Antisense Non-Coding RNA in the INK4 Locus (ANRIL) in Human Cancers

authors:

avatar Mohammad Taheri 1 , avatar Soudeh Ghafouri-Fard ORCID 2 , *

Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran

how to cite: Taheri M, Ghafouri-Fard S. Antisense Non-Coding RNA in the INK4 Locus (ANRIL) in Human Cancers. Int J Cancer Manag. 2018;11(10):e67864. https://doi.org/10.5812/ijcm.67864.

Abstract

Context:

ANRIL (antisense non-coding RNA in the INK4 locus) is one of the oncogenic long non-coding RNAs (lncRNAs), which is located at the 9p21.3. It has a regulatory epigenetic function on its neighboring gene cluster p15/CDKN2B-p16/CDKN2A-p14/ARF. Such regulatory role has led to contribution of ANRIL in the pathogenesis of many human cancers.

Evidence Acquisition:

We searched the PubMed, Google Scholar, EMBASE, and Web of Science databases till February 2018 with the key words "ANRIL" OR "antisense non-coding RNA in the INK4 locus" AND "cancer" OR "tumor" OR "malignancy".

Results:

ANRIL over-expression is documented in several cancers. The underlying mechanism for regulatory effect of ANRIL on the expression of its neighboring tumor suppressors has been clarified in some malignancies. Moreover, ANRIL over-expression has been associated with cisplatin and 5-fluorouracil (5-FU)-resistance in some cancers.

Conclusions:

ANRIL is regarded as a potential biomarker in diverse cancer types and a therapeutic target in certain malignancies.

1. Context

Long non-coding RNAs (lncRNAs) are a group of transcripts with sizes greater than 200 nucleotides that instead of being translated into protein, themselves exert crucial functional roles (1). Almost any fundamental aspect of cell physiology is a subject of regulation by lncRNAs (2-5). Their participation in a wide range of cancers has also been evaluated (6-11). ANRIL (antisense non-coding RNA in the INK4 locus) is one of the oncogenic lncRNAs, which is located at the 9p21.3 locus (12). This locus has been recognized as a common genetic susceptibility locus for coronary disease, intracranial aneurysm, type 2 diabetes, and several malignancies through genome-wide association studies (GWAS) (13). The 9p21.3 locus encompasses p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster as well. ANRIL has been shown to regulate the expression of this gene cluster through recruitment of polycomb repressive complexes PRC2 and PRC1 to this locus (14) (Figure 1). ANRIL-mediated recruitment of these complexes alters the epigenetic chromatin shape and, hence, suppresses the expression of this gene cluster in a cis-acting manner (15). PRC1 and PRC2 participate in chromatin remodeling, suppress transcription of several genes and interact with key signaling pathways. Their regulatory role on stem cell pluripotency provides further evidences for their participation in cancer pathogenesis. PRC2 and PRC1 suppressive regulatory function is exerted through two diverse histone modifications, trimethylation of lysine 27 on histone H3 (H3K27me3) and monoubiquitination of histone H2A (H2AK119ub) (16). ANRIL-mediated silencing of the CDKN2B/CDKN2A/ARF locus is exerted though ANRIL interaction with EZH2 (PRC2) and CBX7 (PRC1) (17). The observed up-regulation of ANRIL and CBX7 in prostate cancer tissues and the ability of ANRIL to bind with CBX7 provided further evidences for contribution of ANRIL in suppression of the CDKN2A/B locus (14). Moreover, EZH2 is a histone methyltransferase, which participates in silencing of p15/CDKN2B-p16/CDKN2A-p14/ARF cluster by tri-methylating H3K27 in the corresponding promoters (18). The p15/CDKN2B-p16/CDKN2A-p14/ARF-ANRIL genes contribute in the regulation of senescence and apoptosis. Alterations in tumor suppressor genes encoded by p15/CDKN2B-p16/CDKN2A-p14/ARF locus have been associated with a wide range of malignancies (19-21), implying the parallel role of ANRIL in the pathogenesis of these malignancies. Consequently, several studies have focused on the evaluation of ANRIL expression or genomic variations in human cancers.

The 9p21.3 locus encompasses p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster as well. ANRIL regulates expression of this gene cluster through recruitment of polycomb repressive complexes PRC2 and PRC1 to this locus. ANRIL suppresses p16/INK4A thereby reverses its effects on growth suppression.
The 9p21.3 locus encompasses p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster as well. ANRIL regulates expression of this gene cluster through recruitment of polycomb repressive complexes PRC2 and PRC1 to this locus. ANRIL suppresses p16/INK4A thereby reverses its effects on growth suppression.

2. Evidence Acquisition

We searched the PubMed, Google Scholar, EMBASE, and Web of Science databases till February 2018 with the key words "ANRIL" OR "antisense non-coding RNA in the INK4 locus" AND "cancer" OR "tumor" OR "malignancy". We only included original articles written in English, described the mechanism of ANRIL participation in malignancies and provided enough patients' data. We excluded other types of papers from the study.

3. Results

Several studies have pointed the significance of ANRIL in diverse human malignancies. Table 1 shows the summary of these studies.

Table 1.

Expression, Genomic Variants, and Function of ANRIL in Human Cancers

Cancer TypeExpressionRoleTarget GenesVariant Associated with Cancer RiskVariant Associated with Therapeutic ResponseReferences
Lung cancerUpAssociated with TNM stage, cisplatin resistance and poor prognosisc-Myc, KLF2, P21-rs1333049 (associated with severe overall toxicity of cisplatin), rs10120688 and rs1333049 (associated with response to platinum) (22-25)
Hepatocellular carcinomaUpPromotes cell proliferationmiR-122-5p, KLF2-- (26, 27)
Prostate cancerUpPromotes cell proliferation and migrationCBX7rs4977574, rs1333048, rs10757278- (14, 28, 29)
Breast cancerUpPromotes cell proliferationp15/CDKN2B-p16/CDKN2A-p14/ARF gene clusterAn rs1333045, rs4977574, rs1333048 and rs10757278 haplotype, rs11515- (12, 53, 30)
Gastric cancerUpPromotes cell proliferation, Associated with cisplatin- and 5-fluorouracil (5-FU)-resistanceMDR1, MRP1, miR-449a, p15Ink4B, p16Ink4A-- (31, 32)
Colorectal cancerUpPromotes cell proliferation, migration and invasion--- (33)
Cervical cancerUpPromotes cell proliferationmiR-186, PI3K/Akt pathway, p15, p16-- (34)
Ovarian cancerUpPromotes cell proliferationP15INK4, Bcl-2-- (35)
Oral squamous cell carcinomaUpPromotes cell proliferation, inhibits metastasis, associated with cisplatin- resistance---
Esophageal squamous cell carcinoma (ESCC)UpPromotes cell proliferationp15INK4b, transforming growth factor β1 (TGFβ1)-- (36)
Nasopharyngeal carcinoma (NPC)UpPromotes cell proliferation, decreases apoptosis, increases radioresistance, reprograms cell glucose metabolism and induces side-population stem-like cancer cellsmiR-125a, miR-let--- (37, 38)
Acute lymphoblastic leukemia (ALL)----rs564398 correlated with ALL phenotype (39)
Multiple myeloma (MM)-Associated with melphalan-mediated apoptosisp14ARF, p53-rs2151280 associated with worse progression-free survival (40)

3.1. ANRIL in Breast Cancer

In a large cohort of patients with breast cancer (n = 456), ANRIL over-expression has been detected in 19.7% of invasive breast carcinomas, particularly in the aggressive triple negative breast cancer (TNBC) subtype (16). Moreover, its expression level was correlated with estrogen receptor (ER) and progesterone receptor (PR) status but no patients' survival. A strong positive correlation has been detected between mRNA expression levels of the ANRIL, ARF, p15, and p16 pointing to the presence of a complicated network between these genes (16). In spite of the proposed role for ANRIL in epithelial-mesenchymal transition (EMT), no significant associations were found between ANRIL and EMT-associated markers such as TWIST1, ZEB1, ZEB2, SNAI1/Snail, SNAI2/Slug, VIM/Vimentin, CDH2/N-cadherin, CDH1/E-cadherin, and ZO-1 (16).

More recently, ANRIL has been shown to be over-expressed in TNBC tumor tissue and cell lines compared to noncancerous tissue and non-TNBC cells. Such elevated expression was accompanied by poor prognosis. In vivo functional studies confirmed the role of ANRIL as molecular 'sponge' for miR-199a in TNBC (30).

The rs11515 SNP located in the 5' region of ANRIL has been shown to be associated with breast cancer risk in a sample of 200 patients and 200 healthy subjects from New Zealand. Notably, the CG genotype of this SNP has been correlated with increased ANRIL and decreased p16INK4a expression levels suggestive of the decreased expression of p16INK4a as a putative mechanism for contribution of rs11515 CG genotype in cancer risk (41). Recently, we genotyped rs1333045, rs4977574, rs1333048, and rs10757278 SNPs in 122 patients with breast cancer and 200 normal age-matched subjects and found TCGA haplotype to be associated with breast cancer risk in Iranian population (12).

3.2. ANRIL in Prostate Cancer

The elevated expression of ANRIL as well as chromobox 7 (CBX7) within the PRC1, which interacts with ANRIL, has been demonstrated in prostate cancer tissues. In prostate tissues, ANRIL binding with CBX7 has been shown to be crucial for suppression of the INK4b/ARF/INK4a locus (14). Moreover, the rs4977574, rs1333048, and rs10757278 within ANRIL have been shown to be associated with prostate cancer risk in Iranian population (29).

3.3. ANRIL in Hepatocellular Carcinoma (HCC)

The assessment of ANRIL expression in 77 HCC tissues and corresponding normal tissues have shown its up-regulation in HCC tissues and correlation of its transcript levels with tumor size and Barcelona Clinic Liver Cancer (BCLC) stage. Its silencing could suppress cell proliferation and invasion and trigger cell apoptosis both in vitro and in vivo. Moreover, functional studies showed the role of ANRIL in suppression of Kruppel-like factor 2 (KLF2) transcription in HCC cells by recruitment of PRC2 to the KLF2 promoter region (42). More recently, in vitro studies have shown a negative correlation between ANRIL and miR-122-5p expressions in HCC tissues, which was confirmed through the detection of directly interaction between ANRIL and miR-122-5p. More importantly, in vivo studies showed the effect of ANRIL knockdown in suppression of tumor growth (26).

3.4. ANRIL in Lung Cancer

ANRIL is over-expressed in H1299 cancer cells compared to normal fibroblasts. Its silencing increased p15 expression with no effect on p16 or ARF expression, led to cell-cycle arrest at the G2/M phase and suppressed proliferation of these cells (43). Besides, its knock-down in A549, SPC-A1, NCI-H1650 lung cancer cells suppressed cell proliferation, migration, and invasion in vitro (44). In addition, its silencing in NSCLC cells could weaken cell proliferation and trigger cell apoptosis both in vitro and in vivo. Its effect on decreasing p15 expression in PC9 cells was exerted through silencing of KLF2 and P21 transcription (23). The expression of lncRNA ANRIL has been up-regulated by heparin-binding growth factor secreted from cancer-associated fibroblasts (CAF) in lung tumors. ANRIL is also involved in proliferation, apoptosis, and cisplatin resistance of the tumor cells via modulation of the drug transporters MRP1 and ABCC2 (45). The physical interaction between c-Myc and ANRIL has also been validated in vitro. The effect of ANRIL silencing in suppression of NSCLC cell proliferation potentiate this lncRNA as a putative therapeutic target for patients with NSCLC (24). However, a single study in lung cancer H460 cells indicated ANRIL as an lncRNA responsible in anti-tumorigenesis caused by PLD inhibition and suggested the addition of ANRIL into PLD inhibition as a novel therapeutic modality for controlling lung cancer (46). The discrepancy between these two studies might be due to the difference in cultured cells or other biological variables. The results of clinical studies are in line with oncogenic role of this lncRNA. For instance, the expression level of ANRIL has been shown to be elevated in NSCLC tissues and lung cancer cells than in adjacent non-tumor tissues and normal human bronchial epithelial cells. Such over-expression in clinical tissues was associated with higher TNM stage, advanced lymph node metastasis as well as poor overall survival (23, 44).

Moreover, in a population of 467 Chinese patients with lung cancer treated with platinum-based chemotherapy, ANRIL rs1333049 was a predictor of both severe overall toxicity and severe gastrointestinal toxicity (22). Besides, ANRIL rs10120688 and rs1333049 were associated with response to platinum-based chemotherapy in patients with adenocarcinoma and small-cell lung cancer, respectively, in 498 Chinese patients with lung cancer (25).

3.5. ANRIL in Cervical Caner

ANRIL over-expression has been detected in HeLa cancer cells compared to normal fibroblasts. Its silencing increased p15 expression with no effect on p16 or ARF expression, led to cell-cycle arrest at the G2/M phase and suppressed proliferation of these cells (43). Moreover, ANRIL expression has been elevated in cervical cancer tumors compared with non-cancerous tissues (34, 47). The role of this lncRNA, as a sponge for miR-186, has been confirmed by both bioinformatics tools and luciferase assay. Consequently, the ANRIL/miR-186 axis has been suggested as a culprit in the pathogenesis of cervical cancer (34). Moreover, the higher expression of this lncRNA in clinical samples was associated with advanced International Federation of Gynecologists and Obstetricians stage (FIGO) stage, lymph node metastasis, and poor overall survival indicating a role for ANRIL expression as an independent prognostic factor in cervical cancer. ANRIL silencing has suppressed cell proliferation, migration, and invasion of cervical cancer via the inhibition of PI3K/Akt pathway. Consequently, this lncRNA has been suggested as a putative prognostic biomarker and therapeutic target in cervical cancer (47).

3.6. ANRIL in Ovarian Cancer

The expression of lncRNA ANRIL is increased by heparin-binding growth factor secreted from cancer-associated fibroblasts (CAF) in ovarian cancer cells (45). ANRIL expression was up-regulated in epithelial ovarian cancer (EOC) tissues compared with normal controls in association with advanced FIGO stage and high histological grade. Such over-expression has been demonstrated to be an independent prognostic factor for overall survival in EOC. The role of ANRIL in the stimulation of EOC cell proliferation has been confirmed both by in vitro and in vivo. Such role is suggested to be exerted through down-regulation of P15INK4B and up-regulation of Bcl-2 (35).

3.7. ANRIL in Oral Squamous Cell Carcinoma (OSCC)

The expression of ANRIL is increased by heparin-binding growth factor secreted from cancer-associated fibroblasts (CAF) in OSCC. Moreover, its over-expression has been correlated with both high TNM stage and lymph node metastasis in OSCC. On the other hand, its silencing suppressed proliferation, induced apoptosis, and increased cisplatin cytotoxicity of the tumor cells via diminishing the drug transporters MRP1 and ABCC2 (45).

3.8. ANRIL in Esophageal Cancer

The elevated expression of ANRIL has been detected in human esophageal squamous cell carcinoma (ESCC) tissues compared with the corresponding adjacent non-tumor tissues. In vitro studies revealed the role of ANRIL silencing in the enhancement of the expression of p15 (INK4b) and transforming growth factor β1 (TGFβ1) as well as the suppression of cellular proliferation (36). However, the assessment of association between ANRIL rs2151280 in 380 ESCC and 380 cancer-free controls from Chinese Han populations cases revealed no association between this polymorphism and ESCC (48).

3.9. ANRIL in Gastric Cancer

The elevated expression of ANRIL has been demonstrated in gastric cancer tissues of cisplatin-resistant and 5-fluorouracil (5-FU)-resistant patients as well as cisplatin-resistant cells (BGC823/DDP) and 5-FU-resistant cells (BGC823/5-FU). ANRIL silencing in these cells decreased their survival and invasion capability, increased their apoptosis, and down-regulated the expression of multi-drug resistance (MDR) related genes (31).

Ten-eleven translocation 2 (TET2), which converts 5-methylcytosine to 5-hydroxymethylcytosine in DNA, has been shown to bind to the promoter the region of ANRIL and modulate the expression of ANRIL, INK4a, INK4b, and ARF in gastric cancer cells. Besides, ANRIL silencing inhibits the effects of TET2 on gastric cancer cell proliferation and colony formation (49).

Besides, in a cohort of 120 patients with gastric cancer, ANRIL over-expression has been correlated with a higher TNM stage and tumor size being an independent predictor for overall survival. ANRIL silencing decreased cell proliferation both in vitro and in vivo. E2F1 has been demonstrated to be responsible for the induction of ANRIL expression and ANRIL-mediated cell proliferation through the epigenetic suppression of miR-99a/miR-449a in gastric cancer cells (32).

3.10. ANRIL in Colorectal Cancer (CRC)

The expression analysis of ANRIL in 97 paired CRC and adjacent non neoplastic tissue samples have shown its over-expression in CRC samples compared with the adjacent non neoplastic tissues in association with decreased survival rate. Moreover, its silencing in HT29 and RKO human CRC cell lines reduced proliferation while suppressed migration and invasion (33).

3.11. ANRIL in Nasopharyngeal Carcinoma (NPC)

ANRIL over-expression has been demonstrated in advanced-stage cancer in a cohort of 88 NPC patients indicating a role for it an independent predictor of overall survival and disease-free survival. In vitro studies have shown ANRIL effect on the induction of side population cells (SP cells) in NPC. Moreover, ANRIL has a role in the enhancement of glucose uptake for glycolysis, through the regulation of the mTOR signaling pathway (50). The over-expression of ANRIL has been demonstrated in NPC tissues and cell lines. ANRIL silencing in CNE2 and HONE1 cells inhibited proliferation, triggered apoptosis, and improved radiosensitivity of cancer cells. In vitro studies suggested miR-125a as a target of ANRIL in these cells (37). Another study has shown the elevated expression of ANRIL while down-regulation of let-7a in NPC tissues and cells. The effect of ANRIL in down-regulation of miR-let-7a expression has been confirmed by luciferase assay. The role of ANRIL silencing in suppression of tumorigenicity and improvement of cisplatin-induced cytotoxicity has offered a hopeful treatment modality for NPC patients (38).

3.12. ANRIL in Hematological Malignancies

The rs564398 within ANRIL has been shown to be associated with acute lymphoblastic leukemia (ALL) phenotype in an association study enrolling 149 leukemia patients, including Philadelphia positive (Ph (+)) acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) samples. This association might show the ability of some ANRIL polymorphisms to alter ANRIL expression and, subsequently, modify CDKN2A/B expression signatures, which would contribute in atypical cell proliferation and ALL susceptibility (39). Another association study in Iranian patients with acute myeloid leukemia (AML) showed no association between rs1333048, rs4977574, rs1333045, and rs10757278 polymorphisms and AML (51). ANRIL genotyping in 108 multiple myeloma patients treated with high-dose melphalan and subsequent hematopoietic stem cell transplantation (HSCT) have shown the association between the rs2151280 (C/T) genotypes and worse progression-free survival. The TT variant was also associated with elevated ANRIL expression and down-regulation of p15, p14ARF, and p16 compared to the TC/CC variants. Consequently, ANRIL might contribute in melphalan-mediated apoptosis through modulation of p14ARF and subsequent p53 (40).

3.13. ANRIL in Osteosarcoma (OS)

The transcript levels of ANRIL have been elevated in OS tissues compared with the adjacent normal tissues in association with tumor size and patients’ outcome. Si-RNA mediated ANRIL silencing has resulted in the induction of cell apoptosis and suppression of cell proliferation and invasion both in vitro and in vivo. Nude mice injected with siANRIL-transfected MNNG/HOS cells have developed smaller tumors as compared with control group (52), which might highlight the role of ANRIL silencing in cancer control.

4. Conclusions

ANRIL role in epigenetic regulation of expression of its adjacent tumor suppressor genes CDKN2B, CDKN2A, and ARF has suggested its contribution in carcinogenesis. This hypothesis has been supported by the observed over-expression of this lncRNA in a wide range of cancers. However, the associations between ANRIL and CDKN2A/CDKNB/ARF expression seem to be context-dependent. For instance, in the context of prostate cancer CBX7 and ANRIL over-expressions have been correlated with CDKN2A/CDKNB/ARF down-regulation (14). On the contrary, in breast tumors, CBX7 has been shown to be down-regulated, while both ANRIL and CDKN2A/CDKNB/ARF have been over-expressed (16). Such observations have led to the supposition of both tumor suppressive and oncogenic roles for CBX7 based on type of the malignancy, microenvironment, and existence of interacting proteins (16).

In addition, certain SNPs within this gene have been shown to confer cancer risk in distinct populations. However, tissue- or disease-specific functions have been suggested for at least some SNPs (41), which might explain lack of associations with some types of cancer despite significant association with other types of malignancies.

The over-expression of ANRIL in cancer cells promotes cell cycle progression through the alteration of signaling pathways such as Akt pathways or oncogenes such as Myc. In addition, ANRIL has a role in the inhibition of apoptosis and enhancement of cell migration and invasion possibly through the modulation of EMT pathways. Consequently, ANRIL has been shown to influence various aspects of carcinogenesis including the cell cycle, apoptosis, and metastasis. The results of in vitro and in vivo studies support the effectiveness of ANRIL silencing in the inhibition of carcinogenesis-related pathways. However, as any signaling pathway is itself affected by several lncRNAs and miRNAs, ANRIL silencing should be considered only as a single part of a comprehensive treatment modality for the effective eradication of cancer.

Clinical and in vitro studies have shown an intricate interactive network between ANRIL, numerous miRNAs, PRC2/PRC1 subunits, and p15/CDKN2B-p16/CDKN2A-p14/ARF gene cluster (16). Uncovering such interactions would facilitate the design of appropriate targeted therapies for different kinds of human malignancies. The crucial role of ANRIL in conferring MDR in cancer cell as being discovered in gastric cancer (31) suggests the combination of ANRIL targeting therapies with conventional chemotherapeutic regimens as a promising option for cancer treatment.

Acknowledgements

References

  • 1.

    Soudyab M, Iranpour M, Ghafouri-Fard S. The role of long non-coding rnas in breast cancer. Arch Iran Med. 2016;19(7):508-17. [PubMed ID: 27362246].

  • 2.

    Dianatpour A, Ghafouri-Fard S. The role of long non coding RNAs in the repair of DNA double strand breaks. Int J Mol Cell Med. 2017;6(1):1-12. [PubMed ID: 28868264]. [PubMed Central ID: PMC5568187].

  • 3.

    Eftekharian MM, Ghafouri-Fard S, Soudyab M, Omrani MD, Rahimi M, Sayad A, et al. Expression analysis of long non-coding RNAs in the blood of multiple sclerosis patients. J Mol Neurosci. 2017;63(3-4):333-41. [PubMed ID: 28967047]. https://doi.org/10.1007/s12031-017-0982-1.

  • 4.

    Dianatpour A, Ghafouri-Fard S. Long non coding RNA expression intersecting cancer and spermatogenesis: A systematic review. Asian Pac J Cancer Prev. 2017;18(10):2601-10. [PubMed ID: 29072050]. [PubMed Central ID: PMC5747377]. https://doi.org/10.22034/APJCP.2017.18.10.2601.

  • 5.

    Faramarzi S, Dianatpour A, Ghafouri-Fard S. Discovering the role of long non-coding RNAs in regulation of steroid receptors signaling in cancer. J Biol Today World. 2017;6(12). https://doi.org/10.15412/j.jbtw.01061202.

  • 6.

    Taheri M, Omrani MD, Ghafouri-Fard S. Long non-coding RNA expression in bladder cancer. Biophys Rev. 2018;10(4):1205-13. [PubMed ID: 29222807]. [PubMed Central ID: PMC6082308]. https://doi.org/10.1007/s12551-017-0379-y.

  • 7.

    Nikpayam E, Soudyab M, Tasharrofi B, Sarrafzadeh S, Iranpour M, Geranpayeh L, et al. Expression analysis of long non-coding ATB and its putative target in breast cancer. Breast Dis. 2017;37(1):11-20. [PubMed ID: 28598827]. https://doi.org/10.3233/BD-160264.

  • 8.

    Taheri M, Habibi M, Noroozi R, Rakhshan A, Sarrafzadeh S, Sayad A, et al. HOTAIR genetic variants are associated with prostate cancer and benign prostate hyperplasia in an Iranian population. Gene. 2017;613:20-4. [PubMed ID: 28259691]. https://doi.org/10.1016/j.gene.2017.02.031.

  • 9.

    Nikpayam E, Tasharrofi B, Sarrafzadeh S, Ghafouri-Fard S. The role of long non-coding RNAs in ovarian cancer. Iran Biomed J. 2017;21(1):3-15. [PubMed ID: 27664137]. [PubMed Central ID: PMC5141251]. https://doi.org/10.6091/.21.1.24.

  • 10.

    Iranpour M, Soudyab M, Geranpayeh L, Mirfakhraie R, Azargashb E, Movafagh A, et al. Expression analysis of four long noncoding RNAs in breast cancer. Tumour Biol. 2016;37(3):2933-40. [PubMed ID: 26409453]. https://doi.org/10.1007/s13277-015-4135-2.

  • 11.

    Taheri M, Omrani M, Ghafouri-Fard S. Long non-coding RNAs expression in renal cell carcinoma. J Biol Today World. 2017;6(12). https://doi.org/10.15412/j.jbtw.01061201.

  • 12.

    Khorshidi HR, Taheri M, Noroozi R, Sarrafzadeh S, Sayad A, Ghafouri-Fard S. ANRIL genetic variants in Iranian breast cancer patients. Cell J. 2017;19(Suppl 1):72-8. [PubMed ID: 28580310]. [PubMed Central ID: PMC5448323]. https://doi.org/10.22074/cellj.2017.4496.

  • 13.

    Pasmant E, Sabbagh A, Vidaud M, Bieche I. ANRIL, a long, noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J. 2011;25(2):444-8. [PubMed ID: 20956613]. https://doi.org/10.1096/fj.10-172452.

  • 14.

    Yap KL, Li S, Munoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, et al. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell. 2010;38(5):662-74. [PubMed ID: 20541999]. [PubMed Central ID: PMC2886305]. https://doi.org/10.1016/j.molcel.2010.03.021.

  • 15.

    Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M, et al. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene. 2011;30(16):1956-62. [PubMed ID: 21151178]. [PubMed Central ID: PMC3230933]. https://doi.org/10.1038/onc.2010.568.

  • 16.

    Meseure D, Vacher S, Alsibai KD, Nicolas A, Chemlali W, Caly M, et al. Expression of ANRIL-polycomb complexes-CDKN2A/B/ARF genes in breast tumors: Identification of a two-gene (EZH2/CBX7) signature with independent prognostic value. Mol Cancer Res. 2016;14(7):623-33. [PubMed ID: 27102007]. https://doi.org/10.1158/1541-7786.MCR-15-0418.

  • 17.

    Popov N, Gil J. Epigenetic regulation of the INK4b-ARF-INK4a locus: In sickness and in health. Epigenetics. 2010;5(8):685-90. [PubMed ID: 20716961]. [PubMed Central ID: PMC3052884].

  • 18.

    Zhao R, Choi BY, Lee MH, Bode AM, Dong Z. Implications of genetic and epigenetic alterations of CDKN2A (p16(INK4a)) in cancer. EBioMedicine. 2016;8:30-9. [PubMed ID: 27428416]. [PubMed Central ID: PMC4919535]. https://doi.org/10.1016/j.ebiom.2016.04.017.

  • 19.

    Bostrom J, Meyer-Puttlitz B, Wolter M, Blaschke B, Weber RG, Lichter P, et al. Alterations of the tumor suppressor genes CDKN2A (p16(INK4a)), p14(ARF), CDKN2B (p15(INK4b)), and CDKN2C (p18(INK4c)) in atypical and anaplastic meningiomas. Am J Pathol. 2001;159(2):661-9. [PubMed ID: 11485924]. [PubMed Central ID: PMC1850553]. https://doi.org/10.1016/S0002-9440(10)61737-3.

  • 20.

    Simon M, Koster G, Menon AG, Schramm J. Functional evidence for a role of combined CDKN2A (p16-p14(ARF))/CDKN2B (p15) gene inactivation in malignant gliomas. Acta Neuropathol. 1999;98(5):444-52. [PubMed ID: 10541865].

  • 21.

    Rousseau E, Ruchoux MM, Scaravilli F, Chapon F, Vinchon M, De Smet C, et al. CDKN2A, CDKN2B and p14ARF are frequently and differentially methylated in ependymal tumours. Neuropathol Appl Neurobiol. 2003;29(6):574-83. [PubMed ID: 14636164].

  • 22.

    Gong WJ, Peng JB, Yin JY, Li XP, Zheng W, Xiao L, et al. Association between well-characterized lung cancer lncRNA polymorphisms and platinum-based chemotherapy toxicity in Chinese patients with lung cancer. Acta Pharmacol Sin. 2017;38(4):581-90. [PubMed ID: 28260796]. [PubMed Central ID: PMC5386317]. https://doi.org/10.1038/aps.2016.164.

  • 23.

    Nie FQ, Sun M, Yang JS, Xie M, Xu TP, Xia R, et al. Long noncoding RNA ANRIL promotes non-small cell lung cancer cell proliferation and inhibits apoptosis by silencing KLF2 and P21 expression. Mol Cancer Ther. 2015;14(1):268-77. [PubMed ID: 25504755]. https://doi.org/10.1158/1535-7163.MCT-14-0492.

  • 24.

    Lu Y, Zhou X, Xu L, Rong C, Shen C, Bian W. Long noncoding RNA ANRIL could be transactivated by c-Myc and promote tumor progression of non-small-cell lung cancer. Onco Targets Ther. 2016;9:3077-84. [PubMed ID: 27307748]. [PubMed Central ID: PMC4888724]. https://doi.org/10.2147/OTT.S102658.

  • 25.

    Gong WJ, Yin JY, Li XP, Fang C, Xiao D, Zhang W, et al. Association of well-characterized lung cancer lncRNA polymorphisms with lung cancer susceptibility and platinum-based chemotherapy response. Tumour Biol. 2016;37(6):8349-58. [PubMed ID: 26729200]. https://doi.org/10.1007/s13277-015-4497-5.

  • 26.

    Ma J, Li T, Han X, Yuan H. Knockdown of LncRNA ANRIL suppresses cell proliferation, metastasis, and invasion via regulating miR-122-5p expression in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2018;144(2):205-14. [PubMed ID: 29127494]. https://doi.org/10.1007/s00432-017-2543-y.

  • 27.

    Huang MD, Chen WM, Qi FZ, Xia R, Sun M, Xu TP, et al. Erratum to: Long non-coding RNA ANRIL is upregulated in hepatocellular carcinoma and regulates cell proliferation by epigenetic silencing of KLF2. J Hematol Oncol. 2017;10(1):143. [PubMed ID: 28950881]. [PubMed Central ID: PMC5530579]. https://doi.org/10.1186/s13045-017-0513-0.

  • 28.

    Zhao B, Lu YL, Yang Y, Hu LB, Bai Y, Li RQ, et al. Overexpression of lncRNA ANRIL promoted the proliferation and migration of prostate cancer cells via regulating let-7a/TGF-beta1/ Smad signaling pathway. Cancer Biomark. 2018;21(3):613-20. [PubMed ID: 29278879]. [PubMed Central ID: PMC5859458]. https://doi.org/10.3233/CBM-170683.

  • 29.

    Taheri M, Pouresmaeili F, Omrani MD, Habibi M, Sarrafzadeh S, Noroozi R, et al. Association of ANRIL gene polymorphisms with prostate cancer and benign prostatic hyperplasia in an Iranian population. Biomark Med. 2017;11(5):413-22. [PubMed ID: 28621612]. https://doi.org/10.2217/bmm-2016-0378.

  • 30.

    Xu ST, Xu JH, Zheng ZR, Zhao QQ, Zeng XS, Cheng SX, et al. Long non-coding RNA ANRIL promotes carcinogenesis via sponging miR-199a in triple-negative breast cancer. Biomed Pharmacother. 2017;96:14-21. [PubMed ID: 28961506]. https://doi.org/10.1016/j.biopha.2017.09.107.

  • 31.

    Lan WG, Xu DH, Xu C, Ding CL, Ning FL, Zhou YL, et al. Silencing of long non-coding RNA ANRIL inhibits the development of multidrug resistance in gastric cancer cells. Oncol Rep. 2016;36(1):263-70. [PubMed ID: 27121324]. https://doi.org/10.3892/or.2016.4771.

  • 32.

    Zhang EB, Kong R, Yin DD, You LH, Sun M, Han L, et al. Long noncoding RNA ANRIL indicates a poor prognosis of gastric cancer and promotes tumor growth by epigenetically silencing of miR-99a/miR-449a. Oncotarget. 2014;5(8):2276-92. [PubMed ID: 24810364]. [PubMed Central ID: PMC4039162]. https://doi.org/10.18632/oncotarget.1902.

  • 33.

    Sun Y, Zheng ZP, Li H, Zhang HQ, 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. 2016;14(2):1714-20. [PubMed ID: 27314206]. https://doi.org/10.3892/mmr.2016.5409.

  • 34.

    Zhang JJ, Wang DD, Du CX, Wang Y. Long noncoding RNA ANRIL promotes cervical cancer development by acting as a sponge of miR-186. Oncol Res. 2017. [PubMed ID: 28550682]. https://doi.org/10.3727/096504017X14953948675449.

  • 35.

    Qiu JJ, Wang Y, Liu YL, Zhang Y, Ding JX, Hua KQ. The long non-coding RNA ANRIL promotes proliferation and cell cycle progression and inhibits apoptosis and senescence in epithelial ovarian cancer. Oncotarget. 2016;7(22):32478-92. [PubMed ID: 27095571]. [PubMed Central ID: PMC5078027]. https://doi.org/10.18632/oncotarget.8744.

  • 36.

    Chen D, Zhang Z, Mao C, Zhou Y, Yu L, Yin Y, et al. ANRIL inhibits p15(INK4b) through the TGFbeta1 signaling pathway in human esophageal squamous cell carcinoma. Cell Immunol. 2014;289(1-2):91-6. [PubMed ID: 24747824]. https://doi.org/10.1016/j.cellimm.2014.03.015.

  • 37.

    Hu X, Jiang H, Jiang X. Downregulation of lncRNA ANRIL inhibits proliferation, induces apoptosis, and enhances radiosensitivity in nasopharyngeal carcinoma cells through regulating miR-125a. Cancer Biol Ther. 2017;18(5):331-8. [PubMed ID: 28402230]. [PubMed Central ID: PMC5499759]. https://doi.org/10.1080/15384047.2017.1310348.

  • 38.

    Wang Y, Cheng N, Luo J. Downregulation of lncRNA ANRIL represses tumorigenicity and enhances cisplatin-induced cytotoxicity via regulating microRNA let-7a in nasopharyngeal carcinoma. J Biochem Mol Toxicol. 2017;31(7). [PubMed ID: 28117929]. https://doi.org/10.1002/jbt.21904.

  • 39.

    Iacobucci I, Sazzini M, Garagnani P, Ferrari A, Boattini A, Lonetti A, et al. A polymorphism in the chromosome 9p21 ANRIL locus is associated to Philadelphia positive acute lymphoblastic leukemia. Leuk Res. 2011;35(8):1052-9. [PubMed ID: 21414664]. https://doi.org/10.1016/j.leukres.2011.02.020.

  • 40.

    Poi MJ, Li J, Sborov DW, VanGundy Z, Cho YK, Lamprecht M, et al. Polymorphism in ANRIL is associated with relapse in patients with multiple myeloma after autologous stem cell transplant. Mol Carcinog. 2017;56(7):1722-32. [PubMed ID: 28150872]. https://doi.org/10.1002/mc.22626.

  • 41.

    Royds JA, Pilbrow AP, Ahn A, Morrin HR, Frampton C, Russell IA, et al. The rs11515 polymorphism is more frequent and associated with aggressive breast tumors with increased ANRIL and decreased p16 (INK4a) expression. Front Oncol. 2015;5:306. [PubMed ID: 26835415]. [PubMed Central ID: PMC4720739]. https://doi.org/10.3389/fonc.2015.00306.

  • 42.

    Huang MD, Chen WM, Qi FZ, Xia R, Sun M, Xu TP, et al. Long non-coding RNA ANRIL is upregulated in hepatocellular carcinoma and regulates cell apoptosis by epigenetic silencing of KLF2. J Hematol Oncol. 2015;8:50. [PubMed ID: 25966845]. [PubMed Central ID: PMC4434820]. https://doi.org/10.1186/s13045-015-0146-0.

  • 43.

    Naemura M, Murasaki C, Inoue Y, Okamoto H, Kotake Y. Long noncoding RNA ANRIL regulates proliferation of non-small cell lung cancer and cervical cancer cells. Anticancer Res. 2015;35(10):5377-82. [PubMed ID: 26408699].

  • 44.

    Lin L, Gu ZT, Chen WH, Cao KJ. Increased expression of the long non-coding RNA ANRIL promotes lung cancer cell metastasis and correlates with poor prognosis. Diagn Pathol. 2015;10:14. [PubMed ID: 25889788]. [PubMed Central ID: PMC4599723]. https://doi.org/10.1186/s13000-015-0247-7.

  • 45.

    Zhang D, Ding L, Li Y, Ren J, Shi G, Wang Y, et al. Midkine derived from cancer-associated fibroblasts promotes cisplatin-resistance via up-regulation of the expression of lncRNA ANRIL in tumour cells. Sci Rep. 2017;7(1):16231. [PubMed ID: 29176691]. [PubMed Central ID: PMC5701200]. https://doi.org/10.1038/s41598-017-13431-y.

  • 46.

    Kang YH, Kim D, Jin EJ. Down-regulation of phospholipase d stimulates death of lung cancer cells involving up-regulation of the long ncRNA ANRIL. Anticancer Res. 2015;35(5):2795-803. [PubMed ID: 25964559].

  • 47.

    Zhang D, Sun G, Zhang H, Tian J, Li Y. Long non-coding RNA ANRIL indicates a poor prognosis of cervical cancer and promotes carcinogenesis via PI3K/Akt pathways. Biomed Pharmacother. 2017;85:511-6. [PubMed ID: 27899255]. https://doi.org/10.1016/j.biopha.2016.11.058.

  • 48.

    Kang M, Sang Y, Gu H, Zheng L, Wang L, Liu C, et al. Long noncoding RNAs POLR2E rs3787016 C/T and HULC rs7763881 A/C polymorphisms are associated with decreased risk of esophageal cancer. Tumour Biol. 2015;36(8):6401-8. [PubMed ID: 25874495]. https://doi.org/10.1007/s13277-015-3328-z.

  • 49.

    Deng W, Wang J, Zhang J, Cai J, Bai Z, Zhang Z. TET2 regulates LncRNA-ANRIL expression and inhibits the growth of human gastric cancer cells. IUBMB Life. 2016;68(5):355-64. [PubMed ID: 27027260]. https://doi.org/10.1002/iub.1490.

  • 50.

    Zou ZW, Ma C, Medoro L, Chen L, Wang B, Gupta R, et al. LncRNA ANRIL is up-regulated in nasopharyngeal carcinoma and promotes the cancer progression via increasing proliferation, reprograming cell glucose metabolism and inducing side-population stem-like cancer cells. Oncotarget. 2016;7(38):61741-54. [PubMed ID: 27557514]. [PubMed Central ID: PMC5308687]. https://doi.org/10.18632/oncotarget.11437.

  • 51.

    Sayad A, Hajifathali A, Taheri M. Association of ANRIL gene polymorphisms with acute myeloid leukemia in an Iranian population. Int J Cancer Manag. 2017;10(10). https://doi.org/10.5812/ijcm.11176.

  • 52.

    Yu G, Liu G, Yuan D, Dai J, Cui Y, Tang X. Long non-coding RNA ANRIL is associated with a poor prognosis of osteosarcoma and promotes tumorigenesis via PI3K/Akt pathway. J Bone Oncol. 2018;11:51-5. [PubMed ID: 29520337]. [PubMed Central ID: PMC5842283]. https://doi.org/10.1016/j.jbo.2018.02.002.

  • 53.

    Meseure D, Vacher S, Alsibai KD, Nicolas A, Chemlali W, Pasmant E. Expression of ANRIL "Polycomb Complexes" CDKN2A/B/ARF genes in breast tumours: Identification of a two-gene (EZH2/CBX7) signature with independent prognostic value. 29th European Congress of Pathology. RAI Amsterdam, The Netherlands. 2017.