Oncotarget

Research Papers:

This article has been retracted. Retraction in: Oncotarget. 2024; 15:255-255.

Knockdown of NEAT1 restrained the malignant progression of glioma stem cells by activating microRNA let-7e

PDF |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:62208-62223. https://doi.org/10.18632/oncotarget.11403

Metrics: PDF 3032 views  |   HTML 3066 views  |   ?  

Wei Gong, Jian Zheng, Xiaobai Liu, Jun Ma, Yunhui Liu and Yixue Xue _

Abstract

Wei Gong1,2, Jian Zheng3,4, Xiaobai Liu3,4, Jun Ma1,2, Yunhui Liu3,4, Yixue Xue1,2

1Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, People’s Republic of China

2Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110122, People’s Republic of China

3Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People’s Republic of China

4Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang 110004, People’s Republic of China

Correspondence to:

Yixue Xue, email: [email protected]

Keywords: lncRNAs, NEAT1, glioma stem cells, let-7e, NRAS

Received: March 18, 2016    Accepted: August 09, 2016    Published: August 19, 2016

ABSTRACT

Nuclear paraspeckle assembly transcript 1 (NEAT1), a long non-coding RNA, promotes oncogenesis in various tumors, including human gliomas. Herein, we studied the expression and function of NEAT1 in glioma stem cells (GSCs). Quantitative real-time PCR demonstrated that NEAT1 was upregulated in GSCs. NEAT1 knockdown inhibited GSC cell proliferation, migration and invasion and promoted GSC apoptosis. A potential binding region between NEAT1 and microRNA let-7e was confirmed by dual-luciferase assays. Upregulation of NEAT1 reduced the expression of let-7e, and there was reciprocal repression between NEAT1 and let-7e in an Argonaute 2-dependent manner. Let-7e expression was lower expression in glioblastoma tissues and GSCs than in normal brain tissues and cells. Restoration of let-7e suppressed tumor function by inhibiting proliferation, migration and invasion while promoting apoptosis in GSCs. NEAT1 knockdown and let-7e overexpression both reduced NRAS protein expression. NRAS was identified as a direct target of let-7e and promoted oncogenesis in GSCs. As NEAT1 promoted oncogenesis by downregulating let-7e expression, both of these genes could be considered for application in glioma therapy.


INTRODUCTION

Glioma is one of the most prevalent and aggressive primary brain tumors in adults. Despite treatment with advanced therapeutic strategies, patients with this disease only have a median survival of 15 months [1]. Glioma stem cells (GSCs) are a subpopulation of glioma cells, and are characterized by self-renewal, promotion of angiogenesis, and multi-differentiation [2]. GSCs are important contributors to the malignant progression of glioma, from its development to therapeutic resistance and recurrence [3]. Therefore, it is urgent to discover the molecular mechanisms by which GSCs are maintained, as this would provide a new focus for the development of glioma treatments.

Long non-coding RNAs (lncRNAs, ~200nt) are a class of RNAs that do not encode proteins [4]. LncRNAs are widely expressed in various human somatic tissues, and are involved in diverse cellular events, including epigenetic regulation, gene transcription, mRNA processing and gene translation [5]. LncRNAs are ubiquitously dysregulated in tumor cells and have crucial regulatory roles in the malignant progression of tumor cells, such as promotes or suppresses proliferation, migration and invasion, and apoptosis [6]. Nuclear paraspeckle assembly transcript 1 (NEAT1) is a 4-kb lncRNA, and has been reported to localize to the nucleus, where it serves as a core component of the paraspeckle sub-organelles [79]. NEAT1 is upregulated and has important functions in a variety of cancers including glioma, such as favors cell proliferation, migration and invasion and impaired apoptosis [1013]. However, whether NEAT1 is associated with the malignant progression of GSCs remains unclear.

MicroRNAs (miRNAs, ~22nt) are a group of small non-coding RNAs with aberrant expression in various tumors. MiRNAs are involved in diverse biological processes, such as cell growth, migration, apoptosis and differentiation, by binding to the 3′-UTRs of mRNAs [14, 15]. The let-7 family of miRNAs comprises 13 members, one of which is let-7e [16]. The let-7 family is characterized by regulating diverse biological processes in cancer cells, including inhibited proliferation and promoted apoptosis [17]. The downregulation of let-7e is known to be a prognostic marker of squamous-cell lung carcinoma [18]. Further, let-7e is repressed in medulloblastoma [19]. Let-7e expression is reduced in deep vein thrombosis and prevents cell endothelial progenitor function by binding to FASLG [20]. Remarkably, let-7e can bind to MMP9 and induce adipose-derived stem cell differentiation [21]. However, the expression and function of let-7e in GSCs remain unclear.

The RAS gene is recognized to encode three isoforms - HRAS, KRAS and NRAS [22] - and is involved in diverse cellular events and signaling pathways. NRAS is approximately 4.3 kb in length and is aberrantly expressed in many tumors, including colorectal cancer and cutaneous melanoma [2325]. However, little has been studied about the function of NRAS in GSCs.

In the current study, we sought to determine the expression and function of NEAT1, let-7e and NRAS in glioma tissues and GSCs. We also investigated the interactions among them in the regulation of GSC malignant behavior and the potential molecular pathways involved.

RESULTS

Isolation and identification of GSCs

Cells isolated from U87 and U251 cell lines were cultured in serum-free medium and allowed to form cell spheres (Figure 1A (a, c)). In an effort to verify the self-renewing abilities of the cells, we harvested the spheres and performed a second round of the sphere-forming assay. As expected, spheres were generated again from single cells (Figure 1A (b, d)). The positive staining of Nestin and CD133 confirmed that most cells within the spheres expressed these neural stem cell lineage markers on their membranes (Figure 1B). Moreover, the cell spheres stained positively for glial fibrillary acidic protein (GFAP) and beta-tubulin-III (lineage markers), suggesting that they were undergoing typical morphological differentiation towards astrocytic and neuronal lineages (Figure 1C). Further, GSCs-U87 and GSCs-U251 induced larger xenografted tumors in mice than non-GSCs, indicating their stronger tumorigenicity (Figure 1D).

Isolation and identification of GSCs.

Figure 1: Isolation and identification of GSCs. A. a and c: U87 and U251 glioma cells formed spheres in serum-free medium. b and d: single cells formed spheres again in a second-round sphere-forming assay. B. GSCs-U87 and GSCs-U251 stained for Nestin (green) and CD133 (red) by immunofluorescence analysis. C. GSC-U87 and GSC-U251 spheres were differentiated and then stained for GFAP (green) and beta-tubulin III (red) by immunofluorescence. D. Subcutaneously implanted GSCs-U87 or GSCs-U251 formed xenografts in nude mice. Data are presented as the mean ± SD (n=5, each group). **P<0.01 vs. GSC-U87 group; ##P<0.01 vs. GSC-U251 group.

NEAT1 was upregulated while let-7e was downregulated in GSCs

As previously reported, NEAT1 was upregulated in glioblastoma (GBM) tissues [10]. Also, we found NEAT1 was upregulated in GBM U87 and U251 cell lines (Figure 2A and 2B). Quantitative real-time PCR (qRT-PCR) was conducted to determine the expression of NEAT1 in two additional glioma cell lines and GSCs. As shown in Figure 2A and 2B, NEAT1 expression was significantly upregulated in T98, A172, GSC-U87 and GSC-U251 cells. On the contrary, let-7e expression was significantly lower in GBM tissues and GBM cell lines than in normal brain tissues and normal human astrocytes, and correlated negatively with the glioma pathological grade (Figure 2C2E). These results suggested that NEAT1 promotes oncogenesis in GSCs, while let-7e functions as a tumor suppressor.

NEAT1 and let-7e levels in glioma tissues and GSCs.

Figure 2: NEAT1 and let-7e levels in glioma tissues and GSCs. A. Expression of NEAT1 in human normal astrocytes and glioma cell lines (n=5, each group). **P<0.01 vs. Normal human astrocyte group. B. Expression of NEAT1 in non-GSCs and GSCs (n=5, each group). **P<0.01 vs. non-GSC-U87 group; ##P<0.01 vs. non-GSC-U251 group. C. Expression of let-7e in glioma tissues of different grades and NBTs (n=30, each group). **P<0.01 vs. NBT group; ##P<0.01 vs. Grade I group; ΔΔP<0.01 vs. Grade II group;ΨΨP<0.01 vs. Grade III group. D. Expression of let-7e in human normal astrocytes and glioma cell lines (n=5, each group). **P<0.01 vs. normal human astrocyte group. E. Expression of let-7e in non-GSCs and GSCs (n=5, each group). **P<0.01 vs. non-GSC-U87 group; ##P<0.01 vs. non-GSC-U251 group. All data are presented as the mean ± SD

Knockdown of NEAT1 impaired the malignant progression of GSCs

To determine the effect of NEAT1 on GSCs, we divided cells into three groups: the control group, sh-NC group (transfected with the sh-NC plasmid), sh-NEAT1 group (transfected with the sh-NEAT1 plasmid) and the sh-NC group (transfected with the sh-NC plasmid). The Cell Counting Kit-8 (CCK-8) assay indicated that GSC proliferation was lower in the NEAT1-knockdown group than in the sh-NC group (Figure 3A). The migration and invasion of GSCs were also significantly lower in the sh-NEAT1 group than in the sh-NC group (Figure 3B). Moreover, GSCs treated with sh-NEAT1 exhibited weaker migration abilities than control cells in 3D Spheroid-based tumor migration assays (Figure 3C). Flow cytometry analysis revealed that the inhibition of NEAT1 significantly increased GSC apoptosis (Figure 3D). These results indicated that NEAT1 might act as an oncogene in GSCs.

Knockdown of NEAT1 restrained cell proliferation, migration and invasion and promoted apoptosis of GSCs.

Figure 3: Knockdown of NEAT1 restrained cell proliferation, migration and invasion and promoted apoptosis of GSCs. A. ACCK-8 assay was used to determine the effect of NEAT1 on GSC proliferation. B. Quantification of the migration and invasion of NEAT1-knockdown GSCs. Representative images and accompanying statistical plots are presented. C. 3D Spheroid-based tumor migration assays of the effect of NEAT1 expression on GSC migration. Scale bars, 60 μm. D. Flow cytometry analysis of the effect of NEAT1 knockdown on GSCs. Data are presented as the mean ± SD (n=5, each group). *P<0.05 vs. sh-NC group. Scale bars, 20 μm.

Let-7e functioned as a tumor suppressor

Similarly, to determine the effect of let-7e on GSCs, we divided cells into five groups: the control group, pre-NC group (transfected with let-7e agomir NC), pre-let-7e group (transfected with the let-7e agomir), anti-NC group (transfected with let-7e antagomir NC) and anti-let-7e (transfected with the let-7e antagomir). A CCK-8 assay revealed that GSC proliferation was lower in the let-7e overexpression group (pre-let-7e) than in the pre-NC group (Figure 4A). Transwell assays were conducted to assess the effects of let-7e overexpression on the invasiveness and migratory abilities of GSCs. The migration and invasion of GSCs were lower in the pre-let-7e group than in the pre-NC group (Figure 4B). Similarly, GSC spheroid migration was attenuated (Figure 4C) and apoptosis was enhanced in the let-7e overexpression group relative to the anti-let-7e group. Thus, it is conceivable that let-7e, in contrast to NEAT1, functions as a tumor suppressor in GSCs (Figure 4D).

Restoration of let-7e inhibited cell proliferation, migration and invasion and facilitated apoptosis of GSCs.

Figure 4: Restoration of let-7e inhibited cell proliferation, migration and invasion and facilitated apoptosis of GSCs. A. The CCK-8 assay was employed to determine the effect of let-7e on GSC proliferation. B. Quantification of GSC migration and invasion in groups according to let-7e expression. Representative images and accompanying statistical plots are presented. C. 3D Spheroid-based tumor migration assays of the effect let-7e expression on GSC migration. Scale bars, 60 μm. D. Flow cytometry analysis of GSCs in groups according to let-7e expression. Data are presented as the mean ± SD (n=5, each group). *P<0.05 vs. pre-NC group; #P<0.05 vs. anti-NC group. Scale bars, 20 μm.

NEAT1 is a direct target of let-7e

There is increasing evidence that lncRNAs could be competing endogenous RNAs (ceRNAs) or molecular sponges in down-regulating the expression and biological functions of miRNAs, Using a bioinformatics database (Starbase), we determined that NEAT1 harbors two putative binding sites for let-7e (Figure 5B). To validate our hypothesis that let-7e could directly bind to NEAT1, we first measured the expression of let-7e in sh-NEAT1 GSCs by qRT-PCR. Let-7e expression was elevated in the sh-NEAT1 group but not in the sh-NEAT1-Mut group. In contrast, NEAT1 expression was reduced in the pre-let-7e group (Figure 5A). Further, dual-luciferase gene reporter assays were used to assess the binding sites of NEAT1 and let-7e. The luciferase activity was significantly lower in the NEAT1-Wt1+pre-let-7e group than in the control group (Figure 5C), while the luciferase activity did not differ between the NEAT1-Mut1+pre-let-7e group and the control group, suggesting that binding site 1 between NEAT1 and let-7e was functional.

Binding and reciprocal repression between let-7e and NEAT1 determined GSC malignant behavior.

Figure 5: Binding and reciprocal repression between let-7e and NEAT1 determined GSC malignant behavior. A. qRT-PCR analysis demonstrating the negative correlation between let-7e and NEAT1 expression in GSCs. **P<0.01 vs. sh-NC group; **P<0.01 vs. pre-NC group; ##P<0.01 vs. sh-NC group. B. NEAT1 harbored two putative let-7e binding sites; the designed mutant sequences are indicated. C. Dual-luciferase reporter assay of HEK 293T cells co-transfected with NEAT1-Wt1 (or NEAT1-Wt2) and let-7e-NC; NEAT1-Wt1 (or NEAT1-Wt2) and let-7e; NEAT1-Mut1 (or NEAT1-Mut2) and let-7e-NC; or NEAT1-Mut1 (or NEAT1-Mut2) and let-7e. *P<0.05 vs. NEAT1-Wt1+let-7e-NC group. D. Let-7e was identified in the NEAT1-RISC complex. NEAT1 and let-7e levels were measured by qRT-PCR. &&P < 0.01 vs. the anti-normal IgG group. E. The CCK-8 assay was applied to evaluate the effects of NEAT1 and let-7e on GSC proliferation. F. Quantification of the migration and invasion of GSCs in groups according to NEAT1 and let-7e expression. Representative images and accompanying statistical plots are presented. G. 3D Spheroid-based tumor migration assays of the effect of NEAT1 and let-7e co-transfection on GSC migration. Scale bars, 60 μm. H. Flow cytometry analysis of GSCs in groups according to NEAT1 and let-7e expression. *P<0.05 vs. sh-NC+pre-NC group. Scale bars, 20 μm. For A, C, D, E and H, data are presented as the mean ± SD (n=5, each group).

An RNA-binding protein immunoprecipitation (RIP) assay was conducted to determine whether NEAT1 and let-7e were in the expected RNA-induced silencing complex (RISC). We explored RNA levels by qRT-PCR, and found NEAT1 and let-7e immunoprecipitated with Argonaute 2 were repressed than those in the control group, respectively (Figure 5D). Thus, NEAT1 inhibition was confirmed to restore let-7e expression in a RISC-dependent manner, and there was a reciprocal repression feedback loop between NEAT1 and let-7e.

NEAT1 inhibition hindered the malignant progression of GSCs by upregulating let-7e

Further, to explore the mechanism whereby NEAT1 promoted malignant behavior in GSCs by attenuating let-7e expression, we divided cells into five groups: the control group, the sh-NC+pre-NC group (cells stably expressing sh-NC, co-transfected with pre-NC), sh-NEAT1+pre-let-7e group (cells stably expressing sh-NEAT1, co-transfected with pre-let-7e), sh-NC+anti-NC group (cells stably expressing sh-NC, co-transfected with anti-NC) and sh-NEAT1+anti-let-7e group (cells stably expressing sh-NEAT1, co-transfected with anti-let-7e). GSC proliferation was markedly lower in the sh-NEAT1+pre-let-7e group than in the sh-NC+pre-NC group (Figure 5E). Moreover, the numbers of migrating and invading GSCs were significantly lower in the sh-NEAT1+pre-let-7e group than in the control group (Figure 5F). In addition, GSCs treated with sh-NEAT1+pre-let-7e exhibited weaker migration abilities than controls in 3D Spheroid-based tumor migration assays (Figure 5G). The apoptotic ratio of GSCs with knockdown of NEAT1 and overexpression of let-7e was robustly elevated compared with that in the sh-NC+pre-NC group (Figure 5H). These results indicated that knockdown of NEAT1 hindered malignant behavior in GSCs by upregulating let-7e.

NRAS was upregulated in glioma tissues and GSCs, and facilitated GSC proliferation, migration and invasion and reduced GSC apoptosis

NRAS was previously identified as an oncogene in several cancers. NRAS protein levels in normal brain tissues, glioma tissues and GSCs were investigated by immunohistochemistry and Western blot. As shown in Figure 6A, NRAS localized to the cytoplasm and was upregulated in glioma tissues compared with normal brain tissues. Similarly, the Western blot revealed greater protein levels of NRAS in glioma tissues and GSCs than in normal brain tissues and non-GSCs (Figure 6B and 6C). Thus, we inferred that NRAS promotes the malignant progression of GSCs. NRAS has been reported to act as an oncogene in various tumor cells. To determine the effect of NRAS on GSCs, we divided cells into five groups: the control group, NRAS (+)-NC group (transfected with the empty plasmid), NRAS (+) group (transfected with the NRAS full-length plasmid), NRAS (-)-NC group (transfected with the empty plasmid) and NRAS (-) group, (transfected with the short-hairpin NRAS plasmid). CCK-8 assays revealed that GSC proliferation was enhanced in the NRAS overexpression group compared to the NRAS (+)-NC group (Figure 6D). Transwell assays demonstrated that GSC migration and invasion were greater in the NRAS overexpression group than in the NRAS (+)-NC group (Figure 6E). Further, overexpression of NRAS induced the migration of GSCs in a 3D Spheroid-based tumor migration assay (Figure 6F). NRAS overexpression also significantly reduced GSC apoptosis (Figure 6G).

NRAS was upregulated in glioma tissues, glioma cell lines and GSCs.

Figure 6: NRAS was upregulated in glioma tissues, glioma cell lines and GSCs. And NRAS promoted cell proliferation, migration and invasion and inhibited apoptosis of GSCs. A. Immunohistochemistry of NRAS protein levels in non-tumorous brain, Grade I-II glioma, and Grade III-IV glioma tissues. Original magnification: 100× (above), 200× (below). Scale bar, 50 μm. B. NRAS protein expression in non-tumorous brain tissues and glioma tissues, with GAPDH as an endogenous control. Representative protein expression and integrated light density values of NRAS in non-tumorous brain tissues, low-grade glioma tissues (WHO I-II), and high-grade glioma tissues (WHO III-IV) are shown. Data are presented as the mean ± SD (n=15, each group). *P<0.05 vs. non-tumorous brain tissue group. C. Western blot of NRAS expression in non-GSCs and GSCs, with GAPDH as an endogenous control. *P<0.05 vs. non-GSC group. D. The CCK-8 assay was employed to determine the effect of NRAS on GSC proliferation. E. Quantification of GSC migration and invasion upon NRAS inhibition. Representative images and accompanying statistical plots are presented. F. 3D Spheroid-based tumor migration assays of the effect of NRAS expression on GSC migration. Scale bars, 60 μm. G. Flow cytometry analysis of the effects of NRAS overexpression or downregulation on GSCs. Data are presented as the mean ± SD (n=5, each group). *P<0.05 vs. NRAS (+)-NC group; #P<0.05 vs. NRAS (-)-NC group. Scale bars, 20 μm.

NRAS was involved in the NEAT1/let-7e-dependent malignant progression of GSCs

Bioinformatics databases (Targetscan, Starbase and miRanda) predicted that several genes would be downstream targets of let-7e, including NRAS. We first determined the effect of let-7e on the protein levels of NRAS by Western blot, and found that NRAS expression was significantly downregulated, among several downstream molecules of let-7e predicted by the bioinformatics databases. Next, the mRNA and protein levels of NRAS were detected in GSCs treated with sh-NEAT1, pre-let-7e or anti-let-7e by qRT-PCR and Western blot. NRAS mRNA and protein levels were lower in the sh-NEAT1 group than in the sh-NC group (Figure 7A and 7E). On the contrary, GSCs treated with anti-let-7e exhibited higher mRNA and protein levels of NRAS than those treated with anti-NC (Figure 7B and 7F). NRAS expression was significantly lower in the sh-NEAT1+pre-let-7e group than in the sh-NC+pre-NC group (Figure 7G).

NRAS was a target gene of Let-7e.

Figure 7: NRAS was a target gene of Let-7e. And the expression of NRAS was regulated by both NEAT1 and let-7e. A. qRT-PCR analysis of the effect of NEAT1 on NRAS mRNA expression in GSCs. *P<0.05 vs. sh-NC group. B. qRT-PCR analysis of the effect of let-7e on NRAS mRNA expression in GSCs. *P<0.05 vs. pre-NC group; #P<0.05 vs. anti-NC group. C. NRAS harbored one putative let-7e binding site, the designed mutant sequence is indicated. D. Dual-luciferase reporter assay of HEK 293T cells co-transfected with NRAS-Wt and let-7e-NC; NRAS-Wt and let-7e; NRAS-Mut and let-7e-NC; or NRAS-Mut and let-7e. *P<0.05 vs. NRAS-Wt+let-7e-NC group. E. Western blot analysis for NRAS in NEAT1-knockdown GSCs, with GAPDH as an endogenous control. *P<0.05 vs. sh-NC group. F. Western blot analysis for NRAS in let-7e-overexpression and let-7e-inhibition GSCs, with GAPDH as an endogenous control. *P<0.05 vs. pre-NC group; #P<0.05 vs. anti-NC group. G. Western blot analysis for NRAS in GSCs co-transfected with sh-NEAT1 and pre-let-7e or anti-let-7e, with GAPDH as an endogenous control. *P<0.05 vs. sh-NC+pre-NC group. H. Western blot analysis for NRAS in let-7e-Mut-overexpression or let-7e-Mut-inhibition GSCs, with GAPDH as an endogenous control. For G-K, data are presented as the mean ± SD (n=5, each group).

Having confirmed that both NEAT1 and let-7e influence the malignant behavior of GSCs, we performed a dual-luciferase reporter assay to verify the putative binding site (Figure 6C) between let-7e and NRAS. Luciferase activity was lower in the NRAS-Wt+let-7e group than in the NRAS-Wt+let-7e-NC group, but did not differ significantly between the NRAS-Mut+let-7e and NRAS-Mut+let-7e-NC groups (Figure 7D).

To further confirm the binding sites between let-7e and NRAS, we mutated the expected binding sequence in let-7e. As shown in Figure 7H, NRAS protein levels did not change in the pre-let-7e-Mut group than in the pre-NC group.

Let-7e impaired NRAS-induced malignant behavior in GSCs by binding to its 3′-UTR

To discover whether let-7e prevented GSC malignant evolution by binding to a specific sequence in NRAS, we investigated the extent of proliferation, migration, invasion and apoptosis in GSCs stably expressing let-7e+NRAS (non-3′UTR). We divided cells into four groups: the let-7e-NC+NRAS-NC group (cells stably expressing pre-NC, co-transfected with the NRAS-NC plasmid), let-7e+NRAS-NC group (cells stably expressing pre-let-7e, co-transfected with NRAS-NC), let-7e+NRAS group (cells stably expressing pre-let-7e, co-transfected with NRAS (+)) and let-7e+NRAS (non-3′UTR) group (cells stably expressing let-7e, co-transfected with NRAS (without 3′-UTR) plasmid).

The proliferation of GSCs was significantly greater in the let-7e+NRAS (non-3′UTR) group than in the let-7e+NRAS group (Figure 8A). Further, in Transwell assays, there were significantly more migrating and invading GSCs in the let-7e+NRAS (non-3′UTR) group than in the let-7e+NRAS group (Figure 8B). Further, GSCs treated with let-7e+NRAS (non-3′UTR) had stronger migration abilities (Figure 8C) and a lower extent of apoptosis (Figure 8D) than those treated with let-7e+NRAS.

Let-7e inhibited GSC malignant progression by binding to the NRAS 3&#x2032;-UTR.

Figure 8: Let-7e inhibited GSC malignant progression by binding to the NRAS 3′-UTR. A. The CCK-8 assay was used to evaluate the effects of let-7e and NRAS (+) on U87 and U251 cell proliferation. B. Quantification of cell migration and invasion in groups according to let-7e and NRAS (+)-NC expression. Representative images and accompanying statistical plots are presented. C. Migration abilities of GSCs in groups according to let-7e and NRAS (+) expression. Scale bars, 60 μm. D. Flow cytometry analysis of GSCs in groups according to let-7e and NRAS (+) expression. Data are presented as the mean ± SD (n=5, each group). *P<0.05 vs. let-7e+NRAS group. Scale bars, 40 μm.

DISCUSSION

In this study, we demonstrated that NEAT1 was upregulated in GSCs. NEAT1 inhibition impaired the malignant behavior of GSCs and attenuated NRAS expression. On the contrary, let-7e expression was downregulated in glioma tissues and GSCs. Restoration of let-7e suppressed proliferation, migration and invasion, promoted apoptosis, and reduced NRAS expression in GSCs. Moreover, let-7e was found to bind to NEAT1 in a sequence-specific manner, and there was reciprocal repression between let-7e and NEAT1, possibly induced by the RISC. NRAS was identified as a direct target of let-7e and was involved in the NEAT1-induced malignant progression of GSCs. Further, NRAS was confirmed to promote oncogenesis in GSCs by stimulating cell proliferation, migration and invasion while inhibiting apoptosis.

There is much evidence that lncRNAs are aberrantly expressed in various tumors. Due to their involvement in tumorigenesis and cancer progression, lncRNAs could be used as diagnostic or prognostic biomarkers and potential therapeutic targets. NEAT1 is an lncRNA confirmed to be upregulated in gliomas, and promotes cell proliferation, migration and invasion while suppressing apoptosis in glioma U87 and U251 cells [10].

GSCs are a subgroup of glioma cells characterized by self-renewal, promotion of angiogenesis and multi-differentiation. Conventional therapies against glioma may be limited mainly due to the existence of GSCs, which facilitate the recurrence, drug-resistance, rapid growth, invasion and metastasis of glioma [2]. Therefore, therapy against GSCs has become an urgent and promising field. In the current study, we found that NEAT1 was upregulated in GSCs, suggesting that it might promote GSC progression. Indeed, inhibition of NEAT1 reduced the malignant progression of GSCs. Consistent with our results, NEAT1 was reported to be upregulated in non-small cell lung cancers and to promote lung cancer cell growth [26]. Likewise, the overexpression of NEAT1 contributes to the malignant behavior of laryngeal squamous cell cancer by downregulating the miR-107/CDK6 pathway [27]. We further examined the expression of NRAS, which promotes oncogenesis in various cancers, and found that NEAT1 knockdown significantly reduced NRAS expression in GSCs. However, whether NRAS is also involved in the NEAT1-induced enhancement of GSC progression needs to be further investigated.

We also found that let-7e, a conserved miRNA, was downregulated in glioma tissues and GSCs, which suggested that let-7e might be associated with GSC progression. As reported earlier, low expression of let-7e may be a biomarker of non-small cell lung cancer [28]. Also, let-7e expression is significantly reduced in retinoblastoma [29], and low let-7e expression contributes to cisplatin resistance in epithelial ovarian cancer [30]. To explore the function of diminished let-7e expression in GSCs, we studied the effects of restoring or inhibiting of let-7e expression in GSCs. Our results indicated that overexpression of let-7e suppressed the proliferation, migration and invasion and promoted the apoptosis of GSCs, whereas inhibition of let-7e facilitated the malignant progression of GSCs. In addition, NRAS expression was reduced in GSCs that overexpressed let-7e.

LncRNAs have been confirmed to function as endogenous miRNA sponges by binding to miRNAs and restraining their function [31]. We used bioinformatics software (Starbase) to determine whether NEAT1 harbors a let-7e binding site, and identified two putative binding sites (Figure 5B). Additional experiments revealed that let-7e expression was significantly rescued when NEAT1 was inhibited. Let-7e overexpression reduced the expression of NEAT1, while let-7e inhibition increased NEAT1 expression. This suggested that NEAT1 and let-7e participate in a reciprocal repression feedback loop. Dual-luciferase reporter assays verified that let-7e binds to NEAT1 in a sequence-specific manner. Further, an RIP experiment confirmed the involvement of the RISC complex in the reciprocal repression between NEAT1 and let-7e. This finding was consistent with a previous report that XIST functions as tumor suppressor by upregulating miR-152 in a RISC-dependent manner, and harbors a miR-152 binding site that allows XIST function as a ceRNA of miR-152 in GBM stem cells [32]. HOTAIR is confirmed to promote the malignancy of renal carcinoma cells, and to stimulate oncogenesis by suppressing miR-141 in the RISC complex [33].

To validate our hypothesis that NEAT1 promoted the malignant progression of GSCs by suppressing let-7e, we established GSCs in which NEAT1 was stably inhibited and let-7e was overexpressed or inhibited. The inhibition of let-7e in these cells largely reversed the suppression of GSC progression resulting from the knockdown of NEAT1. Furthermore, in NEAT1-knockdown GSCs, the inhibition of let-7e restored NRAS expression. These results indicated that NEAT1 promotes oncogenesis by downregulating let-7e in GSCs.

MiRNAs regulate cellular events by binding to the 3′-UTRs of downstream genes. For instance, miR-186 inhibits GSC growth by binding to the 3′-UTRs of XIAP and PAK7[34]. As our bioinformatics analysis and dual-luciferase assay demonstrated, NRAS is a direct target of let-7e and promotes malignant behavior in GSCs. For over four decades, scientists have searched for a safe and effective method of hindering the aberrant activity of RAS in human cancers [22]. NRAS belongs to the RAS gene family, and its encoded protein is essential for controlling the activity of several cellular signaling pathways, including PI3K/AKT/mTOR and MEK/ERK [3537]. It is well known that the PI3K/AKT/mTOR and MEK/ERK pathways can directly and effectively promote cancer cell proliferation, migration and invasion, while inhibiting the apoptosis of tumor cells [3841]. NRAS has been confirmed to activate these two important pathways, suggesting that therapies targeting NRAS may be promising anticancer treatment methods. Moreover, high-frequency NRAS mutations occur in various tumors, such as melanoma, juvenile myelomonocytic leukemia and colorectal cancer [42, 43]. NRAS mutations contribute to the response to cetuximab treatment in colorectal cancer cells by reducing patients survival [43]. Localized immunotherapies exhibit enhanced activity in high-grade melanoma and may be especially effective in those with NRAS mutations [44]. Further, NRAS protein levels are dysregulated in several tumor cells. NRAS is overexpressed in lung cancer cells and can be directly downregulated by miR-515-5p [45]. MiR-421 can inhibit prostate cancer progression by attenuating NRAS protein expression [46]. Dual-luciferase reporter assays confirmed our hypothesis that let-7e could bind to NRAS in a sequence-specific manner, and NRAS expression was inversely affected by the overexpression or downregulation of let-7e in GSCs. Importantly, the NRAS 3′-UTR reversed the let-7e-induced impairment of malignant behavior in GSCs. Whether NRAS protein is a viable clinical therapy target for patients with glioma remains to be investigated.

In conclusion, we have shown that the knockdown of NEAT1 impaired malignant behavior in GSCs by upregulating let-7e expression. Moreover, let-7e suppressed GSC malignant behavior by binding to NRAS. The significance of the correlation among NEAT1,let-7e and NRAS expression was highlighted for the first time. Thus, therapies targeting the NEAT1/let-7e/NRAS axis may be promising options for the treatment of human glioma.

MATERIALS AND METHODS

Clinical tissues

All glioma tissues and normal brain tissues (NBTs) were collected from surgical resections of human glioma in the Department of Neurosurgery, Shengjing Hospital of China Medical of University, from January 2013 to September 2015. Informed consent was gathered from all patients, and the research method was approved by the Ethics Committee of Shengjing Hospital of China Medical University. Grades of glioma were assigned by neuropathologists according to World Health Organization (WHO) classification. These tissues were divided into four groups: grade I (n=30), grade II (n=30), grade III (n=30) and grade IV (n=30). NBTs were used as the negative control group (n=30), and were obtained from patients who had been in traffic accidents that required immediate partial resections of brain tissue to reduce intracranial pressure, but unfortunately died after surgery. The tissues we obtained were far from the trauma tissue. All informed consent forms were obtained from relatives of the trauma victims. The specimens were immediately frozen and preserved in liquid nitrogen until use in this research.

Cell culture

Human glioma cell lines (U87, T98G, U251, A272 and U373) and human embryonic kidney (HEK) 293T cells were purchased from the Chinese Academy of Medical Sciences (Beijing, China). They were cultured in high-glucose Dulbecco’s Modified Eagle Medium (DMEM) with 10% fetal bovine serum (FBS, Gibco, Carlsbad, CA, USA). Primary normal human astrocytes were purchased from Sciencell Research Laboratories (Carlsbad, CA, USA) and cultured in astrocyte medium (Sciencell Research Laboratories). All cells were incubated at 37°C in a humidified incubator with 5% CO2.

Isolation and identification of GSCs

GSCs were obtained and isolated as described previously [47]. Briefly, GSCs were cultured in DMEM/F-12 medium (Life Technologies Corporation, Grand Island, NY, USA) supplemented with basic fibroblast growth factor (20 ng/mL, Life Technologies Corporation, Carlsbad, CA, USA), epidermal growth factor (20 ng/mL, Life Technologies Corporation, Gaithersburg, MD, USA) and 2% B27 (Life Technologies Corporation, Grand Island, NY, USA). As previously described, sphere cells were dissociated in 96-well plates for the limiting dilution assay and primary sphere formation assay [48, 49]. Sphere cells were plated onto glass coverslips coated with poly-L-ornithine (BD Biosciences, Franklin Lakes, NJ, USA) in medium containing 10% FBS for the differentiation assay. For immunostaining of undifferentiated spheres, cells were incubated with antibodies against Nestin and CD133 (also known as prominin-1) (1:100, Santa Cruz Biotechnology, Santa Cruz, CA, USA). For immunostaining of differentiated spheres, cells were stained with antibodies against GFAP (1:100, Abcam, Cambridge, MA, USA) and beta-tubulin III (1:100, Santa Cruz Biotechnology). The primary antibody complexes were visualized with anti-rabbit Alexa Fluor 488 and anti-mouse Alexa Fluor 555 secondary antibodies (Beyotime Institute of Biotechnology, Jiangsu, China). Nuclei were counterstained with 4’, 6-diamidino-2-phenylindole (DAPI).

Tumor xenografts

Four-week-old male nude mice were purchased from the National Laboratory Animal Center (Beijing, China). All mice were given free access to autoclaved food and water during the experiment. All experiments with nude mice were performed strictly in accordance with a protocol approved by the Administrative Panel on Laboratory Animal Care of the China Medical University. The mice were subcutaneously injected with 5 × 104 GSC-U87 (or GSC-U251) and non-GSC-U87 (or non-GSC-U251) cells (n = 6 in each group). The subcutaneous tumor-bearing mice were sacrificed six weeks after injection. The tumor volume was calculated by the formula: volume (mm3) = length × width2/2.

Cell transfections

The NEAT1 knockdown (sh-NEAT1) plasmid and the respective non-targeting sequence (negative control, sh-NC), as well as the let-7e agomir (pre-let-7e), let-7e antagomir (anti-let-7e) and their respective non-targeting sequences (negative controls: pre-NC and anti-NC) were synthesized (GenePharma, Shanghai, China). The NRAS full-length (with 3′-UTR) plasmid (NRAS (+)), short-hairpin NRAS plasmid (NRAS (-)), NRAS (without 3′-UTR) plasmid (NRAS (non-3′UTR)) and their respective non-targeting sequences (negative controls: NRAS (+)-NC and NRAS (-)-NC) were synthesized (Life technology, Carlsbad, CA, USA). Cells were transfected through the use of Opti-MEM and Lipofectamine 3000 (Life Technologies Corporation, Carlsbad, CA, USA) according to the manufacturer’s instructions when cells were at 50-70% confluence. The applicable stably transfected cell lines were established by selection with G418 screening. The transfection efficiency was verified by qRT-PCR.

RNA extraction and qRT-PCR

Total RNA was extracted from clinical specimens and cells with the Trizol reagent (Life Technologies Corporation, Carlsbad, CA, USA). The primers for NEAT1 and GAPDH were synthesized by Takara Bio (Japan). A One-Step SYBR PrimeScript RT-PCR Kit (Perfect Real Time) (Takara Bio) was used for qRT-PCR. The primers for NEAT1: forward 5′-ATGCCACAACGCAGATTGAT-3′, reverse 5′-CGAGAAACGCACAAGAAGG-3′; GAPDH: forward 5′-TGCACCACCAACTGCTTAGC-3′, reverse 5′-GGCATGCACTGTGGTCATGAG-3′. cDNA was generated from miRNA with a TaqMan miRNA Reverse Transcription kit (Applied Biosystems, Foster City, CA, USA). TaqMan Universal Master Mix II was used to perform TaqMan microRNA assays for let-7e and U6 (Applied Biosystems, Foster City, CA, USA) on the ABI 7500 Fast Real-Time PCR System (Applied Biosystems). GAPDH and U6 were used as endogenous controls for gene and miRNA expression, respectively. Gene and miRNA expression were normalized to those of the respective endogenous controls, and the fold-change in gene expression was calculated as 2−ΔΔCt.

Cell proliferation assay

After transfection efficacy was confirmed, GSCs were dissociated with Accutase (Life Technologies Corporation, Carlsbad, CA, USA), resuspended, and seeded in 96-well plates at 3000 cells per well. The CCK-8 assay (Dojin, Japan) was used to measure GSC proliferation. CCK-8 solution (10 μL) was added to each well and the plate was incubated for 2 h at 37°C. The absorbance was recorded at 450 nm on a SpectraMax M5 microplate reader (Molecular Devices, USA).

Cell migration and invasion assay

A 24-well insert with an 8-mm pore size (Corning, USA) was employed for the GSC migration and invasion assays. GSCs were dissociated with Accutase, resuspended in 100 μL serum-free medium and placed in the upper chamber (without or pre-coated with 500 ng/mL Matrigel solution (BD, Franklin Lakes, NJ, USA) for the migration and invasion assays, respectively), while 600 μL of 10% FBS medium was placed in the lower chamber. After incubation at 37°C for 48 h, the cells on the upper membrane surface were scraped off. The cells on the lower side of the member were fixed and then stained with 10% Giemsa. Cells were counted from five random vision fields under a microscope for statistics.

3D Spheroid-based tumor migration assays

For further analysis of the invasion abilities of GSCs, 3D Spheroid-based tumor migration assays were performed. Briefly, a Matrigel matrix (BD, State of New Jersey) was added to each well of a 96-well black, clear-bottom spheroid microplate (Corning, New York). The plate was then transferred to a 37°C/5% CO2 incubator for 1 h to initiate gel formation. GSCs were diluted to a concentration of 3×104/mL in medium, and 100 μL of this dilution was plated to each of the appropriate wells. After 48 h, cells were monitored from five random vision fields under a microscope for statistics.

Apoptosis detection

Apoptosis was measured with Annexin V-PE/7AAD staining (Southern Biotech, Birmingham, AL, USA) according to the manufacturer’s instructions. After being washed twice with 37°C PBS and stained with Annexin V-PE/7AAD, cells were analyzed by flow cytometry (FACScan, BD Biosciences), and apoptotic fractions were recorded with CELL Quest 3.0 software.

Dual-luciferase reporter assays

NEAT1 and NRAS 3′-UTR sequences were amplified by PCR and cloned into a pmirGlo Dual-luciferase miRNA Target Expression Vector (Promega, Madison, WI, USA) to construct luciferase reporter vectors (NEAT1-Wt and NRAS-Wt) (GenePharma). The theoretical let-7e binding sequences in NEAT1 and NRAS were mutated as indicated (NEAT1-Mut and NRAS-Mut). HEK-293T cells were co-transfected with the combinations of plasmids described below when they were at 50-70% confluence. A dual-luciferase reporter assay kit (Promega) was used to determine the luciferase activity 48 h after transfection. For the NEAT1 binding assay, the cells were divided into five groups: the control group, NEAT1-Wt+let-7e-NC group, NEAT1-Wt+let-7e group, NEAT1-Mut+let-7e-NC group, and NEAT1-Mut+let-7e group. Likewise, for the NRAS binding assay, the cells were divided into five groups: the control group, NRAS-Wt+let-7e-NC group, NRAS-Wt+let-7e group, NRAS-Mut+let-7e-NC group, and NRAS-Mut+let-7e group.

RNA immunoprecipitation

To investigate whether NEAT1 was associated with the RISC, we performed RNA immunoprecipitation. GSCs were lysed in complete RNA lysis buffer containing protease inhibitor and RNase inhibitor from an EZ-Magna RIP RNA-binding protein immunoprecipitation kit (Millipore, Billerica, MA, USA) according to the manufacturer’s protocol. Whole cell lysates from the control groups and let-7e groups were incubated with RIP immunoprecipitation buffer containing magnetic beads conjugated with human anti-Argonaute 2 antibody (Millipore) and the negative control (normal mouse IgG; Millipore). Samples were incubated with Proteinase K buffer, and then the immunoprecipitated RNA was isolated. The RNA concentration was measured with a NanoDrop (Thermo Scientific) and the RNA quality was assessed with a bioanalyser (Agilent, Santa Clara, CA, USA). Purified RNA was obtained, and qRT-PCR was performed with the primers mentioned above to demonstrate the presence of the binding targets.

Immunohistochemistry assays

Slides of specimens (4 μm thick) were dewaxed, rehydrated, and incubated in 0.3% H2O2 for 10 minutes to inhibit endogenous peroxidase activity. Slides were then blocked with 10% normal goat serum (MXB, Fuzhou, China) for 30 minutes and incubated overnight at 4°C with a rabbit polyclonal antibody against NRAS (1:50, Abcam, UK) and XIAP (1:50, Abcam, UK). Slides were washed with PBS three times and then incubated with biotinylated rabbit anti-rabbit IgG for 1 h at room temperature. After incubation with an avidin-biotin-peroxidase complex for 10 minutes, samples were stained with 3,3’-diaminobenzidine. Slides were imaged under a light microscope (Olympus, Japan) at 100× and 200× magnification.

Western blot

Total proteins were extracted from cells with RIPA buffer containing protease inhibitors (Beyotime Institute of Biotechnology) on ice, and these proteins were then subjected to SDS-PAGE and electrophoretically transferred to PVDF membranes. After non-specific binding was blocked with 5% nonfat milk at room temperature for 2 h, membranes were incubated with primary antibodies as follows: NRAS (1:2000, Abcam, UK) and GAPDH (1:1000, Santa Cruz Biotechnology). Then, the membranes were incubated with HRP-conjugated secondary antibodies (1:5000 goat anti-rabbit or goat anti-mouse, respectively; Santa Cruz Biotechnology) at room temperature for 2 h. Immunoblots were visualized by enhanced chemiluminescence (ECL kit, Santa Cruz Biotechnology) and recorded with ChemImager 5500 V2.03 software. The relative integrated density values were calculated with GAPDH as an internal control.

Statistical analysis

Data are presented as the mean ± standard deviation (SD). All experimental results were statistically analyzed with Student’s t-test or one-way analysis of variance (ANOVA). All statistical analyses were performed with SPSS 18.0 statistical software, with P<0.05 considered as statistically significant.

ACKNOWLEDGMENTS

This work is supported by grants from the Natural Science Foundation of China (81272564, 81372484 and 81573010), Liaoning Science and Technology Plan Project (No. 2015225007), and Shenyang Science and Technology Plan Projects (Nos. F15-199-1-30 and F15-199-1-57).

CONFLICTS OF INTEREST

The authors declare no conflict of interest.

REFERENCES

1. Thorne AH, Meisen WH, Russell L, Yoo JY, Bolyard CM, Lathia JD, Rich J, Puduvalli VK, Mao H, Yu J, Caligiuri MA, Tridandapani S and Kaur B. Role of cysteine-rich 61 protein (CCN1) in macrophage-mediated oncolytic herpes simplex virus clearance. Mol Ther. 2014; 22:1678-1687.

2. Visvader JE and Lindeman GJ. Cancer stem cells: current status and evolving complexities. Cell stem cell. 2012; 10:717-728.

3. Spina R, Voss DM, Asnaghi L, Sloan A and Bar EE. Atracurium Besylate and other neuromuscular blocking agents promote astroglial differentiation and deplete glioblastoma stem cells. Oncotarget. 2016; 7:459-472. doi: 10.18632/oncotarget.6314.

4. Liz J and Esteller M. lncRNAs and microRNAs with a role in cancer development. Biochimica et biophysica acta. 2016; 1859:169-176.

5. Wang JY, Liu XF, Wu HC, Ni PH, Gu ZD, Qiao YX, Chen N, Sun FY and Fan QS. CREB up-regulates long non-coding RNA, HULC expression through interaction with microRNA-372 in liver cancer. Nucleic Acids Res. 2010; 38:5366-5383.

6. Zhao X, Wang P, Liu J, Zheng J, Liu Y, Chen J and Xue Y. Gas5 Exerts Tumor-suppressive Functions in Human Glioma Cells by Targeting miR-222. Mol Ther. 2015.

7. Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, Chess A and Lawrence JB. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Molecular cell. 2009; 33:717-726.

8. Hutchinson JN, Ensminger AW, Clemson CM, Lynch CR, Lawrence JB and Chess A. A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains. BMC genomics. 2007; 8:39.

9. Fox AH and Lamond AI. Paraspeckles. Cold Spring Harbor perspectives in biology. 2010; 2:a000687.

10. Zhen L, Yun-Hui L, Hong-Yu D, Jun M and Yi-Long Y. Long noncoding RNA NEAT1 promotes glioma pathogenesis by regulating miR-449b-5p/c-Met axis. Tumour biology. 2015.

11. Guo S, Chen W, Luo Y, Ren F, Zhong T, Rong M, Dang Y, Feng Z and Chen G. Clinical implication of long non-coding RNA NEAT1 expression in hepatocellular carcinoma patients. International journal of clinical and experimental pathology. 2015; 8:5395-5402.

12. Chakravarty D, Sboner A, Nair SS, Giannopoulou E, Li R, Hennig S, Mosquera JM, Pauwels J, Park K, Kossai M, MacDonald TY, Fontugne J, Erho N, et al. The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nature communications. 2014; 5:5383.

13. Kim YS, Hwan JD, Bae S, Bae DH and Shick WA. Identification of differentially expressed genes using an annealing control primer system in stage III serous ovarian carcinoma. BMC cancer. 2010; 10:576.

14. Frixa T, Donzelli S and Blandino G. Oncogenic MicroRNAs: Key Players in Malignant Transformation. Cancers. 2015; 7:2466-2485.

15. Wang P, Liu YH, Yao YL, Li Z, Li ZQ, Ma J and Xue YX. Long non-coding RNA CASC2 suppresses malignancy in human gliomas by miR-21. Cellular signalling. 2015; 27:275-282.

16. Roush S and Slack FJ. The let-7 family of microRNAs. Trends in cell biology. 2008; 18:505-516.

17. Boyerinas B, Park SM, Hau A, Murmann AE and Peter ME. The role of let-7 in cell differentiation and cancer. Endocrine-related cancer. 2010; 17:F19-36.

18. Zhang YK, Zhu WY, He JY, Chen DD, Huang YY, Le HB and Liu XG. miRNAs expression profiling to distinguish lung squamous-cell carcinoma from adenocarcinoma subtypes. Journal of cancer research and clinical oncology. 2012; 138:1641-1650.

19. Wang Y, Hu X, Greshock J, Shen L, Yang X, Shao Z, Liang S, Tanyi JL, Sood AK and Zhang L. Genomic DNA copy-number alterations of the let-7 family in human cancers. PloS one. 2012; 7:e44399.

20. Kong L, Du X, Hu N, Li W, Wang W, Wei S, Zhuang H, Li X and Li C. Downregulation of let-7e-5p contributes to endothelial progenitor cell dysfunction in deep vein thrombosis via targeting FASLG. Thrombosis research. 2016; 138:30-36.

21. Ventayol M, Vinas JL, Sola A, Jung M, Brune B, Pi F, Mastora C and Hotter G. miRNA let-7e targeting MMP9 is involved in adipose-derived stem cell differentiation toward epithelia. Cell death & disease. 2014; 5:e1048.

22. Zhang F and Cheong JK. The renewed battle against RAS-mutant cancers. Cellular and molecular life sciences. 2016.

23. Wang T, Li C, Xia C, Dong Y, Yang D, Geng Y, Cai J, Zhang J, Zhang X and Wang J. Oncogenic NRAS hyper-activates multiple pathways in human cord blood stem/progenitor cells and promotes myelomonocytic proliferation in vivo. American journal of translational research. 2015; 7:1963-1973.

24. Zhang J, Zheng J, Yang Y, Lu J, Gao J, Lu T, Sun J, Jiang H, Zhu Y, Zheng Y, Liang Z and Liu T. Molecular spectrum of KRAS, NRAS, BRAF and PIK3CA mutations in Chinese colorectal cancer patients: analysis of 1,110 cases. Scientific reports. 2015; 5:18678.

25. Saldanha G, Potter L, Lee YS, Watson S, Shendge P and Pringle JH. MicroRNA-21 expression and its pathogenetic significance in cutaneous melanoma. Melanoma research. 2016; 26:21-28.

26. You J, Zhang Y, Liu B, Li Y, Fang N, Zu L, Li X and Zhou Q. MicroRNA-449a inhibits cell growth in lung cancer and regulates long noncoding RNA nuclear enriched abundant transcript 1. Indian journal of cancer. 2014; 51:e77-81.

27. Wang P, Wu T, Zhou H, Jin Q, He G, Yu H, Xuan L, Wang X, Tian L, Sun Y, Liu M and Qu L. Long noncoding RNA NEAT1 promotes laryngeal squamous cell cancer through regulating miR-107/CDK6 pathway. Journal of experimental & clinical cancer research. 2016; 35:22.

28. Zhu WY, Luo B, An JY, He JY, Chen DD, Xu LY, Huang YY, Liu XG, Le HB and Zhang YK. Differential expression of miR-125a-5p and let-7e predicts the progression and prognosis of non-small cell lung cancer. Cancer investigation. 2014; 32:394-401.

29. Liu SS, Wang YS, Sun YF, Miao LX, Wang J, Li YS, Liu HY and Liu QL. Plasma microRNA-320, microRNA-let-7e and microRNA-21 as novel potential biomarkers for the detection of retinoblastoma. Biomedical reports. 2014; 2:424-428.

30. Cai J, Yang C, Yang Q, Ding H, Jia J, Guo J, Wang J and Wang Z. Deregulation of let-7e in epithelial ovarian cancer promotes the development of resistance to cisplatin. Oncogenesis. 2013; 2:e75.

31. Zhou M, Wang X, Shi H, Cheng L, Wang Z, Zhao H, Yang L and Sun J. Characterization of long non-coding RNA-associated ceRNA network to reveal potential prognostic lncRNA biomarkers in human ovarian cancer. Oncotarget. 2016; 7:12598-611. doi: 10.18632/oncotarget.7181.

32. Yao Y, Ma J, Xue Y, Wang P, Li Z, Liu J, Chen L, Xi Z, Teng H, Wang Z, Li Z and Liu Y. Knockdown of long non-coding RNA XIST exerts tumor-suppressive functions in human glioblastoma stem cells by up-regulating miR-152. Cancer Lett. 2015; 359:75-86.

33. Chiyomaru T, Fukuhara S, Saini S, Majid S, Deng G, Shahryari V, Chang I, Tanaka Y, Enokida H, Nakagawa M, Dahiya R and Yamamura S. Long non-coding RNA HOTAIR is targeted and regulated by miR-141 in human cancer cells. J Biol Chem. 2014; 289:12550-12565.

34. Zheng J, Li XD, Wang P, Liu XB, Xue YX, Hu Y, Li Z, Li ZQ, Wang ZH and Liu YH. CRNDE affects the malignant biological characteristics of human glioma stem cells by negatively regulating miR-186. Oncotarget. 2015; 6:25339-25355. doi: 10.18632/oncotarget.4509.

35. Gyorffy B and Schafer R. Biomarkers downstream of RAS: a search for robust transcriptional targets. Current cancer drug targets. 2010; 10:858-868.

36. Orouji E, Orouji A, Gaiser T, Larribere L, Gebhardt C and Utikal J. MAP kinase pathway gene copy alterations in NRAS/BRAF wild-type advanced melanoma. International journal of cancer Journal international du cancer. 2016; 138:2257-2262.

37. Vieira GC, Chockalingam S, Melegh Z, Greenhough A, Malik S, Szemes M, Park JH, Kaidi A, Zhou L, Catchpoole D, Morgan R, Bates DO, Gabb PD and Malik K. LGR5 regulates pro-survival MEK/ERK and proliferative Wnt/beta-catenin signalling in neuroblastoma. Oncotarget. 2015; 6:40053-40067. doi: 10.18632/oncotarget.5548.

38. Bartholomeusz C and Gonzalez-Angulo AM. Targeting the PI3K signaling pathway in cancer therapy. Expert opinion on therapeutic targets. 2012; 16:121-130.

39. Park S, Chapuis N, Tamburini J, Bardet V, Cornillet-Lefebvre P, Willems L, Green A, Mayeux P, Lacombe C and Bouscary D. Role of the PI3K/AKT and mTOR signaling pathways in acute myeloid leukemia. Haematologica. 2010; 95:819-828.

40. Paternot S and Roger PP. Combined inhibition of MEK and mammalian target of rapamycin abolishes phosphorylation of cyclin-dependent kinase 4 in glioblastoma cell lines and prevents their proliferation. Cancer research. 2009; 69:4577-4581.

41. Stuart DD and Sellers WR. Targeting RAF-MEK-ERK kinase-scaffold interactions in cancer. Nature medicine. 2013; 19:538-540.

42. Johnson DB and Puzanov I. Treatment of NRAS-mutant melanoma. Current treatment options in oncology. 2015; 16:15.

43. Hsu HC, Thiam TK, Lu YJ, Yeh CY, Tsai WS, You JF, Hung HY, Tsai CN, Hsu A, Chen HC, Chen SJ and Yang TS. Mutations of KRAS/NRAS/BRAF predict cetuximab resistance in metastatic colorectal cancer patients. Oncotarget. 2016; 7:22257-70. doi: 10.18632/oncotarget.8076.

44. Johnson DB, Lovly CM, Flavin M, Panageas KS, Ayers GD, Zhao Z, Iams WT, Colgan M, DeNoble S, Terry CR, Berry EG, Iafrate AJ, Sullivan RJ, Carvajal RD and Sosman JA. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer immunology research. 2015; 3:288-295.

45. Pardo OE, Castellano L, Munro CE, Hu YL, Mauri F, Krell J, Lara R, Pinho FG, Choudhury T, Frampton AE, Pellegrino L, Pshezhetskiy D, Wang YL, Waxman J, Seckl MJ and Stebbing J. miR-515-5p controls cancer cell migration through MARK4 regulation. Embo Rep. 2016; 17:570-584.

46. Meng D, Yang S, Wan X, Zhang Y, Huang W, Zhao P, Li T, Wang L, Huang Y, Li T and Li Y. A transcriptional target of androgen receptor, miR-421 regulates proliferation and metabolism of prostate cancer cells. The international journal of biochemistry & cell biology. 2016; 73:30-40.

47. Zheng J, Liu X, Wang P, Xue Y, Ma J, Qu C and Liu Y. CRNDE promotes malignant progression of glioma by attenuating miR-384/PIWIL4/STAT3 axis. Mol Ther. 2016.

48. Yao Y, Ma J, Xue Y, Wang P, Li Z, Li Z, Hu Y, Shang X and Liu Y. MiR-449a exerts tumor-suppressive functions in human glioblastoma by targeting Myc-associated zinc-finger protein. Molecular oncology. 2015; 9:640-656.

49. Ciceroni C, Bonelli M, Mastrantoni E, Niccolini C, Laurenza M, Larocca LM, Pallini R, Traficante A, Spinsanti P, Ricci-Vitiani L, Arcella A, De Maria R, Nicoletti F, Battaglia G and Melchiorri D. Type-3 metabotropic glutamate receptors regulate chemoresistance in glioma stem cells, and their levels are inversely related to survival in patients with malignant gliomas. Cell death and differentiation. 2013; 20:396-407.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11403