Skip to main content
Top
Published in: Cancer Cell International 1/2020

01-12-2020 | Glioblastoma | Primary research

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

Authors: Xuejiao Liu, Yiming Tu, Yifeng Wang, Di Zhou, Yulong Chong, Lin Shi, Guanzheng Liu, Xu Zhang, Sijin Wu, Huan Li, Shangfeng Gao, Mingshan Niu, Rutong Yu

Published in: Cancer Cell International | Issue 1/2020

Login to get access

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
2.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
4.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
5.
go back to reference 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 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
6.
7.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
8.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
9.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
10.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
11.
go back to reference Wang AY, Liu H. The past, present, and future of CRM1/XPO1 inhibitors. Stem cell Invest. 2019;6:6.CrossRef Wang AY, Liu H. The past, present, and future of CRM1/XPO1 inhibitors. Stem cell Invest. 2019;6:6.CrossRef
12.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
13.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
14.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
15.
16.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
17.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
18.
go back to reference 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.CrossRef 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.CrossRef
19.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
20.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
21.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
22.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
23.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
24.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
25.
go back to reference 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.PubMed 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.PubMed
26.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
27.
go back to reference 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.CrossRef 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.CrossRef
28.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
29.
go back to reference 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.CrossRef 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.CrossRef
30.
go back to reference 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.CrossRef 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.CrossRef
31.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
32.
go back to reference 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.PubMed 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.PubMed
33.
go back to reference 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. 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.
34.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
35.
go back to reference 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.PubMedCrossRefPubMedCentral 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.PubMedCrossRefPubMedCentral
36.
go back to reference 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.CrossRef 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.CrossRef
37.
go back to reference Squatrito M, Holland EC. DNA damage response and growth factor signaling pathways in gliomagenesis and therapeutic resistance. Cancer Res. 2011;71(18):5945–9.PubMedCrossRef Squatrito M, Holland EC. DNA damage response and growth factor signaling pathways in gliomagenesis and therapeutic resistance. Cancer Res. 2011;71(18):5945–9.PubMedCrossRef
38.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
40.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
41.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
42.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
43.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
44.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
45.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
Metadata
Title
Reversible inhibitor of CRM1 sensitizes glioblastoma cells to radiation by blocking the NF-κB signaling pathway
Authors
Xuejiao Liu
Yiming Tu
Yifeng Wang
Di Zhou
Yulong Chong
Lin Shi
Guanzheng Liu
Xu Zhang
Sijin Wu
Huan Li
Shangfeng Gao
Mingshan Niu
Rutong Yu
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2020
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-020-01186-y

Other articles of this Issue 1/2020

Cancer Cell International 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine