Overexpression of TOB1 confers radioprotection to bronchial epithelial cells through the MAPK/ERK pathway

  • Authors:
    • Jun Che
    • Yan-Wei Lu
    • Ke-Kang Sun
    • Chan Feng
    • Ai-Jing Dong
    • Yang Jiao
  • View Affiliations

  • Published online on: June 11, 2013     https://doi.org/10.3892/or.2013.2536
  • Pages: 637-642
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of this study was to investigate the effects and mechanisms of antiproliferative transducer of erbB2, 1 (TOB1) on the radiosensitivity of the normal human bronchial epithelial cell line HBE. After exposure to different doses of irradiation or a certain dose for different time intervals, the expression of TOB1 mRNA and protein in HBE cells was determined by semi-quantitative RT-PCR and western blot analysis. Liposome-induced recombinant plasmid transfection and G418 selection were performed to establish a stably transfected TOB1-overexpressing HBE cell line. A clonogenic assay was used to determine the radiosensitivity of the HBE cells with different TOB1 expression statuses. The cell cycle distribution was detected by flow cytometry. The ionizing radiation (IR)-induced γ-H2AX foci formation was detected by immunofluorescence assay. The related mechanism was explored by western blot analysis. TOB1 expression in the HBE cells was not induced by IR, neither dose-dependently nor time-dependently. Compared to the parental or ‘mock’ transfected HBE cells, the radiosensitivity of HBE cells overexpressing TOB1 was significantly decreased (P<0.05). Exogenous TOB1 prevented HBE cells from apoptosis after IR, in contrast to the control cells (P<0.05), and significantly decreased the IR-induced γ-H2AX foci formation. After IR, the expression of DNA damage repair proteins such as XRCC1, MRE11, FEN1 and ATM was increased in the TOB1‑overexpressing HBE cells when compared with the expression levels in the control cells. HBE/TOB1 cells presented a much higher phosphorylated ERK1/2 and phosphorylated p53 when compared with the levels in the control cell lines when receiving 6 Gy of X-rays. Notably, the increased expression of phosphorylated p53 in HBE/TOB1 cells after IR was sufficiently blocked by U0126, a specific inhibitor of MEK1/2. Different from its functions in several lung cancer cell lines, TOB1 demonstrated a radioprotective function in the immortalized normal human bronchial epithelial cell line HBE via the MAPK/ERK signaling pathway.

Introduction

In thoracic tumor radiotherapy, radiation-induced pulmonary injury (RIPI) is the main dose restrictive and common complication which seriously impairs the local control of thoracic tumors, particularly non-small cell lung cancer that is unsuitable for surgery (1,2). Moreover, RIPI affects the prognosis of lymphoma or breast cancer patients who were confirmed to have extended survival following radiotherapy (2,3). Thus, it is necessary to overcome the restraint of RIPI, in order to improve the efficacy of thoracic radiotherapy in the future.

The transducer of erbB2, 1 (TOB1) gene is located on chromosome 17q21, and was originally discovered as a member of the antiproliferative protein family TOB/BTG (4). Numerous studies have revealed the tumor-suppressor properties of TOB1 in different types of human malignancies (59). Recently, our laboratory also confirmed that TOB1 may radiosensitize human cancer cell lines such as cervical cancer cell line HeLa, breast cancer cell lines MCF-7 and MDA-MB-231 (10,11), and human lung cancer cell line A549 (unpublished data). However, few studies have investigated the influence of TOB1 on the radiosensitivity of normal tissues or cell lines.

In the present study, parental normal human epithelial cell line HBE and HBE-overexpressing TOB1 were used as cell models. RT-PCR, western blot analysis, clonogenic assay and immunofluorescence assay were used to reveal the effects of TOB1 in regards to the radiation protective effects on normal cells. Flow cytometric assay, western blot analysis and pathway-specific inhibitors were applied to elucidate the related mechanisms. Together with previous studies, we report the theoretical and experimental data on clinical lung cancer radiotherapy and radiation protection.

Materials and methods

Cell culture, transfection and irradiation

Human bronchial epithelial cells were originally obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA), maintained in our laboratory and cultured with Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, CA, USA) with 10% fetal bovine serum (Invitrogen), at 37°C in an atmosphere of 5% CO2. Medium was replaced every 2 days, and cells were subcultured every 3 days. Recombinant plasmid pcDNA3.0/TOB1 was constructed by our laboratory as previously described (10). HBE cells were transfected with the vector and pcDNA3.0/TOB1 using Lipofectamine (Invitrogen) and maintained in medium containing 500 μg/ml G418 (Sigma Aldrich, MO, USA) until resistant cell clones were formed. Irradiation was performed with 6-MeV X-ray linear accelerator (Siemens KD2, Germany) with a dose rate of 200 cGy/min, with an irradiating area of 20×20 cm and 100-cm source-skin distance.

Clonogenic assay

The exponentially growing cells were plated at different cell densities and irradiated with 0, 0.5, 1, 2, 4, 6 Gy of X-rays. After 12–14 days of incubation at 37°C, cells were fixed in methanol followed by Giemsa staining. The number of colonies per dish was counted, and the surviving fractions were calculated as the ratio of plating efficiencies for irradiated and unirradiated cells. Plating efficiency is defined as the colony number divided by the number of cells plated for unirradiated controls. Experiments were conducted in triplicate, and data are presented as means ± standard deviation (SD) from three independent experiments. All surviving fractions were fitted into the linear quadratic model using GraphPad Prism 5.0 software (GraphPad Software Inc., La Jolla, CA, USA).

Flow cytometric assay

Cells were removed with trypsin and collected into centrifuge tubes together with the culture medium. The detailed methods for flow cytometry and Annexin V-FITC apoptosis analysis have been previously described (10). The cell cycle distribution and apoptotic rate were calculated from 10,000 cells using ModFit LT software (Becton-Dickinson, San Jose, CA, USA) using FACSCalibur (Becton-Dickinson).

Immunofluorescence detection for phosphorylated γ-H2AX

Cells were grown on sterile glass chambers (Becton-Dickinson and Company, Franklin Lakes, NJ, USA). After a single dose of X-ray exposure, all cells were fixed in 4% formaldehyde before being permeabilized in 0.25% Triton X-100. The chamber was blocked with 1% BSA for at least 1 h. The primary anti-phosphorylation monoclonal antibody γ-H2AX (1:200, Epitomics, Burlingame, CA, USA) was added and incubated for 2 h at room temperature. Specific staining was visualized with a secondary antibody conjugated to FITC 488. Hochest 33342 (Sigma Aldrich, St. Louis, MO, USA) was utilized for nuclear staining. Fluorescence was observed using a laser scanning confocal microscope at a wavelength of 488 nm within 30 min. The number of foci in 20 cells in each sample was counted randomly.

Western blot analysis

Western blot analysis was performed as previously described (10). Briefly, the following primary antibodies were used for immunoblotting: β-actin (C-4), TOB1 (N-20), XRCC 1 (33-2-5), FEN1 (B-4), ATM (C-20), MRE11 (C-16) (1:1000 dilution; all from Santa Cruz Biotechnology, Santa Cruz, CA, USA). The protein bands were visualized using an enhanced chemiluminescence system (Hangzhou Youither Bioscience Co., Ltd., Hangzhou, China) with prestained markers as molecular size standards.

RT-PCR assay

TOB1 mRNA expression was determined using semi-quantitative RT-PCR assays. The PCR reaction conditions and cycle numbers were rigorously adjusted so that each reaction occurred within the linear range of amplification. The detailed methods for RNA isolation, cDNA synthesis and RT-PCR analyses have been previously described (10,12). For specific intent genes, the PCR primers were as follows: GAPDH sense, 5′-CAA CTA CAT GGT CTA CAT GTT CC-3′ and antisense, 5′-CAA CCT GGT CCT CAG TGT AG-3′; TOB1 sense, 5′-GGA TCG ACC CAT TTG AGG TTT CT-3′ and anti-sense-5′-CTA CCC AAG CCA AGC CCA TAC AG-3′. The PCR products were analyzed via electrophoresis through 1% agrose gels containing 0.1 mg/ml ethidium bromide (EB). The gels were photographed under ultraviolet light.

Statistical comparisons

The data are presented as means and standard deviations. Statistical comparisons of the experimental results between the treated group and the control group were carried out using the two-tailed Student’s t-test. All statistical tests were performed using the SPSS version 17.0. P-value <0.05 between groups was considered statistically significant.

Results

TOB1 expression in the normal human bronchial epithelial cell line HBE is not induced by ionizing radiation (IR)

HBE cells were respectively irradiated with 0, 2, 4 and 8 Gy of X-rays generated by a linear accelerator and harvested after 24 h. The cell samples were also collected 2, 12 and 24 h after exposed to 6-Gy irradiation. The relevant expression level of TOB1 mRNA and protein was determined by semi-quantitative RT-PCR and western blot analysis. As shown in Fig. 1, IR did not increase the expression level of TOB1, neither in a dose-dependent nor in a time-dependent manner.

Overexpression of TOB1 reduces the sensitivity of HBE cells to IR

To investigate whether TOB1 overexpression affects the radiosensitivity of HBE cells, the TOB1 stable transfectant (HBE/TOB1) was used. The TOB1 expression was determined by western blot analysis (Fig. 2A). As shown in Fig. 2B, the parental HBE cell line and ‘mock’ transfectant HBE/PC3 both showed a typical clonogenic survival curve with a shoulder representing the potential DNA damage repair, while exogenous TOB1 significantly protected HBE cells from the IR-induced damage.

Flow cytometry assay was also performed 24 h after a single-dose irradiation of 6-Gy of X-rays. As shown in Fig. 2C, all cell lines exhibited typical IR-induced cell cycle G2/M arrest without significant difference (P>0.05). However, the apoptotic cell portion of HBE/TOB1 (24.1%) was obviously less than HBE (32.98%) and HBE/PC3 (34.39%) after IR (P<0.05) (Fig. 2D).

Exogenous TOB1 increases DNA damage repair in HBE cells

In order to gain insight into the molecular mechanisms of the radioprotective effect of TOB1 and to investigate its effect on the initial DNA damage response to IR, the induction of DNA double-strand breaks (DSBs), as analyzed by the formation of phospho-γ-H2AX foci, was measured 2 h after irradiation of the HBE cells, HBE/PC3, and HBE/TOB1 cells. The results showed that IR-induced γ-H2AX foci were decreased in HBE/TOB1 cells 2 h post-radiation, in contrast to the control cells (Fig. 3A and B) suggesting facilitation of DNA damage repair.

Furthermore, we explored the underlying molecular mechanisms related to different IR-induced DNA damage responses due to TOB1 status. As shown in Fig. 3C and D, compared with the HBE/PC3 and parental cells, the expression levels of important DNA repair proteins such as XRCC1, MRE11, FEN1 and ATM were increased in the HBE/TOB1 cells after IR.

TOB1 regulates the radiosensitivity of HBE via the MAPK/ERK signaling pathway

Phosphorylation and subsequent activation of p53, as well as its transcription induction, are key initial responses to stress responses (13). Therefore, western blot analysis was used to investigate the related mechanisms through which TOB1 is involved in radioprotection of normal cells. In the present study, our results revealed that overexpression of TOB1 significantly increased IR-induced phosphorylation of p53 and ERK1/2 (Fig. 4).

We then explored the specific signaling cascade involved in this response by using the MEK1/2 inhibitor (U0126) specific to the MAPK/ERK signaling pathway. The increased phosphorylation of p53 increased by TOB1 overexpression after IR was blocked by U0126 (Fig. 4B).

Discussion

At present, although 70% of cancer patients require radiotherapy, it is still a difficult task to protect adjacent normal tissue and to increase the precision of dose delivery to the target tumor for radiotherapy (14,15). Gene therapy targeted by radiation may be a promising solution in which the complementary DNA for a cytotoxic gene is ligated downstream of IR-inducible promoters, which involve the expression of immediate early genes (16). In our previous study, it was demonstrated that TOB1 was IR-inducible in lung cancer cell lines A549 and NCI-H1975, and that TOB1 radiosensitized lung cancer cells through certain signaling pathways (unpublished data). However, for the first time, we revealed the different biological functions of TOB1 in the immortalized normal cell line HBE. The expression of TOB1 was neither induced by X-rays, nor did it increase the radiosensitivity of HBE cells (Figs. 1 and 2). On the contrary, TOB1 overexpression obviously decreased the sensitivity of HBE to X-ray irradiation (Fig. 2B). All of these results together provide us with clues to the potential gene therapy application of TOB1 in clinical radiotherapy.

A double-strand break (DSB) of DNA, the main biological target of IR, determines mutation or cell death after IR (1719). The ability to repair DSBs determines the sensitivity of mammalian cells to IR (18,20). The recruitment of several proteins to the damage site is the initial cellular response to DSBs, through which γ-H2AX focus formation is one of the earliest event (19,21). In our study, exogenous TOB1 reduced the amount of γ-H2AX foci induced by IR in the HBE cells (Fig. 3A and B), and decreased the percentage of apoptotic HBE cells after IR (Fig. 2D), which indicated either a decrease in DNA damage or an increase in DNA repair.

In response to IR-induced DSBs, DNA damage signals are transmitted through several pathways, from which ataxia telangiectasia-mutated (ATM) signaling plays perhaps the most important role (13,22,23). IR-induced ATM activation leads to the phosphorylation not only of γ-H2AX, but also of other important downstream effectors of DDR (apoptosis-related p53 and MDM2; cell cycle checkpoint-related CHK1/2; DNA repair-related MRE11) (13,2426). In our study, exogenous TOB1 obviously increased the expression of ATM and its downstream DNA repair effectors XRCC1, MRE11, and FEN1 (Fig. 3C and D). These data suggest that TOB1 overexpression promotes IR-induced DNA repair, thus, further confirming the feasibility of TOB1 application for thoracic radiotherapy.

Our previous studies demontrated that the MAPK/ERK pathway plays a significant role in cell survival after IR regulated by TOB1 (unpublished data). We demonstrated in this study that TOB1 overexpression enhanced the phosphorylation status of ERK1/2 in HBE cells, which was mainly associated with MAPK activation (Fig. 4A). After exposure to IR the activated MAPK pathway leads to the phosphorylation of p53 at multiple sites, which are key initial responses to cell cycle distribution and DNA damage (27,28). Our results revealed that exogenous TOB1 strongly enhanced IR-induced p53 phosphorylation in normal HBE cells, suggesting these cells have a more robust DNA damage surveillance and repair mechanism helping them either adapt to or overcome critical IR-induced DNA damage (Fig. 4A). Furthermore, the MAPK/ERK pathway-specific inhibitor U0126 was used just following IR. As shown in Fig. 4B, the serine 15 phosphorylation of p53 after IR was significantly inhibited by U0126 in the HBE/TOB1 cells.

In conclusion, although the mechanisms of how TOB1 is involved in IR-induced DNA damage are still to be fully elucidated, the results of the present study together with previous results in lung cancer cell lines, suggest that gene therapeutic approaches enhancing TOB1 expression may alleviate the side effects of radiotherapy as well as enhance radiotherapeutic benefit.

Acknowledgements

The present study was supported by grants from the Program for the Doctoral Fund of the Ministry of Education of China (20103201120016), the Hospital Management Center of Medical Science and Technology Development Foundation of Wuxi (YGM1101), the National Science Foundation of China (81170468) and the PAPD.

References

1 

Hunter NR, Valdecanas D, Liao Z, Milas L, Thames HD and Mason KA: Mitigation and treatment of radiation-induced thoracic injury with a cyclooxygenase-2 inhibitor, celecoxib. Int J Radiat Oncol Biol Phys. 85:472–476. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Eldh T, Heinzelmann F, Velalakan A, Budach W, Belka C and Jendrossek V: Radiation-induced changes in breathing frequency and lung histology of C57BL/6J mice are time- and dose-dependent. Strahlenther Onkol. 188:274–281. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Ma J, Zhang J, Zhou S, et al: Regional lung density changes after radiation therapy for tumors in and around thorax. Int J Radiat Oncol Biol Phys. 76:116–122. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Jia S and Meng A: Tob genes in development and homeostasis. Dev Dyn. 236:913–921. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Yanagie H, Tanabe T, Sumimoto H, et al: Tumor growth suppression by adenovirus-mediated introduction of a cell-growth-suppressing gene tob in a pancreatic cancer model. Biomed Pharmacother. 63:275–286. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Tzachanis D and Boussiotis VA: Tob, a member of the APRO family, regulates immunological quiescence and tumor suppression. Cell Cycle. 8:1019–1025. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Kitagawa K, Kotake Y and Kitagawa M: Ubiquitin-mediated control of oncogene and tumor suppressor gene products. Cancer Sci. 100:1374–1381. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Helms MW, Kemming D, Contag CH, et al: TOB1 is regulated by EGF-dependent HER2 and EGFR signaling, is highly phosphorylated, and indicates poor prognosis in node-negative breast cancer. Cancer Res. 69:5049–5056. 2009. View Article : Google Scholar : PubMed/NCBI

9 

O’Malley S, Su H, Zhang T, Ng C, Ge H and Tang CK: TOB suppresses breast cancer tumorigenesis. Int J Cancer. 125:1805–1813. 2009.

10 

Jiao Y, Ge CM, Meng QH, Cao JP, Tong J and Fan SJ: Adenovirus-mediated expression of Tob1 sensitizes breast cancer cells to ionizing radiation. Acta Pharmacol Sin. 28:1628–1636. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Jiao Y, Xu JY, Che J and Fan SJ: Study on the effects of anti-proliferative protein Tob1 on the radio-sensitivity of human cervix cancer cell line HeLa. J Radiat Res Radiat Proc. 28:193–196. 2010.(In Chinese).

12 

Jiao Y, Sun KK, Zhao L, Xu JY, Wang LL and Fan SJ: Suppression of human lung cancer cell proliferation and metastasis in vitro by the transducer of ErbB-2.1 (TOB1). Acta Pharmacol Sin. 33:250–260. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Gannon HS, Woda BA and Jones SN: ATM phosphorylation of Mdm2 Ser394 regulates the amplitude and duration of the DNA damage response in mice. Cancer Cell. 21:668–679. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Guckenberger M, Baier K, Polat B, et al: Dose-response relationship for radiation-induced pneumonitis after pulmonary stereotactic body radiotherapy. Radiother Oncol. 97:65–70. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Hart JP, McCurdy MR, Ezhil M, et al: Radiation pneumonitis: correlation of toxicity with pulmonary metabolic radiation response. Int J Radiat Oncol Biol Phys. 71:967–971. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Nokisalmi P, Rajecki M, Pesonen S, et al: Radiation-induced upregulation of gene expression from adenoviral vectors mediated by DNA damage repair and regulation. Int J Radiat Oncol Biol Phys. 83:376–384. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Parplys AC, Petermann E, Petersen C, Dikomey E and Borgmann K: DNA damage by X-rays and their impact on replication processes. Radiother Oncol. 102:466–471. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Neumaier T, Swenson J, Pham C, et al: Evidence for formation of DNA repair centers and dose-response nonlinearity in human cells. Proc Natl Acad Sci USA. 109:443–448. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Hlatky L: Double-strand break motions shift radiation risk notions. Proc Natl Acad Sci USA. 109:351–352. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Schneider L, Fumagalli M and d’Adda dFF: Terminally differentiated astrocytes lack DNA damage response signaling and are radioresistant but retain DNA repair proficiency. Cell Death Differ. 19:582–591. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zlobinskaya O, Dollinger G, Michalski D, et al: Induction and repair of DNA double-strand breaks assessed by gamma-H2AX foci after irradiation with pulsed or continuous proton beams. Radiat Environ Biophys. 51:23–32. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Khoronenkova SV, Dianova II, Ternette N, Kessler BM, Parsons JL and Dianov GL: ATM-dependent downregulation of USP7/HAUSP by PPM1G activates p53 response to DNA damage. Mol Cell. 45:801–813. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Shouse GP, Nobumori Y, Panowicz MJ and Liu X: ATM-mediated phosphorylation activates the tumor-suppressive function of B56gamma-PP2A. Oncogene. 30:3755–3765. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Cheng Q, Cross B, Li B, Chen L, Li Z and Chen J: Regulation of MDM2 E3 ligase activity by phosphorylation after DNA damage. Mol Cell Biol. 31:4951–4963. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Gajjar M, Candeias MM, Malbert-Colas L, et al: The p53 mRNA-Mdm2 interaction controls Mdm2 nuclear trafficking and is required for p53 activation following DNA damage. Cancer Cell. 21:25–35. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Chiu YJ, Hour MJ, Lu CC, et al: Novel quinazoline HMJ-30 induces U-2 OS human osteogenic sarcoma cell apoptosis through induction of oxidative stress and up-regulation of ATM/p53 signaling pathway. J Orthop Res. 29:1448–1456. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Yang Y, Xia F, Hermance N, et al: A cytosolic ATM/NEMO/RIP1 complex recruits TAK1 to mediate the NF-kappaB and p38 mitogen-activated protein kinase (MAPK)/MAPK-activated protein 2 responses to DNA damage. Mol Cell Biol. 31:2774–2786. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Rutkowski R, Dickinson R, Stewart G, et al: Regulation of Caenorhabditis elegans p53/CEP-1-dependent germ cell apoptosis by Ras/MAPK signaling. PLoS Genet. 7:e10022382011.PubMed/NCBI

Related Articles

Journal Cover

August 2013
Volume 30 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
Che J, Lu Y, Sun K, Feng C, Dong A and Jiao Y: Overexpression of TOB1 confers radioprotection to bronchial epithelial cells through the MAPK/ERK pathway. Oncol Rep 30: 637-642, 2013
APA
Che, J., Lu, Y., Sun, K., Feng, C., Dong, A., & Jiao, Y. (2013). Overexpression of TOB1 confers radioprotection to bronchial epithelial cells through the MAPK/ERK pathway. Oncology Reports, 30, 637-642. https://doi.org/10.3892/or.2013.2536
MLA
Che, J., Lu, Y., Sun, K., Feng, C., Dong, A., Jiao, Y."Overexpression of TOB1 confers radioprotection to bronchial epithelial cells through the MAPK/ERK pathway". Oncology Reports 30.2 (2013): 637-642.
Chicago
Che, J., Lu, Y., Sun, K., Feng, C., Dong, A., Jiao, Y."Overexpression of TOB1 confers radioprotection to bronchial epithelial cells through the MAPK/ERK pathway". Oncology Reports 30, no. 2 (2013): 637-642. https://doi.org/10.3892/or.2013.2536