Open Access

Identification of aberrantly methylated‑differentially expressed genes and gene ontology in prostate cancer

  • Authors:
    • Linbang Wang
    • Bing Wang
    • Zhengxue Quan
  • View Affiliations

  • Published online on: December 11, 2019     https://doi.org/10.3892/mmr.2019.10876
  • Pages: 744-758
  • 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

Prostate cancer (PCa) is the most frequent urological malignancy in men worldwide. DNA methylation has an essential role in the etiology and pathogenesis of PCa. The purpose of the present study was to identify the aberrantly methylated‑differentially expressed genes and to determine their potential roles in PCa. The important node genes identified were screened by integrated analysis. Gene expression microarrays and gene methylation microarrays were downloaded and aberrantly methylated‑differentially expressed genes were obtained. Enrichment analysis and protein‑protein interactions (PPI) were obtained, their interactive and visual networks were created, and the node genes in the PPI network were validated. A total of 105 hypomethylation‑high expression genes and 561 hypermethylation‑low expression genes along with their biological processes were identified. The top 10 node genes obtained from the PPI network were identified for each of the two gene groups. The methylation and gene expression status of node genes in TCGA database, GEPIA tool, and the HPA database were generally consistent with those of our results. In conclusion, the present study identified 20 aberrantly methylated‑differentially expressed genes in PCa by combining bioinformatics analyses of gene expression and gene methylation microarrays, and concurrently, the survival of these genes was analyzed. Notably, methylation is a reversible biological process, which makes it of great biological significance for the diagnosis and treatment of prostate cancer using bioinformatics technology to determine abnormal methylation gene markers. The present study provided novel therapeutic targets for the treatment of PCa.

Introduction

Globally, prostate cancer (PCa) is the second most frequent cancer. It is the fifth leading cause of cancer-related deaths in males, and is the most frequently diagnosed cancer among men in more than half of all countries (1). It has been established that age, race, and family history are risk factors associated with PCa (2). Previous studies have implicated the initiation and development of PCa as a multistep complex process, driven by changes in the expression of a large number of genes with epigenetic alterations influencing the expression of crucial genes (3,4).

Among the epigenetic modifications, methylation, phosphorylation, acetylation, and ubiquitination have been identified (5,6). A study by De Carvalho et al (7) revealed that DNA methylation can genetically alter gene expression without a change in the DNA sequence. Hypermethylation of a promoter may downregulate gene expression and influence the progression of human cancer (8). Recently, studies have revealed that DNA methylation can identify invasive lesions and silence tumor suppressor genes in PCa, providing a new direction for the treatment of PCa (9,10).

Bioinformatics analysis based on high-throughput platform microarray technology has been extensively used to predict biomarkers of cancers over the last few decades (1113). Numerous gene expression microarrays have been used to identify potential target genes and their functions in PCa (1416). However, the aforementioned studies focused on gene expression microarrays, the number of which is limited, preventing the accurate identification of target genes and their functions in PCa. Therefore, an approved approach includes the combination of gene expression and gene methylation microarray data.

The purpose of this study was to identify aberrantly methylated-differentially expressed genes based on gene expression and gene methylation microarray datasets. The important node genes were screened by integrated analysis with the goal of identifying a novel therapeutic target for the treatment of PCa. The screening steps for determining the aberrantly methylated-differentially expressed genes in PCa are summarized in Fig. 1.

Materials and methods

Data sources

In the present study, the raw data were selected from the Gene Expression Omnibus (GEO), which is an international public repository that can be found on the National Center for Biotechnology Information (NCBI) home page (https://www.ncbi.nlm.nih.gov/geo/). Microarray gene expression data found at accession GSE55945 involved data from 13 PCa samples and eight normal samples, and accession GSE69223 encompassed 15 PCa samples and 15 normal samples, with the platform GPL570 of the two datasets ([HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array). Methylation profile data in GSE47915 comprised four PCa samples and four normal samples, while GSE76938 contained 73 PCa samples and 63 normal samples. The platform of both datasets (GSE47915 and GSE76938) was based on GPL13534 (Illumina HumanMethylation450 BeadChip).

Data processing

The raw data analysis was carried out using GEO2R, which can separately screen differentially methylated genes (DMGs) and differentially expressed genes (DEGs) between normal and cancer prostate sample datasets (17). DMGs and DEGs were obtained using the criteria|t|>2 and P<0.05. The intersection of DMGs and DEGs was derived using the FunRich Venn function (http://www.funrich.org) (18), followed by obtaining the hypomethylation-high expression genes and hypermethylation-low expression genes.

Gene ontology (GO) term enrichment analysis

The GO terms, including the hypomethylation-high expression genes and hypermethylation-low expression genes, were enriched using the Database for Annotation, Visualization, and Integrated Discovery (DAVID, http://david.niaid.nih.gov), and P-values <0.05 were considered statistically significant. The chord plots from the GO results were created using R language with ggplot2 and GOplot packages (19).

Construction of PPI networks

Protein-protein interactions (PPI) are critical events in signaling pathways, especially when interpreting the molecular mechanisms of cellular activities during carcinogenesis. The PPI relationships of the hypomethylation-high expression genes and hypermethylation-low expression genes were obtained by FunRich, and their interactive and visual networks were created using Cytoscape v3.5.0 software (https://cytoscape.org/) (20).

Node gene validation

Gene expression profiling data (HTSeq-FPKM) and methylation sequencing data (Illumina Human Methylation 450) were downloaded from the Cancer Genome Atlas (TCGA) database (https://cancergenome.nih.gov/). DMGs and DEGs were obtained using the criteria |t|>2 and P<0.05. The hypomethylated-high expression genes and hypermethylated-low expression genes obtained from the GEO database were respectively validated in TCGA database, and the methylation and gene expression status of the top ten node genes were also validated. Subsequently, the expression status, translational levels and the mRNA levels of the top ten node genes were validated using the Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancer-pku.cn) and The Human Protein Atlas database (HPA; http://www.proteinatlas.org/), respectively. In addition, the Disease-Free Survival (RFS) method of the GEPIA online tool was used to analyze 20 aberrantly-methylated genes including hypomethylated-high expression genes and hypermethylated-low expression genes. Group cutoff was set as Quartile (Cutoff-High (%) was 75% and Cutoff-Low (%) was 25%). The confidence interval (CU) was 95%. High and low expression genes were respectively represented in red and blue colour (21,22).

Results

Methylated DEGs in PCa screening results

A total of 3,550 high-expression and 3,706 low-expression genes were obtained from GSE55945, while 4,055 high-expression and 4,412 low-expression genes were obtained from GSE69223. In addition, 10,209 hypermethylated and 5,207 hypomethylated genes were obtained from GSE47915 and 2,959 hypermethylated and 1,268 hypomethylated genes were obtained from GSE76938 by GEO2R. Using the FunRich Venn function, 105 hypomethylation-high expression genes and 561 hypermethylation-low expression genes were identified, as revealed in Fig. 2.

GO term enrichment analysis

The top six GO annotation results of aberrantly methylated-differentially expressed genes are summarized in Table I. The ontology enrichment analysis of hypomethylation-high expression genes is presented in Fig. 3 in the form of cluster chord diagrams. The diagrams of cluster chord A-D, E-F and I-L respectively show the biological processes, cellular components and molecular functions of GO analysis. The four columns of chord diagrams include the first column including A, E and I, the second column including B, F and J, the third column including C, G and K, and the fourth column including D, H and L respectively enriched the GO of GSE47915, GSE55945, GSE69223 and GSE769938 datasets. Similarly, Fig. 4 reveals the enrichment of hypermethylation-low expression genes. Among the hypomethylation-high expression genes, the biological processes (BP) were mainly associated with translational initiation, translation, SRP-dependent co-translational protein targeting to membrane, ribosome biogenesis, regulation of oxidative stress-induced intrinsic apoptotic signaling pathways, and viral transcription. The major cell components (CC) included: Cytosol, large ribosomal subunit, extracellular exosome, cytosolic large ribosomal subunit, mitochondrial membrane, and neuronal cell body, with the most important being the cytosol. The molecular functions (MF) were primarily focused on GTPase activator activity, ribosome structure, protein histidine kinase activity, protein homodimerization activity, protein binding, and aldehyde dehydrogenase [NAD(P)+] activity.

Table I.

GO analysis of aberrantly methylated-differentially expressed genes in prostate cancer.

Table I.

GO analysis of aberrantly methylated-differentially expressed genes in prostate cancer.

A, Hypomethylation and high expression

GO analysisTermGene countPercentage (%)P-value
BPTranslational initiation54.76 5.15×10−3
BPTranslation65.71 9.21×10−3
BPSRP-dependent co-translational protein targeting to membrane43.81 1.21×10−2
BPRibosome biogenesis32.86 1.43×10−2
BPRegulation of oxidative stress-induced intrinsic apoptotic signaling pathway21.90 1.51×10−2
BPViral transcription43.81 1.92×10−2
CCCytosol3432.38 1.94×10−5
CCLarge ribosomal subunit32.86 2.07×10−3
CCExtracellular exosome2523.81 3.56×10−3
CCCytosolic large ribosomal subunit43.81 4.49×10−3
CCMitochondrial membrane43.81 1.10×10−2
CCNeuronal cell body65.71 1.91×10−2
MFGTPase activator activity76.67 2.67×10−3
MFStructural constituent of ribosome65.71 5.00×10−3
MFProtein histidine kinase activity21.90 9.93×10−3
MFProtein homodimerization activity109.52 1.03×10−2
MFProtein binding5552.38 1.56×10−2
MFAldehyde dehydrogenase [NAD(P)+] activity21.90 2.95×10−2

B, Hypermethylation and low expression

BPCell adhesion396.96 2.49×10−8
BPExtracellular matrix organization234.11 1.46×10−7
BPResponse to hypoxia183.21 2.09×10−5
BPMuscle contraction142.50 2.24×10−5
BPPositive regulation of cell migration183.21 4.97×10−5
BPRegulation of phosphatidylinositol 3-kinase signaling111.96 1.25×10−4
CCFocal adhesion539.46 6.76×10−20
CCZ disc223.93 3.87×10−11
CCProteinaceous extracellular matrix325.71 1.08×10−10
CCExtracellular matrix305.36 2.00×10−8
CCActin cytoskeleton254.46 3.70×10−8
CCCytoskeleton325.71 2.49×10−7
MFActin binding285.00 1.03×10−7
MFIntegrin binding152.68 3.34×10−6
MFCollagen binding111.96 1.15×10−5
MFActin filament binding152.68 4.73×10−5
MFSignal transducer activity173.04 4.92×10−4
MFScaffold protein binding81.43 5.61×10−4

[i] GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function.

For the hypermethylation-low expression genes, cell adhesion, extracellular matrix organization, response to hypoxia, muscle contraction, positive regulation of cell migration, and regulation of phosphatidylinositol 3-kinase signaling were the predominant BP. The CC of these genes were primarily distributed in focal adhesions, Z discs, proteinaceous extracellular matrices, other extracellular matrices, and the cytoskeleton. The MF mainly included actin binding, integrin binding, collagen binding, actin filament binding, signal transducer activity, and scaffold protein binding.

Selection of PPI network node genes

FunRich was used to predict the relationships between genes and proteins of the hypomethylation-high expression and hypermethylation-low expression gene groups. Subsequently, their interaction networks were visualized by Cytoscape v3.5.0 software as revealed Fig. 5A and B. Cytoscape is a software focused on open source web visualization and analysis. Its core is to provide the basic functional layout and query network, and is based on the combination of basic data into a visual network. Derived from systems biology, Cytoscape is used to integrate biomolecular interaction networks with high-throughput gene expression data and other molecular state information. Its most powerful function is for the analysis of large-scale PPIs, protein-DNA and genetic interactions (21). The top ten node genes in the hypomethylation-high expression and hypermethylation-low expression gene groups are listed separately in Tables II and III, based on the degree of distribution of the nodes.

Table II.

Top ten node genes in the hypomethylation-high expression genes.

Table II.

Top ten node genes in the hypomethylation-high expression genes.

NameDegree
ESR1234
NPM181
NME277
NME1-NME264
UBQLN147
CTBP145
TRIM2741
CCT340
ERBB335
P4HB33

Table III.

Top ten node genes in the hypermethylation-low expression genes.

Table III.

Top ten node genes in the hypermethylation-low expression genes.

NameDegree
CREBBP160
SMAD3159
MCC142
VIM92
ATXN179
CAV168
FLNA68
MAP3K1455
PRKCB53
FHL252
Validation of the node genes

To further validate the present results, TCGA database, the GEPIA tool, and the HPA database were employed. Venn diagram analysis of the GEO database and TCGA database is presented in Fig. 6, from which it can be observed that most of the aberrantly methylated and expressed genes in GEO were contained within TCGA dataset. The methylation and gene expression status of the top ten node genes were similar between GEO and TCGA databases, as revealed in Tables IV and V. Using the GEPIA tool and HPA database, the expression of node genes in tumor and normal prostate samples were further verified and the immunohistochemical staining combined with patient information, node gene expression, and the mRNA levels, respectively, were obtained, as revealed in Figs. 7 and 8 and Tables II and III. In Figs. 7 and 8, immunohistochemical images and data were generated by the online GEPIA tool, log2(TPM+1) for log-scale was used to express gene expression on the y-axis. Red and gray were set as the colors of tumor and normal data sets respectively. The size of jitter across the box was as 0.4. It was revealed that the expression of node genes was generally consistent with previous results.

Table IV.

Validation of the top ten hypomethylation-high expression genes in TCGA database.

Table IV.

Validation of the top ten hypomethylation-high expression genes in TCGA database.

GeneMethylation statusP-valueExpression statusP-value
ESR1 Hypomethylation 3.70×10−143Upregulated 1.20×10−1
NPM1 Hypomethylation 5.51×10−119Upregulated 3.12×10−15
NME2 Hypomethylation 2.76×10−119Upregulated 1.71×10−5
NME1-NME2 Hypomethylation 6.35×10−132Upregulated 6.43×10−9
UBQLN1 Hypomethylation 4.24×10−117Upregulated 8.87×10−1
CTBP1 Hypomethylation 1.85×10−126Upregulated 2.01×10−7
TRIM27 Hypomethylation 5.11×10−129Upregulated 5.39×10−27
CCT3 Hypomethylation 1.92×10−129Upregulated 2.40×10−18
ERBB3 Hypomethylation 1.46×10−121Upregulated 2.89×10−18
P4HB Hypomethylation 2.12×10−119Upregulated 4.29×10−18

[i] TCGA, The Cancer Genome Atlas.

Table V.

Validation of the top ten hypermethylation-low expression genes in TCGA database.

Table V.

Validation of the top ten hypermethylation-low expression genes in TCGA database.

GeneMethylation statusP-valueExpression statusP-value
CREBBP Hypermethylation 1.06×10−123Downregulated 1.64×10−1
SMAD3 Hypermethylation 6.27×10−121Downregulated 1.34×10−10
MCC Hypermethylation 2.39×10−126Downregulated 2.13×10−20
VIM Hypermethylation 1.11×10−134Downregulated 1.26×10−5
ATXN1 Hypermethylation 3.54×10−134Downregulated 2.13×10−4
CAV1 Hypermethylation 1.31×10−133Downregulated 7.54×10−26
FLNA Hypermethylation 8.64×10−126Downregulated 1.12×10−15
MAP3K14 Hypermethylation 2.39×10−123Downregulated 1.79×10−7
PRKCB Hypermethylation 1.24×10−133Downregulated 6.56×10−20
FHL2 Hypermethylation 1.74×10−123Downregulated 1.71×10−12

[i] TCGA, The Cancer Genome Atlas.

Survival analysis of the node genes

In addition, the Disease-Free Survival (RFS) method of GEPIA online tool was used to analyze 20 aberrantly-methylated genes. Group Cutoff was set as Quartile (Cutoff-High (%) was 75% and Cutoff-Low (%) was 25%). The CI was 95%. Hypomethylated-high expression genes (Fig. 9) and hypermethylated-low expression genes (Fig. 10) were respectively represented in red and blue colour. It was revealed that the survival analysis of the P4HB gene exhibited a statistically significant difference in survival between cancer and normal groups (log-rank P-value 0.022).

P4HB is a key protein of disulfide isomerase (protein di-sulphideisomerase, PDI), which is also referred to as a post-translational modifications collagen synthase, that is involved in antioxidant and detoxification reactions. Its encoding gene is on chromosome 17 q25 (22,23). P4HB was revealed to be highly expressed in glioblastoma and liver cancer (24,25). Notably, Xia et al (25) revealed that high expression of P4HB in liver cancer tissues was associated with poor survival.

Discussion

In recent years, DNA methylation is a major part of epigenetic modification and plays an important role in maintaining chromosome stability and gene expression in mammals. It refers to the process of transferring methyl groups to specific bases catalyzed by DNA methyl transfer enzymes (DNMTs) using s-adenosine methionine (SAM) as the methyl donor. Increasing studies (26) have revealed that aberrant DNA methylation is intimately associated with tumorigenesis. It was revealed that the overall methylation level of DNA in tumor cells was lower than that in normal cells, however some specific gene CpG islands were hypermethylated (27). DNA hypomethylation activates proto-oncogenes and abnormal proliferation of cancer cells. However, hypermethylation of CpG islands in the promoter region can inhibit gene expression and inactivate tumor suppressor genes, thus promoting the occurrence and development of tumors. Therefore, methylation is considered to be another mechanism of tumorigenesis (28). DNA methylation is not only involved in the regulation of the cell cycle, proliferation, apoptosis and metastasis, but also in the regulation of drug resistance and intracellular signal transduction pathway of tumor cells (29).

At present, some progress has been made in genetics and in the molecular pathogenesis of PCa, however effective diagnosis and treatment of PCa still require further advances (30). In the present study, the gene expression and methylation datasets of PCa were analyzed using a variety of online tools. A total of 105 hypomethylation-high expression genes and 561 hypermethylation-low expression genes were obtained.

GO annotations of the hypomethylation-high expression gene group in PCa predominantly included translational initiation, translation, SRP-dependent co-translational protein targeting to the membrane, ribosome biogenesis, regulation of oxidative stress-induced intrinsic apoptotic signaling pathways, viral transcription, GTPase activator activity, structural constituents of ribosomes, protein histidine kinase activity, protein homodimerization activity, protein binding, and aldehyde dehydrogenase [NAD(P)+] activity. Multiple translation initiation factors were regularly magnified or diminished in tumors, promoting proliferation, survival, angiogenesis, and metastasis (31,32). Ribosome biogenesis is a process closely correlated to cell growth and proliferation, which are upregulated in the vast majority of cancers including PCa. This leads to downregulation of the expression and activity of p53, and thus promotes tumorigenesis (33,34). As a GTPase activator, DOCK4 can promote intercellular adhesion by activating Rap GTPase (35).

According to the PPI network for the hypomethylation-high expression gene group constructed by Cytoscape, the node degree of each gene was obtained and the top 10 node genes were selected, including ESR1, NPM1, NME2, NME1-NME2, UBQLN1, CTBP1, TRIM27, CCT3, ERBB3, and P4HB. Toy et al (36) suggested that activation of the ESR1 ligand binding domain mutations had different effects on the efficacy of estrogen receptor antagonists. Nucleophosmin (NPM1) is generally overexpressed, mutated, and rearranged in cancer and has been revealed to be overexpressed in PCa (37,38). In addition, NPM1 was revealed to participate in the progression, invasion, and metastasis of tumors in high-grade serous ovarian adenocarcinoma (39). Underexpression of NME2 can inhibit the metastasis of lung cancer and other studies concluded that hypomethylation and high expression of NME2 can cause apoptosis of the mouse testes cells (40,41). UBQLN1 was revealed to be overexpressed in gastric cancer. Reduced UBQLN1 expression can cause enhanced cell migration and invasion, actin cytoskeleton rearrangements, and stimulation of the epithelial-mesenchymal transition (EMT) (42,43). Scholars have revealed that CTBP1 is carcinogenic in PCa and other adenomas, and overexpression of CTBP1 is thought to be involved in cell survival, proliferation, migration, invasion, and EMT (4446). TRIM27 was initially considered to be part of a fusion gene RET (rearranged during transfection), which is a proto-oncogene that is upregulated in multitudinal cancers, including PCa (47,48). CCT3 was significantly correlated with cancer cell proliferation and was revealed to be upregulated in osteosarcoma and hepatocellular carcinoma (4951). ERBB3 is the only gene identified whose impaired kinase domain may be involved in the progression and invasion of PCa (52,53). P4HB was overexpressed in colon cancer and downregulation of expression may promote cancer cell apoptosis (54).

For the hypermethylation-low expression gene group in this PCa study, the BP revealed from the GO annotations were associated with cell adhesion, extracellular matrix organization, response to hypoxia, muscle contraction, positive regulation of cell migration, and regulation of phosphatidylinositol 3-kinase signaling. Loss of cell adhesion is one of the critical steps in tumor progression (55). Several studies have demonstrated that the extracellular matrix, hypoxia, cell migration, and the phosphatidylinositol 3-kinase signaling pathway may play crucial roles in cancer metastasis (5658). Furthermore, the phosphatidylinositol 3-kinase signaling pathway is involved in tumorigenesis and progression (58). The molecular functions have primarily focused on actin, integrin, collagen, scaffold protein, and actin filament binding, as well as signal transducer activity. Previous studies have revealed that actin-binding proteins have inhibitory roles in PCa cell growth and metastasis (59,60). Integrins play a pivotal role in cell adhesion by connecting the cytoskeleton and extracellular matrix, promoting tumor cell proliferation, metastasis, and invasion (61,62).

In the present study, the top 10 hypermethylation-low expression node genes that were obtained from the PPI network included CREBBP, SMAD3, MCC, VIM, ATXN1, CAV1, FLNA, MAP3K14, PRKCB, and FHL2. There is evidence that CREBBP is downregulated in PCa and is a tumor suppressor gene in small-cell lung cancer and other neuroendocrine tumors (63,64). Smad3/Sox5/Twist1 was reported to promote EMT and cancer progression in PCa (65). MCC, a candidate tumor suppressor gene, was revealed to be often silenced by hypermethylation of its promoter in colorectal cancer (66). Several studies have revealed that the EMT marker, VIM, was positively associated with bone metastasis in PCa, and its promoter is often hypermethylated in cervical cancer cells (67,68). It has been reported that downregulation of ATXN1 induced EMT in cervical cancer, but its role in PCa remains to be elucidated (69). Studies have revealed that CAV1 can promote growth and metastasis of PCa, and the hypermethylation of its promoter could make CAV1 a tumor suppressor gene in PCa (7072). The results of FLNA expression are consistent with those of down-regulation in PCa, indicating that FLNA may play a critical role as a negative regulator in PCa (73). MAP3K14, also known as NIK, is a regulatory component of the mitochondrial division machinery and has a dominant effect on cancer cell invasion (74). PRKCB is a member of the PKC family, and when its expression is downregulated, cell apoptosis and suppression of tumorigenesis occur (75,76). A previous study reported that FHL2 could be considered as a PCa biomarker (77).

GEPIA is a TCGA database-based online tool that can be used to study the prognostic effects of genes in various cancers. Survival analysis was conducted on 20 aberrantly-methylated genes which exhibit potential as diagnostic markers of PCa using GEPIA. Notably, the expression of P4HB was the only one among the 20 genes that was significantly correlated with survival. P4HB is found to be highly expressed in glioblastoma and liver cancer, decreasing the apoptosis of tumor cells (24,25). Xia et al (25) have revealed that high expression of P4HB in liver cancer tissues was associated with poor survival. However, there is also evidence suggesting that the low expression of P4HB could lead to proteasome inhibition-induced autophagy (78), since the boundary between apoptosis and autophagy has always been controversial due to tumor heterogeneity (79). Based on the present results revealing that the patients with hypermethylation and low expression of P4HB had poor prognosis, it was hypothesized that hypermethylation of P4HB in prostate cancer would activate autophagy of prostate cancer cells and promote the tumor progression. Further experiments are required to verify the hypothesis.

To validate the present results, TCGA database, the GEPIA tool, and the HPA database were employed to determine node gene methylation and gene expression status, and the results were generally consistent with our previous results, revealing the reliability of this study. However, there are still several limitations in the present study, including the focus on data mining and analysis without experimental confirmation. In addition, the methylation status of the node genes was only verified in TCGA database. Therefore, further experiments are required to verify our results.

In conclusion, the present study identified 20 aberrantly methylated-differentially expressed genes that may be used as biomarkers in PCa. The bioinformatics approach included the analysis of both gene expression and gene methylation microarrays, providing a novel and practical approach for the diagnosis, treatment, and prognosis of PCa. However, based on the limitations of the present study, additional molecular biologal experiments are required to confirm these findings.

It is anticipated that further study on the relationship between DNA methylation and histone codes and their mutual regulatory mechanism could provide new breakthroughs in the gene regulation of tumor genesis. Prostate cancer has high morbidity and mortality, however its pathogenesis has not been fully elucidated, and its therapeutic effect is not satisfactory. The study of the relationship between DNA methylation and histone codes is crucial to improve understanding of some signaling pathways which widely participate in cell physiology and pathology, making subtle adjustments to the complex signaling network, yet there have been few studies conducted on its clinical application. With the deeper understanding of the role of DNA methylation in tumor and the increasing number of high-throughput data of DNA methylation in tumors, bioinformatics technology has laid the foundation for the early diagnosis of tumors, the search for molecular markers for prognosis evaluation and new tumor therapeutic targets.

In the present study, high throughput microarray data of biological databases was mined by bioinformatics technology, to the best of our knowledge for the first time, to determine the aberrantly-methylated biomarkers of prostate cancer. The strategy applied was the selection of the intersecting high methylation and low expression genes as biological markers. The low methylation and high expression genes were analyzed by the same procedure. The markers were further enriched to display their related signaling pathways. Thus, the carcinogenic mechanism of prostate cancer could be clarified, to provide theories for clinical research and treatment.

Acknowledgements

Not applicable.

Funding

No funding was received.

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

ZQ conceived and designed the present study. LW and BW collected, extracted and analyzed the data. LW and BW wrote the manuscript. LW and ZQ reviewed the final manuscript. All authors read and approved the final manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

PCa

prostate cancer

DEGs

differentially expressed genes

GEO

Gene Expression Omnibus

PPI

protein-protein interaction

TCGA

The Cancer Genome Atlas

GEPIA

Gene Expression Profiling Interactive Analysis

HPA

Human Protein Atlas

NCBI

National Center for Biotechnology Information

DMGs

differentially methylated genes

GO

Gene Ontology

DAVID

Database for Annotation, Visualization, and Integrated Discovery

BP

biological processes

EMT

epithelial-mesenchymal transition

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. 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 

Feinberg AP, Ohlsson R and Henikoff S: The epigenetic progenitor origin of human cancer. Nat Rev Genet. 7:21–33. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Nephew KP and Huang TH: Epigenetic gene silencing in cancer initiation and progression. Cancer Lett. 190:125–133. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Nakao M: Epigenetics: Interaction of DNA methylation and chromatin. Gene. 278:25–31. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Strahl BD and Allis CD: The language of covalent histone modifications. Nature. 403:41–45. 2000. View Article : Google Scholar : PubMed/NCBI

7 

De Carvalho DD, Sharma S, You JS, Su SF, Taberlay PC, Kelly TK, Yang X, Liang G and Jones PA: DNA methylation screening identifies driver epigenetic events of cancer cell survival. Cancer Cell. 21:655–667. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Jones PA and Baylin SB: The fundamental role of epigenetic events in cancer. Nat Rev Genet. 3:415–428. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Mundbjerg K, Chopra S, Alemozaffar M, Duymich C, Lakshminarasimhan R, Nichols PW, Aron M, Siegmund KD, Ukimura O, Aron M, et al: Identifying aggressive prostate cancer foci using a DNA methylation classifier. Genome Biol. 18:32017. View Article : Google Scholar : PubMed/NCBI

10 

Xia D, Wang D, Kim SH, Katoh H and DuBois RN: Prostaglandin E2 promotes intestinal tumor growth via DNA methylation. Nat Med. 18:224–226. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Iizuka N, Oka M, Yamada-Okabe H, Nishida M, Maeda Y, Mori N, Takao T, Tamesa T, Tangoku A, Tabuchi H, et al: Oligonucleotide microarray for prediction of early intrahepatic recurrence of hepatocellular carcinoma after curative resection. Lancet. 361:923–929. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Kulasingam V and Diamandis EP: Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat Clin Pract Oncol. 5:588–599. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Verma M, Khoury MJ and Ioannidis JP: Opportunities and challenges for selected emerging technologies in cancer epidemiology: Mitochondrial, epigenomic, metabolomic, and telomerase profiling. Cancer Epidemiol Biomarkers Prev. 22:189–200. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Lu W and Ding Z: Identification of key genes in prostate cancer gene expression profile by bioinformatics. Andrologia. 51:e131692019. View Article : Google Scholar : PubMed/NCBI

15 

Ye Y, Li SL and Wang SY: Construction and analysis of mRNA, miRNA, lncRNA, and TF regulatory networks reveal the key genes associated with prostate cancer. PLoS One. 13:e01980552018. View Article : Google Scholar : PubMed/NCBI

16 

He Z, Tang F, Lu Z, Huang Y, Lei H, Li Z and Zeng G: Analysis of differentially expressed genes, clinical value and biological pathways in prostate cancer. Am J Transl Res. 10:1444–1456. 2018.PubMed/NCBI

17 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res. 41 (Database Issue):D991–D995. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Pathan M, Keerthikumar S, Ang CS, Gangoda L, Quek CY, Williamson NA, Mouradov D, Sieber OM, Simpson RJ, Salim A, et al: FunRich: An open access standalone functional enrichment and interaction network analysis tool. Proteomics. 15:2597–2601. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Lin P, He RQ, Dang YW, Wen DY, Ma J, He Y, Chen G and Yang H: An autophagy-related gene expression signature for survival prediction in multiple cohorts of hepatocellular carcinoma patients. Oncotarget. 9:17368–17395. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Morris JH, Wu A, Yamashita RA, Marchler-Bauer A and Ferrin TE: cddApp: A Cytoscape app for accessing the NCBI conserved domain database. Bioinformatics. 31:134–136. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Galligan JJ and Petersen DR: The human protein disulfide isomerase gene family. Hum Genomics. 6:62012. View Article : Google Scholar : PubMed/NCBI

23 

Pajunen L, Jones TA, Goddard A, Sheer D, Solomon E, Pihlajaniemi T and Kivirikko KI: Regional assignment of the human gene coding for a multifunctional polypeptide (P4HB) acting as the beta-subunit of prolyl 4-hydroxylase and the enzyme protein disulfide isomerase to 17q25. Cytogenet Cell Genet. 56:165–168. 1991. View Article : Google Scholar : PubMed/NCBI

24 

Goplen D, Wang J, Enger PØ, Tysnes BB, Terzis AJ, Laerum OD and Bjerkvig R: Protein disulfide isomerase expression is related to the invasive properties of malignant glioma. Cancer Res. 66:9895–9902. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Xia W, Zhuang J, Wang G, Ni J, Wang J and Ye Y: P4HB promotes HCC tumorigenesis through downregulation of GRP78 and subsequent upregulation of epithelial-to-mesenchymal transition. Oncotarget. 8:8512–8521. 2017.PubMed/NCBI

26 

Li Y and Tollefsbol TO: Impact on DNA methylation in cancer prevention and therapy by bioactive dietary components. Curr Med Chem. 17:2141–2151. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Ehrlich M: DNA methylation in cancer: Too much, but also too little. Oncogene. 21:5400–5413. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Davis CD and Uthus EO: DNA methylation, cancer susceptibility, and nutrient interactions. Exp Biol Med (Maywood). 229:988–995. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Toyota M, Itoh F and Imai K: DNA methylation and gastrointestinal malignancies: Functional consequences and clinical implications. J Gastroenterol. 35:727–734. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Bjurlin MA and Taneja SS: Prostate cancer. Urol Clin North Am. 44:xv–xvi. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Ruggero D: Translational control in cancer etiology. Cold Spring Harb Perspect Biol. 5(pii): a0123362013.PubMed/NCBI

32 

Chu J, Cargnello M, Topisirovic I and Pelletier J: Translation initiation factors: Reprogramming protein synthesis in cancer. Trends Cell Biol. 26:918–933. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Ray S, Johnston R, Campbell DC, Nugent S, McDade SS, Waugh D and Panov KI: Androgens and estrogens stimulate ribosome biogenesis in prostate and breast cancer cells in receptor dependent manner. Gene. 526:46–53. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Derenzini M, Montanaro L and Trerè D: Ribosome biogenesis and cancer. Acta Histochem. 119:190–197. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Yajnik V, Paulding C, Sordella R, McClatchey AI, Saito M, Wahrer DC, Reynolds P, Bell DW, Lake R, van den Heuvel S, et al: DOCK4, a GTPase activator, is disrupted during tumorigenesis. Cell. 112:673–684. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Toy W, Weir H, Razavi P, Lawson M, Goeppert AU, Mazzola AM, Smith A, Wilson J, Morrow C, Wong WL, et al: Activating ESR1 mutations differentially affect the efficacy of ER antagonists. Cancer Discov. 7:277–287. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Box JK, Paquet N, Adams MN, Boucher D, Bolderson E, O'Byrne KJ and Richard DJ: Nucleophosmin: From structure and function to disease development. BMC Mol Biol. 17:192016. View Article : Google Scholar : PubMed/NCBI

38 

Destouches D, Sader M, Terry S, Marchand C, Maillé P, Soyeux P, Carpentier G, Semprez F, Céraline J, Allory Y, et al: Implication of NPM1 phosphorylation and preclinical evaluation of the nucleoprotein antagonist N6L in prostate cancer. Oncotarget. 7:69397–69411. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Fan X, Wen L, Li Y, Lou L, Liu W and Zhang J: The expression profile and prognostic value of APE/Ref-1 and NPM1 in high-grade serous ovarian adenocarcinoma. APMIS. 125:857–862. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Thakur RK, Yadav VK, Kumar A, Singh A, Pal K, Hoeppner L, Saha D, Purohit G, Basundra R, Kar A, et al: Non-metastatic 2 (NME2)-mediated suppression of lung cancer metastasis involves transcriptional regulation of key cell adhesion factor vinculin. Nucleic Acids Res. 42:11589–11600. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Gu Y, Xu W, Nie D, Zhang D, Dai J, Zhao X, Zhang M, Wang Z, Chen Z and Qiao Z: Nicotine induces Nme2-mediated apoptosis in mouse testes. Biochem Biophys Res Commun. 472:573–579. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Bao J, Jiang X, Zhu X, Dai G, Dou R, Liu X, Sheng H, Liang Z and Yu H: Clinical significance of ubiquilin 1 in gastric cancer. Medicine. 97:e97012018. View Article : Google Scholar : PubMed/NCBI

43 

Shah PP, Lockwood WW, Saurabh K, Kurlawala Z, Shannon SP, Waigel S, Zacharias W and Beverly LJ: Ubiquilin1 represses migration and epithelial-to-mesenchymal transition of human non-small cell lung cancer cells. Oncogene. 34:1709–1717. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Porretti J, Dalton GN, Massillo C, Scalise GD, Farré PL, Elble R, Gerez EN, Accialini P, Cabanillas AM, Gardner K, et al: CLCA2 epigenetic regulation by CTBP1, HDACs, ZEB1, EP300 and miR-196b-5p impacts prostate cancer cell adhesion and EMT in metabolic syndrome disease. Int J Cancer. 143:897–906. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Phelps RA, Chidester S, Dehghanizadeh S, Phelps J, Sandoval IT, Rai K, Broadbent T, Sarkar S, Burt RW and Jones DA: A two-step model for colon adenoma initiation and progression caused by APC loss. Cell. 137:623–634. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Blevins MA, Huang M and Zhao R: The role of CtBP1 in oncogenic processes and its potential as a therapeutic target. Mol Cancer Ther. 16:981–990. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Ma Y, Wei Z, Bast RC Jr, Wang Z, Li Y, Gao M, Liu Y and Wang X, Guo C, Zhang L and Wang X: Downregulation of TRIM27 expression inhibits the proliferation of ovarian cancer cells in vitro and in vivo. Lab Invest. 96:37–48. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Shaikhibrahim Z, Lindstrot A, Ochsenfahrt J, Fuchs K and Wernert N: Epigenetics-related genes in prostate cancer: Expression profile in prostate cancer tissues, androgen-sensitive and -insensitive cell lines. Int J Mol Med. 31:21–25. 2013.PubMed/NCBI

49 

Zhang Y, Wang Y, Wei Y, Wu J, Zhang P, Shen S, Saiyin H, Wumaier R, Yang X, Wang C and Yu L: Molecular chaperone CCT3 supports proper mitotic progression and cell proliferation in hepatocellular carcinoma cells. Cancer Lett. 372:101–109. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Xiong Y, Wu S, Du Q, Wang A and Wang Z: Integrated analysis of gene expression and genomic aberration data in osteosarcoma (OS). Cancer Gene Ther. 22:524–529. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Cui X, Hu ZP, Li Z, Gao PJ and Zhu JY: Overexpression of chaperonin containing TCP1, subunit 3 predicts poor prognosis in hepatocellular carcinoma. World J Gastroenterol. 21:8588–8604. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Jaiswal BS, Kljavin NM, Stawiski EW, Chan E, Parikh C, Durinck S, Chaudhuri S, Pujara K, Guillory J, Edgar KA, et al: Oncogenic ERBB3 mutations in human cancers. Cancer Cell. 23:603–617. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Koumakpayi IH, Le Page C, Delvoye N, Saad F and Mes-Masson AM: Macropinocytosis inhibitors and Arf6 regulate ErbB3 nuclear localization in prostate cancer cells. Mol Carcinog. 50:901–912. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Zhou Y, Yang J, Zhang Q, Xu Q, Lu L, Wang J and Xia W: P4HB knockdown induces human HT29 colon cancer cell apoptosis through the generation of reactive oxygen species and inactivation of STAT3 signaling. Mol Med Rep. 19:231–237. 2019.PubMed/NCBI

55 

Le Bras GF, Taubenslag KJ and Andl CD: The regulation of cell-cell adhesion during epithelial-mesenchymal transition, motility and tumor progression. Cell Adh Migr. 6:365–373. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Gilkes DM, Semenza GL and Wirtz D: Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat Rev Cancer. 14:430–439. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Jacquemet G, Hamidi H and Ivaska J: Filopodia in cell adhesion, 3D migration and cancer cell invasion. Curr Opin Cell Biol. 36:23–31. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Liu X, Xu Y, Zhou Q, Chen M, Zhang Y, Liang H, Zhao J, Zhong W and Wang M: PI3K in cancer: Its structure, activation modes and role in shaping tumor microenvironment. Future Oncol. 14:665–674. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Haffner MC, Esopi DM, Chaux A, Gürel M, Ghosh S, Vaghasia AM, Tsai H, Kim K, Castagna N, Lam H, et al: AIM1 is an actin-binding protein that suppresses cell migration and micrometastatic dissemination. Nat Commun. 8:1422017. View Article : Google Scholar : PubMed/NCBI

60 

Vanaja DK, Grossmann ME, Cheville JC, Gazi MH, Gong A, Zhang JS, Ajtai K, Burghardt TP and Young CY: PDLIM4, an actin binding protein, suppresses prostate cancer cell growth. Cancer Invest. 27:264–272. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Ramovs V, Te Molder L and Sonnenberg A: The opposing roles of laminin-binding integrins in cancer. Matrix Biol. 57-58:213–243. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Desgrosellier JS and Cheresh DA: Integrins in cancer: Biological implications and therapeutic opportunities. Nat Rev Cancer. 10:9–22. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Jia D, Augert A, Kim DW, Eastwood E, Wu N, Ibrahim AH, Kim KB, Dunn CT, Pillai SPS, Gazdar AF, et al: Crebbp loss drives small cell lung cancer and increases sensitivity to HDAC inhibition. Cancer Discov. 8:1422–1437. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Shaikhibrahim Z, Lindstrot A, Buettner R and Wernert N: Analysis of laser-microdissected prostate cancer tissues reveals potential tumor markers. Int J Mol Med. 28:605–611. 2011.PubMed/NCBI

65 

Hu J, Tian J, Zhu S, Sun L, Yu J, Tian H, Dong Q, Luo Q, Jiang N, Niu Y and Shang Z: Sox5 contributes to prostate cancer metastasis and is a master regulator of TGF-β-induced epithelial mesenchymal transition through controlling Twist1 expression. Br J Cancer. 118:88–97. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Kohonen-Corish MR, Sigglekow ND, Susanto J, Chapuis PH, Bokey EL, Dent OF, Chan C, Lin BP, Seng TJ, Laird PW, et al: Promoter methylation of the mutated in colorectal cancer gene is a frequent early event in colorectal cancer. Oncogene. 26:4435–4441. 2007. View Article : Google Scholar : PubMed/NCBI

67 

Lang SH, Hyde C, Reid IN, Hitchcock IS, Hart CA, Bryden AA, Villette JM, Stower MJ and Maitland NJ: Enhanced expression of vimentin in motile prostate cell lines and in poorly differentiated and metastatic prostate carcinoma. Prostate. 52:253–263. 2002. View Article : Google Scholar : PubMed/NCBI

68 

Jung S, Yi L, Kim J, Jeong D, Oh T, Kim CH, Kim CJ, Shin J, An S and Lee MS: The role of vimentin as a methylation biomarker for early diagnosis of cervical cancer. Mol Cells. 31:405–411. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Kang AR, An HT, Ko J and Kang S: Ataxin-1 regulates epithelial-mesenchymal transition of cervical cancer cells. Oncotarget. 8:18248–18259. 2017.PubMed/NCBI

70 

Li L, Yang G, Ebara S, Satoh T, Nasu Y, Timme TL, Ren C, Wang J, Tahir SA and Thompson TC: Caveolin-1 mediates testosterone-stimulated survival/clonal growth and promotes metastatic activities in prostate cancer cells. Cancer Res. 61:4386–4392. 2001.PubMed/NCBI

71 

Tahir SA, Yang G, Goltsov AA, Watanabe M, Tabata K, Addai J, Fattah el MA, Kadmon D and Thompson TC: Tumor cell-secreted caveolin-1 has proangiogenic activities in prostate cancer. Cancer Res. 68:731–739. 2008. View Article : Google Scholar : PubMed/NCBI

72 

Cui J, Rohr LR, Swanson G, Speights VO, Maxwell T and Brothman AR: Hypermethylation of the caveolin-1 gene promoter in prostate cancer. Prostate. 46:249–256. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Sun GG, Lu YF, Zhang J and Hu WN: Filamin A regulates MMP-9 expression and suppresses prostate cancer cell migration and invasion. Tumour Biol. 35:3819–3826. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Jung JU, Ravi S, Lee DW, McFadden K, Kamradt ML, Toussaint LG and Sitcheran R: NIK/MAP3K14 regulates mitochondrial dynamics and trafficking to promote cell invasion. Curr Biol. 26:3288–3302. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Hagiwara K, Ito H, Murate T, Miyata Y, Ohashi H and Nagai H: PROX1 overexpression inhibits protein kinase C beta II transcription through promoter DNA methylation. Genes Chromosomes Cancer. 51:1024–1036. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Surdez D, Benetkiewicz M, Perrin V, Han ZY, Pierron G, Ballet S, Lamoureux F, Rédini F, Decouvelaere AV, Daudigeos-Dubus E, et al: Targeting the EWSR1-FLI1 oncogene-induced protein kinase PKC-β abolishes ewing sarcoma growth. Cancer Res. 72:4494–4503. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Kahl P, Gullotti L, Heukamp LC, Wolf S, Friedrichs N, Vorreuther R, Solleder G, Bastian PJ, Ellinger J, Metzger E, et al: Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res. 66:11341–11347. 2006. View Article : Google Scholar : PubMed/NCBI

78 

Rui YN, Xu Z, Chen Z and Zhang S: The GST-BHMT assay reveals a distinct mechanism underlying proteasome inhibition-induced macroautophagy in mammalian cells. Autophagy. 11:812–832. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Ouyang L, Shi Z, Zhao S, Wang FT, Zhou TT, Liu B and Bao JK: Programmed cell death pathways in cancer: A review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 45:487–498. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 21 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang L, Wang B and Quan Z: Identification of aberrantly methylated‑differentially expressed genes and gene ontology in prostate cancer. Mol Med Rep 21: 744-758, 2020
APA
Wang, L., Wang, B., & Quan, Z. (2020). Identification of aberrantly methylated‑differentially expressed genes and gene ontology in prostate cancer. Molecular Medicine Reports, 21, 744-758. https://doi.org/10.3892/mmr.2019.10876
MLA
Wang, L., Wang, B., Quan, Z."Identification of aberrantly methylated‑differentially expressed genes and gene ontology in prostate cancer". Molecular Medicine Reports 21.2 (2020): 744-758.
Chicago
Wang, L., Wang, B., Quan, Z."Identification of aberrantly methylated‑differentially expressed genes and gene ontology in prostate cancer". Molecular Medicine Reports 21, no. 2 (2020): 744-758. https://doi.org/10.3892/mmr.2019.10876