Long non‑coding RNA MLK7‑AS1 promotes proliferation in human colorectal cancer via downregulation of p21 expression

  • Authors:
    • Rui Zhang
    • Jibin Li
    • Xiaofei Yan
    • Keer Jin
    • Wenya Li
    • Xin Liu
    • Jianfeng Zhao
    • Wen Shang
    • Xiang Zhao
  • View Affiliations

  • Published online on: November 28, 2018     https://doi.org/10.3892/mmr.2018.9702
  • Pages: 1210-1221
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Current studies have highlighted long non‑coding RNAs (lncRNAs) as critical regulators in various cancers, including colorectal cancer (CRC). By utilizing publicly available data from The Cancer Genome Atlas dataset, MLK7 antisense RNA 1 (MLK7‑AS1) was identified as a novel lncRNA that correlated with CRC progression. The results of reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) revealed a significant upregulation of MLK7‑AS1 in both CRC tissue samples and cell lines. In addition, a positive correlation was observed between increased MLK7‑AS1 expression and several clinicopathological factors in patients with CRC. Importantly, MLK7‑AS1 knockdown suppressed CRC cell proliferation and promoted G1/G0 phase arrest and apoptosis in vitro, whereas MLK7‑AS1 overexpression exhibited opposite effects. Consistently, decreased MLK7‑AS1 expression inhibited tumor growth in vivo. Furthermore, RT‑qPCR and western blot assays revealed that p21 may be a potential downstream target of MLK7‑AS1. To the best of the authors' knowledge, this is the first study to report that MLK7‑AS1 has potential as a biomarker and may promote proliferation in CRC partially through downregulating p21 expression.

Introduction

Colorectal cancer (CRC) is one of the common malignancies, with an estimation of 693,900 deaths in 2012 (1). Patients with CRC are frequently diagnosed at an advanced stage due to lack of effective diagnostic biomarkers and the prognosis is unsatisfactory even with comprehensive therapies (24). Thus, it is necessary to identify novel biomarkers and therapeutic targets for CRC patients. Identification of crucial molecules will help to accelebrate the research on CRC pathogenesis.

With the improvement of high-resolution microarray and RNA sequencing technology, it has been verified that non-coding RNAs (ncRNAs) occupies a higher rate of 98% in transcripts of human genome (57). Long non-coding RNAs (lncRNAs), newly identified counterparts of ncRNAs, have been demonstrated to be dysregulated and serve as critical regulators in various tumors (810). Current investigations have focused on the pathogenesis of lncRNAs in multiple cancers (1114). Dysregulation of lncRNAs can alter the process of many biological events, such as cell cycle, apoptosis, invasion, and epigenetic regulation (15,16). Nie et al (17), found an obvious upregulation of lncRNA ANRIL in non-small-cell lung cancer (NSCLC) tissue samples. ANRIL acts as an oncogene in CRC partly via decreasing p21 and Kruppel like factor 2 (KLF2) expression (17). In contrast, increased lncRNA-LET inhibits cell proliferation, invasion, and migration in lung cancer (18). Thus, lncRNAs may be oncogenes or tumor suppressors to mediate cancer progression. There is a critical need to investigate correlations between lncRNAs and carcinogenesis, especially CRC. However, only a small portion of lncRNAs have been functionally studied, more lncRNAs should be identified (19).

To detect aberrantly expressed lncRNAs associated with CRC progression, we analyzed The Cancer Genome Atlas (TCGA) colon cancer and normal tissue RNA sequencing data (49 normal and 648 cancer samples), and focused on a remarkably overexpressed lncRNA termed MLK7 antisense RNA 1 (MLK7-AS1). LncRNA MLK7-AS1 is located on human chromosome 2 with a length of 2551 bp. It was firstly reported to be significantly upregulated in gastric cancer (GC) tissues (20). Further, MLK7-AS1 can serve as an independent prognosis indicator in GC patients (20). However, the expression pattern, functional role, and clinical significance of MLK7-AS1 in CRC still remain uncharacterized. In this study, we show the first reporting of expression pattern and funtional role associated with MLK7-AS1 in CRC. MLK7-AS1 is significantly increased in CRC tissues and cells. Further, MLK7-AS1 overexpression exhibits tight associations with clinicopathologic factors in CRC patients. In vitro and in vivo experiments suggested that decreased MLK7-AS1 expression inhibited cell proliferative capacities, and promoted cell cycle arrest and apoptosis in CRC. Consistently, MLK7-AS1 overexpression showed opposite effects.

Cyclin-dependent kinase inhibitors (CKIs) are known to modulate cell cycle progression and function as tumor suppressors (21,22). p21 is an essential member of CKIs family, which includes p15, p16, p27 and p57 (21). Mounting studies reveal that lncRNAs can alter cancer cell phenotypes through silencing tumor suppressors and CKIs are involved in biological functions induced by lncRNAs (10,23,24). Thus, we further investigated the alteration of CKIs family expression levels in DLD-1 and LOVO cells with MLK7-AS1 knockdown. Our findings revealed that decreased MLK7-AS1 expression levels remarkably activated p21 expression at both mRNA and protein levels. Therefore, p21 is partly involved in MLK7-AS1-induced proliferation in CRC. Overall, MLK7-AS1 has potential as a biomarker for CRC patients and promotes CRC cells proliferation partly through suppressing p21 expression.

Materials and methods

Expression profiling data retrieval and analysis of lncRNAs in colorectal cancer

Expression profiling data of CRC and normal tissue samples were downloaded from the TCGA dataset. The expression data are collected from cancer patients before therapeutic intervention (25). The BAM files and normalized probe-level intensity files can be achieved from the Atlas of Non-coding RNAs in Cancer (TANRIC, http://bioinformatics.mdanderson.org/main/TANRIC:Overview) database (25). Gene annotations accords to GENCODE Release 19 annotation for lncRNAs.Reads per kilobase per million mapped reads (RPKM) values were calculated using TCGA RNA-sequencing data in the BAM files.

Tumor tissue samples and matched non-tumor tissue samples

The CRC tissues (n=50) and matched non-tumor tissue samples (n=50) were obtained from CRC patients with surgical resection in Cancer Hospital of China Medical University. The patients who participated in this study do so in the context of informed consents. Median value was selected as cut-off point to better compare correlations between clinical pathological factors and the expression level of certain gene in patients with cancer (10,26,27). In the present study, we divided 50 PC patients into two groups: the high MLK7-AS1 group (n=25, fold change above the median value); and the low MLK7-AS1 group (n=25, fold change below the median value) according to the median value of MLK7-AS1 levels. The study was approved by the Cancer Hospital of China Medical University Ethics Committee. The collected tissue samples were instantly stored in a liquid nitrogen, and then transferred to be kept at −80°C.

Cell culture

Human colonic epithelial cells (HCoEpiC) and CRC cells (SW480, HCT116, LOVO and DLD-1) are all cultured in DMEM medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), 100 mg/ml streptomycin, and 100 U/ml penicillin (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C with 5% O2.

Reverse transcription-quantitative polymerase chain reaction PCR (RT-qPCR) analyses

A Reverse Transcription Kit (Thermo Fisher Scientific Inc.) was used to reverse total RNA into cDNA. Then, cDNA and primers were used to perform qRT-PCR assays on 7500 Real-Time PCR System (Thermo Fisher Scientific Inc.). The specific primers are listed in Table II. The results downloaded from this instrument were then normalized to GAPDH expression. The collected data were analyzed, expressed relative to threshold cycle values, and then switched to fold changes. Each sample was analyzed in triplicate.

Table II.

Sequences of specific primers for reverse transcription-quantitative polymerase chain reaction.

Table II.

Sequences of specific primers for reverse transcription-quantitative polymerase chain reaction.

Primer sequences (5′-3′)

GeneForwardReverse
MLK7-AS1 CAGCCTCCCGAGTTGAGTAA CAAATGACACGAGCCTTCCT
GAPDH GAAGAGAGAGACCCTCACGCTG ACTGTGAGGAGGGGAGATTCAGT
p15 ACGGAGTCAACCGTTTCGGGAG GGTCGGGTGAGAGTGGCAGG
p16 ATGGAGCCTTCGGCTGACT GGCCTCCGACCGTAACTATT
p21 CAGCAGAGGAAGACCATGTG GGCGTTTGGAGTGGTAGAAA
p27 TGCAACCGACGATTCTTCTACTCAA CAAGCAGTGATGTATCTGATAAACAAGG
p57 CACGATGGAGCGTCTTGTC CCTGCTGGAAGTCGTAATCC
Cell transfection

CRC cell lines were transfected with three individual MLK7-AS1 (MLK7-AS1 no. 1, no. 2 and no. 3), scrambled negative control (NC) small interfering RNAs (siRNAs), as well as vectors such as pcDNA-MLK7-AS1, empty vector, and sh-MLK7-AS1-vector. SiRNAs were transfected into cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Plasmid vectors were extracted by DNA Midiprep kit (Qiagen GmbH, Hilden, Germany) and transfected into cells using Fugene (Roche Diagnostics, Basel, Switzerland). The full-length complementary DNA of MLK7-AS1 was synthesized by Realgene (China), and subcloned into the pcDNA3.1(+) vector (Invitrogen; Thermo Fisher Scientific, Inc.). Up to transfection after 48 h, collected cells were used to conduct qRT-PCR and western blot experiments. The sequences for siRNAs and shRNAs are listed in Table III.

Table III.

Sequences of siRNAs and shRNAs.

Table III.

Sequences of siRNAs and shRNAs.

siRNAs
  si-MLK7-AS1 no. 1 GAGAGUACUUUGGUCACCACGGGAA
  si-MLK7-AS1 no. 2 CCAAGGGCUCUGUUCAUAAACUGUU
  si-MLK7-AS1 no. 3 CCAAGCUACUUGUAAUCCUUCCAAA
shRNAs
  sh-MLK7-AS1 no. 1 CACCGTTCCCGTGGTGACCAAAGTACTCTCCGAAGAGAGTACTTTGGTCACCACGGGAA
  sh-MLK7-AS1 no. 2 CACCGAACAGTTTATGAACAGAGCCCTTGGCGAACCAAGGGCTCTGTTCATAAACTG
  sh-MLK7-AS1 no. 3 CACCGTTTGGAAGGATTACAAGTAGCTTGGCGAACCAAGCTACTTGTAATCCTTCCAAA

[i] Si, small interfering; sh, short hairpin.

Assays of MTT and colony formation

The cells are cultured in 96-well plates. MTT experiments were conducted to test cell viability at 490-nm-wavelength by Microplate Reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). For assays of colony formation, cells with transfection after 48 h are placed in six-well plates. Up to 14 days, methanol-fixed colonies were stained with crystal violet of 0.1% and then colony numbers in each group can be counted.

EdU assay

5-ethynyl2-deoxyuridine (EdU) labeling/detection kit (Guangzhou RiboBio Co., Ltd., Guangzhou, China) was used to assess proliferating cells. SW480 and LOVO cells were cultivated in 96-well plates and transfected with siRNAs for 48 h. Then, the cells were cultured in EdU labeling medium and incubated for 2 h at 37°C under 5% CO2. After treatment with 4% paraformaldehyde and 0.5% Triton X-100, anti-EdU working solution was ued to stain cells. And DAPI was used to label cell nuclei. The percentage of EdU-positive cells was calculated from five random fields in three wells.

Flow cytometry

DLD1 and LOVO cells with 48 h transfection of si-MLK7-AS1 no. 1, no. 2 and si-NC were stained with FITC-Annexin-V and propidium iodide (PI). Then, flow cytometry (FACScan®; BD Biosciences, Franklin Lakes, NJ, USA) was used to detect alterations of cell cycle. Furthermore, cells were classified into viable cells, dead cells, early apoptotic cells, and apoptotic cells through analysis of flow cytometry.

Western blot analysis and antibodies

RIPA buffer with proteinase inhibitor cocktail (Medchem Express, NJ, USA) was used to lyse cells. The membranes were incubated with antibodies against CDK2, CDK4 and P21. GAPDH was used as a reference control. All the antibodies are obtained from Cell Signaling Technology, Inc., (Danvers, MA, USA).

Xenograft model in nude mice

Male athymic BALB/c nude mice of 4 weeks old were purchased from Institute of laboratory animal medicine, Chinese Academy of Medical Sciences. The study accords with rules of Cancer Hospital of China Medical University Ethics Committee. A total of 100 µl empty-vector-transfected or sh-MLK7-AS1-transfected DLD-1 cells was respectively injected into a single side of each mouse. Measurement of tumor volume was conducted every four days. Up to twenty-four days after injection, the mice were killed and tumors removed from the mice were kept in 4% paraformaldehyde for further research.

Immunohistochemical (IHC) analysis

The tumor tissues derived from control group and sh-MLK7-AS1 group were immunostained for H&E and Ki67. Anti-Ki67 from Santa Cruz Biotechnology, Inc., (Dallas, TX, USA) was used to present the percentage of positive cells, further revealing the proliferative activities in tumor tissues.

Statistical analysis

All assays were repeated for three times. The data were analyzed with SPSS v17.0 software program (SPSS, Inc., Chicago, IL, USA) and presented as mean ± SD (standard deviation). Student's t test (two tailed) was used to compare data derived from different groups. P<0.05 was considered to indicate a statistically significant difference.

Results

MLK7-AS1 is obviously upregulated in CRC tissue samples, and increased MLK7-AS1 expression is significantly correlated with several clinicopathological factors and poor prognosis in patients with CRC

To investigate aberrantly expressed lncRNAs involved in CRC, we analyzed the data downloaded from the TCGA dataset, and found that lncRNA MLK7-AS1 exhibited obvious upregulation in CRC tissue samples relative to normal tissues (Fig. 1A). Then, qRT-PCR experiments were used to determine the expression levels of MLK7-AS1 in 50 pairs CRC tissue samples and matched non-tumor tissue samples. MLK7-AS1 exhibits significant upregulation in CRC tissues. Fold change >1.5 was recognized to be significant. Then, we divided CRC patients into high (above the median value, n=25) and low (below the median value, n=25) MLK7-AS1 expression groups to better study correlations between MLK7-AS1 expression levels and clinicopathological features (Fig. 1B). As shown in Fig. 1D and Table I, increased MLK7-AS1 expression was obviously linked with tumor size (P=0.011), TNM stage (P=0.031), and lymph node metastasis (P=0.031) in CRC patients.

Table I.

Correlation between MLK7-AS1 expression and clinicopathological factors of colorectal cancer patients.

Table I.

Correlation between MLK7-AS1 expression and clinicopathological factors of colorectal cancer patients.

MLK7-AS1 expression

VariablesHighLowP-value
Age (years)
  >6011160.156
  ≤6014  9
Gender
  Male17171.000
  Female  8  8
Tumor size
  ≤5 cm  9180.011a
  >5 cm16  7
TNM stage
  I/II  4110.031a
  III/IV2114
Lymph node metastasis
  Positive21140.031a
  Negative  411

a P<0.05.

To detect the correlation between MLK7-AS1 expression and the prognosis of CRC patients, Kaplan-Meier analysis and log-rank test were used to explore the effects of MLK7-AS1 on overall survival of CRC patients (Fig. 1G). The median survival time for cases with high MLK7-AS1 expression was 14 months, whereas it was 25 months for low MLK7-AS1 expression. Furthermore, the overall survival rate over 2 years for the low MLK7-AS1 expression group was 32%, while it was 16% for the high MLK7-AS1 expression group. Our findings show that MLK7-AS1 is an unfavourable prognostic factor for CRC patients.

Regulation of MLK7-AS1 expression in CRC cell lines

We detected MLK7-AS1 expression levels in CRC cell lines, found that MLK7-AS1 exhibited higher levels in DLD-1 and LOVO cells (Fig. 1C). In attempt to evaluate the role of MLK7-AS1 in CRC cells, MLK7-AS1 expression was decreased by transfection with siRNAs or shRNA vector. And qPCR assays were used to test the interference efficiencies of three siRNAs transfected in CRC cells. Si-MLK7-AS1 no. 1 and si-MLK7-AS1 no. 2 exhibited more efficient silencing abilities than si-MLK7-AS1 no. 3 (Fig. 1E). Thus, we selected si-MLK7-AS1 no. 1 and si-MLK7-AS1 no. 2 for all subsequent assays. Moreover, we also examine the expression levels of MLK7-AS1 in DLD-1 and LOVO cells transfected with pcDNA-MLK7-AS1. Compared with the NC, MLK7-AS1 expression significantly increased in pcDNA-MLK7-AS1-transfected CRC cells (Fig. 1F).

The effects of MLK7-AS1 dysregulation on cell viability and colony-formation ability in CRC cells

MTT experiments were performed to test CRC cells viability, and the results demonstrated that cell viabilities of DLD1 and LOVO cells following transfection with si-MLK7-AS1 no. 1 or si-MLK7-AS1 no. 2 were obviously suppressed compared with control cells (Fig. 2A). Furthermore, MLK7-AS1 knockdown impaired CRC cells clonogenic survival in DLD-1 and LOVO cells (Fig. 2B). Consistently, the results of MLK7-AS1 overexpression showed opposite effects (Fig. 2A and B). These findings indicated the effects of MLK7-AS1 on CRC proliferation.

The function of MLK7-AS1 in cell cycle progression and apoptosis of CRC cell lines

To explore whether MLK7-AS1 is involved in cell cycle regulations, flow cytometry assays were performed in DLD-1 and LOVO cell lines. Compared with control cells, DLD1 and LOVO cells with MLK7-AS1 knockdown showed an obvious G1/G0 phase arrest (Fig. 3A and B). Importantly, flow cytometry experiments were further used to study the effects of MLK7-AS1 overexpression on cell cycle regulation in DLD-1 and LOVO cell lines. Consistently, the results showed that DLD-1 and LOVO cells with MLK7-AS1 overexpression had a significant decrease in G1/G0 phase and an obvious increase in G2/S phase (Fig. 4A and B). Edu staining assays also revealed the proliferation promotion mediated by MLK7-AS1 in CRC (Fig. 5A). As known to all, CKIs exert indispensable roles in cell cycle progression and serve as tumor suppressors in many cancers, including CRC (28) Moreover, western blot assays revealed the significant alteration of cyclin dependent kinase 2 (CDK2) and cyclin dependent kinase 4 (CDK4) in DLD-1 and LOVO cells with MLK7-AS1 knockdown (Fig. 4C).

To comfirm the findings that MLK7-AS1 exerts regulatory roles in cell cycle regulation, we investigated the alteration of CKIs family in DLD-1 and LOVO cells following transfection with si-MLK7-AS1 no. 1 or si-MLK7-AS1 no. 2. Interestingly, qPCR and western blot experiments both demonstrated that p21 was dramatically increased in DLD-1 and LOVO cells with MLK7-AS1 knockdown (Fig. 5B). These results highlighted p21 as a novel target gene of MLK7-AS1. Next, we use flow cytometry to study whether MLK7-AS1 could induce CRC cells apoptosis. The results showed that downregulation of MLK7-AS1 by MLK7-AS1 siRNAs significantly increased apoptotic abilities of CRC cells (Fig. 3C and D). These findings suggest that MLK7-AS1 promotes CRC cells proliferative abilities partly via silencing p21 expression.

MLK7-AS1 downregulation inhibited tumor growth in mice

Sh-MLK7-AS1-transfected or empty-vector-transfected DLD-1 cells are injected into nude mice to establish in vivo models (10,29). Up to 24 days after injection, the size of tumor formed from sh-MLK7-AS1-transfected DLD-1 cells was dramatically smaller than the size of tumor in control group (Fig. 6A). Additionally, the tumor volumes and weights were obviously decreased compared with the controls (Fig. 6C and D). The expression of MLK7-AS1 in tumors derived from DLD-1 cells with MLK7-AS1 knockdown exhibited remarkable downregulation, relative to that of control (Fig. 6B). Immunohistochemistry (IHC) assays demonstrated that the tumor tissues formed from DLD1/sh-MLK7-AS1 cells displayed lower Ki-67 expression (Fig. 6E). These results suggested that MLK7-AS1 knockdown could inhibit CRC cells growth in vivo.

Discussion

Advances in sequencing technologies facilitate the completion of new massively human sequencing projects, such as the Encyclopedia of DNA Elements (ENCODE) and GENCODE (30,31). LncRNAs are known as newly identified ncRNAs, which occupies a higher rate of 98% in transcripts of human genome (8). Emerging evidence has highlighted lncRNAs as critical regulators in multiple biological processes, and the dysregulation of lncRNAs has been found to exert important roles in various cancers (8,16,17,23). For example, lncRNA HOXA11-AS is found to be significantly upregulated in GC tissues and alters GC cells phenotypes by modulating cell cycle, apoptosis, and invasion (32). Importantly, lncRNAs exhibit different cell phenotypes in various cancer, and its expression patterns are tissue-specific (32). However, only a few of lncRNAs have been well-studied in cancer progression (33,34). Thus, more lncRNAs should be investigated in tumors, especially CRC.

Here, we utilize publicly available data from TCGA dataset and focus on the overexpressed lncRNAs. LncRNA MLK7-AS1 exhibited obvious upregulation in expression data of CRC and was screened out as a potential oncogene in CRC progression. Then, qRT-PCR experiments were performed to validate the expression levels of MLK7-AS1 in a cohort of 50 paired CRC tissue samples and matched non-tumor samples. Overexpression of MLK7-AS1 is significantly correlated with tumor size, TNM stage, and lymph node metastasis in CRC patients. Functional studies revealed that MLK7-AS1 knockdown promoted CRC cells apoptotic abilities, suppressed proliferation in vitro, and contributed to tumor growth inhibition in vivo. Consistently, upregulation of MLK7-AS1 showed opposite effects. These findings indicated that MLK7-AS1 may be an oncogene in CRC progression, suggesting its utilities as a potential biomarker and a therapeutic target.

Mounting investigations have focused on the antisense transcripts and its corresponding protein-coding genes. For example, lncRNA KRT7-AS promotes GC cell progression by increasing KRT7 expression (35). LncRNA FEZF1-AS1 facilitates cell proliferation and migration in CRC through mediating FEZF1 expression (36). These studies suggested a novel mechanism that sense gene regulation can be controlled by antisense transcripts through forming duplex. LncRNA MLK7-AS1 is located on the antisense chain of the gene coding MLK7 protein. MLK7 has been demonstrated to be significantly upregulated in CRC tissues (37,38). CRC RNA-sequencing data showed that MLK7 increased by about threefold in CRC tissue samples (8). Both MLK7 and MLK7-AS1 exhibited obvious upregulation in CRC tissues. However, the regulatory relationship between MLK7-AS1 and MLK7 remains undefined. The potential mechanism between MLK7-AS1 and MLK7 deserves to be focused in the future research.

Recently, it has been revealed that lncRNAs can increase or inhibit the expression levels of target genes to exert effects in cancer progression (24). CKIs are known to be critical tumor suppressors in various cancers and play key roles in cell cycle regulation (10,22,28) Importantly, CKIs are involved in the biological role of lncRNAs (28,39,40). The abnormal methylation in promoter regions of CKIs leads to gene expression inhibition, thus contributing to alteration of cell cycle (41,42). For example, linc00668-mediated cell proliferation in GC is partly via silencing the expressions of CKIs at epigenetical levels (24). Thus, we further examined the expression levels of CKIs in CRC cells after MLK7-AS1 knockdown. Interestingly, p21 expression was obviously upregulated at both mRNA and protein levels. Current studies have highlighted p21 as a critical tumor suppressor in many cancers, including CRC (21,43). For example, BRAF activated non-coding RNA (BANCR) could promote CRC proliferation by silencing p21 (44). The significant increase of p21 can partly explain the finding that decreased MLK7-AS1 contributes to significant increase in G1/G0 phase, and activates apoptosis in CRC cells. Thus, MLK7-AS1 could promote CRC cell proliferation and induce apoptosis partly via silencing p21 expression. SH2 Domain-Containing Inositol-5′-Phosphatase 1 (SHIP1) has been reported to be involved in pathways associated with p21 (45). Additionally, dysregulated SHIP1 exerts regulatory effects on cell viability and cell death in many diseases, including CRC (4648). It is also of great significance to investigate the relationship between MLK7-AS1 and other potential target gene, including SHIP1.

P21, also known as cyclin dependent kinase inhibitor 1A (CDKN1A), is a crucial component of CKIs family, and has been elucidated to mediate tumor-suppressive activities in many cancers (21,49,50). Current studies have showed the effects of lncRNAs on cell cycle regulation (51). Identifications of lncRNAs and associated regulatory target genes in CRC are important, and will help to explore CRC pathogenesis. Our findings represent first reporting of remarkable upregulation of MLK7-AS1 in CRC tissue samples and cell lines. MLK7-AS1 inhibition can decrease cell proliferative abilities and promote apoptosis in CRC. Importantly, p21 acts as a novel downstream target of MLK7-AS1. The oncogenic role of MLK7-AS1 in CRC progression is partly via silencing p21 expression. Additionally, overexpressed MLK7-AS1 expression may be an unfavourable prognostic factor in CRC patients. Although the functional role of MLK7-AS1 has been investigated, mechanistic investigations between MLK7-AS1 and p21 are required to further the understanding of regulatory mechanism. More numbers of mice can be used to study the effects of MLK7-AS1 dysregulation on tumor growth in vivo in the future. Our results support the idea that lncRNA MLK7-AS1 promotes the proliferation in human CRC partly via downregulating p21 expression and suggest that lncRNA MLK7-AS1 may be a potential therapeutic target for CRC patients. Theses findings will shed new light on CRC pathogenesis and promote the development of lncRNAs-directed diagnosis and treatments.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Fund from the National Natural Science Foundation of China (grant no. 81672427); the project of ‘Liaoning clinical research center for colorectal cancer’ (grant no. 2015225005); ‘Liaoning BaiQianWan Talents Program’ [2017] No. B44 and ‘Clinical capability construction project for Liaoning Provincial Hospitals (grant no. LNCCC-D42-2015)’.

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

XZ, RZ, JBL and XFY made substantial contributions to conception and design. RZ, KJ and WYL were responsible for the analysis and interpretation of data. RZ, XL, JFZ and SW were involved the experimental design, drafting the manuscript and revising it critically for important intellectual content. XZ agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The study was approved by the Cancer Hospital of China Medical University Ethics Committee. Informed consent was obtained from participants.

Consent for publication

Informed consent was obtained from participants.

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 

Garcia-Foncillas J and Diaz-Rubio E: Progress in metastatic colorectal cancer: Growing role of cetuximab to optimize clinical outcome. Clin Transl Oncol. 12:533–542. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Dienstmann R, Salazar R and Tabernero J: Personalizing colon cancer adjuvant therapy: Selecting optimal treatments for individual patients. J Clin Oncol. 33:1787–1796. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Matuchansky C: Colorectal cancer: Some present aspects of its epidemiology, prevention and screening. Presse Med. 46:141–144. 2017.(In French). View Article : Google Scholar : PubMed/NCBI

5 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Xu J, Zhao J, Liu F and Zhang R: Analysis of mechanism and feature genes of colorectal cancer by bioinformatic methods. Minerva Med. 108:94–95. 2017.PubMed/NCBI

7 

Zhao B, Lu M, Wang D, Li H and He X: Genome-wide identification of long noncoding RNAs in human intervertebral disc degeneration by RNA sequencing. Biomed Res Int. 2016:36848752016. View Article : Google Scholar : PubMed/NCBI

8 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar : PubMed/NCBI

9 

He Y, Meng XM, Huang C, Wu BM, Zhang L, Lv XW and Li J: Long noncoding RNAs: Novel insights into hepatocelluar carcinoma. Cancer Lett. 344:20–27. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Su J, Zhang E, Han L, Yin D, Liu Z, He X, Zhang Y, Lin F, Lin Q, Mao P, et al: Long noncoding RNA BLACAT1 indicates a poor prognosis of colorectal cancer and affects cell proliferation by epigenetically silencing of p15. Cell Death Dis. 8:e26652017. View Article : Google Scholar : PubMed/NCBI

11 

Wu X, Yan T, Wang Z, Wu X, Cao G and Zhang C: LncRNA ZEB2-AS1 promotes bladder cancer cell proliferation and inhibits apoptosis by regulating miR-27b. Biomed Pharmacother. 96:299–304. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Z, Fu C, Xu Q and Wei X: Long non-coding RNA CASC7 inhibits the proliferation and migration of colon cancer cells via inhibiting microRNA-21. Biomed Pharmacother. 95:1644–1653. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Kondo Y, Shinjo K and Katsushima K: Long non-coding RNAs as an epigenetic regulator in human cancers. Cancer Sci. 108:1927–1933. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Kwok ZH and Tay Y: Long noncoding RNAs: Lincs between human health and disease. Biochem Soc Trans. 45:805–812. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Gutschner T and Diederichs S: The hallmarks of cancer: A long non-coding RNA point of view. Rna Biol. 9:703–719. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Dey BK, Mueller AC and Dutta A: Long non-coding RNAs as emerging regulators of differentiation, development, and disease. Transcription. 5:e9440142014. View Article : Google Scholar : PubMed/NCBI

17 

Nie FQ, Sun M, Yang JS, Xie M, Xu TP, Xia R, Liu YW, Liu XH, Zhang EB, Lu KH and Shu YQ: Long noncoding RNA ANRIL promotes non-small cell lung cancer cell proliferation and inhibits apoptosis by silencing KLF2 and P21 expression. Mol Cancer Ther. 14:268–277. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Liu B, Pan CF, He ZC, Wang J, Wang PL, Ma T, Xia Y and Chen YJ: Long noncoding RNA-LET suppresses tumor growth and EMT in lung adenocarcinoma. Biomed Res Int. 2016:46934712016. View Article : Google Scholar : PubMed/NCBI

19 

Guil S and Esteller M: Cis-acting noncoding RNAs: Friends and foes. Nat Struct Mol Biol. 19:1068–1075. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Ren W, Zhang J, Li W, Li Z, Hu S, Suo J and Ying X: A tumor-specific prognostic long non-coding RNA signature in gastric cancer. Med Sci Monit. 22:3647–3657. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Abbas T and Dutta A: p21 in cancer: Intricate networks and multiple activities. Nat Rev Cancer. 9:400–414. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Cress WD, Yu P and Wu J: Expression and alternative splicing of the cyclin-dependent kinase inhibitor-3 gene in human cancer. Int J Biochem Cell Biol. 91:98–101. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Sun CC, Li SJ, Li G, Hua RX, Zhou XH and Li DJ: Long intergenic noncoding RNA 00511 acts as an oncogene in non-small-cell lung cancer by binding to EZH2 and suppressing p57. Mol Ther Nucleic Acids. 5:e3852016. View Article : Google Scholar : PubMed/NCBI

24 

Zhang E, Yin D, Han L, He X, Si X, Chen W, Xia R, Xu T, Gu D, De W, et al: E2F1-induced upregulation of long noncoding RNA LINC00668 predicts a poor prognosis of gastric cancer and promotes cell proliferation through epigenetically silencing of CKIs. Oncotarget. 7:23212–23226. 2016.PubMed/NCBI

25 

Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, Weinstein JN and Liang H: TANRIC: An interactive open platform to explore the function of lncRNAs in cancer. Cancer Res. 75:3728–3737. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Zhao L, Guo H, Zhou B, Feng J, Li Y, Han T, Liu L, Li L, Zhang S, Liu Y, et al: Long non-coding RNA SNHG5 suppresses gastric cancer progression by trapping MTA2 in the cytosol. Oncogene. 35:5770–5780. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Ma HW, Xie M, Sun M, Chen TY, Jin RR, Ma TS, Chen QN, Zhang EB, He XZ, De W and Zhang ZH: The pseudogene derived long noncoding RNA DUXAP8 promotes gastric cancer cell proliferation and migration via epigenetically silencing PLEKHO1 expression. Oncotarget. 8:52211–52224. 2016.PubMed/NCBI

28 

Kato S, Schwaederle M, Daniels GA, Piccioni D, Kesari S, Bazhenova L, Shimabukuro K, Parker BA, Fanta P and Kurzrock R: Cyclin-dependent kinase pathway aberrations in diverse malignancies: Clinical and molecular characteristics. Cell Cycle. 14:1252–1259. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Kim T, Jeon YJ, Cui R, Lee JH, Peng Y, Kim SH, Tili E, Alder H and Croce CM: Role of MYC-regulated long noncoding RNAs in cell cycle regulation and tumorigenesis. J Natl Cancer Inst. 107(pii): dju5052015.PubMed/NCBI

30 

Esteller M: Non-coding RNAs in human disease. Nat Rev Genet. 12:861–874. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, Oyama R, Ravasi T, Lenhard B, Wells C, et al: The transcriptional landscape of the mammalian genome. Science. 309:1559–1563. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Sun M, Nie F, Wang Y, Zhang Z, Hou J, He D, Xie M, Xu L, De W, Wang Z and Wang J: LncRNA HOXA11-AS promotes proliferation and invasion of gastric cancer by scaffolding the chromatin modification factors PRC2, LSD1, and DNMT1. Cancer Res. 76:6299–6310. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Li S, Li B, Zheng Y, Li M, Shi L and Pu X: Exploring functions of long noncoding RNAs across multiple cancers through co-expression network. Sci Rep. 7:7542017. View Article : Google Scholar : PubMed/NCBI

34 

Wang Q, Yang H, Wu L, Yao J, Meng X, Jiang H, Xiao C and Wu F: Identification of specific long non-coding RNA expression: profile and analysis of association with clinicopathologic characteristics and BRAF mutation in papillary thyroid cancer. Thyroid. 26:1719–1732. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Huang B, Song JH, Cheng Y, Abraham JM, Ibrahim S, Sun Z, Ke X and Meltzer SJ: Long non-coding antisense RNA KRT7-AS is activated in gastric cancers and supports cancer cell progression by increasing KRT7 expression. Oncogene. 35:4927–4936. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Chen N, Guo D, Xu Q, Yang M, Wang D, Peng M, Ding Y, Wang S and Zhou J: Long non-coding RNA FEZF1-AS1 facilitates cell proliferation and migration in colorectal carcinoma. Oncotarget. 7:11271–11283. 2016.PubMed/NCBI

37 

Liu J, McCleland M, Stawiski EW, Gnad F, Mayba O, Haverty PM, Durinck S, Chen YJ, Klijn C, Jhunjhunwala S, et al: Integrated exome and transcriptome sequencing reveals ZAK isoform usage in gastric cancer. Nat Commun. 5:38302014. View Article : Google Scholar : PubMed/NCBI

38 

Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, Chaudhuri S, Guan Y, Janakiraman V, Jaiswal BS, et al: Recurrent R-spondin fusions in colon cancer. Nature. 488:660–664. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Lim S and Kaldis P: Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development. 140:3079–3093. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Prensner JR and Chinnaiyan AM: The emergence of lncRNAs in cancer biology. Cancer Discov. 1:391–407. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Liu C, Li S, Dai X, Ma J, Wan J, Jiang H, Wang P, Liu Z and Zhang H: PRC2 regulates RNA polymerase III transcribed non-translated RNA gene transcription through EZH2 and SUZ12 interaction with TFIIIC complex. Nucleic Acids Res. 43:6270–6284. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Ferrè F, Colantoni A and Helmer-Citterich M: Revealing protein-lncRNA interaction. Brief Bioinform. 17:106–116. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Yu B, Ye X, Du Q, Zhu B, Zhai Q and Li XX: The long non-coding RNA CRNDE promotes colorectal carcinoma progression by competitively binding miR-217 with TCF7L2 and Enhancing the Wnt/β-catenin signaling pathway. Cell Physiol Biochem. 41:2489–2502. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Shi Y, Liu Y, Wang J, Jie D, Yun T, Li W, Yan L, Wang K and Feng J: Downregulated long noncoding RNA BANCR promotes the proliferation of colorectal cancer cells via downregualtion of p21 expression. PLoS One. 10:e1226792015.

45 

An H, Xu H, Zhang M, Zhou J, Feng T, Qian C, Qi R and Cao X: Src homology 2 domain-containing inositol-5-phosphatase 1 (SHIP1) negatively regulates TLR4-mediated LPS response primarily through a phosphatase activity- and PI-3K-independent mechanism. Blood. 105:4685–4692. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Fuhler GM, Brooks R, Toms B, Iyer S, Gengo EA, Park MY, Gumbleton M, Viernes DR, Chisholm JD and Kerr WG: Therapeutic potential of SH2 domain-containing inositol-5′-phosphatase 1 (SHIP1) and SHIP2 inhibition in cancer. Mol Med. 18:65–75. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Hoekstra E, Das AM, Willemsen M, Swets M, Kuppen PJ, van der Woude CJ, Bruno MJ, Shah JP, Ten Hagen TL, Chisholm JD, et al: Lipid phosphatase SHIP2 functions as oncogene in colorectal cancer by regulating PKB activation. Oncotarget. 7:73525–73540. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Rouquette-Jazdanian AK, Kortum RL, Li W, Merrill RK, Nguyen PH, Samelson LE and Sommers CL: miR-155 controls lymphoproliferation in LAT mutant mice by restraining T-cell apoptosis via SHIP-1/mTOR and PAK1/FOXO3/BIM pathways. PLoS One. 10:e1318232015. View Article : Google Scholar

49 

Won KY, Kim GY, Kim HK, Choi SI, Kim SH, Bae GE, Lim JU and Lim SJ: Tumoral FOXP3 expression is associated with favorable clinicopathological variables and good prognosis in gastric adenocarcinoma: The tumor suppressor function of tumoral FOXP3 is related with the P21 expression in gastric adenocarcinoma. Hum Pathol. 68:112–118. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Chen Z, Chen X, Chen P, Yu S, Nie F, Lu B, Zhang T, Zhou Y, Chen Q, Wei C, et al: Long non-coding RNA SNHG20 promotes non-small cell lung cancer cell proliferation and migration by epigenetically silencing of P21 expression. Cell Death Dis. 8:e30922017. View Article : Google Scholar : PubMed/NCBI

51 

Kitagawa M, Kitagawa K, Kotake Y, Niida H and Ohhata T: Cell cycle regulation by long non-coding RNAs. Cell Mol Life Sci. 70:4785–4794. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 19 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang R, Li J, Yan X, Jin K, Li W, Liu X, Zhao J, Shang W and Zhao X: Long non‑coding RNA MLK7‑AS1 promotes proliferation in human colorectal cancer via downregulation of p21 expression. Mol Med Rep 19: 1210-1221, 2019
APA
Zhang, R., Li, J., Yan, X., Jin, K., Li, W., Liu, X. ... Zhao, X. (2019). Long non‑coding RNA MLK7‑AS1 promotes proliferation in human colorectal cancer via downregulation of p21 expression. Molecular Medicine Reports, 19, 1210-1221. https://doi.org/10.3892/mmr.2018.9702
MLA
Zhang, R., Li, J., Yan, X., Jin, K., Li, W., Liu, X., Zhao, J., Shang, W., Zhao, X."Long non‑coding RNA MLK7‑AS1 promotes proliferation in human colorectal cancer via downregulation of p21 expression". Molecular Medicine Reports 19.2 (2019): 1210-1221.
Chicago
Zhang, R., Li, J., Yan, X., Jin, K., Li, W., Liu, X., Zhao, J., Shang, W., Zhao, X."Long non‑coding RNA MLK7‑AS1 promotes proliferation in human colorectal cancer via downregulation of p21 expression". Molecular Medicine Reports 19, no. 2 (2019): 1210-1221. https://doi.org/10.3892/mmr.2018.9702