CircWEE1/miR-138 axis promotes the malignant progression of glioma by regulating SIRT1
Original Article

CircWEE1/miR-138 axis promotes the malignant progression of glioma by regulating SIRT1

Jinhui Guo1, Lifang Yu2

1Department of Neurosurgery, The Second Hospital of Shanxi Medical University, Taiyuan, China; 2Department of Neurosurgery, Jin Zhong City Chinese Hospital, Jinzhong, China

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Jinhui Guo. Department of Neurosurgery, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Xinghualing District, Taiyuan 030001, China. Email: Guojinhuijz@163.com.

Background: This study aims to reveal the potential impact of circWEE1 on the malignant progression of gliomas and its mechanism.

Methods: Real-time quantitative polymerase chain reaction (qRT-PCR) were used to detect circWEE1 levels in glioma tissues and cell lines. The relationship between circWEE1 expression and glioma metastasis was analyzed.

Results: After knocking out or over-expressing circWEE1, the effects on glioma cells were examined separately. Subsequently, the regulatory relationship of circWEE1 to miR-138 was detected by a dual luciferase reporter gene. In addition, we evaluated the role of silent information regulator 1 (SIRT1) in the progression of gliomas affected by circWEE1 through a rescue experiment. CircWEE1 was significantly up-regulated in glioma tissues and cell lines. At the same time, its expression level was significantly higher in glioma patients with lymphatic or distant metastasis than in glioma patients with non-metastasis. The down-regulation of circWEE1 reduced the viability, migration, and invasion ability of T98-G cells. The expression of miR-138 is negatively regulated by WEE1, while miR-138 directly targets and regulates the expression of SIRT1.

Conclusions: The rescue experiment confirmed that SIRT1 is a regulator of circWEE1 in the malignant progression of glioma. In summary, the CircWEE1/miR-138 axis may regulate SIRT1 to promote malignant progression in glioma.

Keywords: Glioma; circWEE1; miR-138; silent information regulator 1 (SIRT1); metastasis


Submitted Jan 11, 2021. Accepted for publication Apr 15, 2021.

doi: 10.21037/tcr-21-251


Introduction

Glioblastoma is the most common adult central nervous system tumor accounting for 45–50% of CNS tumors (1-4). While gliomas include astrocytoma, oligodendroglioma, and ependymoma, glioblastoma accounts for more than half of these, and is the most malignant. Most gliomas occur slowly, usually in weeks to months from symptom onset to treatment. Since glioma seriously affects the quality of life of patients, its treatment is also the focus of significant clinical research.

Competitive endogenous RNA (ceRNA) is a transcription product that achieves mutual regulation through the competitive binding of miRNA reaction elements to common miRNA, and this mechanism is known as the “ceRNA” hypothesis (5). Since any transcription product with miRNA reaction element structure can theoretically function as ceRNA, ceRNA theory is of great significance for integrating the pathogenesis of diseases, especially for tumor research. CircRNA is a kind of non-coding RNA produced by special selective shearing. Because it is a closed circular structure, circRNA has no free end and is not affected by RNA exonuclease, so it is more stable than linear RNA (6). While there are few studies on circRNA at present and the mechanism has not been clarified, there are still experiments (7) that confirm that some circRNA are rich in miRNA binding sites and play the role of competitive binding of miRNA in cells, which is a form of efficient ceRNA (8-12).

MicroRNAs (miRNAs) are a class of endogenous, non-coding single-stranded small RNA with a length of about 18–24 nucleotides, which are involved in various stages of tumor development and metastasis. MiR-138 is an important regulatory factor in tumors, and its expression is low in glioma cells and tissues. However, its role in glioma patients is not fully understood. As a member of the Sirtuin family, silent information regulator 1 (SIRT1) is a protein deacetylase that relies on NAD to deacetylate lysine residues of various proteins. Sirtuin is highly conserved in evolution from bacteria to eukaryotes and plays an important role in a variety of cellular functions in mammals, such as gene silencing, control of cell cycle and apoptosis, and energy balance (13). SIRT1 has a wide range of biological functions, such as growth regulation, response to stress response, tumor formation, and life extension. SIRT1 might be a target for the treatment of glioma. Several compounds have been shown to treat gliomas by inhibiting SIRT1. Such as, melatonin modulates the microenvironment of glioblastoma multiforme by targeting sirtuin 1 (14). However, at present, there are few studies on the role of SIRT1 in the pathogenesis of glioma.

In this study, an in vitro overexpression and knockout model of circWEE1 was constructed. At the same time, the target gene and regulation of SIRT1 on glioma progression were further investigated. The objective of this study was to describe the regulatory role of circWEE1/miR-138/SIRT1 axis in glioma and to describe the cellular/molecular mechanism of this axis function. This study is the first to report the biological function of circWEE1 in glioma. Our study reveals new aspects of cell function and pathophysiology of circWEE1 and miR-138. All of them can be considered as potential molecular targets for the treatment of glioma.

We present the following article in accordance with the ARRIVE reporting checklist (available at http://dx.doi.org/10.21037/tcr-21-251).


Methods

Patient and glioma samples

A total of 10 matched glioma and adjacent tissues were obtained between September 2017 and October 2019 from The Second Hospital of Shanxi Medical University. The tumor staging of gliomas was evaluated according to criteria proposed by the UICC. The experiment was approved by the ethics committee of Shanxi Medical University in line with the Declaration of Helsinki (as revised in 2013), and informed consent was obtained from all patients.

Cell culture

Human glioma cell lines (U251, U87, T98-G) and human brain normal glial cell lines (HEB) were provided by ATCC (American Type Culture Collection; Manassas, Virginia, USA). All cells were cultured in DMEM medium (DMEM; Thermo Fisher Scientific, Wal-Mart, Massachusetts) containing 10% fetal bovine serum (FBS; Gibco, Rockville, MD, USA) at 37 °C in a humidified incubator with 5% CO2.

Cell transfection

MiR-138 inhibitor or mimics and NC inhibitor or mimics were purchased from Genechem (Shanghai, China). Cell transfection was performed according to the instructions of Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). Cells were seeded into 6-well plates, cultured until confluence reached 60% for transfection and then harvested 48 hours after transfection for functional experiments and detection of protein expression.

Cell proliferation assay

Transfected cells were seeded into 96-well plates at a density of 2×103 cells per well. On days 1, 2, 3, and 4, a CCK-8 kit (Dojindo Laboratories, Kumamoto, Japan) was added to each well and incubated for 2 hours in the dark. Absorbance was recorded at 450 nm with a microplate reader (Japan, Nikon) and after the detection value was obtained, the cell proliferation rate was calculated.

Transwell experiment

Transfected cells were seeded into 24-well plates at 2×105 cells per well. 200 µL of the cell suspension was added to the Transwell upper chamber (Millipore, Billerica, MA, USA), and 600 µL of complete medium with 10% FBS added to the Transwell lower chamber. After 48 hours of incubation, the cells were fixed in methanol for 15 minutes and stained with crystal violet for 10 minutes. Cell migration was then observed under a microscope (15,16).

Scratch test

Cells were seeded into a 24-well plate at 5.0×105 cells/well. After the cells adhered, an artificial wound was formed in the fused cell monolayer using a 100 µL pipette tip. Wound closure images were taken at 0 hours (cell adhesion) and 48 hours using an inverted microscope and the percentage of wound closure was calculated.

Real-time quantitative polymerase chain reaction (qRT-PCR)

Total cellular RNA was extracted using TRIzol reagent (Invitrogen, Carlsbad, California, USA). After total RNA was purified by DNase I treatment, the extracted RNA was reverse transcribed into cDNA using PrimeScript RT reagent (TaKaRa, Otsu, Shiga, Japan). QRTPCR was performed on the obtained cDNA using SYBR Premix Ex Taq™ (TaKaRa, Ostu, Shiga, Japan). The β-actin and U6 were used as internal references and each sample was repeated three times. The relative expression levels of genes were calculated by the 2−ΔΔCt method.

Double-Luciferase reporter assay

After co-transfection of the plasmid for 36–48 h, the medium was discarded, and the cells were washed with 100 µL of 1× PBS. Deionized water diluted the 5× PLB (lysate) to 1× PLB and placed it at normal temperature before use. A 50 µL portion of the diluted 1× PLB was then added to each well and placed on a shaker for 20–30 minutes to ensure that the lysis buffer completely lysed the cells. A white opaque 96-well microtiter plate was then added with 10 µL of supernatant to each well, and 100 µL of premixed Luciferase Assay Reagent II was added to measure the data to measure the luciferase reaction intensity. After the measurement was completed, 100 µL of premixed Stop & Glo Reagent was added to each well and allowed to stand for 2 s, and the data was then measured to determine the intensity of the internal reference Renilla luciferase reaction. The ratio of the two sets of data was then calculated.

In vivo study

Six female nude mice (18–22 g) were purchased from Kay Biological Technology Co. Ltd (Shanghai, China). The BALB/c mice were randomly divided into a sh-NC group and a sh-CircWEE1group. The sh-NC group were injected with 100 µL of phosphate buffered sodium only as control and the sh-CircWEE1group were injected with 100 µL of T98-G cells by subcutaneous injection. After two weeks, mice were dissected and the tumors were obtained and weighed. Experiments were performed under a project license (NO.: IRB-2018-012) granted by ethics board of Shanxi Medical University, in compliance with NIH guidelines for the care and use of animals.

Statistical analysis

SPSS 22.0 (SPSS IBM Corp., Armonk, NY, USA) was used for all statistical analyses. Experimental data are expressed as mean ± standard deviation. Differences between groups were analyzed by t test and multiple groups were compared using ANOVA. P<0.05 was considered statistically significant (17).


Results

CircWEE1 is up-regulated in glioma tissues and cell lines

CircWEE1 expression was significantly up-regulated in glioma tissues compared to adjacent cancer tissues (Figure 1A). Similarly, the expression level of circWEE1 in glioma cells was significantly higher than that of normal glioma cells (Figure 1B). As T98-G expressed the highest circWEE1 abundance among the three glioma cell lines, it was used in subsequent experiments. CircWEE1 expression is related to metastasis and prognosis in glioma patients. We found that circWEE1 levels were positively correlated with lymphatic and distant metastases and lymphatic or distant metastatic patients with glioma had higher circWEE1 expression (Figure 1C,D).

Figure 1 CircWEE1 expression in glioma and adjacent tissues and analysis of clinical information. (A) CircWEE1 level in para-cancerous tissues and glioma tissues; (B) circWEE1 level in normal glial cells and glioma cells; (C) circWEE1 level in glioma patients either with lymphatic metastasis or not; (D) circWEE1 level in glioma patients either with distant metastasis or not. **P<0.01.

CircWEE1 promotes the progression of gliomas in vivo and in vitro

We transfected T98-G cells with si-circWEE1 to detect the mechanism of circWEE1 in gliomas. Figure 2A shows that si-circWEE1 markedly decreased the level of circWEE1, which indicated that the transfection was effective, and Transwell assay and cell scratch test results suggested that si-circWEE1 inhibited the invasion and migration abilities of T98-G cells (Figure 2B,C). Further experiment also indicated that sh-circWEE1 inhibited the growth of gliomas in vivo (Figure 2D). Collectively, these results show circWEE1 can promote the progression of gliomas in vivo and in vitro.

Figure 2 CircWEE1 promotes the progression of gliomas in vivo and in vitro. (A) RT-qPCR was used to test the level of CircWEE1; (B) si-CircWEE1 decreased the number of invasive cells (Transwell assay, Magnification 100×); (C) si-CircWEE1 inhibited cell migration; (D) Downregulation of CircWEE1 inhibited the growth of tumor in vivo. *P<0.05, **P<0.01.

MiR-138 is the target gene of circWEE1

To further study the mechanism of circWEE1 in gliomas, we first analyzed the miRNAs bound by circWEE1. The results show that circWEE1 binds to miR-138 (Figure 3A) and further detection of luciferase reporter gene confirmed the binding site between circWEE1 and miR-138 (Figure 3B). To verify the regulatory effect of circWEE1 on miR-138, we overexpressed and knocked down circWEE1, respectively (Figure 3C). The expression of miR-138 showed that over-expression of circWEE1 could inhibit the expression of miR-138, while knockdown of circWEE1 could promote the expression of miR-138 (Figure 3D). The co-expression test results of circWEE1 and miR-138 showed that they had a negative co-expression correlation (Figure 3E).

Figure 3 CircWEE1 sponge as miR-138. (A) CircWEE1 and miR-138 binding site information maps; (B) dual luciferase report detects circWEE1 binding to miR-138; (C) circWEE1 overexpression and expression verification after knockdown; (D) circWEE1 inhibits miR-138 expression; (E) correlation analysis between circWEE1 and miR-138 co-expression. *P<0.05, **P<0.01, ***P<0.001.

MiR-138 reduces malignant progression of glioma

Figure 4A suggests that miR-138 mimics upregulated the expression of miR-138 and miR-138 inhibitor down-regulated the expression of miR-138. Cell Transwell experiments showed that miR-138 mimics added to glioma cell lines reduced cell invasion capacity and adding miR-138 inhibitors increased cell invasion (Figure 4B). Cell migration experiments showed that miR-138 mimics added to glioma cell lines reduced cell migration capacity, while adding miR-138 inhibitors enhances cell migration capacity (Figure 4C). All results indicated that miR-138 could inhibit the malignant progression of glioma.

Figure 4 MiR-138 reduces malignant evolution of glioma. (A) RT-qPCR was used to test the level of miR-138; (B) cell invasion experiment. miR-138 overexpression inhibited cell invasion ability and miR-138 knockdown promoted cell invasion ability (Transwell assay, magnification 100×); (C) cell migration experiments. miR-138 overexpression inhibited cell migration ability and miR-138 knockdown promoted cell migration ability, *P<0.05, **P<0.01.

MiR-138 inhibits SIRT1 expression

To investigate the target genes of miR-138, we used TargetScan to predict the target gene information of miR-138 and Figure 5A shows a map of the binding sites of the two. Luciferase reporter assay suggested that miR-138 can bind to SIRT1 (Figure 5B). We transfected T98G cells with mimics NC, miR-138 mimics, inhibitor NC, and miR-138 inhibitor and detected the expression of miR-138 by PT-qPCR and the results indicated that the transfection is effective (Figure 5C).

Figure 5 MiR-138 inhibits SIRT1 expression. (A) Information maps of miR-138 and SIRT1 binding sites; (B) dual luciferase report assay detects the relationship between miR-138 and SIRT1; (C) when miR-138 mimics was added to the glioma cell line, SIRT1 content decreased. With the addition of miR-138 inhibitor, SIRT1 content increased. **P<0.01.

CircWEE1 promotes the malignant evolution of glioma cells by up-regulating the expression of SIRT1 through sponge adsorption of miR-138

To further study the regulatory mechanism of ceRNA between circWEE1 and miR-138, we first tested the expression of SIRT1 and the experimental results showed that si-circWEE1 could inhibit the expression of SIRT1. However, miR-138 inhibitor could reverse the effect of si-circWEE1 on SIRT1 expression (Figure 6A). Cell proliferation test results showed that si-circWEE1 could inhibit the proliferation of glioma cells but miR-138 inhibitor could reverse the inhibitory effect of si-circWEE1 (Figure 6B). Cell Transwell and scratch detection results showed that si-circWEE1 could inhibit the invasion and migration ability of glioma cells but miR-138 inhibitor could reverse the inhibitory effect of si-circWEE1 (Figure 6C,D). E-cadherin expression was consistent with SIRT1 expression (Figure 6E), while Vimentin expression was correlated with SIRT1 expression (Figure 6F). The experimental results showed that circRNA up-regulates the expression of SIRT1 by sponge-adsorbed miR-138 to promote the malignant evolution of glioma cells.

Figure 6 CircWEE1 promotes the malignant evolution of glioma cells by up-regulating the expression of SIRT1 through the adsorption of miR-138 by sponge effect. (A) Detection of SIRT1 expression; (B) cell proliferation detection; (C) detection of cell invasion (Transwell assay, magnification 100×); (D) cell migration detection; (E) detection of E-cadherin expression; (F) detection of Vimentin expression. *P<0.05, **P<0.01.

Discussion

Glioma is one of the most common intracranial malignant tumors. The WHO categorizes glioma according to its malignancy degree into four levels. While levels I-II level are considered as low-grade glioma, levels III-IV are high-grade glioma, and the both are categorized by an unclear pathogenesis, high mortality and short survival time (18,19). Therefore, it is particularly important to further understand the occurrence and development of glioma, identify the relevant factors affecting the degree of glioma malignancy, and find early diagnosis and treatment methods (20-22).

In this study, we found that circWEE1 was significantly up-regulated in gliomas and its level was positively correlated with high metastasis rate and poor prognosis in glioma patients. Therefore, circWEE1 may play a carcinogenic role in the progression of glioma. In vitro experiments showed that knockout of circWEE1 reduced the viability, migration ability, and wound closure rate of glioma cells. To date, several ceRNAs, including PTENP1 and KRAS, have been shown to be related to tumor development (23-26) and the clinical application of ceRNA in tumors is another future research direction. Cerebellar degeneration-related protein 1 antisense (CDR1as) consists of about 1500 nucleotides and has more than 70 miR-7 binding sites. Overexpression of CDR1 will inhibit the activity of miR-7, and this phenomenon is particularly evident in the CNS (6). MiR-7 is abnormally expressed in malignant tumors such as glioblastoma, gastric cancer, and colorectal cancer, and through the competitive binding of miR-7, miR-7 target gene expression is increased. We have reason to suspect that CDR1 may be related to the progress of related tumors and play an important regulatory function (27). In esophageal squamous cell carcinoma, circ-ITCH regulates ITCH levels through competitive binding to miR-7, 17, 214. Barbagallo et al. (28) found that compared with normal brain tissue, circ-TTBK2 was significantly up-regulated in gliomas, inhibited tumor cell apoptosis, and was positively correlated with the pathological grade of gliomas. Using normal astrocytes for comparison, we found that miR-217 was down-regulated in glioma U87 and U251 cell lines, and that returning miR-217 to normal expression could inhibit the proliferation of glioma cells. On this basis, we speculate that miR-217 plays a tumor suppressive role in gliomas. Our results also show that circ-TTBK2 reduced the expression of miR-217 in glioma cells in the manner of RNA-induced silencing complex (RISC).

MiRNA is a member of the non-coding RNA family. In recent years, research has found that miRNA plays an important role in the occurrence and development of tumors and has great clinical diagnostic value (29). Lu et al. (30) found that down-regulation of the miR-573 expression level promoted the occurrence of gastric cancer and can be used as a tumor marker in its early diagnosis. MiR-138 is a new class of miRNAs with tumor regulation showing low expression in a variety of malignant tumors (31,32). Therefore, miR-138 usually acts as a tumor suppressor gene during tumor formation. Stojcheva et al. (33) showed that miR-138 can inhibit autophagy-mediated temozolomide chemotherapy resistance to glioma cells by targeting the inhibitor of apoptosis protein BIM. Studies have also shown that miR-138 down-regulates hTERT protein and its expression inhibits the proliferation and migration of glioma cells. On this basis, miR-138 is considered a new tumor suppressor for gliomas.

The occurrence of tumors is closely related to the overexpression of oncogenes or the low expression of tumor suppressor genes. Type III histone deacetylase SIRT1 gene expression and deacetylase activity are up-regulated in tumor cells and it is speculated that these may cause tumorigenesis. SIRT1 can also cause deacetylation of tumor suppressors and promote tumorigenesis. In the occurrence, development, and maintenance of various characteristics of tumors, SIRT1 is likely to play an important role because it can promote tumor cell proliferation and inhibit apoptosis and aging. Studies have reported that SIRT1 is significantly overexpressed in human prostate cancer, human primary colon cancer, and acute myeloid leukemia (34-36). In addition, SIRT1 is also highly expressed in human squamous cell carcinoma and basal cell carcinoma (37-38) and can silence growth and apoptosis of human epithelial cancer cells by silencing SIRT1 (39). FOXM1 is an upstream regulator of SIRT1 in glioma cells. FOXM1 is highly expressed in gliomas. The expression of FOXM1 in glioma cells was overexpressed by gene transfection, and the mRNA and protein levels of SIRT1 in glioma cells were significantly increased. Silencing the expression of FOXM1 with siRNA reduced the expression of SIRT1 in glioma cells. At the transcriptional level, FOXM1 was found to bind to the promoter region of SIRT1 gene in glioma cells and activate SIRT1 promoter activity in a dose-dependent manner. The regulation of FOXM1 on SIRT1 in glioma was further confirmed (40).

In summary, circWEE1 is upregulated in glioma tissues and is related to distant metastases and poor prognosis in glioma patients. CircWEE1 up-regulates SIRT1 expression by targeting miR-138, aggravating the malignant progression of glioma.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the ARRIVE reporting checklist. Available at http://dx.doi.org/10.21037/tcr-21-251

Data Sharing Statement: Available at http://dx.doi.org/10.21037/tcr-21-251

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr-21-251). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The experiment involving human was approved by the ethics committee of Shanxi Medical University in line with the Declaration of Helsinki (as revised in 2013), and informed consent was obtained from all patients. Experiments were performed under a project license (NO.: IRB-2018-012) granted by ethics board of Shanxi Medical University, in compliance with NIH guidelines for the care and use of animals.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Liu Y, Ma C, Qin X, et al. Circular RNA circ_001350 regulates glioma cell proliferation, apoptosis, and metastatic properties by acting as a miRNA sponge. J Cell Biochem 2019;120:15280-7. [Crossref] [PubMed]
  2. Liu J, Zhao K, Huang N, et al. Circular RNAs and human glioma. Cancer Biol Med 2019;16:11-23. [Crossref] [PubMed]
  3. Li G, Yang H, Han K, et al. A novel circular RNA, hsa_circ_0046701, promotes carcinogenesis by increasing the expression of miR-142-3p target ITGB8 in glioma. Biochem Biophys Res Commun 2018;498:254. [Crossref] [PubMed]
  4. Xi X, Liu N, Wang Q, et al. ACT001, a novel PAI-1 inhibitor, exerts synergistic effects in combination with cisplatin by inhibiting PI3K/AKT pathway in glioma. Cell Death Dis 2019;10:757. [Crossref] [PubMed]
  5. Wang Y, Lin G. TP53INP1 3'-UTR functions as a ceRNA in repressing the metastasis of glioma cells by regulating miRNA activity. Biotechnol Lett 2016;38:1699-707. [Crossref] [PubMed]
  6. Conn SJ, Pillman KA, Toubia J, et al. The RNA Binding Protein Quaking Regulates Formation of circRNAs. Cell 2015;160:1125-34. [Crossref] [PubMed]
  7. Hansen TB, Jensen TI, Clausen BH, et al. Natural RNA circles function as efficient microRNA sponges. Nature 2013;495:384-8. [Crossref] [PubMed]
  8. Yang Y, Gao X, Zhang M, et al. Novel Role of FBXW7 Circular RNA in Repressing Glioma Tumorigenesis. J Natl Cancer Inst 2018;110:304-15. [Crossref] [PubMed]
  9. Xu H, Yu Z, Ling Q, et al. NFIX Circular RNA Promotes Glioma Progression by Regulating miR-34a-5p via Notch Signaling Pathway. Front Mol Neurosci 2018;11:225. [Crossref] [PubMed]
  10. Xiong J, Wang T, Tang H, et al. Circular RNA circMAN2B2 facilitates glioma progression by regulating the miR05/S100A8 axis. J Cell Physiol 2019;234:22996-3004. [Crossref] [PubMed]
  11. Xie G. Circular RNA hsa-circ-0012129 Promotes Cell Proliferation and Invasion in 30 Cases of Human Glioma and Human Glioma Cell Lines U373, A172, and SHG44, by Targeting MicroRNA-661 (miR-661). Med Sci Monit 2018;24:2497-507. [Crossref] [PubMed]
  12. Zhong W, Sun B, Lu C, et al. Problems and Solutions in Click Chemistry Applied to Drug Probes. Sci Rep 2016;6:35579. [Crossref] [PubMed]
  13. Yamamoto H, Schoonjans K, Auwerx J. Sirtuin Functions in Health and Disease. Mol Endocrinol 2007;21:1745-55. [Crossref] [PubMed]
  14. Lai S-W, Liu Y-S, Lu D-Y, Tsai C-F. Melatonin modulates the microenvironment of glioblastoma multiforme by targeting sirtuin 1. Nutrients 2019;11:1343. [Crossref] [PubMed]
  15. Wang H, Zhong W, Zhao J, et al. Oleanolic Acid Inhibits Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma by Promoting iNOS Dimerization. Mol Cancer Ther 2019;18:62-74. [Crossref] [PubMed]
  16. Xiao T, Zhong W, Zhao J, et al. Polyphyllin I suppresses the formation of vasculogenic mimicry via Twist1/VE-cadherin pathway. Cell Death Dis 2018;9:906. [Crossref] [PubMed]
  17. Zhong W, Sun B, Gao W, et al. Salvianolic acid A targeting the transgelin-actin complex to enhance vasoconstriction. EBioMedicine 2018;37:246-58. [Crossref] [PubMed]
  18. Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 2002;359:1011-8. [Crossref] [PubMed]
  19. Kruser TJ, Mehta MP, Robins HI. Pseudoprogression after glioma therapy: a comprehensive review. Expert Rev Neurother 2013;13:389-403. [Crossref] [PubMed]
  20. Rooney AG, Alan C, Robin G. Depression in Cerebral Glioma Patients: A Systematic Review of Observational Studies. J Natl Cancer Inst 2011;103:61-76. [Crossref] [PubMed]
  21. Perry J, Chambers A, Spithoff K, et al. Gliadel wafers in the treatment of malignant glioma: A systematic review. Curr Oncol 2007;14:189-94. [Crossref] [PubMed]
  22. Zhong W, Yang W, Qin Y, et al. 6-Gingerol stabilized the p-VEGFR2/VE-cadherin/beta-catenin/actin complex promotes microvessel normalization and suppresses tumor progression. J Exp Clin Cancer Res 2019;38:285. [Crossref] [PubMed]
  23. Ma CC, Xiong Z, Zhu GN, et al. Long non-coding RNA ATB promotes glioma malignancy by negatively regulating miR-200a. J Exp Clin Cancer Res 2016;35:90. [Crossref] [PubMed]
  24. Li DX, Fei XR, Dong YF, et al. The long non-coding RNA CRNDE acts as a ceRNA and promotes glioma malignancy by preventing miR-136-5p-mediated downregulation of Bcl-2 and Wnt2. Oncotarget 2017;8:88163-78. [Crossref] [PubMed]
  25. Cui B, Li B, Liu Q, et al. lncRNA CCAT1 Promotes Glioma Tumorigenesis by Sponging miR1b. J Cell Biochem 2017;118:4548-4557. [Crossref] [PubMed]
  26. Zhong W, Liu P, Zhang Q, et al. Structure-based QSAR, molecule design and bioassays of protease-activated receptor 1 inhibitors. J Biomol Struct Dyn 2017;35:2853-67. [Crossref] [PubMed]
  27. Hansen TB, Kjems J, Damgaard CK. Circular RNA and miR-7 in Cancer. Cancer Res 2013;73:5609-12. [Crossref] [PubMed]
  28. Barbagallo D, Condorelli AG, Ragusa M, et al. Dysregulated miR-671-5p/CDR1-AS/CDR1/VSNL1 axis is involved in glioblastoma multiforme. Oncotarget 2016;7:4746-59. [Crossref] [PubMed]
  29. Cazzoli R, Buttitta F, Nicola MD, et al. microRNAs Derived from Circulating Exosomes as Noninvasive Biomarkers for Screening and Diagnosing Lung Cancer. J Thorac Oncol 2013;8:1156-62. [Crossref] [PubMed]
  30. Lu Z, Luo T, Nie M, et al. TSPAN1 functions as an oncogene in gastric cancer and is downregulated by miR-573. FEBS Lett 2015;589:1988-94. [Crossref] [PubMed]
  31. Xiao L, Zhou H, Li XP, et al. MicroRNA-138 acts as a tumor suppressor in non small cell lung cancer via targeting YAP1. Oncotarget 2016;7:40038-46. [Crossref] [PubMed]
  32. Li B, Yang XX, Wang D, et al. MicroRNA-138 inhibits proliferation of cervical cancer cells by targeting c-Met. Eur Rev Med Pharmacol Sci 2016;20:1109-14. [PubMed]
  33. Stojcheva N, Schechtmann G, Sass S, et al. MicroRNA-138 promotes acquired alkylator resistance in glioblastoma by targeting the Bcl-2-interacting mediator BIM. Oncotarget 2016;7:12937-50. [Crossref] [PubMed]
  34. Chakrabarti Mrinmay, Naren L. Banik, Swapan K Ray. miR-138 overexpression is more powerful that hTERT knockdown to poteniate apogenin for apoptosis in neuroblastoma in vitro and vivo. Exp Cell Res 2013;319:1575-85. [Crossref] [PubMed]
  35. Huffman DM, Grizzle WE, Bamman MM, et al. SIRT1 is Significantly Elevated in Mouse and Human Prostate Cancer. Cancer Res 2007;67:6612-8. [Crossref] [PubMed]
  36. Bradbury CA, Khanim FL, Hayden R, et al. Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia 2005;19:1751-9. [Crossref] [PubMed]
  37. Stünkel W, Peh BK, Tan YC, et al. Function of the SIRT1 protein deacetylase in cancer. Biotechnol J 2007;2:1360-8. [Crossref] [PubMed]
  38. Deng CX. SIRT1, Is It a Tumor Promoter or Tumor Suppressor? Int J Biol Sci 2009;5:147-52. [Crossref] [PubMed]
  39. Ford J, Jiang M, Milner J. Cancer-Specific Functions of SIRT1 Enable Human Epithelial Cancer Cell Growth and Survival. Cancer Res 2005;65:10457-63. [Crossref] [PubMed]
  40. Zhu GY, Shi BZ, Li Y. FoxM1 regulates Sirt1 expression in glioma cells. Eur Rev Med Pharmacol Sci 2014;18:205-11. [PubMed]

(English Language Editor: B. Draper)

Cite this article as: Guo J, Yu L. CircWEE1/miR-138 axis promotes the malignant progression of glioma by regulating SIRT1. Transl Cancer Res 2021;10(4):1863-1873. doi: 10.21037/tcr-21-251

Download Citation