ANRIL is associated with the survival rate of patients with colorectal cancer, and affects cell migration and invasion in vitro

  • Authors:
    • Yi Sun
    • Zhao‑Peng Zheng
    • Hang Li
    • Han‑Qun Zhang
    • Fa‑Qiang Ma
  • View Affiliations

  • Published online on: June 17, 2016     https://doi.org/10.3892/mmr.2016.5409
  • Pages: 1714-1720
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Antisense noncoding RNA in the INK4 locus (ANRIL) has been reported to be upregulated in various types of human cancer, and is also highly expressed in normal human tissue. The aim of the present study was to identify whether ANRIL may be a possible target for colorectal cancer (CRC) therapy. Reverse transcription‑quantitative polymerase chain reaction was used to quantify the expression levels of the long noncoding RNA (lncRNA) ANRIL in 97 paired CRC and adjacent non‑neoplastic tissue samples. In addition, the HT29 and RKO human CRC cell lines underwent ANRIL RNA interference, and knockdown efficiency was evaluated by western blotting. Cell viability, and migratory and invasive ability were subsequently assessed. The CRC tissues were revealed to express higher levels of ANRIL lncRNA compared with the adjacent non‑neoplastic tissues (P<0.05). Furthermore, high ANRIL expression was significantly associated with reduced survival rate (P<0.05). ANRIL gene expression was successfully silenced in human CRC cells. ANRIL knockdown decreased proliferation, inhibited migration and invasion, and reduced the colony‑forming ability of the cells. These data indicated that the lncRNA ANRIL is upregulated in CRC tissues, and is associated with CRC cell pathogenesis. Furthermore, the underlying mechanisms of these effects may be exploited for therapeutic benefit.

Introduction

Colorectal cancer (CRC) is currently the third most common malignancy worldwide (1,2), thus representing a significant health burden. Despite advances in treatment options and availability, minimal improvements have been made regarding the survival rate of patients with CRC. This lack of improvement may be due to the absence of early detection techniques and optimal predictions regarding disease outcome. Therefore, exploring the biomarkers associated with CRC, and increasing knowledge regarding the molecular mechanisms of CRC is required (3).

Long noncoding RNAs (lncRNAs) are transcribed RNA molecules, >200 nt long, which are not translated into protein. LncRNAs regulate gene expression at several levels including epigenetic, transcriptional and post-transcriptional levels. In addition, lncRNAs have been hypothesized to participate in physiological and pathological cell processes (4,5). Previous studies have reported on the role of lncRNAs in the regulation of various genes (610). Their downstream effector genes are associated with transcriptional initiation (11), protein structure (12), and suppression (13,14). LncRNAs comprise a large family; however, the number of lncRNAs that have been well-studied with regards to their established function is small. Previous studies have reported that numerous lncRNAs are closely correlated with various types of malignancies, acting as oncogenes or tumor suppressors (1518).

Research regarding the effects of lncRNAs on CRC is currently in the preliminary stage, and associated studies are rare (19). In addition, despite the growing interest regarding lncRNA as a potential therapeutic target (20,21), the exact function of lncRNAs in CRC tumorigenesis remains unknown. The present study aimed to investigate the regulatory role of the lncRNA antisense noncoding RNA in the INK4 locus (ANRIL) in CRC patient outcome, and the effects of ANRIL knockdown on CRC cell proliferation, and migratory and invasive ability.

Materials and methods

Patient samples

A total of 97 patients, including 46 males and 51 females, with an average age of 66 years old, were recruited from Guizhou Provincial People's Hospital (Guizhou, China). The patients were in the late clinical stages of CRC, and had undergone surgery between May 2010 and May 2013. Adjacent non-neoplastic tissue and primary CRC tumor tissue samples were collected from each of the patients. The Institutional Review Board Affiliated to Guiyang Medical University (Kaili, China) approved the protocol of the present study to be used for the patients at Guizhou Medical University, and informed consent was collected from each patient prior to surgery. Liquid nitrogen was used to promptly freeze the tissue samples following the surgical procedure. The patients were free of any other diseases, and had no prior experience of chemotherapy or radiotherapy. A minimum of two senior pathologists examined the CRC tissues, in order to confirm diagnosis of the disease.

Cell culture

Four commonly used CRC cell lines, HT29, HCT116, RKO and SW480 cells were obtained from American Type Culture Collection (Manassas, VA, USA). A human colonic epithelial cell line, HCoEpiC was obtained from Sciencell Research Laboratories (Carlsbad, CA, USA). The cells were maintained in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA USA) supplemented with 10% fetal bovine serum (FBS; Sigma-Aldrich, St. Louis, MO, USA) in an environment containing 5% CO2 at 37°C.

ANRIL gene interference

Lentivirus vector construction (pGCSIL-green fluorescent protein vector) and transfection was conducted according to the manufacturer's instructions (Shanghai HollyBio, Shanghai, China). ANRIL short hairpin RNA (shRNA) and the negative control shRNA (shControl) were cloned into the pGCSIL-green fluorescent protein vector (Shanghai HollyBio, Shanghai, China), which was then incubated for 24 h at 37°C. The shRNA sequences (Shanghai HollyBio) were designed as follow: ANRIL, 5′-GCAGTTGCTACAAGTTAGACTCGAGTTGCTACAAGTTAGTACGCTTTTT-3′; and shControl, 5′-GGAGATATTCTTTCAAACCCTCATTCTTTCAAACCCTCCGCTTTTTT-3′.

HT29 and RKO cells were subsequently transfected with the reconstructed shRNA plasmids along with two helper plasmids (pVSVG-I and pCMVR8.92; Shanghai HollyBio) using Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The recombinant virus was then cultured in DMEM for 72 h at 37°C. Finally, 6×103 cells/well were incubated with the ANRIL shRNA-expressing lentivirus or negative control for 24 h at 37°C.

Cell viability assay

The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method was used to determine cell viability. Briefly, 1×104 shRNA-transfected cells were seeded in 96-well plates for 48 h at 37°C, and 100 µl MTT (Sigma-Aldrich) was then added to each well. The plates were incubated for 4 h at 37°C, until purple formazan crystals developed. Subsequently, each well was washed with 100 µl dimethyl sulfoxide and was air-dried at room temperature for 30 min. Cell viability was quantified by measuring the absorbance of the plate at A490 using a Universal Microplate Spectrophotometer (Thermo Fisher Scientific, Inc.).

Cell migration and invasion assays

The migratory and invasive capabilities of the CRC cells were measured in a Transwell chamber system. In the upper chamber, a solution of 60 µl diluted extracellular matrix (ECM; Corning Life Science, Lowell, CA, USA) was added. For the migration assay, the same method was used; however the ECM membrane was not added to the upper chamber. The chamber was incubated at 37°C for 4 h. Subsequently, 1×105 cells/well were seeded into the upper chamber in 100 µl DMEM containing 1% FBS. The lower chambers of the Transwell were filled with 500 µl DMEM containing 10% FBS. To allow for cell migration, the Transwell was incubated for 20 h at 37°C in an atmosphere containing 5% CO2. Once incubation was complete a cotton swab was used to carefully remove the cells from the upper chamber. Cells at the bottom of the ECM gel-coated membrane were fixed and stained with paraformaldehyde and crystal violet. After dissolving the crystal violet in 200 µl glacial acetic acid the plate was incubated for 30 min at room temperature. The number of migrated and invaded cells on the bottom side of five randomly selected visual fields (×100 magnification) per well was counted using an inverted microscope (Nikon Eclipse TS100, Nikon, Tokyo, Japan).

Western blot analysis

Protein was prepared with a lysis buffer by adding protease and phosphatase inhibitors (Thermo Fisher Scientific, Inc.) followed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis separation. Protein was then transferred to nitrocellulose membranes under semi-dry system conditions (Bio-Rad Laboratories, Hercules, CA, USA). Incubate membrane in 25 ml of blocking buffer for 1 h at room temperature and wash three times for 5 min each with 15 ml of 1X Tris-buffered saline with Tween-20 (TBST) (Santa Cruz Biotechnology Inc, Dallas, TX, USA). Next, the membranes were incubated with primary antibodies anti-p15/CDKN2B (cat. no. Sc-612) and mouse anti-β-actin (cat. no. sc-47778) at 4°C overnight. Following two continuous washes for 5 min each with TBST, the membranes were then incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG (cat. no. sc-2004) and horseradish peroxidase-conjugated goat anti-mouse IgG (sc-2005) secondary antibody. All of the antibodies were purchased from the Santa Cruz Biotechnology, Inc. Protein expression was visualized with a gel imaging system (Bio-Rad Laboratories).

Cell colony formation

Untreated negative control (shControl) cells and cells with ANRIL silencing (shANRIL) were seeded into a 6-well plate with 100 cells per well and cultured for 72 h at 37°C. After washing twice with phosphate-buffered saline (Sigma-Aldrich), cells were continually cultured for 7 days at 37°C and the medium was refreshed every three days. Then the cells were fixed in methyl alcohol and stained by 10% Giemsa (Sigma-Aldrich). Only colonies with >50 cells were counted under an inverted microscope (Nikon Eclipse TS100).

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

RNA was extracted from homogenized cells and tissues. Total RNA was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and was stored at −80°C until further use. RT-qPCR was performed with SYBR Premix Ex Taq II kit, according to the manufacturer's protocol (Takara Biotechnology Co., Ltd., Dalian, China). qPCR was conducted using SYBR green fluorescent dye and the ABI 7900 fast sequence detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.).

The following primers were used: ANRIL, forward 5′-GAATTGCGTCATTTAAAGCCTAGTT-3′, reverse 5′-GTTTCATCCTACCACTCCCAATTAAT-3′; and β-actin, forward 5′-AGAGCTACGAGCTGCCTGAC-3′ and reverse 5′-AGCACTGTGTTGGCGTACAG-3′ (Takara Biotechnology Co., Ltd., Dalian, China). The total reaction volume was 20 µl containing 2 µl cDNA, 0.4 µl each primer (10 µM), 0.4 µl ROX reference dye (50X), 6.8 µl sterile distilled water, and 2X SYBR Premix Ex Taq II. The PCR conditions were as follows: 95°C for 30 sec, followed by 40 cycles of 5 sec at 95°C, and extension for 45 sec at 60°C. The relative expression of ANRIL was calculated with according to the 2−ΔΔCq method: ΔΔCt = ΔCt target gene - ΔCt control gene, where ΔCt = Ct of target gene - Ct reference sample (22).

Statistical analysis

SPSS 13.0 (SPSS Inc., Chicago, IL, USA) was used to statistically analyze the data. Numerical data are presented as the mean ± standard deviation. Statistical analysis was carried out using Student's t-test to evaluate the statistical differences between normal and gene knockdown cells. One-way analysis of variance followed by the post-hoc multiple comparisons least significant difference test were performed to compare the significant differences between three or more groups. The χ2 test and Fisher's exact test was used to compare percentage distribution between the different groups. Each experiment was repeated at least twice or was performed in triplicate. The Kaplan-Meier method was used to draw a survival curve and evaluate the prognostic value of the ANRIL expression level in colorectal cancer. P<0.05 was considered to indicate a statistically significant difference.

Results

Correlation of patient clinical characteristics and ANRIL expression in tissue samples

The results of RT-qPCR analysis demonstrated that the expression levels of ANRIL were increased in CRC tissues compared with in the adjacent normal tissues (Fig. 1A). However, no significant association was detected between ANRIL expression and gender, age and TNM status (Table I). Relative ANRIL expression >1.5 (tumor/normal) was set as a criteria for high expression, and the 97 patients were divided into ANRIL-high (n=53) and ANRIL-low groups (n=44). Generation of a survival curve indicated that high expression levels of ANRIL are a significant predictor of a worse clinical outcome compared with low expression levels (P<0.05; Fig. 1B).

Table I

ANRIL expression and clinical characteristics in colorectal cancer.

Table I

ANRIL expression and clinical characteristics in colorectal cancer.

CharacteristicCase number (n=97)ANRIL expression
P-value
High (n=53)Low (n=44)
Gender0.207
 Male462719
 Female512625
Age0.144
 >45 years643133
 <45 years332211
TNM stage
 T status0.435
  T1–2332112
  T3–4643232
 N status0.127
  N0432716
  N1–3542628
Histologic grade0.134
  G1392118
  G2352114
  G3231112

[i] ANRIL, antisense noncoding RNA in the INK4 locus; T, Primary tumor; N, Regional lymph nodes; G1, well differentiated (low grade); G2, moderately differentiated (intermediate grade); G3, poorly differentiated (high grade).

Evaluation of knockdown efficiency of ANRIL shRNA in CRC cells

Initially ANRIL expression was detected in four selected CRC cell lines: SW480, HCT116, HT29 and RKO, and in the HCoEpiC human colonic epithelial cell line (Control). The results indicated that ANRIL expression levels were highest in the HT29 and RKO cells, compared with the other two CRC cell lines and the control (Fig. 2A). Therefore, HT29 and RKO cells were used in subsequent experiments. Analysis of knockdown efficiency demonstrated that the constructed lentivirus inhibited ANRIL expression. Since ANRIL, also known as CDKN2B-AS, is a long non-coding RNA located within p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster and closely associated with p15/CDKN2B, p16/CDKN2A and p14/ARF expression. p15/CDKN2B protein (Sc-612, Santa cruz, USA) was used in the western blot analysis to confirm the ANRIL knockdown efficiency in HT-29 and RKO cells. p15/CDKN2B protein expression was inhibited in the majority of cells 72 h post-lentiviral infection (Fig. 2B). In addition, ANRIL mRNA expression was significantly decreased in the infected cells (P<0.05; Fig. 2C).

Lentivirus-mediated ANRIL shRNA significantly inhibits growth of CRC cells

Cell proliferation was evaluated using the MTT method. As shown in Fig. 3A, the growth rate of knockdown cells was significantly impaired compared with the shControl group (P<0.05). A colony formation assay was also performed. A reduced number of cell colonies was observed following ANRIL knockdown. Compared with the shControl cells the ANRIL shRNA-treated cells exhibited decreased colony-forming ability (Fig. 3B).

ANRIL knockdown prevents cell invasion and migration

Compared with the shControl group, there was an evident decrease in the number of ANRIL shRNA-treated cells that crossed the membrane (Fig. 4). Cells that invaded through the membrane were stained with crystal violet. In addition, the relative number of migrated cells was reduced in the ANRIL shRNA group (P<0.05; Fig. 4C). The migratory capability of cells in the ANRIL shRNA group was decreased compared with in the shControl group. The relative number of invasive cells was also decreased in the ANRIL shRNA group compared with the shControl group (P<0.05; Fig. 4D). Compared with the shControl group, the invasive capability of cells in the ANRIL shRNA group was decreased.

Discussion

The role of lncRNAs in carcinogenesis has garnered a lot of attention, and previous studies have examined the relationship between lncRNAs and tumor growth, epithelial-mesenchymal transition and metastasis (14,16,23). However, there seems to be a lack of evidence demonstrating the relationship between CRC and lncRNAs. Previous studies have revealed the vital role lncRNAs can have in CRC; the lncRNAs HOX transcript antisense RNA, metastasis associated lung adenocarcinoma transcript 1, H19 and colon cancer associated transcript 1 have been shown to enhance the progression of CRC and may be used as biomarkers for CRC prognosis (24,25). The results of the present study demonstrated that higher levels of the lncRNA ANRIL are present in CRC tissue compared with adjacent non-neoplastic tissues.

The relatively high expression of ANRIL was significantly associated with decreased survival rate. In addition, ANRIL gene expression was successfully silenced in human CRC cells. ANRIL knockdown decreased proliferation, inhibited migration and invasion, and reduced the colony-forming ability of CRC cells.

The ANRIL gene (NR_003529), also known as CDKN2B antisense RNA 1, is comprised of 19 exons and has a span of 126.3 kb. The ANRIL gene is contained within the antisense gene cluster p15/CDKN2B-p16/CDKN2A-p14/ADP-ribosylation factor 1 (ARF), which produces a 3,834-bp mRNA transcript. The two exons of p15/CDKN2B are overlapped by ANRIL intron 1. The ANRIL gene has a 5′ end on its first exon, which is located ~300 bp upstream of the p14/ARF gene transcription start site. Both genes, ANRIL and p14/ARF, may share corresponding transcription due to their clustered expression in normal human tissues, as well as within various tumors (26). Novel insights into the transcriptional regulation of ANRIL and p14/ARF have suggested it is due to the mapping of CCCTC-binding factor (CTCF)-binding sites on the CpG island, which overlap the ANRIL-p14/ARF promoters (27). The CTCF insulator protein is a highly conserved zinc finger protein, which is a critical factor essential for transcription of the p15/CDKN2B-p16/CDKN2A-p14/ARF locus due to its chromatin barrier activity (28). In neurofibromatosis type 1, ANRIL has a role in the development of plexiform neurofibromas (29). In addition, ANRIL helps to suppress and downregulate the tumor gene p15 (INK4B) through the process of attaching itself and recruiting the suppressor of zester 12 homolog (Drosophila) (12), which is a component of the polycomb repressive complex 2 (30). The ATM-E2F transcription factor 1 signaling pathway is activated when DNA damage occurs via ANRIL upregulation (31,32). In the present study, RT-qPCR was conducted to measure the expression levels of ANRIL in clinical tissues and cell lines. A total of 46.3% of CRC tissues exhibited ANRIL overexpression, predominantly in progressed tumor tissues that are large in size with lymph node metastasis. These results indicated that ANRIL may be a primary participant in the advancement of CRC, and may not be present during initial onset, suggesting that ANRIL may be a likely biomarker for late stage CRC with metastasis. Inhibition of ANRIL suppressed tumor growth in vitro, and inhibited cell migration and invasion, further supporting the cancerous role of ANRIL in CRC. In addition, these results suggested that elimination of ANRIL may inhibit CRC cell proliferation, and block the migratory and invasive ability of the cells.

Qiu et al (33) demonstrated that small interfering RNA-mediated ANRIL silencing in ovarian cancer cells suppressed migration and invasion of these cells, thus suggesting that the lncRNA ANRIL has a critical role in the invasion or metastasis of serous ovarian cancer.

Hua et al (34) performed in vitro assays, which revealed that decreased expression of ANRIL may suppress the proliferation, migration and invasion of hepatocellular carcinoma cells. Furthermore, Huang et al (35) reported that reduced ANRIL expression in vitro and in vivo may reduce cell growth and invasion, and induce cell apoptosis. These findings, alongside those of the present study, illustrate how ANRIL is a major player in CRC. In conclusion, further research is still required in order to examine the underlying mechanisms and functions of ANRIL in CRC, as well as the possibility of using ANRIL as a therapeutic target for CRC.

References

1 

Jaffe N: Colorectal cancer: Review of the past, impact on the future. The American experience. Cancer Treat Res. 152:239–262. 2009. View Article : Google Scholar

2 

Meyers PA, Schwartz CL, Krailo M, Kleinerman ES, Betcher D, Bernstein ML, Conrad E, Ferguson W, Gebhardt M, Goorin AM, et al: Colorectal cancer: A randomized, prospective trial of the addition of ifosfamide and/or muramyl tripeptide to cisplatin, doxorubicin, and high-dose methotrexate. J Clin Oncol. 23:2004–2011. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Gong C, Liao H, Wang J, Lin Y, Qi J, Qin L, Tian LQ and Guo FJ: LY294002 induces G0/G1 cell cycle arrest and apoptosis of cancer stem-like cells from human osteosarcoma via down-regulation of PI3K activity. Asian Pac J Cancer Prev. 13:3103–3107. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Wang K and Zhang XC: Inhibition of SENP5 suppresses cell growth and promotes apoptosis in colorectal cancer cells. Exp Ther Med. 7:1691–1695. 2014.PubMed/NCBI

5 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Zhang Q, Geng PL, Yin P, Wang XL, Jia JP and Yao J: Down-regulation of long non-coding RNA TUG1 inhibits colorectal cancer cell proliferation and promotes apoptosis. Asian Pac J Cancer Prev. 14:2311–2315. 2013. View Article : Google Scholar

7 

Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, et al: Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci USA. 106:11667–11672. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Wang KC and Chang HY: Molecular mechanisms of long noncoding RNAs. Mol Cell. 43:904–914. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Nagano T, Mitchell JA, Sanz LA, Pauler FM, Ferguson-Smith AC, Feil R and Fraser P: The air noncoding RNA epigenetically silences transcription by targeting G9a to chromatin. Science. 322:1717–1720. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Zhao J, Sun BK, Erwin JA, Song JJ and Lee JT: Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science. 322:750–756. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Ørom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, et al: Long noncoding RNAs with enhancer-like function in human cells. Cell. 143:46–58. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M and Xiong Y: Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene. 30:1956–1962. 2011. View Article : Google Scholar

14 

Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, Khalil AM, Zuk O, Amit I, Rabani M, et al: A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 142:409–419. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Feldstein O, Nizri T, Doniger T, Jacob J, Rechavi G and Ginsberg D: The long non-coding RNA ERIC is regulated by E2F and modulates the cellular response to DNA damage. Mol Cancer. 12:1312013. View Article : Google Scholar : PubMed/NCBI

16 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Kino T, Hurt DE, Ichijo T, Nader N and Chrousos GP: Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal. 3:ra82010. View Article : Google Scholar : PubMed/NCBI

18 

Zhang X, Gejman R, Mahta A, Zhong Y, Rice KA, Zhou Y, Cheunsuchon P, Louis DN and Klibanski A: Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression. Cancer Res. 70:2350–2358. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Pandey RR, Mondal T, Mohammad F, Enroth S, Redrup L, Komorowski J, Nagano T, Mancini-Dinardo D and Kanduri C: Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol Cell. 32:232–246. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Pasic I, Shlien A, Durbin AD, Stavropoulos DJ, Baskin B, Ray PN, Novokmet A and Malkin D: Recurrent focal copy-number changes and loss of heterozygosity implicate two noncoding RNAs and one tumor suppressor gene at chromosome 3q13.31 in colorectal cancer. Cancer Res. 70:160–171. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Zhu L and Xu C: Down regulated LncRNA-ANCR promotes osteoblast differentiation by targeting EZH2 and regulating Runx2 expression. Biochem Biophys Res Commun. 432:612–617. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

23 

Kam Y, Rubinstein A, Naik S, Djavsarov I, Halle D, Ariel I, Gure AO, Stojadinovic A, Pan H, Tsivin V, et al: Detection of a long non-coding RNA (CCAT1) in living cells and human adenocarcinoma of colon tissues using FIT-PNA molecular beacons. Cancer Lett. 352:90–96. 2014. View Article : Google Scholar

24 

Ye LC, Zhu X, Qiu JJ, Xu J and Wei Y: Involvement of long non-coding RNA in colorectal cancer: From bench top to bedside (Review). Oncol Lett. 9:1039–1045. 2015.PubMed/NCBI

25 

Nakano S, Murakami K, Meguro M, Soejima H, Higashimoto K, Urano T, Kugoh H, Mukai T, Ikeguchi M and Oshimura M: Expression profile of LIT1/KCNQ1OT1 and epigenetic status at the KvDMR1 in colorectal cancers. Cancer Sci. 97:1147–1154. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Pasmant E, Laurendeau I, Héron D, Vidaud M, Vidaud D and Bièche I: Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: Identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF. Cancer Res. 67:3963–3969. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Rodriguez C, Borgel J, Court F, Cathala G, Forné T and Piette J: CTCF is a DNA methylation-sensitive positive regulator of the INK/ARF locus. Biochem Biophys Res Commun. 392:129–134. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Pasmant E, Sabbagh A, Vidaud M and Bièche I: ANRIL, a long, noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J. 25:444–448. 2011. View Article : Google Scholar

29 

Dereure O: Role of non-coding RNA ANRIL in the genesis of plexiform neurofibromas in neurofibromatosis type 1. Ann Dermatol Venereol. 139:421–422. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M and Xiong Y: Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene. 30:1956–1962. 2011. View Article : Google Scholar

31 

Wan G, Mathur R and Hu X: Long non-coding RNA ANRIL (CDKN2B-AS) is induced by the ATM-E2F1 signaling pathway. Cell Signal. 25:1086–1095. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Tang JY, Lee JC, Chang YT, Hou MF, Huang HW, Liaw CC and Chang HW: Long noncoding RNAs-related diseases, cancers, and drugs. Scientific World Journal. 2013:9435392013. View Article : Google Scholar : PubMed/NCBI

33 

Qiu JJ, Lin YY, Ding JX, Feng WW, Jin HY and Hua KQ: Long non-coding RNA ANRIL predicts poor prognosis and promotes invasion/metastasis in serous ovarian cancer. Int J Oncol. 46:2497–2505. 2015.PubMed/NCBI

34 

Hua L, Wang CY, Yao KH, Chen JT, Zhang JJ and Ma WL: High expression of long non-coding RNA ANRIL is associated with poor prognosis in hepatocellular carcinoma. Int J Clin Exp Pathol. 8:3076–3082. 2015.PubMed/NCBI

35 

Huang MD, Chen WM, Qi FZ, Xia R, Sun M, Xu TP, Yin L, Zhang EB, De W and Shu YQ: Long non-coding RNA ANRIL is upregulated in hepatocellular carcinoma and regulates cell apoptosis by epigenetic silencing of KLF2. J Hematol Oncol. 8:502015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2016
Volume 14 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
Sun Y, Zheng ZP, Li H, Zhang HQ and Ma FQ: ANRIL is associated with the survival rate of patients with colorectal cancer, and affects cell migration and invasion in vitro. Mol Med Rep 14: 1714-1720, 2016
APA
Sun, Y., Zheng, Z., Li, H., Zhang, H., & Ma, F. (2016). ANRIL is associated with the survival rate of patients with colorectal cancer, and affects cell migration and invasion in vitro. Molecular Medicine Reports, 14, 1714-1720. https://doi.org/10.3892/mmr.2016.5409
MLA
Sun, Y., Zheng, Z., Li, H., Zhang, H., Ma, F."ANRIL is associated with the survival rate of patients with colorectal cancer, and affects cell migration and invasion in vitro". Molecular Medicine Reports 14.2 (2016): 1714-1720.
Chicago
Sun, Y., Zheng, Z., Li, H., Zhang, H., Ma, F."ANRIL is associated with the survival rate of patients with colorectal cancer, and affects cell migration and invasion in vitro". Molecular Medicine Reports 14, no. 2 (2016): 1714-1720. https://doi.org/10.3892/mmr.2016.5409