Skip to main content
Erschienen in: Cellular Oncology 6/2017

19.09.2017 | Original Article

Glucosidase II beta subunit (GluIIβ) plays a role in autophagy and apoptosis regulation in lung carcinoma cells in a p53-dependent manner

verfasst von: Worapong Khaodee, Nichanan Inboot, Suruk Udomsom, Warunee Kumsaiyai, Ratchada Cressey

Erschienen in: Cellular Oncology | Ausgabe 6/2017

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Glucosidase II plays a major role in regulating the post-translational modification of N-linked glycoproteins. Previously, we found that the beta subunit of glucosidase II (GluIIβ) levels are significantly increased in lung carcinoma tissues, indicating a potential role in lung tumorigenesis. Here, we investigated the role of GluIIβ in the regulation of autophagy and apoptosis in lung carcinoma- and immortalized human bronchial epithelial-derived cells.

Methods

A selective glucosidase II inhibitor, bromoconduritol, was used to inhibit GluII enzyme activity and a siRNA-based technology was used to suppress the expression of the GluIIβ encoding gene PRKCSH in lung carcinoma cells differing in p53 status. Cell viability was assessed using a MTT assay, cell cycle progression was assessed using flow cytometry, autophagy was assessed using Western blotting and apoptosis was assessed using an annexin V-FITC/PI double labeling method.

Results

We found that GluIIβ inhibition resulted in the induction of autophagy in all cell lines tested, but apoptosis in only wild-type p53 cells. We also found that GluIIβ inhibition dose-dependently decreased activation of the EGFR/RTK and PI3K/AKT signaling pathways. Although the apoptosis inducing effect of GluIIβ inhibition appeared to be p53-dependent, we found that a combined treatment with lysosomal inhibitors to block autophagy enhanced the apoptotic effect of GluIIβ inhibition in both wild-type p53 and p53-null cells.

Conclusions

Our data indicate that GluIIβ inhibition results in autophagy and apoptosis in lung carcinoma-derived cells, supporting the hypothesis that this enzyme may play a role in blocking these two tumor suppressive processes. Since blocking autophagy by lysosomal inhibitors enhanced the apoptosis-inducing effect of bromoconduritol, independent of p53 status, their combined use may hold promise for the treatment of cancer, particularly lung cancer.
Literatur
1.
Zurück zum Zitat B. Suradej, S. Pata, W. Kasinrerk, R. Cressey, Glucosidase II exhibits similarity to the p53 tumor suppressor in regards to structure and behavior in response to stress signals: a potential novel cancer biomarker. Oncol. Rep. 30, 2511–2519 (2013)CrossRefPubMed B. Suradej, S. Pata, W. Kasinrerk, R. Cressey, Glucosidase II exhibits similarity to the p53 tumor suppressor in regards to structure and behavior in response to stress signals: a potential novel cancer biomarker. Oncol. Rep. 30, 2511–2519 (2013)CrossRefPubMed
2.
Zurück zum Zitat S.C. Taylor, P. Thibault, D.C. Tessier, J.J. Bergeron, D.Y. Thomas, Glycopeptide specificity of the secretory protein folding sensor UDP-glucose glycoprotein:glucosyltransferase. EMBO Rep. 4, 405–411 (2003)CrossRefPubMedPubMedCentral S.C. Taylor, P. Thibault, D.C. Tessier, J.J. Bergeron, D.Y. Thomas, Glycopeptide specificity of the secretory protein folding sensor UDP-glucose glycoprotein:glucosyltransferase. EMBO Rep. 4, 405–411 (2003)CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat J.P. Drenth, J.A. Martina, R. van de Kerkhof, J.S. Bonifacino, J.B. Jansen, Polycystic liver disease is a disorder of cotranslational protein processing. Trends Mol. Med. 11, 37–42 (2005)CrossRefPubMed J.P. Drenth, J.A. Martina, R. van de Kerkhof, J.S. Bonifacino, J.B. Jansen, Polycystic liver disease is a disorder of cotranslational protein processing. Trends Mol. Med. 11, 37–42 (2005)CrossRefPubMed
4.
Zurück zum Zitat A. Li, S. Davila, L. Furu, Q. Qian, X. Tian, P.S. Kamath, B.F. King, V.E. Torres, S. Somlo, Mutations in PRKCSH cause isolated autosomal dominant polycystic liver disease. Am. J. Hum. Genet. 72, 691–703 (2003)CrossRefPubMedPubMedCentral A. Li, S. Davila, L. Furu, Q. Qian, X. Tian, P.S. Kamath, B.F. King, V.E. Torres, S. Somlo, Mutations in PRKCSH cause isolated autosomal dominant polycystic liver disease. Am. J. Hum. Genet. 72, 691–703 (2003)CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat R. Palmirotta, F. Guadagni, A. Savonarola, G. Ludovici, M.L. De Marchis, D. Palli, M. Falchetti, L. Ottini, PRKCSH GAG trinucleotide repeat is a mutational target in gastric carcinomas with high-level microsatellite instability. Clin. Genet. 79, 397–398 author reply 399-400 R. Palmirotta, F. Guadagni, A. Savonarola, G. Ludovici, M.L. De Marchis, D. Palli, M. Falchetti, L. Ottini, PRKCSH GAG trinucleotide repeat is a mutational target in gastric carcinomas with high-level microsatellite instability. Clin. Genet. 79, 397–398 author reply 399-400
6.
Zurück zum Zitat R. Forough, L. Lindner, C. Partridge, B. Jones, G. Guy, G. Clark, Elevated 80K-H protein in breast cancer: a role for FGF-1 stimulation of 80K-H. Int. J. Biol. Markers 18, 89–98 (2003)CrossRefPubMed R. Forough, L. Lindner, C. Partridge, B. Jones, G. Guy, G. Clark, Elevated 80K-H protein in breast cancer: a role for FGF-1 stimulation of 80K-H. Int. J. Biol. Markers 18, 89–98 (2003)CrossRefPubMed
7.
Zurück zum Zitat K.C. Goh, Y.P. Lim, S.H. Ong, C.B. Siak, X. Cao, Y.H. Tan, G.R. Guy, Identification of p90, a prominent tyrosine-phosphorylated protein in fibroblast growth factor-stimulated cells, as 80K-H. J. Biol. Chem. 271, 5832–5838 (1996)CrossRefPubMed K.C. Goh, Y.P. Lim, S.H. Ong, C.B. Siak, X. Cao, Y.H. Tan, G.R. Guy, Identification of p90, a prominent tyrosine-phosphorylated protein in fibroblast growth factor-stimulated cells, as 80K-H. J. Biol. Chem. 271, 5832–5838 (1996)CrossRefPubMed
8.
Zurück zum Zitat J. Yang, Y. Zhao, K. Ma, F.J. Jiang, W. Liao, P. Zhang, J. Zhou, B. Tu, L. Wang, H.H. Kampinga, Z. Xie, W.G. Zhu, Deficiency of hepatocystin induces autophagy through an mTOR-dependent pathway. Autophagy 7, 748–759 (2011)CrossRefPubMed J. Yang, Y. Zhao, K. Ma, F.J. Jiang, W. Liao, P. Zhang, J. Zhou, B. Tu, L. Wang, H.H. Kampinga, Z. Xie, W.G. Zhu, Deficiency of hepatocystin induces autophagy through an mTOR-dependent pathway. Autophagy 7, 748–759 (2011)CrossRefPubMed
9.
Zurück zum Zitat J. Cui, B. Chen, H. Wang, Y. Han, X. Chen, W. Zhang, Glucosidase II beta-subunit, a novel substrate for caspase-3-like activity in rice, plays as a molecular switch between autophagy and programmed cell death. Sci. Rep. 6, 31764 (2016)CrossRefPubMedPubMedCentral J. Cui, B. Chen, H. Wang, Y. Han, X. Chen, W. Zhang, Glucosidase II beta-subunit, a novel substrate for caspase-3-like activity in rice, plays as a molecular switch between autophagy and programmed cell death. Sci. Rep. 6, 31764 (2016)CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat S. Jin, Autophagy, mitochondrial quality control, and oncogenesis. Autophagy 2, 80–84 (2006)CrossRefPubMed S. Jin, Autophagy, mitochondrial quality control, and oncogenesis. Autophagy 2, 80–84 (2006)CrossRefPubMed
15.
Zurück zum Zitat W.J. Buchser, T.C. Laskow, P.J. Pavlik, H.M. Lin, M.T. Lotze, Cell-mediated autophagy promotes cancer cell survival. Cancer Res. 72, 2970–2979 (2012)CrossRefPubMedPubMedCentral W.J. Buchser, T.C. Laskow, P.J. Pavlik, H.M. Lin, M.T. Lotze, Cell-mediated autophagy promotes cancer cell survival. Cancer Res. 72, 2970–2979 (2012)CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat M.J. Hayat, N. Howlader, M.E. Reichman, B.K. Edwards, Cancer statistics, trends, and multiple primary cancer analyses from the Surveillance, Epidemiology, and End Results (SEER) program. Oncologist 12, 20–37 (2007)CrossRefPubMed M.J. Hayat, N. Howlader, M.E. Reichman, B.K. Edwards, Cancer statistics, trends, and multiple primary cancer analyses from the Surveillance, Epidemiology, and End Results (SEER) program. Oncologist 12, 20–37 (2007)CrossRefPubMed
17.
Zurück zum Zitat J.R. Jett, D.E. Midthun, Screening for lung cancer: current status and future directions: Thomas A. Neff lecture. Chest 125, 158S–162S (2004)CrossRefPubMed J.R. Jett, D.E. Midthun, Screening for lung cancer: current status and future directions: Thomas A. Neff lecture. Chest 125, 158S–162S (2004)CrossRefPubMed
18.
Zurück zum Zitat P. Chen, J. Li, Y.C. Chen, H. Qian, Y.J. Chen, J.Y. Su, M. Wu, T. Lan, The functional status of DNA repair pathways determines the sensitization effect to cisplatin in non-small cell lung cancer cells. Cell. Oncol. 39, 511–522 (2016)CrossRef P. Chen, J. Li, Y.C. Chen, H. Qian, Y.J. Chen, J.Y. Su, M. Wu, T. Lan, The functional status of DNA repair pathways determines the sensitization effect to cisplatin in non-small cell lung cancer cells. Cell. Oncol. 39, 511–522 (2016)CrossRef
19.
Zurück zum Zitat M. Abend, Reasons to reconsider the significance of apoptosis for cancer therapy. Int. J. Radiat. Biol. 79, 927–941 (2003)CrossRefPubMed M. Abend, Reasons to reconsider the significance of apoptosis for cancer therapy. Int. J. Radiat. Biol. 79, 927–941 (2003)CrossRefPubMed
20.
Zurück zum Zitat A. Melet, K. Song, O. Bucur, Z. Jagani, A.R. Grassian, R. Khosravi-Far, Apoptotic pathways in tumor progression and therapy. Adv. Exp. Med. Biol. 615, 47–79 (2008)CrossRefPubMed A. Melet, K. Song, O. Bucur, Z. Jagani, A.R. Grassian, R. Khosravi-Far, Apoptotic pathways in tumor progression and therapy. Adv. Exp. Med. Biol. 615, 47–79 (2008)CrossRefPubMed
21.
Zurück zum Zitat S.W. Han, J. Roman, Targeting apoptotic signaling pathways in human lung cancer. Curr. Cancer Drug Targets 10, 566–574 (2010)CrossRefPubMed S.W. Han, J. Roman, Targeting apoptotic signaling pathways in human lung cancer. Curr. Cancer Drug Targets 10, 566–574 (2010)CrossRefPubMed
22.
Zurück zum Zitat G. Cheng, D. Kong, X. Hou, B. Liang, M. He, N. Liang, S. Ma, X. Liu, The tumor suppressor, p53, contributes to radiosensitivity of lung cancer cells by regulating autophagy and apoptosis. Cancer Biother. Radiopharm. 28, 153–159 (2013)CrossRefPubMedPubMedCentral G. Cheng, D. Kong, X. Hou, B. Liang, M. He, N. Liang, S. Ma, X. Liu, The tumor suppressor, p53, contributes to radiosensitivity of lung cancer cells by regulating autophagy and apoptosis. Cancer Biother. Radiopharm. 28, 153–159 (2013)CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Y. Takeda, K. Totani, I. Matsuo, Y. Ito, The action of bromoconduritol on ER glucosidase II. Bioorg. Med. Chem. Lett. 20, 5357–5359 (2010)CrossRefPubMed Y. Takeda, K. Totani, I. Matsuo, Y. Ito, The action of bromoconduritol on ER glucosidase II. Bioorg. Med. Chem. Lett. 20, 5357–5359 (2010)CrossRefPubMed
25.
26.
Zurück zum Zitat M. Jung, J. Lee, H.Y. Seo, J.S. Lim, E.K. Kim, Cathepsin inhibition-induced lysosomal dysfunction enhances pancreatic beta-cell apoptosis in high glucose. PLoS One 10, e0116972 (2015)CrossRefPubMedPubMedCentral M. Jung, J. Lee, H.Y. Seo, J.S. Lim, E.K. Kim, Cathepsin inhibition-induced lysosomal dysfunction enhances pancreatic beta-cell apoptosis in high glucose. PLoS One 10, e0116972 (2015)CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat R. Itotani, S. Marumo, M. Fukui, 164P: A review of prognostic factors for patients with epidermal growth factor receptor mutation positive advanced non small cell lung cancer (EGFR+NSCLC). J. Thorac. Oncol. 11, S129 (2016)CrossRefPubMed R. Itotani, S. Marumo, M. Fukui, 164P: A review of prognostic factors for patients with epidermal growth factor receptor mutation positive advanced non small cell lung cancer (EGFR+NSCLC). J. Thorac. Oncol. 11, S129 (2016)CrossRefPubMed
28.
Zurück zum Zitat Y.X. Bao, X.D. Zhao, H.B. Deng, C.L. Lu, Y. Guo, X. Lu, L.L. Deng, Schedule-dependent cytotoxicity of sunitinib and TRAIL in human non-small cell lung cancer cells with or without EGFR and KRAS mutations. Cell. Oncol. 39, 343–352 (2016)CrossRef Y.X. Bao, X.D. Zhao, H.B. Deng, C.L. Lu, Y. Guo, X. Lu, L.L. Deng, Schedule-dependent cytotoxicity of sunitinib and TRAIL in human non-small cell lung cancer cells with or without EGFR and KRAS mutations. Cell. Oncol. 39, 343–352 (2016)CrossRef
29.
Zurück zum Zitat J.N. Contessa, M.S. Bhojani, H.H. Freeze, A. Rehemtulla, T.S. Lawrence, Inhibition of N-linked glycosylation disrupts receptor tyrosine kinase signaling in tumor cells. Cancer Res. 68, 3803–3809 (2008)CrossRefPubMedPubMedCentral J.N. Contessa, M.S. Bhojani, H.H. Freeze, A. Rehemtulla, T.S. Lawrence, Inhibition of N-linked glycosylation disrupts receptor tyrosine kinase signaling in tumor cells. Cancer Res. 68, 3803–3809 (2008)CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat C. Yewale, D. Baradia, I. Vhora, S. Patil, A. Misra, Epidermal growth factor receptor targeting in cancer: a review of trends and strategies. Biomaterials 34, 8690–8707 (2013)CrossRefPubMed C. Yewale, D. Baradia, I. Vhora, S. Patil, A. Misra, Epidermal growth factor receptor targeting in cancer: a review of trends and strategies. Biomaterials 34, 8690–8707 (2013)CrossRefPubMed
31.
Zurück zum Zitat Y. Wei, Z. Zou, N. Becker, M. Anderson, R. Sumpter, G. Xiao, L. Kinch, P. Koduru, C.S. Christudass, R.W. Veltri, N.V. Grishin, M. Peyton, J. Minna, G. Bhagat, B. Levine, EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell 154, 1269–1284 (2013)CrossRefPubMedPubMedCentral Y. Wei, Z. Zou, N. Becker, M. Anderson, R. Sumpter, G. Xiao, L. Kinch, P. Koduru, C.S. Christudass, R.W. Veltri, N.V. Grishin, M. Peyton, J. Minna, G. Bhagat, B. Levine, EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell 154, 1269–1284 (2013)CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat H. Fernandes, S. Cohen, S. Bishayee, Glycosylation-induced conformational modification positively regulates receptor-receptor association: a study with an aberrant epidermal growth factor receptor (EGFRvIII/DeltaEGFR) expressed in cancer cells. J. Biol. Chem. 276, 5375–5383 (2001)CrossRefPubMed H. Fernandes, S. Cohen, S. Bishayee, Glycosylation-induced conformational modification positively regulates receptor-receptor association: a study with an aberrant epidermal growth factor receptor (EGFRvIII/DeltaEGFR) expressed in cancer cells. J. Biol. Chem. 276, 5375–5383 (2001)CrossRefPubMed
33.
Zurück zum Zitat H.B. Guo, M. Randolph, M. Pierce, Inhibition of a specific N-glycosylation activity results in attenuation of breast carcinoma cell invasiveness-related phenotypes: inhibition of epidermal growth factor-induced dephosphorylation of focal adhesion kinase. J. Biol. Chem. 282, 22150–22162 (2007)CrossRefPubMed H.B. Guo, M. Randolph, M. Pierce, Inhibition of a specific N-glycosylation activity results in attenuation of breast carcinoma cell invasiveness-related phenotypes: inhibition of epidermal growth factor-induced dephosphorylation of focal adhesion kinase. J. Biol. Chem. 282, 22150–22162 (2007)CrossRefPubMed
34.
Zurück zum Zitat X. Wang, J. Gu, H. Ihara, E. Miyoshi, K. Honke, N. Taniguchi, Core fucosylation regulates epidermal growth factor receptor-mediated intracellular signaling. J. Biol. Chem. 281, 2572–2577 (2006)CrossRefPubMed X. Wang, J. Gu, H. Ihara, E. Miyoshi, K. Honke, N. Taniguchi, Core fucosylation regulates epidermal growth factor receptor-mediated intracellular signaling. J. Biol. Chem. 281, 2572–2577 (2006)CrossRefPubMed
35.
Zurück zum Zitat A.M. Scott, F.T. Lee, N. Tebbutt, R. Herbertson, S.S. Gill, Z. Liu, E. Skrinos, C. Murone, T.H. Saunder, B. Chappell, A.T. Papenfuss, A.M. Poon, W. Hopkins, F.E. Smyth, D. MacGregor, L.M. Cher, A.A. Jungbluth, G. Ritter, M.W. Brechbiel, R. Murphy, A.W. Burgess, E.W. Hoffman, T.G. Johns, L.J. Old, A phase I clinical trial with monoclonal antibody ch806 targeting transitional state and mutant epidermal growth factor receptors. Proc. Natl. Acad. Sci. U. S. A. 104, 4071–4076 (2007)CrossRefPubMedPubMedCentral A.M. Scott, F.T. Lee, N. Tebbutt, R. Herbertson, S.S. Gill, Z. Liu, E. Skrinos, C. Murone, T.H. Saunder, B. Chappell, A.T. Papenfuss, A.M. Poon, W. Hopkins, F.E. Smyth, D. MacGregor, L.M. Cher, A.A. Jungbluth, G. Ritter, M.W. Brechbiel, R. Murphy, A.W. Burgess, E.W. Hoffman, T.G. Johns, L.J. Old, A phase I clinical trial with monoclonal antibody ch806 targeting transitional state and mutant epidermal growth factor receptors. Proc. Natl. Acad. Sci. U. S. A. 104, 4071–4076 (2007)CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat M. Sudo, S. Mori, V. Madan, H. Yang, G. Leong, H.P. Koeffler, Short-hairpin RNA library: identification of therapeutic partners for gefitinib-resistant non-small cell lung cancer. Oncotarget 6, 814–824 (2015)CrossRefPubMed M. Sudo, S. Mori, V. Madan, H. Yang, G. Leong, H.P. Koeffler, Short-hairpin RNA library: identification of therapeutic partners for gefitinib-resistant non-small cell lung cancer. Oncotarget 6, 814–824 (2015)CrossRefPubMed
37.
Zurück zum Zitat G.C. Trudel, A. Herscovics, P.C. Holland, Inhibition of myoblast fusion by bromoconduritol. Biochem. Cell Biol. 66, 1119–1125 (1988)CrossRefPubMed G.C. Trudel, A. Herscovics, P.C. Holland, Inhibition of myoblast fusion by bromoconduritol. Biochem. Cell Biol. 66, 1119–1125 (1988)CrossRefPubMed
38.
Zurück zum Zitat K.T. Yeo, T.K. Yeo, K. Olden, Bromoconduritol treatment delays intracellular transport of secretory glycoproteins in human hepatoma cell cultures. Biochem. Biophys. Res. Commun. 161, 1013–1019 (1989)CrossRefPubMed K.T. Yeo, T.K. Yeo, K. Olden, Bromoconduritol treatment delays intracellular transport of secretory glycoproteins in human hepatoma cell cultures. Biochem. Biophys. Res. Commun. 161, 1013–1019 (1989)CrossRefPubMed
39.
Zurück zum Zitat G. Pulverer, J. Beuth, H.L. Ko, A. Yassin, Y. Ohshima, K. Roszkowski, G. Uhlenbruck, Glycoprotein modifications of sarcoma L-1 tumor cells by tunicamycin, swainsonine, bromoconduritol or 1-desoxynojirimycin treatment inhibits their metastatic lung colonization in Balb/c-mice. J. Cancer Res. Clin. Oncol. 114, 217–220 (1988)CrossRefPubMed G. Pulverer, J. Beuth, H.L. Ko, A. Yassin, Y. Ohshima, K. Roszkowski, G. Uhlenbruck, Glycoprotein modifications of sarcoma L-1 tumor cells by tunicamycin, swainsonine, bromoconduritol or 1-desoxynojirimycin treatment inhibits their metastatic lung colonization in Balb/c-mice. J. Cancer Res. Clin. Oncol. 114, 217–220 (1988)CrossRefPubMed
40.
Zurück zum Zitat S. Li, L. Wang, Y. Hu, R. Sheng, Autophagy regulators as potential cancer therapeutic agents: a review. Curr. Top. Med. Chem. 15, 720–744 (2015)CrossRefPubMed S. Li, L. Wang, Y. Hu, R. Sheng, Autophagy regulators as potential cancer therapeutic agents: a review. Curr. Top. Med. Chem. 15, 720–744 (2015)CrossRefPubMed
41.
Zurück zum Zitat V. Kaminskyy, B. Zhivotovsky, Proteases in autophagy. Biochim. Biophys. Acta 1824, 44–50 (2012)CrossRefPubMed V. Kaminskyy, B. Zhivotovsky, Proteases in autophagy. Biochim. Biophys. Acta 1824, 44–50 (2012)CrossRefPubMed
42.
Zurück zum Zitat Y.P. Yang, L.F. Hu, H.F. Zheng, C.J. Mao, W.D. Hu, K.P. Xiong, F. Wang, C.F. Liu, Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol. Sin. 34, 625–635 (2013)CrossRefPubMedPubMedCentral Y.P. Yang, L.F. Hu, H.F. Zheng, C.J. Mao, W.D. Hu, K.P. Xiong, F. Wang, C.F. Liu, Application and interpretation of current autophagy inhibitors and activators. Acta Pharmacol. Sin. 34, 625–635 (2013)CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat E. Kim, A. Giese, W. Deppert, Wild-type p53 in cancer cells: when a guardian turns into a blackguard. Biochem. Pharmacol. 77, 11–20 (2009)CrossRefPubMed E. Kim, A. Giese, W. Deppert, Wild-type p53 in cancer cells: when a guardian turns into a blackguard. Biochem. Pharmacol. 77, 11–20 (2009)CrossRefPubMed
44.
45.
Zurück zum Zitat D. Crighton, S. Wilkinson, J. O'Prey, N. Syed, P. Smith, P.R. Harrison, M. Gasco, O. Garrone, T. Crook, K.M. Ryan, DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 126, 121–134 (2006)CrossRefPubMed D. Crighton, S. Wilkinson, J. O'Prey, N. Syed, P. Smith, P.R. Harrison, M. Gasco, O. Garrone, T. Crook, K.M. Ryan, DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 126, 121–134 (2006)CrossRefPubMed
46.
Zurück zum Zitat X. Liu, PARP inhibition as a prototype for synthetic lethal screens. Methods Mol. Biol. 986, 123–137 (2013)CrossRefPubMed X. Liu, PARP inhibition as a prototype for synthetic lethal screens. Methods Mol. Biol. 986, 123–137 (2013)CrossRefPubMed
Metadaten
Titel
Glucosidase II beta subunit (GluIIβ) plays a role in autophagy and apoptosis regulation in lung carcinoma cells in a p53-dependent manner
verfasst von
Worapong Khaodee
Nichanan Inboot
Suruk Udomsom
Warunee Kumsaiyai
Ratchada Cressey
Publikationsdatum
19.09.2017
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 6/2017
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-017-0349-1

Weitere Artikel der Ausgabe 6/2017

Cellular Oncology 6/2017 Zur Ausgabe

Neu im Fachgebiet Pathologie

Molekularpathologische Untersuchungen im Wandel der Zeit

Open Access Biomarker Leitthema

Um auch an kleinen Gewebeproben zuverlässige und reproduzierbare Ergebnisse zu gewährleisten ist eine strenge Qualitätskontrolle in jedem Schritt des Arbeitsablaufs erforderlich. Eine nicht ordnungsgemäße Prüfung oder Behandlung des …

Vergleichende Pathologie in der onkologischen Forschung

Pathologie Leitthema

Die vergleichende experimentelle Pathologie („comparative experimental pathology“) ist ein Fachbereich an der Schnittstelle von Human- und Veterinärmedizin. Sie widmet sich der vergleichenden Erforschung von Gemeinsamkeiten und Unterschieden von …

Gastrointestinale Stromatumoren

Open Access GIST CME-Artikel

Gastrointestinale Stromatumoren (GIST) stellen seit über 20 Jahren ein Paradigma für die zielgerichtete Therapie mit Tyrosinkinaseinhibitoren dar. Eine elementare Voraussetzung für eine mögliche neoadjuvante oder adjuvante Behandlung bei …

Personalisierte Medizin in der Onkologie

Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender …