Skip to main content
  • Primary research
  • Open access
  • Published:

Reversible inhibitor of CRM1 sensitizes glioblastoma cells to radiation by blocking the NF-κB signaling pathway

Abstract

Background

Activation of nuclear factor-kappa B (NF-κΒ) through DNA damage is one of the causes of tumor cell resistance to radiotherapy. Chromosome region 1 (CRM1) regulates tumor cell proliferation, drug resistance, and radiation resistance by regulating the nuclear-cytoplasmic translocation of important tumor suppressor proteins or proto-oncoproteins. A large number of studies have reported that inhibition of CRM1 suppresses the activation of NF-κΒ. Thus, we hypothesize that the reversible CRM1 inhibitor S109 may induce radiosensitivity in glioblastoma (GBM) by regulating the NF-κΒ signaling pathway.

Methods

This study utilized the cell counting kit-8 (CCK-8), 5-ethynyl-2′-deoxyuridine (EdU), and colony formation assay to evaluate the effect of S109 combined with radiotherapy on the proliferation and survival of GBM cells. The therapeutic efficacy of S109 combined with radiotherapy was evaluated in vivo to explore the therapeutic mechanism of S109-induced GBM radiosensitization.

Results

We found that S109 combined with radiotherapy significantly inhibited GBM cell proliferation and colony formation. By regulating the levels of multiple cell cycle- and apoptosis-related proteins, the combination therapy induced G1 cell cycle arrest in GBM cells. In vivo studies showed that S109 combined with radiotherapy significantly inhibited the growth of intracranial GBM and prolonged survival. Importantly, we found that S109 combined with radiotherapy promoted the nuclear accumulation of IκΒα, and inhibited phosphorylation of p65 and the transcriptional activation of NF-κΒ.

Conclusion

Our findings provide a new therapeutic regimen for improving GBM radiosensitivity as well as a scientific basis for further clinical trials to evaluate this combination therapy.

Background

Glioblastoma (GBM) is the most common malignant brain tumor in adults and has the poorest prognosis [1, 2]. Maximum surgical resection is the most common treatment method for GBM, followed by postoperative adjuvant chemotherapy and other comprehensive treatments [3]. The prognosis of patients with GBM is very unsatisfactory, with an average survival time of only 15–23 months [4, 5]. Because most GBMs are resistant to radiotherapy, screening for appropriate targeted drugs and increasing GBM radiosensitivity are the most important issues regarding GBM treatment. These strategies may also be the keys to improving overall therapeutic outcomes of GBM.

The transport of proteins between the nucleus and cytoplasm is critical for maintaining the homeostasis of normal cells. Exportin 1 (XPO1), also known as chromosome region 1 (CRM1), is a regulatory factor necessary for exporting cargo proteins that have the nuclear export signal (NES) from the nucleus to the cytoplasm [6, 7]. CRM1 regulates the nuclear export of more than 200 tumor suppressor proteins and proto-oncoproteins, including forkhead box protein O1 (Foxo1), Rb1, p53, p21, p27, survivin, and nuclear factor-kappa B (NF-κΒ) [8]. In general, CRM1-mediated substrate protein transportation is strictly controlled, but in cancer cells, abnormally high expression of CRM1 often causes abnormal regulation of nuclear-cytoplasmic translocation of proteins, promoting tumorigenesis and tumor progression. CRM1 is widely expressed in tissues and cells, and its high expression is closely related to tumorigenesis and drug resistance [9, 10]. In many malignant tumors, such as ovarian cancer, osteosarcoma, and pancreatic cancer, high CRM1 expression is associated with poor prognosis in patients [11]. Patients with high CRM1 expression have a relatively short median survival time. Our previous study showed that CRM1 was highly expressed in GBM, which was closely related to the poor prognosis of GBM patients. Inhibition of CRM1 simultaneously blocks three core signaling pathways (RTK/AKT/Foxo1, p53, and pRb) closely related to GBM tumorigenesis and progression, thereby suppressing GBM growth [12]. In view of the potential of CRM1 as an anticancer therapeutic target, a number of small-molecule inhibitors against CRM1 have recently been developed [11]. In recent years, our research team has developed a novel reversible CRM1 inhibitor (S109) that induces the degradation of CRM1 protein. S109 has shown significant antitumor effects in a preclinical mouse orthotopic GBM model and significantly prolongs the survival of tumor-bearing mice. S109 can effectively pass through the blood–brain barrier [12]. However, long-term S109 chemotherapy alone may induce drug resistance. The combination of radiotherapy and chemotherapy for GBM is a classic and more effective method in clinical practice.

In recent years, clinical evidence has shown that the low sensitivity of postoperative tumor cells to radiotherapy and chemotherapy is associated with increased NF-κΒ activity [13]. NF-κΒ is a class of nuclear transcriptional regulators with specific DNA-binding sequences that is widely distributed in many cells and participates in cell responses to various stimuli, such as growth factors and ionizing radiation [14, 15]. NF-κΒ is also known to prevent apoptosis through its involvement in cell cycle arrest and DNA repair, and plays an important role in tumor proliferation and apoptosis [16, 17]. In many types of tumors, structural activation of NF-κΒ is involved in tumor resistance to chemotherapy and radiotherapy [18, 19]. NF-κΒ and NF-κΒ-regulated gene products, including Bcl-xl, cyclin D1, matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF), and cyclooxygenase-2 (COX-2), are involved in tumor cell resistance to radiotherapy [20, 21]. IκΒα is an inhibitor of NF-κΒ that binds to NF-κΒ in the resting state, inducing NF-κΒ to remain in the cytoplasm [22]. When cells are exposed to external stimuli or stress, IκΒ kinase (IKK) is activated, phosphorylating and degrading IκΒ to release NF-κΒ to the nucleus where it can drive activation of target transcription factors [23, 24]. Nuclear activation of NF-κΒ rapidly induces transcription of the gene encoding IκΒα. The newly synthesized IκΒα enters the nucleus and forms a complex with NF-κB, dissociating NF-κB from DNA and rapidly transporting NF-κB out of the nucleus [25]. The IκΒα protein contains a NES sequence, and NF-κB nuclear export by IκΒα is regulated by CRM1. Studies have shown that the inhibition of IκΒα nuclear export suppresses NF-κΒ activation when cells are stimulated [26]. Based on the above studies, we hypothesized that the application of a CRM1 inhibitor would block IκΒα nuclear export and prevent GBM resistance to radiation.

It is unclear if S109 enhances the radiosensitivity of GBM in vivo. In this study, we validated this hypothesis by evaluating the effect of S109 on the growth of GBM xenografts in nude mice exposed to radiation. Our results showed that S109 promoted the nuclear accumulation of IκΒα and inhibited transcriptional activation of NF-κΒ by downregulating NF-κΒ activity and mediating GBM radiosensitization, thereby suppressing GBM growth.

Materials and methods

Cell culture and reagents

Human U87 GBM cells and rat C6 glioma cells were obtained from the Shanghai Cell Bank, Chinese Academy of Sciences for this study. Cells were maintained in Dulbecco’s modified Eagle’s medium supplemented with 10% FBS at 37 °C in a humidified incubator with 5% CO2. Antibodies against Cyclin D1 (cat. no.2922S), p27 (cat. no.3686S), Bcl-2 (cat. no.2872S), Bax (cat. no.2772S), γ-H2AX (cat. no.9718S), β-actin (cat. no.8457S), NF-κB p65 (cat. no.8242S), p- NF-κB p65 (Ser536) (cat. no.3033S), Histone H3 (cat.no.4499 s) and IκΒα (cat. no.4814S) were purchased from Cell Signaling Technology (CST, Beverly, MA, USA). Anti-CRM1 (sc-74454) and anti-H2AX (sc-54606) primary antibodies were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The S109 compound was synthesized by the Suzhou Komanda Drug Development Company. S109 was dissolved in DMSO to create a 10 mmol/L solution, which was diluted to different concentrations of medium before use.

CCK-8 assay

Cell viability was examined using a Cell Counting Kit-8 (Dojindo, Kumamoto, Japan) as previously described [27]. U87 or C6 cells were seeded in triplicate on 96-well plates (3000 cells per well). After overnight culture, different concentrations of S109 were added. After 72 h culture, 10 μL of CCK-8 solution was added to each well. The absorbance at the OD450 nm was detected with a microplate reader after incubation for 2 h. The experiment was repeated three times.

EdU incorporation assay

The Cell-Light EdU Cell Proliferation Detection Kit (Ruibo Biotech, Guangzhou, China) was used to measure cell proliferation. C6 and U87 cells were seeded into 96-well plates. When the cells attached, they were treated with 0, 0.125, 0.25 and 0.5 μΜ S109, followed by treatment with radiotherapy at a dose of 0 Gy and 2 Gy. After 24 h, 50 μΜ EdU was added and incubated with the cells for 4 h. The cells were then fixed with 4% paraformaldehyde for 15 min, followed by treatment with 0.5% Triton X-100 for 20 min. Subsequently, the cells were incubated with 1 × Apollo® reaction cocktail for 30 min and stained with 4′,6-diamidino-2-phenylindole (DAPI, Sigma) for 15 min. After washing three times with PBS, the cells were subjected to imaging under a fluorescent inverted microscope. This experiment was performed three times.

Clonogenic survival

The sensitivity of GBM cells to S109 or radiotherapy was evaluated using clonogenic survival assays. Control groups were treated with DMSO or mock irradiated. For combination studies, cells were plated at clonal density in six-well plates (cell numbers ranging from 600 to 2000 cells per well depending on radiation dose) and irradiated the next day. The different concentrations of S109 were added before irradiation, and the cells were irradiated with 0–6 Gy. After treatment with S109 for 24 h, the media were changed, and the cells were incubated for another 10–14 day with fresh drug-free medium. The cells were washed with PBS and fixed them with methanol, then stained with 0.1% crystal violet solution. All colonies with over 50 cells were counted. The surviving fraction (SF) was calculated as mean colonies/(cells inoculated plating efficiency). Cell survival curves were obtained by GraphPad Prism 6.0 software. Compusyn software was used to determine the synergistic effect of S109 combined with IR [28].

Cell cycle assay

We examined the effect of S109 and/or irradiation treatment on cell cycle distribution by flow cytometry as previously described [29]. Briefly, C6 and U87 cells were seeded into 6-cm culture dishes. Once the cells were adhered, 0.5 μΜ or 1 μΜ S109 was added, and then the cells were exposed to a single dose of radiation (0 and 2 Gy) or both for 24 h. The cells were collected and fixed with 70% ice-cold ethanol and subsequently washed twice with PBS. Cells were stained with 50 μg/mL propidium iodide (PI) solution containing 25 μg/mL ribonuclease (RNase) for 30 min. Finally, the cycle distribution was assessed by flow cytometry and analyzed using CellQuest Pro software (Becton–Dickinson).

Luciferase reporter assay

To assess the promoter activity of NF-κB, luciferase assay was performed. U87 cells were co-transfected with NF-κB-luc and renilla luciferase reporter vector using Lipofectamine 2000 (Invitrogen) in serum free DMEM media for 6 h. Then, the serum free media was changed with DMEM media containing 10% FBS, and the cells were incubated for another 24 h. The transfected cells were treated with 0.1% DMSO or S109 and/or IR. Luciferase reporter assay was performed using the Dual Luciferase Assay kit (Promega). Relative activity of NF-κB promoter was calculated relative to Renilla activity using luminometer.

Western blot assay

U87 and C6 cells were treated with S109 at different concentrations. Radiotherapy was performed after dosing, and the radiotherapy dose was 0 Gy or 2 Gy. Control groups were treated with DMSO or mock irradiated. After 24 h, the total proteins, nuclear protein or cytoplasmic protein were collected for western blotting as previously described [30, 31]. The expression levels of Cyclin D1 (dilution 1:1000), p27 (dilution 1:750), Bcl-2 (dilution 1:500), Bax (dilution 1:1000), γ-H2AX (dilution 1:500), H2AX (dilution 1:500), CRM1 (dilution 1:1000), NF-κB p65 (dilution 1:500), p-NF-κB p65 (dilution 1:500) and IκΒα (dilution 1:1000) were examined, and β-actin (dilution 1:2000) and Histone H3 (dilution 1:2000) were used as loading control.

In vivo studies

All animal protocols were approved by the Ethics Committee of the Xuzhou Medical University (Jiangsu Province, China). Seventy-two male athymic BALB/c nude mice aged 5–6 weeks were purchased from Beijing Vital River Experimental Animal Technology Co. Ltd., China. Luciferase-labeled U87 cells were injected orthotopically into the right striatum of nude mice using a small animal stereotaxic instrument (5 × 105 cells per mouse). Five days after tumor cell inoculation, the nude mice were randomly divided into six groups (n = 12 per group). The grouping was as follows: (i) control group (vehicle alone), (ii) S109 treatment alone group (50 mg/kg), (iii) irradiation (IR) alone group (dose of 6 Gy), (iv) combination of S109 with 6 Gy radiation, (v) dose of 10 Gy radiation group, (vi) combination of S109 with 10 Gy radiation. The S109 and vehicle were injected intraperitoneally once a day. The radiotherapy groups were given radiation every other day with 2 Gy fractions three or five times. Tumor growth was monitored at regular intervals (day 10, day 18, and day 26) by injecting 75 mg/kg D-luciferin (Beetle Luciferin Potassium Salt, Promega) in 100 μL of PBS 10 min prior to imaging using a NightOWL LB 983 small-animal in vivo imaging system (Berthold Technologies, Germany). After 28 days of treatment, five randomly selected mice were euthanized and whole brains were collected after perfusion to observe tumor size. The other seven mice in each group were used for survival analysis.

Histopathology and immunofluorescence staining

The whole mouse brains of the control and treatment groups were post-fixed in 4% paraformaldehyde for 24 h, followed by continuous dehydration in 20% and 30% sucrose solutions (24 h each) until the whole brains sank to the bottom of the solution. The frozen GBM tissues were serially sectioned into brain slices with a 12 µm thickness using a cryostat. The brain slices were photographed under a microscope. The brain tumor slices were then incubated with PBS containing 0.3% Triton X-100 at room temperature for 30 min, followed by direct blocking in 10% goat serum for 1 h. The brain tumor slices were subsequently incubated with anti-Ki67 (RM-9106, Thermo Scientific, 1:400) overnight and with secondary antibodies for an hour on the next day. DAPI dye was used for nuclear staining. The immunofluorescent signals were observed and photographed under an inverted fluorescence microscope.

Statistical analysis

Each experiment was independently performed three or more times. GraphPad Prism 6.0 software was used for statistical analysis of the experimental results. Data are presented as mean ± standard deviation of the mean. Independent sample t test was used to compare the difference between two samples. The Kaplan–Meier method was used for survival analysis. Log-rank test was used to compare the difference in survival time between two groups. α = 0.05 was chosen as the test level, and a *P < 0.05 was considered statistically significant.

Results

S109 combined with radiotherapy significantly inhibited GBM cell proliferation

To evaluate the therapeutic efficacy of the CRM1 inhibitor S109 as a radiosensitizer in GBM, the CCK-8 was used to assess the effect of S109 on cell viability in U87 and C6 cells. The results showed that S109 significantly inhibited the growth of U87 and C6 cells in a dose-dependent manner (Fig. 1a, b).

Fig. 1
figure 1

S109 combined with radiotherapy significantly inhibited GBM cell proliferation. U87 a and C6 b cell lines were treated with varying doses of S109, and cell viability was assessed by CCK-8 assay at 72 h after treatment. c, e Measurement of anti-proliferation effects of S109 and/or IR by EdU incorporation assay. Represent images were showed. The EdU incorporation rate was presented as the ratio of EdU positive cells to total DAPI positive cells, scale bar: 100 μm. d, f Quantification of the percentage of EdU-positive cells. All the results were presented as the mean ± SD from 3 independent experiments, *P < 0.05, **P < 0.01, ***P < 0.001

To further evaluate the effect of S109 combined with radiotherapy on GBM cell proliferation, U87 and C6 cells were treated with fixed doses of S109 (0.125–0.5 μΜ) and/or 2 Gy radiation, followed by use of the EdU incorporation assay to determine cell proliferation. As shown in Fig. 1c–f, in U87 cells, compared with the control group, the cell proliferation rates were reduced to 80.7%, 63.7%, and 28.7% in the cells with 0.125, 0.25, and 0.5 μM S109 treatments, respectively. Under the 2 Gy radiotherapy, the cell proliferation rates were reduced to 79.7%, 42.7%, 27.3%, and 4.0% in the cells with 0, 0.125, 0.25, and 0.5 μM S109 treatments, respectively. Similar results were observed in C6 cells; when compared with the control group, the cell proliferation rates were reduced to 87.7%, 78.0%, and 73.0% in the cells with 0.125, 0.25, and 0.5 μM S109 treatments, respectively. Under the 2 Gy radiotherapy, the cell proliferation rates were reduced to 68.3%, 47.0%, 37.7%, and 28.7% in the C6 cells with 0, 0.125, 0.25, and 0.5 μM S109 treatments, respectively. These findings suggest that S109 combined with radiotherapy significantly inhibits the proliferation of GBM cells.

S109 combined with radiotherapy inhibited colony formation of GBM cells

The effect of S109 on radiosensitization in U87 and C6 cells was detected by colony formation assay. The results showed that colony formation rates in the U87 and C6 cell lines decreased gradually as the irradiation dose increased. At the same irradiation dose, the colony formation rates of the cells treated with S109 combined with radiotherapy were significantly lower compared with the cells treated with radiotherapy alone (Fig. 2a, c). The survival curves of U87 and C6 cell lines were fitted by a single-hit, multi-target model and a linear-quadratic model. The results showed that compared with the cells treated with radiotherapy alone, the survival curves of the U87 and C6 cells down-shifted after 0.25 μM S109 plus radiotherapy and 0.5 μM S109 plus radiotherapy. The S109-induced radiosensitization ratios of U87 and C6 cells were calculated from the fitted curves, indicating as 1.29 and 1.88 in U87 cells and as 1.16 and 1.94 in C6 cells after 0.25 μM and 0.5 μM S109 treatments, respectively (Fig. 2b, d). Then, we further analyzed the synergistic effect of S109 and IR. The results showed that the combination indexes (CI) at different concentrations were less than 1, indicating a synergistic effect between S109 and IR (Additional file 1: Tables S1 and S2). These findings indicated that CRM1 inhibition combined with radiotherapy synergistically reduced the proliferation of U87 and C6 cells in a dose-dependent manner.

Fig. 2
figure 2

S109 enhances radiation sensitivity of GBM cells. a U87 cells and (C) C6 cells were cultured in 6-well plates. S109 (0.25 μM or 0.5 μM) or vehicle (DMSO) were added to culture media and the cells were irradiated the next day. Colony forming efficiency was determined 10–14 days later. b, d Killing curves were generated after normalizing for cell killing by S109. Values represent the mean ± SD of 3 independent experiments

S109 combined with radiotherapy induced cell cycle arrest in the G1 phase and enhanced radiation-induced DNA damage

To further investigate the mechanism by which S109 combined with radiotherapy inhibited GBM cell proliferation, flow cytometry was used to analyze the distribution of cells in the cell cycle. As shown in Fig. 3a, b, compared to the cells with no treatment, U87 and C6 cells were arrested in the G1 phase after S109 treatment. Importantly, the number of cells in the G1 phase was not significantly increased after 2 Gy radiotherapy alone. However, the number of cells in the G1 phase was significantly increased after S109 combined with radiotherapy, with a corresponding reduction in the number of cells in the S and G2 phases. These results indicated that the decrease in cell proliferation induced by S109 combined with radiotherapy was due to cell cycle arrest in the G1 phase.

Fig. 3
figure 3

Combination of S109 and radiation results in cell cycle arrest and enhances radiation-induced DNA damage. The distribution of cell cycle after S109 and/or IR treatment and quantitative analyses were examined by flow cytometry in U87 cells a and C6 cells b. The data represent the mean ± SD from 3 independent experiments, *P < 0.05, **P < 0.01. c–f The expression levels of multiple cell-cycle- and apoptosis-associated proteins were assessed by western blot assay in time- and does-manner. g Lysates of S109-treated U87 cells were collected at 24 h after radiation (2 Gy) for Western blot analysis. The levels of γ-H2AX and H2AX were examined

To further explore the mechanism by which S109 combined with radiotherapy regulated the cell cycle, we measured the level of several regulators that affect cell cycle progression by western blot analysis. As shown in Fig. 3c–f, S109 combined with radiotherapy increased the level of the cell cycle inhibitor p27 and reduced the level of cyclin D1 in a time- and dose-dependent manner compared to the radiotherapy alone treatment groups. Our data also showed that S109 combined with radiotherapy reduced the level of the anti-apoptotic protein Bcl-2 and steadily increased the level of the pro-apoptotic protein Bax. Importantly, compared with the S109 alone group, the changes in the level of the above cell cycle- and apoptosis-associated proteins were more pronounced after treatment with S109 combined with radiotherapy. These data demonstrated that S109 combined with radiotherapy induced cell cycle arrest in the G1 phase by modulating multiple cell-cycle- and apoptosis-associated proteins.

To determine the effect of S109 on radiation-induced DNA damage, the expression level of phosphorylated γH2AX was evaluated. As shown in Fig. 3g, the level of phosphorylated γH2AX was not changed in the cells following S109 treatment alone but was increased in the cells following 2 Gy radiotherapy alone. The level of phosphorylated γH2AX expression was even higher in the cells treated with S109 combined with radiotherapy than in the radiotherapy alone groups. Therefore, S109 treatment enhanced radiation-induced DNA damage in the cells.

CRM1 inhibition combined with radiotherapy slows down the growth of GBM in vivo

To validate the therapeutic efficacy of S109 combined with radiotherapy in GBM, we established a mouse orthotopic GBM model by inoculating luciferase-labeled U87 cells into nude mouse brains and periodically observed tumor growth using a small animal imaging system. As shown in Fig. 4a, the animals were intraperitoneally injected daily with 50 mg/kg S109 5 days after tumor cell inoculation. The animals were then divided into six groups and received radiotherapy once every other day starting at day 10, with 2 Gy fractions three or five times. The small animal imaging results showed that compared with the control group, the growth of U87 cells in the mice with S109 treatment alone or radiotherapy alone was significantly inhibited. However, the therapeutic effect was greater in the S109 combined with radiotherapy group, which significantly slowed the growth of U87 orthotopic xenografts in the mice (Fig. 4b, c). Compared with the control group, the median survival was extended 13 days in the S109 treatment group, 6 days in the 6-Gy radiotherapy group, 11 days in the 10-Gy radiotherapy group, 23 days in the 50 mg/kg S109 plus 6-Gy radiotherapy group, and 34 days in the 50 mg/kg S109 plus 10-Gy radiotherapy group (Fig. 4d). Compared with control or radiotherapy alone, S109 combined with radiotherapy significantly prolonged the median survival time of the tumor-bearing mice (Fig. 4d), suggesting that S109 increased the sensitivity of glioma cells to radiotherapy.

Fig. 4
figure 4

CRM1 inhibition synergizes with radiation in in vivo models of GBM. a Schematic diagram of mice treated with S109 combined with radiotherapy. bIn vivo efficacy in U87-Luci GBM model in mice. U87-Luci-bearing mice received daily injection of S109 at a dose of 50 mg/kg, and these mice were irradiated on days 10, 12, 14, 16 and 18 with 2 Gy. Bioluminescent signal changes correlating to tumor growth were showed. c Quantification of the tumor bioluminescence signal (n = 3 mice per group). (D) Kaplan–Meier survival curve for the mice (n = 7, **P < 0.01, ***P < 0.001). e Representative images of Ki-67 immunostaining of tumors dissected from control, S109-treated mice, IR-treated mice and S109+IR groups. Scale bar: 250 μm. f Quantitative analyses of the percentages of Ki-67 positive cells. Data are presented as mean ± SD from 3 independent experiments, *P < 0.05, **P < 0.01, ***P < 0.001

We further analyzed Ki67 expression in the brain tissue sections in the different groups and results showed that compared with the control group, the percentage of Ki67-positive cells was 62.7% and 35.7% in the 6-Gy and 10-Gy radiotherapy groups, respectively. The percentage of Ki67-positive cells in cells treated with 50 mg/kg S109 combined with 0 Gy, 6 Gy, and 10 Gy radiotherapy was 44.3%, 17.0%, and 7.0%, respectively, compared with the control group (Fig. 4e, f). The above results showed that S109 combined with radiotherapy significantly inhibited the growth of tumor cells in tumor-bearing mice and prolonged the survival of these mice.

Combination of radiation and CRM1 inhibitor treatment results in nuclear retention of IκBα and reduces the level of p-p65 and NF-κΒ transcriptional activity

Numerous studies have shown that radiotherapy activates NF-κΒ signaling, which is one of the causes of tumor cell resistance to radiotherapy [32, 33]. IκBα, the inhibitory protein of NF-κΒ, is a well-known CRM1 target protein. CRM1 regulates the nuclear export of IκBα, thereby affecting the activation of NF-κΒ signaling [34, 35]. To investigate the therapeutic mechanism of S109 in enhancing sensitivity to radiotherapy, this study investigated if S109 exerted a radiosensitizing effect through regulation of IκBα/NF-κΒ signaling. As shown in Fig. 5a, b, the level of CRM1 expression was significantly reduced in U87 and C6 cells after S109 and/or 2-Gy radiotherapy. However, the CRM1 level was not affected in the cells treated with radiotherapy alone. Following S109 treatment alone, radiotherapy alone, or the combination therapy, total levels of the NF-κΒ p65 subunit did not change. Although S109 treatment alone or radiotherapy alone reduced the phosphorylation of p65, this reduction was not significant. S109 combined with radiotherapy significantly reduced the phosphorylation of p65 in a dose-dependent manner. We also evaluated the effect of radiotherapy on the level and activity of p65 at different time points after the treatment. We found that the p65 phosphorylation level was gradually reduced in cells treated with S109 alone. The reduction in p65 phosphorylation level was more significant after treatment with S109 combined with radiotherapy, with no effect on total p65 expression level (Fig. 5c, d).

Fig. 5
figure 5

Combination of S109 and radiation reduces NF-κB transcriptional activation and promotes the nuclear accumulation of IκΒα. U87 and C6 cells were treated with S109 and/or IR. Lysates of cells were collected at 24 h after radiation for Western blot. The expression levels of CRM1, p-p65 and p-65 were assessed in a dose- and time-manner ad. e The effects of S109 and/or IR treatment on the subcellular location of IκΒα. The cytoplasmic and nuclear protein extracts were used for immunoblotting with the indicated antibodies. f The transactivation ability of NF-κB were inhibited when combination of S109 and radiation. Relative NF-κB luciferase activity normalized with respect to corresponding renilla luciferase activity is shown. All the Data are presented as mean ± SD, *P < 0.05, **P < 0.01

We further isolated the nucleoplasmic proteins to examine the localization of the target protein IκBα, which is regulated by CRM1. As shown in Fig. 5e, cells treated with S109 alone had a gradual reduction of IκBα in the cytoplasm and a gradual increase of IκBα in the nuclei. The IκBα cytoplasmic reduction and nuclear aggregation in the cells was more significant after treatment with S109 combined with radiotherapy. The results of NF-κΒ-Luciferase reporter gene assays also showed that the transcriptional activation of NF-κΒ in the cells was significantly reduced after S109 treatment combined with radiotherapy (Fig. 5f). The above results indicated that S109 combined with radiotherapy significantly increased the nuclear accumulation of IκBα, which inhibited p-p65 expression and the transcriptional activation ability of NF-κΒ, thereby suppressing the proliferation of GBM cells.

Discussion

Radiotherapy is one of the main therapeutic regimens for glioma in clinical practice. However, 90% of patients with high-grade GBM relapse, mainly because high-grade gliomas develop resistance to chemotherapy and radiotherapy, leading to insensitivity of the tumor cells toward chemotherapy and radiotherapy and reducing the curative rate. Screening for effective chemotherapeutic drugs and improving sensitivity to radiotherapy are hot research topics in current cancer research. Our previous study results have indicated that S109, a novel reversible inhibitor of CRM1, effectively inhibits the growth of GBM cells and prolongs the survival of tumor-bearing mice. The aim of the present study was to determine whether S109 could sensitize GBM to radiotherapy. Results of the current study indicated that S109 combined with radiotherapy inhibited the growth of GBM cells through suppressing NF-κΒ signaling. In vivo experiments showed that S109 combined with radiotherapy significantly inhibited the growth of intracranial GBM and prolonged survival, suggesting that S109 increased the sensitivity of glioma cells to radiotherapy.

The DNA-damage response (DDR) pathway plays an important role in glioma resistance to radiotherapy and is involved in the detection of DNA damage, the initiation of DNA repair, and regulation of the cell cycle and apoptosis [36, 37]. Defects in the DDR pathway lead to GBM tumorigenesis and an increase in GBM resistance to radiotherapy [38]. The key histone that regulates DNA damage, H2AX, is a subtype of histone H2A, which is different from other H2A subtypes due to its highly protected serine-glutamine-glutamic acid motif at the C-terminus. Within the domain, the serine 139 can be phosphorylated by members of the phospholipid inositol-3 kinase family including ataxia-telangiectasia-mutated (ATM), ATM and Rad3-Related (ATR), and DNA-dependent protein kinases (DNA-PKS) [39, 40]. Both in vivo and in vitro studies have shown that H2AX phosphorylation plays a central role in regulating cell damage caused by DNA double-strand breaks, including DNA repair, cell cycle checkpoint, and inhibition of tumor growth [41, 42]. In this study, we evaluated the effect of S109 on radiation-induced DNA damage and showed that the level of phosphorylated γH2AX in cells treated with S109 combined with radiotherapy was significantly increased, indicating enhanced DNA damage. The results of cell cycle assays showed that compared with the S109 treatment alone and radiotherapy alone, S109 combined with radiotherapy significantly increased the number of cells in the G1 phase, indicating significant cell cycle arrest in the G1 phase. Our results are consistent with a recent report on the XPO1 inhibitor, selinexor, which enhanced GBM radiosensitivity [43]. Our data also suggest that S109 has radiosensitizing effects in GBM.

In recent years, clinical evidence has shown that postoperative tumors are insensitive to radiotherapies and chemotherapies, which is associated with increased NF-κΒ activity [19, 32]. Anticancer radiotherapies generally eliminate tumor cells by damaging DNA, inducing apoptosis, and blocking tumor cell proliferation. DNA damage-activated NF-κΒ signaling is one of the main mechanisms of radiotherapy-induced apoptosis [44, 45]. However, the NF-κΒ pathway plays a dual role in DNA damage-induced apoptosis, in which it promotes apoptosis while inducing the expression of anti-apoptotic genes. Once NF-κΒ activity is abnormally increased or continuously activated, NF-κΒ exhibits anti-apoptotic effects by blocking apoptosis and promoting the expression of target genes related to cell proliferation. This results in accelerated cell cycle progression, enhanced anti-apoptotic ability, tumor invasion, and metastasis, thereby causing resistance to radiotherapy [45]. Therefore, blocking NF-κΒ activation is one of the strategies for reducing tumor resistance to radiotherapy. IκBα is an inhibitory protein of NF-κΒ, and IκBα in the nuclei can form a complex with NF-κΒ, causing NF-κΒ to dissociate from DNA and rapidly export from the nuclei [25]. The IκΒα protein contains an NES sequence. IκΒα-NF-κB complexes are recognized by CRM1, which mediates nuclear export. Studies have shown that inhibition of IκΒα nuclear export suppresses NF-κΒ activation in cells that are stimulated. In our study, we observed that NF-κΒ activation required a higher radiation dose (4 to 8 Gy) than other studies (dose of 2 Gy). It is possible that different experiments may require different treatment times to produce the same effect at the same dose. To our surprise, results of our study showed that in cells treated with S109 combined with radiotherapy, the level of p-p65 was suppressed, nuclear accumulation of IκΒα was promoted, the transcriptional activation ability of NF-κB was attenuated, and the expression of the NF-κB downstream target genes cyclin D1 and Bcl-2 was reduced. The above results fully indicated that inhibition of the IκΒα-NF-κB pathway by S109 overcame GBM cell resistance to radiation and improved the radio-therapeutic effect.

Conclusion

Taken together, results of this study showed that S109 enhanced the efficacy of radiotherapy in GBM. S109 combined with radiotherapy significantly inhibited tumor cell colony formation, induced cell cycle arrest, and inhibited tumor cell proliferation in vitro. Results of in vivo experiments showed that S109 combined with radiotherapy effectively extended the survival of tumor-bearing mice and inhibited the growth of tumor cells. Importantly, S109 combined with radiotherapy effectively inhibited NF-κB activation induced by radiation, thereby blocking NF-κB signaling and enhancing GBM radiosensitivity. Therefore, this study provides a theoretical foundation and experimental basis for further clinical trials.

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article.

Abbreviations

GBM:

Glioblastoma multiforme

CRM1:

Chromosome region 1

NES:

Nuclear export signal

CCK-8:

Cell counting kit-8

EdU:

5-ethynyl-2′-deoxyuridine

DAPI:

4′,6-diamidino-2-phenylindole

References

  1. Cantrell JN, Waddle MR, Rotman M, Peterson JL, Ruiz-Garcia H, Heckman MG, Quinones-Hinojosa A, Rosenfeld SS, Brown PD, Trifiletti DM. Progress toward long-term survivors of glioblastoma. Mayo Clin Proc. 2019;94(7):1278–86.

    Article  PubMed  Google Scholar 

  2. Liu YP, Dong FX, Chai X, Zhu S, Zhang BL, Gao DS. Role of autophagy in capsaicin-induced apoptosis in U251 glioma cells. Cell Mol Neurobiol. 2016;36(5):737–43.

    Article  PubMed  CAS  Google Scholar 

  3. Weller M, Le Rhun E, Preusser M, Tonn JC, Roth P. How we treat glioblastoma. ESMO Open. 2019;4(Suppl 2):e000520.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Liu X, Chen X, Shi L, Shan Q, Cao Q, Yue C, Li H, Li S, Wang J, Gao S, et al. The third-generation EGFR inhibitor AZD9291 overcomes primary resistance by continuously blocking ERK signaling in glioblastoma. J Exp Clin Cancer Res. 2019;38(1):219.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Qu DW, Liu Y, Wang L, Xiong Y, Zhang CL, Gao DS. Glial cell line-derived neurotrophic factor promotes proliferation of neuroglioma cells by up-regulation of cyclins PCNA and Ki-67. Eur Rev Med Pharmacol Sci. 2015;19(11):2070–5.

    PubMed  Google Scholar 

  6. Liu X, Chong Y, Liu H, Han Y, Niu M. Novel reversible selective inhibitor of CRM1 for targeted therapy in ovarian cancer. J Ovarian Res. 2015;8:35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Liu X, Niu M, Xu X, Cai W, Zeng L, Zhou X, Yu R, Xu K. CRM1 is a direct cellular target of the natural anti-cancer agent plumbagin. J Pharmacol Sci. 2014;124(4):486–93.

    Article  CAS  PubMed  Google Scholar 

  8. Niu M, Chong Y, Han Y, Liu X. Novel reversible selective inhibitor of nuclear export shows that CRM1 is a target in colorectal cancer cells. Cancer Biol Ther. 2015;16(7):1110–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Delman M, Avci ST, Akcok I, Kanbur T, Erdal E, Cagir A. Antiproliferative activity of (R)-4′-methylklavuzon on hepatocellular carcinoma cells and EpCAM(+)/CD133(+) cancer stem cells via SIRT1 and Exportin-1 (CRM1) inhibition. Eur J Med Chem. 2019;180:224–37.

    Article  CAS  PubMed  Google Scholar 

  10. Abeykoon JP, Paludo J, Nowakowski KE, Stenson MJ, King RL, Wellik LE, Wu X, Witzig TE. The effect of CRM1 inhibition on human non-Hodgkin lymphoma cells. Blood Cancer J. 2019;9(3):24.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Wang AY, Liu H. The past, present, and future of CRM1/XPO1 inhibitors. Stem cell Invest. 2019;6:6.

    Article  CAS  Google Scholar 

  12. Liu X, Chong Y, Tu Y, Liu N, Yue C, Qi Z, Liu H, Yao Y, Liu H, Gao S, et al. CRM1/XPO1 is associated with clinical outcome in glioma and represents a therapeutic target by perturbing multiple core pathways. J Hematol Oncol. 2016;9(1):108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chung AS, Wu X, Zhuang G, Ngu H, Kasman I, Zhang J, Vernes JM, Jiang Z, Meng YG, Peale FV, et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med. 2013;19(9):1114–23.

    Article  CAS  PubMed  Google Scholar 

  14. Man X, Piao C, Lin X, Kong C, Cui X, Jiang Y. USP13 functions as a tumor suppressor by blocking the NF-kB-mediated PTEN downregulation in human bladder cancer. J Exp Clin Cancer Res. 2019;38(1):259.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Mao G, Smyth SS, Morris AJ. Regulation of PLPP3 gene expression by NF-kB family transcription factors. J Biol Chem. 2019;294(38):14009–19.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Wu ZH, Wong ET, Shi Y, Niu J, Chen Z, Miyamoto S, Tergaonkar V. ATM- and NEMO-dependent ELKS ubiquitination coordinates TAK1-mediated IKK activation in response to genotoxic stress. Mol Cell. 2010;40(1):75–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li L, Xu TD, Du YP, Pan DF, Wu WL, Zhu H, Zhang YB, Li DY. Salvianolic acid A attenuates cell apoptosis, oxidative stress, akt and NF-kappa B activation in angiotensin-II induced murine peritoneal macrophages. Curr Pharm Biotechnol. 2016;17(3):283–90.

    Article  CAS  PubMed  Google Scholar 

  18. Patel M, Horgan PG, McMillan DC, Edwards J. NF-kappaB pathways in the development and progression of colorectal cancer. Transl Res: J Lab Clin Med. 2018;197:43–56.

    Article  CAS  Google Scholar 

  19. Strzeszewska A, Alster O, Mosieniak G, Ciolko A, Sikora E. Insight into the role of PIKK family members and NF-small ka, CyrillicB in DNAdamage-induced senescence and senescence-associated secretory phenotype of colon cancer cells. Cell Death Dis. 2018;9(2):44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Xia F, Powell SN. The molecular basis of radiosensitivity and chemosensitivity in the treatment of breast cancer. Semin Radiat Oncol. 2002;12(4):296–304.

    Article  PubMed  Google Scholar 

  21. de Heer P, Gosens MJEM, de Bruin EC, Dekker-Ensink NG, Putter H, Marijnen CAM, van den Brule AJC, van Krieken JHJM, Rutten HJT, Kuppen PJK, et al. Cyclooxygenase 2 expression in rectal cancer is of prognostic significance in patients receiving preoperative radiotherapy. Clin Cancer Res. 2007;13(10):2955–60.

    Article  PubMed  CAS  Google Scholar 

  22. Hay RT, Vuillard L, Desterro JM, Rodriguez MS. Control of NF-kappa B transcriptional activation by signal induced proteolysis of I kappa B alpha. Philos Trans R Soc Lond B Biol Sci. 1999;354(1389):1601–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sethi G, Ahn KS, Chaturvedi MM, Aggarwal BB. Epidermal growth factor (EGF) activates nuclear factor-kappaB through IkappaBalpha kinase-independent but EGF receptor-kinase dependent tyrosine 42 phosphorylation of IkappaBalpha. Oncogene. 2015;34(42):5407.

    Article  CAS  PubMed  Google Scholar 

  24. Meng J, Zhang QY, Yang C, Xiao L, Xue ZZ, Zhu J. Duloxetine, a balanced serotonin-norepinephrine reuptake inhibitor, improves painful chemotherapy-induced peripheral neuropathy by inhibiting activation of p38 MAPK and NF-kappa B. Front Pharmacol. 2019;10:365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Arenzana-Seisdedos F, Turpin P, Rodriguez M, Thomas D, Hay RT, Virelizier J-L, Dargemont C. Nuclear localization of I kappa B alpha promotes active transport of NF-kappa B from the nucleus to the cytoplasm. J Cell Sci. 1997;110(3):369–78.

    CAS  PubMed  Google Scholar 

  26. Silk RN, Bowick GC, Abrams CC, Dixon LK. African swine fever virus A238L inhibitor of NF-kappaB and of calcineurin phosphatase is imported actively into the nucleus and exported by a CRM1-mediated pathway. J Gen Virol. 2007;88(Pt 2):411–9.

    Article  CAS  PubMed  Google Scholar 

  27. Mei PJ, Bai J, Miao FA, Li ZL, Chen C, Zheng JN, Fan YC. Relationship between expression of XRCC1 and tumor proliferation, migration, invasion, and angiogenesis in glioma. Invest New Drug. 2019;37(4):646–57.

    Article  CAS  Google Scholar 

  28. Ferreiro-Neira I, Torres NE, Liesenfeld LF, Chan CH, Penson T, Landesman Y, Senapedis W, Shacham S, Hong TS, Cusack JC. XPO1 inhibition enhances radiation response in preclinical models of rectal cancer. Clin Cancer Res. 2016;22(7):1663–73.

    Article  CAS  PubMed  Google Scholar 

  29. Miao FA, Chu K, Chen HR, Zhang M, Shi PC, Bai J, You YP. Increased DKC1 expression in glioma and its significance in tumor cell proliferation, migration and invasion. Invest New Drug. 2019;37(6):1177–86.

    Article  CAS  Google Scholar 

  30. Yue C, Niu M, Shan QQ, Zhou T, Tu Y, Xie P, Hua L, Yu R, Liu X. High expression of Bruton’s tyrosine kinase (BTK) is required for EGFR-induced NF-κB activation and predicts poor prognosis in human glioma. J Exp Clin Canc Res. 2017;36(1):132.

    Article  CAS  Google Scholar 

  31. Han ZX, Wang XX, Zhang SN, Wu JX, Qian HY, Wen YY, Tian H, Pei DS, Zheng JN. Downregulation of PAK5 inhibits glioma cell migration and invasion potentially through the PAK5-Egr1-MMP2 signaling pathway. Brain Tumor Pathol. 2014;31(4):234–41.

    Article  CAS  PubMed  Google Scholar 

  32. Voboril R, Weberova-Voborilova J. Constitutive NF-kappaB activity in colorectal cancer cells: impact on radiation-induced NF-kappaB activity, radiosensitivity, and apoptosis. Neoplasma. 2006;53(6):518–23.

    CAS  PubMed  Google Scholar 

  33. Shentu S, Aggarwal BB, Krishnan S. Curcumin sensitizes human colorectal cancer Xenografts in nude mice to;-radiation by targeting nuclear factor-KB regulated gene products. Clin Cancer Res. 2008;2008(14):7.

    Google Scholar 

  34. Tam WF, Lee LH, Davis L, Sen R. Cytoplasmic sequestration of rel proteins by IκBα requires CRM1-dependent nuclear export. Mol Cell Biol. 2000;20(6):2269–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Huang TT, Kudo N, Yoshida M, Miyamoto S. A nuclear export signal in the N-terminal regulatory domain of IκBα controls cytoplasmic localization of inactive NF-κB/IκBα complexes. Proc Natl Acad Sci. 2000;97(3):1014–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Perez RL, Nicolay NH, Wolf J-C, Frister M, Schmezer P, Weber K-J, Huber PE. DNA damage response of clinical carbon ion versus photon radiation in human glioblastoma cells. Radiother Oncol. 2019;133:77–86.

    Article  CAS  Google Scholar 

  37. Squatrito M, Holland EC. DNA damage response and growth factor signaling pathways in gliomagenesis and therapeutic resistance. Cancer Res. 2011;71(18):5945–9.

    Article  CAS  PubMed  Google Scholar 

  38. Ahmed SU, Carruthers R, Gilmour L, Yildirim S, Watts C, Chalmers AJ. Selective inhibition of parallel DNA damage response pathways optimizes radiosensitization of glioblastoma stem-like cells. Cancer Res. 2015;75(20):4416–28.

    Article  CAS  PubMed  Google Scholar 

  39. Podhorecka M, Skladanowski A, Bozko P. H2AX phosphorylation: its role in DNA damage response and cancer therapy. J Nucleic Acids. 2010. https://doi.org/10.4061/2010/920161.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Nowsheen S, Aziz K, Luo K, Deng M, Qin B, Yuan J, Jeganathan KB, Yu J, Zhang H, Ding W. ZNF506-dependent positive feedback loop regulates H2AX signaling after DNA damage. Nat Commun. 2018;9(1):2736.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Manoel-Caetano FS, Rossi AFT, de Morais GC, Severino FE, Silva AE. Upregulation of the APE1 and H2AX genes and miRNAs involved in DNA damage response and repair in gastric cancer. Genes Dis. 2019;6(2):176–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Deng Y, Li F, He P, Yang Y, Yang J, Zhang Y, Liu J, Tong Y, Li Q, Mei X. Triptolide sensitizes breast cancer cells to Doxorubicin through the DNA damage response inhibition. Mol Carcinog. 2018;57(6):807–14.

    Article  CAS  PubMed  Google Scholar 

  43. Wahba A, Rath BH, O’Neill JW, Camphausen K, Tofilon PJ. The XPO1 inhibitor selinexor inhibits translation and enhances the radiosensitivity of glioblastoma cells grown in vitro and in vivo. Mol Cancer Ther. 2018;17(8):1717–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sandur SK, Deorukhkar A, Pandey MK, Pabón AM, Shentu S, Guha S, Aggarwal BB, Krishnan S. Curcumin modulates the radiosensitivity of colorectal cancer cells by suppressing constitutive and inducible NF-κB activity. Int J Radiat Oncol Biol Phys. 2009;75(2):534–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kunnumakkara AB, Diagaradjane P, Guha S, Deorukhkar A, Shentu S, Aggarwal BB, Krishnan S. Curcumin sensitizes human colorectal cancer xenografts in nude mice to γ-radiation by targeting nuclear factor-κB–regulated gene products. Clin Cancer Res. 2008;14(7):2128–36.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Zengtian Sun for the technical assistance.

Funding

The research was supported by National Natural Science Foundation of China (No. 81772658, 81670142, 81972345); Jiangsu Provincial Key Research and Development Program (BE2017636, BE2017638); Natural Science Foundation of Jiangsu Province (BK20180104); The Foundation of Jiangsu Province Six Talents Peak (No. 2016-WSN-136, No. 2017-WSN-120); Jiangsu Qing Lan Project for Mingshan Niu; College Students’ Innovation and Entrepreneurship Training Program (No. 201610313003, 20161031Z). The 2019 Suqian industrial development guide fund project (No. K201902).

Author information

Authors and Affiliations

Authors

Contributions

RY and XL designed this study. XL, YT and YW performed the main experimental procedures. DZ, YC, LS, GL, XZ, SW and HL carried out partial experiments. MN and SG constructed the data analysis. XL wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Rutong Yu.

Ethics declarations

Ethics approval and consent to participate

The experimental protocol for animal studies was reviewed and approved by ethics committee of Xuzhou Medical University.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Additional file 1: Table S1.

Combination index (CI) for the synergistic combination of S109 and irradiation in U87 cells. Table S2. Combination index (CI) for the synergistic combination of S109 and irradiation in C6 cells.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X., Tu, Y., Wang, Y. et al. Reversible inhibitor of CRM1 sensitizes glioblastoma cells to radiation by blocking the NF-κB signaling pathway. Cancer Cell Int 20, 97 (2020). https://doi.org/10.1186/s12935-020-01186-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-020-01186-y

Keywords