Open Access

MicroRNA‑423‑3p promotes glioma growth by targeting PANX2

Retraction in: /10.3892/ol.2021.12833

  • Authors:
    • Jing Xu
    • Jian He
    • He Huang
    • Renjun Peng
    • Jian Xi
  • View Affiliations

  • Published online on: May 4, 2018     https://doi.org/10.3892/ol.2018.8636
  • Pages: 179-188
  • Copyright: © Xu 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

Previously, a number of microRNAs (miRs) have been identified to participate in the development and progression of glioma via the regulation of their target genes. However, the molecular mechanisms underlying the effect of miR‑423‑3p in glioma growth remain unclear. In the present study, the reverse transcription‑quantitative polymerase chain reaction and western blotting were used to assess the mRNA and protein expression levels of miR‑423‑3p, respectively. An MTT assay and flow cytometry were performed to determine cell proliferation and apoptosis, respectively. A luciferase reporter gene assay was performed to determine the target association between pannexin 2 (PANX2) and miR‑423‑3p. It was revealed that miR‑423‑3p was significantly upregulated in glioma tissues compared with normal brain tissues, and the increased expression of miR‑423‑3p was significantly associated with an advanced grade as well as a poorer prognosis of patients with glioma. Inhibition of miR‑423‑3p using an miR‑423‑3p inhibitor resulted in the decreased proliferation of glioma U251 and U87MG Uppsala cells, and the induction of apoptosis. PANX2 was identified as a novel target gene of miR‑423‑3p, and the expression of PANX2 was revealed to be increased in U251 and U87MG Uppsala cells when miR‑423‑3p was inhibited. Knockdown of PANX2 attenuated the effects of miR‑423‑3p inhibition on glioma cell proliferation and apoptosis. Furthermore, PANX2 was significantly downregulated in glioma tissues compared with normal brain tissues, and its levels were markedly lower in World Health Organization (WHO) stage III‑IV gliomas compared with WHO stage I‑II gliomas. Additionally, the expression levels of PANX2 were identified to be inversely correlated with miR‑423‑3p expression levels in glioma tissues. Consequently, targeting miR‑423‑3p may inhibit glioma growth via the upregulation of PANX2.

Introduction

Malignant glioma is the most common type of cancer in the brain, accounting for 80% of all malignant brain tumors worldwide (1,2). In previous years, the deregulation of numerous oncogenes and tumor suppressors including microRNAs (miRs) has been observed in glioma, and investigating the underlying molecular mechanism may be beneficial for developing effective therapeutic strategies for this disease (24). miRs are a class of non-coding RNAs that are 18–25 nucleotides in length, and have been demonstrated to act as key regulators of gene expression by directly binding to the complementary regions of their target mRNA, leading to mRNA degradation or translational inhibition (5). Through the inhibition of the expression of their target mRNA, a multitude of miRs participate in various physiological and pathological biological processes, including differentiation, development, angiogenesis and tumorigenesis (57). Glioma, like other types of cancer, possesses a distinct miR expression signature, and previous studies have identified that a number of miRs are involved in the regulation of glioma cell proliferation, survival, cell cycle progression, migration and invasion (810). Furthermore, a number of miRs have been suggested as potential therapeutic targets in the treatment of glioma (8,10,11).

miR-423 is involved in a number of physiological and pathological progresses, including skeletal muscle development (12), childhood obesity (13), heart failure (14), idiopathic pulmonary fibrosis (15) and acute graft-versus-host disease (16). Furthermore, the deregulation of miR-423 has been identified to be involved in several types of human cancer (17,18). For instance, miR-423 was upregulated in head and neck squamous cell carcinoma tissues compared with normal tissues (17). It may also promote the proliferation of hepatocellular carcinoma cells through regulating the G1/S transition by targeting cyclin-dependent kinase inhibitor 1 (p21Cip1/Waf1) (18). Additionally, miR-423 promotes cell proliferation in breast cancer cell lines through its miR-423-3p strand as opposed to its miR-423-5p strand (19). Previously, miR-423-5p was demonstrated to be significantly upregulated in glioma, and promoted the malignant phenotypes of glioma cells as well as their temozolomide resistance (20), suggesting that miR-423-5p serves an oncogenic function in glioma. However, the regulatory function of miR-423-3p in glioma remains unclear.

In the present study, the molecular mechanisms underlying the effect of miR-423-3p on glioma growth were investigated.

Materials and methods

Tissue collection

The present study was approved by the Ethics Committee of Xiangya Hospital, Central South University (Changsha, China). A total of 58 cases of glioma specimens and 10 cases of normal brain tissues were obtained from Xiangya Hospital, Central South University (Changsha, China) between January 2010 and March 2012. Written informed consent was obtained from all patients prior to the study. Tissues were snap-frozen in liquid nitrogen following surgical resection, and stored in liquid nitrogen until use. The clinicopathological information of the patients with glioma included in the present study is summarized in Table I.

Table I.

Associations between miR-423-3p expression levels and the clinicopathological characteristics of patients with glioma.

Table I.

Associations between miR-423-3p expression levels and the clinicopathological characteristics of patients with glioma.

VariableCases (n=58)Low miR-423-3p level (n=31)High miR-423-3p level (n=27)P-value
Age, years 0.596
  <55251213
  ≥55331914
Sex 0.270
  Male392316
  Female19  811
WHO grade 0.026
  I–II2015  5
  III–IV381622
KPS 0.013
  >902116  5
  ≤90371522

[i] KPS, Karnofsky performance scale; WHO, World Health Organization; miR, microRNA.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the tissues and cells using TRIzol® reagent (Thermo Fisher Scientific, Inc., Waltham, MA, USA), according to the manufacturer's protocol. The MirVana™ RT-PCR microRNA detection kit (Thermo Fisher Scientific, Inc.) was used to examine miR expression, according to the manufacturer's protocol. U6 was used as an internal reference. The standard SYBR Green RT-PCR Kit (Takara Bio, Inc., Otsu, Japan) was used to examine mRNA expression, according to the manufacturer's protocol. GAPDH was used as an internal reference. The primers used were as follows: miR-423 forward, 5′-ATGGTTCGTGGGTGAGGGGCAGAGAGCGAGAGCAGGGTCCGAGGTATTCG-3′ and reverse, 5′-GTGCAGGGTCCGAGGT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; PANX2 forward, 5′-CCAAGAACTTCGCAGAGGAAC-3′ and reverse, 5′-GGGCAGGAACTTGTGCTCA-3′; GAPDH forward, 5′-GGAGCGAGATCCCTCCAAAAT-3′; and reverse, 5′-GGCTGTTGTCATACTTCTCATGG-3′. The reaction mixture included cDNA solution (1 µl), PCR master mix (10 µl), primers (2 µl) and water (7 µl). The reaction conditions were 9°C for 3 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 30 sec. The relative expression level was quantified using the 2−ΔΔCq method (21).

Cell culture

Human glioma U251 and U87MG Uppsala cell lines were obtained from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in Dulbecco's modified Eagle's medium (Thermo Fisher Scientific, Inc.) with 10% fetal bovine serum (Thermo Fisher Scientific, Inc.) and were maintained at 37°C in a humidified incubator (Thermo Fisher Scientific, Inc.) containing 5% CO2.

Cell transfection

Lipofectamine 2000 transfection reagent (Thermo Fisher Scientific, Inc.) was used to perform transfection, according to the manufacturer's protocol. Briefly, U251 and U87MG Uppsala cells were transfected with a negative control (NC) inhibitor, miR-423-3p inhibitor, non-specific small interfering RNA (siRNA) or PANX2-specific siRNA, respectively were purchased from Guangzhou FulenGen Co. Ltd. (Guangzhou, China). Following transfection at 37°C for 48 h, the expression assay was performed.

Western blotting

U251 and U87MG Uppsala cells were lysed in RIPA Lysis Buffer (Beyotime Institute of Biotechnology, Haimen, China). The protein concentration was quantified using a bicinchinonic acid protein assay kit (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Protein (50 µg) was separated by SDS-PAGE (12% gel), transferred onto a polylvinylidene fluoride membrane (Thermo Fisher Scientific, Inc.), and then blocked using 5% non-fat dried milk (Yili Group, Beijing, China) in Tris-buffered saline with Tween-20 (TBST; Beyotime Institute of Biotechnology) at room temperature for 3 h. The membrane was incubated with rabbit polyclonal anti-PANX2 primary antibody (1:100; cat no. ab55917; Abcam, Cambridge, MA, USA) or rabbit polyclonal anti-GAPDH primary antibody (1:100; cat no. ab9485; Abcam) at room temperature for 3 h, and then washed three times using TBST. Subsequently, the membrane was incubated with goat monoclonal anti-rabbit secondary antibody (1:5,000; cat no. ab190492; Abcam) for 1 h at room temperature, and washed three times using TBST. The immune complexes were detected using an enhanced chemiluminescence western blotting kit (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Image J software (version 1.0, National Institutes of Health, Bethesda, MD, USA) was used to analyze the relative protein expression, represented as the density ratio relative to GAPDH.

Cell proliferation analysis

U251 and U87MG Uppsala cells (2×103 cells per well) were seeded in 96-well plates, and 100 µl fresh serum-free DMEM with 0.5 g/l MTT solution (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added. Following incubation at 37°C for 0, 24, 48 and 72 h, the medium containing MTT solution was removed, and 50 µl dimethylsulfoxide (Sigma-Aldrich; Merck KGaA) was added. Following incubation at 37°C for 10 min, the absorbance at 570 nm of each sample was determined using a plate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Cell apoptosis analysis

A flow cytometer was used to determine the cell apoptosis with an Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection kit (Sigma-Aldrich; Merck KGaA). Cells were harvested and washed with ice-cold PBS twice, and 106 cells were resuspended in 200 µl binding buffer with 10 µl annexin-V-FITC and 5 µl propidium iodide-phycoerythrin, prior to incubation in the dark at 4°C for 30 min. Subsequently, 300 µl binding buffer was added, followed by analysis using flow cytometry (BD Accuri C6 software 1.0, C6; BD Biosciences, Franklin Lakes, NJ, USA).

Bioinformatics analysis and luciferase reporter assay

TargetScan Human 5.1 software (22) (www.targetscan.org) was used to determine the putative target of miR-423-3p. The wild-type (WT) PANX2 3′-untranslated region (UTR) was constructed by PCR, which was performed by Yearthbio (Changsha, China), and inserted into the pMIR-REPORT miRNA Expression Reporter vector (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The mutant type (MT) of PANX2 3′-UTR was constructed using the Easy Mutagenesis System kit (Promega Corporation, Madison, WI, USA), according to the manufacturer's protocol, and then inserted into the pMIR-REPORT miRNA Expression Reporter vector (Thermo Fisher Scientific, Inc.). U251 and U87MG Uppsala cells were co-transfected with WT PANX2-3′UTR plasmid or MT PANX2-3′UTR plasmid, and miR-NC or miR-423-3p mimics, using Lipofectamine 2000 transfection reagent (Thermo Fisher Scientific, Inc.). Following transfection at 37°C for 48 h, the activity of Renilla luciferase and firefly luciferase were determined using a dual-luciferase reporter assay system (Promega Corporation), 48 h after transfection. The activity of Renilla luciferase was normalized to that of the firefly luciferase.

Statistical analysis

Data are expressed as the mean ± standard deviation. The differences between two groups were analyzed using two-tailed Student's t-test. Statistical analysis was performed using SPSS software (version 17.0; SPSS, Inc., Chicago, IL, USA). Pearson's correlation analysis was performed to examine the correlation between the miR-423-3p and PANX2 expression in glioma tissues. Kaplan Meier analysis with log rank tests were used for the survival analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Upregulation of miR-423-3p is associated with glioma progression

In the present study, qPCR was used to determine the expression of miR-423-3p in glioma tissues. Normal brain tissues were used as controls. As indicated in Fig. 1A, miR-423-3p expression levels were significantly increased in glioma tissues compared with normal brain tissues. Furthermore, the expression of miR-423-3p was increased in World Health Organization (WHO) III–IV grade glioma compared with WHO I–II grade glioma (Fig. 1B). These glioma tissues were further divided into two groups, a high miR-423-3p group and a low miR-423-3p group, on the basis of mean expression value. It was observed that high expression of miR-423-3p was significantly associated with an advanced grade of glioma as well as a low Karnofsky performance score (KPS), but not with age and sex (Table I). Furthermore, as presented in Fig. 1C, the patients with glioma with high miR-423-3p expression levels had a shorter survival time, compared with those with low miR-423-3p expression levels. Therefore, miR-423-3p upregulation may contribute to the malignant progression of glioma as well as poor prognosis in patients with glioma.

Knockdown of miR-423-3p decreases U251 and U87MG Uppsala cell proliferation and induces cell apoptosis

As miR-423-3p was upregulated in glioma, glioma U251 and U87MG Uppsala cells were transfected with an miR-423-3p inhibitor in order to knock down miR-423-3p expression. Transfection with an NC inhibitor was used as the control group. Following transfection with miR-423-3p inhibitor, miR-423-3p expression levels were significantly decreased compared with the NC inhibitor group (Fig. 2A and B). An MTT assay further indicated that the inhibition of miR-423-3p led to the decreased proliferation of U251 and U87MG Uppsala cells (Fig. 2C and D). Cell apoptosis was further examined and it was revealed that the knockdown of miR-423-3p resulted in a significant increase in U251 and U87MG Uppsala cell apoptosis (Fig. 2E and F). Therefore, the results of the present study demonstrated that the knockdown of miR-423-3p decreases U251 and U87MG Uppsala cell proliferation and induces cell apoptosis.

PANX2 is a novel target of miR-423-3p in U251 and U87MG Uppsala cells

Bioinformatics analysis indicated that PANX2 was a putative target gene of miR-423-3p (Fig. 3A). To the best of our knowledge, this targeting association has never previously been reported. In the present study, luciferase reporter plasmids were constructed containing WT or MT PANX2 3′-UTR (Fig. 3B and C). The luciferase reporter gene assay was then performed in U251 and U87MG Uppsala cells. The results of the present study indicated that luciferase activity was significantly decreased in U251 and U87MG Uppsala cells co-transfected with the WT PANX2 3′-UTR plasmid and miR-423-3p mimic compared with the control group, which was eliminated during transfection with the MT PANX2 3′-UTR plasmid (Fig. 3D and E). Subsequently, it was revealed that the inhibition of miR-423-3p significantly increased the expression of PANX2 protein in U251 and U87MG Uppsala cells (Fig. 3F and G). Therefore, PANX2 is a potential novel target gene of miR-423-3p in U251 and U87MG Uppsala cells.

Knockdown of PANX2 attenuates the effects of miR-423-3p inhibition on U251 and U87MG Uppsala cells

On the basis of the aforementioned results, it was hypothesized that PANX2 may be involved in miR-423-3p-mediated glioma growth. To investigate this hypothesis, U251 and U87MG Uppsala cells were co-transfected with miR-423-3p inhibitor and PANX2 siRNA, or miR-423-3p inhibitor and NC siRNA. Following transfection, the mRNA and protein levels of PANX2 were significantly decreased in the miR-423-3p inhibitor + PANX2 siRNA group, compared with the miR-423-3p inhibitor + NC siRNA group (Fig. 4A-D). An MTT assay further demonstrated that the proliferation of U251 and U87MG Uppsala cells was significantly increased in the miR-423-3p inhibitor + PANX2 siRNA group, compared with the miR-423-3p inhibitor + NC siRNA group (Fig. 5A and B), indicating that knockdown of PANX2 attenuates the suppressive effects of miR-423-3p inhibition on glioma cell proliferation. Cell apoptosis was then assessed. It was revealed that the apoptosis of U251 and U87MG Uppsala cells was significantly decreased in the miR-423-3p inhibitor + PANX2 siRNA group, compared with the miR-423-3p inhibitor + NC siRNA group (Fig. 5C and D), indicating that the knockdown of PANX2 attenuates the effect of miR-423-3p inhibition on glioma cell apoptosis. As a result, it may be suggested that the knockdown of miR-423-3p at least partially inhibits proliferation and induces the apoptosis of glioma cells, via the direct targeting of PANX2.

PANX2, which is downregulated in glioma, is inversely correlated with the miR-423-3p expression

Finally, RT-qPCR was performed to examine the mRNA expression levels of PANX2 in glioma. The expression of PANX2 mRNA was demonstrated to be significantly decreased in glioma tissues compared with normal brain tissues (Fig. 6A). Furthermore, PANX2 mRNA expression levels in WHO III–IV grade glioma were decreased compared with in WHO I–II grade glioma (Fig. 6B). Notably, the PANX2 mRNA expression levels were inversely correlated with the miR-423-3p expression levels in glioma tissues (Fig. 6C). The decreased expression of PANX2 may be due to the upregulation of miR-423-3p in glioma.

Discussion

The exact function of miR-423-3p in glioma growth as well as the underlying molecular mechanism remain unclear. In the present study, it was demonstrated that miR-423-3p was significantly upregulated in glioma tissues compared with normal brain tissues, and the increased expression of miR-423-3p was significantly associated with an advanced grade of glioma in addition to a poorer prognosis in patients with glioma. Further investigation suggested that miR-423-3p serves a promoting function in glioma growth via the direct targeting of PANX2. Furthermore, PANX2 was significantly downregulated in glioma tissues compared with normal brain tissues, and PANX2 expression levels were inversely correlated with miR-423-3p expression levels in glioma tissues.

miR-423-3p has been demonstrated to serve a promoting function in several types of human cancer (23,24). For example, Guan et al (23) identified that miR-423-3p was upregulated in laryngeal carcinoma cells, and that the inhibition of miR-423-3p resulted in a significant decrease in cell proliferation, clonogenicity, cell migration and invasion. Additionally, miR-423-3p was upregulated in colorectal cancer (CRC), and promoted CRC cell proliferation via enhancing the G1/S transition by targeting p21Cip1/Waf1 (24). Previously, miR-423-5p was demonstrated to be significantly upregulated in glioma, and the overexpression of miR-423-5p promoted glioma cell proliferation, angiogenesis and invasion by increasing the activities of protein kinase B and mitogen-activated protein kinase signaling pathways and suppressing the expression of inhibitor of growth family member 4 (20). In addition, miR-423-5p upregulation enhanced glioblastoma neurosphere formation and rendered glioma cells resistant to temozolomide (20). However, the exact function of miR-423-3p in glioma has not been uncovered. In the present study, it was identified that miR-423-3p was downregulated in glioma tissues compared with normal brain tissues, and its downregulation was associated with an advanced pathological grade and lower KPS in glioma. Furthermore, the patients with glioma with high miR-423-3p levels had a shorter survival time compared with patients with low miR-423-3p levels. As a result, it may be suggested that the upregulation of miR-423-3p contributes to glioma progression and a poorer prognosis in patients with glioma.

PANX2 was also identified as a novel target gene of miR-423-3p using bioinformatics analysis and a luciferase reporter assay, and knockdown of miR-423-3p increased the protein expression levels of PANX2 in U251 and U87MG Uppsala cells. PANX2 encodes the protein pannexin 2, belonging to the innexin family, the members of which are the structural components of gap junctions (25). Previous studies have demonstrated that PANX2 is abundantly expressed in the central neuronal system, and participates in neuronal development and adult neurogenesis (26,27). Furthermore, the upregulation and downregulation of PANX2 are associated with the development and progression of certain diseases, including neoplasms, multiple sclerosis, migraines and hypertension (28). Previously, gene array analysis indicated that there was a significant decrease in PANX2 expression in glioma, and that the decreased expression of PANX2 was associated with a poorer prognosis in patients with glioma (29). Furthermore, the expression of PANX2 was also lower in human glioma cell lines compared with normal brain tissues and astrocytes (29). Consistent with this previous study, the results of the present study identified that PANX2 was downregulated in glioma tissues compared with normal brain tissues, and its expression levels in WHO III–IV grade glioma were decreased compared with those in WHO I–II grade glioma. Furthermore, Lai et al (29) identified that the restoration of PANX2 expression significantly decreased monolayer saturation density and anchorage-independent growth of rat C6 glioma cells in vitro, as well as tumor growth in vivo. However, the regulatory mechanism of PANX2 in glioma remains unknown. In the present study, it was demonstrated that the knockdown of PANX2 attenuated the effects of miR-423-3p inhibition on the proliferation and apoptosis of U251 and U87MG Uppsala cells, suggesting that miR-423-3p promotes glioma cell proliferation by directly targeting PANX2. Furthermore, the expression levels of PANX2 were inversely correlated with the miR-423-3p expression levels in glioma tissues, suggesting that the decreased expression of PANX2 may be caused by the upregulation of miR-423-3p.

In summary, the results of the present study demonstrated that miR-423-3p serves an oncogenic function in glioma cell proliferation by directly targeting PANX2, suggesting that miR-423-3p may be a potential therapeutic target for glioma.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of China (grant no. 81201740) and the Project of Science and Technology Department of the Hunan Province (grant no. 2012FJ6075).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

JXi and JH collected clinical tissues. JXu, HH and RP performed the in vitro experiments. JXi wrote the manuscript. JXu designed the study and revised the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Xiangya Hospital, Central South University (Changsha, China). Written informed consent was obtained from all patients prior to the study.

Consent for publication

Written informed consents for the publication of this data were obtained from all patients in the present study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Goodenberger ML and Jenkins RB: Genetics of adult glioma. Cancer Genet. 205:613–621. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Yan Y and Jiang Y: RACK1 affects glioma cell growth and differentiation through the CNTN2-mediated RTK/Ras/MAPK pathway. Int J Mol Med. 37:251–257. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Marumoto T and Saya H: Molecular biology of glioma. Adv Exp Med Biol. 746:2–11. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Zhang R, Wang R, Chen Q and Chang H: Inhibition of autophagy using 3-methyladenine increases cisplatininduced apoptosis by increasing endoplasmic reticulum stress in U251 human glioma cells. Mol Med Rep. 12:1727–1732. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Zheng K, Liu W, Liu Y, Jiang C and Qian Q: Microrna-133a suppresses colorectal cancer cell invasion by targeting fascin1. Oncol Lett. 9:869–874. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Liang ML, Hsieh TH, Ng KH, Tsai YN, Tsai CF, Chao ME, Liu DJ, Chu SS, Chen W, Liu YR, et al: Downregulation of miR-137 and miR-6500-3p promotes cell proliferation in pediatric high-grade gliomas. Oncotarget. 7:19723–19737. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Xu J, Xu W and Zhu J: Propofol suppresses proliferation and invasion of glioma cells by upregulating microRNA-218 expression. Mol Med Rep. 12:4815–4820. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Liu C, Liang S, Xiao S, Lin Q, Chen X, Wu Y and Fu J: MicroRNA-27b inhibits Spry2 expression and promotes cell invasion in glioma U251 cells. Oncol Lett. 9:1393–1397. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Wang H, Tao T, Yan W, Feng Y, Wang Y, Cai J, You Y, Jiang T and Jiang C: Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma. Sci Rep. 5:130722015. View Article : Google Scholar : PubMed/NCBI

12 

McDaneld TG, Smith TP, Doumit ME, Miles JR, Coutinho LL, Sonstegard TS, Matukumalli LK, Nonneman DJ and Wiedmann RT: MicroRNA transcriptome profiles during swine skeletal muscle development. BMC Genomics. 10:772009. View Article : Google Scholar : PubMed/NCBI

13 

Prats-Puig A, Ortega FJ, Mercader JM, Moreno-Navarrete JM, Moreno M, Bonet N, Ricart W, López-Bermejo A and Fernández-Real JM: Changes in circulating microRNAs are associated with childhood obesity. J Clin Endocrinol Metab. 98:E1655–E1660. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Kumarswamy R, Anker SD and Thum T: MicroRNAs as circulating biomarkers for heart failure: Questions about MiR-423-5p. Circ Res. 106:e82010. View Article : Google Scholar : PubMed/NCBI

15 

Oak SR, Murray L, Herath A, Sleeman M, Anderson I, Joshi AD, Coelho AL, Flaherty KR, Toews GB, Knight D, et al: A micro RNA processing defect in rapidly progressing idiopathic pulmonary fibrosis. PLoS One. 6:e212532011. View Article : Google Scholar : PubMed/NCBI

16 

Xiao B, Wang Y, Li W, Baker M, Guo J, Corbet K, Tsalik EL, Li QJ, Palmer SM, Woods CW, et al: Plasma microRNA signature as a noninvasive biomarker for acute graft-versus-host disease. Blood. 122:3365–3375. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Hui AB, Lenarduzzi M, Krushel T, Waldron L, Pintilie M, Shi W, Perez-Ordonez B, Jurisica I, O'Sullivan B, Waldron J, et al: Comprehensive MicroRNA profiling for head and neck squamous cell carcinomas. Clin Cancer Res. 16:1129–1139. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Lin J, Huang S, Wu S, Ding J, Zhao Y, Liang L, Tian Q, Zha R, Zhan R and He X: MicroRNA-423 promotes cell growth and regulates G(1)/S transition by targeting p21Cip1/Waf1 in hepatocellular carcinoma. Carcinogenesis. 32:1641–1647. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Zhao H, Gao A, Zhang Z, Tian R, Luo A, Li M, Zhao D, Fu L, Fu L, Dong JT and Zhu Z: Genetic analysis and preliminary function study of miR-423 in breast cancer. Tumour Biol. 36:4763–4771. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Li S, Zeng A, Hu Q, Yan W, Liu Y and You Y: miR-423-5p contributes to a malignant phenotype and temozolomide chemoresistance in glioblastomas. Neuro Oncol. 19:55–65. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Arocho A, Chen B, Ladanyi M and Pan Q: Validation of the 2-DeltaDeltaCt calculation as an alternate method of data analysis for quantitative PCR of BCR-ABL P210 transcripts. Diagn Mol Pathol. 15:56–61. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Guan G, Zhang D, Zheng Y, Wen L, Yu D, Lu Y and Zhao Y: microRNA-423-3p promotes tumor progression via modulation of AdipoR2 in laryngeal carcinoma. Int J Clin Exp Pathol. 7:5683–5691. 2014.PubMed/NCBI

24 

Li HT, Zhang H, Chen Y, Liu XF and Qian J: MiR-423-3p enhances cell growth through inhibition of p21Cip1/Waf1 in colorectal cancer. Cell Physiol Biochem. 37:1044–1054. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Tang W, Ahmad S, Shestopalov VI and Lin X: Pannexins are new molecular candidates for assembling gap junctions in the cochlea. Neuroreport. 19:1253–1257. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Swayne LA and Bennett SA: Connexins and pannexins in neuronal development and adult neurogenesis. BMC Cell Biol. 17 Suppl 1:S102016. View Article : Google Scholar

27 

Swayne LA, Sorbara CD and Bennett SA: Pannexin 2 is expressed by postnatal hippocampal neural progenitors and modulates neuronal commitment. J Biol Chem. 285:24977–24986. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Penuela S, Harland L, Simek J and Laird DW: Pannexin channels and their links to human disease. Biochem J. 461:371–381. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Lai CP, Bechberger JF and Naus CC: Pannexin2 as a novel growth regulator in C6 glioma cells. Oncogene. 28:4402–4408. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 16 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu J, He J, Huang H, Peng R and Xi J: MicroRNA‑423‑3p promotes glioma growth by targeting PANX2 Retraction in /10.3892/ol.2021.12833. Oncol Lett 16: 179-188, 2018
APA
Xu, J., He, J., Huang, H., Peng, R., & Xi, J. (2018). MicroRNA‑423‑3p promotes glioma growth by targeting PANX2 Retraction in /10.3892/ol.2021.12833. Oncology Letters, 16, 179-188. https://doi.org/10.3892/ol.2018.8636
MLA
Xu, J., He, J., Huang, H., Peng, R., Xi, J."MicroRNA‑423‑3p promotes glioma growth by targeting PANX2 Retraction in /10.3892/ol.2021.12833". Oncology Letters 16.1 (2018): 179-188.
Chicago
Xu, J., He, J., Huang, H., Peng, R., Xi, J."MicroRNA‑423‑3p promotes glioma growth by targeting PANX2 Retraction in /10.3892/ol.2021.12833". Oncology Letters 16, no. 1 (2018): 179-188. https://doi.org/10.3892/ol.2018.8636