Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

Tunicamycin specifically aggravates ER stress and overcomes chemoresistance in multidrug-resistant gastric cancer cells by inhibiting N-glycosylation

Authors: Jian Wu, Sheng Chen, Hao Liu, Zhe Zhang, Zhen Ni, Jie Chen, Zhiping Yang, Yongzhan Nie, Daiming Fan

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Multidrug resistance remains a major obstacle to successful treatment for patients with gastric cancer (GC). Recently, glycosylation has been demonstrated to play a vital role in the acquisition of multidrug resistance. As a potent inhibitor of glycosylation, tunicamycin (Tu) has shown marked antitumor activities in various cancers. In the present study, we attempted to determine the exact effect of Tu on the chemoresistance of GC.

Methods

The cytotoxic effects of drugs on GC cells were evaluated by cell viability assays, and apoptosis was detected by flow cytometry. PCR, western blot analysis, immunofluorescence staining and canonical inhibitors were employed to identify the underlying mechanisms of the specific effects of Tu on multidrug-resistant (MDR) GC cells.

Results

For the first time, we found that MDR GC cells were more sensitive to Tu-induced cell death than the parental cells and that the increased sensitivity might correlate with basal endoplasmic reticulum (ER) stress. In addition, Tu dramatically increased chemotherapy-induced apoptosis by evoking ER stress in GC cells, particularly MDR cells. Further study indicated that these effects were highly dependent on glycosylation inhibition by Tu, rather than its role as a canonical ER stress inducer. Besides, autophagy was markedly triggered by Tu, and blocking autophagy enhanced the combined effects of Tu and chemotherapy on MDR GC cells.

Conclusions

Our results suggest that tumor-targeted glycosylation inhibition may be a feasible strategy to reverse chemoresistance in GC patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.CrossRef Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.CrossRef
2.
go back to reference Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.CrossRef Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.CrossRef
3.
go back to reference Camargo MC, Kim WH, Chiaravalli AM, Kim KM, Corvalan AH, Matsuo K, et al. Improved survival of gastric cancer with tumour Epstein-Barr virus positivity: an international pooled analysis. Gut. 2014;63:236–43.CrossRef Camargo MC, Kim WH, Chiaravalli AM, Kim KM, Corvalan AH, Matsuo K, et al. Improved survival of gastric cancer with tumour Epstein-Barr virus positivity: an international pooled analysis. Gut. 2014;63:236–43.CrossRef
4.
go back to reference Very N, Lefebvre T, El Yazidi-Belkoura I. Drug resistance related to aberrant glycosylation in colorectal cancer. Oncotarget. 2018;9:1380–402.CrossRef Very N, Lefebvre T, El Yazidi-Belkoura I. Drug resistance related to aberrant glycosylation in colorectal cancer. Oncotarget. 2018;9:1380–402.CrossRef
5.
go back to reference Legler K, Rosprim R, Karius T, Eylmann K, Rossberg M, Wirtz RM, et al. Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer. Br J Cancer. 2018;118:847–56.CrossRef Legler K, Rosprim R, Karius T, Eylmann K, Rossberg M, Wirtz RM, et al. Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer. Br J Cancer. 2018;118:847–56.CrossRef
6.
go back to reference Jones RB, Dorsett KA, Hjelmeland AB, Bellis SL. The ST6Gal-I sialyltransferase protects tumor cells against hypoxia by enhancing HIF-1alpha signaling. J Biol Chem. 2018;293:5659–67.CrossRef Jones RB, Dorsett KA, Hjelmeland AB, Bellis SL. The ST6Gal-I sialyltransferase protects tumor cells against hypoxia by enhancing HIF-1alpha signaling. J Biol Chem. 2018;293:5659–67.CrossRef
7.
go back to reference Cui J, Huang W, Wu B, Jin J, Jing L, Shi WP, et al. N-glycosylation by N-acetylglucosaminyltransferase V enhances the interaction of CD147/basigin with integrin beta1 and promotes HCC metastasis. J Pathol. 2018;245:41–52.CrossRef Cui J, Huang W, Wu B, Jin J, Jing L, Shi WP, et al. N-glycosylation by N-acetylglucosaminyltransferase V enhances the interaction of CD147/basigin with integrin beta1 and promotes HCC metastasis. J Pathol. 2018;245:41–52.CrossRef
8.
go back to reference Chakraborty A, Dorsett KA, Trummell HQ, Yang ES, Oliver PG, Bonner JA, et al. ST6Gal-I sialyltransferase promotes chemoresistance in pancreatic ductal adenocarcinoma by abrogating gemcitabine-mediated DNA damage. J Biol Chem. 2018;293:984–94.CrossRef Chakraborty A, Dorsett KA, Trummell HQ, Yang ES, Oliver PG, Bonner JA, et al. ST6Gal-I sialyltransferase promotes chemoresistance in pancreatic ductal adenocarcinoma by abrogating gemcitabine-mediated DNA damage. J Biol Chem. 2018;293:984–94.CrossRef
9.
go back to reference Rao TD, Fernandez-Tejada A, Axelrod A, Rosales N, Yan X, Thapi S, et al. Antibodies against specific MUC16 glycosylation sites inhibit ovarian Cancer growth. ACS Chem Biol. 2017;12:2085–96.CrossRef Rao TD, Fernandez-Tejada A, Axelrod A, Rosales N, Yan X, Thapi S, et al. Antibodies against specific MUC16 glycosylation sites inhibit ovarian Cancer growth. ACS Chem Biol. 2017;12:2085–96.CrossRef
10.
go back to reference Agrawal P, Fontanals-Cirera B, Sokolova E, Jacob S, Vaiana CA, Argibay D, et al. A systems biology approach identifies FUT8 as a driver of melanoma metastasis. Cancer Cell. 2017;31:804–19.CrossRef Agrawal P, Fontanals-Cirera B, Sokolova E, Jacob S, Vaiana CA, Argibay D, et al. A systems biology approach identifies FUT8 as a driver of melanoma metastasis. Cancer Cell. 2017;31:804–19.CrossRef
11.
go back to reference Yen HY, Liu YC, Chen NY, Tsai CF, Wang YT, Chen YJ, et al. Effect of sialylation on EGFR phosphorylation and resistance to tyrosine kinase inhibition. Proc Natl Acad Sci U S A. 2015;112:6955–60.CrossRef Yen HY, Liu YC, Chen NY, Tsai CF, Wang YT, Chen YJ, et al. Effect of sialylation on EGFR phosphorylation and resistance to tyrosine kinase inhibition. Proc Natl Acad Sci U S A. 2015;112:6955–60.CrossRef
12.
go back to reference Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15:540–55.CrossRef Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer. 2015;15:540–55.CrossRef
13.
go back to reference Li K, Sun Z, Zheng J, Lu Y, Bian Y, Ye M, et al. In-depth research of multidrug resistance related cell surface glycoproteome in gastric cancer. J Proteome. 2013;82:130–40.CrossRef Li K, Sun Z, Zheng J, Lu Y, Bian Y, Ye M, et al. In-depth research of multidrug resistance related cell surface glycoproteome in gastric cancer. J Proteome. 2013;82:130–40.CrossRef
14.
go back to reference Wu J, Qin H, Li T, Cheng K, Dong J, Tian M, et al. Characterization of site-specific glycosylation of secreted proteins associated with multi-drug resistance of gastric cancer. Oncotarget. 2016;7:25315–27.PubMedPubMedCentral Wu J, Qin H, Li T, Cheng K, Dong J, Tian M, et al. Characterization of site-specific glycosylation of secreted proteins associated with multi-drug resistance of gastric cancer. Oncotarget. 2016;7:25315–27.PubMedPubMedCentral
15.
go back to reference Surani MA. Glycoprotein synthesis and inhibition of glycosylation by tunicamycin in preimplantation mouse embryos: compaction and trophoblast adhesion. Cell. 1979;18:217–27.CrossRef Surani MA. Glycoprotein synthesis and inhibition of glycosylation by tunicamycin in preimplantation mouse embryos: compaction and trophoblast adhesion. Cell. 1979;18:217–27.CrossRef
16.
go back to reference Yoo J, Mashalidis EH, Kuk ACY, Yamamoto K, Kaeser B, Ichikawa S, et al. GlcNAc-1-P-transferase-tunicamycin complex structure reveals basis for inhibition of N-glycosylation. Nat Struct Mol Biol. 2018;25:217–24.CrossRef Yoo J, Mashalidis EH, Kuk ACY, Yamamoto K, Kaeser B, Ichikawa S, et al. GlcNAc-1-P-transferase-tunicamycin complex structure reveals basis for inhibition of N-glycosylation. Nat Struct Mol Biol. 2018;25:217–24.CrossRef
17.
go back to reference Hakulinen JK, Hering J, Branden G, Chen H, Snijder A, Ek M, et al. MraY-antibiotic complex reveals details of tunicamycin mode of action. Nat Chem Biol. 2017;13:265–7.CrossRef Hakulinen JK, Hering J, Branden G, Chen H, Snijder A, Ek M, et al. MraY-antibiotic complex reveals details of tunicamycin mode of action. Nat Chem Biol. 2017;13:265–7.CrossRef
18.
go back to reference Jung YH, Lim EJ, Heo J, Kwon TK, Kim YH. Tunicamycin sensitizes human prostate cells to TRAIL-induced apoptosis by upregulation of TRAIL receptors and downregulation of cIAP2. Int J Oncol. 2012;40:1941–8.CrossRef Jung YH, Lim EJ, Heo J, Kwon TK, Kim YH. Tunicamycin sensitizes human prostate cells to TRAIL-induced apoptosis by upregulation of TRAIL receptors and downregulation of cIAP2. Int J Oncol. 2012;40:1941–8.CrossRef
19.
go back to reference Contessa JN, Bhojani MS, Freeze HH, Rehemtulla A, Lawrence TS. Inhibition of N-linked glycosylation disrupts receptor tyrosine kinase signaling in tumor cells. Cancer Res. 2008;68:3803–9.CrossRef Contessa JN, Bhojani MS, Freeze HH, Rehemtulla A, Lawrence TS. Inhibition of N-linked glycosylation disrupts receptor tyrosine kinase signaling in tumor cells. Cancer Res. 2008;68:3803–9.CrossRef
20.
go back to reference de-Freitas-Junior JC, Bastos LG, Freire-Neto CA, Rocher BD, Abdelhay ES, Morgado-Diaz JA. N-glycan biosynthesis inhibitors induce in vitro anticancer activity in colorectal cancer cells. J Cell Biochem. 2012;113:2957–66.CrossRef de-Freitas-Junior JC, Bastos LG, Freire-Neto CA, Rocher BD, Abdelhay ES, Morgado-Diaz JA. N-glycan biosynthesis inhibitors induce in vitro anticancer activity in colorectal cancer cells. J Cell Biochem. 2012;113:2957–66.CrossRef
21.
go back to reference Hou H, Sun H, Lu P, Ge C, Zhang L, Li H, et al. Tunicamycin potentiates cisplatin anticancer efficacy through the DPAGT1/Akt/ABCG2 pathway in mouse xenograft models of human hepatocellular carcinoma. Mol Cancer Ther. 2013;12:2874–84.CrossRef Hou H, Sun H, Lu P, Ge C, Zhang L, Li H, et al. Tunicamycin potentiates cisplatin anticancer efficacy through the DPAGT1/Akt/ABCG2 pathway in mouse xenograft models of human hepatocellular carcinoma. Mol Cancer Ther. 2013;12:2874–84.CrossRef
22.
go back to reference Wojtowicz K, Januchowski R, Nowicki M, Zabel M. Inhibition of protein glycosylation reverses the MDR phenotype of cancer cell lines. Biomed Pharmacother. 2015;74:49–56.CrossRef Wojtowicz K, Januchowski R, Nowicki M, Zabel M. Inhibition of protein glycosylation reverses the MDR phenotype of cancer cell lines. Biomed Pharmacother. 2015;74:49–56.CrossRef
23.
go back to reference Gan PP, Zhou YY, Zhong MZ, Peng Y, Li L, Li JH. Endoplasmic reticulum stress promotes autophagy and apoptosis and reduces chemotherapy resistance in mutant p53 lung Cancer cells. Cell Physiol Biochem. 2017;44:133–51.CrossRef Gan PP, Zhou YY, Zhong MZ, Peng Y, Li L, Li JH. Endoplasmic reticulum stress promotes autophagy and apoptosis and reduces chemotherapy resistance in mutant p53 lung Cancer cells. Cell Physiol Biochem. 2017;44:133–51.CrossRef
24.
go back to reference Fu YF, Liu X, Gao M, Zhang YN, Liu J. Endoplasmic reticulum stress induces autophagy and apoptosis while inhibiting proliferation and drug resistance in multiple myeloma through the PI3K/Akt/mTOR signaling pathway. Oncotarget. 2017;8:61093–106.PubMedPubMedCentral Fu YF, Liu X, Gao M, Zhang YN, Liu J. Endoplasmic reticulum stress induces autophagy and apoptosis while inhibiting proliferation and drug resistance in multiple myeloma through the PI3K/Akt/mTOR signaling pathway. Oncotarget. 2017;8:61093–106.PubMedPubMedCentral
25.
go back to reference Ma Y, Hendershot LM. The role of the unfolded protein response in tumour development: friend or foe? Nat Rev Cancer. 2004;4:966–77.CrossRef Ma Y, Hendershot LM. The role of the unfolded protein response in tumour development: friend or foe? Nat Rev Cancer. 2004;4:966–77.CrossRef
26.
go back to reference Hetz C, Papa FR. The unfolded protein response and cell fate control. Mol Cell. 2018;69:169–81.CrossRef Hetz C, Papa FR. The unfolded protein response and cell fate control. Mol Cell. 2018;69:169–81.CrossRef
27.
go back to reference Urra H, Dufey E, Avril T, Chevet E, Hetz C. Endoplasmic reticulum stress and the hallmarks of Cancer. Trends Cancer. 2016;2:252–62.CrossRef Urra H, Dufey E, Avril T, Chevet E, Hetz C. Endoplasmic reticulum stress and the hallmarks of Cancer. Trends Cancer. 2016;2:252–62.CrossRef
28.
go back to reference Carlberg M, Dricu A, Blegen H, Kass GE, Orrenius S, Larsson O. Short exposures to tunicamycin induce apoptosis in SV40-transformed but not in normal human fibroblasts. Carcinogenesis. 1996;17:2589–96.CrossRef Carlberg M, Dricu A, Blegen H, Kass GE, Orrenius S, Larsson O. Short exposures to tunicamycin induce apoptosis in SV40-transformed but not in normal human fibroblasts. Carcinogenesis. 1996;17:2589–96.CrossRef
29.
go back to reference Qin L, Wang Z, Tao L, Wang Y. ER stress negatively regulates AKT/TSC/mTOR pathway to enhance autophagy. Autophagy. 2010;6:239–47.CrossRef Qin L, Wang Z, Tao L, Wang Y. ER stress negatively regulates AKT/TSC/mTOR pathway to enhance autophagy. Autophagy. 2010;6:239–47.CrossRef
30.
go back to reference Huang Z, Zhou L, Chen Z, Nice EC, Huang C. Stress management by autophagy: implications for chemoresistance. Int J Cancer. 2016;139:23–32.CrossRef Huang Z, Zhou L, Chen Z, Nice EC, Huang C. Stress management by autophagy: implications for chemoresistance. Int J Cancer. 2016;139:23–32.CrossRef
31.
go back to reference Hoyer-Hansen M, Jaattela M. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ. 2007;14:1576–82.CrossRef Hoyer-Hansen M, Jaattela M. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ. 2007;14:1576–82.CrossRef
32.
go back to reference Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012;13:89–102.CrossRef Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012;13:89–102.CrossRef
33.
go back to reference Yan MM, Ni JD, Song D, Ding M, Huang J. Interplay between unfolded protein response and autophagy promotes tumor drug resistance. Oncol Lett. 2015;10:1959–69.CrossRef Yan MM, Ni JD, Song D, Ding M, Huang J. Interplay between unfolded protein response and autophagy promotes tumor drug resistance. Oncol Lett. 2015;10:1959–69.CrossRef
34.
go back to reference Rodvold JJ, Chiu KT, Hiramatsu N, Nussbacher JK, Galimberti V, Mahadevan NR, et al. Intercellular transmission of the unfolded protein response promotes survival and drug resistance in cancer cells. Sci Signal. 2017;10:482.CrossRef Rodvold JJ, Chiu KT, Hiramatsu N, Nussbacher JK, Galimberti V, Mahadevan NR, et al. Intercellular transmission of the unfolded protein response promotes survival and drug resistance in cancer cells. Sci Signal. 2017;10:482.CrossRef
35.
go back to reference Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, et al. Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. J Clin Invest. 2014;124:1406–17.CrossRef Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, et al. Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. J Clin Invest. 2014;124:1406–17.CrossRef
36.
go back to reference Lee AS. GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 2007;67:3496–9.CrossRef Lee AS. GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 2007;67:3496–9.CrossRef
37.
go back to reference Higa A, Taouji S, Lhomond S, Jensen D, Fernandez-Zapico ME, Simpson JC, et al. Endoplasmic reticulum stress-activated transcription factor ATF6alpha requires the disulfide isomerase PDIA5 to modulate chemoresistance. Mol Cell Biol. 2014;34:1839–49.CrossRef Higa A, Taouji S, Lhomond S, Jensen D, Fernandez-Zapico ME, Simpson JC, et al. Endoplasmic reticulum stress-activated transcription factor ATF6alpha requires the disulfide isomerase PDIA5 to modulate chemoresistance. Mol Cell Biol. 2014;34:1839–49.CrossRef
38.
go back to reference Gifford JB, Huang W, Zeleniak AE, Hindoyan A, Wu H, Donahue TR, et al. Expression of GRP78, master regulator of the unfolded protein response, increases Chemoresistance in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2016;15:1043–52.CrossRef Gifford JB, Huang W, Zeleniak AE, Hindoyan A, Wu H, Donahue TR, et al. Expression of GRP78, master regulator of the unfolded protein response, increases Chemoresistance in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2016;15:1043–52.CrossRef
39.
go back to reference Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007;67:9809–16.CrossRef Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007;67:9809–16.CrossRef
40.
go back to reference Avril T, Vauleon E, Chevet E. Endoplasmic reticulum stress signaling and chemotherapy resistance in solid cancers. Oncogene. 2017;6:e373.CrossRef Avril T, Vauleon E, Chevet E. Endoplasmic reticulum stress signaling and chemotherapy resistance in solid cancers. Oncogene. 2017;6:e373.CrossRef
41.
go back to reference Han X, Zhang X, Li H, Huang S, Zhang S, Wang F, et al. Tunicamycin enhances the antitumor activity of trastuzumab on breast cancer in vitro and in vivo. Oncotarget. 2015;6:38912–25.PubMedPubMedCentral Han X, Zhang X, Li H, Huang S, Zhang S, Wang F, et al. Tunicamycin enhances the antitumor activity of trastuzumab on breast cancer in vitro and in vivo. Oncotarget. 2015;6:38912–25.PubMedPubMedCentral
42.
go back to reference Sehgal P, Szalai P, Olesen C, Praetorius HA, Nissen P, Christensen SB, et al. Inhibition of the sarco/endoplasmic reticulum (ER) ca (2+)-ATPase by thapsigargin analogs induces cell death via ER ca (2+) depletion and the unfolded protein response. J Biol Chem. 2017;292:19656–73.CrossRef Sehgal P, Szalai P, Olesen C, Praetorius HA, Nissen P, Christensen SB, et al. Inhibition of the sarco/endoplasmic reticulum (ER) ca (2+)-ATPase by thapsigargin analogs induces cell death via ER ca (2+) depletion and the unfolded protein response. J Biol Chem. 2017;292:19656–73.CrossRef
43.
go back to reference Kornfeld R, Kornfeld S. Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem. 1985;54:631–64.CrossRef Kornfeld R, Kornfeld S. Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem. 1985;54:631–64.CrossRef
44.
45.
go back to reference Sampath D, Varki A, Freeze HH. The spectrum of incomplete N-linked oligosaccharides synthesized by endothelial cells in the presence of brefeldin a. J Biol Chem. 1992;267:4440–55.PubMed Sampath D, Varki A, Freeze HH. The spectrum of incomplete N-linked oligosaccharides synthesized by endothelial cells in the presence of brefeldin a. J Biol Chem. 1992;267:4440–55.PubMed
46.
go back to reference Peyroche A, Antonny B, Robineau S, Acker J, Cherfils J, Jackson CL. Brefeldin a acts to stabilize an abortive ARF-GDP-Sec7 domain protein complex: involvement of specific residues of the Sec7 domain. Mol Cell. 1999;3:275–85.CrossRef Peyroche A, Antonny B, Robineau S, Acker J, Cherfils J, Jackson CL. Brefeldin a acts to stabilize an abortive ARF-GDP-Sec7 domain protein complex: involvement of specific residues of the Sec7 domain. Mol Cell. 1999;3:275–85.CrossRef
47.
go back to reference Klausner RD, Donaldson JG, Lippincott-Schwartz J, Brefeldin A. Insights into the control of membrane traffic and organelle structure. J Cell Biol. 1992;116:1071–80.CrossRef Klausner RD, Donaldson JG, Lippincott-Schwartz J, Brefeldin A. Insights into the control of membrane traffic and organelle structure. J Cell Biol. 1992;116:1071–80.CrossRef
48.
go back to reference Britain CM, Holdbrooks AT, Anderson JC, Willey CD, Bellis SL. Sialylation of EGFR by the ST6Gal-I sialyltransferase promotes EGFR activation and resistance to gefitinib-mediated cell death. J Ovarian Res. 2018;11:12.CrossRef Britain CM, Holdbrooks AT, Anderson JC, Willey CD, Bellis SL. Sialylation of EGFR by the ST6Gal-I sialyltransferase promotes EGFR activation and resistance to gefitinib-mediated cell death. J Ovarian Res. 2018;11:12.CrossRef
49.
go back to reference Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, et al. Mechanisms of cisplatin resistance and targeting of cancer stem cells: adding glycosylation to the equation. Drug Resist Updat. 2016;24:34–54.CrossRef Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, et al. Mechanisms of cisplatin resistance and targeting of cancer stem cells: adding glycosylation to the equation. Drug Resist Updat. 2016;24:34–54.CrossRef
50.
go back to reference Feng X, Zhao L, Gao S, Song X, Dong W, Zhao Y, et al. Increased fucosylation has a pivotal role in multidrug resistance of breast cancer cells through miR-224-3p targeting FUT4. Gene. 2016;578:232–41.CrossRef Feng X, Zhao L, Gao S, Song X, Dong W, Zhao Y, et al. Increased fucosylation has a pivotal role in multidrug resistance of breast cancer cells through miR-224-3p targeting FUT4. Gene. 2016;578:232–41.CrossRef
51.
go back to reference da Fonseca LM, da Silva VA, Freire-de-Lima L, Previato JO, Mendonca-Previato L, Capella MA. Glycosylation in Cancer: interplay between multidrug resistance and epithelial-to-mesenchymal transition? Front Oncol. 2016;6:158.CrossRef da Fonseca LM, da Silva VA, Freire-de-Lima L, Previato JO, Mendonca-Previato L, Capella MA. Glycosylation in Cancer: interplay between multidrug resistance and epithelial-to-mesenchymal transition? Front Oncol. 2016;6:158.CrossRef
52.
go back to reference Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, et al. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007;14:230–9.CrossRef Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, et al. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007;14:230–9.CrossRef
53.
go back to reference Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000;287:664–6.CrossRef Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000;287:664–6.CrossRef
54.
go back to reference Wang J, Kang R, Huang H, Xi X, Wang B, Wang J, et al. Hepatitis C virus core protein activates autophagy through EIF2AK3 and ATF6 UPR pathway-mediated MAP1LC3B and ATG12 expression. Autophagy. 2014;10:766–84.CrossRef Wang J, Kang R, Huang H, Xi X, Wang B, Wang J, et al. Hepatitis C virus core protein activates autophagy through EIF2AK3 and ATF6 UPR pathway-mediated MAP1LC3B and ATG12 expression. Autophagy. 2014;10:766–84.CrossRef
55.
go back to reference Mizushima N. Autophagy: process and function. Genes Dev. 2007;21:2861–73.CrossRef Mizushima N. Autophagy: process and function. Genes Dev. 2007;21:2861–73.CrossRef
56.
go back to reference Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 2011;27:107–32.CrossRef Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 2011;27:107–32.CrossRef
57.
go back to reference Singh SS, Vats S, Chia AY, Tan TZ, Deng S, Ong MS, et al. Dual role of autophagy in hallmarks of cancer. Oncogene. 2018;37:1142–58.CrossRef Singh SS, Vats S, Chia AY, Tan TZ, Deng S, Ong MS, et al. Dual role of autophagy in hallmarks of cancer. Oncogene. 2018;37:1142–58.CrossRef
58.
go back to reference Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17:528–42.CrossRef Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17:528–42.CrossRef
59.
go back to reference Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S, et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol. 2006;26:9220–31.CrossRef Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S, et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol. 2006;26:9220–31.CrossRef
60.
go back to reference Cheng X, Liu H, Jiang CC, Fang L, Chen C, Zhang XD, et al. Connecting endoplasmic reticulum stress to autophagy through IRE1/JNK/beclin-1 in breast cancer cells. Int J Mol Med. 2014;34:772–81.CrossRef Cheng X, Liu H, Jiang CC, Fang L, Chen C, Zhang XD, et al. Connecting endoplasmic reticulum stress to autophagy through IRE1/JNK/beclin-1 in breast cancer cells. Int J Mol Med. 2014;34:772–81.CrossRef
61.
go back to reference Yang YP, Hu LF, Zheng HF, Mao CJ, Hu WD, Xiong KP, et al. Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol Sin. 2013;34:625–35.CrossRef Yang YP, Hu LF, Zheng HF, Mao CJ, Hu WD, Xiong KP, et al. Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol Sin. 2013;34:625–35.CrossRef
Metadata
Title
Tunicamycin specifically aggravates ER stress and overcomes chemoresistance in multidrug-resistant gastric cancer cells by inhibiting N-glycosylation
Authors
Jian Wu
Sheng Chen
Hao Liu
Zhe Zhang
Zhen Ni
Jie Chen
Zhiping Yang
Yongzhan Nie
Daiming Fan
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0935-8

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 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