Open Access

Mechanism analysis of colorectal cancer according to the microRNA expression profile

  • Authors:
    • Hong Li
    • Huichao Zhang
    • Gang Lu
    • Qingjing Li
    • Jifeng Gu
    • Yuan Song
    • Shejun Gao
    • Yawen Ding
  • View Affiliations

  • Published online on: August 16, 2016     https://doi.org/10.3892/ol.2016.5027
  • Pages: 2329-2336
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The present study aimed to identify specific microRNAs (miRs) and their predicted target genes to clarify the molecular mechanisms of colorectal cancer (CRC). An miR expression profile (array ID, GSE39833), which consisted of 88 CRC samples with various tumor‑necrosis‑metastasis stages and 11 healthy controls, was downloaded from the Gene Expression Omnibus database. Subsequently, the differentially expressed miRs and their target genes were screened. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways of target genes were analyzed using the Database for Annotation Visualization and Integrated Discovery. A protein‑protein interaction (PPI) network of the target genes was constructed using the Search Tool for the Retrieval of Interacting Genes database. The present study identified a total of 18 differentially expressed miRs (upregulated, 8; downregulated, 10) in the sera of the CRC patients compared with the healthy controls. Of these, 3 upregulated (let‑7b, miR‑1290 and miR‑126) and 2 downregulated (miR‑16 and miR‑760) differentially expressed miRs and their target genes, including cyclin D1 (CCND1), v‑myc avian myelocytomatosis viral oncogene homolog (MYC), phosphoinositide‑3‑kinase, regulatory subunit 2 (beta) (PIK3R2) and SMAD family member 3 (SMAD3), were significantly enriched in the CRC developmental pathway. All these target genes had higher node degrees in the PPI network. In conclusion, let‑7b, miR‑1290, miR‑126, miR‑16 and miR‑760 and their target genes, CCND1, MYC, PIK3R2 and SMAD3, may be important in the molecular mechanisms for the progression of CRC.

Introduction

Colorectal cancer (CRC) is a global health challenge with >1.2 million novel cases and 600,000 mortalities every year (1). Survival rates of CRC greatly depend on the stage at primary diagnosis, and the 5-year survival rate is 93 and 8% for stage I and IV, respectively (2). Systematic methods for diagnosis, including flexible sigmoidoscopy and implementation of the fecal occult blood test, may contribute to a high detection rate of patients in the early stages of CRC, thus reducing mortality rates (3). There are numerous screening methods, drugs and chemotherapy regimens, including irinotecan and oxaliplatin, available; however, the majority of these have low sensitivity and treatment-limiting side effects (46). Therefore, the development of highly sensitive and CRC-specific diagnostic and prognostic biomarkers is required for the diagnosis and treatment of early stage CRC.

MicroRNAs (miRs) have emerged as important molecules involved in tumorigenesis and disease progression and metastasis in various cancers, including CRC (7). miRs are endogenous, short non-coding RNAs that effectively inhibit target gene activity by binding to target mRNAs and hindering their translation (8). Each miR achieves functional specificity by targeting a core network of genes and has potential effects on almost every genetic pathway (9). Increasing evidence suggests that aberrant miR expression is clearly associated with the initiation and progression of certain cancers, and miRs may act as oncogenes and/or tumor suppressor genes (10). miR-21 downregulates tumor suppressor programmed cell death 4 at a post-transcriptional level and promotes the invasion, intravasation and metastasis of CRC (11). miR-335 exhibits anti-tumoral activity in various tumors, including gastric cancer (12), ovarian cancer (13) and CRC (14). Therefore, pivotal miRs may be informative biomarkers for detection, diagnosis and prognosis of various types of cancer. Furthermore, miRs have emerged as highly tissue-specific biomarkers (15,16), and previous studies have paved the way for miR-based cancer tissue classification (17,18). However, miRs associated with CRC and their roles in the molecular pathogenesis of CRC remain unclear.

To date, one of the most interesting findings is that secreted miRs potentially influence target cell function via exosomes (19). Exosomes are small (30–90 nm) membrane vesicles that carry mRNAs, proteins, lipids and miRs, depending on the origin of the secreting cells (20,21). Previous studies have demonstrated that exosomes are the newest family members of ‘bioactive vesicles’ that promote intercellular communication and immunoregulatory processes via shuttling molecules between cells (22,23). miRs carried in exosomes are secreted from cancer cells into body fluids, such as blood, urine, breast milk and saliva (21). Therefore, exosomal miRs in body fluids may be useful diagnostic and prognostic biomarkers of cancers (24). However, research on the association between exosomal miR profiles in blood and the pathological condition of cancer patients are limited.

In a previous study, the exosomal miR profile (GSE39833) (25) has been used to highlight the most abundant miRs in the blood of patients with CRC using an arbitrary boundary. In silico analysis of these miRs was performed to explore the function of miRs as tumor surveillance mechanisms exerting continuous inhibition on tumor formation. There was no systematic and comprehensive analysis for the miR functions in CRC. Therefore, the present study used microarray analysis to screen the differentially expressed miRs in the sera of patients with CRC. Additionally, in order to improve the understanding of miR functions, the predicted target genes of miRs were identified. Comprehensive bioinformatics methods were used to investigate the function and pathways of target genes of miRs and construct a protein-protein interaction (PPI) network to identify the hub target genes and miRs associated with CRC. The present study aimed to investigate the key role of potentially important miRs and their target genes in the initiation and progression of CRC. The present study may provide a basis for screening novel biomarkers of CRC for therapeutic interventions.

Materials and methods

Microarray data of miRs

The miR expression profile was downloaded from the Gene Expression Omnibus (GEO) database (www.ncbi.nlm.nih.gov/geo/) using the series accession number GSE39833, which was deposited by Ogata-Kawata et al (3). The GSE39833 dataset consisted of 88 CRC patients with various tumor-necrosis-metastasis stages (stage I, 20 patients; II, 20 patients; IIIa, 20 patients; IIIb, 16 patients; IV, 12 patients) between the ages of 45–65 years and 11 healthy controls. An Agilent Human miRNA Microarray platform was used (product no., G4470C; design ID, 021827; Agilent Technologies, Inc., Santa Clara, CA, USA).

Data preprocessing and screening of differentially expressed miRs

Limma (26) (version 2.7.10; bioconductor.org/packages/release/bioc/html/limma.html) package in R (version 2.3.1; www.r-project.org/) was used for data preprocessing and normalization. A T-test (26) in Limma package was used to identify the significantly differentially expressed miRs in the CRC group compared with the healthy control group. P<0.05 and |log2 fold change|>1.5 were considered to indicate a statistically significant difference.

Prediction of the target genes for miRs

Identification of the predicted target genes of miRs is conducive to improve the understanding of miR functions. miRecords (version 3; c1.accurascience.com/miRecords/) and miRWalk (verison 2.0; zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2/) were used to retrieve the target genes of the differentially expressed miRs. miRecords (27), an integrated database for miR-target interactions (MTIs), hosts a large, high-quality, manually curated database of experimentally validated MTIs. miRWalk (28), a comprehensive database on miRs, hosts predicted and validated miR binding sites and information on all known genes of human, mouse and rat. Only miR-target interactions present in the two databases were used, since target genes shared by the two databases were considered to be more reliable.

Function and pathway enrichment analysis

Functional analysis of large gene lists from high-throughput genomic, proteomic and bioinformatics scanning approaches increases the likelihood for researchers to identify biological processes most associated with their study (29). Gene Ontology (GO) analysis is increasingly applied for functional studies of microarray data (30). Kyoto Encyclopedia of Genes and Genomes (KEGG) is the major public pathway-associated database (31), and Database for Annotation Visualization and Integrated Discovery (DAVID) is a tool for providing functional annotation behind large-scale genomic or transcriptomic data (32). DAVID online analytical tool (version 6.7; david.ncifcrf.gov/) was utilized to analyze the significantly enriched GO terms (geneontology.org/) and KEGG pathways (www.genome.jp/kegg/) for target genes of miRs. Each GO term or KEGG pathway included at least two genes, and P<0.05 was considered to indicate a statistically significant difference.

Construction of a PPI network

Search Tool for the Retrieval of Interacting Genes (STRING) (33) is a database providing the predicted protein interaction information in a given cell context. PPI networks are increasingly becoming the focus of research in the identification of the cellular function of proteins in various organisms (34). Based on the information of the STRING database (version 8.3; string-db.org/), a protein interaction network was constructed by mapping the target genes of miRs. The interaction pairs with a combined score of 0.4 were selected in this network.

Results

Screening of differentially expressed miRs

In the present study, the expression levels of 851 miRs were acquired following normalization. Following a comparison of the expression between the CRC and healthy control samples using a t-test, 18 differentially expressed miRs were identified (Table I). Among them, 8 miRs were upregulated (Table IA) and 10 were downregulated (Table IB).

Table I.

Differentially expressed miRs between colorectal cancer and healthy control samples.

Table I.

Differentially expressed miRs between colorectal cancer and healthy control samples.

A, Upregulated differentially expressed miRs

miRLog2, FCP-value
hsa-let-7b0.58796649 2.89×10−3
hsa-miR-12462.59391919   7.14×10−10
hsa-miR-1263.16477901 6.27×10−9
hsa-miR-12901.32767795 3.49×10−5
hsa-miR-181b1.08748377 3.88×10−2
hsa-miR-181d1.41294860 2.45×10−2
hsa-miR-23a1.87225376 6.94×10−8
hsa-miR-654-5p1.35980450 1.25×10−3

B, Downregulated differentially expressed miRs

miRLog2, FCP-value

hsa-miR-1225-5p−0.81943754 9.73×10−4
hsa-miR-1287−0.72720715 2.84×10−3
hsa-miR-1295−1.44266151 5.07×10−5
hsa-miR-1299−1.22393055 4.15×10−3
hsa-miR-144−2.48669140 1.47×10−9
hsa-miR-16−1.35722402 2.42×10−5
hsa-miR-513a-5p−0.90185107 3.77×10−4
hsa-miR-575−0.86229919   5.94×10−10
hsa-miR-652−1.77364130   8.49×10−12
hsa-miR-760−0.91368658 1.36×10−2

[i] miR, microRNA; FC, fold change.

Prediction of the target genes for miRs

Based on the information of miRecords and miRTarBase database, 27 target genes corresponding to the 8 upregulated miRs (Fig. 1) and 79 target genes corresponding to 4 out of 10 downregulated miRs (Fig. 2) were obtained. In total, 6 out of 10 downregulated miRs, miR-1287, miR-1295, miR-1299, miR-144, miR-575 and miR-652, did not have their target genes identified.

Function and pathway enrichment analysis

In the present study, 15 KEGG pathways corresponding to the upregulated miRs and 9 KEGG pathways corresponding to the downregulated miRs were enriched, respectively, using a cutoff criteria of P<0.05 (Table II). The present results revealed that the pathways enriched by differentially expressed miRs were not only associated with various types of cancer, including chronic myeloid leukemia, small cell lung cancer, endometrial cancer and CRC, but also with important signaling pathways, including the ErbB, Wnt, Jak-STAT and p53 signaling pathways (Table II). Notably, the present results demonstrated that the target genes of upregulated miRs, such as cyclin D1 (CCND1), v-myc avian myelocytomatosis viral oncogene homolog (MYC) and phosphoinositide-3-kinase, regulatory subunit 2 (beta) (PIK3R2) (Table IIA), and target genes of downregulated miRs, such as CCND1, mutS homolog 2 (MSH2), jun proto-oncogene (JUN), B-cell CLL/lymphoma 2 (BCL2) and SMAD family member 3 (SMAD3) (Table IIB), were all enriched in CRC pathways; therefore, these target genes may be closely associated with CRC. Additionally, the upregulated miRs of these target genes were let-7b (CCND1), miR-1290 (MYC) and miR-126 (PIK3R2). The downregulated miRs of these target genes were miR-16 (CCND1, MSH2, JUN, BCL2) and miR-760 (SMAD3).

Table II.

Enriched KEGG pathways of target genes in colorectal cancer samples.

Table II.

Enriched KEGG pathways of target genes in colorectal cancer samples.

A, Enriched KEGG pathways of upregulated target genes

KEGG IDDescriptionCountP-valueGenes
hsa05220Chronic myeloid leukemia5 1.33×10−5CCND1, CRKL, CDK6, MYC, PIK3R2
hsa05200Pathways in cancer5 3.90×10−3CCND1, CRKL, CDK6, MYC, PIK3R2
hsa05222Small cell lung cancer4 6.52×10−4CCND1, CDK6, MYC, PIK3R2
hsa04110Cell cycle4 2.07×10−3CCND1, CDK6, MYC, CDC25A
hsa05213Endometrial cancer3 5.32×10−3CCND1, MYC, PIK3R2
hsa05223Non-small cell lung cancer3 5.73×10−3CCND1, CDK6, PIK3R2
hsa05221Acute myeloid leukemia3 6.58×10−3CCND1, MYC, PIK3R2
hsa05214Glioma3 7.73×10−3CCND1, CDK6, PIK3R2
hsa05218Melanoma3 9.75×10−3CCND1, CDK6, PIK3R2
hsa05212Pancreatic cancer3 1.00×10−2CCND1, CDK6, PIK3R2
hsa05210Colorectal cancer3 1.35×10−2CCND1, MYC, PIK3R2
hsa04012ErbB signaling pathway3 1.44×10−2CRKL, MYC, PIK3R2
hsa04670Leukocyte transendothelial migration3 2.56×10−2VCAM1, CXCL12, PIK3R2
hsa04310Wnt signaling pathway3 4.04×10−2CCND1, NLK, MYC
hsa04630Jak-STAT signaling pathway3 4.24×10−2CCND1, MYC, PIK3R2

B, Enriched KEGG pathways of downregulated target genes

KEGG IDDescriptionCountP-valueGenes

hsa05200Pathways in cancer9 5.85×10−4E2F1, CCNE1, CCND1, HSP90B1, XIAP, MSH2, JUN, BCL2, SMAD3
hsa04110Cell cycle7 8.70×10−5E2F1, CCNE1, CCND1, CCND3, CDC14B, SMAD3, GADD45B
hsa04510Focal adhesion6 6.74×10−3CCND1, CCND3, XIAP, JUN, BCL2, KDR
hsa05210Colorectal cancer5 1.55×10−3CCND1, MSH2, JUN, BCL2, SMAD3
hsa05222Small cell lung cancer5 1.55×10−3E2F1, CCNE1, CCND1, XIAP, BCL2
hsa05215Prostate cancer5 1.92×10−3E2F1, CCNE1, CCND1, HSP90B1, BCL2
hsa04310Wnt signaling pathway5 1.26×10−2CCND1, CSNK2A1, CCND3, JUN, SMAD3
hsa04144Endocytosis5 2.43×10−2VPS45, HSPA1A, PDCD6IP, HLA-G, KDR
hsa04115p53 signaling pathway4 7.87×10−3CCNE1, CCND1, CCND3, GADD45B

[i] Count, number of up/downregulated target genes enriched in KEGG pathway; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Furthermore, based on GO enrichment analysis associated with the biological processes of target genes, 35 and 96 GO terms were identified, which were enriched corresponding to the up/downregulated miRs, respectively (Table III). The results revealed that the target genes of differentially expressed miRs were significantly associated with cell cycle processes and response to hormone stimulus.

Table III.

Ten of the most enriched GO terms associated with the biological process of target genes in colorectal cancer samples.

Table III.

Ten of the most enriched GO terms associated with the biological process of target genes in colorectal cancer samples.

A, Enriched GO terms of upregulated target genes

GO IDDescriptionCountP-value
GO:0045449Regulation of transcription10 2.14×10−2
GO:0010033Response to organic substance  7 1.04×10−3
GO:0022402Cell cycle  6 2.25×10−3
GO:0007049Cell cycle  6 8.69×10−3
GO:0010604Positive regulation of macromolecule metabolic process  6 1.30×10−2
GO:0043627Response to estrogen stimulus  5 2.71×10−5
GO:0048545Response to steroid hormone stimulus  5 2.82×10−4
GO:0045596Negative regulation of cell differentiation  5 4.41×10−4
GO:0009725Response to hormone stimulus  5 3.14×10−3
GO:0043627Response to estrogen stimulus  5 3.23×10−3

B, Enriched GO terms of downregulated target genes

GO IDDescriptionCountP-value

GO:0006915Apoptosis13 1.80×10−5
GO:0012501Programmed cell death13 2.08×10−5
GO:0008219Cell death13 1.01×10−4
GO:0016265Cell death13 1.08×10−4
GO:0007049Cell cycle12 8.20×10−4
GO:0051726Regulation of cell cycle11 2.71×10−6
GO:0006793Phosphorus metabolic process11 1.38×10−2
GO:0006796Phosphate metabolic process11 1.38×10−2
GO:0016310 Phosphorylation10 1.13×10−2
GO:0042981Regulation of apoptosis10 1.17×10−2

[i] GO ID, identification number of enriched GO term associated with a biological process; Count, number of up/downregulated target genes; GO, Gene Ontology.

PPI network construction of differentially expressed miR target genes

According to the information from STRING database, a PPI network of differentially expressed miR target genes was constructed (Fig. 3). The results demonstrated that the top 6 target genes, from high to low expression based on node degree, were estrogen receptor 1 (degree=19), MYC (degree=18), JUN (degree=17), CCND1 (degree=15), cell division cycle 25A (degree=10) and H3 histone, family 3B (degree=10). Notably, all the target genes enriched in the CRC pathway, including CCND1, MYC, PIK3R2, MSH2, JUN, BCL2 and SMAD3, had higher node degrees in the PPI network and the node degrees of these genes was >4.

Discussion

CRC is a complex disease with genetic and epigenetic alterations in numerous key oncogenes and tumor suppressor genes (35). However, the precise molecular mechanisms leading to anticancer activities in CRC remain unclear. miR biology has increasingly become a focus in the field of cancer research, and secreted miRs embedded in exosomes have emerged as diagnostic biomarkers for cancer detection (24). The present study analyzed the exosomal miR profile in the sera of CRC patients, derived from a GEO database, to screen differentially expressed miRs between CRC patients and healthy controls, and their target genes for therapeutic intervention.

A total of 18 differentially expressed miRs were identified in the sera of CRC patients compared to healthy controls. Furthermore, the present results revealed that 3 upregulated (let-7b, miR-1290 and miR-126) and 2 downregulated (miR-16 and miR-760) differentially expressed miRs and their target genes, including CCND1, MYC, PIK3R2 and SMAD3, were significantly enriched in the CRC pathway. All these target genes had higher node degrees in a PPI network constructed in the present study.

CCND1 was the identified crucial target gene of let-7b and miR-16 in the network of miRs and target genes. let-7b is a member of the let-7 family, which may inhibit cancer cell growth and promote cell apoptosis via post-transcriptional repression of cytochrome P450 epoxygenase 2J2 (CYP2J2) (36). CYP2J2 has been observed to have a tumor-promotion function in human cancer (36). In addition, miR-16 is a regulator of microtubule-associated protein 7 in modulating the proliferation in several types of cancer cell (37). In vitro experiments reveal that overexpression of miR-16 represses the proliferation and induces apoptosis of CRC cells by regulating the p53/survivin signaling pathway (38). Furthermore, CCND1 is widely recognized to be involved in cancer cell cycle progression and is vital in controlling G1/S cell cycle transition (39). CCND1 regulates the switch between proliferation and growth arrest, and the CCND1 G870A polymorphism contributes to CRC susceptibility (40). Additionally, CCND1 is regulated by the miR-16 family, and the miR-16 family induces G1 arrest by regulating the expression of CCND1 (41). let-7b inhibits cell cycle progression and anchorage-independent growth of melanoma cells via the downregulation of CCND1 (42). The results of the present study are similar to those of previous studies, as let-7b and miR-16 could directly target CCND1. The present results additionally suggest that let7b and miR-16 inhibit CRC progression by targeting CCND1.

miR-1290 was also demonstrated to be upregulated in the present study. miR-1290 and its target gene MYC are considered to be crucial in the regulation of CRC. Upregulation of miR-1290 in colon cancer cells impairs cytokinesis, which is the final step of cell division (43). Furthermore, overexpression of miR-1290 activates the Wnt pathway (43), which has been demonstrated in several types of cancer, including CRC (44), and is important in tumorigenesis (45). The MYC proto-oncogene encodes a prototypical oncogenic transcription factor that is crucial in the genesis of various cancers (46). MYC is part of the Mad-Max network that regulates the cell cycle, and cell differentiation and apoptosis (47,48). Previous experimental data indicates that even a slight inhibition of c-MYC expression may permanently inhibit tumor growth and enhance the regression of tumors (49). A previous study by Wu et al (43) demonstrated that miR-1290 was involved in the reprogramming of colon cancer cells by increasing the expression of c-MYC. Thus, miR-1290 may be a putative oncogenic miR in promoting CRC progression.

Similarly, the upregulated miR-126 and target gene PIK3R2 were also identified in the network of miRs and target genes. miR-126 is upregulated in primary CRC tissues and cell lines; a previous study verified that epigenetic silencing of miR-126 in CRC cells effectively leads to cell growth, migration and invasion (50,51). PIK3R2 is a key regulator in the phosphoinositide 3-kinase (PI3K) pathway, which is a key signal transduction system that associates oncogenes with numerous cellular functions and has a role in human cancer (52). In addition, miR-126 may be important in tumorigenesis and metastasis by targeting PIK3R2 to regulate the vascular endothelial growth factor/PI3K/protein kinase B signaling pathway in human breast cancer (53). On the basis of the present results, the present authors hypothesise that miR-126 may be an important molecule in the pathogenesis of CRC via the regulation of PIK3R2.

In the present study miR-760 was downregulated, and was demonstrated to interact with SMAD3. SMAD3 is a downstream mediator of the transforming growth factor-β (TGF-β) signaling pathway (54). TGF-β signaling regulates tumorigenesis, and its signaling pathway is crucial in tumor progression in human cancer (55). Additionally, SMAD3 expression decreases susceptibility to tumorigenicity in the early stages of gastric carcinogenesis (56). In the present study, miR-760 was downregulated, which was similar to a previous finding that miR-760 had a lower expression in CRC tissues and might be a biomarker for the early detection of CRC (57). Therefore, miR-760 may indirectly inhibit tumorigenesis and progression of CRC by targeting SMAD3. However, few functional studies have been performed on miR-760; therefore, additional experimental validation and high throughput data analysis are required.

In conclusion, the present study has identified differentially expressed miRs in the sera of CRC patients compared with healthy controls. let-7b, miR-16, miR-1290, miR-126 and miR-760, and their target genes, CCND1, MYC, PIK3R2 and SMAD3, may be important in the initiation and progression of CRC. let-7b and miR-16 may regulate cell cycle processes by interacting with CCND1, thus inhibiting cancer cell proliferation and growth and induce cell apoptosis. miR-1290 and miR-126 may function in promoting the tumorigenesis and metastasis of CRC cells via the regulation of MYC and PIK3R2, respectively, while miR-760 may indirectly prevent the progression of CRC by targeting SMAD3. Therefore, these 5 miRs and their target genes may be candidate biomarkers for the diagnosis and treatment of CRC. In addition, the present study provides intriguing clues as to the identification of potential candidate biomarkers. The target genes of 6 downregulated miRs, including miR-1287, miR-1295, miR-1299, miR-144, miR-575 and miR-652, were not identified in the present study; therefore, whether they are associated with CRC requires addition study. Overall, the present study may aid additional clinical molecular target therapy experiments concerning CRC.

References

1 

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

2 

Løvf M, Nome T, Bruun J, Eknaes M, Bakken AC, Mpindi JP, Kilpinen S, Rognum TO, Nesbakken A, Kallioniemi O, et al: A novel transcript, VNN1-AB, as a biomarker for colorectal cancer. Int J Cancer. 135:2077–2084. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Ogata-Kawata H, Izumiya M, Kurioka D, Honma Y, Yamada Y, Furuta K, Gunji T, Ohta H, Okamoto H, Sonoda H, et al: Circulating exosomal microRNAs as biomarkers of colon cancer. Plos One. 9:e929212014. View Article : Google Scholar : PubMed/NCBI

4 

Bretthauer M: Evidence for colorectal cancer screening. Best Pract Res Clin Gastroenterol. 24:417–425. 2010. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Chen CC, Yang SH, Lin JK, Lin TC, Chen WS, Jiang JK, Wang HS and Chang SC: Is it reasonable to add preoperative serum level of CEA and CA19-9 to staging for colorectal cancer? J Surg Res. 124:169–174. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Garzon R, Calin GA and Croce CM: MicroRNAs in cancer. Annu Rev Med. 60:167–179. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Esquela-Kerscher A and Slack FJ: Oncomirs-microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Mencía A, Modamio-Høybjør S, Redshaw N, Morín M, Mayo-Merino F, Olavarrieta L, Aguirre LA, del Castillo I, Steel KP, Dalmay T, et al: Mutations in the seed region of human miR-96 are responsible for nonsyndromic progressive hearing loss. Nat Genet. 41:609–613. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S and Allgayer H: MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 27:2128–2136. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Li H, Xie S, Liu M, Chen Z, Liu X, Wang L, Li D and Zhou Y: The clinical significance of downregulation of mir-124-3p, mir-146a-5p, mir-155-5p and mir-335-5p in gastric cancer tumorigenesis. Int J Oncol. 45:197–208. 2014.PubMed/NCBI

13 

Cao J, Cai J, Huang D, Han Q, Yang Q, Li T, Ding H and Wang Z: miR-335 represents an invasion suppressor gene in ovarian cancer by targeting Bcl-w. Oncol Rep. 30:701–706. 2013.PubMed/NCBI

14 

Sun Z, Zhang Z, Liu Z, Qiu B, Liu K and Dong G: MicroRNA-335 inhibits invasion and metastasis of colorectal cancer by targeting ZEB2. Med Oncol. 31:9822014. View Article : Google Scholar : PubMed/NCBI

15 

Farh KK, Grimson A, Jan C, Lewis BP, Johnston WK, Lim LP, Burge CB and Bartel DP: The widespread impact of mammalian MicroRNAs on mRNA repression and evolution. Science. 310:1817–1821. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Baskerville S and Bartel DP: Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. RNA. 11:241–247. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Montecalvo A, Larregina AT, Shufesky WJ, Stolz DB, Sullivan ML, Karlsson JM, Baty CJ, Gibson GA, Erdos G, Wang Z, et al: Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 119:756–766. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Ogawa Y, Kanai-Azuma M, Akimoto Y, Kawakami H and Yanoshita R: Exosome-like vesicles with dipeptidyl peptidase IV in human saliva. Biol Pharm Bull. 31:1059–1062. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Hu G, Drescher KM and Chen XM: Exosomal miRNAs: Biological properties and therapeutic potential. Front Genet. 3:562012. View Article : Google Scholar : PubMed/NCBI

22 

Théry C, Ostrowski M and Segura E: Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 9:581–593. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, de Gruijl TD, Würdinger T and Middeldorp JM: Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci USA. 107:6328–6333. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT Jr, Carter BS, Krichevsky AM and Breakefield XO: Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 10:1470–1476. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Igaz I and Igaz P: Tumor surveillance by circulating microRNAs: A hypothesis. Cell Mol Life Sci. 71:4081–4087. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Smyth GK: Limma: Linear models for microarray dataBioinformatics and Computational Biology Solutions using R and Bioconductor. Gentleman R, Carey V, Dudoit S, Irizarry R and Huber W: Springer; New York, PA: pp. 397–420. 2005, View Article : Google Scholar

27 

Xiao F, Zuo Z, Cai G, Kang S, Gao X and Li T: miRecords: An integrated resource for microRNA-target interactions. Nucleic Acids Res. 37:(Database Issue). D105–D110. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk-database: Prediction of possible miRNA binding sites by ‘walking’ the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Huangda W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Hulsegge I, Kommadath A and Smits MA: Globaltest and GOEAST: Two different approaches for gene ontology analysis. BMC Proc. 3(Suppl 4): S102009. View Article : Google Scholar : PubMed/NCBI

31 

Kanehisa M: The KEGG database. Novartis Found Symp. 247:91–101; discussion 101–103, 119–128, 244–252. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Huangda W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, von Mering C and Jensen LJ: STRING v9. 1: Protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41:(Database Issue). D808–D815. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Patil A and Nakamura H: Filtering high-throughput protein-protein interaction data using a combination of genomic features. BMC Bioinformatics. 6:1002005. View Article : Google Scholar : PubMed/NCBI

35 

Takahashi M, Sung B, Shen Y, Hur K, Link A, Boland CR, Aggarwal BB and Goel A: Boswellic acid exerts antitumor effects in colorectal cancer cells by modulating expression of the let-7 and miR-200 microRNA family. Carcinogenesis. 33:2441–2449. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Chen F, Chen C, Yang S, Gong W, Wang Y, Cianflone K, Tang J and Wang DW: Let-7b inhibits human cancer phenotype by targeting cytochrome P450 epoxygenase 2J2. Plos One. 7:e391972012. View Article : Google Scholar : PubMed/NCBI

37 

Yan X, Liang H, Deng T, Zhu K, Zhang S, Wang N, Jiang X, Wang X, Liu R, Zen K, et al: The identification of novel targets of miR-16 and characterization of their biological functions in cancer cells. Mol Cancer. 12:922013. View Article : Google Scholar : PubMed/NCBI

38 

Ma Q, Wang X, Li Z, Li B, Ma F, Peng L, Zhang Y, Xu A and Jiang B: microRNA-16 represses colorectal cancer cell growth in vitro by regulating the p53/survivin signaling pathway. Oncol Rep. 29:1652–1658. 2013.PubMed/NCBI

39 

Yang Y, Wang F, Shi C, Zou Y, Qin H and Ma Y: Cyclin D1 G870A polymorphism contributes to colorectal cancer susceptibility: Evidence from a systematic review of 22 case-control studies. Plos One. 7:e368132012. View Article : Google Scholar : PubMed/NCBI

40 

Probst-Hensch NM, Sun CL, Van Den Berg D, Ceschi M, Koh WP and Yu MC: The effect of the cyclin D1 (CCND1) A870G polymorphism on colorectal cancer risk is modified by glutathione-S-transferase polymorphisms and isothiocyanate intake in the Singapore Chinese Health Study. Carcinogenesis. 27:2475–2482. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Liu Q, Fu H, Sun F, Zhang H, Tie Y, Zhu J, Xing R, Sun Z and Zheng X: miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes. Nucleic Acids Res. 36:5391–5404. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Schultz J, Lorenz P, Gross G, Ibrahim S and Kunz M: MicroRNA let-7b targets important cell cycle molecules in malignant melanoma cells and interferes with anchorage-independent growth. Cell Res. 18:549–557. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Wu J, Ji X, Zhu L, Jiang Q, Wen Z, Xu S, Shao W, Cai J, Du Q, Zhu Y and Mao J: Up-regulation of microRNA-1290 impairs cytokinesis and affects the reprogramming of colon cancer cells. Cancer Lett. 329:155–163. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Behrens J: The role of the Wnt signalling pathway in colorectal tumorigenesis. Biochem Soc Trans. 33:672–675. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Karim RZ, Tse GM, Putti TC, Scolyer RA and Lee CS: The significance of the Wnt pathway in the pathology of human cancers. Pathology. 36:120–128. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Zeller KI, Jegga AG, Aronow BJ, O'Donnell KA and Dang CV: An integrated database of genes responsive to the Myc oncogenic transcription factor: Identification of direct genomic targets. Genome Biol. 4:R692003. View Article : Google Scholar : PubMed/NCBI

47 

Pelengaris S, Khan M and Evan G: c-MYC: More than just a matter of life and death. Nat Rev Cancer. 2:764–776. 2002. View Article : Google Scholar : PubMed/NCBI

48 

González V and Hurley LH: The c-MYC NHE III(1): Function and regulation. Annu Rev Pharmacol Toxicol. 50:111–129. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Hermeking H: The MYC oncogene as a cancer drug target. Curr Cancer Drug Targets. 3:163–175. 2003. View Article : Google Scholar : PubMed/NCBI

50 

Li XM, Wang AM, Zhang J and Yi H: Down-regulation of miR-126 expression in colorectal cancer and its clinical significance. Med Oncol. 28:1054–1057. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Zhang Y, Wang X, Xu B, Wang B, Wang Z, Liang Y, Zhou J, Hu J and Jiang B: Epigenetic silencing of miR-126 contributes to tumor invasion and angiogenesis in colorectal cancer. Oncol Rep. 30:1976–1984. 2013.PubMed/NCBI

52 

Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Zhu N, Zhang D, Xie H, Zhou Z, Chen H, Hu T, Bai Y, Shen Y, Yuan W, Jing Q and Qin Y: Endothelial-specific intron-derived miR-126 is down-regulated in human breast cancer and targets both VEGFA and PIK3R2. Mol Cell Biochem. 351:157–164. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Kang HY, Lin HK, Hu YC, Yeh S, Huang KE and Chang C: From transforming growth factor-beta signaling to androgen action: Identification of Smad3 as an androgen receptor coregulator in prostate cancer cells. Proc Natl Acad Sci USA. 98:3018–3023. 2001. View Article : Google Scholar : PubMed/NCBI

55 

Bierie B and Moses HL: TGF-beta and cancer. Cytokine Growth Factor Rev. 17:29–40. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Han SU, Kim HT, Seong DH, Kim YS, Park YS, Bang YJ, Yang HK and Kim SJ: Loss of the Smad3 expression increases susceptibility to tumorigenicity in human gastric cancer. Oncogene. 23:1333–1341. 2004. View Article : Google Scholar : PubMed/NCBI

57 

Liu G, Fang Y, Zhang H, Li Y, Li X, Yu J and Wang X: Computational identification and microarray-based validation of microRNAs in Oryctolagus cuniculus. Mol Biol Rep. 37:3575–3581. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 12 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li H, Zhang H, Lu G, Li Q, Gu J, Song Y, Gao S and Ding Y: Mechanism analysis of colorectal cancer according to the microRNA expression profile. Oncol Lett 12: 2329-2336, 2016
APA
Li, H., Zhang, H., Lu, G., Li, Q., Gu, J., Song, Y. ... Ding, Y. (2016). Mechanism analysis of colorectal cancer according to the microRNA expression profile. Oncology Letters, 12, 2329-2336. https://doi.org/10.3892/ol.2016.5027
MLA
Li, H., Zhang, H., Lu, G., Li, Q., Gu, J., Song, Y., Gao, S., Ding, Y."Mechanism analysis of colorectal cancer according to the microRNA expression profile". Oncology Letters 12.4 (2016): 2329-2336.
Chicago
Li, H., Zhang, H., Lu, G., Li, Q., Gu, J., Song, Y., Gao, S., Ding, Y."Mechanism analysis of colorectal cancer according to the microRNA expression profile". Oncology Letters 12, no. 4 (2016): 2329-2336. https://doi.org/10.3892/ol.2016.5027