Open Access

lncRNA SNHG3 promotes breast cancer progression by acting as a miR‑326 sponge

Retraction in: /10.3892/or.2022.8255

  • Authors:
    • Haipeng Zhang
    • Na Wei
    • Wei Zhang
    • Lishennan Shen
    • Rongbo Ding
    • Qian Li
    • Simin Li
    • Ye Du
  • View Affiliations

  • Published online on: July 15, 2020     https://doi.org/10.3892/or.2020.7690
  • Pages: 1502-1510
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Accumulating evidence suggests that long noncoding RNA (lncRNA) small nucleolar RNA host gene 3 (SHNG3) plays crucial roles in the initiation and progression of various types of malignant cancers. Yet, the role played by SNHG3 in breast cancer as well as the associated mechanisms remain largely unclear. The expression of SNHG3 was detected in breast cancer tissues and cell lines by reverse‑transcription quantitative PCR (RT‑qPCR). Cell proliferation, colony formation, cell cycle distribution, migration and invasion abilities were detected by Cell Counting Kit‑8, colony formation assay, flow cytometry, wound‑healing and Matrigel invasion assays, respectively. The regulatory relationships between SNHG3 and miR‑326 were explored by luciferase reporter assay. A nude mouse model was established to investigate the effect of SNHG3 in vivo. The results showed an upregulation of SNHG3 in breast cancer tissues and cell lines. Loss‑of‑function assays revealed significant suppression of breast cancer behaviors such as: Abilities to proliferate, form colonies, migrate and invade in vitro coupled with a delayed growth of tumors in vivo when SNHG3 was knocked down. Mechanically, it was shown that SNHG3 served as a competing endogenous RNA (ceRNA) of miR‑326 that in turn is a tumor suppressor in this cancer. The correlation between the expression of SNHG3 and miR‑326 was found to be strongly negative in these samples. Additionally, we found that inhibition of SNHG3 caused a partially reversal in the inhibition exerted by miR‑326 on the ability of these cells to proliferate, form colonies, migrate and invade. Collectively, these findings suggest the functioning of SNHG3 as a ceRNA to enhance the ability of breast cancer cells to proliferate and metastasize to putatively serve as a new target to explore therapeutic intervention of this malignancy.

Introduction

A ubiquitous form of malignancy associated with the largest number of fatalities among women is breast cancer (1). The health burden of breast cancer is increasing in China, with more than 1.6 million individuals being diagnosed and 1.2 million mortalities each year (2). Breast cancer frequently causes multi-organ distant metastasis such as lung, bone and brain (3). Although the disease prognosis has been prolonged by improvements in radical surgery along with adjuvant therapy, the overall survival rate of patients with advanced breast cancer remains poor mainly due to recurrence and metastasis (3). Therefore, identifying the molecular mechanisms associated with breast cancer progression is of great importance in order to identify novel diagnostic and therapeutic targets for patients with breast cancer.

Long non-coding RNAs (lncRNAs) are a class of noncoding RNAs longer than 200 nucleotides that do not possess a significant ability to code for proteins (4,5). Reports indicate the involvement of lncRNAs in several processes of physiology and pathology (68). It has been shown that lncRNAs act as oncogenes or tumor suppressor genes to control the ability of cells to proliferate, differentiate, invade and migrate as well as their apoptosis (9,10). An increasing number of lncRNAs have been reported to be involved vitally in tumorigenesis and breast cancer progression (11,12). For example, Zhang et al revealed that lncRNA MEG3 inhibits breast cancer progression partially via the activation of the endoplasmic reticulum (ER) stress, NF-κB and p53 pathways (13). Qiao et al revealed that TALNEC2 functions as a breast cancer oncogene in order to target p57KIP2 by binding to EZH2 via the p-p38 MAPK and NF-κB pathways (14). Kong et al found that lncRNA-CDC6 acts as a competing endogenous RNA (ceRNA) that sponges miR-215 to single out CDC6 resulting in enhancement of breast cancer progression and stage (15). These studies suggest that lncRNAs could be used in breast cancer as diagnostic markers or as targets in therapeutic approaches.

Small nucleolar RNA host gene 3 (SHNG3), a recently reported lncRNA, has been implicated in the vital aspects of the origin and progression of several types of human malignancies, including lung cancer (16), colorectal cancer (17), hepatocellular carcinoma (18), ovarian cancer (19) and glioma (20). A recent study demonstrated that SNHG3 expression is upregulated in breast cancer tissues and cell lines (21), yet the functioning and associated mechanism of SHNG3 in breast cancer remains to be fully characterized. The present study involved the assessment of SHNG3 expression in breast cancer tissues as well as cell lines. The functioning of this lncRNA in the ability of tumor cells to proliferate, migrate and invade as well as a putative mechanism was investigated in breast cancer. The results demonstrated that SHNG3 promoted breast cancer progression by sponging miR-326, which may offer novel targets for breast cancer therapy.

Materials and methods

Breast cancer samples

The breast tissues and corresponding adjacent normal breast tissues (the samples were collected such that there was a minimum 2-cm distance between the tumor edge and healthy tissue) were harvested from 48 cases of patients with breast cancer who underwent surgery at the First Hospital of Jilin University (Changchun, China) between 2016 and 2017. The patients were 30–62 years of age (mean, 46±4.18) and did not receive any form of treatment (radiotherapy, chemotherapy or any other treatment) prior to surgery. The specimens (tumor and adjacent healthy tissues) from surgery were subjected to instant freezing in liquid nitrogen, and stored at −80°C until RNA was extracted. All the subjects yielded written informed consent while the study received approval from the Ethics Committees of our The First Hospital of Jilin University (Changchun, Jilin, China).

Cell culture and transfection

American Type Culture Collection (ATCC) was the source of 4 breast cancer lines: MCF-7, MDA-MB-231, MDA-MB-468 and BT-474 as well as a healthy epithelial cell line of the breast called MCF-10A. Cell culture involved the use of RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) plus 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) supplemented with U/ml penicillin plus 0.1 mg/ml streptomycin in a 5% CO2 atmosphere at 37°C.

Synthesis of a short hairpin (sh)RNA called sh-SNHG3 that targets SNHG3 and the scramble negative control (sh-NC) were designed followed by cloning in pGreenPuro™ Vector (System Biosciences), followed by transfection into MCF-7 cells with Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) in adherence to the prescribed procedure. Puromycin (1 µg/ml) was used to select the cells that showed stable sh-SNHG3 and sh-NC transfection. miR-326 mimic plus the control (miR-NC), the miR-326 inhibitor with corresponding negative control mimics (anti-miR-NC) were obtained from GenePharma Co., Ltd., followed by transfection into MCF-7 cells with Lipofectamine 2000 in accordance to the prescribed procedures.

Extraction of RNA and quantitative reverse transcription polymerase chain reaction (RT-qPCR)

TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was utilized to isolate total RNA from all the samples and cell cultures followed by purification using an RNeasy Maxi kit (Qiagen). TaqMan MicroRNA assay Kit (Thermo Fisher Scientific, Inc.) was employed to check the expression of miR-326 under an ABI 7900 real-time PCR system (Thermo Fisher Scientific, Inc.) in accordance to the prescribed protocols. Total RNA (1 µg) was reverse transcribed into cDNA with PrimeScript™ RT Reagent Kit (Takara Bio Technology Co., Ltd.) to detect the mRNA of SNHG3 that was subject to amplification with SYBR Premix Ex Taq II (Takara Bio Technology Co., Ltd.) in the system mentioned above. The primers utilized have been elucidated by previous publications (16,22). The endogenous control for miR-326 was U6 while it was GAPDH for SNHG3. The 2−ΔΔCq method was utilized to calculate relative expression levels by ABI 7500 software v3.2 (v3.2; Applied Biosystems) (23).

Examining cell proliferation

Cell Counting Kit-8 (CCK-8; Dojindo Laboratories) was utilized to assess the ability of cells to proliferate in adherence with the prescribed procedure. Briefly, 96-well plates were seeded with 5×103 cells/well followed by culture for 24–72 h. Addition of 10 µl of CCK-8 reagent was carried out/well at days 1, 2 and 3 respectively, followed by a 4-h incubation at 37°C. A Benchmark Plus microplate spectrometer (Bio-Rad Laboratories) was utilized to record the absorbance (450 nm).

Clonogenic assay

Six-well plates were seeded with sh-SNHG3 or sh-NC-stably transfected MCF-7 cells (1,000 cells in total) in the medium described earlier for 2 weeks. Following this interval, cells were subjected to fixation using 96% ethanol for 30 min followed by crystal violet (1%) staining for 5 min at 37°C. The colonies were manually imaged and counted at in an inverted microscope (magnification ×10, Olympus Corp.). Colonyforming efficiency was calculated using the following equation: Visible cell colonies of experiment group/Visible cell Colonies of experiment group ×100 (%).

Cell cycle assay

Cell cycle analysis was determined using a cell cycle detection kit (Beyotime Institution of Biotechnology) in adherence to the prescribed procedure. Cell cycle distribution was determined under a Beckman-Coulter FC 500 MCL flow cytometer (Beckman Coulter, Inc.) using MultiCycle for Windows 32-bit software (Beckman Coulter, Inc.).

Assay for wound healing assay

The association of SNHG3 and the ability of cells to migrate were assessed by a wound healing assay. Briefly, 6-well plates were seeded with 2×105 transfected cells/well in the indicated medium with FBS to reach 100% confluence. This confluent monolayer was scratched and incubated in medium minus FBS for 24 h. Wounds were observed at time 0 and 24 h post scratching using a IX71 Olympus light microscope (magnification ×4; Olympus). The migration index was analyzed using ImageJ (FIJI distribution, version 1.52n; National Institutes of Health).

Transwell invasion assay

BD BioCoat™ Matrigel invasion chambers (Becton-Dickinson Labware) were used to study the association of SNHG3 and the ability of cells to invade in adherence to prescribed protocols. Briefly, the upper chamber with Matrigel (BD Biosciences) precoating of the aforementioned system was seeded with transfected cells in medium minus serum while the lower chamber was coated with medium that had serum (10%). Following a 48-h incubation at 37°C, the cells that invaded the lower chambers were subjected to 4% paraformaldehyde fixation and crystal violet (1%) staining for 5 min at 37°C. A Nikon phase-contrast microscope (magnification ×200) was used to observe and enumerate the cells across more than 5 fields.

Luciferase activity assay

The putative miRNAs that target SNHG3 were identified using Starbase 2.0 software (http://www.sysu.edu.cn/). Synthesis of miRNA binding sites: Wild-type (WT) or mutant (MT) was carried out followed by insertion into a psiCHECK™-2 luciferase reporter vector (Promega Corp.), represented as WT-SNHG3 and MT-SNHG3. For the luciferase assay, 5×103 cells were transfected with the WT-SNHG3 or MT-SNHG3 reporter vector and the miR-326 mimic or miR-NC in a 24-well plate with Lipofectamine 2000 in adherence to the prescribed procedure. The relative activity of luciferase was determined 48 h with a dual assay post transfection. The activity of Renilla luciferase was subject to normalization against that of firefly.

Tumor growth in vivo

The animal experiments were approved by the Animal Care and Use Committee of Jilin University (Grant no: JL2018426). The SLAC Animal Laboratory Center of this University was the source of 4–6 week old female BALB nude mice (18–20 g; n=10) that were bred in standard mouse irradiated food and tap water ad libitum, and maintained under conditions of 25°C and 50% humidity with a 12-h light/dark cycle. All mice were handled according to the requirements of the National Institutes of Health guidelines for care and use of laboratory animals. Animal health and behavior were monitored every day. Then, the left abdominal wall of the mice received subcutaneous injection of 2×106 stable SNHG3-depleted MCF-7 cells or sh-NC-MCF-7 cells, respectively (n=5). Measurement of the length (L) and width (W) of the tumor every fifth day, 10 days post injection was performed in order to calculate the size of the tumors. The tumor volume (V) was quantified using the expression: V=0.5 × width2 length. If tumor burden was >10% of the body weight in each mouse, the longest tumor diameter exceeded 2 cm, or tumors became ulcerated, necrotic or infected, euthanasia was used to halt the experiments. After 35 days, all mice were anesthetized by intraperitoneal injection with 10% chloral hydrate (300 mg/kg), and then euthanized by intraperitoneal injection of 200 mg/kg pentobarbital sodium (SigmaAldrich; Merck KGaA). The tumors were excised and weighed after the heartbeat and respiratory arrest of the mice. All mice did not exhibit multiple subcutaneous tumors before they were sacrificed. The diameters of the length and width of all tumor were <2 cm. A part of each tissue was sorted to detect SNHG3 and miR-326 expression by RT-qPCR. Moreover, other tumor portions were subjected to neutral formalin-fixation and paraffin-embedding for immunohistochemistry (IHC) analysis of Ki-67 as described previously (24).

Statistical analyses

Data are shown in the form of mean ± standard deviation (SD). All experiments were repeated at least thrice. SPSS v. 19.0 (IBM Corp., USA) was utilized for all analyses. Comparisons between two groups were conducted by unpaired or paired Student's t-test. One-way analysis of variance followed by the Tukey's post hoc test was utilized for for all the analyses involving three groups. Differences among 4 groups were assessed using mixed ANOVA or two-way ANOVA followed by Bonferroni test. Pearson's correlation analysis was used to analyze the correlation of SNHG3 and miR-326 in breast cancer tissues. In all cases, P-value <0.05 was considered statistically significant.

Results

Overexpression of lncRNA SNHG3 in breast cancer samples and cell lines

RT-qPCR was utilized to examine the expression of SNHG3 in the cases of breast cancer using the tissues described above. SNHG3 was significantly elevated in the cancer samples in comparison to that noted in the adjacent healthy ones (Fig. 1A). A similar trend was observed in the case of the cancer cell lines listed in this work in comparison to the control cell line MCF-10A (Fig. 1B). The observations are suggestive of the role of lncRNA as an oncogene in breast cancer progression.

Knockdown of SNHG3 inhibits cell proliferation and colony formation of breast cancer cells

As a next step to classify SNHG3 functioning, we downregulated SNHG3 by sh-SNHG3 transfection. qRT-PCR was used as a confirmation of the efficiency of transfection (Fig. 2A). Cell proliferation was inhibited to a significant extent as shown by CCK-8 assay when SNHG3 was downregulated in the MCF-7 cells when compared to the sh-NC group (Fig. 2B). The ability of sh-SNHG3-transfected MCF-7 cells to form colonies showed a significant reduction in comparison to this ability in the sh-NC-transfected MCF-7 cells (Fig. 2C). Moreover, flow cytometry revealed that knockdown of SNHG3 in MCF-7 cells significantly increased cell cycle arrest at the G1 stage while that at S phase was significantly decreased (Fig. 2D).

Knockdown of SNHG3 inhibits cell migration and invasion of breast cancer cells

Next, we sought an understanding of the link between SNHG3 and the ability of MCF-7 cells to invade and migrate by assays for wound healing and Transwell invasion, respectively. SNHG3-knockdown resulted in significant suppression in the ability of these cells to both invade and migrate (Fig. 3A and B).

miR-326 is a target of SNHG3

It is well known that lncRNAs may serve as sponges to modulate the expression and activity of miRNAs (25,26). To investigate whether the expression of SNHG3 is regulated by miRNA, a target prediction tool Starbase 2.0 was utilized in order to assess putative miRNAs that interact with SNHG3. This tool demonstrated that SNHG3 possessed a putative miR-326 binding site (Fig. 4A). The luciferase reporter assays further revealed that miR-326 expression caused a significant decrease in enzyme activity of WT-SNHG3 3′-UTR that was not observed in the case of the MT-SNHG3-3′-UTR (Fig. 4B), suggesting that miR-326 directly targets SNHG3. Furthermore, it was shown that overexpression of miR-326 significantly suppressed SNHG3 expression in MCF-7 cells (Fig. 4C), while SNHG3- knockdown significantly increased miR-326 expression in the MCF-7 cells (Fig. 4D). Moreover, it was found that the expression of miR-326 was negatively correlated with SNHG3 in the breast cancer tissues (r=−0.489, P=0.001; Fig. 4E).

miR-326 inhibition abolishes SNHG3-knockdown-mediated suppression of cell proliferation, colony formation, cell cycle arrest and migration and invasion abilities

The role of miR-326 as a downstream regulator in the inhibition of the ability of MCF-7 to proliferate, migrate and invade was examined more in detail by miR-326 inhibitor transfection of these cells. RT-qPCR assay revealed that SNHG3 knockdown significantly increased miR-326 expression when compared to the sh-NC group, but simultaneous use of the miR-326 inhibitor caused a partial reversal of the miR-326 upregulation caused by depletion of SNHG3 (Fig. 5A). Additionally, the inhibition of the ability of MCF-7 to proliferate, form colonies, migrate and invade along with cell cycle distribution by SNHG3 knockdown was significantly reversed by inhibiting miR-326 (all P<0.05, Fig. 5B-F). To summarize, overall these data are indicative of inhibited breast progression by SNHG3 knockdown via regulation of miR-326.

Knockdown of SNHG3 suppresses tumor growth in vivo

Athymic mice received injection of MCF-7 cells with stable SNHG3 depletion as described above in order to assess the role of SNHG3 in breast cancer growth in vivo. In comparison to controls, the tumors with depleted SNHG3 showed a significantly retarded pace of growth (Fig. 6A). 30 days post injection, the tumors were subjected to excision followed by imaging as shown in Fig. 6B. In comparison with the controls, the average weight of the SNHG3-depleted tumors was significantly lower (Fig. 6C). RT-qPCR was utilized to assess the levels of SNHG3, miR-326. While the level of SNHG3 in the tumor group with depleted SNHG3 was significantly lower in comparison to the controls (P<0.01, Fig. 6D), that of miR-326 was higher in the same group (Fig. 6D). Additionally, the expression of Ki-67 showed a significant reduction in the SNHG3-deleption MCF-7 tumor cells in comparison to the controls (Fig. 6E). The observations in this experiment are suggestive of suppressed breast cancer growth in vivo due to the knockdown of SNHG3.

Discussion

Many long non-coding RNAs (lncRNAs) have been identified to be deregulated and hence associated with the occurrence and development of breast cancer (11,12). The present study examined the role of small nucleolar RNA host gene 3 (SHNG3) in the origin and development of breast cancer using in vitro and in vivo assays. The results revealed an upregulation of SNHG3 in breast cancer tissues and cell lines, and downregulation of SNHG3 significantly inhibited the malignant progression of tumor cells.

An upregulation of SNHG3 as well as its role in oncogenesis in several types of cancer has been previously indicated (1621). For instance, Zhang et al reported that SNHG3 overexpression augmented the ability of hepatocellular carcinoma cells to invade, undergo epithelial-mesenchymal transition (EMT), and develop sorafenib resistance via regulation of the miR-128/CD151 axis (18). Fei et al showed that the upregulation of SNHG3 promoted glioma cell proliferation, accelerated cell cycle progression, and repressed cell apoptosis through an epigenetic repression of KLF2 and p21 via enhancer of zeste homolog 2 recruitment to the promoter of KLF2 and p21 (20). Huang et al found that SNHG3 promoted progression of colorectal cancer via miR-182-5p sponging that upregulated c-Myc along with its target genes (17). Consistent with these findings, the present study reports an elevated SNHG3 expression in breast cancer cell lines and tissue samples in comparison with the relevant normal controls. Knockdown of SNHG3 showed a distinct inhibition of the ability of tumor cells to proliferate, form colonies, migrate and invade in the laboratory while the growth of tumors was delayed in a the mouse model used. This is suggestive of a function of SNHG3 as an oncogene in breast cancer progression.

Growing evidence points to the function of lncRNAs as competing endogenous RNAs (ceRNAs) that sponge microRNAs in order to modulate their functions in turn to affect the manifestations observed in malignancies (25,26). SNHG3 has been reported to act as a ceRNA to sponge for miR-128 (18), miR-182-5p (17) and miR-384 (27). For example, SNHG3 promoted colorectal cancer progression via sponging miR-182-5p and upregulating c-Myc and its target genes (17). SNHG3 was found to accelerate papillary thyroid carcinoma progression by regulation of the miR-214-3p/PSMD10 axis (28). SNHG3 was also found to function as a miRNA sponge to promote hepatocellular carcinoma growth (29). Thus, it is necessary to identify target miRNAs of SNHG3 to clarify the molecular mechanism of SNHG3 in breast cancer. Through Starbase2.0 software, it was found that SNHG3 binds with miR-326. Previous studies have demonstrated the tumor-suppressor function of miR-326 in multiple cancers by regulating the ability of cells to proliferate, migrate and invade (30,31). Recent studies have found that the levels of miR-326 are lower in breast cancer tissues (32,33). In particular, a recent study by our team found that miR-326 functions as a tumor suppressor in breast cancer by targeting SOX12 (34). The RT-qPCR results showed that overexpression of miR-326 significantly decreased SNHG3 expression whereas depletion of SNHG3 obviously increased miR-326 levels in the assayed cells. SNHG3 and miR-326 were found to possess an inverse correlation in this sample set. The observations are suggestive of SNHG3 targeting miR-326 in breast cancer. Importantly, we found that inhibition of miR-326 caused a conspicuous reversal of the SNHG3 knockdown-mediated suppression in terms of MCF-7 cell proliferation, colony formation, migration, and invasion as well as arrest of the cell cycle. These results suggest that SNHG3 functions as a ceRNA via the sponging of miR-326 in breast cancer.

Some limitations exist in this study. First, the sample size of the breast cancer tissues was small. Thus, we may harvest the data of TCGA and GEO to investigate the clinical significance of SNHG3 in breast cancer in the future. We may also investigate the association of SNHG3 expression and overall survival of patients with breast cancer in the future. Second, we evaluated the biological role of SNHG3 in breast cancer using MCF-7 cells (a luminal cell line). We may further test the function of SNHG3 in breast cancer using two or more cell lines. Third, SOX12 was identified as a direct target of miR-326 in breast cancer; thus, the associations among SNHG3, miR-326 and SOX12 in breast cancer need further exploration.

Taken together, the present findings are a first to show upregulation of SNHG3 in breast cancer tissues and cell lines. Knockdown of SNHG3 caused a distinct inhibition of tumorigenesis via miR-326, suggesting that SNHG3 may be explored to be utilized in therapeutic applications for breast cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Education Department of Jilin Province (JJKH20170833KJ) and Jilin Province Department of Science and Technology (20180520055JH).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

HZ and YD conceived the study concept and design. NW, WZ, LS and QL performed the experiments. RD and SL analyzed the data and QL and SL wrote the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Jilin University (Changchun, China) based on 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 

Fan L, Strasser-Weippl K, Li JJ, St Louis J, Finkelstein DM, Yu KD, Chen WQ, Shao ZM and Goss PE: Breast cancer in China. Lancet Oncol. 15:e279–e289. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Zhang T, Li J, He Y, Yang F, Hao Y, Jin W, Wu J, Sun Z, Li Y, Chen Y, et al: A small molecule targeting myoferlin exerts promising anti-tumor effects on breast cancer. Nat Commun. 9:37262018. View Article : Google Scholar : PubMed/NCBI

4 

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

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 

Bouckenheimer J, Assou S, Riquier S, Hou C, Philippe N, Sansac C, Lavabre-Bertrand T, Commes T, Lemaître JM, Boureux A and Vos JD: Long non-coding RNAs in human early embryonic development and their potential in ART. Hum Reprod Update. 23:19–40. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Archer K, Broskova Z, Bayoumi AS, Teoh JP, Davila A, Tang Y, Su H and Kim IM: Long non-coding RNAs as master regulators in cardiovascular diseases. Int J Mol Sci. 16:23651–23667. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Heward JA and Lindsay MA: Long non-coding RNAs in the regulation of the immune response. Trends Immunol. 35:408–419. 2014. 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 

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

11 

Tian T, Wang M, Lin S, Guo Y, Dai Z, Liu K, Yang P, Dai C, Zhu Y, Zheng Y, et al: The impact of lncRNA dysregulation on clinicopathology and survival of breast cancer: A systematic eeview and meta-analysis. Mol Ther Nucleic Acids. 12:359–369. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Cerk S, Schwarzenbacher D, Adiprasito JB, Stotz M, Hutterer GC, Gerger A, Ling H, Calin GA and Pichler M: Current status of long non-coding RNAs in human breast cancer. Int J Mol Sci. 17:14852016. View Article : Google Scholar

13 

Zhang Y, Wu J, Jing H, Huang G, Sun Z and Xu S: Long noncoding RNA MEG3 inhibits breast cancer growth via upregulating endoplasmic reticulum stress and activating NF-κB and p53. J Cell Biochem. 120:6789–6797. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Qiao E, Chen D, Li Q, Feng W, Yu X, Zhang X, Xia L, Jin J and Yang H: Long noncoding RNA TALNEC2 plays an oncogenic role in breast cancer by binding to EZH2 to target p57KIP2 and involving in p-p38 MAPK and NF-κB pathways. J Cell Biochem. 120:3978–3988. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, Liu Y, Zhang N and Yang Q: LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. J Cell Physiol. 234:9105–9117. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Liu L, Ni J and He X: Upregulation of the long noncoding RNA SNHG3 promotes lung adenocarcinoma proliferation. Dis Markers. 2018:57367162018. View Article : Google Scholar : PubMed/NCBI

17 

Huang W, Tian Y, Dong S, Cha Y, Li J, Guo X and Yuan X: The long non-coding RNA SNHG3 functions as a competing endogenous RNA to promote malignant development of colorectal cancer. Oncol Rep. 38:1402–1410. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Zhang PF, Wang F, Wu J, Wu Y, Huang W, Liu D, Huang XY, Zhang XM and Ke AW: LncRNA SNHG3 induces EMT and sorafenib resistance by modulating the miR-128/CD151 pathway in hepatocellular carcinoma. J Cell Physiol. 234:2788–2794. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Hong L, Chen W, Wu D and Wang Y: Upregulation of SNHG3 expression associated with poor prognosis and enhances malignant progression of ovarian cancer. Cancer Biomark. 22:367–374. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Fei F, He Y, He S, He Z, Wang Y, Wu G and Li M: lncRNA SNHG3 enhances the malignant progress of glioma through silencing KLF2 and p21. Biosci Rep. 38:BSR201804202018. View Article : Google Scholar : PubMed/NCBI

21 

Ma Q, Qi X, Lin X, Li L, Chen L and Hu W: LncRNA SNHG3 promotes cell proliferation and invasion through the miR-384/hepatoma-derived growth factor axis in breast cancer. Hum Cell. 33:232–242. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Liang X, Li Z, Men Q, Li Y, Li H and Chong T: Mir-326 functions as a tumor suppressor in human prostatic carcinoma by targeting mucin1. Biomed Pharmacother. 108:574–583. 2018. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

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

25 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The rosetta stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Chan JJ and Tay Y: Noncoding RNA: RNA regulatory networks in cancer. Int J Mol Sci. 19:13102018. View Article : Google Scholar

27 

Wang L, Su K, Wu H, Li J and Song D: LncRNA SNHG3 regulates laryngeal carcinoma proliferation and migration by modulating the miR-384/WEE1 axis. Life Sci. 232:1165972019. View Article : Google Scholar : PubMed/NCBI

28 

Sui G, Zhang B, Fei D, Wang H, Guo F and Luo Q: The lncRNA SNHG3 accelerates papillary thyroid carcinoma progression via the miR-214-3p/PSMD10 axis. J Cell Physiol. 11:295572020.

29 

Wu J, Liu L, Jin H, Li Q, Wang S and Peng B: LncSNHG3/miR-139-5p/BMI1 axis regulates proliferation, migration, and invasion in hepatocellular carcinoma. Onco Targets Ther. 12:6623–6638. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Wang J, Cao L, Wu J and Wang Q: Long non-coding RNA SNHG1 regulates NOB1 expression by sponging miR-326 and promotes tumorigenesis in osteosarcoma. Int J Oncol. 52:77–88. 2018.PubMed/NCBI

31 

Li Y, Gao Y, Xu Y, Ma H and Yang M: Down-Regulation of miR-326 is associated with poor prognosis and promotes growth and metastasis by targeting FSCN1 in gastric cancer. Growth Factors. 33:267–274. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Liang Z, Wu H, Xia J, Li Y, Zhang Y, Huang K, Wagar N, Yoon Y, Cho HT, Scala S and Shim H: Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem Pharmacol. 79:817–824. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Ghaemi Z, Soltani BM and Mowla SJ: MicroRNA-326 functions as a tumor suppressor in breast cancer by targeting ErbB/PI3K signaling pathway. Front Oncol. 9:6532019. View Article : Google Scholar : PubMed/NCBI

34 

Du Y, Shen L, Zhang W, Ding R, Li Q, Li S and Zhang H: Functional analyses of microRNA-326 in breast cancer development. Biosci Rep. 39:BSR201907872019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 44 Issue 4

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
Zhang H, Wei N, Zhang W, Shen L, Ding R, Li Q, Li S and Du Y: lncRNA SNHG3 promotes breast cancer progression by acting as a miR‑326 sponge Retraction in /10.3892/or.2022.8255. Oncol Rep 44: 1502-1510, 2020
APA
Zhang, H., Wei, N., Zhang, W., Shen, L., Ding, R., Li, Q. ... Du, Y. (2020). lncRNA SNHG3 promotes breast cancer progression by acting as a miR‑326 sponge Retraction in /10.3892/or.2022.8255. Oncology Reports, 44, 1502-1510. https://doi.org/10.3892/or.2020.7690
MLA
Zhang, H., Wei, N., Zhang, W., Shen, L., Ding, R., Li, Q., Li, S., Du, Y."lncRNA SNHG3 promotes breast cancer progression by acting as a miR‑326 sponge Retraction in /10.3892/or.2022.8255". Oncology Reports 44.4 (2020): 1502-1510.
Chicago
Zhang, H., Wei, N., Zhang, W., Shen, L., Ding, R., Li, Q., Li, S., Du, Y."lncRNA SNHG3 promotes breast cancer progression by acting as a miR‑326 sponge Retraction in /10.3892/or.2022.8255". Oncology Reports 44, no. 4 (2020): 1502-1510. https://doi.org/10.3892/or.2020.7690