; Thermo Fisher Scientific, Inc.), 1 µl gene-specific primers/probe and 7.67 µl of nuclease-free water in a final volume of 20 µl. Quantitative PCR (TaqMan) analysis was performed on a TaqMan ABI Prism 7000 Sequence Detection System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The reaction mixtures were incubated at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min. The relative expression was normalized to the expression of β-actin (forward, 5′-ACAGAGCCTCGCCTTTGC-3′ and reverse, 5′-GCGGCGATATCATCATCC-3′). Relative fold changes of gene expression were calculated by the 2−ΔΔCq method (21,22). The methylation primer was as follows: Forward, 5′-CGTTTTATTTCGGTTTTGTTTTC-3′ and reverse, 5′-CTCGAAATTTAATAAAAACTTCACG-3′. The amplified DNA was cut into segments by restriction endonuclease (Bethesda Research Laboratories, Inc., Gaithersburg, MD, USA) and the DNA fragments were precipitated by precipitation solution PM1 (Promega Corp., Madison, WI, USA) and isopropanol (Sigma-Aldrich; Merck KGaA). The sedimentary DNA was suspended again in precipitation solution PM1 and isopropanol. The resuspended DNA samples were dispersed on beadchip chips and Human HT-12 v. 4.0 BeadChip (Illumina, Inc., San Diego, CA, USA) was used for hybridization. Illumina iScan 148 (Illumina, Inc.) was applied to image the BeadChips. Before using this, Illumina data were reserved on the basis of the Minimum Information about a Microarray Experiment guidelines in the public Gene Expression Omnibus database (ncbi.nlm.nih.gov/geo/), accession number GSE 54690.

DNA extraction and quantitative methylation specific PCR (QMSP)

A Multisource Genomic DNA Miniprep kit (Axygen Scientific; Thermo Fisher Scientific, Inc.) was applied to extract total DNA from DLD-1 cells, according to the recommendations of manufacturer. A CpGenome™ DNA Modification kit (Chemicon; Merck KGaA) was employed to modify each genomic DNA from samples using sodium bisulfate according to the manufacturer's protocol. The methylation levels of MLH, phosphatase and tensin homolog (PTEN), runt-related transcription factor (RUNX), RASSF, cadherin-1 (CDH1), O-6-methylguanine-DNA-methyltransferase (MGMT) and P16 were determined. The regions of β-actin that were short of any CpG dinucleotide were amplified using a Cyclogene thermal cycler machine (Techne, Cambridge, UK): 65°C for 5 min, 96°C for 2 min, 65°C for 4 min, 96°C for 1 min, 65°C for 1 min, and 96°C for 30 sec. The primers used were as follows: MLH forward 5′-GGTTGGATATTTYGTATTTTTYGAG-3′ and reverse 5′-AATTACTAAATCTCTTCRTCCCTCC-3′; RUNX forward, 5′-CCTTACGTAGAGGTCACAGTAG-3′ and reverse 5′-CTCCAAGCTGCAAAGTCAC-3′; CDH1 forward, 5′-AATTTTAGGTTAGAGGGTTATCGCGT-3′ and reverse, 5′-TCCCCAAAACGAAACTAACGAC-3′; MGMT forward, 5′-GGGTTATTTGGTAAATTAAGGTATAGAG-3′ and reverse, 5′-CACCTAAAAATAAAACAAAAACTACCAC-3′; P16 forward, 5′-GAGGGGGTAGGGGGATAT-3′ and reverse, 5′-ACCAATCAACCAAAAACTCCATACTA-3′; β-actin forward, 5′-ACAGAGCCTCGCCTTTGC-3′ and reverse, 5′-GCGGCGATATCATCATCC-3′.

Analysis of differential gene transcription

Principal component analysis (PCA) may be used to analyze data tables whose content contains some inter-correlated quantitative dependent variables. Partek Genomics Suite v. 6.5 (Partek, Inc., St Louis, MO, USA) was used to analyze the gene expression data. The analyzed data were then corrected and normalized by quantile normalization and summarization (23). PCA was used for global visualization of all datasets.

Functional enrichment and pathway enrichment analysis

MetaCore Bioinformatics software (www.portal.genego.com/) was used to identify the significant pathways. Data for Annotation, Visualization, and Integrated Discovery (DAVID; (http://david.niaid.nih.gov),) was used to conduct Gene Ontology (GO; http://www.geneontology.org/) and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg/ or http://www.kegg.jp/) pathway enrichment analysis. DAVID is able to combine integrate functional genomic annotations with intuitive graphical summaries. This contributed to the explanation on genome-scale datasets by promoting the change from data collection to biological meaning. GO terms and KEGG pathways with P<0.01 were selected (24). The P-values were calculated as follows:

p=(a+ba)(c+dc)(na+c)

Where, n is the number of background genes; a′ is the gene number of one gene set in the gene lists; a′ + b is the number of genes in the gene list and one gene set was also included; a′ + c is the gene number of one gene list in the background genes; and a′ is replaced with a = a′-1.

Statistical analysis

Scatter plots of the genome-wide methylation changes in Tamoxifen-resistant lines and parental was performed. SPSS v. 19.0 (IBM Corp., Armonk, NY, USA) and GraphPad Prism 6 (GraphPad Software, Inc., La Jolla, CA, USA) software were employed to analyze data. Two sample t-tests were used to differentiate the mean methylation scores between two samples. A paired t-test was applied to determine the differences of average sib pair in methylation scores. P<0.05 was considered to indicate a statistically significant difference.

Results

Analysis of differential gene transcription

Partek Genomics Suite v. 6.5 was used to analyze the gene expression data. The analyzed data were then corrected and normalized by quantile normalization and summarization. The datasets were deposited for CRC cells (DLD-1) in 2D, 3D and Tis stages. PCA was used for global visualization of all datasets (Fig. 1A). PCA is one of the most important and powerful methods in chemometrics as well as in a wealth of other areas (25). It is a one-sample technique applied to data with no groupings among the observations and no partitioning of the variables into sub-vectors y and × (26). PCA analysis revealed the close connection between the DLD1-3D, DLD1-2D and DLD1-Tis groups. Gene lists were established with a cut-off of relative ± 1.5-fold change and P≤0.05. A Venn diagram of modulated samples in the DLD1-3D, DLD1-2D and DLD1-Tis groups was created. The result demonstrated that 23 genes were commonly found in the DLD1-3D vs. DLD1-2D group, DLD1-3D vs. DLD1-Tis group and DLD1-Tis vs. DLD1-2D group. Apart from the common 23 genes, 95 genes were commonly identified in the DLD1-3D vs. DLD1-2D group and DLD1-Tis vs. DLD1-2D group, 44 genes were commonly identified in the DLD1-3D vs. DLD1-2D group and DLD1-3D vs. DLD1-Tis group, and 18,912 genes were commonly identified in the DLD1-3D vs DLD1-Tis group and DLD1-Tis vs. DLD1-2D group (Fig. 1B). Furthermore, results demonstrated that the overlapping section in the three populations of samples presented high methylation. Unsupervised clustering analysis of the CpG location revealed that 119 CpGs presented different levels of methylation in the three groups of samples and 16 CpGs all appeared with a highly methylated status in the three groups of samples (Fig. 1C).

Analysis on different CpGs

To analyze the different CpG locations in CRC cells cultured in 2D, 3D and Tis stage cultures, scatter plots were created to compare all CpG sites among the DLD1-3D, DLD1-2D and DLD1-Tis groups (Fig. 2). The areas outlined in grey on each of the scatter plots in Fig. 2A-C included data points for demethylated CpG sites that demonstrated a 2-fold change and had average β values of >0.2. The methylation patterns between DLD1-3D and DLD1-2D (Fig. 2A) were more similar than those of DLD1-3D and DLD1-Tis (Fig. 2B), and DLD1-Tis and DLD1-2D (Fig. 2C). Fig. 2D-F demonstrated the column distribution of β values in the three groups of samples. AVG_β>0.2 stated that the degree of methylation differences was large.

Functional analysis of differentially methylated DNA

To identify the biological function, cellular component and molecular function of the genes identified, GO and pathway analysis were utilized. From GO, it was demonstrated that there were differentially expressed genes in various over-represented cellular processes. In terms of biological process, DNA-dependent transcription, apoptosis, ion transport, cell differentiation and transmembrane transport were identified to be very important. In terms of cellular components, membrane fraction, cytosol, integral to plasma membrane, perinuclear region of cytoplasm and cytoskeleton were demonstrated to be important. As for molecular function, there were some key functions identified, including signal transducer activity, adenosine 5′-triphosphate binding, calcium ion binding, protein homedimerization activity and nucleotide binding (Fig. 3A). To further refine the biological functions of genes corresponding by differential methylation sites, these genes were placed into cellular or metabolic pathways, based on their roles. KEGG may be used to systematically analyze gene functions based on the networks of genes and molecules. Pathway analyses of the corresponding genes recognized 13 significantly over-represented cellular pathways (P=0.032; Fig. 3B). Among these 13 pathways, four pathways were more significant than the others (P=0.008). These pathways were cancer pathways, the mitogen-activated protein kinase (MAPK) signaling pathway, axon guidance and the insulin signaling pathway.

Culture condition has no effect on methylation

To determine whether different cell culture methods affected important genes, a QMSP experiment was used. It has been reported that PTEN has a role in regulating the formation of tumors, and its sequence was similar to cytoskeletal protein tensin (27). RUNX genes have been identified as tumor suppressors or oncogenes based on their roles in regulating cell fate and their ambivalent influences in cancer (28). RASSF genes are tumor suppressors and RASSF expression is decreased in various types of cancer (29). The results of the present QMSP experiment demonstrated that the methylation of the MLH, PTEN, RUNX, RASSF, CDH1, MGMT and P16 genes and the related genes had no obvious difference in 2D, 3D and Tis culture conditions (Fig. 4A and B).

Discussion

CRC has a high incidence and mortality rate worldwide (30). Studies have demonstrated that the occurrence rate of CRC in China has increased yearly and the rate will continue to increase with time (31,32). At present, ~1.25 million worldwide have CRC and >600,000 patients will lose their life as a result each year (33). In human CRC, it has been demonstrated that DNA methylation of CpG islands is able to silence the gene when the methylation occurs in a promoter region (34). In normal cells, CG nucleotides have been identified in promoter regions of several tissue-specific genes with no change in the methylation pattern; however, in cancer cells, this pattern was altered (35). The present study aimed to explore the influence of culture method on DNA methylation in CRC.

First, human CRC DLD-1 cells were obtained and cultured at 2D, 3D and Tis stage cultures. The differentially methylated DNA in each sample was selected and the relationship between them was determined by PCA. Results demonstrated that the differentially methylated DNA in the three different samples was closely related. In order to determine groups of differentially methylated sites common in the three samples or belonging to one sample, a Venn diagram was used for intersection and union analysis with different comparisons. Results identified that 23 differentially methylated sites were common among the three groups of samples, and the common sections presented high methylation in the three samples. Unsupervised clustering analysis was then used to explore the methylation status of CpG regions. Results demonstrated that 119 CpGs presented different levels of methylation in the three groups of samples and 16 CpGs all appeared to have a high methylation status in the three groups of samples. Previous research has investigated abnormal DNA methylation in promoters with a CpG region, and CpG region shores were reported have a key role in hiding the alteration of DNA methylation in human CRC (36). In the present study, scatter plots were created to compare all CpG sites among the DLD1-3D, DLD1-2D and DLD1-Tis groups and to analyze the different CpG locations in CRC cells cultured in 2D, 3D and Tis stage. Results demonstrated that methylation patterns of DLD1-3D and DLD1-2D were more similar than those of DLD1-3D and DLD1-Tis, and DLD1-Tis and DLD1-2D.

The results of GO indicated that differentially expressed genes were involved in molecular function, cellular component and biological function. KEGG pathway analysis creates manually curated pathway maps that present content on biological networks in graphical forms (37). The present results demonstrated that genes were enriched in 13 pathways, and four of these pathways were more evident than the rest. These pathways included cancer pathways, the MAPK signaling pathway, axon guidance and the insulin signaling pathway. PTEN is a tumor suppressor that may negatively affect the phosphatidylinositol-3 kinase/protein kinase B signaling pathway (38). In CRC, abnormal expression of PTEN is useful for the damage responses to cetuximab (39). Furthermore, PTEN downregulation is related to liver metastasis and low survival rate in CRC (40). RUNX may regulate a variety of biological processes, such as growth and differentiation of lymphocytes and hematopoietic cells (41). In CRC cells, RUNX2 may regulate the transcription of a metastatic gene, osteopontin (42). RASSF gene family genes have been reported to be epigenetically silenced with promoter methylation (43). Ras proteins have an important role in human cancer and may activate mutations in Ras, which occur in ~30% of tumors (44). To explore whether the different cell culture methods had an effect on important genes, a QMSP experiment was performed in the present study. Results demonstrated that the methylation of the MLH, PTEN, RUNX, RASSF, CDH1, MGMT and P16 genes and the related genes had no obvious difference in the 2D, 3D and Tis culture conditions.

Considering the results and discussion above, in conclusion, DNA methylation was associated with the development of CRC; however, this was not altered under 2D, 3D or Tis culture conditions. However, previous research has demonstrated that DNA methylation and gene expression in squamous cell carcinoma had significant differences between 2D and 3D culture systems (45). Compared with squamous cell carcinoma, CRC may have particular characteristics due to a unique microenvironment. It is extremely complex to study the molecular mechanisms of CRC in a tumor model in situ; therefore, 3D cell culture, may replace animal models as a novel experimental method to study the progression of CRC. However, the effects of different culture methods on other cancer types requires further research.

References

1 

Lobert VH, Mouradov D and Heath JK: Focusing the spotlight on the zebrafish intestine to illuminate mechanisms of colorectal cancer. Adv Exp Med Biol. 916:411–437. 2016. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Girard P, Ducreux M, Baldeyrou P, Rougier P, Le Chevalier T, Bougaran J, Lasser P, Gayet B, Ruffié P and Grunenwald D: Surgery for lung metastases from colorectal cancer: Analysis of prognostic factors. J Clin Oncol. 14:2047–2053. 1996. View Article : Google Scholar : PubMed/NCBI

4 

Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pintér T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 360:1408–1417. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Jiang W, Wang PG, Zhan Y and Zhang D: Prognostic value of p16 promoter hypermethylation in colorectal cancer: A meta-analysis. Cancer Invest. 32:43–52. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Zou HZ, Yu BM, Wang ZW, Sun JY, Cang H, Gao F, Li DH, Zhao R, Feng GG and Yi J: Detection of aberrant p16 methylation in the serum of colorectal cancer patients. Clin Cancer Res. 8:188–191. 2002.PubMed/NCBI

7 

Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC, Baylin SB and Sidransky D: 5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers. Nat Med. 1:686–692. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Shen L, Kondo Y, Rosner GL, Xiao L, Hernandez NS, Vilaythong J, Houlihan PS, Krouse RS, Prasad AR, Einspahr JG, et al: MGMT promoter methylation and field defect in sporadic colorectal cancer. J Natl Cancer Inst. 97:1330–1338. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Müller HM, Oberwalder M, Fiegl H, Morandell M, Goebel G, Zitt M, Mühlthaler M, Ofner D, Margreiter R and Widschwendter M: Methylation changes in faecal DNA: A marker for colorectal cancer screening? Lancet. 363:1283–1285. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Herman JG, Graff JR, Myöhänen S, Nelkin BD and Baylin SB: Methylation-specific PCR: A novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA. 93:pp. 9821–9826. 1996; View Article : Google Scholar : PubMed/NCBI

11 

Deaton AM and Bird A: CpG islands and the regulation of transcription. Genes Dev. 25:1010–1022. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Costello JF, Frühwald MC, Smiraglia DJ, Rush LJ, Robertson GP, Gao X, Wright FA, Feramisco JD, Peltomäki P, Lang JC, et al: Aberrant CpG-island methylation has non-random and tumour-type-specific patterns. Nat Genet. 24:132–138. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Holmquist GP and Ashley T: Chromosome organization and chromatin modification: Influence on genome function and evolution. Cytogenet Genome Res. 114:96–125. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Jones PA and Takai D: The role of DNA methylation in mammalian epigenetics. Science. 293:1068–1070. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Wistuba II, Mao L and Gazdar AF: Smoking molecular damage in bronchial epithelium. Oncogene. 21:7298–7306. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Chai H and Brown RE: Field effect in cancer-an update. Ann Clin Lab Sci. 39:331–337. 2009.PubMed/NCBI

17 

Coppedè F, Migheli F, Lopomo A, Failli A, Legitimo A, Consolini R, Fontanini G, Sensi E, Servadio A, Seccia M, et al: Gene promoter methylation in colorectal cancer and healthy adjacent mucosa specimens: Correlation with physiological and pathological characteristics, and with biomarkers of one-carbon metabolism. Epigenetics. 9:621–633. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Godugu C and Singh M: AlgiMatrix™-based 3D cell culture system as an in vitro tumor model: An important tool in cancer research. Methods Mol Biol. 1379:117–128. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Hiler D, Chen X, Hazen J, Kupriyanov S, Carroll PA, Qu C, Xu B, Johnson D, Griffiths L, Frase S, et al: Quantification of retinogenesis in 3D cultures reveals epigenetic memory and higher efficiency in iPSCs derived from rod photoreceptors. Cell Stem Cell. 17:101–115. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Fidler IJ: The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 3:453–459. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Bolstad BM, Irizarry RA, Åstrand M and Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 19:185–193. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Ma CH, Lv Q, Cao Y, Wang Q, Zhou XK, Ye BW and Yi CQ: Genes relevant with osteoarthritis by comparison gene expression profiles of synovial membrane of osteoarthritis patients at different stages. Eur Rev Med Pharmacol Sci. 18:431–439. 2014.PubMed/NCBI

25 

Bro R and Smilde AK: Principal component analysis. Anal Met. 6:2812–2831. 2014. View Article : Google Scholar

26 

Rencher AC: Principal component analysis. Met Multi Anal (Second). 1–407. 2002.

27 

Stambolic V, Suzuki A, De La Pompa JL, Brothers GM, Mirtsos C, Sasaki T, Ruland J, Penninger JM, Siderovski DP and Mak TW: Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell. 95:29–39. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Blyth K, Cameron ER and Neil JC: The RUNX genes: Gain or loss of function in cancer. Nat Rev Cancer. 5:376–387. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Allen NP, Donninger H, Vos MD, Eckfeld K, Hesson L, Gordon L, Birrer MJ, Latif F and Clark GJ: RASSF6 is a novel member of the RASSF family of tumor suppressors. Oncogene. 26:6203–6211. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Atkin WS, Edwards R, Kralj-Hans I, Wooldrage K, Hart AR, Northover JM, Parkin DM, Wardle J, Duffy SW and Cuzick J: UK Flexible Sigmoidoscopy Trial Investigators: Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: A multicentre randomised controlled trial. Lancet. 375:1624–1633. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Dai Z, Zheng RS, Zou XN, Zhang SW, Zeng HM, Li N and Chen WQ: Analysis and prediction of colorectal cancer incidence trend in China. Zhonghua Yu Fang Yi Xue Za Zhi. 46:598–603. 2012.(In Chinese). PubMed/NCBI

32 

Ni S, Peng J, Huang D, Xu M, Wang L, Tan C, SUN H, Cai S and Sheng W: HER2 overexpression and amplification in patients with colorectal cancer: A large-scale retrospective study in Chinese population. Am Society Clin Oncol. 2017.

33 

Mu WP, Wang J, Niu Q, Shi N and Lian HF: Clinical significance and association of RUNX3 hypermethylation frequency with colorectal cancer: A meta-analysis. Onco Targets Ther. 7:1237–1245. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH, Widschwendter M, Weener D, Buchanan D, et al: CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 38:787–793. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Yuan J, Luo RZ, Fujii S, Wang L, Hu W, Andreeff M, Pan Y, Kadota M, Oshimura M, Sahin AA, et al: Aberrant methylation and silencing of ARHI, an imprinted tumor suppressor gene in which the function is lost in breast cancers. Cancer Res. 63:4174–4180. 2003.PubMed/NCBI

36 

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

37 

Zhang JD and Wiemann S: KEGGgraph: A graph approach to KEGG PATHWAY in R and bioconductor. Bioinformatics. 25:1470–1471. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Arico S, Petiot A, Bauvy C, Dubbelhuis PF, Meijer AJ, Codogno P and Ogier-Denis E: The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem. 276:35243–35246. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Perrone F, Lampis A, Orsenigo M, Di Bartolomeo M, Gevorgyan A, Losa M, Frattini M, Riva C, Andreola S, Bajetta E, et al: PI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patients. Ann Oncol. 20:84–90. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Sawai H, Yasuda A, Ochi N, Ma J, Matsuo Y, Wakasugi T, Takahashi H, Funahashi H, Sato M and Takeyama H: Loss of PTEN expression is associated with colorectal cancer liver metastasis and poor patient survival. BMC Gastroenterol. 8:562008. View Article : Google Scholar : PubMed/NCBI

41 

Miyazono K, Maeda S and Imamura T: Coordinate regulation of cell growth and differentiation by TGF-beta superfamily and Runx proteins. Oncogene. 23:4232–4237. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Wai PY, Mi Z, Gao C, Guo H, Marroquin C and Kuo PC: Ets-1 and runx2 regulate transcription of a metastatic gene, osteopontin, in murine colorectal cancer cells. J Biol Chem. 281:18973–18982. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Djos A, Martinsson T, Kogner P and Carén H: The RASSF gene family members RASSF5, RASSF6 and RASSF7 show frequent DNA methylation in neuroblastoma. Mol Cancer. 11:402012. View Article : Google Scholar : PubMed/NCBI

44 

van der Weyden L and Adams DJ: The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 1776:58–85. 2007.PubMed/NCBI

45 

DesRochers TM, Shamis Y, Alt-Holland A, Kudo Y, Takata T, Wang G, Jackson-Grusby L and Garlick JA: The 3D tissue microenvironment modulates DNA methylation and E-cadherin expression in squamous cell carcinoma. Epigenetics. 7:34–46. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang S, Huang Y, Mu X, Qi T, Qiao S, Lu Z and Li H: DNA methylation is a common molecular alteration in colorectal cancer cells and culture method has no influence on DNA methylation. Exp Ther Med 15: 3173-3180, 2018
APA
Wang, S., Huang, Y., Mu, X., Qi, T., Qiao, S., Lu, Z., & Li, H. (2018). DNA methylation is a common molecular alteration in colorectal cancer cells and culture method has no influence on DNA methylation. Experimental and Therapeutic Medicine, 15, 3173-3180. https://doi.org/10.3892/etm.2018.5809
MLA
Wang, S., Huang, Y., Mu, X., Qi, T., Qiao, S., Lu, Z., Li, H."DNA methylation is a common molecular alteration in colorectal cancer cells and culture method has no influence on DNA methylation". Experimental and Therapeutic Medicine 15.4 (2018): 3173-3180.
Chicago
Wang, S., Huang, Y., Mu, X., Qi, T., Qiao, S., Lu, Z., Li, H."DNA methylation is a common molecular alteration in colorectal cancer cells and culture method has no influence on DNA methylation". Experimental and Therapeutic Medicine 15, no. 4 (2018): 3173-3180. https://doi.org/10.3892/etm.2018.5809