Hsa-miR-137, hsa-miR-520e and hsa-miR-590-3p perform crucial roles in Lynch syndrome

  • Authors:
    • Changyu Zhou
    • Jiayu Li
    • Jiarui Li
    • Yingchun Wan
    • Tao Li
    • Piyong Ma
    • Yingjian Wang
    • Haiyan Sang
  • View Affiliations

  • Published online on: July 6, 2016     https://doi.org/10.3892/ol.2016.4816
  • Pages: 2011-2017
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to identify the differentially expressed microRNAs (DEMs) between Lynch syndrome (LS) and the normal colonic (N-C) control samples, predict the target genes (TGs) and analyze the potential functions of the DEMs and TGs. The miRNA expression dataset GSE30454, which included data of 13 LS and 20 N‑C tissue samples, was downloaded from the Gene Expression Omnibus. The classical t‑test in Linear Models for Microarray Data package was used for DEM identification. TG prediction was performed using 5 databases. The regulatory network of the DEMs and their TGs was constructed using Cytoscape. Functional and pathway enrichment analysis was performed. The transcription factors (TFs), tumor‑associated genes (TAG) and tumor suppressor genes (TSGs) were then identified. Three key DEMs hsa‑miR‑137, hsa-miR-520e, and hsa-miR-590-3p were identified. Hsa-miR-520e and hsa-miR-137 had 4 common TGs, including SNF related kinase, metal-regulatory transcription factor 1 (MTF1), round spermatid basic protein 1 and YTH N6‑methyladenosine RNA binding protein 3; hsa-miR-590-3p and hsa-miR‑137 had 14 common TGs, including NCK adaptor protein 1 (NCK1), EPH receptor A7, and stress-associated endoplasmic reticulum protein 1; hsa‑miR‑590‑3p and hsa‑miR‑520e had 12 common TGs, including Krüppel‑like factor (KLF) 13, twinfilin actin binding protein 1, and nuclear factor I B. Through the functional and pathway enrichments analysis, MTF1 was involved in regulation of gene expression and metabolic processes, and sequence‑specific DNA binding TF activity. KLF13 was involved in regulation of gene expression and regulation of cellular metabolic processes. NCK1 was enriched in the axon guidance pathway. In addition, the functional and pathway enrichment analysis showed certain TGs, such as hypoxia‑inducible factor 1α, AKT serine/threonine kinase 2, and rapamycin‑insensitive companion of mammalian target of rapamycin, participated in the mTOR signaling pathway. The 3 key DEMs hsa‑miR‑137, hsa‑miR‑520e, and hsa‑miR‑590‑3p may have important roles in the process of LS.

Introduction

Colorectal cancer (CRC) is one of the most common causes of cancer-associated mortality and is the second and third leading cause of cancer-associated mortality in Western countries and the USA, respectively (1,2). In 2014, ~137,000 individuals in the USA were diagnosed with CRC and ~50,000 patients succumbed to CRC. In addition, more than one-third of all CRC-associated mortalities (29% in men and 43% in women) occurred in individuals aged ≥80 years (1). Lynch syndrome (LS), previously termed hereditary non-polyposis colorectal cancer (HNPCC), accounts for 3% of all CRCs and is caused by a germline mutation in one of the mismatch repair (MMR) genes mutL homolog 1 (MLH1), mutS homolog (MSH) 2, MSH6 and PMS1 homolog 2, mismatch repair system component (PMS2), and LS is the most common hereditary form of CRC (35).

Numerous studies have been published since the first case of hereditary CRC was reported in 1861 (6). Numerous microRNAs (miRNAs) and genes involved in the tumorigenesis of LS have been identified, such as miR-622, miR-1238 (3), insulin like growth factor 2 (7), phosphatase and tensin homolog (PTEN) (8), and PMS2 (4). In addition, certain cellular signaling pathways involved in the tumorigenesis of LS have been identified, including the AKT/mammalian target of rapamycin (mTOR) signaling pathways (8), axon guidance (9), and DNA repair pathways, such as the p53 pathway (10). In LS, somatic mutations of PTEN, which results in the upregulation of the AKT pathway, have been found and intervention against AKT or towards downstream targets, such as mTOR, resulted in a decreased incidence of LS, suggesting that AKT may be an effective approach for the prevention of HNPCC in patients (8,9). However, the molecular mechanisms of LS remain to be elucidated.

In the study performed by Balaguer et al (3), hsa-miR-622, hsa-miR-1238 and hsa-miR-192 were identified as differentially expressed miRNAs (DEMs) in LS, compared with the sporadic microsatellite instability. However, the signatures of the identified target genes (TGs) of DEMs were not analyzed. In order to study the regulatory mechanisms of LS, the microarray data deposited by Balaguer et al were downloaded to identify key DEMs and their TGs. In addition, functional and pathway enrichment analyses were performed for TGs.

Materials and methods

miRNA microarray data

miRNA expression microarray data of GSE30454 (3) was downloaded from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/), based on platform GPL8179 (Illumina Human v2 MicroRNA expression beadchip; Illumina Inc., San Diego, CA, USA). A total of 20 normal colonic tissue samples (N-C group) and 13 LS tumor samples, consisting of 4 with a germline mutation in MLH1, 5 with a germline mutation in MSH2, 3 with a germline mutation in MSH6 and 1 with EpCam deletion (LS group) were selected (3). The raw data and the probe annotation files were downloaded for further analysis.

Data preprocessing and identification of LS-associated DEMs

Firstly, probe sets were mapped to the corresponding miRNAs. If there were multiple probe sets that corresponded to the same miRNA, the expression values of those probe sets were averaged. Then, the t-test method in the Linear Models for Microarray Data package of R (11) (limma version 3.22.7; www.bioconductor.org/packages/3.0/bioc/html/limma.html)was used to identify the DEMs between the N-C and LS groups. Next, the t-test P-value was adjusted to the false discovery rate (FDR) by the Benjamini-Hochberg procedure (12). The cut-off criteria for DEMs were |log2 fold change (FC) |>1 and FDR <0.01. Finally, the LS-associated DEMs were screened using the Human microRNA Disease Database (http://cmbi.bjmu.edu.cn/hmdd), which is a collection of experimentally supported human miRNA and disease associations (13).

Predication of TGs

From the standpoint of high confidence, the TGs of the LS-associated DEMs were predicted using 5 databases, as follows: miRanda (14); MirTarget2 (15); PicTar (16); PITA (17); and TargetScan (18). The intersections of the 5 databases were regarded as the final predicted TGs.

Functional and pathway enrichment analysis

Gene ontology (GO) analysis (http://www.geneontology.org/) is a functional method for the analysis of large-scale transcriptomic or genomic data (19). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database (http://www.genome.jp/kegg/pathway.html) contains information on the mechanism of molecules or genes (20). In order to investigate the biofunction of TGs in tumor progression, the Database for Annotation, Visualization and Integrated Discovery (DAVID; https://david.ncifcrf.gov/), a high-throughput and integrated data-mining environment (21), was used to perform the GO functional and KEGG pathway enrichment analyses for the TGs, based on the hypergeometric distribution. P<0.01 was selected as the threshold.

Identification of transcription factors (TFs), tumor-associated genes (TAGs) and tumor suppressor genes (TSGs) among TGs

The TRANSFAC database (http://www.gene-regulation.com/pub/databases.html) is a database of eukaryotic transcription-regulating DNA sequence elements and the TFs binding to and acting through these elements (22). In order to determine whether the TGs had transcription regulation function, the TRANSFAC database was used to identify the TFs.

The Tumor Associated Gene (TAG) database (http://www.binfo.ncku.edu.tw/TAG/) is a semi-automatic information retrieving engine that was built to collect specific information on TGs from various resources (23). Additionally, Tumor Suppressor Gene (TSGene) database (http://bioinfo.mc.vanderbilt.edu/TSGene/) provides not only a comprehensive resource of TSGs for studies on cancer and to further experimental design, but also a comprehensive TSG catalog biology-based analyses of advanced systems (24). In order to identify the functions of the TGs that may function during genesis of LS, the TSGene and TAG databases were used to perform the identification of TSGs and TAGs among the TGs, respectively.

Construction of regulatory network. DEMs with >100 TGs were considered as key DEMs. According to the regulatory associations between key DEMs and TGs, the regulatory network containing key DEMs and their TGs was constructed and visualized by Cytoscape 2.8.2 software (25).

Statistical analysis

The t-test method in the Linear Models for Microarray Data package of R (limma, version 3.22.7) was used for the DEM selection. The P-value is adjusted as the FDR, and the selection criteria were |log2 fold change (FC)|>1 and FDR <0.01. DAVID, based on the hypergeometric distribution, was used to perform GO and KEGG pathway enrichment analyses. P<0.01 was considered to indicate a statistically significant difference.

Results

DEM identification and TG predication

A total of 159 DEMs were identified between the N-C and LS groups. There were 105 upregulated DEMs and 54 downregulated DEMs, of which only 13 DEMs were predicted to be associated with LS. Furthermore, 3 key DEMs were identified, including 2 upregulated miRNAs (hsa-miR-137 and hsa-miR-590-3p) and 1 downregulated miRNA (hsa-miR-520e). A total of 159 TGs for hsa-miR-137, including NCK adaptor protein 1 (NCK1), AKT serine/threonine kinase 2 (AKT2), Krüppel-like factor (KLF) 4, metal-responsive transcription factor 1 (MTF1) and zinc finger protein (ZNF) 217, 273 TGs for hsa-miR-590-3p, including NCK1, hypoxia-inducible factor 1α (HIF1A), KLF13, nuclear factor I B (NFIB) and ZNF800 and 132 TGs for hsa-miR-520e, such as KLF13, NFIB, MTF1, ZNF800 and rapamycin-insensitive companion of mTOR (RICTOR), were identified.

Functional and pathway enrichment analysis

Through the GO functional enrichment analysis of biological processes, certain TGs, including NCK1, HIF1A, MTF1, KLF13, KLF4 and AKT2, were found to be involved in the regulation of gene expression (P<0.001). Certain TGs, including NCK1, MTF1, KLF4, KLF13, HIF1A, AKT2 and RICTOR, were involved in the regulation of macromolecule metabolic processes (P<0.001) and the regulation of primary metabolic processes (P<0.001) (Table I). Through the GO functional enrichment analysis of cellular components, certain TGs, including MTF1, KLF13, NCK1, HIF1A and AKT2, were found to be enriched in the nucleus (P<0.001). Certain TGs, including HIF1A, MTF1, KLF4, KLF13 and ZNF800, were enriched in intracellular membrane-bound organelles (P<0.001); certain TGs, including MTF1, KLF13, NCK1, HIF1A and ZNF800, were enriched in membrane-bound organelles (P<0.001) (Table I). Through the GO functional enrichment analysis of molecular function, certain TGs, such as MTF1, KLF4, KLF13, HIF1A, ZNF800, and cancer susceptibility candidate 3 (CASC3), were involved in nucleic acid binding (P<0.001). In addition, certain TGs, such as MTF1, KLF4, TEA domain transcription factor 1 (TEAD1), zinc finger protein 148, and HIF1A, were involved in nucleic acid binding TF activity (P<0.001). Certain TGs, such as MTF1, KLF4, KLF13, HIF1A, AKT2 and ZNF800, were involved in heterocyclic compound binding (P<0.001) (Table I).

Table I.

GO enrichment analysis of the target genes.

Table I.

GO enrichment analysis of the target genes.

CategoryIDDescriptionCountGenesP-value
BiologicalGO:0010468Regulation of gene expression183MTF1, NCK1, AKT2, HIF1A, ZNF800, KLF4, KLF13<0.001
processGO:0060255Regulation of macromolecule metabolic process211ZNF800, EPHA7, NCK1, AKT2, MTF1, RICTOR, HIF1A, KLF4, KLF13<0.001
GO:0080090Regulation of primary metabolic process216ZNF800, NCK1, KLF4, KLF13, RICTOR, HIF1A, AKT2, MTF1<0.001
GO:0031323Regulation of cellular metabolic process216ZNF800, KLF4, KLF13, HMGA2, RICTOR, HIF1A, EPHA7, AKT2, MTF1<0.001
GO:0044260Cellular macromolecule metabolic process286ZNF146, HMGA2, NCK1, HIF1A, RICTOR, KLF4, KLF13, EPHA7, MTF1<0.001
CellularGO:0005634Nucleus274KLF4, KLF13, MEF2A, NCK1, ZNF800, HIF1A, AKT2, MTF1<0.001
componentGO:0031981Nuclear lumen109KLF4, MEF2A, HMGA2, USP7, HIF1A, MTF1, FYN<0.001
GO:0044428Nuclear part123KLF4, TEAD1, MTF1, MEF2A, ZNF146, HMGA2, HIF1A, RNF6, NCK1<0.001
GO:0043231Intracellular membrane-bounded organelle342KLF4, KLF13, TEAD1, ZNF800, HIF1A, MTF1<0.001
GO:0043227Membrane-bounded organelle342NCK1, ZNF800, HIF1A, KLF4, KLF13, MTF1<0.001
MolecularGO:0003676Nucleic acid binding168ZNF800, KLF4, KLF13, ZNF146, HMGA2, CASC3, MTF1<0.001
functionGO:0001071Nucleic acid binding transcription factor activity  78TEAD1, MEF2A, HIF1A, HMGA2, MTF1, KLF4<0.001
GO:1901363Heterocyclic compound binding230EPHA7, KLF4, KLF13, ZNF800, HIF1A, AKT2, MTF1<0.001
GO:0003700Sequence-specific DNA binding transcription factor activity  77HIF1A, MEF2A, HMGA2, ZNF148, ZNF396, MTF1, KLF4<0.001
GO:0097159Organic cyclic compound binding231KLF4, KLF13, MTF1 HIF1A, AKT2, EPHA7<0.001

[i] GO, gene ontology; MTF1, metal-regulatory transcription factor 1; NCK1, NCK adaptor protein 1; AKT2, AKT serine/threonine kinase 2; HIF1A, hypoxia-inducible factor 1α; ZNF, zinc finger protein; KLF, Krüppel-like factor; EPHA7, EPH receptor A7; RICTOR, rapamycin-insensitive companion of mammalian target of rapamycin; RNF6, ring finger protein 6; TEAD1, TEA domain transcription factor 1; MEF2A, myocyte enhancer factor 2A; HMGA2, high mobility group AT-hook 2.

According to the KEGG pathway enrichment analysis, TGs were significantly enriched in 3 pathways: mTOR signaling pathway, including RICTOR, HIF1A and AKT2 (P=0.001); RNA transport, including eukaryotic translation initiation factor (EIF) 3 subunit E, CASC3 and EIF3J (P=0.008); and axon guidance, including NCK1, FYN proto-oncogene, Src family tyrosine kinase (FYN) and EPH receptor A7 (EPHA7) (P=0.010) (Table II).

Table II.

Kyoto Encyclopedia of Genes and Genomes pathway enrichment of the target genes.

Table II.

Kyoto Encyclopedia of Genes and Genomes pathway enrichment of the target genes.

IDDescriptionCountGenesP-value
4150mTOR signaling pathway6CAB39, RICTOR, HIF1A, AKT2, RPS6KA3, ULK10.001
3013RNA transport9PAIP1, EIF3E, EIF4A2, EIF5, PABPC1, CASC3, EIF3J, NXT2, RANBP20.008
4360Axon guidance8NCK1, FYN, EPHA7, SEMA3C, SEMA3E, PAK7, PPP3R1, CFL20.010

[i] mTOR, mammalian target of rapamycin; CAB39, calcium binding protein 39; RICTOR, rapamycin-insensitive companion of mammalian target of rapamycin; HIF1A, hypoxia-inducible factor 1α; AKT2, AKT serine/threonine kinase 2; RPS6KA3, ribosomal protein S6 kinase A3; ULK1, unc-51 like autophagy activating kinase 1; PAIP1, poly(A) binding protein interacting protein 1; EIF, eukaryotic translation initiation factor; PABPC1, poly(A) binding protein cytoplasmic 1; CASC3, cancer susceptibility candidate 3; NXT2, nuclear transport factor 2 like export factor 2; RANBP2, RAN binding protein 2; NCK1, NCK adaptor protein 1; FYN, FYN proto-oncogene, Src family tyrosine kinase; EPHA7, EPH receptor A7; SEMA, semaphorin; PAK7, p21 (RAC1) activated kinase 7; PPP3R1, protein phosphatase 3 regulatory subunit B, α; CFL2, cofilin 2.

Identification of TFs, TSGs and TAGs

A total of 12 TGs, consisting of TEAD1, ZNF146, high mobility group AT-hook 2 (HMGA2), FYN, v-crk avian sarcoma virus CT10 oncogene homolog-like, receptor-like tyrosine kinase, ZNF217, KIT proto-oncogene receptor tyrosine kinase, AKT2, chromosome segregation 1 like, interferon regulatory factor 2 and B-cell CLL/lymphoma 6 (BCL6), were identified as oncogenes, according to the TSGene database and TAG database. Among these identified oncogenes, 4 genes, consisting of TEAD1, ZNF146, HMGA2 and BCL6, were identified as TFs based on the TRANSFAC database.

Regulatory network construction

The visualized regulatory network of the key DEMs and the TGs of these DEMs showed that hsa-miR-520e and hsa-miR-137 had 4 common TGs, consisting of SNF related kinase (SNRK), MTF1, round spermatid basic protein 1 (RSBN1) and YTH N6-methyladenosine RNA binding protein 3 (YTHDF3), hsa-miR-590-3p and hsa-miR-137 had 14 common TGs, including NCK1, EPHA7, stress-associated endoplasmic reticulum protein 1 (SERP1) and myocyte enhancer factor 2A (MEF2A), and hsa-miR-590-3p and hsa-miR-520e had 12 common TGs, including KLF13, twinfilin actin binding protein 1 (TWF1), NFIB and ZNF800 (Fig. 1).

Discussion

In the current study, 2 upregulated miRNAs, consisting of hsa-miR-137 and hsa-miR-590-3p, and 1 downregulated miRNA, hsa-miR-520e, were screened as key DEMs. Hsa-miR-520e and hsa-miR-137 had 4 common TGs, consisting of SNRK, MTF1, RSBN1 and YTHDF3. Through functional enrichment analysis, MTF1 was identified as involved in the regulation of gene expression and macromolecule metabolic processes, intracellular membrane-bound organelles, and sequence-specific DNA binding TF activity. As previously reported, MTF1 binds specifically to heavy metal-responsive DNA sequence elements in the enhancer/promoter region of metallothionein (MT) genes to regulate their expression (26,27). MT genes, such as MT1X and MT2A, encode small cysteine-rich proteins that scavenge heavy metals, such as Zn (II), Cd (II) and Cu (I), and reactive oxygen species (28,29). Arriaga et al (30) found that the expression of 5 isoforms of MTs, consisting of MT1G, MT1E, MT1F, MT1H and MT1M, were lost during the transition between normal colorectal mucosa and CRC, and the expression of MTs was associated with a shorter survival time in CRC patients (30). In addition, the MT1X gene showed a high sensitivity for the identification of patients with LS or CRC with sporadic defective in MMR (31). This suggests that hsa-miR-520e and hsa-miR-137 may have crucial roles in LS by regulating TGs.

In the present study, hsa-miR-590-3p and hsa-miR-520e had 12 common TGs, including KLF13, TWF1, NFIB and ZNF800, while KLF4 was the TG of hsa-miR-137. Using functional enrichment analysis, KLF4 and KLF13 were identified as involved in the regulation of gene expression, heterocyclic compound binding, and regulation of the cellular metabolic process. LOR-253, a compound that stimulates KLF4 through the inhibition of the human MTF1, is currently used in an early stage of colon cancer (32). The KLF proteins are zinc finger-containing TFs that exert important functions in regulating diverse biological processes, such as growth and cell proliferation (33). KLF4, formerly termed GKLF, encodes a TF associated with tumor suppression and oncogenesis (34). In LS, KLF4 and KLF5 were expressed and predominantly localized to the epithelial cells of the tumors (35). Overexpression of KLF4 in the colon cancer cell lines resulted in reduced tumorigenesis (36). Additionally, a previous study reported that the loss of heterozygosity in the KLF6 locus in patients with sporadic CRC and LS was 0–35% and 0%, respectively (37). These data suggested that loss of heterozygosity of the KLF6 locus was rarely involved in the carcinogenesis of CRC in patients with LS (38). This revealed that hsa-miR-590-3p and hsa-miR-520e, in addition to hsa-miR-137, may have important roles in the cellular metabolic process and proliferation in LS by targeting their TGs.

Additionally, the common TGs, including NCK1, EPHA7, SERP1 and MEF2A, were simultaneously regulated by hsa-miR-590-3p and hsa-miR-137. The enrichment analysis showed that NCK1 was significantly enriched in the axon guidance pathway. As previously reported, AKT executes double roles in protecting motoneuronal survival and promoting nerve regeneration in vivo, and the dominant overexpression of AKT in adult hypoglossal neurons showed accelerated axonal regeneration (39). However, the phosphorylation of AKT was partially reduced in NCK1-deficient B cells, and it was almost completely absent in NCK1-NCK2-deficient B cells. This revealed that the activation of the phosphoinositide 3-kinase (PI3K)/AKT pathway was restrained in the NCK1-NCK2-knockout B cells (40). Furthermore, ectopic expression of hsa-miR-137 in CRC cells inhibited the phosphorylation of mitogen-activated protein kinase (MAPK) and AKT, which reduced the invasiveness of CRC cells by inhibiting signaling via the P13K/AKT and MAPK pathways (41). Hsa-miR-590-3p was also found to activate the PI3K/AKT signaling pathway by downregulating PTEN, a TSG-activated P13K pathway, and then provide strong growth and survival signals to tumor cells (42). Thus, these results suggested that hsa-miR-590-3p and hsa-miR-137 may play important roles in LS by regulating their target genes through the axon guidance pathway.

The functional and pathway enrichment analyses showed that certain TGs of the key DEMs, including HIF1A, AKT2 and RICTOR, participated in the mTOR signaling pathway. mTOR is a member of the PI3K/AKT/mTOR pathway, whose activation stimulates protein and lipid biosynthesis and it is constitutively activated in LS (43). The mTOR signaling pathway senses and integrates a variety of environmental cues to regulate numerous major cellular processes (44,45). As previously shown, the mTOR complex 2 protein RICTOR was highly overexpressed in CRC tissues, and the inhibition of RICTOR resulted in growth inhibition and induced apoptosis in CRCs (46,47). Additionally, AKT2, a component of the PI3K/AKT/mTOR pathway, is proposed as an oncogene for pancreatic cancer that always occurs in the context of LS (48), and the overexpression of the AKT2 proto-oncogene is an early event during sporadic colon cancer (47,49). In addition, mTOR directly stimulated HIF1A, downstream of PI3K/AKT/mTOR, and indirectly causes other metabolic changes by activating HIF1A (50,51). The subsequent HIF1-dependent metabolic changes are a major determinant of the glycolytic phenotype, including the Warburg effect (50,51). The Warburg effect in CRC results in the accumulation of a glycolytic metabolite, pyruvate, which provides the CRC cells with an environment that is a competitive advantage for invasion (52). Overexpression of HIF1A has also been reported to be significantly associated with shorter CRC-specific survival and overall survival times (53). In the present study, AKT2 was a TG of hsa-miR-137, while RICTOR and HIF1A were TGs of hsa-miR-590-3p. All these genes were enriched in the regulation of the metabolic process. Therefore, it may be hypothesized that the important roles of hsa-miR-137 and hsa-miR-590-3p in regulating metabolic process occur by regulating the expression of their TGs in LS.

In the present study, 3 key DEMs, consisting of hsa-miR-520e, hsa-miR-590-3p and hsa-miR-137 were identified. Hsa-miR-590-3p and hsa-miR-520e had 12 common TGs, including KLF13. Hsa-miR-590-3p and hsa-miR-137 had 14 common TGs, including NCK1. Hsa-miR-520e and hsa-miR-137 had 4 common TGs, including MTF1. Functional and pathway enrichment analysis showed that these TGs were significantly enriched in important functions, such as the regulation of metabolic processes, including regulation of macromolecule metabolic process, regulation of primary metabolic process, regulation of cellular metabolic process and cellular macromolecule metabolic process, mTOR signaling, and the axon guidance pathway, which were crucial for the growth and regulation of LS cancer cells. The present study showed the potential crucial role of hsa-miR-520e, hsa-miR-590-3p and hsa-miR-137 in LS.

In conclusion, the present study identified that hsa-miR-137, hsa-miR-520e and hsa-miR-590-3p are key DEMs. Hsa-miR-520e and hsa-miR-137 had 4 common TGs, consisting of SNRK, MTF1, RSBN1 and YTHDF3. Hsa-miR-590-3p and hsa-miR-137 had 14 common TGs, including NCK1, EPHA7 and SERP1. Hsa-miR-590-3p and hsa-miR-520e had 12 common TGs, including KLF13, TWF1 and NFIB. These regulatory interactions may provide biomarkers for LS detection and prognosis.

References

1 

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

2 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Balaguer F, Moreira L, Lozano JJ, Link A, Ramirez G, Shen Y, Cuatrecasas M, Arnold M, Meltzer SJ, Syngal S, et al: Colorectal cancers with microsatellite instability display unique miRNA profiles. Clin Cancer Res. 17:6239–6249. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch JF and Boland CR: Review of the Lynch syndrome: History, molecular genetics, screening, differential diagnosis and medicolegal ramifications. Clin Genet. 76:1–18. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Lynch HT, Boland CR, Gong G, Shaw TG, Lynch PM, Fodde R, Lynch JF and de la Chapelle A: Phenotypic and genotypic heterogeneity in the Lynch syndrome: Diagnostic, surveillance and management implications. Eur J Hum Genet. 14:390–402. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Luschka H: Ueber polypöse Vegetationen der gesammten Dickdarmschleimhaut. Virchow's Arch f path Anat. 20:133–142. 1861. View Article : Google Scholar

7 

Jass JR: Heredity and DNA methylation in colorectal cancer. Gut. 56:154–155. 2007.PubMed/NCBI

8 

Qian CN, Furge KA, Knol J, Huang D, Chen J, Dykema KJ, Kort EJ, Massie A, Khoo SK, Vanden Beldt K, et al: Activation of the PI3K/AKT pathway induces urothelial carcinoma of the renal pelvis: Identification in human tumors and confirmation in animal models. Cancer Res. 69:8256–8264. 2009. View Article : Google Scholar : PubMed/NCBI

9 

van Engeland M, Derks S, Smits KM, Meijer GA and Herman JG: Colorectal cancer epigenetics: Complex simplicity. J Clin Oncol. 29:1382–1391. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Murphy ME: Polymorphic variants in the p53 pathway. Cell Death Differ. 13:916–920. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Wettenhall JM and Smyth GK: limmaGUI: A graphical user interface for linear modeling of microarray data. Bioinformatics. 20:3705–3706. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: A practical and powerful approach to multiple testing. J Roy Stat Soc B. 57:289–300. 1995.

13 

Leidinger P, Keller A, Borries A, Reichrath J, Rass K, Jager SU, Lenhof HP and Meese E: High-throughput miRNA profiling of human melanoma blood samples. BMC Cancer. 10:2622010. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Wang X and El Naqa IM: Prediction of both conserved and nonconserved microRNA targets in animals. Bioinformatics. 24:325–332. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M and Rajewsky N: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Betel D, Koppal A, Agius P, Sander C and Leslie C: Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites. Genome Biol. 11:R902010. View Article : Google Scholar : PubMed/NCBI

18 

Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP and Burge CB: Prediction of mammalian microRNA targets. Cell. 115:787–798. 2003. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

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

21 

da Huang 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

22 

Wingender E, Dietze P, Karas H and Knüppel R: TRANSFAC: A database on transcription factors and their DNA binding sites. Nucleic Acids Res. 24:238–241. 1996. View Article : Google Scholar : PubMed/NCBI

23 

Chen JS, Hung WS, Chan HH, Tsai SJ and Sun HS: In silico identification of oncogenic potential of fyn-related kinase in hepatocellular carcinoma. Bioinformatics. 29:420–427. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Zhao M, Sun J and Zhao Z: TSGene: A web resource for tumor suppressor genes. Nucleic Acids Res. 41(Database issue): D970–D976. 2013. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Boudreault F, Pinilla M, Kho A, Baron R and Tschumperlin D: Metallothionein is a stretch-induced gene that confers protection during mechanical ventilation. Technology. 1:32011.

27 

Günther V, Lindert U and Schaffner W: The taste of heavy metals: Gene regulation by MTF-1. Biochim Biophys Acta. 1823:1416–1425. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Saini N, Georgiev O and Schaffner W: The parkin mutant phenotype in the fly is largely rescued by metal-responsive transcription factor (MTF-1). Mol Cell Biol. 31:2151–2161. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Günther V, Davis AM, Georgiev O and Schaffner W: A conserved cysteine cluster, essential for transcriptional activity, mediates homodimerization of human metal-responsive transcription factor-1 (MTF-1). Biochim Biophys Acta. 1823:476–483. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Arriaga JM, Levy EM, Bravo AI, Bayo SM, Amat M, Aris M, Hannois A, Bruno L, Roberti MP, Loria FS, et al: Metallothionein expression in colorectal cancer: Relevance of different isoforms for tumor progression and patient survival. Hum Pathol. 43:197–208. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Morandi L, de Biase D, Visani M, Monzoni A, Tosi A, Brulatti M, Turchetti D, Baccarini P, Tallini G and Pession A: T ([20]) repeat in the 3′-untranslated region of the MT1X gene: A marker with high sensitivity and specificity to detect microsatellite instability in colorectal cancer. Int J Colorectal Dis. 27:647–656. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Constant S, Huang S, Wiszniewski L and Mas C: Colon cancer: Current treatments and preclinical models for the discovery and development of new therapies. Drug Discovery. El-Shemy HA: InTech. (Rijeka). 433–458. 2013.PubMed/NCBI

33 

Ghaleb AM and Yang VW: The pathobiology of Krüppel-like factors in colorectal cancer. Current Colorectal Cancer Rep. 4:59–64. 2008. View Article : Google Scholar

34 

Rowland BD, Bernards R and Peeper DS: The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol. 7:1074–1082. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Shao J, Yang VW and Sheng H: Prostaglandin E2 and Krüppel-like transcription factors synergistically induce the expression of decay-accelerating factor in intestinal epithelial cells. Immunology. 125:397–407. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Dang DT, Chen X, Feng J, Torbenson M, Dang LH and Yang VW: Overexpression of Krüppel-like factor 4 in the human colon cancer cell line RKO leads to reduced tumorigenecity. Oncogene. 22:3424–3430. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Saunders IW, Ross J, Macrae F, Young GP, Blanco I, Brohede J, Brown G, Brookes D, Lockett T, Molloy PL, et al: Evidence of linkage to chromosomes 10p15.3-p15.1, 14q24.3-q31.1 and 9q33.3-q34.3 in non-syndromic colorectal cancer families. Eur J Hum Genet. 20:91–96. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Miyaki M, Yamaguchi T, Iijima T, Funata N and Mori T: Difference in the role of loss of heterozygosity at 10p15 (KLF6 locus) in colorectal carcinogenesis between sporadic and familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer patients. Oncology. 71:131–135. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Namikawa K, Honma M, Abe K, Takeda M, Mansur K, Obata T, Miwa A, Okado H and Kiyama H: Akt/protein kinase B prevents injury-induced motoneuron death and accelerates axonal regeneration. J Neurosci. 20:2875–2886. 2000.PubMed/NCBI

40 

Leu CM: Nck, a missing adaptor between the B-cell receptor complex and the BCAP/PI3K/Akt pathway. Cell Mol Immunol. 11:120–122. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Liang L, Li X, Zhang X, Lv Z, He G, Zhao W, Ren X, Li Y, Bian X, Liao W, et al: MicroRNA-137, an HMGA1 target, suppresses colorectal cancer cell invasion and metastasis in mice by directly targeting FMNL2. Gastroenterology. 144:624.e4–635.e4. 2013. View Article : Google Scholar

42 

Yang H, Zheng W, Zhao W, Guan C and An J: Roles of miR-590-5p and miR-590-3p in the development of hepatocellular carcinoma. Nan Fang Yi Ke Da Xue Xue Bao. 33:804–811. 2013.(In Chinese). PubMed/NCBI

43 

Ekstrand AI, Jönsson M, Lindblom A, Borg A and Nilbert M: Frequent alterations of the PI3K/AKT/mTOR pathways in hereditary nonpolyposis colorectal cancer. Fam Cancer. 9:125–129. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Laplante M and Sabatini DM: mTOR signaling in growth control and disease. Cell. 149:274–293. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Wullschleger S, Loewith R and Hall MN: TOR signaling in growth and metabolism. Cell. 124:471–484. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Gulhati P, Cai Q, Li J, Liu J, Rychahou PG, Qiu S, Lee EY, Silva SR, Bowen KA, Gao T and Evers BM: Targeted inhibition of mammalian target of rapamycin signaling inhibits tumorigenesis of colorectal cancer. Clin Cancer Res. 15:7207–7216. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Johnson SM, Gulhati P, Rampy BA, Han Y, Rychahou PG, Doan HQ, Weiss HL and Evers BM: Novel expression patterns of PI3K/Akt/mTOR signaling pathway components in colorectal cancer. J Am Coll Surg. 210:767–776. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Schneider G and Schmid RM: Genetic alterations in pancreatic carcinoma. Mol Cancer. 2:152003. View Article : Google Scholar : PubMed/NCBI

49 

Roy HK, Olusola BF, Clemens DL, Karolski WJ, Ratashak A, Lynch HT and Smyrk TC: AKT proto-oncogene overexpression is an early event during sporadic colon carcinogenesis. Carcinogenesis. 23:201–205. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Cairns RA, Harris IS and Mak TW: Regulation of cancer cell metabolism. Nat Rev Cancer. 11:85–95. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Mellinghoff IK and Sawyers CL: TORward AKTually useful mouse models. Nat Med. 10:579–580. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Bi X, Lin Q, Foo TW, Joshi S, You T, Shen HM, Ong CN, Cheah PY, Eu KW and Hew CL: Proteomic analysis of colorectal cancer reveals alterations in metabolic pathways: Mechanism of tumorigenesis. Mol Cell Proteomics. 5:1119–1130. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Baba Y, Nosho K, Shima K, Irahara N, Chan AT, Meyerhardt JA, Chung DC, Giovannucci EL, Fuchs CS and Ogino S: HIF1A overexpression is associated with poor prognosis in a cohort of 731 colorectal cancers. Am J Pathol. 176:2292–2301. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 12 Issue 3

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
Zhou C, Li J, Li J, Wan Y, Li T, Ma P, Wang Y and Sang H: Hsa-miR-137, hsa-miR-520e and hsa-miR-590-3p perform crucial roles in Lynch syndrome. Oncol Lett 12: 2011-2017, 2016
APA
Zhou, C., Li, J., Li, J., Wan, Y., Li, T., Ma, P. ... Sang, H. (2016). Hsa-miR-137, hsa-miR-520e and hsa-miR-590-3p perform crucial roles in Lynch syndrome. Oncology Letters, 12, 2011-2017. https://doi.org/10.3892/ol.2016.4816
MLA
Zhou, C., Li, J., Li, J., Wan, Y., Li, T., Ma, P., Wang, Y., Sang, H."Hsa-miR-137, hsa-miR-520e and hsa-miR-590-3p perform crucial roles in Lynch syndrome". Oncology Letters 12.3 (2016): 2011-2017.
Chicago
Zhou, C., Li, J., Li, J., Wan, Y., Li, T., Ma, P., Wang, Y., Sang, H."Hsa-miR-137, hsa-miR-520e and hsa-miR-590-3p perform crucial roles in Lynch syndrome". Oncology Letters 12, no. 3 (2016): 2011-2017. https://doi.org/10.3892/ol.2016.4816