Skip to main content
Erschienen in: Radiation Oncology 1/2015

Open Access 01.12.2015 | Research

RETRACTED ARTICLE: IL-6 signaling promotes DNA repair and prevents apoptosis in CD133+ stem-like cells of lung cancer after radiation

verfasst von: Yuhchyau Chen, Fuquan Zhang, Ying Tsai, Xiadong Yang, Li Yang, Shanzhou Duan, Xin Wang, Peter Keng, Soo Ok Lee

Erschienen in: Radiation Oncology | Ausgabe 1/2015

Abstract

Background

Local tumor control by standard fractionated radiotherapy (RT) remains poor because of tumor resistance to radiation (radioresistance). It has been suggested that cancer stem cells (CSCs) are more radioresistant than non-CSCs. In previous studies, we have shown IL-6 promotes self-renewal of CD133+ CSC-like cells. In this study, we investigated whether IL-6 plays roles not only in promoting self-renewal of CD133+ cells after radiation, but also in conferring radioresistance of CD133+ cells in NSCLC.

Materials and methods

To compare radiation sensitivity of CSCs and non-CSCs, CD133+ CSC-like and CD133- cell populations were isolated from two NSCLC cell lines, A549 and H157, by immunomagnetic separation and their sensitivities to ionizing radiation were investigated using the clonogenic survival assay. To further study the IL-6 effect on the radiosensitivity of CD133+ CSC-like cells, CD133+ cells were isolated from A549IL-6si/sc and H157IL-6si/sc cells whose intracellular IL-6 levels were manipulated via the lentiviral transduction with IL-6siRNA. Post-irradiation DNA damage was analyzed by γ-H2AX staining and Comet assay. Molecular mechanisms by which IL-6 regulates the molecules associated with DNA repair and anti-apoptosis after radiation were analyzed by Western blot and immunofluoresecence (IF) staining analyses.

Results

NSCLC CD133+ CSC-like cells were enriched upon radiation. Survival of NSCLC CD133+ cells after radiation was higher than that of CD133- cells. Survival of IL-6 expressing NSC LC CD133+ cells (sc) was higher than that of IL-6 knocked-down cells (IL-6si) after radiation. IL-6 played a role in protecting NSCLC CD133+ cells from radiation-induced DNA damage and apoptosis.

Conclusions

IL-6 signaling promotes DNA repair while protecting CD133+ CSC-like cells from apoptotic death after radiation for lung cancer. A combined therapy of radiation and agents that inhibit IL-6 signaling (or its downstream signaling) is suggested to reduce CSC-mediated radioresistance in lung cancer.
Hinweise
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s13014-022-02030-5.
This article has been retracted. Please see the retraction notice for more detail:https://​doi.​org/​10.​1186/​s13014-022-02030-5

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SL and YC conceived the study, participated in its design and coordination, performed the statistical analysis and drafted the manuscript. SD, FZ, YT, XW, and YL participated in experiments, analysis, and interpretation of data. PK and YC critically reviewed the article. All authors read and approved the final manuscript.
Abkürzungen
IL-6
Interleukin-6
NSCLC
Non-small cell lung cancer
SCLC
Small cell lung cancer
RT
Radiotherapy
CSC
Cancer stem cell
A549IL-6si
A549/IL-6 knocked down with IL-6siRNA
A549sc
A549 scramble control
H157IL-6si
H157/IL-6 knocked down with IL-6siRNA
ELISA
Enzyme-linked immunosorbent assay
ATM
Ataxia telangiectasia mutated
CHK
Checkpoint
ECL
Electrochemiluminescence
IF
Immunofluorescence
MAPK
Mitogen-activated protein kinase

Background

Lung cancer is the predominant cause of cancer death in both men and women [1]. It is heterogeneous and histologically divided into two types: small cell lung carcinomas (SCLCs) and non-small cell lung carcinomas (NSCLCs), with the latter comprising 85 % of lung cancer cases [2]. Radiotherapy (RT) is the standard primary treatment for patients diagnosed with localized unresectable NSCLC. However, local tumor control by standard fractionated RT remains poor primarily due to tumor resistance to radiation.
Accumulating evidence indicates that cancer stem cells (CSCs) exist as a very minor population in NSCLC tumors [36]. It has been suggested by several investigators that CSCs are more radioresistant than non-CSCs. Hittelman et al. [7] and Zhang et al. [8] showed that cancer cell colonies surviving radiation treatment exhibited stem cell features, and Gomez-Casal et al. [9] reported that NSCLC cells surviving radiation treatment displayed CSC and epithelial-mesenchymal transition (EMT) phenotypes. In addition, Baumann et al. [10] suggested that local tumor control by RT was affected by the number of CSCs in the tumors.
According to Hittelman et al. [7], radioresistance can be influenced by different intrinsic and extrinsic factors, including proliferation or quiescence, activated radiation response mechanisms (e.g., enhanced DNA repair, upregulated cell cycle control, and increased free-radical scavengers), and a surrounding microenvironment that enhances cell survival (e.g., hypoxia and interaction with stromal elements).
Implications of cytokines in radioresistance have been suggested. Liu et al. [11] reported that the IL-6 class cytokine leukemia inhibitory factor (LIF) promoted radioresistance of nasopharyngeal carcinoma. It was also shown that the inhibition of IL-4 or IL-10 resulted in sensitization of colorectal cancer cells to radiation [12]. In addition, Zhou et al. [13] suggested that cytokines can shift the balance between tumor cells and tumor microenvironment after irradiation.
Our laboratory recently found that IL-6 played a promoter role in the self-renewal of CD133+, CSC-like cells in A549 and H157 cell lines, not in CD133- cells (manuscript submitted), demonstrating that it may promote growth of the surviving CD133+ CSC-like cells after radiation. We further investigated whether IL-6 plays roles not only in promoting self-renewal of CD133+ cells, but also in conferring radioresistance of CD133+ cells in NSCLC.

Methods

Cell culture

A549 and H157 cell lines were purchased from the American Type Culture Collection (ATCC, Manassas, VA) and cultured in RPMI 1640 containing 10 % FBS. CD133+ CSCs were cultured in DMEM/F12 medium supplemented with 20 ng/ml EGF (Invitrogen), and 20 ng/ml FGF (Invitrogen). All cells were maintained in a humidified 5 % CO2 environment at 37 °C.

Flow cytometric analysis of CD133+ cells after radiation

A549 and H157 cell lines were irradiated with 6 Gy Cs-137 gamma rays and then grown as a monolayer culture for 7 days. After 7 days, the percentage of CD133+ population (CD133+ positively stained cells) in the culture was determined by flow cytometric analysis using the Canto II system (Becton-Dickinson, San Antonio, TX). The non-irradiated cells were used as control.

Development of IL-6 knocked down and sc control cells by lentiviral transduction

For incorporation of IL-6 siRNA or scramble (sc) control plasmids into A549 and H157 cells, lentivirus construct carrying either sc or IL-6 siRNA (pLenti-II vector, Applied Biological Materials Inc, Canada) was transfected into 293 T cells with a mixture of pLent-II-IL-6 siRNA, psPAX2 (virus-packaging plasmid), and pMD2G (envelope plasmid) (4:3:2 ratio) using PolyFect Transfection reagent (Qiagen, Valencia, CA). After A549 and H157 cells were infected with virus overnight, the culture media containing the virus were removed and the infected cells were then maintained under normal cell culture media. After sub-culturing, the IL-6 knocked down cells were selected by puromycin (2 μg/ml) (Sigma) and then maintained in media containing 0.1 μg/ml puromycin.

Isolation of CD133+ CSC-like cells using immunomagnetic separation techniques

Cells (2 × 107) were detached from tissue culture plates with 5 mM EDTA, centrifuged, and incubated with magnetic microbeads conjugated with anti-CD133 antibody (Miltenyi Biotec, Cambridge, MA). The bead-bound cells (CD133+) and unbound cells (CD133-) were separated using the Quadro MACSTM Separation Unit (Miltenyi Biotec, Cambridge, MA). The purity of isolated CD133+ cells was confirmed by flow cytometric analyses, and by qPCR analyses. The isolated CD133+ cells were then cultured in stem cell media.

Sphere formation assay

For sphere formation assays, single-cell suspensions (1 × 103 cells) were mixed with cold Matrigel (BD, Franklin Lakes) (1:1 ratio, v/v, total volume of 100 μl)) and the mixture was placed along the rim of the 24-well plates. The culture plates were placed in 37 °C incubator for 10 min to let the mixture solidify, and 500 μl medium was then added into the wells. The number of spheres with diameter greater than 50 μm was counted 7–14 days later using an Olympus light microscope. A minimum of three triplicate experiments were performed.

Cell survival assay after radiation

Cells were exposed to different doses (0, 1, 2, 4, 6, and 8 Gy) of radiation using a Cs-137 source with a dose rate of 180–205 cGy/min. After treatment, clonogenic assay was performed as previously described [14]. Cells were seeded in culture dishes with appropriate dilutions to form colonies after 7–9 days incubation. Colonies were fixed with methanol, stained with crystal violet (0.5 % w/v), and counted using a microscope. Colonies consisting of at least 50 cells were counted and the surviving fraction was calculated from the normalized plating efficiency.

RNA Extraction and Quantitative Real-Time PCR (qPCR) Analysis

Total RNAs were isolated using Trizol reagent (Invitrogen). One μg of total RNA was subjected to reverse transcription using Superscript III transcriptase (Invitrogen). qPCR was conducted using the appropriate primers and the Bio-Rad CFX96 system. SYBR green was used to determine the expression levels of mRNA from genes of interest. Expression levels were normalized to GAPDH level.

IL-6 ELISA

IL-6 in the supernatant of unseparated parental or isolated CD133- and CD133+ cells of A549IL-6si/sc and H157IL-6si/sc pairs was determined by the ELISA kit according to the manufacturer’s instructions (BD, Franklin Lakes). The secreted IL-6 level was normalized by cell number.

Western blot analysis

Cells were lysed in RIPA buffer (50 mM Tris-Cl at pH 7.5, 150 mM NaCl, 1 % NP-40, 0.5 % sodium deoxycholate, 1 mM EDTA, 1 μg/mL leupeptin, 1 μg/mL aprotinin, 0.2 mM PMSF) and proteins (20–40 μg) were isolated and separated on 8–10 % SDS/PAGE gel and then transferred onto PVDF membranes (Millipore, Billerica, MA). After blocking procedure, membranes were incubated with primary antibodies, followed by HRP-conjugated secondary antibodies, and then the proteins of interest were visualized using the Imager (Bio-Rad) and ECL (Thermo Fisher Scientific, Rochester, NY) system. Antibody of GAPDH was purchased from Abcam (Cambridge, MA) and antibodies of Mcl-1and cleaved caspase-3 were obtained from Cell Signaling (Danvers, MA). Bcl-2 antibody was obtained from Santa Cruz (Santa Cruz, CA).

Immunofluoresence (IF) staining

Unseparated parental or isolated CD133- and CD133+ cells of A549IL-6si/sc and H157IL-6si/sc pairs (1 x 103) were mounted on chamber slide, irradiated (6 Gy, with non-irradiated cells as control), and stained with appropriate primary antibodies. Antibodies of ATM and CHK2 were obtained from Bethyl Laboratory (Montgomery, TX), phosphorylated ATM (Ser 1981), and phosphorylated p53 (Ser 20) were from Gene Tex (Irvine CA), and γ-H2AX antibody was purchased from Trevigen (Gaithersburg, MD). Antibodies of Mcl-1 and Bcl-2 were obtained from Cell Signaling (Danvers, MA) and Santa Cruz (Santa Cruz, CA), respectively. After reaction with Alexa flour® 488 anti-goat secondary antibody (Life Technologies, Grand Island, NY), images were recorded using a fluorescent microscope (Zeiss, Germany).

Comet assay

Isolated CD133+ and CD133- cells of IL-6si/sc pairs were irradiated (6 Gy) and at 0 and 30 min after radiation cells were used in the assay following the procedure of Singh et al. [15] with some modifications. Briefly, cells were embedded in low-melting-point agarose in lysis buffer (10 mM Tris–HCl, pH 10, 2.5 M NaCl, 100 mM EDTA, 10 % DMSO, 1 % Triton X-100). The unwinding step was performed for 20 min at 4 °C in unwinding/electrophoresis buffer (300 mM NaOH, 1 mM EDTA, pH = 13). Electrophoresis was performed at 4 °C for 20 min in unwinding/electrophoresis buffer at electric field strength of 25 V and a current of 300 mA. The slides were then neutralized with a neutralizing buffer (0.4 Tris–HCl, pH 7.5) for 20 min, rinsed with distilled water, air-dried, stained with 30 μg/ml ethidium bromide. Images were recorded using a fluorescent microscope (Zeiss, Germany).

ATM-luciferase assay

293HEK and H1299 cells in 24 well plates were transfected with 2 μg/mL ATM reporter plasmid (Addgene, Cambridge, MA) and 0.02 μg/mL phRL-cytomegalovirus Renilla luciferase plasmid (used as control for normalizing transfection efficiencies) using Polyfect (Qiagen, Valencia, CA). After transfection, cells were incubated with or without IL-6. Twenty-four hours later, luciferase activities were measured using the Dual-Luciferase Reporter Assay System (Promega, Madison Wisconsin) according to manufacturer’s instructions. Luciferase activity was measured using theGloMax® 20/20 luminometer (Promega, Madison, WI). For data analysis, the experimental reporter was normalized to the level of constitutive reporter to adjust for the differences in transfection efficiency.

Statistics

The data were presented as the mean ± SEM. Differences in mean values between two groups were analyzed by two-tailed Student’s t test. p ≤ 0.05 was considered statistically significant.

Results

CD133+, CSC-like cells were enriched in A549 and H157 cell cultures upon exposure to radiation

To investigate whether the population of CSCs in NSCLC was altered after radiation, unseparated A549 and H157 parental cells were irradiated with 6 Gy gamma rays and the percentage of CD133+ cells, before and after radiation, was analyzed by flow cytometry. The CD133 molecule was chosen because it is the most widely used surface marker for the CSC of NSCLC [4, 6]. As shown in Fig. 1a, the percentage of CD133+ cells was increased by 3.4 and 4.0 fold at 7 days after irradiation for A549 and H157 cells, respectively. Consistent with the CD133+ population data (Fig. 1a) and the published result by others [9], we also observed a higher number of spheres in the sphere formation assay (Fig. 1b) and expression of CSC markers in NSCLC cell lines following irradiation in IF staining (Fig. 1c).

CD133+, CSC-like cells were more resistant to radiation than CD133- cells

Emerging evidence suggests that CSCs are likely to be more resistant to radiation than non-CSCs [710, 16]. To directly compare the radiation sensitivity of CSC and non-CSC cells in NSCLC, we isolated CD133+ and CD133- cells from A549 and H157 cells by anti-CD133 antibody and the immunomagnetic separation method. Flow cytometric analyses indicated an enrichment of CD133+ cells after separation with a purity of CD133+ greater than 90 % (Fig. 2a). In order to confirm the CSC-like characteristics of the isolated CD133+ cells, we analyzed expression of CSC markers in these cells and in parental cells. The results showed a dramatic increase in CSC markers in CD133+ cells (Fig. 2b, A549 cells). The CD133+ cells were able to grow into spheres using the Matrigel-based sphere formation assay (Fig. 2c), further indicating the isolated CD133+ cells have CSC-like features. The CD133+ cells were maintained under non-adherent culture conditions with stem cell media, and the freshly isolated cells and once passaged cells were used in the entire study.
Radiation sensitivity was determined and compared between the isolated CD133+ and CD133- cells. CD133+ and CD133- cells were isolated by an immunomagnetic separation method one day prior to irradiation experiments and the CD133+ cells were kept in stem cell media. After irradiation to different doses of Cs-137 gamma rays, cells were harvested, trypsinized (in case of CD133- cells), cell numbers counted, and single cell suspensions of indicated numbers of CD133- and CD133+ cells were used in clonogenic assay at 7–9 days post-radiation. We found survival of CD133+ cells was significantly higher than that of CD133- cells in both cell lines (Fig. 2d; top, A549 cells; bottom, H157 cells), indicating that CD133+ cells are more radioresistant than CD133- cells.

Effects of intracellular level of IL-6 on mediating survival of CD133+ cells upon irradiation

In investigating the role of IL-6 in the radiation sensitivity of CSCs, the A549 and H157 cells secreted a high level of IL-6, so it was hard to observe the exogenously added IL-6 effect on survival of CD133+ cells after radiation. So, we investigated the radiation survival of CD133+ CSC-like cells obtained from parental cells whose intracellular IL-6 level was manipulated by the IL-6 siRNA in lentiviral transduction system. We have previously obtained IL-6 knocked down A549 (A549IL-6si) and H157 (H157IL-6si) cells and their corresponding scramble (sc) control (A549sc, H157sc) cells. The IL-6 knockdown efficiency was greater than 90 % (A549) and 72 % (H157) in qPCR analyses (Fig. 3a). The IL-6 knockdown in CD133- and CD133+ cells are shown in ELISA test (Fig. 3b) and IF staining (CD133+ cells, Fig. 3c). CD133+ cells were then isolated from these A549IL-6/sc and H157IL-6si/sc pairs and exposed to various doses of radiation for clonogenic cell survival assay. After exposure to radiation, we found that the survival of CD133+ cells isolated from IL-6 expressing A549sc and H157sc cells was higher than from A549IL-6si and H157IL-6si cells (Fig. 3d), suggesting that intracellular IL-6 is important for maintaining a greater level of radiation survival in CD133+ cells.
We next tested whether IL-6 was also involved in promoting the self-renewal of CD133+ cells after radiation. We irradiated the CD133+ cells isolated from A549IL-6si/sc and H157IL-6si/sc pairs and performed the sphere formation assay at 7 days after irradiation to test their self-renewal abilities. As shown in Fig. 3e, we observed a higher number of spheres from A549sc and H157sc cells than those from A549IL-6si and H157IL-6si cells. Consistent with the spheroid formation results, a higher number of Ki67 positive cells was found in CD133+ of A549sc cells following radiation exposure (Fig. 3f), indicating higher proliferating activity of IL-6 expressing CD133+ cells.The secreted IL-6 level differences in CD133+ and CD133- cells of A549IL-6si/sc and H157IL-6si/sc pairs are shown in Fig. 3g.

IL-6 was important in mediating DNA repair and protecting cells from apoptotic death in CD133+ cells after radiation damage

We then investigated whether DNA damage and repair system in CD133+ cells was affected by IL-6 signaling following irradiation. We irradiated CD133+ cells isolated from A549IL-6si/sc and H157IL-6si/sc pairs and performed the IF staining of γ-H2AX after irradiation. As shown in Fig. 4a, we detected higher levels of γ-H2AX staining in A549IL-6si and H157IL-6si cells, indicating greater amount of DNA damage still left unrepaired when IL-6 was knocked down in CD133+ cells. Different levels of post-irradiation DNA strand breaks in CD133- and CD133+ cells of A549IL-6si/sc and H157IL-6si/sc pairs were also investigated in the Comet assay. When CD133- and CD133+ cells were isolated from A549IL-6si/sc and H157IL-6si/sc pairs and irradiated at 6 Gy, we detected a similar extent of DNA damage in the CD133- and CD133+ cells of IL-6si/sc pair immediately after radiation, but lower DNA breaks in CD133+ cells of IL-6sc cell lines than those of IL-6si cell lines were detected at 30 min after radiation (Fig. 4b), suggesting higher DNA repair in CD133+ cells of IL-6sc cell lines than those of IL-6si cell lines. However, we did not observe a significant difference in DNA repair in CD133- cells of A549IL-6si/sc and H157IL-6si/sc pairs.
We then investigated whether the higher level of unrepaired DNA damage in IL-6 knocked down cells was due to lower DNA repair activity. We investigated expression of ATM [17] and its downstream molecules, including ATM, phosphorylated ATM, CHK2, and phosphorylated p53, in CD133- and CD133+ cells of A549IL-6si/sc and H157IL-6si/sc cell pairs upon radiation. The results of IF staining showed that the CD133+ cells from A549IL-6si and H157IL-6si cell lines exhibited lower expression of these molecules than those isolated from sc cell lines (Fig. 4c). However, not much difference was observed in CD133- cells, whether isolated from IL-6si or sc cell lines.
To further explore whether the ATM and CHK2 are up-regulated in IL-6 expression CD133+ cells after radiation, we compared the mRNA levels of ATM and CHK2 in A549IL-6si/sc and H157IL-6si/sc pairs following radiation exposure. Similar to the IF staining results, higher levels of ATM mRNA were observed in the IL-6 expressing and CD133+ sc cells compared to those of IL-6 knocked down cells (Fig. 4d).
Next, we investigated whether IL-6 regulates ATM at the transcriptional level in the 293HEK cell line using the ATM-luciferase constructs containing the ATM promoter region. We also used non-IL-6 expressing H1299 NSCLC cell line in this assay to observe the exogenously added IL-6 effect. We detected a dose dependent regulation of ATM-luciferase by IL-6 in these two cell lines, suggesting direct regulation of IL-6 on ATM molecule at the transcriptional level (Fig. 4e).
We then investigated whether difference in expression of apoptosis markers could be detected between IL-6 expressing (sc) and knocked down (IL-6si) CD133+ cells. As shown in Fig. 5a, we observed a higher level of cleaved caspase in A549IL-6si cells than A549sc cells, indicating more apoptosis in IL-6 knocked down cells following radiation. When we examined expression of anti-apoptotic markers Bcl-2 and Mcl-1 in the A549IL-6si/sc cell line pair, higher expression of these molecules in IL-6 expressing CD133+ cells were observed in Western blot (Fig. 5a) and IF staining (Fig. 5b) analyses. These results suggest that IL-6 in CD133+ cells modulated up-regulations of these molecules, and protected cells from apoptotic death upon irradiation.
In summary, [1] NSCLC CD133+ CSC-like cells were enriched upon radiation, [2] cell survival of NSCLC CD133+ cells after radiation was higher than that of CD133- cells, [3] cell survival of IL-6 expressing NCSLC CD133+ cells (sc) was higher than that of IL-6 knocked-down cells (IL-6si) after radiation [4] IL-6 played a role in protecting NSCLC CD133+ cells from radiation-induced DNA damage and apoptosis.

Discussion

We first demonstrated that CD133+ CSC-like cells were enriched after radiation in NSCLC cells. This result is consistent with previous reports by Desai et al. [18] showing an increase of CSC in A549 cell line after radiation, and by Gomez-Casal [9] showing an expansion of sphere numbers and increase of CSC marker expression after radiation.
We then investigated the survival of CSCs and non-CSCs upon radiation since a direct comparison showing the difference between CSC and non-CSC originated from same lung cancer cell line has not been studied before. Our in vitro cell survival results clearly demonstrated that the CD133+ cells had higher survival than CD133- cells after radiation (Fig. 2), which is clear evidence suggesting that CSCs are more radioresistant than non-CSCs.
Regarding the molecular mechanisms by which CSCs exhibit higher radioresistance than non-CSCs, Pajonk et al. [19] suggested that the CSC is inherently radioresistant. Matthews et al. [20] proposed that CSC has higher expression of radioresistance-related genes and higher DNA repair ability. However, it is widely accepted that the other factors such as adaptive responses in CSC and microenvironmental changes upon irradiation can contribute to radioresistance in CSCs [21]. Bao et al. [22] showed that glioma stem cells promote radioresistance by preferential activation of the DNA damage response. In addition, several signaling pathways were suggested to be involved in radioresistance of CSCs. Piao et al. [16] showed increased activation of MAPK/PI3K signaling pathway and reduction in reactive oxygen species levels in CD133+ cells of human hepatocarcinoma compared to CD133- cells upon irradiation. Meanwhile, Ettl et al. [23] showed AKT and MET signaling mediates anti-apoptotic radioresistance in head neck cancer cell lines, and Kim et al. [24] suggested that EZH2 is important in radioresistance of CSC in glioblastoma.
In this study, we suggest that IL-6 signaling may be important in promoting radioresistance in NSCLC CD133+ cells. We speculate that intracellular IL-6 may be more critical in protecting cells from radiation-induced damage since we observed higher radioresistance of sc cells compared to IL-6si cells, but could not detect significant effect when IL-6 was added exogenously to the non-IL-6 expressing H1299 cells. Contribution of IL-6 in radioprotection has been suggested previously. In animal studies, Neta et al. [25] showed reduced mortality upon irradiation when mice were pre-treated with IL-6 antibody. In addition, Wu et al. [26] showed that IL-6 plays a role in radioresistance of castration resistant prostate cancer. However, no clear IL-6 role had been addressed in protection of NSCLC CSCs from radiation. In our study, we clearly demonstrated the IL-6 role in mediating radioresistance of NSCLC CD133+ cells.
We suggested that the effect of IL-6 in mediating radioresistance is partially arbitrated through regulation of DNA repair related molecules. Desai et al. [18] also suggested that the radioresistance in CD133+ cells is gone through DNA repair molecules, such as Exo1 and Rad51. Using several different assays, we showed the regulation of IL-6 on the key molecules of DNA repair, ATM and CHK, in CD133+ cells. This result is consistent with the recent observation showing IL-6 regulation of ATM/NFkB signaling in conferring the resistance of lung cancer to chemotherapy [27]. Although we have only studied IL-6 regulation on ATM and CHK, identifying the IL-6 effect on regulation of other DNA repair associated molecules will be carried out in future studies.
In this study, we showed that IL-6 may modulate ATM and CHK at the transcriptional level. However, it may also be possible that the IL-6 effect can go through signaling pathways which is downstream of IL-6 signaling as the ATM level was suggested to be modulated by signaling pathways, such as Akt and Erk [28]. Therefore, whether IL-6 regulates their activation by mediating up-regulation of these molecules should be tested.
In addition to the modulation of DNA repair mechanism, we showed that IL-6 regulated expression of anti-apoptotic proteins Bcl-2 and Mcl-1. The IL-6 regulation of Bcl-2 and Mcl-1 in CD133+ cells, without the radiation effect, has been shown in our previous studies (manuscript submitted). Whether this regulation can be accelerated after radiation will be tested.

Conclusions

Our data suggest that IL-6 contributes to radioresistance of CD133+ CSC-like cells in NSCLC by protecting them against radiation-induced DNA damage and apoptotic death. In light of the high recurrence rate of NSCLC after RT, and considering CSC as the target population with high radioresistance, we suggest a combination of radiation and agents that inhibit IL-6 signaling (or its downstream signaling) to reduce CSC-mediated radioresistance in lung cancer.

Acknowledgement

We thank Mrs. Laura Finger for her editorial assistance as an employee of the Department of Radiation Oncology at the University of Rochester.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SL and YC conceived the study, participated in its design and coordination, performed the statistical analysis and drafted the manuscript. SD, FZ, YT, XW, and YL participated in experiments, analysis, and interpretation of data. PK and YC critically reviewed the article. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Cersosimo RJ. Lung cancer: a review. Am J Health Syst Pharm. 2002;59(7):611–42. PubMed Epub 2002/04/12. eng.CrossRef Cersosimo RJ. Lung cancer: a review. Am J Health Syst Pharm. 2002;59(7):611–42. PubMed Epub 2002/04/12. eng.CrossRef
2.
Zurück zum Zitat Parsons A, Daley A, Begh R, Aveyard P. Influence of smoking cessation after diagnosis of early stage lung cancer on prognosis: systematic review of observational studies with meta-analysis. BMJ. 2010;340:b5569. PubMed Pubmed Central PMCID: 2809841, Epub 2010/01/23. eng.CrossRef Parsons A, Daley A, Begh R, Aveyard P. Influence of smoking cessation after diagnosis of early stage lung cancer on prognosis: systematic review of observational studies with meta-analysis. BMJ. 2010;340:b5569. PubMed Pubmed Central PMCID: 2809841, Epub 2010/01/23. eng.CrossRef
3.
Zurück zum Zitat Akunuru S, James Zhai Q, Zheng Y. Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations and exhibit plasticity. Cell death & disease. 2012;3, e352. PubMed Pubmed Central PMCID: 3406592.CrossRef Akunuru S, James Zhai Q, Zheng Y. Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations and exhibit plasticity. Cell death & disease. 2012;3, e352. PubMed Pubmed Central PMCID: 3406592.CrossRef
4.
Zurück zum Zitat Bertolini G, Roz L, Perego P, Tortoreto M, Fontanella E, Gatti L, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci U S A. 2009;106(38):16281–6. PubMed Pubmed Central PMCID: 2741477.CrossRef Bertolini G, Roz L, Perego P, Tortoreto M, Fontanella E, Gatti L, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci U S A. 2009;106(38):16281–6. PubMed Pubmed Central PMCID: 2741477.CrossRef
5.
Zurück zum Zitat Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15(3):504–14. PubMed Epub 2007/12/01. eng.CrossRef Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15(3):504–14. PubMed Epub 2007/12/01. eng.CrossRef
6.
Zurück zum Zitat Wang S, Xu ZY, Wang LF, Su W. CD133+ cancer stem cells in lung cancer. Front Biosci. 2013;18:447–53. PubMed.CrossRef Wang S, Xu ZY, Wang LF, Su W. CD133+ cancer stem cells in lung cancer. Front Biosci. 2013;18:447–53. PubMed.CrossRef
7.
Zurück zum Zitat Hittelman WN, Liao Y, Wang L, Milas L. Are cancer stem cells radioresistant? Future Oncol. 2010;6(10):1563–76. PubMed Pubmed Central PMCID: 3059151.CrossRef Hittelman WN, Liao Y, Wang L, Milas L. Are cancer stem cells radioresistant? Future Oncol. 2010;6(10):1563–76. PubMed Pubmed Central PMCID: 3059151.CrossRef
8.
Zurück zum Zitat Zhang X, Komaki R, Wang L, Fang B, Chang JY. Treatment of radioresistant stem-like esophageal cancer cells by an apoptotic gene-armed, telomerase-specific oncolytic adenovirus. Clin Cancer Res. 2008;14(9):2813–23. PubMed Pubmed Central PMCID: 2387204.CrossRef Zhang X, Komaki R, Wang L, Fang B, Chang JY. Treatment of radioresistant stem-like esophageal cancer cells by an apoptotic gene-armed, telomerase-specific oncolytic adenovirus. Clin Cancer Res. 2008;14(9):2813–23. PubMed Pubmed Central PMCID: 2387204.CrossRef
9.
Zurück zum Zitat Gomez-Casal R, Bhattacharya C, Ganesh N, Bailey L, Basse P, Gibson M, et al. Non-small cell lung cancer cells survived ionizing radiation treatment display cancer stem cell and epithelial-mesenchymal transition phenotypes. Mol Cancer. 2013;12(1):94. PubMed Pubmed Central PMCID: 3751356.CrossRef Gomez-Casal R, Bhattacharya C, Ganesh N, Bailey L, Basse P, Gibson M, et al. Non-small cell lung cancer cells survived ionizing radiation treatment display cancer stem cell and epithelial-mesenchymal transition phenotypes. Mol Cancer. 2013;12(1):94. PubMed Pubmed Central PMCID: 3751356.CrossRef
10.
Zurück zum Zitat Baumann M, Krause M, Hill R. Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer. 2008;8(7):545–54. PubMed.CrossRef Baumann M, Krause M, Hill R. Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer. 2008;8(7):545–54. PubMed.CrossRef
11.
Zurück zum Zitat Liu SC, Tsang NM, Chiang WC, Chang KP, Hsueh C, Liang Y, et al. Leukemia inhibitory factor promotes nasopharyngeal carcinoma progression and radioresistance. J Clin Invest. 2013;123(12):5269–83. PubMed Pubmed Central PMCID: 3859424.CrossRef Liu SC, Tsang NM, Chiang WC, Chang KP, Hsueh C, Liang Y, et al. Leukemia inhibitory factor promotes nasopharyngeal carcinoma progression and radioresistance. J Clin Invest. 2013;123(12):5269–83. PubMed Pubmed Central PMCID: 3859424.CrossRef
12.
Zurück zum Zitat Voboril R, Weberova-Voborilova J. Sensitization of colorectal cancer cells to irradiation by IL-4 and IL-10 is associated with inhibition of NF-kappaB. Neoplasma. 2007;54(6):495–502. PubMed.PubMed Voboril R, Weberova-Voborilova J. Sensitization of colorectal cancer cells to irradiation by IL-4 and IL-10 is associated with inhibition of NF-kappaB. Neoplasma. 2007;54(6):495–502. PubMed.PubMed
13.
Zurück zum Zitat Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol. 2015;141(4):575–89. PubMed.CrossRef Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol. 2015;141(4):575–89. PubMed.CrossRef
14.
Zurück zum Zitat Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9. PubMed.CrossRef Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9. PubMed.CrossRef
15.
Zurück zum Zitat Singh NP, McCoy MT, Tice RR, Schneider EL. A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res. 1988;175(1):184–91. PubMed.CrossRef Singh NP, McCoy MT, Tice RR, Schneider EL. A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res. 1988;175(1):184–91. PubMed.CrossRef
16.
Zurück zum Zitat Piao LS, Hur W, Kim TK, Hong SW, Kim SW, Choi JE, et al. CD133+ liver cancer stem cells modulate radioresistance in human hepatocellular carcinoma. Cancer Lett. 2012;315(2):129–37. PubMed.CrossRef Piao LS, Hur W, Kim TK, Hong SW, Kim SW, Choi JE, et al. CD133+ liver cancer stem cells modulate radioresistance in human hepatocellular carcinoma. Cancer Lett. 2012;315(2):129–37. PubMed.CrossRef
17.
Zurück zum Zitat Kitagawa R, Kastan MB. The ATM-dependent DNA damage signaling pathway. Cold Spring Harb Symp Quant Biol. 2005;70:99–109. PubMed.CrossRef Kitagawa R, Kastan MB. The ATM-dependent DNA damage signaling pathway. Cold Spring Harb Symp Quant Biol. 2005;70:99–109. PubMed.CrossRef
18.
Zurück zum Zitat Desai A, Webb B, Gerson SL. CD133+ cells contribute to radioresistance via altered regulation of DNA repair genes in human lung cancer cells. Radiother Oncol. 2014;110(3):538–45. PubMed Pubmed Central PMCID: 4004669.CrossRef Desai A, Webb B, Gerson SL. CD133+ cells contribute to radioresistance via altered regulation of DNA repair genes in human lung cancer cells. Radiother Oncol. 2014;110(3):538–45. PubMed Pubmed Central PMCID: 4004669.CrossRef
19.
Zurück zum Zitat Pajonk F, Vlashi E, McBride WH. Radiation resistance of cancer stem cells: the 4 R's of radiobiology revisited. Stem Cells. 2010;28(4):639–48. PubMed Pubmed Central PMCID: 2940232.CrossRef Pajonk F, Vlashi E, McBride WH. Radiation resistance of cancer stem cells: the 4 R's of radiobiology revisited. Stem Cells. 2010;28(4):639–48. PubMed Pubmed Central PMCID: 2940232.CrossRef
20.
Zurück zum Zitat Mathews LA, Cabarcas SM, Farrar WL. DNA repair: the culprit for tumor-initiating cell survival? Cancer Metastasis Rev. 2011;30(2):185–97. PubMed Pubmed Central PMCID: 3078299.CrossRef Mathews LA, Cabarcas SM, Farrar WL. DNA repair: the culprit for tumor-initiating cell survival? Cancer Metastasis Rev. 2011;30(2):185–97. PubMed Pubmed Central PMCID: 3078299.CrossRef
21.
Zurück zum Zitat Rycaj K, Tang DG. Cancer stem cells and radioresistance. Int J Radiat Biol. 2014;90(8):615–21. PubMed Pubmed Central PMCID: 4341971.CrossRef Rycaj K, Tang DG. Cancer stem cells and radioresistance. Int J Radiat Biol. 2014;90(8):615–21. PubMed Pubmed Central PMCID: 4341971.CrossRef
22.
Zurück zum Zitat Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60. PubMed.CrossRef Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60. PubMed.CrossRef
23.
Zurück zum Zitat Ettl T, Viale-Bouroncle S, Hautmann MG, Gosau M, Kolbl O, Reichert TE, et al. AKT and MET signalling mediates antiapoptotic radioresistance in head neck cancer cell lines. Oral Oncol. 2015;51(2):158–63. PubMed.CrossRef Ettl T, Viale-Bouroncle S, Hautmann MG, Gosau M, Kolbl O, Reichert TE, et al. AKT and MET signalling mediates antiapoptotic radioresistance in head neck cancer cell lines. Oral Oncol. 2015;51(2):158–63. PubMed.CrossRef
24.
Zurück zum Zitat Kim SH, Joshi K, Ezhilarasan R, Myers TR, Siu J, Gu C, et al. EZH2 protects glioma stem cells from radiation-induced cell death in a MELK/FOXM1-dependent manner. Stem cell reports. 2015;4(2):226–38. PubMed Pubmed Central PMCID: 4325196.CrossRef Kim SH, Joshi K, Ezhilarasan R, Myers TR, Siu J, Gu C, et al. EZH2 protects glioma stem cells from radiation-induced cell death in a MELK/FOXM1-dependent manner. Stem cell reports. 2015;4(2):226–38. PubMed Pubmed Central PMCID: 4325196.CrossRef
25.
Zurück zum Zitat Neta R, Perlstein R, Vogel SN, Ledney GD, Abrams J. Role of interleukin 6 (IL-6) in protection from lethal irradiation and in endocrine responses to IL-1 and tumor necrosis factor. J Exp Med. 1992;175(3):689–94. PubMed Pubmed Central PMCID: 2119144.CrossRef Neta R, Perlstein R, Vogel SN, Ledney GD, Abrams J. Role of interleukin 6 (IL-6) in protection from lethal irradiation and in endocrine responses to IL-1 and tumor necrosis factor. J Exp Med. 1992;175(3):689–94. PubMed Pubmed Central PMCID: 2119144.CrossRef
26.
Zurück zum Zitat Wu CT, Chen MF, Chen WC, Hsieh CC. The role of IL-6 in the radiation response of prostate cancer. Radiat Oncol. 2013;8:159. PubMed Pubmed Central PMCID: 3717100.CrossRef Wu CT, Chen MF, Chen WC, Hsieh CC. The role of IL-6 in the radiation response of prostate cancer. Radiat Oncol. 2013;8:159. PubMed Pubmed Central PMCID: 3717100.CrossRef
27.
Zurück zum Zitat Yan HQ, Huang XB, Ke SZ, Jiang YN, Zhang YH, Wang YN, et al. Interleukin 6 augments lung cancer chemotherapeutic resistance via ataxia-telangiectasia mutated/NF-kappaB pathway activation. Cancer Sci. 2014;105(9):1220–7. PubMed.CrossRef Yan HQ, Huang XB, Ke SZ, Jiang YN, Zhang YH, Wang YN, et al. Interleukin 6 augments lung cancer chemotherapeutic resistance via ataxia-telangiectasia mutated/NF-kappaB pathway activation. Cancer Sci. 2014;105(9):1220–7. PubMed.CrossRef
28.
Zurück zum Zitat Hein AL, Ouellette MM, Yan Y. Radiation-induced signaling pathways that promote cancer cell survival (review). Int J Oncol. 2014;45(5):1813–9. PubMed Pubmed Central PMCID: 4203326.CrossRef Hein AL, Ouellette MM, Yan Y. Radiation-induced signaling pathways that promote cancer cell survival (review). Int J Oncol. 2014;45(5):1813–9. PubMed Pubmed Central PMCID: 4203326.CrossRef
Metadaten
Titel
RETRACTED ARTICLE: IL-6 signaling promotes DNA repair and prevents apoptosis in CD133+ stem-like cells of lung cancer after radiation
verfasst von
Yuhchyau Chen
Fuquan Zhang
Ying Tsai
Xiadong Yang
Li Yang
Shanzhou Duan
Xin Wang
Peter Keng
Soo Ok Lee
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Radiation Oncology / Ausgabe 1/2015
Elektronische ISSN: 1748-717X
DOI
https://doi.org/10.1186/s13014-015-0534-1

Weitere Artikel der Ausgabe 1/2015

Radiation Oncology 1/2015 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.