MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis

  • Authors:
    • Sha Liu
    • Xiaofen Pan
    • Qin Yang
    • Lu Wen
    • Yao Jiang
    • Yingchao Zhao
    • Guiling Li
  • View Affiliations

  • Published online on: April 27, 2015     https://doi.org/10.3892/or.2015.3929
  • Pages: 2853-2862
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Evidence has demonstrated that microRNAs (miRNAs) are important in the regulation of cellular radiosensitivity of various types of human cancer. The aim of this study was to examine the role of miR-18a in regulating the radiosensitivity of cervical cancer, in order to understand the underlying mechanism and to assess the potential of miR-18a as a biomarker for predicting radiosensitivity. The expression of miR-18a was investigated in 48 cervical cancer patients. The results revealed that miR-18a expression was significantly higher in radiosensitive patients than in radioresistant patients by RT-qPCR (P<0.05). Transient transfection experiments showed that miR-18a was upregulated by the miR-18a mimic and downregulated by the miR-18a inhibitor in the SiHa and HeLa cells. Without irradiation treatment, a similar growth was observed in the cells with or without transfection of miR-18a. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and Hoechst staining assays showed that miR-18a had no effect on the proliferation and apoptosis of cervical cancer cells after transfection. However, the upregulation of miR-18a suppressed the level of ataxia-telangiectasia mutated and attenuated DNA double-strand break repair after irradiation, which re-sensitized the cervical cancer cells to radiotherapy by promoting apoptosis. Taken together, these results demonstrated that miR-18a is a potential molecule predictor of radiosensitivity in cervical cancer patients and played an important role in the response to radiotherapy.

Introduction

Cervical cancer is the third most commonly diagnosed cancer and the fourth leading cause of cancer-associated mortalities among females worldwide (1). Most of these cases and deaths occurred in developing countries (2). Despite showing a high cure potential when diagnosed early, cervical cancer continues to be a significant health threat to females and remains a serious issue of public health concern, especially in developing countries. Cervical cancer is usually treated with surgery, radiotherapy and chemotherapy depending on the stage of disease. Radiotherapy serves as the standard adjuvant approach in the management of cervical cancer across different clinical sub-types. For some early stage patients, radiotherapy can achieve the same effect as radical surgery (3). However, in clinical practice, some patients often develop resistance to the radiation treatment, resulting in treatment failure and eventually poor outcomes. Thus, it is important to improve radiosensitivity in those patients and explore the mechanisms underlying radioresistance (4).

MicroRNAs (miRNAs) are a type of endogenously expressed small, non-coding RNA sequences (~22 nucleotides) with a role in gene expression regulation at the post-transcriptional level (5). miRNAs bind to the 3′-untranslated regions (UTRs) of their target mRNAs and repress the translation or stability of target mRNAs (6). By downregulating the oncogenes or tumor suppressors, miRNAs may play tumor suppressive or oncogenic roles. miRNAs can alter various cell processes, including developmental transitions, organ morphology, bilateral asymmetry, stress response, metabolism, cell proliferation, apoptosis and the response to radiation (5,7,8). In recent years, the relationship between miRNAs and the radiosensitivity of different cancers has been identified (Table I). Those studies showed that miRNAs are potentially useful in manipulating the radiation response in the clinic to enhance susceptibility to or protect cells from radiation. In human cervical cancer, the expression of miR-18a was significantly decreased in the radioresistant tumor samples as compared with that in the radiosensitive tumor samples as identified by microarray analysis (9).

Table I

Differential expression of microRNAs in various types of cancer and the effect on sensitivity to radiotherapy.

Table I

Differential expression of microRNAs in various types of cancer and the effect on sensitivity to radiotherapy.

MicroRNACancer typeTarget gene(s)Effect on sensitivity to radiotherapyRefs.
hsa-let-7 familyLung cancerRASUp/Down(33)
hsa-miR-521Prostate cancerCSAUp(34)
hsa-miR-221/222Gastric cancerPTENDown(35)
hsa-miR-221Colorectal carcinomaPTENDown(36)
hsa-miR-21Esophageal cancerPTENDown(37)
hsa-miR-181aMalignant gliomaBcl-2Up(38)
hsa-miR-34NSCLCp53Up(39)
hsa-miR-148bNHLDNMT3bUp(40)
hsa-miR-9Lung cancerNF-κB1Up(41)
hsa-miR-218Cervical cancerRictor proteinUp(4)
hsa-miR-155Lung cancerFOXO3ADown(42)
hsa-miR-29cNPCBcl-2 and Mcl-1Up(43)
hsa-miR-18aBreast cancerATMUp(13)

[i] NSCLC, non-small cell lung cancer; NHL, non-Hodgkin’s lymphoma; NPC, nasopharyngeal carcinoma; CSA, Cockayne syndrome protein A; ATM, ataxia-telangiectasia mutated.

In the present study, we revealed that the mean expression level of miR-18a was significantly higher in radiosensitive patients compared with radioresistant patients. Moreover, we demonstrated that miR-18a re-sensitizes cervical cancer cells to radiotherapy by impairing DNA damage repair and promoting cell apoptosis. We proved that miR -18a is a potential predictor of radiosensitivity and a therapeutic target in cervical cancer patients.

Materials and methods

Patient characteristics and tissue specimens

Primary cervical carcinoma tissues were collected from Wuhan Union Hospital. The samples were obtained after written informed consent was provided by the patients. The study was approved by the Ethics Committee of Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology. A total of 48 clinical tissue samples were verified by histopathological examination and stored at −80°C prior to use. The tumor samples were obtained from the patients who did not receive any previous therapy. The samples were classified into two groups according to the response to radiotherapy: the radiosensitive group included patients who achieved complete response (CR) after radical radiotherapy, while the radioresistant group included patients who achieved partial response (PR) or relapsed within six months. The patient characteristics are shown in Table II.

Table II

Different clinicopathological characteristics of cervical cancer patients.

Table II

Different clinicopathological characteristics of cervical cancer patients.

Clinicopathological characteristicsAll casesRadiotherapy response
P-value
Radioresistant (%)Radiosensitive (%)
Age (years)
 <50238 (34.8)15 (65.2)0.353
 ≥502512 (48)13 (52)
Histological classification
 Squamous cell carcinoma4416 (36.4)28 (63.6)0.013
 Adenocarcinoma44 (100)0
FIGO stage
 I-IIb3415 (44.1)19 (55.9)0.591
 IIIa-IV145 (35.7)9 (64.3)
Tumor size (cm)
 <4218 (38.1)13 (61.9)0.658
 ≥42712 (44.4)15 (55.6)
Cell culture

The SiHa and HeLa human cervical cancer cell lines were provided by the Gynaecology and Obstetric Laboratory of Wuhan Union Hospital. SiHa cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) with high glucose and supplemented with 10% fetal bovine serum (FBS) (both from Hyclone, Logan, UT, USA). HeLa cells were routinely cultured in RPMI-1640 medium containing 10% FBS (Hyclone). The cell lines were maintained in a humidified incubator at 37°C in a 5% CO2 atmosphere.

Transfection

The cells (2×105 cells/well) were seeded in 6-well plates and transfection was carried out 24 h later using Opti-MEM and Lipofectamine 2000 (Invitrogen, Life Technologies) according to the manufacturer’s instructions until the cells reached a ~30–50% confluence. The miR-18a mimic, inhibitor and their negative controls were purchased from RiboBio (Guangzhou, China). miR-18a expression was increased or decreased in the SiHa and HeLa cells by transient transfection with miR-18a mimic at a final concentration of 50 nM or miR-18a inhibitor at a final concentration of 100 nM. The negative control was included in the parallel experiment. Following transfection, the cells were subjected to further assays or for RNA/protein extraction.

RNA extraction, reverse transcription and quantitative PCR

Total RNA was isolated from the transfected cells and human tissues using TRIzol reagent (Ambion, Life Technologies, Carlsbad, CA, USA) according to the manufacturer’s instructions. miR-18a expression was assessed using SYBR-Green quantitative reverse transcription (RT)-PCR (Takara Biotechnology, Dalian, China) with U6 snRNA as a normalizing control on a StepOne Real-Time PCR system (Applied Biosystems, Foster City, CA, USA). The cycling conditions were: 95°C for 30 sec, 60°C for 5 sec, 95°C for 15 sec, 60°C for 1 min and 95°C 15 sec, which were repeated for 40 cycles. The gene expression was measured using the following primers: miR-18a, (forward), 5′-GTTCCTAAGGTTCATCTAGTGCAGATA-3′ and (reverse), 5′-CAGTGCAGGGTCCGAGGTAT-3′; U6, (forward), 5′-CTCGCTTCGGCAGCACATA-3′ and (reverse), 5′-CGAATTTGCGTGTCATCCT-3′. The relative expression of miR-18a was calculated using the relative quantity (RQ) 2−ΔCT (in tissues) or 2−ΔΔCt (in cells) method against U6 for normalization, where ΔCT = (CTmiR-18a-CTU6snRNA), ΔΔCT = (CTmiR-18a-CTU6snRNA) mimic/inhibitor - (CTmiR-18a-CTU6snRNA) control. Each sample was analyzed in triplicate.

MTT assay

The MTT assay was used to detect the proliferation rate of the cervical cancer cells following transfection. The cells (5×103 cells/well) were seeded in a 96-well plate for 24 h before transfection and then allowed to grow in normal medium. For the MTT assay, the cells were incubated in 20 μl MTT (5 μg/ml) at 37°C for 4 h and lysed in 150 μl dimethyl sulfoxide (DMSO) at room temperature for 10 min with slight agitation. The absorbance in each well was measured at 490 nm (test wavelength) and 630 nm (reference wavelength) using a microplate reader (BioTek Instruments, Inc., Winooski, VT, USA). The experiment was repeated three times and six parallel samples were measured each time.

Hoechst staining

The transfected cells were fixed with 10% absolute methanol for 10 min, and washed twice with PBS. Then cells were stained with Hoechst 33342 (Beyotime, Shanghai, China) for 15 min in the dark and then washed twice with PBS. The nuclear morphology of the cells was visualized using an inverted fluorescence microscope. The cells showing nuclear fragmentation and chromatin condensation in Hoechst staining were considered apoptotic cells (10). Three independent experiments were performed in triplicate.

Western blot analysis

The parental and transfected cells were exposed to 8 Gy X-rays, and then the cells were collected and lysed 24 h after irradiation. Protein was extracted from the lysates and the concentrations was measured using the BCA Protein Assay system (Beyotime). Anti-ATM, anti-γ-H2AX, anti-caspase-3 and anti-PARP antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA). β-actin protein levels were used as a control to verify equal protein loading.

Clonogenic cell survival in vitro

The SiHa and HeLa cells (2×105 cells/well) were seeded in 6-well plates 24 h before transfection. After 48 h of transfection, the cells were seeded in culture dishes and irradiated the following day at distinct doses (0, 2, 4, 6 and 8 Gy). The plates were incubated at 37°C in a 5% CO2 atmosphere for 12 days. The cells were fixed with 4% paraformaldehyde and then stained with 1% crystal violet, and colonies containing ≥50 cells in size were counted. The survival curve was derived from the multi-target single-hit model: SF = 1-(1-e−D/D0)N using the multi-target click model of GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA). The sensitization-enhancing ratio (SER) was then calculated (a ratio of SF2).

Flow cytometric analysis of apoptosis

Transfected cells were irradiated with a dose of 8 Gy. The cells were collected and resuspended 24 h after ionizing radiation (IR) according to the manufacturer’s instructions (Beyotime). Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI) staining assays were conducted to detect the percentage of apoptotic (FITC-stained) and necrotic (PI-stained) cells in a given population. Analyses were performed by a FACScan flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA), the procedures were repeated in triplicate.

Immunofluorescent staining

Transfected cells were exposed to 2 Gy X-rays, an appropriate dose according to most radical radiotherapy protocols (11). The cells were collected and stained with anti-γ-H2AX (Cell Signaling Technology) antibody at different times as previously described (12). Images were captured using a laser scanning confocal microscope (Olympus Optical Co., Tokyo, Japan). Red showed γ-H2AX foci and blue nuclei stained with DAPI. For each treatment, a γ-H2AX foci number of ≥300 cells was counted.

Statistical analysis

Statistical analysis was performed with the SPSS 21.0 statistical software package (SPSS, Chicago, IL, USA). The results were shown as mean ± SD. Differences in measured variables between experimental and control groups were assessed using the Student’s t-test. The receiver operating characteristics (ROC) curve analysis was used to investigate the predictive value of miR-18a expression in cervical cancer radiotherapy response. The Mann-Whitney U test was performed to compare the differences in the miR-18a expression levels between radiosensitive and radioresistant patients. In all cases, P<0.05 was considered to indicate a statistically significant difference.

Results

Detection of high level of miR-18a expression is in radiosensitive patients

To determine whether miR-18a was associated with radiosensitivity in cervical cancer, we first compared the miR-18a expression level in radiosensitive and radioresistant patients. Results are shown in Fig. 1 after normalization to control U6 expression. We detected that the mean expression level of miR-18a was significantly higher in radiosensitive than in radioresistant patients by RT-qPCR. This result revealed that miR-18a had, a linear correlation with radiosensitivity.

Tissue miR-18a as a potential diagnostic marker for the sensitivity of radiotherapy

The ROC curve and the area under the curve (AUC) were used to assess the feasibility of using miR-18a expression level as a diagnostic tool for detecting radiosensitivity. The data using a ROC curve showed obvious separation between the two groups, with an AUC of 0.814 and a 95% CI (confidence interval) of 0.685–0.944 (Fig. 2). At the cut-off value of 6.49×10−6 (relative expression in comparison with U6), the sensitivity was 89.3% and the specificity was 70%. This result indicated that miR-18a is a potential molecular marker for the detection of the sensitivity to radiotherapy.

Overexpression of miR-18a increases sensitivity of cervical cancer cells to radiation treatment

To determine the effect of miR-18a on the sensitivity of cervical cancer cells to radiotherapy, we transfected miR-18a mimic or inhibitor into the SiHa and HeLa cells. A qPCR analysis showed that miR-18a was markedly upregulated or downregulated when transfected with miR-18a mimic or inhibitor, particularly 48 h post-transfection (Fig. 3). Results of the clonogenic assay showed the surviving fractions (SF) at each dose (2, 4, 6 and 8 Gy) decreased in miR-18a overexpressed cells (Fig. 4). The values of D0, Dq and SF2 were lower in the miR-18a-upregulated cells than the control and blank groups (Table III). Therefore, we concluded that the miR-18a increased the cellular sensitivity to radiation treatment in vitro.

Table III

Main parameters of cell survival curves following irradiation.

Table III

Main parameters of cell survival curves following irradiation.

ParametersMock
Mimic-18a NC
Mimic-18a
SiHaHeLaSiHaHeLaSiHaHeLa
D02.042.662.132.581.661.67
Dq4.923.103.562.441.380.94
N3.412.172.681.951.831.56
SF20.790.730.710.680.470.43

[i] D0, mean lethal dose; Dq, quasi-threshold dose; N, extrapolation number; SF2, surviving fraction at 2 Gy.

Overexpression of miR-18a enhances radiation-induced apoptosis

To investigate the effect of miR-18a on the sensitivity of cervical cancer cells to radiotherapy, we examined cell apoptosis following treatment of irradiation with a flow cytometry assay. We detected that miR-18a enhanced the cellular sensitivity to radiation by promoting cell apoptosis following exposure to 8-Gy irradiation. As shown in Fig. 5, the percentage of apoptotic cells was significantly increased in the miR-18a mimic-transfected cells than the control cells, but decreased in the miR-18a inhibitor transfected cells than the control cells. What is more, apoptosis was detected through an increase of cleaved caspase-3 and PARP by western blotting, which were two classic characteristics of cell apoptosis. Western blot analysis demonstrated that 24 h after exposure to 8-Gy irradiation, cleaved caspase-3 and PARP protein expression in the SiHa and HeLa cells transfected with miR-18a mimic were higher than that in cells transfected with a control (Fig. 6).

miR-18a expression has no effect on the proliferation and apoptosis of cervical cancer cells

To determine the biological role of miR-18a in cervical cancer, an MTT assay was performed to examine the relationship between miR-18a expression and cell proliferation. Consequently, ectopic upregulation or downregulation of miR-18a had no significant impact on the proliferative activity of the SiHa and HeLa cells compared with the control groups (Fig. 7). In the Hoechst staining assay, the nucleus which exhibited brightly stained condensed chromatin, indicated typical morphological changes of apoptosis. In this experiment, the number of Hoechst-positive apoptotic cells had no difference in each group (Fig. 8). These data showed that miR-18a had no effect on the proliferation and apoptosis of cervical cancer cells without irradiation treatment.

MiR-18a suppresses ATM expression by targeting 3′-UTR of ATM

miRWalk, GenBank, TargetScan and mirBase were used to predict the target genes of miR-18a to determine how miR-18a modulates the sensitivity of cancer cells to radiation. Analysis with these algorithms, revealed that ATM was a potential target of miR-18a. In addition, the luciferase assay, as previously reported, demonstrated that miR-18a specifically affected the ATM-3′-UTR (13,14). It has been previously reported that radiation treatment upregulated ATM expression, leading to phosphorylation of the repair proteins (15). Therefore, we examined whether miR-18a enhanced the radio-sensitivity of cervical cancer cells by targeting ATM using western blotting after exposure to 8 Gy irradiation. The results showed that the level of ATM protein was significantly down-regulated following miR-18a overexpression but upregulated when miR-18a was suppressed. Results of downstream protein, γ-H2AX, were similar to those of ATM (Fig. 9).

Overexpression of miR-18a impairs the double-strand break (DSB) repair induced by radiation

One of the most important therapeutic functions of radiation is to induce DNA DSB in cancer cells. When cells respond to DNA damage, the initial step is the phosphorylation of repair proteins, such as the ATM and H2AX (11,16,17). If the ATM decreases, the DSB repair signaling pathway is impaired and cell apoptosis is enhanced. We monitored the kinetics of the DSB repair process by immunofluorescent staining of γ-H2AX foci at different time-points post-irradiation to 2 Gy. As shown in Fig. 10, a rapid increase of the number of γ-H2AX foci was evident following irradiation, reaching a peak at 30 min. The level of fluorescence then decreased significanly as the DSB repair was completed. Overexpression of miR-18a downregulated the ATM and reduced DNA repair, while inhibition of miR-18a led to an increase of DNA damage repair by upregulated ATM. The change of γ-H2AX foci was consistent with the western blot results, suggesting that the reduction of ATM led to a low efficiency of DSB repair as we hypothesized. These results further corroborated the role and mechanism of miR-18a in enhancing cancer cell radiosensitivity to radiotherapy.

Discussion

The standard radiotherapy for cervical cancer is the combination of external beam radiotherapy and intracavitary brachytherapy, which have the same efficiency on suppressing tumor growth and inhibiting metastasis as surgery for early patients (18). Radiation therapy serves as a major treatment strategy and is beneficial for cervical cancer patients to prevent recurrence or metastasis. Findings of a recent study showed that patients who received radiotherapy were 40–90% less likely to have a relapse of their cancer within 5 years than patients who did not undergo this therapy (3). However, we found a considerable number of patients suffering from radiation insensitivity in clinical practice and the 5-year survival rate was limited. Although there are constant technological innovations on radiotherapy equipment, its effect remains unsatisfactory, especially with respect to its negative effects of local recurrence and distant metastasis (19). Therefore, it is crucial to find predictive markers of radiation sensitivity.

The clinical value of miRNAs has been assessed because their expression is associated with tumor development, progression and even response to therapy (5,20,21). As previously reported, miRNAs were involved in the regulation of cellular radiosensitivity of various types of cancer. For instance, in breast cancer, miR-18a increased cellular radiosensitivity (13). miR-18a is located in the chromosome 13q31.1 region and belongs to the miR-17–92 cluster (22). A high expression of miR-18a has been found in several cancer types, such as bladder cancer (23), hepatocellular carcinoma (24), colon cancer (25) and nasopharyngeal carcinoma (26). The function of miR-18a in those types of cancer is as oncogene or suppressor. However, the functional role of miR-18a in cervical cancer remains unknown.

In the present study, we investigated the value of miR-18a in predicting the radiosensitivity of cervical cancer and reported, to the best of our knowledge, for the first time that the expression of miR-18a was significantly different between radiosensitive and radioresistant patients. The results showed that patients with a high expression of miR-18a were hypersensite to radiation as compared to patients with a low expression of miR-18a. The ROC curve indicated miR-18a may be a novel biomarker for predicting the radiosensitivity in cervical cancer treatment. In vitro, we demonstrated that overexpression of miR-18a significantly increases cellular sensitivity to radiotherapy using a colony formation assay, while the downregulation of miR-18a induced cell resistance to radiation. Furthermore, we revealed the molecular mechanism of how miR-18a altered the sensitivity of cancer cells to radiotherapy. miR-18a increased the radiosensitivity of cancer cells by targeting the 3′-UTR of the ATM gene, thereby reducing DSB repair. The most lethal form of DNA damage after radiation treatment is known to be the DSB. Cells suffering from DSB must be repaired for cells to survive, or it would lead to genomic instability and eventually apoptosis. DSB can be repaired by the two major pathways: non-homologous end joining (NHEJ) and homologous recombination repair (HRR) (27). DNA damage activates ATM through auto-phosphorylation and the activated ATM is thought to be the main regulator of HRR (16). DNA DSB can be induced after exposure to irradiation. ATM is then recruited to the DSB site and activated by auto-phosphorylation. Activated ATM can phosphorylate the histone variant H2AX on its C-terminal tail on serine 139, forming γ-H2AX foci. A sequence of downstream targets involving events to repair the DNA damage are activated, leading to amplification of the HRR signaling cascade (28,29). ATM has been demonstrated as a direct target of several miRNAs, and overexpression of those miRNAs downregulates ATM expression and sensitizes various cell lines to radiation therapy (3032). It is known that miR-18a suppresses ATM expression by specifically targeting the 3′-UTR of ATM transcripts as previously reported. In this study, we upregulated and downregulated the expression of miR-18a in cervical cancer cells (SiHa and HeLa) by trans-fecting with miR-18a mimic and inhibitor. Tthe results showed that the level of miR-18a had no effect on the proliferation and apoptosis of the SiHa and HeLa cells post-transfection, but affected the expression of ATM after radiotherapy. Western blot analysis showed that overexpression of miR-18a resulted in a significantly decrease of ATM proteins and a sequential downregulation of the phosphorylation level of ATM downstream genes, such as γ-H2AX. Our data also suggest that overexpression of miR-18a increases cellular sensitivity to radiotherapy by enhancing radiation-associated apoptosis. To confirm the role of miR-18a in radiotherapy, we detected the expression level of caspase-3 and PARP. The results show that cleaved caspase-3 and PARP were markedly increased in miR-18a-upregulated cells than the controls, and vice versa.

In summary, we have demonstrated that the expression of miR-18a was significantly higher in the radiosensitive than in the radioresistant patients. ATM, a key protein to DNA damage repair, was the target of miR-18a in cervical cancer lines. In addition, overexpression of miR-18a suppressed the level of ATM and attenuated DNA DSB repair, which re-sensi-tized the cervical cancer cells to radiotherapy by promoting apoptosis. The results have demonstrated that the suppressed DNA damage repair induced by upregulated miR-18a may play an important role in radiation sensitivity. miR-18a may be a potential molecular marker for the detection of the response to radiotherapy in cervical cancer patients and a therapeutic target.

Acknowledgments

The authors thank the Gynecology and Obstetric Laboratory of Wuhan Union Hospital for providing the SiHa and HeLa cells, and Bohan Yang and Tianhe Zeng for their advice and assistance.

Abbreviations:

miRNA

microRNA

ATM

ataxia-telangiectasia mutated

DSB

double-strand break

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Krieger N, Bassett MT and Gomez SL: Breast and cervical cancer in 187 countries between 1980 and 2010. Lancet. 379:1391–1392. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Rogers L, Siu SS, Luesley D, Bryant A and Dickinson HO: Radiotherapy and chemoradiation after surgery for early cervical cancer. Cochrane Database Syst Rev. 5:CD0075832012.PubMed/NCBI

4 

Yuan W, Xiaoyun H, Haifeng Q, Jing L, Weixu H, Ruofan D, Jinjin Y and Zongji S: MicroRNA-218 enhances the radiosensitivity of human cervical cancer via promoting radiation induced apoptosis. Int J Med Sci. 11:691–696. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Metheetrairut C and Slack FJ: MicroRNAs in the ionizing radiation response and in radiotherapy. Curr Opin Genet Dev. 23:12–19. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Bernstein E, Caudy AA, Hammond SM and Hannon GJ: Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature. 409:363–366. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Hummel R, Hussey DJ and Haier J: MicroRNAs: predictors and modifiers of chemo- and radiotherapy in different tumour types. Eur J Cancer. 46:298–311. 2010. View Article : Google Scholar

8 

Zhang B, Wang Q and Pan X: MicroRNAs and their regulatory roles in animals and plants. J Cell Physiol. 210:279–289. 2007. View Article : Google Scholar

9 

Ke G, Liang L, Yang JM, Huang X, Han D, Huang S, Zhao Y, Zha R, He X and Wu X: MiR-181a confers resistance of cervical cancer to radiation therapy through targeting the pro-apoptotic PRKCD gene. Oncogene. 32:3019–3027. 2013. View Article : Google Scholar

10 

Chen HM, Hsu JH, Liou SF, Chen TJ, Chen LY, Chiu CC and Yeh JL: Baicalein, an active component of Scutellaria baicalensis Georgi, prevents lysophosphatidylcholine-induced cardiac injury by reducing reactive oxygen species production, calcium overload and apoptosis via MAPK pathways. BMC Complement Altern Med. 14:2332014. View Article : Google Scholar : PubMed/NCBI

11 

Bourton EC, Plowman PN, Smith D, Arlett CF and Parris CN: Prolonged expression of the γ-H2AX DNA repair biomarker correlates with excess acute and chronic toxicity from radiotherapy treatment. Int J Cancer. 129:2928–2934. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Ohba S, Mukherjee J, See WL and Pieper RO: Mutant IDH1-driven cellular transformation increases RAD51-mediated homologous recombination and temozolomide resistance. Cancer Res. 74:4836–4844. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Song L, Lin C, Wu Z, Gong H, Zeng Y, Wu J, Li M and Li J: miR-18a impairs DNA damage response through downregulation of ataxia telangiectasia mutated (ATM) kinase. PLoS One. 6:e254542011. View Article : Google Scholar : PubMed/NCBI

14 

Wu CW, Dong YJ, Liang QY, He XQ, Ng SS, Chan FK, Sung JJ and Yu J: MicroRNA-18a attenuates DNA damage repair through suppressing the expression of ataxia telangiectasia mutated in colorectal cancer. PLoS One. 8:e570362013. View Article : Google Scholar : PubMed/NCBI

15 

Shiloh Y and Ziv Y: The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. Nat Rev Mol Cell Biol. 14:197–210. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Borgstahl GE, Brader K, Mosel A, Liu S, Kremmer E, Goettsch KA, Kolar C, Nasheuer HP and Oakley GG: Interplay of DNA damage and cell cycle signaling at the level of human replication protein A. DNA Repair (Amst). 21:12–23. 2014. View Article : Google Scholar

17 

Bakkenist CJ and Kastan MB: DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature. 421:499–506. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Nakano T, Kato S, Ohno T, Tsujii H, Sato S, Fukuhisa K and Arai T: Long-term results of high-dose rate intracavitary brachy-therapy for squamous cell carcinoma of the uterine cervix. Cancer. 103:92–101. 2005. View Article : Google Scholar

19 

Qi L, Xing LN, Wei X and Song SG: Effects of VEGF suppression by small hairpin RNA interference combined with radiotherapy on the growth of cervical cancer. Genet Mol Res. 13:5094–5106. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Kloosterman WP and Plasterk RH: The diverse functions of microRNAs in animal development and disease. Dev Cell. 11:441–450. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Esquela-Kerscher A and Slack FJ: Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Tao J, Wu D, Li P, Xu B, Lu Q and Zhang W: microRNA-18a, a member of the oncogenic miR-17–92 cluster, targets Dicer and suppresses cell proliferation in bladder cancer T24 cells. Mol Med Rep. 5:167–172. 2012.

24 

Liu WH, Yeh SH, Lu CC, Yu SL, Chen HY, Lin CY, Chen DS and Chen PJ: MicroRNA-18a prevents estrogen receptor-alpha expression, promoting proliferation of hepatocellular carcinoma cells. Gastroenterology. 136:683–693. 2009. View Article : Google Scholar

25 

Fujiya M, Konishi H, Mohamed Kamel MK, Ueno N, Inaba Y, Moriichi K, Tanabe H, Ikuta K, Ohtake T and Kohgo Y: microRNA-18a induces apoptosis in colon cancer cells via the autophagolysosomal degradation of oncogenic heterogeneous nuclear ribonucleoprotein A1. Oncogene. 33:4847–4856. 2014. View Article : Google Scholar

26 

Luo Z, Dai Y, Zhang L, Jiang C, Li Z, Yang J, McCarthy JB, She X, Zhang W, Ma J, et al: miR-18a promotes malignant progression by impairing microRNA biogenesis in nasopha-ryngeal carcinoma. Carcinogenesis. 34:415–425. 2013. View Article : Google Scholar

27 

Symington LS and Gautier J: Double-strand break end resection and repair pathway choice. Annu Rev Genet. 45:247–271. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Valdiglesias V, Giunta S, Fenech M, Neri M and Bonassi S: γH2AX as a marker of DNA double strand breaks and genomic instability in human population studies. Mutat Res. 753:24–40. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Kinner A, Wu W, Staudt C and Iliakis G: Gamma-H2AX in recognition and signaling of DNA double-strand breaks in the context of chromatin. Nucleic Acids Res. 36:5678–5694. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Hu H, Du L, Nagabayashi G, Seeger RC and Gatti RA: ATM is downregulated by N-Myc-regulated microRNA-421. Proc Natl Acad Sci USA. 107:1506–1511. 2010. View Article : Google Scholar

31 

Yan D, Ng WL, Zhang X, Wang P, Zhang Z, Mo YY, Mao H, Hao C, Olson JJ, Curran WJ, et al: Targeting DNA-PKcs and ATM with miR-101 sensitizes tumors to radiation. PLoS One. 5:e113972010. View Article : Google Scholar : PubMed/NCBI

32 

Ng WL, Yan D, Zhang X, Mo YY and Wang Y: Over-expression of miR-100 is responsible for the low-expression of ATM in the human glioma cell line: M059J. DNA Repair (Amst). 9:1170–1175. 2010. View Article : Google Scholar

33 

Weidhaas JB, Babar I, Nallur SM, Trang P, Roush S, Boehm M, Gillespie E and Slack FJ: MicroRNAs as potential agents to alter resistance to cytotoxic anticancer therapy. Cancer Res. 67:11111–11116. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Josson S, Sung SY, Lao K, Chung LW and Johnstone PA: Radiation modulation of microRNA in prostate cancer cell lines. Prostate. 68:1599–1606. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Chun-Zhi Z, Lei H, An-Ling Z, Yan-Chao F, Xiao Y, Guang-Xiu W, Zhi-Fan J, Pei-Yu P, Qing-Yu Z and Chun-Sheng K: MicroRNA-221 and microRNA-222 regulate gastric carcinoma cell proliferation and radioresistance by targeting PTEN. BMC Cancer. 10:3672010. View Article : Google Scholar : PubMed/NCBI

36 

Xue Q, Sun K, Deng HJ, Lei ST, Dong JQ and Li GX: Anti-miRNA-221 sensitizes human colorectal carcinoma cells to radiation by upregulating PTEN. World J Gastroenterol. 19:9307–9317. 2013. View Article : Google Scholar

37 

Huang S, Li XQ, Chen X, Che SM, Chen W and Zhang XZ: Inhibition of microRNA-21 increases radiosensitivity of esophageal cancer cells through phosphatase and tensin homolog deleted on chromosome 10 activation. Dis Esophagus. 26:823–831. 2013. View Article : Google Scholar

38 

Chen G, Zhu W, Shi D, Lv L, Zhang C, Liu P and Hu W: microRNA-181a sensitizes human malignant glioma U87MG cells to radiation by targeting Bcl-2. Oncol Rep. 23:997–1003. 2010.PubMed/NCBI

39 

He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Zender L, Magnus J, Ridzon D, et al: A microRNA component of the p53 tumour suppressor network. Nature. 447:1130–1134. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Wu Y, Liu GL, Liu SH, Wang CX, Xu YL, Ying Y and Mao P: MicroRNA-148b enhances the radiosensitivity of non-Hodgkin’s lymphoma cells by promoting radiation-induced apoptosis. J Radiat Res (Tokyo). 53:516–525. 2012. View Article : Google Scholar

41 

Arora H, Qureshi R, Jin S, Park AK and Park WY: miR-9 and let-7g enhance the sensitivity to ionizing radiation by suppression of NFκB1. Exp Mol Med. 43:298–304. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Babar IA, Czochor J, Steinmetz A, Weidhaas JB, Glazer PM and Slack FJ: Inhibition of hypoxia-induced miR-155 radiosensitizes hypoxic lung cancer cells. Cancer Biol Ther. 12:908–914. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Zhang JX, Qian D, Wang FW, Liao DZ, Wei JH, Tong ZT, Fu J, Huang XX, Liao YJ, Deng HX, et al: MicroRNA-29c enhances the sensitivities of human nasopharyngeal carcinoma to cisplatin-based chemotherapy and radiotherapy. Cancer Lett. 329:91–98. 2013. View Article : Google Scholar

Related Articles

Journal Cover

June-2015
Volume 33 Issue 6

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
Liu S, Pan X, Yang Q, Wen L, Jiang Y, Zhao Y and Li G: MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis. Oncol Rep 33: 2853-2862, 2015
APA
Liu, S., Pan, X., Yang, Q., Wen, L., Jiang, Y., Zhao, Y., & Li, G. (2015). MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis. Oncology Reports, 33, 2853-2862. https://doi.org/10.3892/or.2015.3929
MLA
Liu, S., Pan, X., Yang, Q., Wen, L., Jiang, Y., Zhao, Y., Li, G."MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis". Oncology Reports 33.6 (2015): 2853-2862.
Chicago
Liu, S., Pan, X., Yang, Q., Wen, L., Jiang, Y., Zhao, Y., Li, G."MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis". Oncology Reports 33, no. 6 (2015): 2853-2862. https://doi.org/10.3892/or.2015.3929