Phosphorothioate‑modified antisense oligonucleotides against human telomerase reverse transcriptase sensitize cancer cells to radiotherapy

  • Authors:
    • Fei Cao
    • Xiaoping Ju
    • Di Chen
    • Lingong Jiang
    • Xiaofei Zhu
    • Shuiwang Qing
    • Fang Fang
    • Yuxin Shen
    • Zhen Jia
    • Huojun Zhang
  • View Affiliations

  • Published online on: June 14, 2017     https://doi.org/10.3892/mmr.2017.6778
  • Pages: 2089-2094
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Emergence of resistance, unavoidable systemic toxicity and unsatisfactory efficacy arethe main obstacles for traditional cancer therapy. Combination with phosphorothioate modified antisense oligonucleotides (PS‑ASODN) against human telomerase reverse transcriptase (hTERT) may enhance the therapeutic effect of irradiation. However, the effect of PS‑ASODN against hTERT on the anti‑tumor effects of irradiation in liver cancer remain unclear. In the current study, Walker 256 cells were transfected with hTERT PS‑ASODN. Cell proliferation and cell viability were measured using the MTT assay and cell senescence was examined by SA‑β‑gal staining. Telomerase activity was determined by telomeric repeat amplification protocol‑polymerase chain reaction‑ELISA. Cell apoptosis was assayed by flow cytometry and DNA damage was determined by the comet assay.The PS‑ASODN was demonstrated to have an inhibitory effect on cell proliferation and accelerated effect on cell senescence by inhibiting telomerase activity. PS‑ASODN promoted the irradiation‑induced inhibition of cell viability and telomerase activity, and irradiation‑induced DNA damage and cell apoptosis via the activation of apoptosis‑associated proteins. Taken together, these results indicated that combined treatment of PS‑ASODN with irradiation significantly enhanced tumor inhibition. Therefore, PS‑ASODN provides an experimental foundation for gene therapy and is proposed for application in clinical treatment of liver cancer combined with radiotherapy.

Introduction

Cancerhas a large influence on human health and economy. Despite the fact that progress has been made in understanding cancer, it remains one of the leading causes of mortality (1,2). Traditional cancer therapies including surgery, radiotherapy and chemotherapy, aim to destroy the tumor and leave normal tissue intact as much as is possible (3). However, the emergence of resistance to these therapies remains a challenge for successful treatment of cancer. Given the unsatisfactory efficacy and unavoidable systemic toxicity of traditional treatment, gene therapy has been investigated as a gene transcription and translation intervention, serving important roles for tumor growth, spread, survival and therapy resistance (4).

As a special type of RNA nuclear protease, telomerase synthesizes telomeric DNA sequences that provide tandem GT-rich repeats (TTAGGG) to compensate for telomere shortening (5,6). Activated telomerase can be observed in more than 90% of cancer cases and beconsidered to permit tumor cell immortalization and promote cell malignant transformation (7,8). Therefore, telomerase can be used as a potential target for gene therapy.

As the most important regulator of telomerase activity, human telomerase reverse transcriptase (hTERT) is a catalytic subunit of telomerase and processes telomere ends, which are over expressed in greater than 90% of tumor cells and contribute to tumor cell proliferation (8). Previous studies have demonstrated that antisense oligodeoxynucleotide (ASODN) gene therapy against hTERT may effectively inhibit telomerase activity and tumor growth (9,10).

In the current study, a phosphorothioate ASODN (PS-ASODN) against hTERT was used to treat Walker 256 cells. The inhibitory effect and the mechanism of the PS-ASODN were explored in the Walker 256 cells to provide novel strategies for cancer gene therapy.

Materials and methods

Cell culture

Walker 256 cells were purchased from Shanghai Institute of Pharmaceutical Industry (Shanghai, China) and cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) that contained 10% (v/v) fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin, within a humidified at mosphere containing 5% CO2 at 37°C.

Cell transfection

The hTERT-targeted PS-ASODN was purchased from Shanghai Biotechnology Corporation (Shanghai, China). Walker 256 cells were transiently transfected with hTERT-targeted PS-ASODN using Lipofectamine® 2000 Transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.).

Cell proliferation and viability assay

Walker 256 cells were seeded onto 96-cell plates and incubated at 37°C overnight. For the cell proliferation assay, cells were treated with PS-ASODN at a final concentration of 0, 1, 2, 3, 4 and 5 µmol/l for 1, 3 and 7 days, respectively. For cell viability assay, cells were treated with 5 µmol/l PS-ASODN for 24 h and then irradiation (IR) was performed by using an RS 2000 X-ray Biological Irradiator (Rad Source, Suwanee, GA, USA) at doses of 0, 2, 4, 6, and 8 Gy for 24 h. MTT solution (20 µl) was added into each well and the plates were incubated for 4 h at 37°C. Then cells were exposed to 150 µl DMSO and incubated for 10 min. The optical density at 490 nm was read by an enzyme-linked immunosorbent assay (ELISA) reader.

Senescence-associated β-gal staining

Walker 256 cells were treated with 5 µmol/l PS-ASODN for 5 days. Senescence-associated β-gal (SA-β-gal) activity was performed using SA-β-gal staining kit (BeyotimeInstitute of Biotechnology, Haimen, China). Briefly, following removal of the RPMI-1640 medium, Walker 256 cells were washed with PBS and fixed with 2% form aldehyde and 0.2% glutaraldehyde for 10 min. Subsequently, cells were incubated with fresh SA-β-gal stain solution overnight at 37°C (without CO2) following washing with PBS. After staining, cells were photographed using a microscope (IX73; Olympus Corporation, Tokyo, Japan), and the percentage of senescence cell was determined via counting five random fields.

Telomerase activity assay

Walker 256 cells were lysed with ice-cold lysis buffer (Takara Bio, Inc., Otsu, Japan) and centrifuged at 12,000 × g for 30 min at 4°C. The Bicinchoninic Acid (BCA) Protein Assay kit (Takara Bio, Inc.) was used to measure protein concentration. Telomerase activity assay was measured using the telomeric repeat amplification protocol (TRAP) with the TeloTAGGG Telomerase PCR ELISA kit (Roche Diagnostics, Basel, Switzerland) according to the manufacturer's instructions.

Cell apoptosis assay

Walker 256 cells were treated with PS-ASODN at final concentrations of 0, 1, 3 and 5 µmol/l for 24 h, respectively, then IR was performed using the RS 2000 X-ray Biological Irradiator at doses of 6 Gy for 24 h. Cell apoptosis following radiation treatment was determined using the Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptotic cell detection kit (BD Biosciences, Franklin Lakes, NJ, USA) according to manufacturer's protocol. The percentage of apoptotic cells was determined via flow cytometry based on Annexin V/PI stain.

Western blot analysis

Walker 256 cells were treated with PS-ASODN at final concentrations of 0, 1, 3 and 5 µmol/l for 24 h, respectively, then IR was performed using the RS 2000 X-ray Biological Irradiator at doses of 6 Gy for 24 h. Cells were collected and lysed by ice-cold lysis buffer (Takara Bio, Inc.). Protein concentrations were determined using the BCA Protein Assay kit (Takara Bio, Inc.). Then, protein extracts were separated using 12% SDS-PAGE and transferred onto polyvinylidene difluoride (PVDF) membranes. Following incubated with 5% nonfat milk, the membranes were incubated with polyclonal rabbit anti-caspase 3 (1:1,000; cat. no. ab13585; Abcam, Cambridge, MA, USA) or polyclonal rabbit anti-caspase 9 antibodies (1:1,000; cat. no. 9502; Cell Signaling Technology, Inc., Danvers, MA, USA) overnight at 4°C. Membranes were then incubated with horseradish peroxidase-conjugated secondary antibodies (1:5,000; cat. no. ab181658; Abcam) for 1 h at room temperature. Finally, the membranes were visualized using ECL Chemiluminescent Substrate Reagent kit (Thermo Fisher Scientific, Inc.).

Comet assay

DNA damage was determined using a comet assay. Walker 256 cells were treated with PS-ASODN at final concentrations of 0, 1, 3 and 5 µmol/l for 24 h, respectively, then IR was performed using the RS 2000 X-ray Biological Irradiator at doses of 6 Gy for 24 h. Cells were collected and mixed with 0.5 % low melting agarose. Following layeringonto agarose-coated slides, the slides were immersed in lysis buffer overnight at 4°C. Subsequently, slides were incubated in electrophoresis buffer for 10 h. Following electrophoresis, slides were neutralized with 0.4 mol/l Tris-HCl buffer (pH=7.5) twice per 5 min and then immersed in 100% ethanol for 20 min. After being air-dried, slides were stained with 2 µg/ml ethidium bromide for 15 min. Finally, slides were observed under a confocal microscope.

Statistical analysis

Data were expressed as the mean ± standard deviation. GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA) was used for the statistical tests. The data were compared between two groups using the two-tailed Student's t-test and between multiple groups using two-way analysis of variance followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Inhibition of PS-ASODN on Walker 256 cell proliferation

The proliferation of Walker 256 cells was inhibited by treating with PS-ASODN (1–5 µM) on days 0, 1, 3 and 7 (Fig. 1). The proliferation rate between the PS-ASODN group and control group was significant. The proliferation rate decreased as the time and PS-ASODN concentration increased, which indicated that PS-ASODN inhibited Walker 256 cells proliferation in a dose- and time-dependent manner.

PS-ASODN induced acceleration of cell senescence by inhibiting telomerase activity

SA-β-gal commonly acts as a biomarker of replicative senescence due to its overexpression in pre-senescent and senescent cells. In order to study the effect of PS-ASODN on senescence of Walker 256 cells, SA-β-gal activity was measured after treating Walker 256 cells with PS-ASODN (5 µmol/l) for 5 days. As presented in Fig. 2A, PS-ASODN accelerated cell senescence.

Telomere shortening and the accumulation of dysfunctional telomeres are associated with cell senescence (11,12). Therefore, following treatment of Walker 256 cells with PS-ASODN (5 µmol/l), the effect of PS-ASODN on telomerase activity was determined. As presented in Fig. 2B, PS-ASODN resulted in a time-dependent decrease in telomerase activity, indicating that PS-ASODN promoted cell senescence by inhibiting telomerase activity.

Enhancement of PS-ASODN on cell sensitivity to IR

In order to evaluate the effect of PS-ASODN on radio sensitization of cells, the cell viability was examined by MTT assay. As presented in Fig. 3, PS-ASODN enhanced the inhibition of IRon cell viability. In addition, PS-ASODN was also identified to improve the inhibition of IR on telomerase activity (Fig. 4).

The apoptosis of Walker 256 cells was further analyzed by flow cytometry. Compared with the control group, the apoptosis rate of PS-ASODN treatment group increased significantly (Fig. 5A), indicating that PS-ASODN may induce cell apoptosis. Compared with the IR treatment group, the apoptosis rate of Walker 256 cells treated with PS-ASODN and IR increased as the PS-ASODN concentration increased (Fig. 5A), suggesting that PS-ASODN promoted IR-induced cell apoptosis. In addition, the results of western blotting indicated that combined treatment of PS-ASODN with IR resulted in increased protein expression levels of cleaved caspase-3 and cleaved caspase-9 in the Walker 256 cells (Fig. 5B), thus indicating that combined treatment of PS-ASODN with IR promoted activation of cleaved caspase-3/9 in the apoptotic signaling pathway.

To confirm the effect of PS-ASODN on IR-induced DNA damage, the comet assay was performed to assess degree of DNA damage. Compared with IR treatment group, the DNA damage of Walker 256 cells treated with PS-ASODN and IR increased as the PS-ASODN concentration increased at 5 µmol/l (Fig. 6), suggesting that PS-ASODN enhanced IR-induced DNA damage.

Taken together, these observations suggested that effect of PS-ASODN may improve Walker 256 cell sensitivity to IR.

Discussion

As a special type of RNA nuclear protease, telomerase maintains the function of the telomere and servesan important role in cell senescence and carcinogenesis. It was reported that high telomerase activity has been identifiedin malignant tumors, and low telomerase activity has been identifiedin the majority of normal tissues and benign tumors (13). hTERT is a subunit of telomerase and its expression levels are indicated to be associated with telomerase activity (1416). Therefore, it is suggested that hTERT may be an appropriate target for anti-sense therapy.

ASODN has become a research focus due to its high specificity, targeting and low toxicity, and may be applied to gene therapy (7). ASODNs are short DNA sequences that hybridize to complementary mRNA sequences based on Watson-Crick base pairing. ASODN gene therapy has been identifiedto effectively target the expression of genes involved in causing cancer, tumor growth inhibition, radiosensitization or chemosensitization (1720). However, because ASODNs exhibit poor solubility, low affinity for their target complementary RNA sequences (21), phosphorothioate modification was adopted for ASODN in the current study. PS-ASODN presents with good aqueous solubility, increased resistance to degradation of oligodeoxynucleotide by nuclease, increased stability and liposome encapsulation (7,22). Previous studies have demonstrated that PS-ASOND suppressed the activity of telomerase and tumor growth (2327).

In the current study, PS-ASODN was demonstrated to have an inhibitory effect on cell proliferation in a time-dependent manner. PS-ASODN accelerated cell senescence and resulted in a time-dependent decrease in telomerase activity, indicating that PS-ASODN improved cell senescence by suppressing telomerase activity. Through treating Walker 256 cells with PS-ASODN and IR in combination, it was identified that PS-ASODN may enhance the inhibition of IRon cell viability and telomerase activity. Besides, PS-ASODN also promoted the induction of IRon cell apoptosis by activating apoptosis-associated proteins, and enhanced the induction of IRon DNA damage.

In conclusion, it is suggested that hTERT PS-ASODN, the inhibitor of telomerase activity, can inhibit cell proliferation, accelerate cell senescence by down-regulating telomerase activity, and enhance the inhibition of IRon cell viability and induction of IRon cell apoptosis and DNA damage, thus resulting in increased sensitivity to IR. These observations suggest that combination of PS-ASODN and IR is of significance in providing an experimental foundation for gene therapy and guiding the radio therapeutic strategy for cancer treatment.

Acknowledgements

The current study was supported by the National Natural Science Foundation of China (grant no. 81171435).

References

1 

Jemal A, Center MM, DeSantis C and Ward EM: Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev. 19:1893–1907. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Lebedeva IV and Stein CA: Antisense oligonucleotides in cancer: Recent advances. BioDrugs. 13:195–216. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Walther W and Schlag PM: Current status of gene therapy for cancer. Curr Opin Oncol. 25:659–664. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Nittis T, Guittat L and Stewart SA: Alternative lengthening of telomeres (ALT) and chromatin: Is there a connection? Biochimie. 90:5–12. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Rao YK, Kao TY, Wu MF, Ko JL and Tzeng YM: Identification of small molecule inhibitors of telomerase activity through transcriptional regulation of hTERT and calcium induction pathway in human lung adenocarcinoma A549 cells. Bioorg Med Chem. 18:6987–6994. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Fan XK, Yan RH, Li BJ, Chen XM, Wei L and Wang Z: Antisense oligodeoxynucleotide against human telomerase reverse transcriptase inhibits the proliferation of Eca-109 esophageal carcinoma cells. Exp Ther Med. 8:1247–1252. 2014.PubMed/NCBI

8 

Ponnala S, Chetty C, Veeravalli KK, Dinh DH, Klopfenstein JD and Rao JS: MMP-9 silencing regulates hTERT expression via β1 integrin-mediated FAK signaling and induces senescence in glioma xenograft cells. Cell Signal. 23:2065–2075. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Folini M, Brambilla C, Villa R, Gandellini P, Vignati S, Paduano F, Daidone MG and Zaffaroni N: Antisense oligonucleotide-mediated inhibition of hTERT, but not hTERC, induces rapid cell growth decline and apoptosis in the absence of telomere shortening in human prostate cancer cells. Eur J Cancer. 41:624–634. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Kraemer K, Fuessel S, Schmidt U, Kotzsch M, Schwenzer B, Wirth MP and Meye A: Antisense-mediated hTERT inhibition specifically reduces the growth of human bladder cancer cells. Clin Cancer Res. 9:3794–3800. 2003.PubMed/NCBI

11 

Sekoguchi S, Nakajima T, Moriguchi M, Jo M, Nishikawa T, Katagishi T, Kimura H, Minami M, Itoh Y, Kagawa K, et al: Role of cell-cycle turnover and oxidative stress in telomere shortening and cellular senescence in patients with chronic hepatitis C. J Gastroenterol Hepatol. 22:182–190. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Kaul Z, Cesare AJ, Huschtscha LI, Neumann AA and Reddel RR: Five dysfunctional telomeres predict onset of senescence in human cells. EMBO Rep. 13:52–59. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL and Shay JW: Specific association of human telomerase activity with immortal cells and cancer. Science. 266:2011–2015. 1994. View Article : Google Scholar : PubMed/NCBI

14 

Sekaran VG, Soares J and Jarstfer MB: Structures of telomerase subunits provide functional insights. Biochim Biophys Acta. 1804:1190–1201. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Donate LE and Blasco MA: Telomeres in cancer and ageing. Philos Trans R Soc Lond B Biol Sci. 366:76–84. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Zvereva MI, Shcherbakova DM and Dontsova OA: Telomerase: Structure, functions, and activity regulation. Biochemistry (Mosc). 75:1563–1583. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Agrawal A, Dang S and Gabrani R: Recent patents on anti-telomerase cancer therapy. Recent Pat Anticancer Drug Discov. 7:102–117. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Nakhlband A, Barar J, Bidmeshkipour A, Heidari HR and Omidi Y: Bioimpacts of anti epidermal growth factor receptor antisense complexed with polyamidoamine dendrimers in human lung epithelial adenocarcinoma cells. J Biomed Nanotechnol. 6:360–369. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Zhao FJ, Zhang SL, Ma L, Gao H and Zong ZH: Inhibitory effects of c-erbB-2 antisense oligonucleotide transfection on uterine endometrial cancer Ishikawa cell lines. Eur J Gynaecol Oncol. 30:54–59. 2009.PubMed/NCBI

20 

Loriot Y, Mordant P, Brown BD, Bourhis J, Soria JC and Deutsch E: Inhibition of BCL-2 in small cell lung cancer cell lines with oblimersen, an antisense BCL-2 oligodeoxynucleotide (ODN): In vitro and in vivo enhancement of radiation response. Anticancer Res. 30:3869–3878. 2010.PubMed/NCBI

21 

Zhang KZ, Xu JH, Huang XW, Wu LX, Su Y and Chen YZ: Curcumin synergistically augments bcr/abl phosphorothioate antisense oligonucleotides to inhibit growth of chronic myelogenous leukemia cells. Acta Pharmacol Sin. 28:105–110. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Yuan Y, Cai H, Yang XJ, Li W, He J, Guo TK and Chen YR: Liposome-mediated induction of apoptosis of human hepatoma cells by c-myc antisense phosphorothioate oligodeoxynucleotide and 5-fluorouracil. Asian Pac J Cancer Prev. 15:5529–5533. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Svinareva LV, Glukhov AI, Moskaleva EY and Shvets VI: Effect of modified DNA and RNA oligonucleotides on telomerase activity and tumor cell survival in vitro. Appl Biochem Micro. 47:718–722. 2011. View Article : Google Scholar

24 

Wang XS, Wang K, Li X and Fu SB: Effects of phosphorothioate anti-sense oligodeoxynucleotides on colorectal cancer cell growth and telomerase activity. World J Gastroenterol. 10:3455–3458. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Gao XD and Chen YR: Inhibition of telomerase with human telomerase reverse transcriptase antisense increases the sensitivity of tumor necrosis factor-alpha-induced apoptosis in prostate cancer cells. Asian J Androl. 9:697–704. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Ji XM, Xie CH, Fang MH, Zhou FX, Zhang WJ, Zhang MS and Zhou YF: Efficient inhibition of human telomerase activity by antisense oligonucleotides sensitizes cancer cells to radiotherapy. Acta Pharmacol Sin. 27:1185–1191. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Fan XK, Yan RH, Li BJ, Chen XM, Wei L and Wang Z: Antisense oligodeoxynucleotide against human telomerase reverse transcriptase inhibits the proliferation of Eca-109 esophageal carcinoma cells. Exp Ther Med. 8:1247–1252. 2014.PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 16 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cao F, Ju X, Chen D, Jiang L, Zhu X, Qing S, Fang F, Shen Y, Jia Z, Zhang H, Zhang H, et al: Phosphorothioate‑modified antisense oligonucleotides against human telomerase reverse transcriptase sensitize cancer cells to radiotherapy. Mol Med Rep 16: 2089-2094, 2017
APA
Cao, F., Ju, X., Chen, D., Jiang, L., Zhu, X., Qing, S. ... Zhang, H. (2017). Phosphorothioate‑modified antisense oligonucleotides against human telomerase reverse transcriptase sensitize cancer cells to radiotherapy. Molecular Medicine Reports, 16, 2089-2094. https://doi.org/10.3892/mmr.2017.6778
MLA
Cao, F., Ju, X., Chen, D., Jiang, L., Zhu, X., Qing, S., Fang, F., Shen, Y., Jia, Z., Zhang, H."Phosphorothioate‑modified antisense oligonucleotides against human telomerase reverse transcriptase sensitize cancer cells to radiotherapy". Molecular Medicine Reports 16.2 (2017): 2089-2094.
Chicago
Cao, F., Ju, X., Chen, D., Jiang, L., Zhu, X., Qing, S., Fang, F., Shen, Y., Jia, Z., Zhang, H."Phosphorothioate‑modified antisense oligonucleotides against human telomerase reverse transcriptase sensitize cancer cells to radiotherapy". Molecular Medicine Reports 16, no. 2 (2017): 2089-2094. https://doi.org/10.3892/mmr.2017.6778