Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01-12-2020 | Acute Myeloid Leukemia | Research

PERK/NRF2 and autophagy form a resistance mechanism against G9a inhibition in leukemia stem cells

Authors: Ji Eun Jang, Ju-In Eom, Hoi-Kyung Jeung, Haerim Chung, Yu Ri Kim, Jin Seok Kim, June-Won Cheong, Yoo Hong Min

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2020

Login to get access

Abstract

Background

The histone methyltransferase G9a has recently been identified as a potential target for epigenetic therapy of acute myeloid leukemia (AML). However, the effect of G9a inhibition on leukemia stem cells (LSCs), which are responsible for AML drug resistance and recurrence, is unclear. In this study, we investigated the underlying mechanisms of the LSC resistance to G9a inhibition.

Methods

We evaluated the effects of G9a inhibition on the unfolded protein response and autophagy in AML and LSC-like cell lines and in primary CD34+CD38 leukemic blasts from patients with AML and investigated the underlying mechanisms. The effects of treatment on cells were evaluated by flow cytometry, western blotting, confocal microscopy, reactive oxygen species (ROS) production assay.

Results

The G9a inhibitor BIX-01294 effectively induced apoptosis in AML cell lines; however, the effect was limited in KG1 LSC-like cells. BIX-01294 treatment or siRNA-mediated G9a knockdown led to the activation of the PERK/NRF2 pathway and HO-1 upregulation in KG1 cells. Phosphorylation of p38 and intracellular generation of reactive oxygen species (ROS) were suppressed. Pharmacological or siRNA-mediated inhibition of the PERK/NRF2 pathway synergistically enhanced BIX-01294-induced apoptosis, with suppressed HO-1 expression, increased p38 phosphorylation, and elevated ROS generation, indicating that activated PERK/NRF2 signaling suppressed ROS-induced apoptosis in KG1 cells. By contrast, cotreatment of normal hematopoietic stem cells with BIX-01294 and a PERK inhibitor had no significant proapoptotic effect. Additionally, G9a inhibition induced autophagy flux in KG1 cells, while autophagy inhibitors significantly increased the BIX-01294-induced apoptosis. This prosurvival autophagy was not abrogated by PERK/NRF2 inhibition.

Conclusions

PERK/NRF2 signaling plays a key role in protecting LSCs against ROS-induced apoptosis, thus conferring resistance to G9a inhibitors. Treatment with PERK/NRF2 or autophagy inhibitors could overcome resistance to G9a inhibition and eliminate LSCs, suggesting the potential clinical utility of these unique targeted therapies against AML.
Appendix
Available only for authorised users
Literature
1.
go back to reference Marcucci G, Haferlach T, Dohner H. Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. J Clin Oncol. 2011;29(5):475–86.PubMedCrossRef Marcucci G, Haferlach T, Dohner H. Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. J Clin Oncol. 2011;29(5):475–86.PubMedCrossRef
2.
go back to reference Ravandi F, Estrov Z. Eradication of leukemia stem cells as a new goal of therapy in leukemia. Clin Cancer Res. 2006;12(2):340–4.PubMedCrossRef Ravandi F, Estrov Z. Eradication of leukemia stem cells as a new goal of therapy in leukemia. Clin Cancer Res. 2006;12(2):340–4.PubMedCrossRef
3.
go back to reference Bruserud O, Aasebo E, Hernandez-Valladares M, Tsykunova G, Reikvam H. Therapeutic targeting of leukemic stem cells in acute myeloid leukemia - the biological background for possible strategies. Expert Opin Drug Discov. 2017;12(10):1053–65.PubMedCrossRef Bruserud O, Aasebo E, Hernandez-Valladares M, Tsykunova G, Reikvam H. Therapeutic targeting of leukemic stem cells in acute myeloid leukemia - the biological background for possible strategies. Expert Opin Drug Discov. 2017;12(10):1053–65.PubMedCrossRef
4.
go back to reference Agirre X, Martinez-Climent JA, Odero MD, Prosper F. Epigenetic regulation of miRNA genes in acute leukemia. Leukemia. 2012;26(3):395–403.PubMedCrossRef Agirre X, Martinez-Climent JA, Odero MD, Prosper F. Epigenetic regulation of miRNA genes in acute leukemia. Leukemia. 2012;26(3):395–403.PubMedCrossRef
5.
go back to reference Yan XJ, Xu J, Gu ZH, Pan CM, Lu G, Shen Y, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2011;43(4):309–15.PubMedCrossRef Yan XJ, Xu J, Gu ZH, Pan CM, Lu G, Shen Y, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2011;43(4):309–15.PubMedCrossRef
6.
go back to reference Wainwright EN, Scaffidi P. Epigenetics and Cancer stem cells: unleashing, hijacking, and restricting cellular plasticity. Trends Cancer. 2017;3(5):372–86.PubMedPubMedCentralCrossRef Wainwright EN, Scaffidi P. Epigenetics and Cancer stem cells: unleashing, hijacking, and restricting cellular plasticity. Trends Cancer. 2017;3(5):372–86.PubMedPubMedCentralCrossRef
7.
go back to reference Jung N, Dai B, Gentles AJ, Majeti R, Feinberg AP. An LSC epigenetic signature is largely mutation independent and implicates the HOXA cluster in AML pathogenesis. Nat Commun. 2015;6:8489.PubMedCrossRef Jung N, Dai B, Gentles AJ, Majeti R, Feinberg AP. An LSC epigenetic signature is largely mutation independent and implicates the HOXA cluster in AML pathogenesis. Nat Commun. 2015;6:8489.PubMedCrossRef
9.
go back to reference Hu L, Zang MD, Wang HX, Zhang BG, Wang ZQ, Fan ZY, et al. G9A promotes gastric cancer metastasis by upregulating ITGB3 in a SET domain-independent manner. Cell Death Dis. 2018;9(3):278.PubMedPubMedCentralCrossRef Hu L, Zang MD, Wang HX, Zhang BG, Wang ZQ, Fan ZY, et al. G9A promotes gastric cancer metastasis by upregulating ITGB3 in a SET domain-independent manner. Cell Death Dis. 2018;9(3):278.PubMedPubMedCentralCrossRef
10.
go back to reference Qin J, Zeng Z, Luo T, Li Q, Hao Y, Chen L. Clinicopathological significance of G9A expression in colorectal carcinoma. Oncol Lett. 2018;15(6):8611–9.PubMedPubMedCentral Qin J, Zeng Z, Luo T, Li Q, Hao Y, Chen L. Clinicopathological significance of G9A expression in colorectal carcinoma. Oncol Lett. 2018;15(6):8611–9.PubMedPubMedCentral
11.
go back to reference Zhang K, Wang J, Yang L, Yuan YC, Tong TR, Wu J, et al. Targeting histone methyltransferase G9a inhibits growth and Wnt signaling pathway by epigenetically regulating HP1alpha and APC2 gene expression in non-small cell lung cancer. Mol Cancer. 2018;17(1):153.PubMedPubMedCentralCrossRef Zhang K, Wang J, Yang L, Yuan YC, Tong TR, Wu J, et al. Targeting histone methyltransferase G9a inhibits growth and Wnt signaling pathway by epigenetically regulating HP1alpha and APC2 gene expression in non-small cell lung cancer. Mol Cancer. 2018;17(1):153.PubMedPubMedCentralCrossRef
12.
go back to reference Mayr C, Helm K, Jakab M, Ritter M, Shrestha R, Makaju R, et al. The histone methyltransferase G9a: a new therapeutic target in biliary tract cancer. Hum Pathol. 2018;72:117–26.PubMedCrossRef Mayr C, Helm K, Jakab M, Ritter M, Shrestha R, Makaju R, et al. The histone methyltransferase G9a: a new therapeutic target in biliary tract cancer. Hum Pathol. 2018;72:117–26.PubMedCrossRef
14.
go back to reference Wojtala M, Macierzynska-Piotrowska E, Rybaczek D, Pirola L, Balcerczyk A. Pharmacological and transcriptional inhibition of the G9a histone methyltransferase suppresses proliferation and modulates redox homeostasis in human microvascular endothelial cells. Pharmacol Res. 2018;128:252–63.PubMedCrossRef Wojtala M, Macierzynska-Piotrowska E, Rybaczek D, Pirola L, Balcerczyk A. Pharmacological and transcriptional inhibition of the G9a histone methyltransferase suppresses proliferation and modulates redox homeostasis in human microvascular endothelial cells. Pharmacol Res. 2018;128:252–63.PubMedCrossRef
15.
go back to reference Chen H, Yan Y, Davidson TL, Shinkai Y, Costa M. Hypoxic stress induces dimethylated histone H3 lysine 9 through histone methyltransferase G9a in mammalian cells. Cancer Res. 2006;66(18):9009–16.PubMedCrossRef Chen H, Yan Y, Davidson TL, Shinkai Y, Costa M. Hypoxic stress induces dimethylated histone H3 lysine 9 through histone methyltransferase G9a in mammalian cells. Cancer Res. 2006;66(18):9009–16.PubMedCrossRef
16.
go back to reference Casciello F, Al-Ejeh F, Kelly G, Brennan DJ, Ngiow SF, Young A, et al. G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis. Proc Natl Acad Sci U S A. 2017;114(27):7077–82.PubMedPubMedCentralCrossRef Casciello F, Al-Ejeh F, Kelly G, Brennan DJ, Ngiow SF, Young A, et al. G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis. Proc Natl Acad Sci U S A. 2017;114(27):7077–82.PubMedPubMedCentralCrossRef
17.
go back to reference Jiang W, Liu P, Li X. G9A performs important roles in the progression of breast cancer through upregulating its targets. Oncol Lett. 2017;13(6):4127–32.PubMedPubMedCentralCrossRef Jiang W, Liu P, Li X. G9A performs important roles in the progression of breast cancer through upregulating its targets. Oncol Lett. 2017;13(6):4127–32.PubMedPubMedCentralCrossRef
18.
go back to reference Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 2010;70(20):7830–40.PubMedCrossRef Chen MW, Hua KT, Kao HJ, Chi CC, Wei LH, Johansson G, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res. 2010;70(20):7830–40.PubMedCrossRef
19.
go back to reference Kim Y, Kim YS, Kim DE, Lee JS, Song JH, Kim HG, et al. BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production. Autophagy. 2013;9(12):2126–39.PubMedCrossRef Kim Y, Kim YS, Kim DE, Lee JS, Song JH, Kim HG, et al. BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production. Autophagy. 2013;9(12):2126–39.PubMedCrossRef
21.
go back to reference Lehnertz B, Pabst C, Su L, Miller M, Liu F, Yi L, et al. The methyltransferase G9a regulates HoxA9-dependent transcription in AML. Genes Dev. 2014;28(4):317–27.PubMedPubMedCentralCrossRef Lehnertz B, Pabst C, Su L, Miller M, Liu F, Yi L, et al. The methyltransferase G9a regulates HoxA9-dependent transcription in AML. Genes Dev. 2014;28(4):317–27.PubMedPubMedCentralCrossRef
22.
go back to reference Pappano WN, Guo J, He Y, Ferguson D, Jagadeeswaran S, Osterling DJ, et al. The histone methyltransferase inhibitor A-366 uncovers a role for G9a/GLP in the epigenetics of leukemia. PLoS One. 2015;10(7):e0131716.PubMedPubMedCentralCrossRef Pappano WN, Guo J, He Y, Ferguson D, Jagadeeswaran S, Osterling DJ, et al. The histone methyltransferase inhibitor A-366 uncovers a role for G9a/GLP in the epigenetics of leukemia. PLoS One. 2015;10(7):e0131716.PubMedPubMedCentralCrossRef
23.
go back to reference Kondengaden SM, Luo LF, Huang K, Zhu M, Zang L, Bataba E, et al. Discovery of novel small molecule inhibitors of lysine methyltransferase G9a and their mechanism in leukemia cell lines. Eur J Med Chem. 2016;122:382–93.PubMedCrossRef Kondengaden SM, Luo LF, Huang K, Zhu M, Zang L, Bataba E, et al. Discovery of novel small molecule inhibitors of lysine methyltransferase G9a and their mechanism in leukemia cell lines. Eur J Med Chem. 2016;122:382–93.PubMedCrossRef
24.
go back to reference San Jose-Eneriz E, Agirre X, Rabal O, Vilas-Zornoza A, Sanchez-Arias JA, Miranda E, et al. Discovery of first-in-class reversible dual small molecule inhibitors against G9a and DNMTs in hematological malignancies. Nat Commun. 2017;8:15424.PubMedPubMedCentralCrossRef San Jose-Eneriz E, Agirre X, Rabal O, Vilas-Zornoza A, Sanchez-Arias JA, Miranda E, et al. Discovery of first-in-class reversible dual small molecule inhibitors against G9a and DNMTs in hematological malignancies. Nat Commun. 2017;8:15424.PubMedPubMedCentralCrossRef
25.
go back to reference Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519–29.PubMedCrossRef Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519–29.PubMedCrossRef
26.
go back to reference Cui J, Sun W, Hao X, Wei M, Su X, Zhang Y, et al. EHMT2 inhibitor BIX-01294 induces apoptosis through PMAIP1-USP9X-MCL1 axis in human bladder cancer cells. Cancer Cell Int. 2015;15(1):4.PubMedPubMedCentralCrossRef Cui J, Sun W, Hao X, Wei M, Su X, Zhang Y, et al. EHMT2 inhibitor BIX-01294 induces apoptosis through PMAIP1-USP9X-MCL1 axis in human bladder cancer cells. Cancer Cell Int. 2015;15(1):4.PubMedPubMedCentralCrossRef
27.
go back to reference Kusio-Kobialka M, Podszywalow-Bartnicka P, Peidis P, Glodkowska-Mrowka E, Wolanin K, Leszak G, et al. The PERK-eIF2alpha phosphorylation arm is a pro-survival pathway of BCR-ABL signaling and confers resistance to imatinib treatment in chronic myeloid leukemia cells. Cell Cycle. 2012;11(21):4069–78.PubMedPubMedCentralCrossRef Kusio-Kobialka M, Podszywalow-Bartnicka P, Peidis P, Glodkowska-Mrowka E, Wolanin K, Leszak G, et al. The PERK-eIF2alpha phosphorylation arm is a pro-survival pathway of BCR-ABL signaling and confers resistance to imatinib treatment in chronic myeloid leukemia cells. Cell Cycle. 2012;11(21):4069–78.PubMedPubMedCentralCrossRef
28.
go back to reference Higa A, Chevet E. Redox signaling loops in the unfolded protein response. Cell Signal. 2012;24(8):1548–55.PubMedCrossRef Higa A, Chevet E. Redox signaling loops in the unfolded protein response. Cell Signal. 2012;24(8):1548–55.PubMedCrossRef
29.
go back to reference Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 1997;236(2):313–22.PubMedCrossRef Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 1997;236(2):313–22.PubMedCrossRef
30.
go back to reference Cubillos-Ruiz JR, Bettigole SE, Glimcher LH. Tumorigenic and immunosuppressive effects of endoplasmic reticulum stress in Cancer. Cell. 2017;168(4):692–706. Cubillos-Ruiz JR, Bettigole SE, Glimcher LH. Tumorigenic and immunosuppressive effects of endoplasmic reticulum stress in Cancer. Cell. 2017;168(4):692–706.
31.
go back to reference Yun CW, Lee SH, The Roles of Autophagy in Cancer. Int J Mol Sci. 2018;1(11):3466. Yun CW, Lee SH, The Roles of Autophagy in Cancer. Int J Mol Sci. 2018;1(11):3466.
32.
go back to reference Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937–51.PubMedCrossRef Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937–51.PubMedCrossRef
33.
go back to reference Jang JE, Eom JI, Jeung HK, Cheong JW, Lee JY, Kim JS, et al. AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells. Clin Cancer Res. 2017;23(11):2781–94.PubMedCrossRef Jang JE, Eom JI, Jeung HK, Cheong JW, Lee JY, Kim JS, et al. AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells. Clin Cancer Res. 2017;23(11):2781–94.PubMedCrossRef
34.
go back to reference Williams BA, Wang XH, Keating A. Clonogenic assays measure leukemia stem cell killing not detectable by chromium release and flow cytometric cytotoxicity assays. Cytotherapy. 2010;12(7):951–60.PubMedCrossRef Williams BA, Wang XH, Keating A. Clonogenic assays measure leukemia stem cell killing not detectable by chromium release and flow cytometric cytotoxicity assays. Cytotherapy. 2010;12(7):951–60.PubMedCrossRef
35.
go back to reference She M, Niu X, Chen X, Li J, Zhou M, He Y, et al. Resistance of leukemic stem-like cells in AML cell line KG1a to natural killer cell-mediated cytotoxicity. Cancer Lett. 2012;318(2):173–9.PubMedCrossRef She M, Niu X, Chen X, Li J, Zhou M, He Y, et al. Resistance of leukemic stem-like cells in AML cell line KG1a to natural killer cell-mediated cytotoxicity. Cancer Lett. 2012;318(2):173–9.PubMedCrossRef
36.
go back to reference Pedranzini L, Mottadelli F, Ronzoni S, Rossella F, Ferracin M, Magnani I, et al. Differential cytogenomics and miRNA signature of the acute myeloid Leukaemia Kasumi-1 cell line CD34+38- compartment. Leuk Res. 2010;34(10):1287–95.PubMedCrossRef Pedranzini L, Mottadelli F, Ronzoni S, Rossella F, Ferracin M, Magnani I, et al. Differential cytogenomics and miRNA signature of the acute myeloid Leukaemia Kasumi-1 cell line CD34+38- compartment. Leuk Res. 2010;34(10):1287–95.PubMedCrossRef
37.
go back to reference Guo R, Su Y, Liu B, Li S, Zhou S, Xu Y. Resveratrol suppresses oxidised low-density lipoprotein-induced macrophage apoptosis through inhibition of intracellular reactive oxygen species generation, LOX-1, and the p38 MAPK pathway. Cell Physiol Biochem. 2014;34(2):603–16.PubMedCrossRef Guo R, Su Y, Liu B, Li S, Zhou S, Xu Y. Resveratrol suppresses oxidised low-density lipoprotein-induced macrophage apoptosis through inhibition of intracellular reactive oxygen species generation, LOX-1, and the p38 MAPK pathway. Cell Physiol Biochem. 2014;34(2):603–16.PubMedCrossRef
38.
go back to reference Kim AH, Khursigara G, Sun X, Franke TF, Chao MV. Akt phosphorylates and negatively regulates apoptosis signal-regulating kinase 1. Mol Cell Biol. 2001;21(3):893–901.PubMedPubMedCentralCrossRef Kim AH, Khursigara G, Sun X, Franke TF, Chao MV. Akt phosphorylates and negatively regulates apoptosis signal-regulating kinase 1. Mol Cell Biol. 2001;21(3):893–901.PubMedPubMedCentralCrossRef
39.
40.
go back to reference Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, et al. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science. 1997;275(5296):90–4.PubMedCrossRef Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, et al. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science. 1997;275(5296):90–4.PubMedCrossRef
41.
go back to reference Axten JM, Medina JR, Feng Y, Shu A, Romeril SP, Grant SW, et al. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-p yrrolo [2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). J Med Chem. 2012;55(16):7193–207.PubMedCrossRef Axten JM, Medina JR, Feng Y, Shu A, Romeril SP, Grant SW, et al. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-p yrrolo [2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). J Med Chem. 2012;55(16):7193–207.PubMedCrossRef
42.
go back to reference Cullinan SB, Diehl JA. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J Biol Chem. 2004;279(19):20108–17.PubMedCrossRef Cullinan SB, Diehl JA. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J Biol Chem. 2004;279(19):20108–17.PubMedCrossRef
43.
go back to reference Furfaro AL, Traverso N, Domenicotti C, Piras S, Moretta L, Marinari UM, et al. The Nrf2/HO-1 Axis in Cancer cell growth and Chemoresistance. Oxidative Med Cell Longev. 2016;2016:1958174.CrossRef Furfaro AL, Traverso N, Domenicotti C, Piras S, Moretta L, Marinari UM, et al. The Nrf2/HO-1 Axis in Cancer cell growth and Chemoresistance. Oxidative Med Cell Longev. 2016;2016:1958174.CrossRef
44.
go back to reference Testa U, Labbaye C, Castelli G, Pelosi E. Oxidative stress and hypoxia in normal and leukemic stem cells. Exp Hematol. 2016;44(7):540–60.PubMedCrossRef Testa U, Labbaye C, Castelli G, Pelosi E. Oxidative stress and hypoxia in normal and leukemic stem cells. Exp Hematol. 2016;44(7):540–60.PubMedCrossRef
45.
go back to reference Prieto-Bermejo R, Romo-Gonzalez M, Perez-Fernandez A, Ijurko C, Hernandez-Hernandez A. Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. J Exp Clin Cancer Res. 2018;37(1):125.PubMedPubMedCentralCrossRef Prieto-Bermejo R, Romo-Gonzalez M, Perez-Fernandez A, Ijurko C, Hernandez-Hernandez A. Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. J Exp Clin Cancer Res. 2018;37(1):125.PubMedPubMedCentralCrossRef
46.
go back to reference Rushworth SA, Zaitseva L, Murray MY, Shah NM, Bowles KM, MacEwan DJ. The high Nrf2 expression in human acute myeloid leukemia is driven by NF-kappaB and underlies its chemo-resistance. Blood. 2012;120(26):5188–98.PubMedCrossRef Rushworth SA, Zaitseva L, Murray MY, Shah NM, Bowles KM, MacEwan DJ. The high Nrf2 expression in human acute myeloid leukemia is driven by NF-kappaB and underlies its chemo-resistance. Blood. 2012;120(26):5188–98.PubMedCrossRef
47.
go back to reference Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis. 2013;4:e838.PubMedPubMedCentralCrossRef Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis. 2013;4:e838.PubMedPubMedCentralCrossRef
48.
go back to reference Kim Y, Eom JI, Jeung HK, Jang JE, Kim JS, Cheong JW, et al. Induction of cytosine arabinoside-resistant human myeloid leukemia cell death through autophagy regulation by hydroxychloroquine. Biomed Pharmacother. 2015;73:87–96.PubMedCrossRef Kim Y, Eom JI, Jeung HK, Jang JE, Kim JS, Cheong JW, et al. Induction of cytosine arabinoside-resistant human myeloid leukemia cell death through autophagy regulation by hydroxychloroquine. Biomed Pharmacother. 2015;73:87–96.PubMedCrossRef
49.
go back to reference Ren A, Qiu Y, Cui H, Fu G. Inhibition of H3K9 methyltransferase G9a induces autophagy and apoptosis in oral squamous cell carcinoma. Biochem Biophys Res Commun. 2015;459(1):10–7.PubMedCrossRef Ren A, Qiu Y, Cui H, Fu G. Inhibition of H3K9 methyltransferase G9a induces autophagy and apoptosis in oral squamous cell carcinoma. Biochem Biophys Res Commun. 2015;459(1):10–7.PubMedCrossRef
50.
go back to reference Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B. Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy. 2010;6(3):322–9.PubMedCrossRef Dalby KN, Tekedereli I, Lopez-Berestein G, Ozpolat B. Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer. Autophagy. 2010;6(3):322–9.PubMedCrossRef
52.
go back to reference Schonthal AH. Endoplasmic reticulum stress and autophagy as targets for cancer therapy. Cancer Lett. 2009;275(2):163–9.PubMedCrossRef Schonthal AH. Endoplasmic reticulum stress and autophagy as targets for cancer therapy. Cancer Lett. 2009;275(2):163–9.PubMedCrossRef
Metadata
Title
PERK/NRF2 and autophagy form a resistance mechanism against G9a inhibition in leukemia stem cells
Authors
Ji Eun Jang
Ju-In Eom
Hoi-Kyung Jeung
Haerim Chung
Yu Ri Kim
Jin Seok Kim
June-Won Cheong
Yoo Hong Min
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2020
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01565-3

Other articles of this Issue 1/2020

Journal of Experimental & Clinical Cancer Research 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