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

01-12-2020 | Ovarian Cancer | Review

Insight into the role of p62 in the cisplatin resistant mechanisms of ovarian cancer

Authors: Xiao-Yu Yan, Xian-Zhi Qu, Long Xu, Si-Hang Yu, Rui Tian, Xin-Ru Zhong, Lian-Kun Sun, Jing Su

Published in: Cancer Cell International | Issue 1/2020

Login to get access

Abstract

Cisplatin is a platinum-based first-line drug for treating ovarian cancer. However, chemotherapy tolerance has limited the efficacy of cisplatin for ovarian cancer patients. Research has demonstrated that cisplatin causes changes in cell survival and death signaling pathways through its interaction with macromolecules and organelles, which indicates that investigation into the DNA off-target effects of cisplatin may provide critical insights into the mechanisms underlying drug resistance. The multifunctional protein p62 works as a signaling hub in the regulation of pro-survival transcriptional factors NF-κB and Nrf2 and connects autophagy and apoptotic signals, which play important roles in maintaining cell homeostasis. In this review, we discuss the role of p62 in cisplatin resistance by exploring p62-associated signaling pathways based on current studies and our work. Insights into these resistance mechanisms may lead to more effective therapeutic strategies for ovarian cancer by targeting p62.
Literature
1.
2.
go back to reference Rosenberg B, Vancamp L, Krigas T. Inhibition of cell division in Escherichia Coli by electrolysis products from a platinum electrode. Nature. 1965;205:698–9.PubMedCrossRef Rosenberg B, Vancamp L, Krigas T. Inhibition of cell division in Escherichia Coli by electrolysis products from a platinum electrode. Nature. 1965;205:698–9.PubMedCrossRef
3.
go back to reference Peng H, Jin H, Zhuo H, Huang H. Enhanced antitumor efficacy of cisplatin for treating ovarian cancer in vitro and in vivo via transferrin binding. Oncotarget. 2017;8(28):45597–611.PubMedPubMedCentralCrossRef Peng H, Jin H, Zhuo H, Huang H. Enhanced antitumor efficacy of cisplatin for treating ovarian cancer in vitro and in vivo via transferrin binding. Oncotarget. 2017;8(28):45597–611.PubMedPubMedCentralCrossRef
4.
go back to reference Pfisterer J, Vergote I, Du Bois A, Eisenhauer E, Ago O, Ncic CTG, Eortc GCG. Combination therapy with gemcitabine and carboplatin in recurrent ovarian cancer. Int J Gynecol Cancer. 2005;15(Suppl 1):36–41.PubMedCrossRef Pfisterer J, Vergote I, Du Bois A, Eisenhauer E, Ago O, Ncic CTG, Eortc GCG. Combination therapy with gemcitabine and carboplatin in recurrent ovarian cancer. Int J Gynecol Cancer. 2005;15(Suppl 1):36–41.PubMedCrossRef
5.
go back to reference Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, Mannel RS, DeGeest K, Hartenbach EM, Baergen R, et al. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2003;21(17):3194–200.PubMedCrossRef Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, Mannel RS, DeGeest K, Hartenbach EM, Baergen R, et al. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2003;21(17):3194–200.PubMedCrossRef
6.
go back to reference Aebi S, Castiglione M, Group EGW. Newly and relapsed epithelial ovarian carcinoma: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2009;20(Suppl 4):21–3. Aebi S, Castiglione M, Group EGW. Newly and relapsed epithelial ovarian carcinoma: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2009;20(Suppl 4):21–3.
7.
go back to reference McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, Look KY, Clarke-Pearson DL, Davidson M. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N Engl J Med. 1996;334(1):1–6.PubMedCrossRef McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, Look KY, Clarke-Pearson DL, Davidson M. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N Engl J Med. 1996;334(1):1–6.PubMedCrossRef
8.
go back to reference Gonzalez-Martin A, Pothuri B, Vergote I, DePont Christensen R, Graybill W, Mirza MR, McCormick C, Lorusso D, Hoskins P, Freyer G, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2019;381(25):2391–402.PubMedCrossRef Gonzalez-Martin A, Pothuri B, Vergote I, DePont Christensen R, Graybill W, Mirza MR, McCormick C, Lorusso D, Hoskins P, Freyer G, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2019;381(25):2391–402.PubMedCrossRef
9.
go back to reference Bible KC, Peethambaram PP, Oberg AL, Maples W, Groteluschen DL, Boente M, Burton JK, Gomez Dahl LC, Tibodeau JD, Isham CR, et al. A phase 2 trial of flavopiridol (Alvocidib) and cisplatin in platin-resistant ovarian and primary peritoneal carcinoma: MC0261. Gynecol Oncol. 2012;127(1):55–62.PubMedPubMedCentralCrossRef Bible KC, Peethambaram PP, Oberg AL, Maples W, Groteluschen DL, Boente M, Burton JK, Gomez Dahl LC, Tibodeau JD, Isham CR, et al. A phase 2 trial of flavopiridol (Alvocidib) and cisplatin in platin-resistant ovarian and primary peritoneal carcinoma: MC0261. Gynecol Oncol. 2012;127(1):55–62.PubMedPubMedCentralCrossRef
10.
go back to reference Melnikov SV, Soll D, Steitz TA, Polikanov YS. Insights into RNA binding by the anticancer drug cisplatin from the crystal structure of cisplatin-modified ribosome. Nucleic Acids Res. 2016;44(10):4978–87.PubMedPubMedCentralCrossRef Melnikov SV, Soll D, Steitz TA, Polikanov YS. Insights into RNA binding by the anticancer drug cisplatin from the crystal structure of cisplatin-modified ribosome. Nucleic Acids Res. 2016;44(10):4978–87.PubMedPubMedCentralCrossRef
11.
go back to reference Russo Krauss I, Ferraro G, Merlino A. Cisplatin-protein interactions: unexpected drug binding to N-terminal amine and lysine side chains. Inorg Chem. 2016;55(16):7814–6.PubMedCrossRef Russo Krauss I, Ferraro G, Merlino A. Cisplatin-protein interactions: unexpected drug binding to N-terminal amine and lysine side chains. Inorg Chem. 2016;55(16):7814–6.PubMedCrossRef
12.
go back to reference Gatti L, Cassinelli G, Zaffaroni N, Lanzi C, Perego P. New mechanisms for old drugs: insights into DNA-unrelated effects of platinum compounds and drug resistance determinants. Drug Resist Updat. 2015;20:1–11.PubMedCrossRef Gatti L, Cassinelli G, Zaffaroni N, Lanzi C, Perego P. New mechanisms for old drugs: insights into DNA-unrelated effects of platinum compounds and drug resistance determinants. Drug Resist Updat. 2015;20:1–11.PubMedCrossRef
13.
go back to reference Galluzzi L, Vitale I, Michels J, Brenner C, Szabadkai G, Harel-Bellan A, Castedo M, Kroemer G. Systems biology of cisplatin resistance: past, present and future. Cell Death Dis. 2014;5:e1257.PubMedPubMedCentralCrossRef Galluzzi L, Vitale I, Michels J, Brenner C, Szabadkai G, Harel-Bellan A, Castedo M, Kroemer G. Systems biology of cisplatin resistance: past, present and future. Cell Death Dis. 2014;5:e1257.PubMedPubMedCentralCrossRef
14.
go back to reference Thakur B, Ray P. Cisplatin triggers cancer stem cell enrichment in platinum-resistant cells through NF-kappaB-TNFalpha-PIK3CA loop. J Exp Clin Cancer Res. 2017;36(1):164.PubMedPubMedCentralCrossRef Thakur B, Ray P. Cisplatin triggers cancer stem cell enrichment in platinum-resistant cells through NF-kappaB-TNFalpha-PIK3CA loop. J Exp Clin Cancer Res. 2017;36(1):164.PubMedPubMedCentralCrossRef
15.
go back to reference Shen DW, Pouliot LM, Hall MD, Gottesman MM. Cisplatin resistance: a cellular self-defense mechanism resulting from multiple epigenetic and genetic changes. Pharmacol Rev. 2012;64(3):706–21.PubMedPubMedCentralCrossRef Shen DW, Pouliot LM, Hall MD, Gottesman MM. Cisplatin resistance: a cellular self-defense mechanism resulting from multiple epigenetic and genetic changes. Pharmacol Rev. 2012;64(3):706–21.PubMedPubMedCentralCrossRef
16.
go back to reference Duran A, Amanchy R, Linares JF, Joshi J, Abu-Baker S, Porollo A, Hansen M, Moscat J, Diaz-Meco MT. p62 is a key regulator of nutrient sensing in the mTORC1 pathway. Mol Cell. 2011;44(1):134–46.PubMedPubMedCentralCrossRef Duran A, Amanchy R, Linares JF, Joshi J, Abu-Baker S, Porollo A, Hansen M, Moscat J, Diaz-Meco MT. p62 is a key regulator of nutrient sensing in the mTORC1 pathway. Mol Cell. 2011;44(1):134–46.PubMedPubMedCentralCrossRef
18.
go back to reference Reina-Campos M, Shelton PM, Diaz-Meco MT, Moscat J. Metabolic reprogramming of the tumor microenvironment by p62 and its partners. Biochem Biophys Acta. 2018;1870(1):88–95. Reina-Campos M, Shelton PM, Diaz-Meco MT, Moscat J. Metabolic reprogramming of the tumor microenvironment by p62 and its partners. Biochem Biophys Acta. 2018;1870(1):88–95.
19.
go back to reference Ju LL, Zhao CY, Ye KF, Yang H, Zhang J. Expression and clinical implication of Beclin1, HMGB1, p62, survivin, BRCA1 and ERCC1 in epithelial ovarian tumor tissues. Eur Rev Med Pharmacol Sci. 2016;20(10):1993–2003.PubMed Ju LL, Zhao CY, Ye KF, Yang H, Zhang J. Expression and clinical implication of Beclin1, HMGB1, p62, survivin, BRCA1 and ERCC1 in epithelial ovarian tumor tissues. Eur Rev Med Pharmacol Sci. 2016;20(10):1993–2003.PubMed
20.
go back to reference Sanchez-Martin P, Saito T, Komatsu M. p62/SQSTM1: ‘Jack of all trades’ in health and cancer. FEBS J. 2019;286(1):8–23.PubMedCrossRef Sanchez-Martin P, Saito T, Komatsu M. p62/SQSTM1: ‘Jack of all trades’ in health and cancer. FEBS J. 2019;286(1):8–23.PubMedCrossRef
21.
go back to reference Lamark T, Svenning S, Johansen T. Regulation of selective autophagy: the p62/SQSTM1 paradigm. Essays Biochem. 2017;61(6):609–24.PubMedCrossRef Lamark T, Svenning S, Johansen T. Regulation of selective autophagy: the p62/SQSTM1 paradigm. Essays Biochem. 2017;61(6):609–24.PubMedCrossRef
22.
go back to reference He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Ann Rev Genet. 2009;43:67–93.PubMedCrossRef He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Ann Rev Genet. 2009;43:67–93.PubMedCrossRef
23.
go back to reference Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath J, Gewirtz DA, Karantza V, et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015;34(7):856–80.PubMedPubMedCentralCrossRef Galluzzi L, Pietrocola F, Bravo-San Pedro JM, Amaravadi RK, Baehrecke EH, Cecconi F, Codogno P, Debnath J, Gewirtz DA, Karantza V, et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015;34(7):856–80.PubMedPubMedCentralCrossRef
24.
go back to reference Zhang XY, Zhang M, Cong Q, Zhang MX, Zhang MY, Lu YY, Xu CJ. Hexokinase 2 confers resistance to cisplatin in ovarian cancer cells by enhancing cisplatin-induced autophagy. Int J Biochem Cell Biol. 2018;95:9–16.PubMedCrossRef Zhang XY, Zhang M, Cong Q, Zhang MX, Zhang MY, Lu YY, Xu CJ. Hexokinase 2 confers resistance to cisplatin in ovarian cancer cells by enhancing cisplatin-induced autophagy. Int J Biochem Cell Biol. 2018;95:9–16.PubMedCrossRef
25.
go back to reference Periyasamy-Thandavan S, Jiang M, Schoenlein P, Dong Z. Autophagy: molecular machinery, regulation, and implications for renal pathophysiology. Am J Physiol Renal Physiol. 2009;297(2):F244–56.PubMedPubMedCentralCrossRef Periyasamy-Thandavan S, Jiang M, Schoenlein P, Dong Z. Autophagy: molecular machinery, regulation, and implications for renal pathophysiology. Am J Physiol Renal Physiol. 2009;297(2):F244–56.PubMedPubMedCentralCrossRef
26.
go back to reference Matsumoto G, Wada K, Okuno M, Kurosawa M, Nukina N. Serine 403 phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of ubiquitinated proteins. Mol Cell. 2011;44(2):279–89.PubMedCrossRef Matsumoto G, Wada K, Okuno M, Kurosawa M, Nukina N. Serine 403 phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of ubiquitinated proteins. Mol Cell. 2011;44(2):279–89.PubMedCrossRef
27.
go back to reference Geetha T, Wooten MW. Structure and functional properties of the ubiquitin binding protein p62. FEBS Lett. 2002;512(1–3):19–24.PubMedCrossRef Geetha T, Wooten MW. Structure and functional properties of the ubiquitin binding protein p62. FEBS Lett. 2002;512(1–3):19–24.PubMedCrossRef
29.
go back to reference Wurzer B, Zaffagnini G, Fracchiolla D, Turco E, Abert C, Romanov J, Martens S. Oligomerization of p62 allows for selection of ubiquitinated cargo and isolation membrane during selective autophagy. Elife. 2015;4:e08941.PubMedPubMedCentralCrossRef Wurzer B, Zaffagnini G, Fracchiolla D, Turco E, Abert C, Romanov J, Martens S. Oligomerization of p62 allows for selection of ubiquitinated cargo and isolation membrane during selective autophagy. Elife. 2015;4:e08941.PubMedPubMedCentralCrossRef
30.
go back to reference Ma L, Xu Y, Su J, Yu H, Kang J, Li H, Li X, Xie Q, Yu C, Sun L, et al. Autophagic flux promotes cisplatin resistance in human ovarian carcinoma cells through ATP-mediated lysosomal function. Int J Oncol. 2015;47(5):1890–900.PubMedCrossRef Ma L, Xu Y, Su J, Yu H, Kang J, Li H, Li X, Xie Q, Yu C, Sun L, et al. Autophagic flux promotes cisplatin resistance in human ovarian carcinoma cells through ATP-mediated lysosomal function. Int J Oncol. 2015;47(5):1890–900.PubMedCrossRef
31.
go back to reference Yan XY, Zhang Y, Zhang JJ, Zhang LC, Liu YN, Wu Y, Xue YN, Lu SY, Su J, Sun LK. p62/SQSTM1 as an oncotarget mediates cisplatin resistance through activating RIP1-NF-kappaB pathway in human ovarian cancer cells. Cancer Sci. 2017;108(7):1405–13.PubMedPubMedCentralCrossRef Yan XY, Zhang Y, Zhang JJ, Zhang LC, Liu YN, Wu Y, Xue YN, Lu SY, Su J, Sun LK. p62/SQSTM1 as an oncotarget mediates cisplatin resistance through activating RIP1-NF-kappaB pathway in human ovarian cancer cells. Cancer Sci. 2017;108(7):1405–13.PubMedPubMedCentralCrossRef
32.
go back to reference Cha-Molstad H, Yu JE, Feng Z, Lee SH, Kim JG, Yang P, Han B, Sung KW, Yoo YD, Hwang J, et al. p62/SQSTM1/Sequestosome-1 is an N-recognin of the N-end rule pathway which modulates autophagosome biogenesis. Nat Commun. 2017;8(1):102.PubMedPubMedCentralCrossRef Cha-Molstad H, Yu JE, Feng Z, Lee SH, Kim JG, Yang P, Han B, Sung KW, Yoo YD, Hwang J, et al. p62/SQSTM1/Sequestosome-1 is an N-recognin of the N-end rule pathway which modulates autophagosome biogenesis. Nat Commun. 2017;8(1):102.PubMedPubMedCentralCrossRef
33.
go back to reference Yan XY, Zhong XR, Yu SH, Zhang LC, Liu YN, Zhang Y, Sun LK, Su J. p62 aggregates mediated Caspase 8 activation is responsible for progression of ovarian cancer. J Cell Mol Med. 2019;23(6):4030–42.PubMedPubMedCentralCrossRef Yan XY, Zhong XR, Yu SH, Zhang LC, Liu YN, Zhang Y, Sun LK, Su J. p62 aggregates mediated Caspase 8 activation is responsible for progression of ovarian cancer. J Cell Mol Med. 2019;23(6):4030–42.PubMedPubMedCentralCrossRef
34.
go back to reference Lee Y, Chou TF, Pittman SK, Keith AL, Razani B, Weihl CC. Keap1/Cullin3 modulates p62/SQSTM1 activity via UBA domain ubiquitination. Cell Rep. 2017;20(8):1994.PubMedCrossRef Lee Y, Chou TF, Pittman SK, Keith AL, Razani B, Weihl CC. Keap1/Cullin3 modulates p62/SQSTM1 activity via UBA domain ubiquitination. Cell Rep. 2017;20(8):1994.PubMedCrossRef
35.
go back to reference Carroll B, Otten EG, Manni D, Stefanatos R, Menzies FM, Smith GR, Jurk D, Kenneth N, Wilkinson S, Passos JF, et al. Oxidation of SQSTM1/p62 mediates the link between redox state and protein homeostasis. Nat Commun. 2018;9(1):256.PubMedPubMedCentralCrossRef Carroll B, Otten EG, Manni D, Stefanatos R, Menzies FM, Smith GR, Jurk D, Kenneth N, Wilkinson S, Passos JF, et al. Oxidation of SQSTM1/p62 mediates the link between redox state and protein homeostasis. Nat Commun. 2018;9(1):256.PubMedPubMedCentralCrossRef
36.
go back to reference Kingnate C, Charoenkwan K, Kumfu S, Chattipakorn N, Chattipakorn SC. Possible roles of mitochondrial dynamics and the effects of pharmacological interventions in chemoresistant ovarian cancer. EBioMedicine. 2018;34:256–66.PubMedPubMedCentralCrossRef Kingnate C, Charoenkwan K, Kumfu S, Chattipakorn N, Chattipakorn SC. Possible roles of mitochondrial dynamics and the effects of pharmacological interventions in chemoresistant ovarian cancer. EBioMedicine. 2018;34:256–66.PubMedPubMedCentralCrossRef
37.
39.
go back to reference Sekine S, Kanamaru Y, Koike M, Nishihara A, Okada M, Kinoshita H, Kamiyama M, Maruyama J, Uchiyama Y, Ishihara N, et al. Rhomboid protease PARL mediates the mitochondrial membrane potential loss-induced cleavage of PGAM5. J Biol Chem. 2012;287(41):34635–45.PubMedPubMedCentralCrossRef Sekine S, Kanamaru Y, Koike M, Nishihara A, Okada M, Kinoshita H, Kamiyama M, Maruyama J, Uchiyama Y, Ishihara N, et al. Rhomboid protease PARL mediates the mitochondrial membrane potential loss-induced cleavage of PGAM5. J Biol Chem. 2012;287(41):34635–45.PubMedPubMedCentralCrossRef
40.
go back to reference Seibenhener ML, Du Y, Diaz-Meco MT, Moscat J, Wooten MC, Wooten MW. A role for sequestosome 1/p62 in mitochondrial dynamics, import and genome integrity. Biochim Biophys Acta. 2013;1833(3):452–9.PubMedCrossRef Seibenhener ML, Du Y, Diaz-Meco MT, Moscat J, Wooten MC, Wooten MW. A role for sequestosome 1/p62 in mitochondrial dynamics, import and genome integrity. Biochim Biophys Acta. 2013;1833(3):452–9.PubMedCrossRef
41.
go back to reference Cullen KJ, Yang Z, Schumaker L, Guo Z. Mitochondria as a critical target of the chemotheraputic agent cisplatin in head and neck cancer. J Bioenergy Biomembr. 2007;39(1):43–50.CrossRef Cullen KJ, Yang Z, Schumaker L, Guo Z. Mitochondria as a critical target of the chemotheraputic agent cisplatin in head and neck cancer. J Bioenergy Biomembr. 2007;39(1):43–50.CrossRef
43.
go back to reference Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, Springer W. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010;12(2):119–31.PubMedCrossRef Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, Springer W. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010;12(2):119–31.PubMedCrossRef
44.
go back to reference Yang W, Liu L, Li C, Luo N, Chen R, Li L, Yu F, Cheng Z. TRIM52 plays an oncogenic role in ovarian cancer associated with NF-kB pathway. Cell Death Dis. 2018;9(9):908.PubMedPubMedCentralCrossRef Yang W, Liu L, Li C, Luo N, Chen R, Li L, Yu F, Cheng Z. TRIM52 plays an oncogenic role in ovarian cancer associated with NF-kB pathway. Cell Death Dis. 2018;9(9):908.PubMedPubMedCentralCrossRef
45.
go back to reference Mabuchi S, Ohmichi M, Nishio Y, Hayasaka T, Kimura A, Ohta T, Saito M, Kawagoe J, Takahashi K, Yada-Hashimoto N, et al. Inhibition of NFkappaB increases the efficacy of cisplatin in in vitro and in vivo ovarian cancer models. J Biol Chem. 2004;279(22):23477–85.PubMedCrossRef Mabuchi S, Ohmichi M, Nishio Y, Hayasaka T, Kimura A, Ohta T, Saito M, Kawagoe J, Takahashi K, Yada-Hashimoto N, et al. Inhibition of NFkappaB increases the efficacy of cisplatin in in vitro and in vivo ovarian cancer models. J Biol Chem. 2004;279(22):23477–85.PubMedCrossRef
46.
go back to reference Mabuchi S, Ohmichi M, Nishio Y, Hayasaka T, Kimura A, Ohta T, Kawagoe J, Takahashi K, Yada-Hashimoto N, Seino-Noda H, et al. Inhibition of inhibitor of nuclear factor-kappaB phosphorylation increases the efficacy of paclitaxel in in vitro and in vivo ovarian cancer models. Clin Cancer Res. 2004;10(22):7645–54.PubMedCrossRef Mabuchi S, Ohmichi M, Nishio Y, Hayasaka T, Kimura A, Ohta T, Kawagoe J, Takahashi K, Yada-Hashimoto N, Seino-Noda H, et al. Inhibition of inhibitor of nuclear factor-kappaB phosphorylation increases the efficacy of paclitaxel in in vitro and in vivo ovarian cancer models. Clin Cancer Res. 2004;10(22):7645–54.PubMedCrossRef
49.
go back to reference Meads MB, Li ZW, Dalton WS. A novel TNF receptor-associated factor 6 binding domain mediates NF-kappa B signaling by the common cytokine receptor beta subunit. J Immunol. 2010;185(3):1606–15.PubMedCrossRef Meads MB, Li ZW, Dalton WS. A novel TNF receptor-associated factor 6 binding domain mediates NF-kappa B signaling by the common cytokine receptor beta subunit. J Immunol. 2010;185(3):1606–15.PubMedCrossRef
51.
go back to reference Duran A, Linares JF, Galvez AS, Wikenheiser K, Flores JM, Diaz-Meco MT, Moscat J. The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. Cancer Cell. 2008;13(4):343–54.PubMedCrossRef Duran A, Linares JF, Galvez AS, Wikenheiser K, Flores JM, Diaz-Meco MT, Moscat J. The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. Cancer Cell. 2008;13(4):343–54.PubMedCrossRef
52.
go back to reference Sanz L, Diaz-Meco MT, Nakano H, Moscat J. The atypical PKC-interacting protein p62 channels NF-kappaB activation by the IL-1-TRAF6 pathway. EMBO J. 2000;19(7):1576–86.PubMedPubMedCentralCrossRef Sanz L, Diaz-Meco MT, Nakano H, Moscat J. The atypical PKC-interacting protein p62 channels NF-kappaB activation by the IL-1-TRAF6 pathway. EMBO J. 2000;19(7):1576–86.PubMedPubMedCentralCrossRef
53.
go back to reference Wooten MW, Geetha T, Seibenhener ML, Babu JR, Diaz-Meco MT, Moscat J. The p62 scaffold regulates nerve growth factor-induced NF-kappaB activation by influencing TRAF6 polyubiquitination. J Biol Chem. 2005;280(42):35625–9.PubMedCrossRef Wooten MW, Geetha T, Seibenhener ML, Babu JR, Diaz-Meco MT, Moscat J. The p62 scaffold regulates nerve growth factor-induced NF-kappaB activation by influencing TRAF6 polyubiquitination. J Biol Chem. 2005;280(42):35625–9.PubMedCrossRef
54.
go back to reference Ofengeim D, Yuan J. Regulation of RIP1 kinase signalling at the crossroads of inflammation and cell death. Nat Rev Mol Cell Biol. 2013;14(11):727–36.PubMedCrossRef Ofengeim D, Yuan J. Regulation of RIP1 kinase signalling at the crossroads of inflammation and cell death. Nat Rev Mol Cell Biol. 2013;14(11):727–36.PubMedCrossRef
55.
go back to reference Liu XY, Lai F, Yan XG, Jiang CC, Guo ST, Wang CY, Croft A, Tseng HY, Wilmott JS, Scolyer RA, et al. RIP1 kinase is an oncogenic driver in melanoma. Cancer Res. 2015;75(8):1736–48.PubMedCrossRef Liu XY, Lai F, Yan XG, Jiang CC, Guo ST, Wang CY, Croft A, Tseng HY, Wilmott JS, Scolyer RA, et al. RIP1 kinase is an oncogenic driver in melanoma. Cancer Res. 2015;75(8):1736–48.PubMedCrossRef
56.
go back to reference Li H, Kobayashi M, Blonska M, You Y, Lin X. Ubiquitination of RIP is required for tumor necrosis factor alpha-induced NF-kappaB activation. J Biol Chem. 2006;281(19):13636–43.PubMedCrossRef Li H, Kobayashi M, Blonska M, You Y, Lin X. Ubiquitination of RIP is required for tumor necrosis factor alpha-induced NF-kappaB activation. J Biol Chem. 2006;281(19):13636–43.PubMedCrossRef
57.
go back to reference Emmerich CH, Ordureau A, Strickson S, Arthur JS, Pedrioli PG, Komander D, Cohen P. Activation of the canonical IKK complex by K63/M1-linked hybrid ubiquitin chains. Proc Natl Acad Sci USA. 2013;110(38):15247–52.PubMedCrossRefPubMedCentral Emmerich CH, Ordureau A, Strickson S, Arthur JS, Pedrioli PG, Komander D, Cohen P. Activation of the canonical IKK complex by K63/M1-linked hybrid ubiquitin chains. Proc Natl Acad Sci USA. 2013;110(38):15247–52.PubMedCrossRefPubMedCentral
58.
go back to reference Christofferson DE, Li Y, Hitomi J, Zhou W, Upperman C, Zhu H, Gerber SA, Gygi S, Yuan J. A novel role for RIP1 kinase in mediating TNFalpha production. Cell Death Dis. 2012;3:e320.PubMedPubMedCentralCrossRef Christofferson DE, Li Y, Hitomi J, Zhou W, Upperman C, Zhu H, Gerber SA, Gygi S, Yuan J. A novel role for RIP1 kinase in mediating TNFalpha production. Cell Death Dis. 2012;3:e320.PubMedPubMedCentralCrossRef
59.
go back to reference Sanz L, Sanchez P, Lallena MJ, Diaz-Meco MT, Moscat J. The interaction of p62 with RIP links the atypical PKCs to NF-kappaB activation. EMBO J. 1999;18(11):3044–53.PubMedPubMedCentralCrossRef Sanz L, Sanchez P, Lallena MJ, Diaz-Meco MT, Moscat J. The interaction of p62 with RIP links the atypical PKCs to NF-kappaB activation. EMBO J. 1999;18(11):3044–53.PubMedPubMedCentralCrossRef
60.
go back to reference Cai PC, Shi L, Liu VW, Tang HW, Liu IJ, Leung TH, Chan KK, Yam JW, Yao KM, Ngan HY, et al. Elevated TAK1 augments tumor growth and metastatic capacities of ovarian cancer cells through activation of NF-kappaB signaling. Oncotarget. 2014;5(17):7549–62.PubMedPubMedCentralCrossRef Cai PC, Shi L, Liu VW, Tang HW, Liu IJ, Leung TH, Chan KK, Yam JW, Yao KM, Ngan HY, et al. Elevated TAK1 augments tumor growth and metastatic capacities of ovarian cancer cells through activation of NF-kappaB signaling. Oncotarget. 2014;5(17):7549–62.PubMedPubMedCentralCrossRef
61.
go back to reference Wangpaichitr M, Wu C, Li YY, Nguyen DJM, Kandemir H, Shah S, Chen S, Feun LG, Prince JS, Kuo MT, et al. Exploiting ROS and metabolic differences to kill cisplatin resistant lung cancer. Oncotarget. 2017;8(30):49275–92.PubMedPubMedCentralCrossRef Wangpaichitr M, Wu C, Li YY, Nguyen DJM, Kandemir H, Shah S, Chen S, Feun LG, Prince JS, Kuo MT, et al. Exploiting ROS and metabolic differences to kill cisplatin resistant lung cancer. Oncotarget. 2017;8(30):49275–92.PubMedPubMedCentralCrossRef
62.
go back to reference Roh JL, Jang H, Kim EH, Shin D. Targeting of the glutathione, thioredoxin, and Nrf2 antioxidant systems in head and neck cancer. Antioxid Redox Signal. 2017;27(2):106–14.PubMedCrossRef Roh JL, Jang H, Kim EH, Shin D. Targeting of the glutathione, thioredoxin, and Nrf2 antioxidant systems in head and neck cancer. Antioxid Redox Signal. 2017;27(2):106–14.PubMedCrossRef
63.
go back to reference Kansanen E, Kuosmanen SM, Leinonen H, Levonen AL. The Keap1-Nrf2 pathway: mechanisms of activation and dysregulation in cancer. Redox Biol. 2013;1:45–9.PubMedPubMedCentralCrossRef Kansanen E, Kuosmanen SM, Leinonen H, Levonen AL. The Keap1-Nrf2 pathway: mechanisms of activation and dysregulation in cancer. Redox Biol. 2013;1:45–9.PubMedPubMedCentralCrossRef
64.
go back to reference Ryoo IG, Kwak MK. Regulatory crosstalk between the oxidative stress-related transcription factor Nfe2l2/Nrf2 and mitochondria. Toxicol Appl Pharmacol. 2018;359:24–33.PubMedCrossRef Ryoo IG, Kwak MK. Regulatory crosstalk between the oxidative stress-related transcription factor Nfe2l2/Nrf2 and mitochondria. Toxicol Appl Pharmacol. 2018;359:24–33.PubMedCrossRef
65.
go back to reference Canet MJ, Merrell MD, Harder BG, Maher JM, Wu T, Lickteig AJ, Jackson JP, Zhang DD, Yamamoto M, Cherrington NJ. Identification of a functional antioxidant response element within the eighth intron of the human ABCC3 gene. Drug Metab Dispos. 2015;43(1):93–9.PubMedPubMedCentralCrossRef Canet MJ, Merrell MD, Harder BG, Maher JM, Wu T, Lickteig AJ, Jackson JP, Zhang DD, Yamamoto M, Cherrington NJ. Identification of a functional antioxidant response element within the eighth intron of the human ABCC3 gene. Drug Metab Dispos. 2015;43(1):93–9.PubMedPubMedCentralCrossRef
66.
go back to reference Bao L, Wu J, Dodson M, Rojo de la Vega EM, Ning Y, Zhang Z, Yao M, Zhang DD, Xu C, Yi X. ABCF2, an Nrf2 target gene, contributes to cisplatin resistance in ovarian cancer cells. Mol Carcinog. 2017;56(6):1543–53.PubMedPubMedCentralCrossRef Bao L, Wu J, Dodson M, Rojo de la Vega EM, Ning Y, Zhang Z, Yao M, Zhang DD, Xu C, Yi X. ABCF2, an Nrf2 target gene, contributes to cisplatin resistance in ovarian cancer cells. Mol Carcinog. 2017;56(6):1543–53.PubMedPubMedCentralCrossRef
67.
go back to reference Wu J, Bao L, Zhang Z, Yi X. Nrf2 induces cisplatin resistance via suppressing the iron export related gene SLC40A1 in ovarian cancer cells. Oncotarget. 2017;8(55):93502–15.PubMedPubMedCentralCrossRef Wu J, Bao L, Zhang Z, Yi X. Nrf2 induces cisplatin resistance via suppressing the iron export related gene SLC40A1 in ovarian cancer cells. Oncotarget. 2017;8(55):93502–15.PubMedPubMedCentralCrossRef
68.
go back to reference Jain A, Lamark T, Sjottem E, Larsen KB, Awuh JA, Overvatn A, McMahon M, Hayes JD, Johansen T. p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J Biol Chem. 2010;285(29):22576–91.PubMedPubMedCentralCrossRef Jain A, Lamark T, Sjottem E, Larsen KB, Awuh JA, Overvatn A, McMahon M, Hayes JD, Johansen T. p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J Biol Chem. 2010;285(29):22576–91.PubMedPubMedCentralCrossRef
69.
go back to reference Jena KK, Kolapalli SP, Mehto S, Nath P, Das B, Sahoo PK, Ahad A, Syed GH, Raghav SK, Senapati S, et al. TRIM16 controls assembly and degradation of protein aggregates by modulating the p62-NRF2 axis and autophagy. EMBO J. 2018;37(18):e98358.PubMedPubMedCentralCrossRef Jena KK, Kolapalli SP, Mehto S, Nath P, Das B, Sahoo PK, Ahad A, Syed GH, Raghav SK, Senapati S, et al. TRIM16 controls assembly and degradation of protein aggregates by modulating the p62-NRF2 axis and autophagy. EMBO J. 2018;37(18):e98358.PubMedPubMedCentralCrossRef
70.
71.
go back to reference Xia M, Yu H, Gu S, Xu Y, Su J, Li H, Kang J, Cui M. p62/SQSTM1 is involved in cisplatin resistance in human ovarian cancer cells via the Keap1-Nrf2-ARE system. Int J Oncol. 2014;45(6):2341–8.PubMedCrossRef Xia M, Yu H, Gu S, Xu Y, Su J, Li H, Kang J, Cui M. p62/SQSTM1 is involved in cisplatin resistance in human ovarian cancer cells via the Keap1-Nrf2-ARE system. Int J Oncol. 2014;45(6):2341–8.PubMedCrossRef
72.
go back to reference Stepkowski TM, Kruszewski MK. Molecular cross-talk between the NRF2/KEAP1 signaling pathway, autophagy, and apoptosis. Free Radic Biol Med. 2011;50(9):1186–95.PubMedCrossRef Stepkowski TM, Kruszewski MK. Molecular cross-talk between the NRF2/KEAP1 signaling pathway, autophagy, and apoptosis. Free Radic Biol Med. 2011;50(9):1186–95.PubMedCrossRef
73.
go back to reference O’Mealey GB, Plafker KS, Berry WL, Janknecht R, Chan JY, Plafker SM. A PGAM5-KEAP1-Nrf2 complex is required for stress-induced mitochondrial retrograde trafficking. J Cell Sci. 2017;130(20):3467–80.PubMedPubMedCentralCrossRef O’Mealey GB, Plafker KS, Berry WL, Janknecht R, Chan JY, Plafker SM. A PGAM5-KEAP1-Nrf2 complex is required for stress-induced mitochondrial retrograde trafficking. J Cell Sci. 2017;130(20):3467–80.PubMedPubMedCentralCrossRef
75.
go back to reference Koo GB, Morgan MJ, Lee DG, Kim WJ, Yoon JH, Koo JS, Kim SI, Kim SJ, Son MK, Hong SS, et al. Methylation-dependent loss of RIP3 expression in cancer represses programmed necrosis in response to chemotherapeutics. Cell Res. 2015;25(6):707–25.PubMedPubMedCentralCrossRef Koo GB, Morgan MJ, Lee DG, Kim WJ, Yoon JH, Koo JS, Kim SI, Kim SJ, Son MK, Hong SS, et al. Methylation-dependent loss of RIP3 expression in cancer represses programmed necrosis in response to chemotherapeutics. Cell Res. 2015;25(6):707–25.PubMedPubMedCentralCrossRef
76.
77.
go back to reference Hernandez L, Kim MK, Noonan AM, Sagher E, Kohlhammer H, Wright G, Lyle LT, Steeg PS, Anver M, Bowtell DD, et al. A dual role for Caspase8 and NF-kappaB interactions in regulating apoptosis and necroptosis of ovarian cancer, with correlation to patient survival. Cell Death Discov. 2015;1:15053.PubMedPubMedCentralCrossRef Hernandez L, Kim MK, Noonan AM, Sagher E, Kohlhammer H, Wright G, Lyle LT, Steeg PS, Anver M, Bowtell DD, et al. A dual role for Caspase8 and NF-kappaB interactions in regulating apoptosis and necroptosis of ovarian cancer, with correlation to patient survival. Cell Death Discov. 2015;1:15053.PubMedPubMedCentralCrossRef
78.
go back to reference Jin Z, Li Y, Pitti R, Lawrence D, Pham VC, Lill JR, Ashkenazi A. Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell. 2009;137(4):721–35.PubMedCrossRef Jin Z, Li Y, Pitti R, Lawrence D, Pham VC, Lill JR, Ashkenazi A. Cullin3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell. 2009;137(4):721–35.PubMedCrossRef
79.
go back to reference Huang S, Okamoto K, Yu C, Sinicrope FA. p62/sequestosome-1 up-regulation promotes ABT-263-induced caspase-8 aggregation/activation on the autophagosome. J Biol Chem. 2013;288(47):33654–66.PubMedPubMedCentralCrossRef Huang S, Okamoto K, Yu C, Sinicrope FA. p62/sequestosome-1 up-regulation promotes ABT-263-induced caspase-8 aggregation/activation on the autophagosome. J Biol Chem. 2013;288(47):33654–66.PubMedPubMedCentralCrossRef
80.
go back to reference Young MM, Takahashi Y, Khan O, Park S, Hori T, Yun J, Sharma AK, Amin S, Hu CD, Zhang J, et al. Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis. J Biol Chem. 2012;287(15):12455–68.PubMedPubMedCentralCrossRef Young MM, Takahashi Y, Khan O, Park S, Hori T, Yun J, Sharma AK, Amin S, Hu CD, Zhang J, et al. Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis. J Biol Chem. 2012;287(15):12455–68.PubMedPubMedCentralCrossRef
81.
go back to reference Iurlaro R, Munoz-Pinedo C. Cell death induced by endoplasmic reticulum stress. FEBS J. 2016;283(14):2640–52.PubMedCrossRef Iurlaro R, Munoz-Pinedo C. Cell death induced by endoplasmic reticulum stress. FEBS J. 2016;283(14):2640–52.PubMedCrossRef
82.
go back to reference Peng H, Yang J, Li G, You Q, Han W, Li T, Gao D, Xie X, Lee BH, Du J, et al. Ubiquitylation of p62/sequestosome1 activates its autophagy receptor function and controls selective autophagy upon ubiquitin stress. Cell Res. 2017;27(5):657–74.PubMedPubMedCentralCrossRef Peng H, Yang J, Li G, You Q, Han W, Li T, Gao D, Xie X, Lee BH, Du J, et al. Ubiquitylation of p62/sequestosome1 activates its autophagy receptor function and controls selective autophagy upon ubiquitin stress. Cell Res. 2017;27(5):657–74.PubMedPubMedCentralCrossRef
83.
go back to reference El-Gazzar A, Wittinger M, Perco P, Anees M, Horvat R, Mikulits W, Grunt TW, Mayer B, Krainer M. The role of c-FLIP(L) in ovarian cancer: chaperoning tumor cells from immunosurveillance and increasing their invasive potential. Gynecol Oncol. 2010;117(3):451–9.PubMedCrossRef El-Gazzar A, Wittinger M, Perco P, Anees M, Horvat R, Mikulits W, Grunt TW, Mayer B, Krainer M. The role of c-FLIP(L) in ovarian cancer: chaperoning tumor cells from immunosurveillance and increasing their invasive potential. Gynecol Oncol. 2010;117(3):451–9.PubMedCrossRef
84.
go back to reference Clarke P, Tyler KL. Down-regulation of cFLIP following reovirus infection sensitizes human ovarian cancer cells to TRAIL-induced apoptosis. Apoptosis. 2007;12(1):211–23.PubMedPubMedCentralCrossRef Clarke P, Tyler KL. Down-regulation of cFLIP following reovirus infection sensitizes human ovarian cancer cells to TRAIL-induced apoptosis. Apoptosis. 2007;12(1):211–23.PubMedPubMedCentralCrossRef
85.
go back to reference Li LC, Jayaram S, Ganesh L, Qian L, Rotmensch J, Maker AV, Prabhakar BS. Knockdown of MADD and c-FLIP overcomes resistance to TRAIL-induced apoptosis in ovarian cancer cells. Am J Obstet Gynecol. 2011;205(4):362 e312-325.CrossRef Li LC, Jayaram S, Ganesh L, Qian L, Rotmensch J, Maker AV, Prabhakar BS. Knockdown of MADD and c-FLIP overcomes resistance to TRAIL-induced apoptosis in ovarian cancer cells. Am J Obstet Gynecol. 2011;205(4):362 e312-325.CrossRef
86.
go back to reference Nazim UM, Moon JH, Lee JH, Lee YJ, Seol JW, Eo SK, Lee JH, Park SY. Activation of autophagy flux by metformin downregulates cellular FLICE-like inhibitory protein and enhances TRAIL—induced apoptosis. Oncotarget. 2016;7(17):23468–81.PubMedPubMedCentralCrossRef Nazim UM, Moon JH, Lee JH, Lee YJ, Seol JW, Eo SK, Lee JH, Park SY. Activation of autophagy flux by metformin downregulates cellular FLICE-like inhibitory protein and enhances TRAIL—induced apoptosis. Oncotarget. 2016;7(17):23468–81.PubMedPubMedCentralCrossRef
87.
go back to reference Xu Y, Ma HB, Fang YL, Zhang ZR, Shao J, Hong M, Huang CJ, Liu J, Chen RQ. Cisplatin-induced necroptosis in TNFalpha dependent and independent pathways. Cell Signal. 2017;31:112–23.PubMedCrossRef Xu Y, Ma HB, Fang YL, Zhang ZR, Shao J, Hong M, Huang CJ, Liu J, Chen RQ. Cisplatin-induced necroptosis in TNFalpha dependent and independent pathways. Cell Signal. 2017;31:112–23.PubMedCrossRef
88.
go back to reference Liu S, Li Y, Choi HMC, Sarkar C, Koh EY, Wu J, Lipinski MM. Lysosomal damage after spinal cord injury causes accumulation of RIPK1 and RIPK3 proteins and potentiation of necroptosis. Cell Death Dis. 2018;9(5):476.PubMedPubMedCentralCrossRef Liu S, Li Y, Choi HMC, Sarkar C, Koh EY, Wu J, Lipinski MM. Lysosomal damage after spinal cord injury causes accumulation of RIPK1 and RIPK3 proteins and potentiation of necroptosis. Cell Death Dis. 2018;9(5):476.PubMedPubMedCentralCrossRef
89.
go back to reference Goodall ML, Fitzwalter BE, Zahedi S, Wu M, Rodriguez D, Mulcahy-Levy JM, Green DR, Morgan M, Cramer SD, Thorburn A. The autophagy machinery controls cell death switching between apoptosis and necroptosis. Devel Cell. 2016;37(4):337–49.CrossRef Goodall ML, Fitzwalter BE, Zahedi S, Wu M, Rodriguez D, Mulcahy-Levy JM, Green DR, Morgan M, Cramer SD, Thorburn A. The autophagy machinery controls cell death switching between apoptosis and necroptosis. Devel Cell. 2016;37(4):337–49.CrossRef
90.
go back to reference Pilie PG, Tang C, Mills GB, Yap TA. State-of-the-art strategies for targeting the DNA damage response in cancer. Nature reviews Clinical oncology. 2019;16(2):81–104.PubMedCrossRefPubMedCentral Pilie PG, Tang C, Mills GB, Yap TA. State-of-the-art strategies for targeting the DNA damage response in cancer. Nature reviews Clinical oncology. 2019;16(2):81–104.PubMedCrossRefPubMedCentral
91.
go back to reference Gao L, Wang Z, Lu D, Huang J, Liu J, Hong L. Paeonol induces cytoprotective autophagy via blocking the Akt/mTOR pathway in ovarian cancer cells. Cell Death Dis. 2019;10(8):609.PubMedPubMedCentralCrossRef Gao L, Wang Z, Lu D, Huang J, Liu J, Hong L. Paeonol induces cytoprotective autophagy via blocking the Akt/mTOR pathway in ovarian cancer cells. Cell Death Dis. 2019;10(8):609.PubMedPubMedCentralCrossRef
92.
go back to reference Liu B, Huang X, Li Y, Liao W, Li M, Liu Y, He R, Feng D, Zhu R, Kurihara H. JS-K, a nitric oxide donor, induces autophagy as a complementary mechanism inhibiting ovarian cancer. BMC Cancer. 2019;19(1):645.PubMedPubMedCentralCrossRef Liu B, Huang X, Li Y, Liao W, Li M, Liu Y, He R, Feng D, Zhu R, Kurihara H. JS-K, a nitric oxide donor, induces autophagy as a complementary mechanism inhibiting ovarian cancer. BMC Cancer. 2019;19(1):645.PubMedPubMedCentralCrossRef
93.
go back to reference Zhang X, Hou G, Liu A, Xu H, Guan Y, Wu Y, Deng J, Cao X. Matrine inhibits the development and progression of ovarian cancer by repressing cancer associated phosphorylation signaling pathways. Cell Death Dis. 2019;10(10):770.PubMedPubMedCentralCrossRef Zhang X, Hou G, Liu A, Xu H, Guan Y, Wu Y, Deng J, Cao X. Matrine inhibits the development and progression of ovarian cancer by repressing cancer associated phosphorylation signaling pathways. Cell Death Dis. 2019;10(10):770.PubMedPubMedCentralCrossRef
94.
go back to reference Bhattacharjee A, Hasanain M, Kathuria M, Singh A, Datta D, Sarkar J, Mitra K. Ormeloxifene-induced unfolded protein response contributes to autophagy-associated apoptosis via disruption of Akt/mTOR and activation of JNK. Sci Rep. 2018;8(1):2303.PubMedPubMedCentralCrossRef Bhattacharjee A, Hasanain M, Kathuria M, Singh A, Datta D, Sarkar J, Mitra K. Ormeloxifene-induced unfolded protein response contributes to autophagy-associated apoptosis via disruption of Akt/mTOR and activation of JNK. Sci Rep. 2018;8(1):2303.PubMedPubMedCentralCrossRef
95.
go back to reference Young AN, Herrera D, Huntsman AC, Korkmaz MA, Lantvit DD, Mazumder S, Kolli S, Coss CC, King S, Wang H, et al. Phyllanthusmin derivatives induce apoptosis and reduce tumor burden in high-grade serous ovarian cancer by late-stage autophagy inhibition. Mol Cancer Ther. 2018;17(10):2123–35.PubMedPubMedCentralCrossRef Young AN, Herrera D, Huntsman AC, Korkmaz MA, Lantvit DD, Mazumder S, Kolli S, Coss CC, King S, Wang H, et al. Phyllanthusmin derivatives induce apoptosis and reduce tumor burden in high-grade serous ovarian cancer by late-stage autophagy inhibition. Mol Cancer Ther. 2018;17(10):2123–35.PubMedPubMedCentralCrossRef
96.
go back to reference Li BX, Wang HB, Qiu MZ, Luo QY, Yi HJ, Yan XL, Pan WT, Yuan LP, Zhang YX, Xu JH, et al. Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, ripoptosome and autophagy mediated cell death pathway. J Exp Clin Cancer Res. 2018;37(1):53.PubMedPubMedCentralCrossRef Li BX, Wang HB, Qiu MZ, Luo QY, Yi HJ, Yan XL, Pan WT, Yuan LP, Zhang YX, Xu JH, et al. Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, ripoptosome and autophagy mediated cell death pathway. J Exp Clin Cancer Res. 2018;37(1):53.PubMedPubMedCentralCrossRef
97.
go back to reference Kao C, Chao A, Tsai CL, Chuang WC, Huang WP, Chen GC, Lin CY, Wang TH, Wang HS, Lai CH. Bortezomib enhances cancer cell death by blocking the autophagic flux through stimulating ERK phosphorylation. Cell Death Dis. 2014;5:e1510.PubMedPubMedCentralCrossRef Kao C, Chao A, Tsai CL, Chuang WC, Huang WP, Chen GC, Lin CY, Wang TH, Wang HS, Lai CH. Bortezomib enhances cancer cell death by blocking the autophagic flux through stimulating ERK phosphorylation. Cell Death Dis. 2014;5:e1510.PubMedPubMedCentralCrossRef
98.
go back to reference Mao J, Ma L, Shen Y, Zhu K, Zhang R, Xi W, Ruan Z, Luo C, Chen Z, Xi X, et al. Arsenic circumvents the gefitinib resistance by binding to P62 and mediating autophagic degradation of EGFR in non-small cell lung cancer. Cell Death Dis. 2018;9(10):963.PubMedPubMedCentralCrossRef Mao J, Ma L, Shen Y, Zhu K, Zhang R, Xi W, Ruan Z, Luo C, Chen Z, Xi X, et al. Arsenic circumvents the gefitinib resistance by binding to P62 and mediating autophagic degradation of EGFR in non-small cell lung cancer. Cell Death Dis. 2018;9(10):963.PubMedPubMedCentralCrossRef
99.
go back to reference Iwadate R, Inoue J, Tsuda H, Takano M, Furuya K, Hirasawa A, Aoki D, Inazawa J. High expression of p62 protein is associated with poor prognosis and aggressive phenotypes in endometrial cancer. Am J Pathol. 2015;185(9):2523–33.PubMedCrossRef Iwadate R, Inoue J, Tsuda H, Takano M, Furuya K, Hirasawa A, Aoki D, Inazawa J. High expression of p62 protein is associated with poor prognosis and aggressive phenotypes in endometrial cancer. Am J Pathol. 2015;185(9):2523–33.PubMedCrossRef
100.
go back to reference Wang J, Garbutt C, Ma H, Gao P, Hornicek FJ, Kan Q, Shi H, Duan Z. Expression and role of autophagy-associated p62 (SQSTM1) in multidrug resistant ovarian cancer. Gynecol Oncol. 2018;150(1):143–50.PubMedCrossRef Wang J, Garbutt C, Ma H, Gao P, Hornicek FJ, Kan Q, Shi H, Duan Z. Expression and role of autophagy-associated p62 (SQSTM1) in multidrug resistant ovarian cancer. Gynecol Oncol. 2018;150(1):143–50.PubMedCrossRef
Metadata
Title
Insight into the role of p62 in the cisplatin resistant mechanisms of ovarian cancer
Authors
Xiao-Yu Yan
Xian-Zhi Qu
Long Xu
Si-Hang Yu
Rui Tian
Xin-Ru Zhong
Lian-Kun Sun
Jing Su
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/s12935-020-01196-w

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