Delineating the underlying molecular mechanisms and key genes involved in metastasis of colorectal cancer via bioinformatics analysis

  • Authors:
    • Chao Qi
    • Yuanlei Chen
    • Yixuan Zhou
    • Xucheng Huang
    • Guoli Li
    • Jin Zeng
    • Zhi Ruan
    • Xinyou Xie
    • Jun Zhang
  • View Affiliations

  • Published online on: March 8, 2018     https://doi.org/10.3892/or.2018.6303
  • Pages: 2297-2305
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is considered the world's fourth most deadly cancer. Its metastasis is associated with poor prognosis and weakens the effects of treatment. However, the potential molecular mechanisms and key genes involved in CRC metastasis have remained to be comprehensively elucidated. The objective of the present study was to identify the key genes and molecular pathways underlying CRC metastasis. Differentially expressed genes (DEGs) between primary CRC tissues and metastatic CRC were identified by analyzing the GSE2509 dataset from the Gene Expression Omnibus database. Subsequently, the DEGs were subjected to Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analyses via the Database for Annotation, Visualization and Integrated Discovery (DAVID). Next, the top ten hub genes were identified in a protein-protein interaction (PPI) network. Sub-network and pathway enrichment analysis were respectively performed with the plugin MCODE and DAVID. Finally, reverse transcription-quantitative polymerase chain reaction assays were performed to corroborate the expression levels of the top five potential metastasis-associated genes in the clinical samples of CRC patients. A total of 7,384 DEGs were obtained, among which 3,949 were upregulated and 3,435 were downregulated. GO and KEGG enrichment analyses identified numerous possible biological processes and pathways that may have a role in the metastasis of CRC. The leading ten hub genes, recognized from the PPI, were epidermal growth factor receptor (EGFR), Has proto-oncogene GTPase (HRas), Wnt family member 5A (Wnt5a), serine/threonine kinase 1 (Akt1), cyclin-dependent kinase inhibitor 1A (CDKN1a), early growth response 1, Ras homolog family member A, cyclin D1 and Ras-related C3 botulinum toxin substrate 1. Sub-network analysis disclosed the most prominent three modules. Ultimately, EGFR, HRas and Akt1 were verified to be upregulated DEGs, while Wnt5a and CDKN1a were downregulated DEGs when compared with the primary controls. In conclusion, the present study revealed several key genes and relevant molecular mechanisms that may enhance the current understanding of CRC metastasis, making them significant potential foci for the discovery of further CRC treatments.

Introduction

Colorectal cancer (CRC), the world's fourth leading cause of cancer-associated mortality, is the third most common type of malignancy in females and the third most common type in males (13). At present, >1 million patients suffer from CRC, and >600,000/year succumb to it worldwide. Although CRC may occur at any age, the risk is highest for individuals aged >50 years (4).

At present, the standard therapies for CRC are surgical resection, radiotherapy, chemotherapy and targeted therapy (5,6). Metastasis is one of the most critical factors impacting the prognosis of CRC patients and the efficiency of their treatment. Even though the 5-year survival rate for CRC patients without metastasis is high, ~10% of them gradually develop metastasis, which eventually leads to death, rendering the above therapeutic therapies inefficient (7). Furthermore, the majority of patients are diagnosed with CRC at stages when the cancer cells have further metastasized to other tissues and organs in the body (8,9).

CRC patients tend to develop lymph node metastasis and distant metastasis at late stages. Previous investigations have corroborated that changes in various processes, including increased cell proliferation, altered cell metabolism, decreased apoptosis, activated protease systems take part in the development of adenoma-carcinoma metastasis (10). However, the underlying molecular mechanisms and key genes involved in CRC progression or metastasis have remained to be fully elucidated. Consequently, the exposure of these molecular mechanisms and key genes is the most critical step towards preventing metastasis. In recent years, bioinformatics analysis, including high-throughput sequencing technology and protein-protein interactions (PPIs), has provided a deeper understanding of the aberrant genetic pathways involved in cancers (11,12). In the present study, a bioinformatics analysis was performed to identify the underlying molecular mechanisms and key genes involved in the metastasis of CRC.

Initially, differentially expressed genes (DEGs) between primary CRC tissues and metastatic CRC were identified from datasets downloaded from the Gene Expression Omnibus (GEO) online database. Subsequently, function and pathway enrichment analysis were performed on the DEGs. The ten leading hub genes which may be involved in the CRC progression of metastasis were selected from the PPI network. Finally, tumor tissues from four patients with metastatic CRC were subjected to reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assays to verify these five leading potential metastasis-associated DEGs.

Materials and methods

Microarray data processing

The GEO dataset GSE2509, including the mRNA expression profile of 3 SW480 and 3 SW620 cell samples, derived from sequencing and subsequent data analysis by Provenzani et al (13), was downloaded from the National Center of Biotechnology Information GEO database (http://www.ncbi.nlm.nih.gov/geo/). The data had been generated using the GPL96 (HG-U133A) platform (Affymetrix Human Genome U133A Array; Affymetrix; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The SW480 cell line originated from a primary rectal gland carcinoma sample and the SW620 cell line was derived one year later from a lymph node metastasis sample of the same patient.

Screening of DEGs

The raw data were uploaded to the Gene-Cloud of Biotechnology Information online laboratory for further analysis (https://www.gcbi.com.cn/gclib/html/index). DEGs were identified between SW480 and SW620. The threshold for the DEGs was defined with a corrected P-value of <0.05 and a fold change (FC) of >1.2.

Function and pathway enrichment analysis of DEGs

Gene Ontology (GO) analysis is a useful Bioinformatics tool used to annotate information about gene product function and identify characteristic biological attributes by analyzing high-throughput genome data (14,15). The Kyoto Encyclopedia of Genes and Genomes (KEGG) is a tool for the systematic analysis of gene function, performed by linking series of genes with networks of interacting cellular molecules, including complex pathways (16). To facilitate the function and pathway analysis, the DEGs were copied into the Database for Annotation, Visualization and Integrated Discovery (DAVID), to perform the GO and KEGG enrichment analyses. The human genome was chosen as the background list. P<0.05 was considered to indicate a statistically significant difference.

PPI network and module analysis

The PPI is described through an undirected diagram with nodes symbolizing the genes and edges symbolizing the mutual interactions of proteins encoded by the corresponding genes (17). In the present study, all of the DEGs were imported into the Search Tool for the Retrieval of Interacting Genes (STRING; http://www.string-db.org) for analysis and only interactions with a combined score of >0.7 were pasted into the Cytoscape plugin to create the network visualization. Subsequently, the PPI network was subjected to module analysis by using the Plugin MCODE with the default parameters (Degree cutoff ≥2, Node score cutoff ≥2, K-core ≥2 and Max depth =100). Finally, function and pathway enrichment analysis of the DEGs in the most prominent three modules were performed via DAVID.

Verification of the leading five potential metastasis-associated DEGs in clinical tumor tissue samples

In the present study, RT-qPCR assays were used to validate the expression levels of the top five potential metastasis-associated genes with the highest degree of interaction and significance in the PPI network. Total RNA was extracted from metastatic and primary CRC samples from four patients who had been diagnosed with metastatic CRC by the Pathology Department of Sir Run Run Shaw Hospital (Hangzhou, China) from October 2016 to March 2017.

First, the mRNA was extracted from the patients' samples using TRIzol reagent (Thermo Fisher Scientific, Inc.) and then stored at −80°C. A Reverse Transcription System (GoTaq® Real-Time PCR System; Promega Corp., Madison, WI, USA) was employed for the synthesis of complementary DNA according to the manufacturer's instructions. The mRNA expression levels of the key genes were measured by quantitative real-time-PCR using the ABI PRISM 7500 Sequence Detector System (Applied Biosystems; Thermo Fisher Scientific, Inc.), and GAPDH was used as an internal standard. The primers used for qPCR were used as follows: Epidermal growth factor receptor (EGFR) forward, 5′-AGGCACGAGTAACAAGCTCAC-3′ and reverse, 5′-ATGAGGACATAACCAGCCACC-3′; cyclin-dependent kinase inhibitor 1A (CDKN1a) forward, 5′-TGTCCGTCAGAACCCATGC-3′ and reverse, 5′-AAAGTCGAAGTTCCATCGCTC-3′; Wnt family member 5A (Wnt5a) forward, 5′-TTCTTGGTGGTCGCTAGGT-3′ and reverse, 5′-TTCTTTGATGCCTGTCTTCG-3′; Has proto-oncogene GTPase (HRas) forward, 5′-TTTGCCATCAACAACACCA-3′ and reverse, 5′-TCCTGAGCCTGCCGAGAT-3′; serine/threonine kinase 1 (Akt1) forward, 5′-TCCTCCTCAAGAATGATGGCA-3′ and reverse, 5′-GTGCGTTCGATGACAGTGGT-3′; GAPDH forward, 5′-AGACAGCCGCATCTTCTTGT-3′ and reverse, 5′-TGATGGCAACAATGTCCACT-3′. The reaction protocol included heating for 3 min at 95°C, followed by 40 cycles of amplification (5 sec at 95°C and 20 sec at 60°C). qPCR reactions were performed in triplicate. The final results were analyzed via the 2−∆∆Cq method with normalization to GAPDH (18).

Statistical analysis

PCR results are presented as the mean ± standard error of the mean (n=3). Differences between the PCR results for the primary and metastatic tumors were determined by the two-tailed unpaired Student's t-test. Prism 4.0 statistical software (GraphPad Inc., La Jolla, CA, USA) was used for statistical analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of DEGs between the SW480 and SW620 CRC cell lines

With the aim of exploring the mechanisms underlying CRC metastasis, publicly accessible microarray datasets were retrieved from the GEO database. In total, 3 cell lines each of SW480 and SW620 pertaining to primary and secondary CRC, respectively, were screened for DEGs. The specified criteria, including FC >1.2 and P<0.05, were met for 7,384 genes, including 3,949 upregulated genes and 3,435 downregulated genes. The DEGs were displayed in a heat map (Fig. 1) and a volcano plot (Fig. 2).

GO and KEGG pathway enrichment analysis

The online biological classification tool DAVID was then employed to analyze the functions and pathways of the 7,384 DEGs. GO analysis suggested that the DEGs were fully involved in 873 biological processes with the premise of false discovery rate <0.05 and P<0.05. The upregulated DEGs were mainly involved in DNA metabolic processes, responses to DNA damage stimuli, cellular responses to stress and the cell cycle (Fig. 3), while the downregulated DEGs were significantly involved in the Wnt signaling pathway, negative regulation of the canonical Wnt signaling pathway, cell-cell adhesion and negative regulation of apoptotic processes (Fig. 3).

The biological functions of cells are complex processes mediated by numerous molecules and genes. Through KEGG pathway enrichment analysis, the upregulated DEGs were identified to be significantly enriched in purine metabolism, arginine and proline metabolism, the cell cycle and DNA replication, which may be associated with tumor proliferation and migration (Fig. 4). As to the downregulated DEGs, pathways involved in cancer, focal adhesions, adhesion junctions and the Hippo signaling pathway were most significant (Fig. 4).

PPI network and module analysis

To investigate the interactions and acquire the hub genes for potential metastasis-associated DEGs in CRC, all of the DEGs were analysed by using STRING. Subsequently, the genes whose combined score were >0.7 revealed close association between genes and were imported into Cytoscape for further analysis. The PPI network contained 954 nodes and 3,326 interactions (data not shown). The ten leading genes defined as the hub genes were as follows: EGFR, HRas, Akt1, Wnt5a, CDKN1a, early growth response 1, Cd44, Ras homolog family member A, cyclin D1 and Ras-related C3 botulinum toxin substrate 1 (data not shown). Next, the Plugin MCODE was employed to recognize the module genes from the PPI analysis (Fig. 5). Function annotation and pathway analysis of the three most significant modules, performed by DAVID, revealed that the module genes were mainly associated with the cell cycle, the forkhead box (FoxO) signaling pathway, the Wnt signaling pathway, ubiquitin-mediated proteolysis, the ERBB2 signaling pathway and the vascular endothelial growth factor (VEGF) signaling pathway (Tables IIII).

Figure 5.

Top 3 modules from analysis of the PPI network. (A) Module 1, (B) module 2 and (C) module 3. EGFR, epidermal growth factor receptor; HRas, Has proto-oncogene GTPase; Akt1, serine/threonine kinase 1; Wnt5a, Wnt family member 5A; CDKN1a, cyclin-dependent kinase inhibitor 1A; ASB4, ankyrin repeat and SOCS box containing 4; FBXO17, F-box protein 17; CTSZ, cathepsin Z; SYNJ1, synaptojanin 1; LMAN2L, lectin, mannose binding 2 like; UBE2H, ubiquitin conjugating enzyme E2 H; STUB1, STIP1 homology and U-box containing protein 1; REPS2, RALBP1 associated Eps domain containing 2; OCRL, inositol polyphosphate-5-phosphatase; ATG7, autophagy related 7; BET1, Bet1 golgi vesicular membrane trafficking protein; SKP1, S-phase kinase associated protein 1; NECAP1, NECAP endocytosis associated 1; FNBP1, formin binding protein 1; AP2S1, adaptor related protein complex 2 sigma 1 subunit; GOSR2, golgi SNAP receptor complex member 2; CLTA, clathrin light chain A; CLTB, clathrin light chain B; VAMP3, vesicle associated membrane protein 3; TGOLN2, trans-golgi network protein 2; F8, coagulation factor VIII; ITSN1, intersectin 1; ARPC1A, actin related protein 2/3 complex subunit 1A; SMURF2, SMAD specific E3 ubiquitin protein ligase 2; RNF6, ring finger protein 6; KLHL21, kelch like family member 21; ASB6, ankyrin repeat and SOCS box containing 6; UBE2C, ubiquitin conjugating enzyme E2 C; HERC5, HECT and RLD domain containing E3 ubiquitin protein ligase 5; ITCH, itchy E3 ubiquitin protein ligase; NOTCH2, notch 2; HSPB1, heat shock protein family B (small) member 1; PRKCZ, protein kinase C ζ; TJP1, tight junction protein 1; MYLK, myosin light chain kinase; PPI, protein-protein interaction.

Table I.

Functional annotation of the significant module 1.

Table I.

Functional annotation of the significant module 1.

AnalysisTermGenesP-value
GOTERM_BP_DIRECTNegative regulation of epidermal growth factor receptor signaling pathwayAP2S1, CLTA, EGFR 1.1×10−3
ERBB2 signaling pathwayHRas, STUB1, EGFR1.2 ×10−3
Positive regulation of MAPK activityHRas, EGFR 7.8×10−2
KEGG PATHWAYCell cycleSKP1, CDC20, CDKN1a3.6 ×10−2
FoxO signaling pathwayHRas, CDKN1a, EGFR4.2 ×10−2

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; GO, Gene Ontology; BP, biological pro-cesses; MAPK, mitogen-activated protein kinase; FoxO, forkhead box O; EGFR, epidermal growth factor receptor; HRas, Has proto-oncogene GTPase; CDKN1a, cyclin-dependent ki-nase inhibitor 1A; AP2S1, adaptor related protein complex 2 sigma 1 subunit; CLTA, clathrin light chain A; STUB1, STIP1 homology and U-box containing protein 1; CDC20, cell division cycle 20; SKP1, S-phase kinase associated protein 1.

Table III.

Functional annotation of the significant module 3.

Table III.

Functional annotation of the significant module 3.

AnalysisTermGenesP-value
GOTERM_BP_DIRECTRegulation of apoptotic processAkt1, HSPB1, NOTCH2, PRKCZ 1.9×10−4
Negative regulation of oxidative stress-induced intrinsic apoptotic signaling pathwayAkt1, HSPB1 4.2×10−3
Positive regulation of blood vessel endothelial cell migrationAkt1, HSPB1 5.6×10−3
KEGG PATHWAYTight junctionAkt1, PPKCZ, TJP1 3.8×10−3
VEGF signaling pathwayAkt1, HSPB1 4.3×10−2

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; GO, Gene Ontology; BP, biological pro-cesses; VEGF, vascular endothelial growth factor; AKT1, serine/threonine kinase 1; HSPB1, heat shock protein family B (small) member 1; PPKCZ, protein kinase C, zeta; TJP1, tight junction protein 1.

Verification of the five most prominent potential metastasis-associated DEGs through analysis of clinical tumor tissue samples

Every DEG analysed with Cytoscape has an interactional degree with others. The five hub genes that had the highest interactional degree of all DEGs were selected as the most prominent potential metastasis-associated DEGs. RT-qPCR analysis was performed to corroborate the expression levels of five potential metastasis-associated genes (EGFR, HRas, Wnt5a, Akt1 and CDKN1a), identified by the above analyses, in four metastatic and four primary CRC samples. EGFR, HRas and Akt1 were upregulated DEGs and their expression levels were identified to be increased, while Wnt5a and CDKN1a were downregulated DEGs and their expression levels were decreased when compared with those in the primary controls. The results of the RT-qPCR analysis are presented in Fig. 6.

Discussion

CRC is considered to be a histological heterogeneous disease, and genetic aberrations have a conspicuous role in its occurrence, progression and metastasis (19,20). However, the underlying molecular mechanisms and key genes involved in the progression or metastasis of CRC have remained to be fully elucidated. Consequently, exposure of these molecular mechanisms and key genes is the most critical step in preventing metastasis.

In the present study, 7,384 DEGs, including 3,949 upregulated and 3,435 downregulated genes, were identified from available gene expression profiles. GO and KEGG pathway enrichment analyses subsequently revealed that the DEGs were mainly involved in the cell cycle, regulation of apoptosis, purine metabolism, DNA metabolic process, as well as the Hippo and Wnt signaling pathways.

It is in accord with the current knowledge that the abnormal regulation of apoptosis and the cell cycle are the causes for cancer initiation and progression (21,22).

Uric acid is the final breakdown product in the cycle of nucleotide metabolism. Xanthine oxidoreductase (XOR) acts as a catalyst when uric acid is produced from xanthine (23). Several prior studies revealed that the absence of XOR in tumor cells is significantly associated with poor clinical prognosis and reduced survival rates in breast, stomach, ovarian, colorectal and non-small cell lung cancer (2426). A recent study, independent of all these variables, revealed that stage IIIA/IIIB CRC patients with high serum uric acid levels may develop early metastasis (27). The present study indicated that DEGs were enriched in purine metabolism, which may correlate with CRC metastasis.

The Hippo pathway participates in the mediation of cell proliferation, differentiation, growth and apoptosis (28). Its negative regulation is often discovered in various types of malignant human tumor, demonstrating that the Hippo signaling pathway is associated with tumor initiation and progression (29,30). In the intestine, the deregulation of the Hippo pathway may lead to neoplastic growth and migration. Furthermore, it has been demonstrated that interactions among the Hippo, Wnt and EGFR signaling pathways have an effect on the development of CRC (31).

In the present study, GO and KEGG pathway enrichment analysis revealed several possible biological processes and pathways, which may be involved in the initiation and development of CRC associated with metastasis.

The results of the module analysis indicated an association of CRC progression with cell cycle activity, negative regulation of the apoptotic process, as well as the Wnt, EGFR, VEGF and FoxO signaling pathways, which is consistent with the above analyses. In addition, the five leading potential metastasis-associated DEGs (EGFR, HRas, WNT5a, Akt1 and CDKN1a) were involved in the three leading modules.

Wnt5a, located at 3p14.2-p21.1, is known as a non-canonical signaling pathway member of the Wnt family (the Wnt signaling pathway can be classified as canonical Wnt signaling and non-canonical Wnt signaling) (32). Prior studies have observed an elevated expression of Wnt5a in numerous types of cancer and particularly in aggressive tumor cells with greater invasive ability than that of the non-aggressive tumor cells. This delineates the important role of Wnt5a in promoting the progression of tumors through the non-canonical Wnt signaling pathway (33). High expression levels of Wnt5a have been reported to be associated with cancer progression and metastasis in gastric cancer, osteosarcoma and glioblastoma (3436). Of note, the various functions of Wnt5a rely on disparate cellular contexts in diverse cancer types (33). Wnt5a also inhibits the canonical Wnt signaling pathway (37). In breast cancer, Wnt5a has been reported to inhibit metastasis in vivo, as well as cell migration and invasion in vitro (38,39). In addition, downregulation of Wnt5a is significantly linked with the malignant characteristics of primary invasive breast cancer (40). However, Wnt5a was reported to act as a tumor suppressor by obstructing canonical Wnt signaling in CRC (41). Studies focusing on the roles of Wnt5a in the metastatic progression of CRC, determining whether a lower expression of Wnt5a is correlated with a poorer clinical outcome, have been rarely conducted. In the present study, it was discovered that Wnt5a was a downregulated DEG and the expression levels of Wnt5a in primary versus metastatic CRC were varied. GO and KEGG analysis indicated that Wnt5a was significantly enriched in Wnt signaling, which may be the mechanism underlying the higher metastatic rate in CRC patients with decreased expression levels of Wnt5a. Consequently, targeting Wnt5a is likely to be a novel therapeutic strategy for metastatic CRC.

HRas is a member of the Ras superfamily, which comprises KRas, HRas and NRas (4244). Since HRas mutations do not appear frequently as do mutations in KRas, few studies have been performed with this regard. Several previous studies have reported that HRas directly activates the Ras/BRAF/mitogen-activated protein kinase (MAPK) kinase (MEK)/MAPK and phosphoinositide-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway, bypassing EGFR-initiated signaling (4547). The activation of HRas is likewise associated with galectin-1. The inhibitors of galectin-1, including OTX008, and of mTOR, including rapamycin, may nearly completely suppress Hras-mutant cancers (48). A recent study has revealed that HRas mutations have the tendency to affect patients' outcomes (hazard ratio =0.545, 95% confidence interval =0.277–1.073, P=0.079) (49). Furthermore, non-mutated HRas has been identified to be upregulated in CRC cells when compared with normal adjacent tissues. A positive association has been noted between HRas expression levels and the up and downstream signaling factors of EGFR, MEK and extracellular signal-regulated kinase, indicating that HRas overexpression contributes to the carcinogenesis of CRC (50). Despite these studies, HRas expression levels have rarely been assessed with regard to the metastatic progression of CRC, and the connection between HRas and metastasis has remained elusive. In the present study, PPI and module analysis disclosed that HRas is a significant DEG participating in the metastatic progression of CRC. KEGG enrichment analysis revealed that the RAS/BRAF/MEK/MAPK pathway is likely to be an important pathway involved in metastasis. Subsequent RT-qPCR assays performed on tissues of CRC patients further confirmed that high expression levels of HRas were positively associated with CRC metastasis. Although HRas mutations are rare, the upregulation of HRas implies CRC progression. Therefore, the prognostic value of this indication calls for further study.

Proteins of the Akt family, including Akt1, 2 and 3, possess high homology but are expressed by different genes. Akt1, known as an essential molecule in the PI3K/Akt signaling pathway, has a vital role in multiple tumor-associated events, including the promotion of cancer cell proliferation, metabolism and survival (51,52). It is implicated in epithelial-mesenchymal transition and metastasis (53,54). Häggblad Sahlberg et al (53) have confirmed that knockout of Akt1 attenuates metastasis and tumor cell growth by upregulating apoptosis and certain metastasis-inhibitory genes in CRC. In addition, Akt1 directly activates focal adhesion kinase via serine phosphorylation, facilitating the metastasis of pressure-induced CRC (55). In the present study, the PPI network revealed that Akt1 exhibited a high degree of connectivity. In addition, the present KEGG enrichment analysis demonstrated that Akt1 participates in regulating the VEGF signaling pathway, while the latter promotes tumor angiogenesis, development and metastasis (56,57). Of note, previous studies have confirmed that inhibition of Akt1 reduces the secretion of VEGF (58). Furthermore, the upregulated expression levels of Akt1 in certain metastatic versus paired primary CRC samples were confirmed in the present study by RT-qPCR assays, which was consistent with the above analyses. In summary, Akt1 is associated with increased cell invasion and motility in CRC. For CRC patients, a high level of Akt1 may herald advanced colorectal tumor progression and unfavorable outcomes.

EGFR may provide cancer cells with self-sufficient growth ability. EGFR has been reported to be overexpressed in >80% of all CRC patients (59), and to be correlated with a higher metastatic risk (60). Simultaneously, an increased expression of EGFR is associated with tumor progression and a lessened survival time of patients with metastatic CRC (61). In addition, the RAS/BRAF/MEK/MAPK and the PI3K/Akt/mTOR pathway, the two major signaling cascades of the EGFR signaling pathway, are activated by phosphorylated EGFR and have an indispensable role in the progression of CRC (10). EGFR-targeting drugs are the first-line therapy and a powerful strategy for treating CRC patients without Ras mutations based on their molecular profiles (62). In the present study, RT-qPCR assays revealed that the expression levels of EGFR were significantly increased in metastatic tissues, implying the possible involvement of EGFR in CRC metastasis.

The CDKN1a gene, which encodes p21, a protein regulating anti-growth signals, is located on chromosome 6 (6p21.2) (63). The expression levels of CDKN1a, a key intermediary agent of the p53 response, are mediated by p53 in response to innumerable stress-stimulating factors. It then binds to and blocks several CDKs, restraining cell cycle progression at the G1- and S-phase (6466). Suppression of CDKN1a leads to the transformation of colorectal adenoma into a malignant tumor and likewise accelerates the proliferation and metastasis of CRC cells (67). Furthermore, downregulation of CDKN1a has been confirmed to be associated with venous involvement, as well as lymph node and liver metastasis (68,69). The present study, consistent with the abovementioned studies, implied that CDKN1a may be a key DEG in the metastasis of CRC. Targeting this gene may be a novel therapeutic strategy.

In conclusion, the present study identified several hub genes and key pathways participating in the progression and metastasis of CRC. These efforts may contribute towards an improved understanding of the molecular mechanisms underlying the progression of CRC, and provide potential biomarkers for clinical surveillance and therapy. However, as the total number of cases analyzed in the present study was insufficient, there may be some bias; further molecular biology experiments are required to further confirm the specific functions of these identified genes involved in CRC metastasis in the future.

Acknowledgements

Not applicable.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

DEGs

differentially expressed genes

GEO

Gene Expression Omnibus

KEGG

Kyoto Encyclopedia of Genes and Genomes

STRING

Search Tool the Retrieval of Interacting Genes

DAVID

Database for Annotation Visualization and Integrated Discovery

GO

Gene Ontology

PPI

protein-protein interaction

RT-qPCR

reverse transcription-quantitative polymerase chain reaction

EGFR

epidermal growth factor receptor

HRas

Has proto-oncogene GTPase

Akt1

serine/threonine kinase 1

Wnt5a

Wnt family member 5A

CDKN1a

cyclin-dependent kinase inhibitor 1A

XOR

xanthine oxidoreductase

References

1 

Choi Y, Sateia HF, Peairs KS and Stewart RW: Screening for colorectal cancer. Semin Oncol. 44:34–44. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Manku G, Hueso A, Brimo F, Chan P, Gonzalez-Peramato P, Jabado N, Gayden T, Bourgey M, Riazalhosseini Y and Culty M: Changes in the expression profiles of claudins during gonocyte differentiation and in seminomas. Andrology. 4:95–110. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Siegel R, Desantis C and Jemal A: Colorectal cancer statistics, 2014. CA Cancer J Clin. 64:104–117. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Basu A, Seth S, Arora K and Verma M: Evaluating estradiol levels in male patients with colorectal carcinoma. J Clin Diagn Res. 9:BC08–BC10. 2015.PubMed/NCBI

5 

Ciombor KK, Wu C and Goldberg RM: Recent therapeutic advances in the treatment of colorectal cancer. Annu Rev Med. 66:83–95. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Yu H, Gao G, Jiang L, Guo L, Lin M, Jiao X, Jia W and Huang J: Decreased expression of miR-218 is associated with poor prognosis in patients with colorectal cancer. Int J Clin Exp Pathol. 6:2904–2911. 2013.PubMed/NCBI

7 

Zhou C, Cui F, Li J, Wang D, Wei Y, Wu Y, Wang J, Zhu H and Wang S: miR-650 represses high-risk non-metastatic colorectal cancer progression via inhibition of AKT2/GSK3β/E-cadherin pathway. Oncotarget. 8:49534–49547. 2017.PubMed/NCBI

8 

De Rosa M, Pace U, Rega D, Costabile V, Duraturo F, Izzo P and Delrio P: Genetics, diagnosis and management of colorectal cancer (Review). Oncol Rep. 34:1087–1096. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Yamasaki M, Takemasa I, Komori T, Watanabe S, Sekimoto M, Doki Y, Matsubara K and Monden M: The gene expression profile represents the molecular nature of liver metastasis in colorectal cancer. Int J Oncol. 30:129–138. 2007.PubMed/NCBI

10 

Hagland HR, Berg M, Jolma IW, Carlsen A and Søreide K: Molecular pathways and cellular metabolism in colorectal cancer. Dig Surg. 30:12–25. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Gao M, Zhong A, Patel N, Alur C and Vyas D: High throughput RNA sequencing utility for diagnosis and prognosis in colon diseases. World J Gastroenterol. 23:2819–2825. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Serratì S, De Summa S, Pilato B, Petriella D, Lacalamita R, Tommasi S and Pinto R: Next-generation sequencing: Advances and applications in cancer diagnosis. Onco Targets Ther. 9:7355–7365. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Provenzani A, Fronza R, Loreni F, Pascale A, Amadio M and Quattrone A: Global alterations in mRNA polysomal recruitment in a cell model of colorectal cancer progression to metastasis. Carcinogenesis. 27:1323–1333. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Gene Ontology Consortium, . The Gene Ontology (GO) project in 2006. Nucleic Acids Res. 34:D322–D326. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Gene Ontology Consortium, . Gene Ontology Consortium: Going forward. Nucleic Acids Res. 43(D1): D1049–D1056. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28(1): 27–30. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Köhler S, Bauer S, Horn D and Robinson PN: Walking the interactome for prioritization of candidate disease genes. Am J Hum Genet. 82:949–958. 2008. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Aran V, Victorino AP, Thuler LC and Ferreira CG: Colorectal Cancer: Epidemiology, disease mechanisms and interventions to reduce onset and mortality. Clin Colorectal Cancer. 15:195–203. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Taylor-Weiner A, Zack T, O'Donnell E, Guerriero JL, Bernard B, Reddy A, Han GC, AlDubayan S, Amin-Mansour A, Schumacher SE, et al: Genomic evolution and chemoresistance in germ-cell tumours. Nature. 540:114–118. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Chang YT, Tseng HC, Huang CC, Chen YP, Chiang HC and Chou FP: Relative down-regulation of apoptosis and autophagy genes in colorectal cancer. Eur J Clin Invest. 41:84–92. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Perez R, Wu N, Klipfel AA and Beart RW Jr: A better cell cycle target for gene therapy of colorectal cancer: Cyclin G. J Gastrointest Surg. 7:884–889. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Fini MA, Elias A, Johnson RJ and Wright RM: Contribution of uric acid to cancer risk, recurrence, and mortality. Clin Transl Med. 1:162012. View Article : Google Scholar : PubMed/NCBI

24 

Ikegami T, Natsumeda Y and Weber G: Decreased concentration of xanthine dehydrogenase (EC 1.1.1.204) in rat hepatomas. Cancer Res. 46:3838–3841. 1986.PubMed/NCBI

25 

Sun AS and Cederbaum AI: Oxidoreductase activities in normal rat liver, tumor-bearing rat liver, and hepatoma HC-252. Cancer Res. 40:4677–4681. 1980.PubMed/NCBI

26 

Tanriverdi O, Cokmert S, Oktay E, Pilanci KN, Menekse S, Kocar M, Sen CA, Avci N, Akman T, Ordu C, et al: Prognostic significance of the baseline serum uric acid level in non-small cell lung cancer patients treated with first-line chemotherapy: A study of the Turkish Descriptive Oncological Researches Group. Med Oncol. 31:2172014. View Article : Google Scholar : PubMed/NCBI

27 

Cetin AO, Omar M, Calp S, Tunca H, Yimaz N, Ozseker B and Tanriverdi O: Hyperuricemia at the time of diagnosis is a factor for poor prognosis in patients with stage II and III colorectal cancer (uric acid and colorectal cancer). Asian Pac J Cancer Prev. 18:485–490. 2017.PubMed/NCBI

28 

Zhao B, Li L, Lei Q and Guan KL: The Hippo-YAP pathway in organ size control and tumorigenesis: An updated version. Genes Dev. 24:862–874. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Harvey KF, Zhang X and Thomas DM: The Hippo pathway and human cancer. Nat Rev Cancer. 13:246–257. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Liu H, Jiang D, Chi F and Zhao B: The Hippo pathway regulates stem cell proliferation, self-renewal, and differentiation. Protein Cell. 3:291–304. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Wierzbicki PM and Rybarczyk A: The Hippo pathway in colorectal cancer. Folia Histochem Cytobiol. 53:105–119. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Serra R, Easter SL, Jiang W and Baxley SE: Wnt5a as an effector of TGFβ in mammary development and cancer. J Mammary Gland Biol Neoplasia. 16:157–167. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Endo M, Nishita M, Fujii M and Minami Y: Insight into the role of Wnt5a-induced signaling in normal and cancer cells. Int Rev Cell Mol Biol. 314:117–148. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Hu B, Wang Q, Wang YA, Hua S, Sauvé CG, Ong D, Lan ZD, Chang Q, Ho YW, Monasterio MM, et al: Epigenetic activation of WNT5A drives glioblastoma stem cell differentiation and invasive growth. Cell. 167:1281–1295. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Kurayoshi M, Oue N, Yamamoto H, Kishida M, Inoue A, Asahara T, Yasui W and Kikuchi A: Expression of Wnt-5a is correlated with aggressiveness of gastric cancer by stimulating cell migration and invasion. Cancer Res. 66:10439–10448. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Yong BC, Lu JC, Xie XB, Su Q, Tan PX, Tang QL, Wang J, Huang G, Han J, Xu HW, et al: LDOC1 regulates Wnt5a expression and osteosarcoma cell metastasis and is correlated with the survival of osteosarcoma patients. Tumour Biol. 39:10104283176911882017. View Article : Google Scholar : PubMed/NCBI

37 

Mikels AJ and Nusse R: Purified Wnt5a protein activates or inhibits beta-catenin-TCF signaling depending on receptor context. PLoS Biol. 4:e1152006. View Article : Google Scholar : PubMed/NCBI

38 

Prasad CP, Chaurasiya SK, Axelsson L and Andersson T: WNT-5A triggers Cdc42 activation leading to an ERK1/2 dependent decrease in MMP9 activity and invasive migration of breast cancer cells. Mol Oncol. 7:870–883. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Säfholm A, Tuomela J, Rosenkvist J, Dejmek J, Härkönen P and Andersson T: The Wnt-5a-derived hexapeptide Foxy-5 inhibits breast cancer metastasis in vivo by targeting cell motility. Clin Cancer Res. 14:6556–6563. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Jönsson M, Dejmek J, Bendahl PO and Andersson T: Loss of Wnt-5a protein is associated with early relapse in invasive ductal breast carcinomas. Cancer Res. 62:409–416. 2002.PubMed/NCBI

41 

Ying J, Li H, Yu J, Ng KM, Poon FF, Wong SC, Chan AT, Sung JJ and Tao Q: WNT5A exhibits tumor-suppressive activity through antagonizing the Wnt/β-catenin signaling, and is frequently methylated in colorectal cancer. Clin Cancer Res. 14:55–61. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Elbjeirami WM and Sughayer MA: KRAS mutations and subtyping in colorectal cancer in Jordanian patients. Oncol Lett. 4:705–710. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Nakano H, Yamamoto F, Neville C, Evans D, Mizuno T and Perucho M: Isolation of transforming sequences of two human lung carcinomas: Structural and functional analysis of the activated c-K-ras oncogenes. Proc Natl Acad Sci USA. 81:71–75. 1984. View Article : Google Scholar : PubMed/NCBI

44 

Santos E, Martin-Zanca D, Reddy EP, Pierotti MA, Della Porta G and Barbacid M: Malignant activation of a K-ras oncogene in lung carcinoma but not in normal tissue of the same patient. Science. 223:661–664. 1984. View Article : Google Scholar : PubMed/NCBI

45 

Espada J, Pérez-Moreno M, Braga VM, Rodriguez-Viciana P and Cano A: H-Ras activation promotes cytoplasmic accumulation and phosphoinositide 3-OH kinase association of beta-catenin in epidermal keratinocytes. J Cell Biol. 146:967–980. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Mendoza MC, Er EE and Blenis J: The Ras-ERK and PI3K-mTOR pathways: Cross-talk and compensation. Trends Biochem Sci. 36:320–328. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Rodriguez-Viciana P, Warne PH, Vanhaesebroeck B, Waterfield MD and Downward J: Activation of phosphoinositide 3-kinase by interaction with Ras and by point mutation. EMBO J. 15:2442–2451. 1996.PubMed/NCBI

48 

Michael JV, Wurtzel JG and Goldfinger LE: Inhibition of galectin-1 sensitizes HRAS-driven tumor growth to rapamycin treatment. Anticancer Res. 36:5053–5061. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Chang YY, Lin PC, Lin HH, Lin JK, Chen WS, Jiang JK, Yang SH, Liang WY and Chang SC: Mutation spectra of RAS gene family in colorectal cancer. Am J Surg. 212:537–544. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Feng J, Hua F, Shuo R, Chongfeng G, Huimian X, Nakajima T, Subao W and Tsuchida N: Upregulation of non-mutated H-ras and its upstream and downstream signaling proteins in colorectal cancer. Oncol Rep. 8:1409–1413. 2001.PubMed/NCBI

51 

Riggio M, Perrone MC, Polo ML, Rodriguez MJ, May M, Abba M, Lanari C and Novaro V: AKT1 and AKT2 isoforms play distinct roles during breast cancer progression through the regulation of specific downstream proteins. Sci Rep. 7:442442017. View Article : Google Scholar : PubMed/NCBI

52 

Hofbauer SW, Krenn PW, Hofbauer Piñόn J, Pucher S, Asslaber D, Egle A, Hartmann TN and Greil R: The AKT1 isoform plays a dominant role in the survival and chemoresistance of chronic lymphocytic leukaemia cells. Br J Haematol. 172:815–819. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Sahlberg Häggblad S, Mortensen AC, Haglöf J, Engskog MK, Arvidsson T, Pettersson C, Glimelius B, Stenerlöw B and Nestor M: Different functions of AKT1 and AKT2 in molecular pathways, cell migration and metabolism in colon cancer cells. Int J Oncol. 50:5–14. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Irie HY, Pearline RV, Grueneberg D, Hsia M, Ravichandran P, Kothari N, Natesan S and Brugge JS: Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J Cell Biol. 171:1023–1034. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Wang S and Basson MD: Akt directly regulates focal adhesion kinase through association and serine phosphorylation: Implication for pressure-induced colon cancer metastasis. Am J Physiol Cell Physiol. 300:C657–C670. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Papadimitriou K, Rolfo C, Dewaele E, VanDe Wiel M, Van den Brande J, Altintas S, Huizing M, Specenier P and Peeters M: Incorporating anti-VEGF pathway therapy as a continuum of care in metastatic colorectal cancer. Curr Treat Options Oncol. 16:182015. View Article : Google Scholar : PubMed/NCBI

57 

Xu WW, Li B, Lam AK, Tsao SW, Law SY, Chan KW, Yuan QJ and Cheung AL: Targeting VEGFR1- and VEGFR2-expressing non-tumor cells is essential for esophageal cancer therapy. Oncotarget. 6:1790–1805. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Wang H, Zhang C, Ning Z, Xu L, Zhu X and Meng Z: Bufalin enhances anti-angiogenic effect of sorafenib via AKT/VEGF signaling. Int J Oncol. 48:1229–1241. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Salomon DS, Brandt R, Ciardiello F and Normanno N: Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol. 19:183–232. 1995. View Article : Google Scholar : PubMed/NCBI

60 

Mayer A, Takimoto M, Fritz E, Schellander G, Kofler K and Ludwig H: The prognostic significance of proliferating cell nuclear antigen, epidermal growth factor receptor, and mdr gene expression in colorectal cancer. Cancer. 71:2454–2460. 1993. View Article : Google Scholar : PubMed/NCBI

61 

Srivatsa S, Paul MC, Cardone C, Holcmann M, Amberg N, Pathria P, Diamanti MA, Linder M, Timelthaler G, Dienes HP, et al: EGFR in tumor-associated myeloid cells promotes development of colorectal cancer in mice and associates with outcomes of patients. Gastroenterology. 153:178–190. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Bardelli A and Siena S: Molecular mechanisms of resistance to cetuximab and panitumumab in colorectal cancer. J Clin Oncol. 28:1254–1261. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Georgakilas AG, Martin OA and Bonner WM: p21: A two-faced genome guardian. Trends Mol Med. 23:310–319. 2017. View Article : Google Scholar : PubMed/NCBI

64 

el-Deiry WS, Harper JW, O'Connor PM, Velculescu VE, Canman CE, Jackman J, Pietenpol JA, Burrell M, Hill DE and Wang Y: WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res. 54:1169–1174. 1994.PubMed/NCBI

65 

el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW and Vogelstein B: WAF1, a potential mediator of p53 tumor suppression. Cell. 75:817–825. 1993. View Article : Google Scholar : PubMed/NCBI

66 

Harper JW, Adami GR, Wei N, Keyomarsi K and Elledge SJ: The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell. 75:805–816. 1993. View Article : Google Scholar : PubMed/NCBI

67 

Yasui W, Akama Y, Yokozaki H, Semba S, Kudo Y, Shimamoto F and Tahara E: Expression of p21WAF1/CIP1 in colorectal adenomas and adenocarcinomas and its correlation with p53 protein expression. Pathol Int. 47:470–477. 1997. View Article : Google Scholar : PubMed/NCBI

68 

Matsushita K, Kobayashi S, Kato M, Itoh Y, Okuyama K, Sakiyama S and Isono K: Reduced messenger RNA expression level of p21 CIP1 in human colorectal carcinoma tissues and its association with p53 gene mutation. Int J Cancer. 69:259–264. 1996. View Article : Google Scholar : PubMed/NCBI

69 

Schwandner O, Bruch HP and Broll R: Prognostic significance of p21 and p27 protein, apoptosis, clinical and histologic factors in rectal cancer without lymph node metastases. Eur Surg Res. 34:389–396. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 39 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qi C, Chen Y, Zhou Y, Huang X, Li G, Zeng J, Ruan Z, Xie X and Zhang J: Delineating the underlying molecular mechanisms and key genes involved in metastasis of colorectal cancer via bioinformatics analysis. Oncol Rep 39: 2297-2305, 2018
APA
Qi, C., Chen, Y., Zhou, Y., Huang, X., Li, G., Zeng, J. ... Zhang, J. (2018). Delineating the underlying molecular mechanisms and key genes involved in metastasis of colorectal cancer via bioinformatics analysis. Oncology Reports, 39, 2297-2305. https://doi.org/10.3892/or.2018.6303
MLA
Qi, C., Chen, Y., Zhou, Y., Huang, X., Li, G., Zeng, J., Ruan, Z., Xie, X., Zhang, J."Delineating the underlying molecular mechanisms and key genes involved in metastasis of colorectal cancer via bioinformatics analysis". Oncology Reports 39.5 (2018): 2297-2305.
Chicago
Qi, C., Chen, Y., Zhou, Y., Huang, X., Li, G., Zeng, J., Ruan, Z., Xie, X., Zhang, J."Delineating the underlying molecular mechanisms and key genes involved in metastasis of colorectal cancer via bioinformatics analysis". Oncology Reports 39, no. 5 (2018): 2297-2305. https://doi.org/10.3892/or.2018.6303