Open Access

The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis

  • Authors:
    • Fangfang Sun
    • Weiwei Liang
    • Jing Qian
  • View Affiliations

  • Published online on: August 14, 2019     https://doi.org/10.3892/mmr.2019.10588
  • Pages: 3583-3596
  • Copyright: © Sun 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

With the increasing rate of chemoresistance in colorectal cancer (CRC) patients with advanced tumor stages, it is a matter of urgent importance to delineate the factors involved in the drug resistance process. In this study, gene expression profiles were downloaded from the Gene Expression Omnibus database and an integrated analysis with the aim of detecting hub long non‑coding RNAs (lncRNAs) and their regulated, differentially expressed genes (DEGs) during treatment with oxaliplatin (OxPt) or irinotecan was conducted. A total of seven differentially expressed lncRNAs were correlated with OxPt resistance and 21 were correlated with resistance to SN‑38, the active metabolite of irinotecan. Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis confirmed that drug resistance was strongly associated with an imbalance between cell proliferation and apoptosis, cell energetic metabolism under hypoxic conditions, and angiogenesis. Moreover, a large number of lncRNA‑targeted DEGs were located in extracellular exosomes. Further analyses identified four hub lncRNAs involved in the process of drug resistance, including CRNDE, H19, UCA1 and HOTAIR, which are predictive factors for treatment sensitivity. Among them, HOTAIR stands out as a strong factor, the elevated expression of which is also associated with advanced tumor node and metastasis stage and poor CRC disease prognosis.

Introduction

Colorectal cancer (CRC) is a frequent malignant tumor in developed countries, with high morbidity and a high death rate. According to statistics published in 2015, CRC is the third most frequently diagnosed cancer in males and ranks second in females (1). According to the American Joint Committee on Cancer, the 5-year overall survival (OS) rate of CRC is 65.2% (2). Common risk factors for CRC include age, obesity, smoking and an unhealthy diet (3). Previous studies have found that several patients present with unresectable stage disease at initial diagnosis (4,5). Standard treatments for CRC include surgical resection, modern chemotherapy and radiation therapy. Although chemotherapy plays an important role in CRC treatment, the chemoresistance-reduced effectiveness of anti-neoplastic agents causes a lower survival rate of advanced-stage CRC patients.

Oxaliplatin (OxPt) is a third-generation platinum-based chemotherapeutic drug that inhibits the DNA replication and transcription of tumor cells (6). When used in combination with fluorouracil (5-FU), leucovorin and folinic acid, it is recognized as the first-line chemotherapy strategy for CRC (7). In recent years, the extensive use of OxPt has caused increased chemoresistance in clinical practice. Previous studies have found that the nucleotide excision repair pathway plays a key role in OxPt resistance (8,9). Moreover, the overexpression of chemoresistance-associated proteins (e.g., transforming growth factor-β and WBSCR22) and microRNAs, and increased messenger RNA levels of XPAC and ERCC1, are believed to predict chemoresistance to OxPt (1012).

Irinotecan, an analogue of camptothecin isolated from the Chinese tree Camptotheca acuminata, was first approved by the Food and Drug Administration for the treatment of CRC in 1996 and is now used as the second choice for CRC chemotherapy (13). Irinotecan selectively inhibits topoisomerase I and induces the degradation of double-stranded DNA via intracellular modifications. Ultimately, irinotecan is activated by carboxylesterases to its active metabolite SN-38, which combines with topoisomerase and induces the breakage of DNA strands, DNA replication failure and cellular apoptosis (14,15). The therapeutic efficacy and toxicity of irinotecan are related to the expression of tumor-specific genes and the intra-tumor accumulation of SN-38 (16). Irinotecan treatment may also cause severe chemoresistance via variable levels of metabolic enzymes, reduced cellular accumulation from active drug efflux and reduced levels of Topo I expression, as well as different mutations and/or the activation of nuclear factor κB (NF-κB) (17).

Long non-coding RNAs (lncRNAs), defined as mRNA-like transcripts with lengths up to 200 nucleotides, are important members of the non-coding RNA family (18). lncRNA expression is regulated by both transcriptional and epigenetic factors. Several studies have revealed that lncRNAs are expressed in various tumor tissues and are involved in a number of cellular functions and developmental processes, such as cell growth, development, invasion, and apoptosis (1921). Increasing lines of evidence suggest that lncRNAs are valuable bio-targets in the diagnosis and treatment of CRC (22). Recent studies have suggested that lncRNAs play a key role in drug function regulation and chemoresistance through various mechanisms in multiple cancers (23,24). At present, at least 70 CRC-related lncRNAs have been recognized, including HOTAIR, CCAT1, CCAT2, MALAT-1 and H19 (25). Changes in the expression of these lncRNAs could lead to chemotherapy and radiotherapy resistance. Therefore, further research and efforts are needed to clarify the chemoresistance mechanism of each lncRNA. Then a simple and reliable screening program based on the expression levels of lncRNAs can be developed to guide the selection of chemotherapy drugs.

In the current study, previously published RNA expression datasets on the chemoresistance of CRC in the Gene Expression Omnibus (GEO) database were searched. A comprehensive bioinformatics analysis was performed by defining the differentially expressed lncRNAs and other RNAs separately according to their chemoresistance to irinotecan or OxPt. The differentially expressed lncRNAs were correlated with differentially expressed genes (DEGs) and potential diseases based on the RNA-associated interactions database (RAID) and the mammalian lncRNA-disease repository (MNDR) database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were also performed and a protein-protein interaction (PPI) network was established to screen for crucial genes and lncRNAs, whose hazard degrees were further demonstrated by evaluating their expression in tumors and according to tumor, node and metastasis (TNM) stage, as well as survival.

Materials and methods

Datasets and DEG identification

Gene expression profiles for chemoresistance to two chemotherapeutic drugs, OxPt and irinotecan (active metabolite SN-38), in three CRC cell lines (HCT116, HT29, and LoVo) were obtained from GSE42387 in the GEO database (26). Gene expression data were explored and visualized in the ggplot2 package of R software (version 3.2.0, http://ggplot2.tidyverse.org) for each sample in the nine groups (normal HCT116, OxPt-resistant HCT116, SN-38-resistant HCT116, normal HT29, OxPt-resistant HT29, SN-38-resistant HT29, normal LoVo, OxPt-resistant LoVo and SN-38-resistant LoVo). A detailed workflow of the data analysis is shown in Fig. S1. The limma package, which includes lmFit, eBayes and top Table functions, was used for the pairwise comparison of DEGs among the nine groups (27). The cutoff criteria were P<0.05 and abs(log2FC)>1, where FC indicates fold-change. The screened DEGs were divided into two groups, differentially expressed mRNAs and lncRNAs, which were further analyzed.

GO annotation and KEGG pathway enrichment analyses of DEGs

As mentioned above, two groups of DEGs: mRNAs and lncRNAs were analyzed. Based on RAID version 2.0 (28), the potential targets (confidence score >0.5) of the differentially expressed lncRNAs were obtained and intersected with DEGs found in the same group. Next, the intersected DEGs were divided into two groups: OxPt resistance and irinotecan resistance. GO annotation is a classic method used to identify the biological attributes of DEGs. This analysis comprises three parts: Biological process (BP), cell component (CC) and molecular function (MF). KEGG, a collection of genome, biological pathway, disease, drug and chemical substance databases, was used to identify the functional attributes of the DEGs. The Database for Annotation, Visualization and Integrated Discovery (DAVID; ver. 6.8) (29,30) was also applied for the functional interpretation of the two large lists of genes derived from previous analyses. Statistical significance was set at P<0.05.

PPI network construction and hub gene identification

A PPI network was constructed for the DEGs using STRING (31), an online functional protein association network tool, to detect potential relationships among the DEGs with confidence scores ≥0.4 and a maximum number of interactors of 1. The generated PPI network data were downloaded and imported into Cytoscape software (ver. 3.5.1), a bioinformatics tool used to create networks of protein interactions and reassess and integrate gene information from numerous embedded applications. The CentiScaPe plugin was used to determine the characteristics of each node in the PPI network, which gave each gene a degree score, the simplest topological index, allowing for immediate evaluation of the average number of edges (interactions) incident to the node (32). Genes with a degree ≥5 in significantly perturbed networks were defined as hub genes (mRNA). The differentially expressed lncRNA-mRNA interaction network was constructed and displayed by Cytoscape (version 3.5, http://www.cytoscape.org/). The lncRNA holding interactions with the greatest number of hub genes (mRNA) in the network were defined as hub lncRNAs.

Constructing the differentially expressed lncRNA-related disease network

Each lncRNA has a disease association profile. According to MNDR version 2.0 (33), a global view of the lncRNA-mediated disease network, which contains all of the differentially expressed lncRNA-related disease data (confidence score >0.5), was downloaded and integrated by R software. The disease network was constructed by Cytoscape. The disease label size was normalized to the number of lncRNAs related to each disease.

Hub lncRNA analysis

The hub lncRNAs were further investigated using Gene Expression Profiling Interactive Analysis, a newly developed interactive web server for analyzing the RNA sequencing expression data of 9,736 tumors and 8,587 normal samples from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression projects (34). The expression of each hub lncRNA in colon adenocarcinoma and rectum adenocarcinoma was compared with in normal tissue and to different tumor stages and plotted. Survival analysis was performed based on information from TCGA database. Relapse-free survival and OS were calculated. Hazard ratios with 95% confidence intervals and P-values calculated based on the Mantel-Cox log-rank test are displayed in Kaplan-Meier plots.

Results

Data quality control and DEG screening

A total of 32,705 genes were detected in each sample; their expression values and data distributions were similar between groups (Fig. S2A) and among samples (Fig. S2B). Principal component analysis showed that samples were easily grouped in different cell lines but not into certain drug resistance patterns (Fig. S2C). Therefore, further bioinformatics analysis should be performed separately based on different cell lines and chemoresistance. This dataset qualified for the following analysis.

Following data processing using limma, 255 DEGs in HCT116-OxPt vs. HCT116, 672 DEGs in HT29-OxPt vs. HT29, 491 DEGs in LoVo-OxPt vs. LoVo, 283 DEGs in HCT116-SN-38 vs. HCT116, 779 DEGs in HT29-SN-38 vs. HT29 and 1,713 DEGs in LoVo-SN-38 vs. LoVo were identified (Fig. S2D and E). The detailed DEGs information were shown in Tables SISVI.

Intersection between the predicted targets of lncRNAs and DEGs

Among all of the DEGs identified in each comparison, the differentially expressed lncRNAs were isolated and are listed in Table I. The target prediction of each lncRNA was searched and obtained using RAID version 2.0. Next, the target genes were intersected with the identified DEGs in each comparison and are displayed as Venn diagrams (Fig. 1A). The intersected DEGs in the same drug management were united to perform GO annotation and KEGG pathway enrichment analyses based on DAVID. All significant (P<0.05) enriched entries in OxPt resistance are shown in a histogram (Fig. 1B), with the top three being protein ubiquitination, SMAD protein signal transduction and negative regulation of protein kinase activity in the BP GO annotation category; extracellular exosome and focal adhesion in the CC category; and GTP binding, receptor binding, and calmodulin binding in the MF category. All significant (P<0.05) enriched entries in irinotecan resistance are also shown in a histogram (Fig. 1C and D), with the top three being positive regulation of transcription from RNA polymerase II promoter, positive regulation of apoptotic process, angiogenesis in the BP GO annotation category; cytoplasm, extracellular exosome, and extracellular space in the CC category; protein homodimerization activity, RNA polymerase II core promoter proximal region sequence-specific DNA binding, and growth factor activity in the MF category; and pathways in cancer, microRNAs in cancer, and TNF signaling pathway in the KEGG pathway. Detailed information on the DEGs involved in the BP GO category and KEGG pathway are listed in Fig. 1E.

Table I.

Drug resistance-related differentially expressed non-coding RNAs.

Table I.

Drug resistance-related differentially expressed non-coding RNAs.

GroupIDSymbollogFCAveExprP-valueChangeGene name
HCT116-OxPt285735LINC003261.467.31 2.1×10−05UPLong intergenic non-protein coding RNA 326
284739LINC001761.118.93 1.3×10−02UPLong intergenic non-protein coding RNA 176
HT29-OxPt283460HNF1A-AS1−1.727.65 5.0×10−05DOWNHNF1A antisense RNA 1
643911CRNDEa−1.9710.82 1.4×10−06DOWNColorectal neoplasia differentially expressed
LoVo-OxPt283120H19a1.007.06 5.4×10−04UPH19, imprinted maternally expressed transcript
64150DIO3OS−1.798.51 2.5×10−06DOWNDIO3 opposite strand/antisense RNA
196047EMX2OS−1.467.87 2.5×10−03DOWNEMX2 opposite strand/antisense RNA
HCT116-SN38652995UCA1a−1.4012.29 4.4×10−06DOWNUrothelial cancer associated 1
HT29-SN38441951ZNFX1-AS11.3510.22 9.2×10−05UPZNFX1 antisense RNA 1
727701LINC00165−1.037.43 5.5×10−05DOWNLong intergenic non-protein coding RNA 165
100169750PRINS−1.509.81 6.8×10−06DOWNPsoriasis associated RNA induced by stress
283460HNF1A-AS1−1.627.70 6.6×10−05DOWNHNF1A antisense RNA 1
554202MIR31HG−1.498.07 1.3×10−05DOWNMIR31 host gene
283131NEAT11.3211.22 9.3×10−04UPNuclear paraspeckle assembly transcript 1
100289410MCF2L-AS1−1.309.95 2.8×10−06DOWNMCF2L antisense RNA 1
643911CRNDEa−1.1711.21 2.9×10−05DOWNColorectal neoplasia differentially expressed
LoVo-SN38100093630SNHG8−1.499.80 1.3×10−03DOWNSmall nucleolar RNA host gene 8
253264ZNF503-AS1−2.287.90 8.6×10−07DOWNZNF503 antisense RNA 1
60674GAS5−1.2115.21 2.1×10−02DOWNGrowth arrest-specific 5
64150DIO3OS1.079.94 4.0×10−05UPDIO3 opposite strand/antisense RNA
196047EMX2OS−1.108.05 8.5×10−03DOWNEMX2 opposite strand/antisense RNA
128439SNHG11−1.3910.12 4.7×10−04DOWNSmall nucleolar RNA host gene 11
100124700HOTAIRa1.587.21 3.1×10−06UPHOX transcript antisense RNA
114915EPB41L4A-AS1−1.4215.27 3.0×10−02DOWNEPB41L4A antisense RNA 1
652995UCA1a−1.457.58 1.6×10−04DOWNUrothelial cancer associated 1
401237LINC003401.997.58 1.7×10−06UPLong intergenic non-protein coding RNA 340
29092LINC00339−1.559.20 4.0×10−03DOWNLong intergenic non-protein coding RNA 339
79992AGPAT4-IT11.508.13 2.5×10−04UPAGPAT4 intronic transcript 1
388815LINC004783.038.11 2.7×10−08UPLong intergenic non-protein coding RNA 478

a Hub ncRNA. OxPt, oxaliplatin; nc, noncoding.

Identification of hub lncRNAs and their associated disease network

There was a positive association between mRNA and protein expression changes; therefore, putative PPI network maps were constructed for the united datasets of chemoresistance using the STRING database and Cytoscape. Excluding the DEGs distributed on the edge of the PPI network, the remaining 26 and 57 central DEGs in the OxPt and SN-38 groups, respectively, were evaluated using the CentiScaPe plugin. The degree index of each central DEG was calculated and hub genes were defined using the criterion that the degree value must be ≥5. In the OxPt group, the hub genes were FOS, VIM, PLAUR and IGF2. In the SN-38 group, the hub genes were VEGFA, JUN, PTGS2, FOS, VIM, STAT1, CDKN1A, MMP3, CCL5, LYN, ATF3, KITLG, MMP1, FN1, CEBPB, CXCL2, MUC1, CRYZ, WNT5A and FOXA2 (Tables SVII and SVIII). These hub genes are marked in red in the PPI network (Figs. 2A and 3A). In a similar manner, the interaction network between each specific lncRNA and its potential targets in DEGs were visualized using Cytoscape in Figs. 2B and 3B. Up- and downregulated DEGs are colored in red and blue, respectively. The log2FC value is also indicated by the size of the node. Each hub lncRNA was identified by calculating its downstream hub genes whose count should be ranked in the top three and larger than 2. During OxPt screening, the lncRNAs CRNDE (target DEGs: PLAUR and FOS) and H19 (target DEGs: IGF2 and VIM) stood out. SN-38 screening revealed the following lncRNAs: CRNDE (target DEGs: STAT1, MUC1, LYN, KITLG, CXCL2, CRYZ, CCL5 and ATF3), HOTAIR (target DEGs: VEGFA, CDKN1A, VIM, MMP3, and MMP1), and UCA1 (target DEGs: CDKN1A, JUN, WNT5A, PTGS2, and FN1).

Information on lncRNA-associated diseases was obtained in full scale according to the MNDR database. By integrating this information in Cytoscape, the major related diseases associated with the differentially expressed lncRNAs were clearly sorted out. The more lncRNAs a disease is connected to, the larger its label. As shown in Fig. 4, cancer is the word that most frequently appears. Among them, CRC is obvious in the OxPt-lncRNA-related disease network, which is strongly connected to CRNDE and H19 (Fig. 4A), whereas the SN-38-lncRNA-related disease network is strongly connected to GAS5, UCA1, NEAT1, and CRNDE (Fig. 4B).

lncRNA expression validation and survival analysis in TCGA

TCGA database was utilized to validate and compare hub lncRNA expression in tumor and normal tissues. Although the results cannot relate the expression levels of these lncRNAs to chemoresistance, it did, to a certain extent, partially confirm the hazard degree of each lncRNA. As indicated in Fig. 5A-D, the expression of CRNDE and UCA1 was significantly increased in tumor tissue compared with in normal tissue (P<0.05). Except for CRNDE, the expression of three other lncRNAs increased as the TNM stage increased. In Fig. 5E-H, only HOTAIR showed a significant relationship with OS and disease-free survival (DFS; P<0.05). Patients with increased expression of HOTAIR suffered a shorter OS (HR=1.9, P=0.0066) and DFS (HR=1.8, P=0.012) compared to those with weaker expression.

Discussion

In recent years, the prevalence of CRC has increased worldwide. Due to the popularity of prophylactic screening, CRC can be diagnosed and treated increasingly early (3). However, many patients are in late stages when diagnosed (≥stage III or high-risk stage II) and require neoadjuvant or adjuvant chemotherapy (7). Some patients must face a tough reality-chemoresistance-which results in a shorter survival owing to limited chemotherapy choices. Although a previous bioinformatics analysis has revealed differences in gene expression patterns in chemoresistant cells, such as original study of GSE42387 done by Jensen et al (26), few have determined pivotal lncRNAs involved in this process. Therefore, a comprehensive analysis at the whole-genome level is necessary to identify key lncRNAs and provide detailed gene signatures for effective chemotherapy. The present study aimed at exploring the relationship between aberrant expressed lncRNAs and mRNAs. The GEO dataset was re-analyzed by relaxing DEG screening condition to gain more information. Only DEGs predicted to be the targets of differential expressed lncRNAs were involved in the following analysis.

To gain further insight into the biological pathways and identify key factors involved in chemoresistance, functional analyses of lncRNA-targeted DEGs, including GO annotation and KEGG pathway enrichment analyses were performed. The present study found that several DEGs are components of the extracellular exosome. Interestingly, the exosome is believed to confer chemoresistance to pancreatic cancer (35), breast cancer (36), gastric cancer (37) and CRC (38). It is believed that due to their cell-to-cell communication, exosomes protect tumor cells from the cytotoxic effects of chemotherapy drugs and transfer chemoresistance properties to nearby cells (39). The items enriched in GO biological processes indicate that chemoresistance occurring during both OxPt and irinotecan therapy is associated with an imbalance between cell proliferation and apoptosis regulation, cell energetic metabolism under hypoxic conditions, and angiogenesis. However, it seems that irinotecan treatment yields more DEGs than OxPt treatment. The present study hypothesized that this is why OxPt is the first choice for the clinical therapy of CRC.

In the present study, four hub lncRNAs were identified: CRNDE, H19, UCA1 and HOTAIR. CRNDE is a 1,910-nt cancer-secreted lncRNA transcribed by human chromosome 16 (16q12.2). Decades of research have shown that the high expression of CRNDE is involved in the progression of several cancers, such as renal cell carcinoma (40), hepatocellular carcinoma (41) and glioma tumors (42). Liu et al (43) found exosomal CRNDE-h in the sera of CRC patients. They also found a positive correlation between serum levels of CRNDE-h and regional lymph node or distant metastasis. Therefore, they believe that exosomal CRNDE can be used as a serum biochemical marker in the diagnosis and prognosis of CRC. Another recent study revealed that the overexpression of CRNDE promoted the development of CRC by suppressing the expression of dual-specificity phosphatase 5 (DUSP5) and CDKN1A. Moreover, the knockdown of CRNDE significantly blocked cell proliferation and induced apoptosis (44). In glioma, CRNDE promoted cell growth and invasion via mTOR signaling (42). The involvement of CRNDE in the chemoresistance of CRC treatment has also been assessed by a number of researchers (45,46). After treating CRC cells with different concentrations of 5-FU, Han et al (45) found that CRNDE-knockdown CRC cells exhibited increased sensitivity to 5-FU and OxPt treatment. Together, these results suggest that CRNDE is closely related to the development and chemoresistance of CRC.

Another hub lncRNA identified in the present study, lncRNA H19, is a key oncogenic mediator coded by chromosome 11. According to previous studies, H19 is overexpressed in various cancers, including breast cancer (47), thyroid carcinoma (48) and lung cancer (49). H19 also plays an oncogenic role in the development of CRC. The expression of H19 is increased in CRC tissues from advanced TNM stage patients (50). Enhanced H19 expression was also found in a CRC cell line and treatment with H19 significantly induced the proliferation of CRC cells, suggesting an underlying relationship between H19 and colorectal carcinogenesis (51). H19 also plays an important role in the migration and invasion of CRC cells (52). Increasing research attention has been given to the chemoresistance of H19 in the treatment of cancer (53,54). Emerging evidence also indicates that H19 knockout in the HT-29 cell line (MTX-resistant colorectal cells) reduces the risk of drug resistance. Further investigations showed that this H19-related chemoresistance is mediated by the Wnt/β-catenin signaling pathway (55).

HOTAIR is a novel oncogenic lncRNA transcribed by the HoxC gene. As with the abovementioned lncRNAs, HOTAIR has also been identified in several cancers, including osteosarcoma (56), cervical cancer (57) and gastric cancer (58). Similarly, HOTAIR exerts its oncogenic role by regulating CRC cell proliferation, invasion, and migration by mediating p21 (59). Elevated HOTAIR expression has also been detected in CRC cells and tissues relative to adjacent control tissues (60,61). The precise role of HOTAIR in chemoresistance in the chemotherapy of CRC was also recently reported. Li et al (62) indicated that HOTAIR promotes the chemoresistance of CRC cells to 5-FU by suppressing miR-218 and activating NF-κB signaling. Another study performed by Xiao et al (63) found that HOTAIR knockout improved the sensitivity of CRC cells to cisplatin and paclitaxel and that this mechanism may, at least in part, be related to the expression of miR-203a-3p and activation of the Wnt/β-catenin signaling pathway.

The regulatory role of UCA1 in chemotherapy drug resistance in CRC was also investigated. The upregulated lncRNA UCA1 is correlated with the progression of lung cancer, esophageal squamous cell carcinoma and CRC (6466) and predicts a poor prognosis. Compared with control tissues, an increased level of UCA1 was found in CRC tissues and cells and the level of UCA1 was positively correlated with tumor size and depth (67). Further in vitro experiments suggested that UCA1 induces CRC cell proliferation and inhibits apoptosis. Recent studies have also revealed the potential role of UCA1 in chemoresistance in CRC. CRC cells with UCA1 knockdown exhibit sensitivity to 5-FU by increasing tumor cell apoptosis. Conversely, UCA1 overexpression may result in increased resistance to 5-FU treatment, suggesting that UCA1 may predict the response to CRC chemotherapy (68). Furthermore, overexpression of UCA1 has a consanguineous connection with radioresistance in CRC treatment (69).

Collectively, the current study identified four pivotal lncRNAs, CRNDE, H19, UCA1 and HOTAIR, which are strongly associated with CRC chemoresistance and can be used as predictive factors for treatment sensitivity and tumor prognosis. Functional enrichment analysis indicated that an imbalance between cell proliferation and apoptosis, cell energetic metabolism under hypoxic conditions, and angiogenesis were the key biological processes whereby lncRNAs perturbed gene expression and altered sensitivity of tumor cells to chemotherapy. However, the results of the present study are based entirely on bioinformatics analyses and lack in vivo and in vitro experimental evidence. Further research is required to delineate their potential roles in chemoresistance.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used during the present study are downloaded from TCGA and GEO datasets and analyzed data is available from the corresponding author on reasonable request.

Authors' contributions

FFS was involved in the conception and design of the study, project administration and drafted the manuscript; FFS and WWL collected data from the internet, performed bioinformatics analyses and organized the figures and tables; WWL, performed the literature search. JQ was involved in the conception and design of the study, supervised the study and critically reviewed the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

O'Connell JB, Maggard MA and Ko CY: Colon cancer survival rates with the new American Joint Committee on cancer sixth edition staging. J Natl Cancer Inst. 96:1420–1425. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A and Jemal A: Colorectal cancer statistics, 2017. CA Cancer J Clin. 67:177–193. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Moriarity A, O'Sullivan J, Kennedy J, Mehigan B and McCormick P: Current targeted therapies in the treatment of advanced colorectal cancer: A review. Ther Adv Med Oncol. 8:276–293. 2016. View Article : Google Scholar : PubMed/NCBI

5 

de Mestier L, Manceau G, Neuzillet C, Bachet JB, Spano JP, Kianmanesh R, Vaillant JC, Bouché O, Hannoun L and Karoui M: Primary tumor resection in colorectal cancer with unresectable synchronous metastases: A review. World J Gastrointest Oncol. 6:156–169. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Raymond E, Faivre S, Woynarowski JM and Chaney SG: Oxaliplatin: Mechanism of action and antineoplastic activity. Semin Oncol. 25:4–12. 1998.PubMed/NCBI

7 

Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Engstrom PF, et al: NCCN guidelines insights: Colon cancer, version 2.2018. J Natl Compr Canc Netw. 16:359–369. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Bahrami A, Amerizadeh F, Hassanian SM, ShahidSales S, Khazaei M, Maftouh M, Ghayour-Mobarhan M, Ferns GA and Avan A: Genetic variants as potential predictive biomarkers in advanced colorectal cancer patients treated with oxaliplatin-based chemotherapy. J Cell Physiol. 233:2193–2201. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Hammond WA, Swaika A and Mody K: Pharmacologic resistance in colorectal cancer: A review. Ther Adv Med Oncol. 8:57–84. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Yan D, Tu L, Yuan H, Fang J, Cheng L, Zheng X and Wang X: WBSCR22 confers oxaliplatin resistance in human colorectal cancer. Sci Rep. 7:154432017. View Article : Google Scholar : PubMed/NCBI

11 

Mao L, Li Y, Zhao J, Li Q, Yang B, Wang Y, Zhu Z, Sun H and Zhai Z: Transforming growth factor-β1 contributes to oxaliplatin resistance in colorectal cancer via epithelial to mesenchymal transition. Oncol Lett. 14:647–654. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Gnoni A, Russo A, Silvestris N, Maiello E, Vacca A, Marech I, Numico G, Paradiso A, Lorusso V and Azzariti A: Pharmacokinetic and metabolism determinants of fluoropyrimidines and oxaliplatin activity in treatment of colorectal patients. Curr Drug Metab. 12:918–931. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Conti JA, Kemeny NE, Saltz LB, Huang Y, Tong WP, Chou TC, Sun M, Pulliam S and Gonzalez C: Irinotecan is an active agent in untreated patients with metastatic colorectal cancer. J Clin Oncol. 14:709–715. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Cecchin E, Corona G, Masier S, Biason P, Cattarossi G, Frustaci S, Buonadonna A, Colussi A and Toffoli G: Carboxylesterase isoform 2 mRNA expression in peripheral blood mononuclear cells is a predictive marker of the irinotecan to SN38 activation step in colorectal cancer patients. Clin Cancer Res. 11:6901–6907. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Chabot GG, Robert J, Lokiec F and Canal P: Irinotecan pharmacokinetics. Bull Cancer. 11–20. 1998.(In French). PubMed/NCBI

16 

Hecht JR: Gastrointestinal toxicity of irinotecan. Oncology (Williston Park). 12:72–78. 1998.PubMed/NCBI

17 

Xu Y and Villalona-Calero M: Irinotecan: Mechanisms of tumor resistance and novel strategies for modulating its activity. Ann Oncol. 13:1841–1851. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Ransohoff JD, Wei Y and Khavari PA: The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol. 19:143–157. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Huarte M: The emerging role of lncRNAs in cancer. Nat Med. 21:1253–1261. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Kim T and Croce CM: Long noncoding RNAs: Undeciphered cellular codes encrypting keys of colorectal cancer pathogenesis. Cancer Lett. 417:89–95. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Li H, Ma SQ, Huang J, Chen XP and Zhou HH: Roles of long noncoding RNAs in colorectal cancer metastasis. Oncotarget. 8:39859–39876. 2017.PubMed/NCBI

22 

Han D, Wang M, Ma N, Xu Y, Jiang Y and Gao X: Long noncoding RNAs: Novel players in colorectal cancer. Cancer Lett. 361:13–21. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Wang M, Han D, Yuan Z, Hu H, Zhao Z, Yang R, Jin Y, Zou C, Chen Y, Wang G, et al: Long non-coding RNA H19 confers 5-Fu resistance in colorectal cancer by promoting SIRT1-mediated autophagy. Cell Death Dis. 9:11492018. View Article : Google Scholar : PubMed/NCBI

24 

Li L, Shang J, Zhang Y, Liu S, Peng Y, Zhou Z, Pan H, Wang X, Chen L and Zhao Q: MEG3 is a prognostic factor for CRC and promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis. Oncol Rep. 38:1383–1392. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Luo J, Qu J, Wu DK, Lu ZL, Sun YS and Qu Q: Long non-coding RNAs: A rising biotarget in colorectal cancer. Oncotarget. 8:22187–22202. 2017.PubMed/NCBI

26 

Jensen NF, Stenvang J, Beck MK, Hanáková B, Belling KC, Do KN, Viuff B, Nygård SB, Gupta R, Rasmussen MH, et al: Establishment and characterization of models of chemotherapy resistance in colorectal cancer: Towards a predictive signature of chemoresistance. Mol Oncol. 9:1169–1185. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

28 

Yi Y, Zhao Y, Li C, Zhang L, Huang H, Li Y, Liu L, Hou P, Cui T, Tan P, et al: RAID v2. 0: An updated resource of RNA-associated interactions across organisms. Nucleic Acids Res. 45:D115–D118. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Huang da 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 

Huang da 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

31 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47:D607–D613. 2018. View Article : Google Scholar :

32 

Scardoni G, Tosadori G, Faizaan M, Spoto F, Fabbri F and Laudanna C: Biological network analysis with CentiScaPe: Centralities and experimental dataset integration. F1000Res. 3:e1392014. View Article : Google Scholar

33 

Cui T, Zhang L, Huang Y, Yi Y, Tan P, Zhao Y, Hu Y, Xu L, Li E and Wang D: MNDR v2.0: An updated resource of ncRNA-disease associations in mammals. Nucleic Acids Res. 46:D371–D374. 2018.PubMed/NCBI

34 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Patel GK, Khan MA, Bhardwaj A, Srivastava SK, Zubair H, Patton MC, Singh S, Khushman M and Singh AP: Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br J Cancer. 116:609–619. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, Ji MH, Hu Q, Luo Z, Wu JZ and Tang JH: Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One. 9:e952402014. View Article : Google Scholar : PubMed/NCBI

37 

Ji R, Zhang B, Zhang X, Xue J, Yuan X, Yan Y, Wang M, Zhu W, Qian H and Xu W: Exosomes derived from human mesenchymal stem cells confer drug resistance in gastric cancer. Cell Cycle. 14:2473–2483. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Hu Y, Yan C, Mu L, Huang K, Li X, Tao D, Wu Y and Qin J: Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS One. 10:e01256252015. View Article : Google Scholar : PubMed/NCBI

39 

Brinton LT, Sloane HS, Kester M and Kelly KA: Formation and role of exosomes in cancer. Cell Mol Life Sci. 72:659–671. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Ding C, Han F, Xiang H, Xia X, Wang Y, Dou M, Zheng J, Li Y, Xue W, Ding X and Tian P: LncRNA CRNDE is a biomarker for clinical progression and poor prognosis in clear cell renal cell carcinoma. J Cell Biochem. 119:10406–10414. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Wang H, Ke J, Guo Q, Barnabo Nampoukime KP, Yang P and Ma K: Long non-coding RNA CRNDE promotes the proliferation, migration and invasion of hepatocellular carcinoma cells through miR-217/MAPK1 axis. J Cell Mol Med. 22:5862–5876. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Wang Y, Wang Y, Li J, Zhang Y, Yin H and Han B: CRNDE, a long-noncoding RNA, promotes glioma cell growth and invasion through mTOR signaling. Cancer Lett. 367:122–128. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Liu T, Zhang X, Gao S, Jing F, Yang Y, Du L, Zheng G, Li P, Li C and Wang C: Exosomal long noncoding RNA CRNDE-h as a novel serum-based biomarker for diagnosis and prognosis of colorectal cancer. Oncotarget. 7:85551–85563. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Ding J, Li J, Wang H, Tian Y, Xie M, He X, Ji H, Ma Z, Hui B, Wang K and Ji G: Long noncoding RNA CRNDE promotes colorectal cancer cell proliferation via epigenetically silencing DUSP5/CDKN1A expression. Cell Death Dis. 8:e29972017. View Article : Google Scholar : PubMed/NCBI

45 

Han P, Li JW, Zhang BM, Lv JC, Li YM, Gu XY, Yu ZW, Jia YH, Bai XF, Li L, et al: The lncRNA CRNDE promotes colorectal cancer cell proliferation and chemoresistance via miR-181a-5p-mediated regulation of Wnt/β-catenin signaling. Mol Cancer. 16:92017. View Article : Google Scholar : PubMed/NCBI

46 

Gao H, Song X, Kang T, Yan B, Feng L, Gao L, Ai L, Liu X, Yu J and Li H: Long noncoding RNA CRNDE functions as a competing endogenous RNA to promote metastasis and oxaliplatin resistance by sponging miR-136 in colorectal cancer. Onco Targets Ther. 10:205–216. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Han J, Han B, Wu X, Hao J, Dong X, Shen Q and Pang H: Knockdown of lncRNA H19 restores chemo-sensitivity in paclitaxel-resistant triple-negative breast cancer through triggering apoptosis and regulating Akt signaling pathway. Toxicol Appl Pharmacol. 359:55–61. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Li M, Chai HF, Peng F, Meng YT, Zhang LZ, Zhang L, Zou H, Liang QL, Li MM, Mao KG, et al: Estrogen receptor β upregulated by lncRNA-H19 to promote cancer stem-like properties in papillary thyroid carcinoma. Cell Death Dis. 9:11202018. View Article : Google Scholar : PubMed/NCBI

49 

Huang Z, Lei W, Hu HB, Zhang H and Zhu Y: H19 promotes non-small-cell lung cancer (NSCLC) development through STAT3 signaling via sponging miR-17. J Cell Physiol. 233:6768–6776. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ and Kwok TT: Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis. 31:350–358. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Han D, Gao X, Wang M, Qiao Y, Xu Y, Yang J, Dong N, He J, Sun Q, Lv G, et al: Long noncoding RNA H19 indicates a poor prognosis of colorectal cancer and promotes tumor growth by recruiting and binding to eIF4A3. Oncotarget. 7:22159–22173. 2016.PubMed/NCBI

52 

Yang W, Redpath RE, Zhang C and Ning N: Long non-coding RNA H19 promotes the migration and invasion of colon cancer cells via MAPK signaling pathway. Oncol Lett. 16:3365–3372. 2018.PubMed/NCBI

53 

Ma H, Yuan L, Li W, Xu K and Yang L: The LncRNA H19/miR-193a-3p axis modifies the radio-resistance and chemotherapeutic tolerance of hepatocellular carcinoma cells by targeting PSEN1. J Cell Biochem. 119:8325–8335. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Si X, Zang R, Zhang E, Liu Y, Shi X, Zhang E, Shao L, Li A, Yang N, Han X, et al: LncRNA H19 confers chemoresistance in ERα-positive breast cancer through epigenetic silencing of the pro-apoptotic gene BIK. Oncotarget. 7:81452–81462. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Wu KF, Liang WC, Feng L, Pang JX, Waye MM, Zhang JF and Fu WM: H19 mediates methotrexate resistance in colorectal cancer through activating Wnt/β-catenin pathway. Exp Cell Res. 350:312–317. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Wang N, Meng X, Liu Y, Chen Y and Liang Q: LPS promote Osteosarcoma invasion and migration through TLR4/HOTAIR. Gene. 680:1–8. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Kim HJ, Lee DW, YIm GW, Nam EJ, Kim S, Kim SW and Kim YT: Long non-coding RNA HOTAIR is associated with human cervical cancer progression. Int J Oncol. 46:521–530. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Xian HP, Zhuo ZL, Sun YJ, Liang B and Zhao XT: Circulating long non-coding RNAs HULC and ZNFX1-AS1 are potential biomarkers in patients with gastric cancer. Oncol Lett. 16:4689–4698. 2018.PubMed/NCBI

59 

Lin K, Jiang H, Zhang LL, Jiang Y, Yang YX, Qiu GD, She YQ, Zheng JT, Chen C, Fang L and Zhang SY: Down-regulated LncRNA-HOTAIR suppressed colorectal cancer cell proliferation, invasion, and migration by mediating p21. Dig Dis Sci. 63:2320–2331. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Lu X, Liu Z, Ning X, Huang L and Jiang B: The long noncoding RNA HOTAIR promotes colorectal cancer progression by sponging miR-197. Oncol Res. 26:473–481. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Luo ZF, Zhao D, Li XQ, Cui YX, Ma N, Lu CX, Liu MY and Zhou Y: Clinical significance of HOTAIR expression in colon cancer. World J Gastroenterol. 22:5254–5259. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Li P, Zhang X, Wang L, Du L, Yang Y, Liu T, Li C and Wang C: lncRNA HOTAIR contributes to 5FU resistance through suppressing miR-218 and activating NF-κB/TS signaling in colorectal cancer. Mol Ther Nucleic Acids. 8:356–369. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Xiao Z, Qu Z, Chen Z, Fang Z, Zhou K, Huang Z, Guo X and Zhang Y: LncRNA HOTAIR is a prognostic biomarker for the proliferation and chemoresistance of colorectal cancer via MiR-203a-3p-mediated Wnt/ss-catenin signaling pathway. Cell Physiol Biochem. 46:1275–1285. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Wang HM, Lu JH, Chen WY and Gu AQ: Upregulated lncRNA-UCA1 contributes to progression of lung cancer and is closely related to clinical diagnosis as a predictive biomarker in plasma. Int J Clin Exp Med. 8:11824–11830. 2015.PubMed/NCBI

65 

Shalini S, Dorstyn L, Dawar S and Kumar S: Old, new and emerging functions of caspases. Cell Death Differ. 22:526–539. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Li JY, Ma X and Zhang CB: Overexpression of long non-coding RNA UCA1 predicts a poor prognosis in patients with esophageal squamous cell carcinoma. Int J Clin Exp Pathol. 7:7938–7944. 2014.PubMed/NCBI

67 

Han Y, Yang YN, Yuan HH, Zhang TT, Sui H, Wei XL, Liu L, Huang P, Zhang WJ and Bai YX: UCA1, a long non-coding RNA up-regulated in colorectal cancer influences cell proliferation, apoptosis and cell cycle distribution. Pathology. 46:396–401. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Bian Z, Jin L, Zhang J, Yin Y, Quan C, Hu Y, Feng Y, Liu H, Fei B, Mao Y, et al: LncRNA-UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting miR-204-5p. Sci Rep. 6:238922016. View Article : Google Scholar : PubMed/NCBI

69 

Yang X, Liu W, Xu X, Zhu J, Wu Y, Zhao K, He S, Li M, Wu Y, Zhang S, et al: Downregulation of long noncoding RNA UCA1 enhances the radiosensitivity and inhibits migration via suppression of epithelialmesenchymal transition in colorectal cancer cells. Oncol Rep. 40:1554–1564. 2018.PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun F, Liang W and Qian J: The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Mol Med Rep 20: 3583-3596, 2019
APA
Sun, F., Liang, W., & Qian, J. (2019). The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis. Molecular Medicine Reports, 20, 3583-3596. https://doi.org/10.3892/mmr.2019.10588
MLA
Sun, F., Liang, W., Qian, J."The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis". Molecular Medicine Reports 20.4 (2019): 3583-3596.
Chicago
Sun, F., Liang, W., Qian, J."The identification of CRNDE, H19, UCA1 and HOTAIR as the key lncRNAs involved in oxaliplatin or irinotecan resistance in the chemotherapy of colorectal cancer based on integrative bioinformatics analysis". Molecular Medicine Reports 20, no. 4 (2019): 3583-3596. https://doi.org/10.3892/mmr.2019.10588