Long intergenic non‑protein coding RNA 152 promotes multiple myeloma progression by negatively regulating microRNA‑497

Retraction in: /10.3892/or.2021.8239

  • Authors:
    • Tianhua Yu
    • Zhihua Xu
    • Xuanhe Zhang
    • Lan Men
    • Haiying Nie
  • View Affiliations

  • Published online on: September 21, 2018     https://doi.org/10.3892/or.2018.6721
  • Pages: 3763-3771
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Long intergenic non‑protein coding RNA 152 (LINC00152, also known as cytoskeleton regulator RNA) is reportedly involved in the development and progression of various types of human malignancy. However, the functional role of LINC00152 in multiple myeloma (MM) and the underlying molecular mechanisms have remained elusive. The present study aimed to investigate the role of LINC00152 in the genesis of MM and the potential molecular mechanisms. It was identified that the expression of LINC00152 was significantly upregulated in plasma cells from patients with MM vs. healthy subjects, and that a high expression of LINC00152 was correlated with a shorter overall survival in patients with MM. Functional assays demonstrated that knockdown of LINC00152 by transfecting MM cells with LINC00152‑specific short hairpin RNA expression plasmids significantly inhibited cell proliferation by inducing cell cycle arrest at the G0/G1 phase. Furthermore, knockdown of LINC00152 promoted caspase‑3/9 activity and apoptosis in MM cells. In addition, knockdown of LINC00152 significantly attenuated tumor growth in mice bearing a myeloma xenograft. A luciferase reporter assay indicated that microRNA (miR)‑497 is a direct target of LINC00152, and its expression levels were inversely correlated with those of LINC00152 in MM tissues. Furthermore, repression of miR‑497 partly abrogated the inhibitory effects of LINC00152 knockdown on MM cells. Collectively, the results indicated that LINC00152 has an oncogenic effect in MM by targeting miR‑497, and may be a novel diagnostic marker and therapeutic target for MM.

Introduction

Multiple myeloma (MM) is the second most common type of hematologic cancer in the world, characterized by the clonal proliferation of malignant plasma cells, osteolytic bone destruction and pathological fractures (1,2). Although significant progress has been made in MM-associated research, the complex biological and core molecular mechanisms that are involved in the pathogenesis of MM have remained to be fully elucidated (3) Therefore, understanding the molecular events involved in the initiation and progression of MM and identifying novel therapies to treat MM remain a critical, albeit unmet goal.

Long non-coding RNAs (lncRNAs) are a class of transcripts of >200 nucleotides in length, which have no protein coding capability (4). Accumulating evidence has revealed that lncRNAs function as master regulators of gene expression at multiple levels, including transcriptional, post-transcriptional and epigenetic modulation (5,6). Several studies have indicated that dysregulated lncRNAs have a crucial role in various biological processes, including cell apoptosis, proliferation and differentiation, by serving as ‘micro (mi)RNA sponges’ (7). Furthermore, the dysregulated expression of lncRNAs has been indicated to be involved in the development and progression of various human cancer types, and these molecules are considered to be promising biomarkers and therapeutic agents for cancer (8,9). Numerous lncRNAs that have been implicated in MM progression and development by functioning as tumor suppressors or oncogenes (10,11). Consequently, understanding the underlying mechanism and biological functions of lncRNAs in MM may provide new approaches for its treatment.

Long intergenic non-protein coding RNA 152 (LINC00152), also known as cytoskeleton regulator RNA, which is located on chromosome 2p11.2 (12), has been reported to be upregulated in colorectal cancer (1315), breast cancer (16), lung cancer (17), tongue squamous cell carcinoma (18), gallbladder cancer (19) and renal cell carcinoma (20). These results emphasize the importance of the LINC00152 gene as an oncogene. However, the expression patterns and biological functions of LINC00152 in the genesis of MM, and the underlying mechanisms remain elusive. Therefore, LINC00152 expression was measured in MM tissues and cell lines, and a series of in vitro and in vivo experiments was performed to determine the biological functions and a possible molecular basis of LINC00152 in MM.

Materials and methods

Human tissue samples and cell lines

Plasma cell samples from 40 MM patients (mean age, 62.5±4.2 years; age range, 51.3–76.8 years; 22 males and 18 females) and from 40 healthy controls (mean age, 58.3.5±3.5 years; age range, 44.2.3–72.4 years; 20 males and 20 females) were obtained from the China-Japan Union Hospital of Jilin University (Changchun, China) between April 2012 and April 2014. The plasma cells were purified from bone marrow aspirates using CD138 MicroBeads (cat. no. 130-051-301; Miltenyi Biotec GmbH, Bergisch Gladbach, Germany), as described previously (19). The present study was approved by the Ethics Committee of Jilin University (Changchun, China) in accordance with the Declaration of Helsinki (2000) and written informed consent was obtained from all participants.

Three MM cell lines H929, MM1S and RPMI8226, and normal plasma cells (nPCs; cat. no. PCS-800-010™) were purchased from the American Type Culture Collection (Manassas, VA, USA). These cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), penicillin (100 U/ml) and streptomycin (100 U/ml) (Thermo Fisher Scientific, Inc.) at 37°C in a humidified atmosphere with 5% CO2.

Plasmid construction and transfection

miR-497 mimics, a miR-497 inhibitor, the corresponding negative control mimics (miR-NC), short hairpin (sh)RNA targeting LINC00152 (sh-LINC00152) and the empty lentiviral vector (sh-NC) were chemically synthesized and constructed by GenePharma Co., Ltd. (Shanghai, China). MM1S cells in the logarithmic growth phase were transfected with the abovementioned products using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. After transfection for 48 h, MM1S cells were screened with puromycin aminonucleoside to obtain those cells that stably expressed sh-LINC00152 after being transfected with the sh-LINC00152 plasmid. The transfection efficiency was assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis.

RT-qPCR

Total RNA was extracted from cultured cells and tissues using a high purity total RNA extraction kit (cat no. K0801; BioTeke Co., Beijing, China) according to the manufacturer's instructions. The miR-497 expression levels were measured using the TaqMan MicroRNA assay Kit (cat no. 4366596; Thermo Fisher Scientific, Inc.) in an ABI 7900 real-time PCR system (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. For the detection of LINC00152, complementary (c)DNAs were synthesized from RNA templates by using the RevertAid First Strand cDNA synthesis kit (Thermo Fisher Scientific, Inc.). The RT products were then amplified using SYBR Green reaction mix (Solarbio, Beijing, China) in an ABI 7900 real-time PCR system. The primers used in the present study were as follows: miR-497 sense, 5′-ACACTCCAGCTGGGCAGCAGCACACTGTGG-3′ and anti-sense, 5′-TGGTGTCGTGGAGTCG-3′; U6 sense, 5′-TGCGGGTGCTCGCTTCGGCAGC-3′ and anti-sense, 5′-CCAGTGCAGGGTCCGAGGT-3′. LINC00152 sense, 5′-TTGATGGCTTGAACATTTGG-3′ and anti-sense, 5′-TCGTGATTTTCGGTGTCTGT-3′; GAPDH sense, 5′-GAGTCAACGATTTGGTCGT-3′ and anti-sense, 5′-GACAAGCTTCCCGTTCTCAG-3′. The following PCR conditions were used: Denaturation at 94°C for 3 min, followed by 40 cycles of amplification (denaturation at 94°C for 10 sec, annealing at 60°C for 30 sec and extension at 72°C for 40 sec). Relative quantification of the target genes was performed with the comparative quantification cycle (Cq) method (2−∆∆Cq) (21). U6 and GAPDH were assessed as endogenous controls.

Cell proliferation and colony formation assays

Transfected cells were seeded in 96-well plates at a density of 5×103 cells/well in RPMI-1640 medium supplemented with 10% FBS. The proliferation of the cancer cells was measured with a Cell Counting Kit-8 (CCK-8; Dojindo Laboratories, Kumamoto, Japan) following the manufacturer's protocol. The optical density (OD) at 450 nm was detected using a Benchmark Plus microplate spectrometer (Bio-Rad Laboratories, Hercules, CA, USA).

MM1S cells stably expressing sh-LINC00152 were seeded on 6-well plates at a density of 500 cells/well and cultured for 10 days. The medium was changed every two days. After being gently washed with PBS, the colonies were then fixed with 4% paraformaldehyde for 30 min at 37°C and stained with 0.1% crystal violet for 5 min at 37°C. The visible colonies, consisting of >50 cells, were manually imaged and counted under a light microscope (Olympus Corp., Tokyo, Japan).

Cell cycle and apoptosis assays

For cell cycle analysis, cells were harvested via trypsinization at 48 h post-transfection, washed with cold PBS and fixed in 70% ice-cold ethanol overnight, followed by staining propidium iodide (PI, 50 mg/ml) in the presence of RNase A (50 mg/ml; Thermo Fisher Scientific, Inc.) for 30 min at 37°C.

For analysis of apoptosis, cells were harvested and re-suspended in fixation fluid at 48 h post-transfection. Apoptosis was determined using an Annexin V-FITC/PI apoptosis detection kit (BD Biosciences, San Jose, CA, USA). Cell cycle and apoptosis were analyzed on a FACSCalibur flow cytometer (BD Biosciences) and evaluation was performed with CellQuest 3.0 software (BD Biosciences).

Caspase-3/-9 activity assay

Caspase-3/-9 activity was determined by a Caspase-3/-9 Activity Assay Kit (cat. no. AAT-22820; Beyotime Institute of Biotechnology, Beijing, China) following the manufacturer's protocol. Caspase-3/-9 activities were determined by measuring the OD at 405 nm using a microplate reader (BioTek Instruments, Inc., Winooski, VT, USA).

Dual luciferase assay

The binding sites between LINC00152 and miR-497 were predicted using miRcode software (http://www.mircode.org). Fragments of the 3′ untranslated region (UTR) of LINC00152 containing the putative wild-type (WT) or mutated (MT) miR-497 binding site were chemically synthesized and cloned into the pmirGLO Dual-Luciferase miRNA Target Expression Vector (Promega Corp., Madison, WI, USA) between the XhoI and NotI sites, and named WT-LINC00152-3′UTR or MT-LINC00152-3′UTR. For the reporter assays, MM cells were co-transfected with WT-LINC00152-3′UTR or MT-LINC00152-3′UTR reporter plasmid and miR-497 mimics or miR-NC using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Firefly and Renilla luciferase activities in cell lysates were measured using the Dual Luciferase Reporter Assay system (Promega Corp.) at 48 h post-transfection. The relative luciferase activity was standardized to the Renilla luciferase activity.

Xenograft tumor model

A total of 10 male BALB/c-nu nude mice (age, 5–6 weeks; weight, 18–20 g) were obtained from the Experimental Animal Center of Jilin University (Changchun, China). All animal experiments were approved by the Ethics Committee of Jilin University (Changchun, China) and complied within the Guidelines for the Welfare and Use of Animals in Cancer Research (ad hoc committee of the National Cancer Research Institute, UK).

MM1S cells stably expressing sh-LINC00152 or sh-NC were subcutaneously injected into nude mice at a dose of 2×106 cells/mouse. The tumor growth was monitored by measuring tumor length (L) and width (W) weekly and calculating the volume (V) by using the formula V=(L × W2)/2. All animals were sacrificed at five weeks after tumor cell inoculation, and the subcutaneous tumors were excised and weighed. The tumor tissues were stored at −80°C until they were used to detect LINC00152 or miR-497 expression by RT-qPCR. Other parts of the tumors were fixed in neutral formalin, dehydrated and embedded in paraffin for further analysis.

Immunohistochemistry (IHC)

IHC was performed on paraffin-embedded xenograft tumors as described previously (20). Primary antibodies to Ki-67 were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA; cat. no. 23900) and used at a 1:400 dilution.

Statistical analysis

Values are expressed as the mean ± standard deviation from at least three independent repeats of the experiments. All statistical analyses were performed using SPSS v. 19.0 (IBM Corp., Armonk, NY, USA). Student's t-test was used for comparisons between two groups. One-way analysis of variance with Tukey's post-hoc test was employed to estimate the significant differences if >2 groups were present. Spearman's correlation analysis was used to analyze correlation in a data-set. The survival time of the patients was analyzed using the Kaplan-Meier method and the log-rank test. P<0.05 was considered to indicate a statistically significant difference between the groups.

Results

LINC00152 is upregulated in plasma cells from MM patients and MM cell lines

First, the expression levels of LIN00152 in plasma cells from 40 MM patients and 40 healthy donors were examined. RT-qPCR demonstrated that the expression of LINC00152 was significantly higher in plasma cells from MM patients than in those from healthy donors (Fig. 1A). Subsequently, the expression of LINC00152 in the three MM cell lines H929, MM1S and RPMI8226, and in the nPCs was evaluated. As presented in Fig. 1B, LINC00152 was significantly upregulated in the three MM cell lines as compared with that in nPCs (P<0.01). These results suggest that LINC00152 may have a role in the genesis of MM. Next, the MM patients were divided into two groups according to the expression levels of LINC00152 by using the median LINC00152 expression value as a cut-off, and it was further assessed whether the expression of LINC00152 is associated with the survival time of the patients using Kaplan-Meier analysis and the log-rank test. The results indicated that high LINC00152 expression corresponded with a significantly shorter overall survival as compared with that in the low expression group (Fig. 1C).

LINC00152 knockdown inhibits MM cell proliferation and induces cell cycle arrest

To assess the effect of LINC00152 on MM cells, the LINC00152 gene was knocked down in MM1S cells using sh-LINC00152 plasmid. The transfection efficiency was verified by RT-qPCR, revealing that MM1S cells transfected with the sh-LINC00152 plasmid had a significantly decreased LINC00152 expression as compared with that in sh-NC-transfected cells (Fig. 2A). The CCK-8 assay indicated that knockdown of LINC00152 in MM1S cells significantly reduced their viability (Fig. 2B) and furthermore, their colony formation ability was significantly impaired (Fig. 2C). To investigate the mechanisms involved in the effect of LINC00152 on the proliferation and clonogenicity of MM cells, the cell cycle was assessed using flow cytometry. The results demonstrated that LINC00152 knockdown induced cell cycle arrest of MM cells at G0/G1 phase and decreased cell cycle progression to the S phase (Fig. 2D).

LINC00152 knockdown induces MM-cell apoptosis

The effect of LINC00152 silencing on the apoptosis of MM1S cells was then explored by fluorescence-assisted cell sorting analysis. As presented in Fig. 3A, LINC00152 knockdown significantly induced apoptosis in MM1S cells (P<0.01). In order to investigate the underlying mechanism, the activities of caspase-3 and caspase-9 were measured with a Caspase-3/-9 Activity Assay Kit. The intracellular activities of caspase-3 and caspase-9 were significantly increased in MM1S cells transfected with sh-LINC00152 as compared with that in sh-NC-transfected cells (Fig. 3B and C).

miR-497 expression is directly regulated by LINC00152

Vast evidence has indicated that lncRNAs function as competing endogenous RNAs (ceRNAs) by competitively binding to miRNAs. To identify miRNAs that are directly targeted by LINC00152 in MM cells, the bioinformatics tool miRcode was used to predict potential target miRs of LINC00152, of which miR-497 was selected as a putative target (Fig. 4A). To confirm the direct binding interaction between LINC00152 and miR-497, a luciferase reporter assay was performed. The results demonstrated that miR-497 mimics reduced the luciferase activity of LINC00152-WT but not of LINC00152-MT vectors (Fig. 4B; P<0.05). Furthermore, LINC00152 knockdown significantly increased the levels of miR-497 in MM cells (Fig. 4C; P<0.05). However, miR-497 mimics or inhibitor did not affect LINC00152 levels in MM cells (Fig. 4D; P>0.05). In addition, in plasma cells from MM patients, the expression of miR-497 was significantly downregulated and was inversely correlated with the expression of LINC00152 (Fig. 4E and F). These results indicate that LINC00152 directly binds to miR-497 to reduce its availability.

Repression of miR-497 restores the inhibitory effects of sh-LINC00152 on MM cells

To investigate whether LINC00152 exerts its oncogenic effect on MM cells through miR-497, a loss-of-function experiment was performed by inhibiting miR-497 expression in LINC00152-knockdown MM1S cells (Fig. 5A). The CCK-8 assay and the colony formation assay indicated that the miR-497 inhibitor promoted the proliferation and colony formation (Fig. 5B and C; P<0.05), and partially abrogated the LINC00152 knockdown-induced reduction in cell proliferation and colony formation ability on MM1S cells (Fig. 5B and C; P<0.05). Flow cytometric analysis further indicated that apoptosis was increased in LINC00152-knockdown MM1S cells, which was partially abrogated by co-transfection with miR-497 inhibitor (Fig. 5D; P<0.05). These results suggest that repression of miR-497 partially restored the inhibitory effects of sh-LINC00152 on MM cells.

LINC00152 knockdown suppresses tumor growth in vivo

To further investigate the role of LINC00152 in tumor growth in vivo, MM1S cells stably expressing sh-LINC00152 or sh-NC were subcutaneously injected into nude mice. Xenograft tumors were evaluated over the duration of the experiment and excised at seven weeks after inoculation. It was indicated that the tumor volumes and final weight in the sh-LINC00152 group were significantly lower than those in the sh-NC group (P<0.05; Fig. 6A-C). Furthermore, LINC00152 and miR-497 expression in tumor tissues was examined by RT-qPCR. The results indicated that LINC00152 expression was significantly decreased, whereas miR-497 expression was increased in the sh-LINC00152 group as compared with that in the sh-NC group (P<0.05; Fig. 6D and E). IHC staining of the tumors revealed that the number of Ki-67-positive cells was obviously reduced in the sh-LINC00152 group as compared with that in the sh-NC group (Fig. 6F). These results suggest that LINC00152 knockdown reduces the tumorigenesis of MM in vivo.

Discussion

Increasing evidence has suggested that dysregulation of lncRNAs is involved in the genesis and progression of MM (10,11). For instance, overexpression of lncRNA imprinted RNA near maternally expressed 3 promoted the osteogenic differentiation of mesenchymal stem cells from MM patients by targeting the transcription of bone morphogenetic protein 4 (22). Knockdown of the lncRNA H19, imprinted maternally expressed transcript by shRNA transfection significantly inhibited the proliferation, viability and colony formation in MM cells, and inactivated the nuclear factor-κB pathway (23). Knockdown of lncRNA metastasis-associated lung adenocarcinoma transcript 1 significantly suppressed the cell proliferation, induced apoptosis, caused cell cycle arrest in G1/S phase and inhibited MM cell growth in vivo through sponging miR-509-5p to modulate forkhead box P1 expression (24). lncRNA colon cancer-associated transcript 1 promoted MM growth in vitro and in vivo by acting as a molecular sponge for miR-181a-5p to modulate homeobox A1 expression (25). In the present study, it was identified that LINC00152 was highly expressed in plasma cells from MM patients and MM cell lines, and the upregulation of LINC00152 in their plasma cells was significantly associated with poor prognosis of MM patients. In addition, it was verified that LINC00152 knockdown significantly suppressed MM cell proliferation and clonogenicity, and induced apoptosis. These results suggest that LINC00152 may serve as a promising target for therapeutic intervention in MM.

Several studies have demonstrated that LINC00152 has considerable functional roles in various human malignancies by regulating cancer cell proliferation, apoptosis, metastasis, migration and invasion (1220). It was reported to be upregulated and to function as an oncogene in various cancer types (26,27). However, the role of LINC00152 in MM and the underlying mechanisms have remained largely elusive. In the present study, RT-qPCR confirmed that LINC00152 was highly expressed in plasma cells from MM patients and MM cell lines, and that high LINC00152 expression in plasma cells was associated with a relatively shorter survival of patients with MM. In addition, knockdown of LINC00152 in MM1S cells significantly suppressed their proliferation and colony formation, while promoting cell apoptosis, as well as and caspase-3 and caspase-9 activity. An in vivo xenograft experiment demonstrated that knockdown of LINC00152 suppressed the growth of tumors derived from MM1S cells in nude mice. These results suggest that LINC00152 functions as an oncogene in human MM.

lncRNAs are known to be involved in a series of cellular biological processes by acting as ceRNAs or molecular sponges to modulate the effect of miRNAs (28,29). Previous studies have demonstrated that LINC00152 has an oncogenic role in colorectal cancer by regulating miR-376c-3p (30), in glioma by regulating miR-103a-3p (31) and in gastric cancer by targeting miR-139-5p (32). In the present study, miR-497 was identified as an inhibitory target of LINC00152 by sequence complementarity analysis and luciferase reporter assays. miR-497, a member of the miR-15 family, has been reported to have a critical role in cancer progression and development (33). A study has indicated that miR-497 expression was downregulated in MM samples and cell lines, and that its overexpression significantly inhibited malignant progression of MM by directly targeting PBX homeobox 3 (34). In line with this, the present results indicated that miR-497 was downregulated in MM samples. In addition, miR-497 expression was inversely correlated with LINC00152 expression. Furthermore, LINC00152 knockdown increased miR-497 expression in MM cells. In addition, it was demonstrated that the miR-497 inhibitor partly abrogated the effect of LINC00152 knockdown on MM growth. These results strongly suggest that LINC00152 directly targets miR-497 and affects the biological characteristics of MM cells by negatively regulating miR-497.

Various limitations of the present study should be considered. First, only one MM cell line was used to examine the biological function of LINC00152 in MM progression, and experiments using further cells lines may be required for further validation. In addition, a gain-of-function study with upregulation of LINC00152 in MM cells should be performed to fully determine the role of LINC00152 in MM. Furthermore, LINC00152 may affect drug resistance of patients with MM, which requires further study.

In conclusion, the present study indicated that LINC00152 is highly expressed in plasma cells from MM patients and MM cell lines. In addition, it was revealed that LINC00152 acts as an oncogene and modulates MM cell growth by targeting miR-497. These results provided insight into the biological and clinical significance of LINC00152. Based on the present results, LINC00152 may potentially be utilized as a biomarker for MM and may serve as a therapeutic target for the treatment of patients with MM.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

TY, ZX and HN conceived the study; XZ and LM performed the experiments; TY and ZX analyzed the data; and TY, ZX and HN wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Jilin University (Changchun, China) in accordance with the Declaration of Helsinki (2000) and written informed consent was obtained from all participants.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Alexander DD, Mink PJ, Adami HO, Cole P, Mandel JS, Oken MM and Trichopoulos D: Multiple myeloma: A review of the epidemiologic literature. Int J Cancer. 12 Suppl 120:S40–S61. 2007. View Article : Google Scholar

3 

Hideshima T, Richardson PG and Anderson KC: Mechanism of action of proteasome inhibitors and deacetylase inhibitors and the biological basis of synergy in multiple myeloma. Mol Cancer Ther. 10:2034–2042. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Kornienko AE, Guenzl PM, Barlow DP and Pauler FM: Gene regulation by the act of long non-coding RNA transcription. BMC Biol. 11:592013. View Article : Google Scholar : 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 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: Insights into functions. Nat Rev Genet. 10:155–159. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Geisler S and Coller J: RNA in unexpected places: Long non-coding RNA functions in diverse cellular contexts. Nat Rev Mol Cell Biol. 14:699–712. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Sun T: Long noncoding RNAs act as regulators of autophagy in cancer. Pharmacol Res. 129:151–155. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Fan Q, Yang L, Zhang X, Peng X, Wei S, Su D, Zhai Z, Hua X and Li H: The emerging role of exosome-derived non-coding RNAs in cancer biology. Cancer Lett. 414:107–115. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Hu AX, Huang ZY, Zhang L and Shen J: Potential prognostic long non-coding RNA identification and their validation in predicting survival of patients with multiple myeloma. Tumour Biol. 39:10104283176945632017. View Article : Google Scholar : PubMed/NCBI

11 

Ronchetti D, Manzoni M, Todoerti K, Neri A and Agnelli L: In silico characterization of miRNA and long non-coding RNA interplay in multiple myeloma. Genes (Basel). 7:E1072016. View Article : Google Scholar : PubMed/NCBI

12 

Liu L, Wen J, Gu X, Wu D, Lu M and Zhao Q: Prognostic role of long non-coding RNA LINC00152 in Chinese cancer patients: A meta-analysis. Oncotarget. 8:93227–93235. 2017.PubMed/NCBI

13 

Bian Z, Zhang J, Li M, Feng Y, Yao S, Song M, Qi X, Fei B, Yin Y, Hua D and Huang Z: Long non-coding RNA LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in colorectal cancer by inhibiting miR-139-5p. Oncogenesis. 6:3952017. View Article : Google Scholar : PubMed/NCBI

14 

Wang X, Yu H, Sun W, Kong J, Zhang L, Tang J, Wang J, Xu E, Lai M and Zhang H: The long non-coding RNA CYTOR drives colorectal cancer progression by interacting with NCL and Sam68. Mol Cancer. 17:1102018. View Article : Google Scholar : PubMed/NCBI

15 

Yue B, Liu C, Sun H, Liu M, Song C, Cui R, Qiu S and Zhong M: A positive feed-forward loop between LncRNA-CYTOR and Wnt/β-catenin signaling promotes metastasis of colon cancer. Mol Ther. 26:1287–1298. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Wu J, Shuang Z, Zhao J, Tang H, Liu P, Zhang L, Xie X and Xiao X: Linc00152 promotes tumorigenesis by regulating DNMTs in triple-negative breast cancer. Biomed Pharmacother. 97:1275–1281. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Zhang Y, Xiang C, Wang Y, Duan Y, Liu C, Jin Y and Zhang Y: lncRNA LINC00152 knockdown had effects to suppress biological activity of lung cancer via EGFR/PI3K/AKT pathway. Biomed Pharmacother. 94:644–651. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Yu J, Liu Y, Guo C, Zhang S, Gong Z, Tang Y, Yang L, He Y, Lian Y, Li X, et al: Upregulated long non-coding RNA LINC00152 expression is associated with progression and poor prognosis of tongue squamous cell carcinoma. J Cancer. 8:523–530. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Cai Q, Wang Z, Wang S, Weng M, Zhou D, Li C, Wang J, Chen E and Quan Z: Long non-coding RNA LINC00152 promotes gallbladder cancer metastasis and epithelial-mesenchymal transition by regulating HIF-1alpha via miR-138. Open Biol. 7:1602472017. View Article : Google Scholar : PubMed/NCBI

20 

Wu Y, Tan C, Weng WW, Deng Y, Zhang QY, Yang XQ, Gan HL, Wang T, Zhang PP, Xu MD, et al: Long non-coding RNA Linc00152 is a positive prognostic factor for and demonstrates malignant biological behavior in clear cell renal cell carcinoma. Am J Cancer Res. 6:285–299. 2016.PubMed/NCBI

21 

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 : PubMed/NCBI

22 

Zhuang W, Ge X, Yang S, Huang M, Zhuang W, Chen P, Zhang X, Fu J, Qu J and Li B: Upregulation of lncRNA MEG3 promotes osteogenic differentiation of mesenchymal stem cells from multiple myeloma patients by targeting BMP4 transcription. Stem Cells. 33:1985–1997. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Sun Y, Pan J, Zhang N, Wei W, Yu S and Ai L: Knockdown of long non-coding RNA H19 inhibits multiple myeloma cell growth via NF-κB pathway. Sci Rep. 7:180792017. View Article : Google Scholar : PubMed/NCBI

24 

Gu Y, Xiao X and Yang S: LncRNA MALAT1 acts as an oncogene in multiple myeloma through sponging miR-509-5p to modulate FOXP1 expression. Oncotarget. 8:101984–101993. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Chen L, Hu N, Wang C, Zhao H and Gu Y: Long non-coding RNA CCAT1 promotes multiple myeloma progression by acting as a molecular sponge of miR-181a-5p to modulate HOXA1 expression. Cell Cycle. 17:319–329. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Ji J, Tang J, Deng L, Xie Y, Jiang R, Li G and Sun B: LINC00152 promotes proliferation in hepatocellular carcinoma by targeting EpCAM via the mTOR signaling pathway. Oncotarget. 6:42813–42824. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Quan FY, Jiang J, Zhai YF, Li B, Wu XH and Nie W: The prognostic effect of LINC00152 for cancer: A meta-analysis. Oncotarget. 8:75427–75433. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Huang C, Yuan N, Wu L, Wang X, Dai J, Song P, Li F, Xu C and Zhao X: An integrated analysis for long noncoding RNAs and microRNAs with the mediated competing endogenous RNA network in papillary renal cell carcinoma. Onco Targets Ther. 10:4037–4050. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Xue M, Zhuo Y and Shan B: MicroRNAs, long noncoding RNAs, and their functions in human disease. Methods Mol Biol. 1617:1–25. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Zhang YH, Fu J, Zhang ZJ, Ge CC and Yi Y: LncRNA-LINC00152 down-regulated by miR-376c-3p restricts viability and promotes apoptosis of colorectal cancer cells. Am J Transl Res. 8:5286–5297. 2016.PubMed/NCBI

31 

Yu M, Xue Y, Zheng J, Liu X, Yu H, Liu L, Li Z and Liu Y: Linc00152 promotes malignant progression of glioma stem cells by regulating miR-103a-3p/FEZF1/CDC25A pathway. Mol Cancer. 16:1102017. View Article : Google Scholar : PubMed/NCBI

32 

Sun K, Hu P and Xu F: LINC00152/miR-139-5p regulates gastric cancer cell aerobic glycolysis by targeting PRKAA1. Biomed Pharmacother. 97:1296–1302. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Yang G, Xiong G, Cao Z, Zheng S, You L, Zhang T and Zhao Y: miR-497 expression, function and clinical application in cancer. Oncotarget. 7:55900–55911. 2016.PubMed/NCBI

34 

Yu T, Zhang X, Zhang L, Wang Y, Pan H, Xu Z and Pang X: MicroRNA-497 suppresses cell proliferation and induces apoptosis through targeting PBX3 in human multiple myeloma. Am J Cancer Res. 6:2880–2889. 2016.PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 40 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu T, Xu Z, Zhang X, Men L and Nie H: Long intergenic non‑protein coding RNA 152 promotes multiple myeloma progression by negatively regulating microRNA‑497 Retraction in /10.3892/or.2021.8239. Oncol Rep 40: 3763-3771, 2018
APA
Yu, T., Xu, Z., Zhang, X., Men, L., & Nie, H. (2018). Long intergenic non‑protein coding RNA 152 promotes multiple myeloma progression by negatively regulating microRNA‑497 Retraction in /10.3892/or.2021.8239. Oncology Reports, 40, 3763-3771. https://doi.org/10.3892/or.2018.6721
MLA
Yu, T., Xu, Z., Zhang, X., Men, L., Nie, H."Long intergenic non‑protein coding RNA 152 promotes multiple myeloma progression by negatively regulating microRNA‑497 Retraction in /10.3892/or.2021.8239". Oncology Reports 40.6 (2018): 3763-3771.
Chicago
Yu, T., Xu, Z., Zhang, X., Men, L., Nie, H."Long intergenic non‑protein coding RNA 152 promotes multiple myeloma progression by negatively regulating microRNA‑497 Retraction in /10.3892/or.2021.8239". Oncology Reports 40, no. 6 (2018): 3763-3771. https://doi.org/10.3892/or.2018.6721