Pro-apoptotic effects of splice-switching oligonucleotides targeting Bcl-x pre-mRNA in human glioma cell lines

  • Authors:
    • Zhaohui Li
    • Qingwei Li
    • Liang Han
    • Nan Tian
    • Qianlei Liang
    • Yanzhe Li
    • Xingli Zhao
    • Chao Du
    • Yu Tian
  • View Affiliations

  • Published online on: November 30, 2015     https://doi.org/10.3892/or.2015.4465
  • Pages: 1013-1019
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Alternative splicing is a near-ubiquitous phenomenon with important roles in human diseases, including cancers. Splice-switching oligonucleotides (SSOs) have emerged as a class of antisense therapeutics that modulate alternative splicing by hybridizing to the pre-mRNA splice site. The Bcl-x gene is alternatively spliced to express anti‑apoptotic Bcl-xL and pro-apoptotic Bcl-xS. Bcl-xL expression is upregulated in many cancers and is considered a general mechanism by which cancer cells evade apoptosis. By redirecting Bcl-x pre-mRNA splicing from Bcl-xL to Bcl-xS, SSO exerted pro-apoptotic and chemosensitizing effects in various cancer cell lines. In this study, we investigated the effects of SSO targeting Bcl-x pre-mRNA in human glioma cell lines. First, we performed reverse transcription-polymerase chain reaction (RT-PCR) and western blotting to determine the mRNA and protein expression levels of Bcl-xL in glioma cell lines (U87 and U251) and a normal human astrocyte cell line (HA1800). Then, the Bcl-x SSO was designed to bind to the downstream 5' alternative splice site of exon 2 in Bcl-x pre-mRNA and was modified using 2'-O-methoxyethyl-phosphorothioate. An oligonucleotide targeting aberrantly spliced human β-globin intron was used as a negative control. The SSOs were delivered with a cationic lipid into glioma and astrocyte cell lines. The antitumor effects of the SSOs were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays and flow cytometry, and the switch in production from Bcl-xL to Bcl-xS was analyzed by RT-PCR and western blotting. Bcl-xL mRNA and protein were highly expressed in both glioma cell lines. The Bcl-x SSO modified Bcl-x pre-mRNA splicing and had pro-apoptotic effects on the glioma cell lines. By contrast, the lipid alone and the control SSO did not affect Bcl-xL expression or induce apoptosis. Our study demonstrated the antitumor activity of an SSO that targets Bcl-x pre-mRNA splicing in glioma cell lines. Bcl-x SSO may be a potential strategy for treating gliomas.

Introduction

Alternative splicing is the process by which splice sites in precursor messenger RNA (pre-mRNA) are differentially selected and paired to produce multiple mature mRNAs and protein isoforms with distinct structural and functional properties. Alternative splicing is a very accurate, efficient, and extraordinarily flexible process that regulates many major aspects of eukaryotic cell biology. Approximately 95% of human genes with multiple exons undergo alternative splicing during pre-mRNA maturation (13). In addition to its role in human proteome diversity, alternative splicing is now accepted to play important roles in human diseases, including diabetes, neurodegenerative diseases, and cancer (46).

Aberrant alternative splicing has two major roles in cancer by promoting the emergence of a cancer-specific isoform or disturbing the balanced expression of normally expressed isoforms in cancer cells (7). During tumor growth and development, and during oncogenesis, cells progress through various stages as they acquire additional oncogenic properties. These stages were described by Hanahan and Weinberg (8) in 2000 and in papers describing the features of cancer, which were updated in 2011 to include ten processes required for tumor development and progression to metastases. These processes were growth factor self-sufficiency; insensitivity to growth inhibitory signals; limitless replicative potential; the ability to evade apoptosis; the ability to sustain angiogenesis; the ability to invade tissues and metastasize; the ability to evade the immune system; the presence of inflammation; the tendency towards genomic instability; and deregulated metabolism. The results of recent studies indicate that alternative splicing regulates many of these processes involved in tumorigenesis and development (917). Accordingly, deregulated alternative splicing is considered to be a key feature of cancer and an opportunity to identify cancer biomarkers (18).

Because of its frequently occurring and important role in cancer, alternative splicing has emerged as an important target for molecular therapies. Therefore, many molecules have been designed and developed in order to inhibit cancer-specific isoforms or isoforms highly expressed in cancer cells, or switch the expression of specific isoforms as cancer cell-specific treatments (19,20). Splice-switching oligonucleotides (SSOs) are antisense oligonucleotides that modify alternative splicing by hybridizing to pre-mRNA sequences, which undergo splicing, and blocking access to the transcript by splicing factors, thereby redirecting the splicing machinery to the alternative pathway. The efficacy of SSOs has been established in various disease models, including β-thalassemia, Duchenne muscular dystrophy, spinal muscular atrophy, inflammatory diseases, and cancer (21,22).

In cancer, the first and most frequently cited demonstration of the antitumor effects of SSOs were of SSOs targeting Bcl-x pre-mRNA. Bcl-x is a member of the Bcl-2 family of proteins, which regulate the permeability of the outer membrane of mitochondria. Bcl-x undergoes splicing at two alternative 5′ splice sites in exon 2, yielding two distinct proteins, Bcl-xL and Bcl-xS, with antagonistic properties (Fig. 1). Bcl-xL exerts anti-apoptotic effects by antagonizing and inhibiting the pro-apoptotic Bax and Bak proteins. Bcl-xL overexpression has been detected in several types of cancer. High Bcl-xL expression is correlated with decreased cellular sensitivity to chemotherapeutic reagents. Bcl-xS has pro-apoptotic effects by directly binding to and inhibiting the pro-apoptotic Bcl-xL and Bcl-2 proteins. High Bcl-xS expression was reported to induce apoptosis in cancer cells from patients with colon or stomach cancers (23,24).

Redirecting the pre-mRNA splicing of Bcl-x from Bcl-xL to Bcl-xS with an SSO had pro-apoptotic and chemosensitizing effects in various cancer cell lines. An SSO targeting Bcl-x pre-mRNA (termed Bcl-x SSO) that blocked the downsteam 5′ alternative splice site in exon 2 of Bcl-x pre-mRNA redirected Bcl-x pre-mRNA splicing from Bcl-xL to Bcl-xS. Accordingly, it increased the expression of pro-apoptotic Bcl-xS and decreasing the expression of anti-apoptotic Bcl-xL in various cancer cell lines. Mercatante et al (25) reported that Bcl-x SSO, which targeted the downstream alternative 5′-splice site in exon 2 of Bcl-x pre-mRNA, shifted splicing from Bcl-xL to Bcl-xS in prostate and breast cancer cells in vitro. They also found that Bcl-xS protein, induced by the SSO, sensitized the cancer cells to treatment with ultraviolet- and γ-irradiation and chemotherapeutic drugs. In other studies, the same group showed that delivery of Bcl-x SSO using a lipid nanoparticle redirected Bcl-x splicing and reduced the tumor burden in melanoma lung metastases in vivo (26,27).

The effects of modifying a target gene's splicing using an SSO vary depending on the expression profile of the target cells. The differences in the cellular responses to Bcl-x SSO-induced modification of Bcl-x pre-mRNA splicing were mainly attributed to the endogenous Bcl-xL expression level. Tumor cells containing higher levels of Bcl-xL were more susceptible to the effects of Bcl-x SSO (9,25). To date, however, the effects of Bcl-x SSO on glioma have not been reported. Previous studies have reported that Bcl-xL is highly expressed in glioma, and confers resistance to chemotherapies (28). Therefore, we hypothesized that Bcl-x SSO can modulate alternative splicing of Bcl-x pre-mRNA and inhibit the growth of glioma cells.

In this study, we examined the effects of Bcl-x SSO on glioma cell lines. First, we measured the endogenous mRNA and protein expression of Bcl-xL in human glioma cell lines and a normal human astrocyte cell line. Then, we determined the effects of Bcl-x SSO on apoptosis and viability of these glioma cell lines. Finally, we measured the shift in expression from Bcl-xL to Bcl-xS in glioma cell lines treated with Bcl-x SSO.

Materials and methods

Ethics

The study was approved by the Ethics Committee of the China-Japan Union Hospital of Jilin University (Changchun, China).

Cell culture

Two human glioma cell lines (U87 and U251) and a normal human astrocyte cell line (HA1800) were purchased from Boster Biological Technology, Ltd. (Wuhan, China). The cell lines were routinely cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), penicillin (100 U/ml), and streptomycin (100 µg/ml) in a 5% CO2 atmosphere at 37°C. DMEM, FBS, and other tissue culture reagents were purchased from Beijing Dingguo Changsheng Biotechnology Co., Ltd. (Beijing, China).

SSO preparation and transfection

2′-O-methoxyethyl-phosphorothioate SSOs were synthesized by Shanghai Sangon Biological Engineering Technology and Services Co., Ltd. (Shanghai, China). The Bcl-x SSO (5′-TGGTTCTTACCCAGCCGCCG-3′) targeted the downstream 5′ alternative splice site of exon 2 of Bcl-x pre-mRNA. An oligonucleotide (5′-GCTATTACCTTAACCCAG-3′) targeting human β-globin pre-mRNA was used as negative control SSO.

The U87 and U251 cells were plated in 6-well culture plates containing antibiotic-free DMEM at a density of 5.0×105 cells/ml. At >60% confluence, the cells were transfected with SSO using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions.

RNA isolation and reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA was isolated from cells using TRIzol reagent (Invitrogen Life Technologies) according to the manufacturer's instructions. Complementary DNA (cDNA) was synthesized using a HiFi-MMLV cDNA kit (Beijing Kang Century Biotechnology Co., Ltd., Beijing, China) and random hexamer primers. The primers were designed using Primer 5 software (Premier Biosoft, Palo Alto, CA, USA) and synthesized by genScript Co., Ltd. (Nanjing, China). The primers 5′-AGCGTAGACAAGGAGATGCAGG-3′ (forward) and 5′-GTGGATGGTCAGTGTCTGGTCA-3′ (reverse) were used to amplify Bcl-xL, and the primers 5′-AGTAAAGCAAGCGCTGAGGGAG-3′ (forward) and 5′-ACTGAAGAGTGAGCCCAGCAGA-3′ (reverse) were used to amplify both Bcl-xL and Bcl-xS. PCR was performed using GoldStar Best DNA Polymerase (Beijing Kang Century Biotechnology Co., Ltd.). The reaction conditions were as follows: 95°C for 10 min followed by 40 cycles of degradation at 94°C for 30 sec, annealing at 58°C for 30 sec, and extension at 72°C for 60 sec, and a final step of 72°C for 10 min.

The PCR products were separated on a 10% non-denaturing polyacrylamide gel (Invitrogen Life Technologies) and bands were visualized on a Typhoon 9400 imager (GE Healthcare, Piscataway, NJ, USA). Images were quantified using imageQuant analysis software (Molecular Dynamics, Sunnyvale, CA, USA). The relative abundance of Bcl-xS in each lane was determined by dividing the intensity of the 250-bp band (Bcl-xS) by the total intensities of the 452-bp (Bcl-xL) and 250-bp (Bcl-xS) bands.

Western blot analysis

Cells in all experimental groups were collected using a cell scraper. Total protein was extracted from cells using protein extraction reagent (Boster Bioengineering, Wuhan, China) containing 1 mM phenylmethanesulfonyl fluoride (PMSF; Roche Molecular Biochemicals, indianapolis, IN, USA). The protein concentration was determined using the BCA protein assay (Nanjing Keygen Biotech Co., Ltd., Nanjing, China). Total protein was electrophoresed on a 15% sodium dodecyl sulfate-polyacrylamide gel and electro-transferred to polyvinylidene difluoride (PVDF) membranes (Pall gelman Laboratory Corporation, Ann Arbor, Mi, USA). The membranes were blocked overnight using 5% skimmed milk powder at 4°C. The membranes were washed in Tris-buffered saline containing Tween-20 (TBST) and incubated for 1 h at room temperature with primary antibodies against target proteins, followed by an additional TBST wash. The membranes were then incubated with appropriate secondary antibodies for 1 h at room temperature and washed again with TBST. The protein bands were detected by enhanced chemiluminescence.

Cell viability assay

The effects of Bcl-x SSO on the viability of human glioma cells were determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. Cells (2×104 cells/well) were cultured in 96-well culture plates for 1 day before SSO transfection. One day later, after the cells had adhered to the culture plates, they were transfected with Bcl-x SSO and control SSO. MTT solution (5 mg/ml, 20 µl; Sigma Chemical Co., St. Louis, MO, USA) was added to each well and the cells were cultured in a CO2 incubator for 4 h. Next, the culture solution was removed and 150 µl of dimethyl sulfoxide was added to each well, and the plates were agitated at room temperature for 10 min. The optical density of each well was measured at 490 nm using a SpectraMax M3 microplate reader (Molecular Devices, Sunnyvale, CA, USA). Each experimental group was prepared in six duplicate wells. The mean values were calculated and growth curves were constructed.

Flow cytometry analysis

Apoptosis was evaluated by flow cytometry analysis (FCM). Cells from all experimental groups were digested in 0.25% trypsin and re-suspended in phosphate-buffered saline to prepare single-cell suspensions. The cell density was adjusted to 1×106 cells/ml. Next, 5 µl of Annexin V-FiTC and 5 µl of propidium iodide were added, and the cells were incubated for 30 min at 4°C before flow cytometry.

Statistical analysis

Data are expressed as the mean ± standard deviation of experiments performed in triplicate. Statistical analysis was performed using one-way analysis of variance (ANOVA) for multiple comparisons and unpaired t-tests for comparisons between pairs of groups. P-values of <0.05 were considered statistically significant.

Results

High expression of Bcl-xL in human glioma cell lines

We first determined the expression profile of Bcl-xL mRNA in two glioma cell lines (U87 and U251) and in a normal astrocyte cell line (HA1800) by RT-PCR. Bcl-xL mRNA expression was significant greater in U87 and U251 cells than in HA1800 (Fig. 2A and B). Likewise, western blot analysis revealed that Bcl-xL protein expression was also increased in U87 and U251 cells (Fig. 2C and D). The results indicate that Bcl-xL mRNA and protein levels are much greater in human glioma cell lines than in the control astrocyte cell line.

Bcl-x SSO inhibited the proliferation of human glioma cell lines

To shift the alternative splicing pathway of Bcl-x pre-mRNA from Bcl-xL to Bcl-xS, the U87 and U251 cells were transfected with Bcl-x SSO, which targeted the downstream 5′ alternative splice site of exon 2, using the cationic lipid Lipofectamine 2000. To investigate whether Bcl-x SSO-mediated splice switching affected cell viability, MTT assays were performed 48 h after transfecting cells with different concentrations of SSO. The results showed that transfection with Bcl-x SSO significantly decreased the viability of U87 and U251 cells compared with untreated cells and cells transfected with a control SSO (Fig. 3). These results indicate that Bcl-x SSO inhibited the proliferation of human glioma cells.

Bcl-x SSO has pro-apoptotic effects on human glioma cell lines

To investigate whether Bcl-x SSO-mediated splice switching has pro-apoptotic effects on human glioma cell lines, flow cytometry was used to determine the rate of apoptosis. Transfection with Bcl-x SSO (200 nM) for 48 h significantly increased the apoptotic rate of U87 and U251 cells. By contrast, the control SSO did not affect the apoptotic rate of either cell line (Fig. 4). These results indicate that Bcl-x SSO has pro-apoptotic effects on human glioma cells.

Bcl-x SSO shifts Bcl-x splicing from Bcl-xL to Bcl-xS in human glioma cell lines

The shift in Bcl-x splicing in human glioma cell lines transfected with Bcl-x SSO (200 nM) for 48 h was determined by RT-PCR using primers to amplify both Bcl-xL and Bcl-xS. RT-PCR analysis of total RNA from U87 and U251 cells at 48 h after transfection showed that Bcl-x SSO caused a significant shift in splicing from the Bcl-xL to Bcl-xS pathway, as indicated by a shift in the ratio of these mRNAs. By contrast, the control SSO did not affect Bcl-x pre-mRNA splicing in either cell line (Fig. 5). Because a shift in the splicing pattern of Bcl-x pre-mRNA from Bcl-xL to Bcl-xS should lead to a change in Bcl-xL and Bcl-xS protein expression, we also conducted western blotting to determine the Bcl-xL and Bcl-xS protein expression levels. Consistent with the results of RT-PCR, the western blot analysis using anti-Bcl-xL and anti-Bcl-xS antibodies revealed a significant reduction in Bcl-xL protein expression and an increase in Bcl-xS protein expression (Fig. 6). Thus, the SSO-induced shift in Bcl-x splicing was confirmed in terms of the mRNA and protein expression levels of Bcl-xL and Bcl-xS.

Discussion

Gliomas are the most aggressive and lethal tumors of the central nervous system. In the past decades, the targeted therapy of glioma has not gained significant breakthrough, mainly due to the lack of an idea molecular target underlying gliomas (29). Alternative splicing is a key process involved in proteomic diversity and is essential for normal cell growth and development. However, deregulation of alternative splicing may occur and is implicated in various human diseases, including cancer. Aberrant alternative splicing is now considered an important event in cancer (16). Recent studies show that alternative splicing also makes important contributions to the genesis and development of gliomas (30,31).

Thus, aberrant alternative splicing has emerged as an important target for molecular therapies, and methods to manipulate alternative splicing are therapeutically valuable. Accordingly, SSOs have been developed to regulate alternative splicing by directing splice site selection. SSO are short oligonucleotides designed to anneal to a specific target region on pre-mRNA to interfere with pre-mRNA splicing. SSOs targeting an exon-intron junction may sterically block access to the splicing machinery, redirecting splicing reaction to an adjacent splicing site. Numerous studies have demonstrated the potential anticancer effects of SSO both in vitro and in vivo (20,32).

In cancer, the first and frequently quoted demonstration of antitumor efficacy of SSO was Bcl-x SSO targeting to Bcl-x pre-mRNA. Bcl-x SSOs block the downstream 5′ alternative splice site in exon 2 of Bcl-x pre-mRNA and thereby redirect Bcl-x pre-mRNA splicing from Bcl-xL to Bcl-xS. Redirection of Bcl-x pre-mRNA splicing from Bcl-xL to Bcl-xS by SSOs had pro-apoptotic and chemosensitizing effects in various cancer cell lines. Mercatante et al were among the first to demonstrate the antitumor effects of SSOs (9). They reported that Bcl-x SSO initiated pro-apoptotic events and promoted cell death by decreasing Bcl-xL expression and increasing Bcl-xS expression in prostate cancer cells and breast cancer cells in vitro. They also reported that Bcl-xS, which was upregulated by SSO, sensitized the cancer cells to irradiation and chemotherapeutic drugs. Soon after, Bauman et al (26,27) demonstrated the in vivo antitumor efficacy of Bcl-x SSO. Using lipid nanoparticles, they administered Bcl-x SSO to a mouse model of metastatic melanoma. Bcl-x SSO efficiently redirected Bcl-x pre-mRNA splicing from Bcl-xL to Bcl-xS, and significantly reduced the tumor burden in mice with rapidly growing and highly tumorigenic lung metastases. However, no studies have investigated the antitumor effects of Bcl-x SSO on glioma cells, until now.

In another study by Mercatante et al (25), they found that the efficiency of splicing modulation and the corresponding antitumor effects of Bcl-x SSO were highly dependent on the expression profile of Bcl-xL. Tumor cells containing higher levels of Bcl-xL were more susceptible to the effects of Bcl-x SSO, which suggests that cancers with high Bcl-xL expression may show the greatest responses to Bcl-x SSO. Previous studies have revealed that Bcl-xL expression was elevated and contributed to chemotherapeutic resistance in glioma (28). In this study, in order to explore whether human gliomas are potential candidates for Bcl-x SSO therapy, we analyzed the mRNA and protein expression levels of Bcl-xL in two human glioma cell lines (U87 and U251) and a normal astrocyte cell line (HA1800). We found that Bcl-xL mRNA and protein expression levels were elevated in both human glioma cell lines, and were significantly higher in these cell lines than in HA1800 cells. Based on the results of previous studies and our results, we speculate that human glioma cell lines are good candidates for Bcl-x SSO therapy, which may modulate alternative splicing of Bcl-x pre-mRNA and inhibit glioma cell growth.

In our study, we designed the Bcl-x SSO to bind to the 5′-splice site of exon 2 in Bcl-x pre-mRNA. We also used a oligonucleotide targeting an aberrantly spliced human β-globin intron as a negative control SSO. The Bcl-x SSO and control SSO were modified using 2′-O-methoxyethyl-phosphorothioate and were delivered into U87 and U251 cells using a cationic lipid. We next examined the cellular effects of Bcl-x SSO on the human glioma cells. In these experiments, administration of Bcl-x SSO significantly reduced the replication rate of human glioma cells, demonstrating that Bcl-x SSO can inhibit the proliferation of glioma cells. We also examined the effects of Bcl-x SSO on cell apoptosis by flow cytometry, and the results revealed that Bcl-x SSO enhanced glioma cell apoptosis, whereas the control SSO had no effects on apoptosis. Finally, we determined the effects of Bcl-x SSO on switching the splicing from Bcl-xL to Bcl-xS in terms of the RNA and protein expression levels of both isoforms. RT-PCR and western blotting revealed that administration of Bcl-x SSO significantly reduced the mRNA and protein expression levels of Bcl-xL, and correspondingly increased the mRNA and protein expression levels of Bcl-xS. In other words, by targeting the alternative splicing of Bcl-x pre-mRNA, Bcl-x SSO reduced Bcl-xL expression and increased Bcl-xS expression at the mRNA and protein levels, and thereby promoted cancer cell apoptosis. Our data are consistent with the predicted mechanism of action of SSOs.

Unlike siRNA and conventional antisense oligonucleotides (ASOs), which degrade RNA via RNA-induced silencing complex (RISC) and RNase H-mediated cleavage, respectively, SSOs block sequences in pre-mRNA without causing RNA degradation. To achieve this, the SSO forms a very stable duplex with its pre-mRNA target sequence. This process can be encouraged by chemically modifying the oligonucleotide sugar-phosphate backbone to improve binding affinity and avoid RNase H cleavage, for example. The oligonucleotide sugar-phosphate backbone can be modified using 2′-O-methyl, 2′-O-methoxyethyl, phosphorodiamidate morpholino oligomers, and peptide nucleic acids (33,34). In our study, we modified the Bcl-xL SSO with 2′-O-methoxyethyl-phosphorothioate.

Two features of splicing modulation that distinguish this approach from downregulation of anti-apoptotic genes using ASO or siRNA are worth mentioning. First, Bcl-x SSO, which shifts the splicing of Bcl-x pre-mRNA from Bcl-xL to Bcl-xS, should be superior to Bcl-xL ASO and siRNA, which down-regulate Bcl-xL mRNA expression. This is because Bcl-x SSO decreases Bcl-xL expression and concomitantly increases the expression of the antagonistic Bcl-xS, and thus amplifies the biological effects of the treatment. Second, Bcl-x SSO shows good specificity because its splicing modulation efficiency and antitumor effect are dependent on the expression of Bcl-xL. These properties suggest that cells showing higher Bcl-x expression are more susceptible to the effects of SSO, by greater upregulation of anti-apoptotic Bcl-xS expression. Accordingly, cells from aggressive cancers with higher Bcl-xL expression levels are likely to be more susceptible to SSO-induced apoptosis than healthy untransformed cells. This counterintuitive mechanism should also help to reduce the undesirable side effects associated with established chemotherapeutic drugs. Accordingly, cancers displaying high Bcl-xL expression and those that depend on Bcl-xL expression for survival represent good candidates for treatment with an SSO targeting Bcl-x. By contrast, ASOs and siRNA are less effective in cells showing high expression of the target gene owing to incomplete inhibition of the target gene in such cells (35,36). Here, we showed that Bcl-xL is highly expressed in human glioma cells, and we proposed the hypothesis that human glioma cells are susceptible to Bcl-xL SSO. We next confirmed that Bcl-x SSO modulates Bcl-x pre-mRNA splicing and has marked pro-apoptotic effects in human glioma cell lines. We also found that Bcl-xL expression was significantly greater in the glioma cell lines than in the normal human astrocyte cell line. Accordingly, we may be able to find a suitable dosage of Bcl-x SSO that has marked antitumor effects on glioma cells without causing undesirable side effects on normal cells.

In conclusion, the present study was the first to explore the pro-apoptotic effects of Bcl-x SSO on glioma cell lines. The results showed that Bcl-x SSO modulated the alternative splicing of Bcl-x pre-mRNA from Bcl-xL to Bcl-xS in glioma cell lines. Bcl-x SSO had antitumor effects by inducing apoptosis and cell death in human glioma cell lines in vitro. These observations indicate that Bcl-x SSO may represent an efficient gene therapy for gliomas.

Acknowledgments

The authors thank Professor Chunlei Fan for technical assistance. The present study was supported by grants from the National Science Foundation of China (no. 30672159) and the Program for New Century Excellent Talents in Chinese Universities (NCET-06-0306).

Abbreviations:

pre-mRNA

precursor messenger RNA

SSO

splice-switching oligonucleotides

PCR

polymerase chain reaction

RT-PCR

reverse transcription PCR

References

1 

Nilsen TW and Graveley BR: Expansion of the eukaryotic proteome by alternative splicing. Nature. 463:457–463. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Gamazon ER and Stranger BE: Genomics of alternative splicing: Evolution, development and pathophysiology. Hum Genet. 133:679–687. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, et al: The landscape of somatic copy-number alteration across human cancers. Nature. 463:899–905. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Kalsotra A and Cooper TA: Functional consequences of developmentally regulated alternative splicing. Nat Rev Genet. 12:715–729. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Barta A and Schümperli D: Editorial on alternative splicing and disease. RNA Biol. 7:388–389. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Oltean S and Bates DO: Hallmarks of alternative splicing in cancer. Oncogene. 33:5311–5318. 2014. View Article : Google Scholar

7 

Pal S, Gupta R and Davuluri RV: Alternative transcription and alternative splicing in cancer. Pharmacol Ther. 136:283–294. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Mercatante DR, Bortner CD, Cidlowski JA and Kole R: Modification of alternative splicing of Bcl-x pre-mRNA in prostate and breast cancer cells. Analysis of apoptosis and cell death. J Biol Chem. 276:16411–16417. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Palve V, Mallick S, Ghaisas G, Kannan S and Teni T: Overexpression of Mcl-1L splice variant is associated with poor prognosis and chemoresistance in oral cancers. PLoS One. 9:e1119272014. View Article : Google Scholar : PubMed/NCBI

11 

Vegran F, Boidot R, Oudin C, Riedinger JM and Lizard-Nacol S: Distinct expression of Survivin splice variants in breast carcinomas. Int J Oncol. 27:1151–1157. 2005.PubMed/NCBI

12 

Bojesen SE, Pooley KA, Johnatty SE, Beesley J, Michailidou K, Tyrer JP, Edwards SL, Pickett HA, Shen HC, Smart CE, et al Australian Cancer Study; Australian Ovarian Cancer Study; Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer (kConFab); Gene Environment interaction and Breast Cancer (GENICA); Swedish Breast Cancer Study (SWE-BRCA); Hereditary Breast and Ovarian Cancer Research group Netherlands (HEBON); Epidemiological study of BRCA1 and BRCA2 Mutation Carriers (EMBRACE); Genetic Modifiers of Cancer Risk in BRCA1/2 Mutation Carriers (GEMO): Multiple independent variants at the TERT locus are associated with telomere length and risks of breast and ovarian cancer. Nat Genet. 45:371–384. e1–e2. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Babic I, Anderson ES, Tanaka K, Guo D, Masui K, Li B, Zhu S, Gu Y, Villa GR, Akhavan D, et al: EGFR mutation-induced alternative splicing of Max contributes to growth of glycolytic tumors in brain cancer. Cell Metab. 17:1000–1008. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Singh A, Karnoub AE, Palmby TR, Lengyel E, Sondek J and Der CJ: Rac1b, a tumor associated, constitutively active Rac1 splice variant, promotes cellular transformation. Oncogene. 23:9369–9380. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Mayer S, Hirschfeld M, Jaeger M, Pies S, Iborra S, Erbes T and Stickeler E: RON alternative splicing regulation in primary ovarian cancer. Oncol Rep. 34:423–430. 2015.PubMed/NCBI

16 

Matsuyama M, Chijiwa T, Inoue Y, Abe Y, Nishi M, Miyazaki N, Furukawa D, Mukai M, Suemizu H, Sekido Y, et al: Alternative splicing variant of vascular endothelial growth factor-A is a critical prognostic factor in non-small cell lung cancer. Oncol Rep. 22:1407–1413. 2009.PubMed/NCBI

17 

David CJ, Chen M, Assanah M, Canoll P and Manley JL: HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 463:364–368. 2010. View Article : Google Scholar :

18 

Kim YJ and Kim HS: Alternative splicing and its impact as a cancer diagnostic marker. Genomics Inform. 10:74–80. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Schuster J, Lai RK, Recht LD, Reardon DA, Paleologos NA, Groves MD, Mrugala MM, Jensen R, Baehring JM, Sloan A, et al: A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: The ACT III study. Neuro Oncol. 17:854–861. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Chen J and Weiss WA: Alternative splicing in cancer: Implications for biology and therapy. Oncogene. 34:1–14. 2015. View Article : Google Scholar

21 

Bonomi S, Gallo S, Catillo M, Pignataro D, Biamonti G and Ghigna C: Oncogenic alternative splicing switches: Role in cancer progression and prospects for therapy. Int J Cell Biol. 2013:9620382013. View Article : Google Scholar : PubMed/NCBI

22 

Kinali M, Arechavala-Gomeza V, Feng L, Cirak S, Hunt D, Adkin C, Guglieri M, Ashton E, Abbs S, Nihoyannopoulos P, et al: Local restoration of dystrophin expression with the morpholino oligomer AVi-4658 in Duchenne muscular dystrophy: A single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol. 8:918–928. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Moore MJ, Wang Q, Kennedy CJ and Silver PA: An alternative splicing network links cell-cycle control to apoptosis. Cell. 142:625–636. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Miura K, Fujibuchi W and Unno M: Splice variants in apoptotic pathway. Exp Oncol. 34:212–217. 2012.PubMed/NCBI

25 

Mercatante DR, Mohler JL and Kole R: Cellular response to an antisense-mediated shift of Bcl-x pre-mRNA splicing and anti-neoplastic agents. J Biol Chem. 277:49374–49382. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Bauman JA, Li SD, Yang A, Huang L and Kole R: Anti-tumor activity of splice-switching oligonucleotides. Nucleic Acids Res. 38:8348–8356. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Bauman J, Jearawiriyapaisarn N and Kole R: Therapeutic potential of splice-switching oligonucleotides. Oligonucleotides. 19:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Weiler M, Bähr O, Hohlweg U, Naumann U, Rieger J, Huang H, Tabatabai G, Krell HW, Ohgaki H, Weller M, et al: BCL-xL: Time-dependent dissociation between modulation of apoptosis and invasiveness in human malignant glioma cells. Cell Death Differ. 13:1156–1169. 2006. View Article : Google Scholar

29 

Alexandru-Abrams D, Jadus MR, Hsu FP, Stathopoulos A and Bota DA: Therapeutic targeting of malignant glioma. Anticancer Agents Med Chem. 14:1075–1084. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Li Z, Tian Y, Tian N, Zhao X, Du C, Han L and Zhang H: Aberrant alternative splicing pattern of ADAR2 downregulates adenosine-to-inosine editing in glioma. Oncol Rep. 33:2845–2852. 2015.PubMed/NCBI

31 

Fontana L, Rovina D, Novielli C, Maffioli E, Tedeschi G, Magnani I and Larizza L: Suggestive evidence on the involvement of polypyrimidine-tract binding protein in regulating alternative splicing of MAP/microtubule affinity-regulating kinase 4 in glioma. Cancer Lett. 359:87–96. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Disterer P, Kryczka A, Liu Y, Badi YE, Wong JJ, Owen JS and Khoo B: Development of therapeutic splice-switching oligo-nucleotides. Hum Gene Ther. 25:587–598. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Sazani P, Astriab-Fischer A and Kole R: Effects of base modifications on antisense properties of 2′-O-methoxyethyl and PNA oligonucleotides. Antisense Nucleic Acid Drug Dev. 13:119–128. 2003. View Article : Google Scholar

34 

Adkin C, Fletcher S and Wilton SD: Optimizing splice-switching oligomer sequences using 2′-O-methyl phosphorothioate chemistry. Methods Mol Biol. 867:169–188. 2012. View Article : Google Scholar

35 

Wan J, Bauman JA, Graziewicz MA, Sazani P and Kole R: Oligonucleotide therapeutics in cancer. Cancer Treat Res. 158:213–233. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Shchelkunova A, Ermolinsky B, Boyle M, Mendez I, Lehker M, Martirosyan KS and Kazansky AV: Tuning of alternative splicing - switch from proto-oncogene to tumor suppressor. Int J Biol Sci. 9:45–54. 2013. View Article : Google Scholar :

Related Articles

Journal Cover

February-2016
Volume 35 Issue 2

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
Li Z, Li Q, Han L, Tian N, Liang Q, Li Y, Zhao X, Du C and Tian Y: Pro-apoptotic effects of splice-switching oligonucleotides targeting Bcl-x pre-mRNA in human glioma cell lines. Oncol Rep 35: 1013-1019, 2016
APA
Li, Z., Li, Q., Han, L., Tian, N., Liang, Q., Li, Y. ... Tian, Y. (2016). Pro-apoptotic effects of splice-switching oligonucleotides targeting Bcl-x pre-mRNA in human glioma cell lines. Oncology Reports, 35, 1013-1019. https://doi.org/10.3892/or.2015.4465
MLA
Li, Z., Li, Q., Han, L., Tian, N., Liang, Q., Li, Y., Zhao, X., Du, C., Tian, Y."Pro-apoptotic effects of splice-switching oligonucleotides targeting Bcl-x pre-mRNA in human glioma cell lines". Oncology Reports 35.2 (2016): 1013-1019.
Chicago
Li, Z., Li, Q., Han, L., Tian, N., Liang, Q., Li, Y., Zhao, X., Du, C., Tian, Y."Pro-apoptotic effects of splice-switching oligonucleotides targeting Bcl-x pre-mRNA in human glioma cell lines". Oncology Reports 35, no. 2 (2016): 1013-1019. https://doi.org/10.3892/or.2015.4465