Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

EIF2A-dependent translational arrest protects leukemia cells from the energetic stress induced by NAMPT inhibition

Authors: Chiara Zucal, Vito G. D’Agostino, Antonio Casini, Barbara Mantelli, Natthakan Thongon, Debora Soncini, Irene Caffa, Michele Cea, Alberto Ballestrero, Alessandro Quattrone, Stefano Indraccolo, Alessio Nencioni, Alessandro Provenzani

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, is one of the major factors regulating cancer cells metabolism and is considered a promising target for treating cancer. The prototypical NAMPT inhibitor FK866 effectively lowers NAD+ levels in cancer cells, reducing the activity of NAD+-dependent enzymes, lowering intracellular ATP, and promoting cell death.

Results

We show that FK866 induces a translational arrest in leukemia cells through inhibition of MTOR/4EBP1 signaling and of the initiation factors EIF4E and EIF2A. Specifically, treatment with FK866 is shown to induce 5′AMP-activated protein kinase (AMPK) activation, which, together with EIF2A phosphorylation, is responsible for the inhibition of protein synthesis. Notably, such an effect was also observed in patients’ derived primary leukemia cells including T-cell Acute Lymphoblastic Leukemia. Jurkat cells in which AMPK or LKB1 expression was silenced or in which a non-phosphorylatable EIF2A mutant was ectopically expressed showed enhanced sensitivity to the NAMPT inhibitor, confirming a key role for the LKB1-AMPK-EIF2A axis in cell fate determination in response to energetic stress via NAD+ depletion.

Conclusions

We identified EIF2A phosphorylation as a novel early molecular event occurring in response to NAMPT inhibition and mediating protein synthesis arrest. In addition, our data suggest that tumors exhibiting an impaired LBK1- AMPK- EIF2A response may be especially susceptible to NAMPT inhibitors and thus become an elective indication for this type of agents.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13(6):472–82.CrossRefPubMed Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13(6):472–82.CrossRefPubMed
2.
go back to reference Tan B, Young DA, Lu Z-H, Wang T, Meier TI, Shepard RL, et al. Pharmacological inhibition of nicotinamide phosphoribosyltransferase (NAMPT), an enzyme essential for NAD+ biosynthesis, in human cancer cells: metabolic basis and potential clinical implications. J Biol Chem. 2013;288(5):3500–11.CrossRefPubMed Tan B, Young DA, Lu Z-H, Wang T, Meier TI, Shepard RL, et al. Pharmacological inhibition of nicotinamide phosphoribosyltransferase (NAMPT), an enzyme essential for NAD+ biosynthesis, in human cancer cells: metabolic basis and potential clinical implications. J Biol Chem. 2013;288(5):3500–11.CrossRefPubMed
3.
go back to reference Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279(49):50754–63.CrossRefPubMed Revollo JR, Grimm AA, Imai S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J Biol Chem. 2004;279(49):50754–63.CrossRefPubMed
4.
go back to reference Skokowa J, Lan D, Thakur BK, Wang F, Gupta K, Cario G, et al. NAMPT is essential for the G-CSF-induced myeloid differentiation via a NAD(+)-sirtuin-1-dependent pathway. Nat Med. 2009;15(2):151–8.CrossRefPubMed Skokowa J, Lan D, Thakur BK, Wang F, Gupta K, Cario G, et al. NAMPT is essential for the G-CSF-induced myeloid differentiation via a NAD(+)-sirtuin-1-dependent pathway. Nat Med. 2009;15(2):151–8.CrossRefPubMed
5.
go back to reference Xiao Y, Elkins K, Durieux JK, Lee L, Oeh J, Yang LX, et al. Dependence of Tumor Cell Lines and Patient-Derived Tumors on the NAD Salvage Pathway Renders Them Sensitive to NAMPT Inhibition with GNE-618. Neoplasia. 2013;15(10):1151–60.CrossRefPubMedPubMedCentral Xiao Y, Elkins K, Durieux JK, Lee L, Oeh J, Yang LX, et al. Dependence of Tumor Cell Lines and Patient-Derived Tumors on the NAD Salvage Pathway Renders Them Sensitive to NAMPT Inhibition with GNE-618. Neoplasia. 2013;15(10):1151–60.CrossRefPubMedPubMedCentral
6.
go back to reference Soncini D, Caffa I, Zoppoli G, Cea M, Cagnetta A, Passalacqua M, et al. Nicotinamide phosphoribosyltransferase promotes epithelial-to-mesenchymal transition as a soluble factor independent of its enzymatic activity. J Biol Chem. 2014;289(49):34189–204.CrossRefPubMedPubMedCentral Soncini D, Caffa I, Zoppoli G, Cea M, Cagnetta A, Passalacqua M, et al. Nicotinamide phosphoribosyltransferase promotes epithelial-to-mesenchymal transition as a soluble factor independent of its enzymatic activity. J Biol Chem. 2014;289(49):34189–204.CrossRefPubMedPubMedCentral
7.
go back to reference Wang B, Hasan MK, Alvarado E, Yuan H, Wu H, Chen WY. NAMPT overexpression in prostate cancer and its contribution to tumor cell survival and stress response. Oncogene. 2011;30(8):907–21.CrossRefPubMed Wang B, Hasan MK, Alvarado E, Yuan H, Wu H, Chen WY. NAMPT overexpression in prostate cancer and its contribution to tumor cell survival and stress response. Oncogene. 2011;30(8):907–21.CrossRefPubMed
8.
go back to reference Nakajima TE, Yamada Y, Hamano T, Furuta K, Gotoda T, Katai H, et al. Adipocytokine levels in gastric cancer patients: resistin and visfatin as biomarkers of gastric cancer. J Gastroenterol. 2009;44(7):685–90.CrossRefPubMed Nakajima TE, Yamada Y, Hamano T, Furuta K, Gotoda T, Katai H, et al. Adipocytokine levels in gastric cancer patients: resistin and visfatin as biomarkers of gastric cancer. J Gastroenterol. 2009;44(7):685–90.CrossRefPubMed
9.
go back to reference Montecucco F, Cea M, Cagnetta A, Damonte P, Nahimana A, Ballestrero A, et al. Nicotinamide phosphoribosyltransferase as a target in inflammation- related disorders. Curr Top Med Chem. 2013;13(23):2930–8.CrossRefPubMed Montecucco F, Cea M, Cagnetta A, Damonte P, Nahimana A, Ballestrero A, et al. Nicotinamide phosphoribosyltransferase as a target in inflammation- related disorders. Curr Top Med Chem. 2013;13(23):2930–8.CrossRefPubMed
10.
go back to reference Bruzzone S, Fruscione F, Morando S, Ferrando T, Poggi A, Garuti A, et al. Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE. PLoS One. 2009;4(11):e7897.CrossRefPubMedPubMedCentral Bruzzone S, Fruscione F, Morando S, Ferrando T, Poggi A, Garuti A, et al. Catastrophic NAD+ depletion in activated T lymphocytes through Nampt inhibition reduces demyelination and disability in EAE. PLoS One. 2009;4(11):e7897.CrossRefPubMedPubMedCentral
11.
go back to reference Zoppoli G, Cea M, Soncini D, Fruscione F, Rudner J, Moran E, et al. Potent synergistic interaction between the Nampt inhibitor APO866 and the apoptosis activator TRAIL in human leukemia cells. Exp Hematol. 2010;38(11):979–88.CrossRefPubMed Zoppoli G, Cea M, Soncini D, Fruscione F, Rudner J, Moran E, et al. Potent synergistic interaction between the Nampt inhibitor APO866 and the apoptosis activator TRAIL in human leukemia cells. Exp Hematol. 2010;38(11):979–88.CrossRefPubMed
12.
go back to reference Nahimana A, Attinger A, Aubry D, Greaney P, Ireson C, Thougaard AV, et al. The NAD biosynthesis inhibitor APO866 has potent antitumor activity against hematologic malignancies. Blood. 2009;113(14):3276–86.CrossRefPubMed Nahimana A, Attinger A, Aubry D, Greaney P, Ireson C, Thougaard AV, et al. The NAD biosynthesis inhibitor APO866 has potent antitumor activity against hematologic malignancies. Blood. 2009;113(14):3276–86.CrossRefPubMed
13.
go back to reference Nencioni A, Cea M, Montecucco F, Longo VD, Patrone F, Carella AM, et al. Autophagy in blood cancers: biological role and therapeutic implications. Haematologica. 2013;98(9):1335–43.CrossRefPubMedPubMedCentral Nencioni A, Cea M, Montecucco F, Longo VD, Patrone F, Carella AM, et al. Autophagy in blood cancers: biological role and therapeutic implications. Haematologica. 2013;98(9):1335–43.CrossRefPubMedPubMedCentral
14.
go back to reference Wosikowski K, Mattern K, Schemainda I, Hasmann M, Rattel B, Löser R. WK175, a novel antitumor agent, decreases the intracellular nicotinamide adenine dinucleotide concentration and induces the apoptotic cascade in human leukemia cells. Cancer Res. 2002;62(4):1057–62.PubMed Wosikowski K, Mattern K, Schemainda I, Hasmann M, Rattel B, Löser R. WK175, a novel antitumor agent, decreases the intracellular nicotinamide adenine dinucleotide concentration and induces the apoptotic cascade in human leukemia cells. Cancer Res. 2002;62(4):1057–62.PubMed
15.
go back to reference Christensen MK, Erichsen KD, Olesen UH, Tjørnelund J, Fristrup P, Thougaard A, et al. Nicotinamide phosphoribosyltransferase inhibitors, design, preparation, and structure-activity relationship. J Med Chem. 2013;56(22):9071–88.CrossRefPubMed Christensen MK, Erichsen KD, Olesen UH, Tjørnelund J, Fristrup P, Thougaard A, et al. Nicotinamide phosphoribosyltransferase inhibitors, design, preparation, and structure-activity relationship. J Med Chem. 2013;56(22):9071–88.CrossRefPubMed
16.
go back to reference Montecucco F, Cea M, Bauer I, Soncini D, Caffa I, Lasigliè D, et al. Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors as therapeutics: rationales, controversies, clinical experience. Curr Drug Targets. 2013;14(6):637–43.CrossRefPubMed Montecucco F, Cea M, Bauer I, Soncini D, Caffa I, Lasigliè D, et al. Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors as therapeutics: rationales, controversies, clinical experience. Curr Drug Targets. 2013;14(6):637–43.CrossRefPubMed
17.
go back to reference Cerna D, Li H, Flaherty S, Takebe N, Coleman CN, Yoo SS. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) activity by small molecule GMX1778 regulates reactive oxygen species (ROS)-mediated cytotoxicity in a p53- and nicotinic acid phosphoribosyltransferase1 (NAPRT1)-dependent manner. J Biol Chem. 2012;287(26):22408–17.CrossRefPubMedPubMedCentral Cerna D, Li H, Flaherty S, Takebe N, Coleman CN, Yoo SS. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) activity by small molecule GMX1778 regulates reactive oxygen species (ROS)-mediated cytotoxicity in a p53- and nicotinic acid phosphoribosyltransferase1 (NAPRT1)-dependent manner. J Biol Chem. 2012;287(26):22408–17.CrossRefPubMedPubMedCentral
18.
go back to reference Del Nagro C, Xiao Y, Rangell L, Reichelt M, O’Brien T. Depletion of the Central Metabolite NAD Leads to Oncosis-mediated Cell Death. J Biol Chem. 2014;289(51):35182–92.CrossRefPubMedPubMedCentral Del Nagro C, Xiao Y, Rangell L, Reichelt M, O’Brien T. Depletion of the Central Metabolite NAD Leads to Oncosis-mediated Cell Death. J Biol Chem. 2014;289(51):35182–92.CrossRefPubMedPubMedCentral
19.
go back to reference Ginet V, Puyal J, Rummel C, Aubry D, Breton C, Cloux A-J, et al. A critical role of autophagy in antileukemia/lymphoma effects of APO866, an inhibitor of NAD biosynthesis. Autophagy. 2014;10(4):603–17.CrossRefPubMedPubMedCentral Ginet V, Puyal J, Rummel C, Aubry D, Breton C, Cloux A-J, et al. A critical role of autophagy in antileukemia/lymphoma effects of APO866, an inhibitor of NAD biosynthesis. Autophagy. 2014;10(4):603–17.CrossRefPubMedPubMedCentral
20.
go back to reference Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F, Visvikis O, et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol. 2013;15(6):647–58.CrossRefPubMedPubMedCentral Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F, Visvikis O, et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol. 2013;15(6):647–58.CrossRefPubMedPubMedCentral
21.
go back to reference Cea M, Cagnetta A, Fulciniti M, Tai Y-T, Hideshima T, Chauhan D, et al. Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase (ERK1/2) inhibition. Blood. 2012;120(17):3519–29.CrossRefPubMedPubMedCentral Cea M, Cagnetta A, Fulciniti M, Tai Y-T, Hideshima T, Chauhan D, et al. Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase (ERK1/2) inhibition. Blood. 2012;120(17):3519–29.CrossRefPubMedPubMedCentral
22.
go back to reference Hardie DG. The AMP-activated protein kinase pathway--new players upstream and downstream. J Cell Sci. 2004;117(Pt 23):5479–87.CrossRefPubMed Hardie DG. The AMP-activated protein kinase pathway--new players upstream and downstream. J Cell Sci. 2004;117(Pt 23):5479–87.CrossRefPubMed
23.
go back to reference Bowlby SC, Thomas MJ, D’Agostino RB, Kridel SJ. Nicotinamide phosphoribosyl transferase (Nampt) is required for de novo lipogenesis in tumor cells. PLoS One. 2012;7(6):e40195.CrossRefPubMedPubMedCentral Bowlby SC, Thomas MJ, D’Agostino RB, Kridel SJ. Nicotinamide phosphoribosyl transferase (Nampt) is required for de novo lipogenesis in tumor cells. PLoS One. 2012;7(6):e40195.CrossRefPubMedPubMedCentral
24.
go back to reference Schuster S, Penke M, Gorski T, Gebhardt R, Weiss TS, Kiess W, et al. FK866-induced NAMPT inhibition activates AMPK and downregulates mTOR signaling in hepatocarcinoma cells. Biochem Biophys Res Commun. 2015;458(2):334–40.CrossRefPubMed Schuster S, Penke M, Gorski T, Gebhardt R, Weiss TS, Kiess W, et al. FK866-induced NAMPT inhibition activates AMPK and downregulates mTOR signaling in hepatocarcinoma cells. Biochem Biophys Res Commun. 2015;458(2):334–40.CrossRefPubMed
25.
go back to reference Wang P, Xu T-Y, Guan Y-F, Tian W-W, Viollet B, Rui Y-C, et al. Nicotinamide phosphoribosyltransferase protects against ischemic stroke through SIRT1-dependent adenosine monophosphate-activated kinase pathway. Ann Neurol. 2011;69(2):360–74.CrossRefPubMed Wang P, Xu T-Y, Guan Y-F, Tian W-W, Viollet B, Rui Y-C, et al. Nicotinamide phosphoribosyltransferase protects against ischemic stroke through SIRT1-dependent adenosine monophosphate-activated kinase pathway. Ann Neurol. 2011;69(2):360–74.CrossRefPubMed
26.
go back to reference Spriggs KA, Stoneley M, Bushell M, Willis AE. Re-programming of translation following cell stress allows IRES-mediated translation to predominate. Biol Cell. 2008;100(1):27–38.CrossRefPubMed Spriggs KA, Stoneley M, Bushell M, Willis AE. Re-programming of translation following cell stress allows IRES-mediated translation to predominate. Biol Cell. 2008;100(1):27–38.CrossRefPubMed
27.
go back to reference Agnusdei V, Minuzzo S, Frasson C, Grassi A, Axelrod F, Satyal S, et al. Therapeutic antibody targeting of Notch1 in T-acute lymphoblastic leukemia xenografts. Leukemia. 2013;28(2):278–88.CrossRefPubMed Agnusdei V, Minuzzo S, Frasson C, Grassi A, Axelrod F, Satyal S, et al. Therapeutic antibody targeting of Notch1 in T-acute lymphoblastic leukemia xenografts. Leukemia. 2013;28(2):278–88.CrossRefPubMed
28.
go back to reference D’Agostino VG, Adami V, Provenzani A. A novel high throughput biochemical assay to evaluate the HuR protein-RNA complex formation. PLoS One. 2013;8(8):e72426.CrossRefPubMedPubMedCentral D’Agostino VG, Adami V, Provenzani A. A novel high throughput biochemical assay to evaluate the HuR protein-RNA complex formation. PLoS One. 2013;8(8):e72426.CrossRefPubMedPubMedCentral
29.
go back to reference Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat Biotechnol. 1997;15(9):871–5.CrossRefPubMed Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat Biotechnol. 1997;15(9):871–5.CrossRefPubMed
30.
go back to reference Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307(5712):1098–101.CrossRefPubMed Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307(5712):1098–101.CrossRefPubMed
31.
go back to reference Moffat J, Grueneberg DA, Yang X, Kim SY, Kloepfer AM, Hinkle G, et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell. 2006;124(6):1283–98.CrossRefPubMed Moffat J, Grueneberg DA, Yang X, Kim SY, Kloepfer AM, Hinkle G, et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell. 2006;124(6):1283–98.CrossRefPubMed
32.
go back to reference Sonenberg N, Hinnebusch AG. Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell Elsevier Inc. 2009;136(4):731–45. Sonenberg N, Hinnebusch AG. Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell Elsevier Inc. 2009;136(4):731–45.
33.
go back to reference Cea M, Cagnetta A, Patrone F, Nencioni A, Gobbi M, Anderson KC. Intracellular NAD(+) depletion induces autophagic death in multiple myeloma cells. Autophagy. 2013;9(3):410–2.CrossRefPubMedPubMedCentral Cea M, Cagnetta A, Patrone F, Nencioni A, Gobbi M, Anderson KC. Intracellular NAD(+) depletion induces autophagic death in multiple myeloma cells. Autophagy. 2013;9(3):410–2.CrossRefPubMedPubMedCentral
34.
go back to reference Shi Y, Yan H, Frost P, Gera J, Lichtenstein A. Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade. Mol Cancer Ther. 2005;4(10):1533–40.CrossRefPubMed Shi Y, Yan H, Frost P, Gera J, Lichtenstein A. Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade. Mol Cancer Ther. 2005;4(10):1533–40.CrossRefPubMed
35.
go back to reference Waskiewicz AJ, Flynn A, Proud CG, Cooper JA. Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J. 1997;16(8):1909–20.CrossRefPubMedPubMedCentral Waskiewicz AJ, Flynn A, Proud CG, Cooper JA. Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J. 1997;16(8):1909–20.CrossRefPubMedPubMedCentral
36.
go back to reference Krishnamoorthy T, Pavitt GD, Zhang F, Dever TE, Hinnebusch AG. Tight binding of the phosphorylated alpha subunit of initiation factor 2 (eIF2alpha) to the regulatory subunits of guanine nucleotide exchange factor eIF2B is required for inhibition of translation initiation. Mol Cell Biol. 2001;21(15):5018–30.CrossRefPubMedPubMedCentral Krishnamoorthy T, Pavitt GD, Zhang F, Dever TE, Hinnebusch AG. Tight binding of the phosphorylated alpha subunit of initiation factor 2 (eIF2alpha) to the regulatory subunits of guanine nucleotide exchange factor eIF2B is required for inhibition of translation initiation. Mol Cell Biol. 2001;21(15):5018–30.CrossRefPubMedPubMedCentral
37.
go back to reference Zhang C-S, Jiang B, Li M, Zhu M, Peng Y, Zhang Y-L, et al. The Lysosomal v-ATPase-Ragulator Complex Is a Common Activator for AMPK and mTORC1, Acting as a Switch between Catabolism and Anabolism. Cell Metab. 2014;20(3):526–40.CrossRefPubMed Zhang C-S, Jiang B, Li M, Zhu M, Peng Y, Zhang Y-L, et al. The Lysosomal v-ATPase-Ragulator Complex Is a Common Activator for AMPK and mTORC1, Acting as a Switch between Catabolism and Anabolism. Cell Metab. 2014;20(3):526–40.CrossRefPubMed
38.
go back to reference Liu X, Chhipa RR, Nakano I, Dasgupta B. The AMPK inhibitor compound C is a potent AMPK-independent antiglioma agent. Mol Cancer Ther. 2014;13(3):596–605.CrossRefPubMedPubMedCentral Liu X, Chhipa RR, Nakano I, Dasgupta B. The AMPK inhibitor compound C is a potent AMPK-independent antiglioma agent. Mol Cancer Ther. 2014;13(3):596–605.CrossRefPubMedPubMedCentral
40.
go back to reference Cagnetta A, Cea M, Calimeri T, Acharya C, Fulciniti M, Tai Y-T, et al. Intracellular NAD+ depletion enhances bortezomib-induced anti-myeloma activity. Blood. 2013;122(7):1243–55.CrossRefPubMedPubMedCentral Cagnetta A, Cea M, Calimeri T, Acharya C, Fulciniti M, Tai Y-T, et al. Intracellular NAD+ depletion enhances bortezomib-induced anti-myeloma activity. Blood. 2013;122(7):1243–55.CrossRefPubMedPubMedCentral
41.
go back to reference Dai RY, Zhao XF, Li JJ, Chen R, Luo ZL, Yu LX, et al. Implication of transcriptional repression in compound C-induced apoptosis in cancer cells. Cell Death Dis. 2013;4:e883.CrossRefPubMedPubMedCentral Dai RY, Zhao XF, Li JJ, Chen R, Luo ZL, Yu LX, et al. Implication of transcriptional repression in compound C-induced apoptosis in cancer cells. Cell Death Dis. 2013;4:e883.CrossRefPubMedPubMedCentral
42.
go back to reference Dagon Y, Avraham Y, Berry EM. AMPK activation regulates apoptosis, adipogenesis, and lipolysis by eIF2alpha in adipocytes. Biochem Biophys Res Commun. 2006;340(1):43–7.CrossRefPubMed Dagon Y, Avraham Y, Berry EM. AMPK activation regulates apoptosis, adipogenesis, and lipolysis by eIF2alpha in adipocytes. Biochem Biophys Res Commun. 2006;340(1):43–7.CrossRefPubMed
43.
go back to reference Shackelford DB, Abt E, Gerken L, Vasquez DS, Seki A, Leblanc M, et al. LKB1 inactivation dictates therapeutic response of non-small cell lung cancer to the metabolism drug phenformin. Cancer Cell. 2013;23(2):143–58.CrossRefPubMedPubMedCentral Shackelford DB, Abt E, Gerken L, Vasquez DS, Seki A, Leblanc M, et al. LKB1 inactivation dictates therapeutic response of non-small cell lung cancer to the metabolism drug phenformin. Cancer Cell. 2013;23(2):143–58.CrossRefPubMedPubMedCentral
44.
go back to reference Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A. 2004;101(10):3329–35.CrossRefPubMedPubMedCentral Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A. 2004;101(10):3329–35.CrossRefPubMedPubMedCentral
45.
go back to reference Mills JR, Hippo Y, Robert F, Chen SMH, Malina A, Lin C-J, et al. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci U S A. 2008;105(31):10853–8.CrossRefPubMedPubMedCentral Mills JR, Hippo Y, Robert F, Chen SMH, Malina A, Lin C-J, et al. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci U S A. 2008;105(31):10853–8.CrossRefPubMedPubMedCentral
46.
go back to reference Zhou T, Li G, Cao B, Liu L, Cheng Q, Kong H, et al. Downregulation of Mcl-1 through inhibition of translation contributes to benzyl isothiocyanate-induced cell cycle arrest and apoptosis in human leukemia cells. Cell Death Dis. 2013;4:e515.CrossRefPubMedPubMedCentral Zhou T, Li G, Cao B, Liu L, Cheng Q, Kong H, et al. Downregulation of Mcl-1 through inhibition of translation contributes to benzyl isothiocyanate-induced cell cycle arrest and apoptosis in human leukemia cells. Cell Death Dis. 2013;4:e515.CrossRefPubMedPubMedCentral
47.
go back to reference Fritsch RM, Schneider G, Saur D, Scheibel M, Schmid RM. Translational repression of MCL-1 couples stress-induced eIF2 alpha phosphorylation to mitochondrial apoptosis initiation. J Biol Chem. 2007;282(31):22551–62.CrossRefPubMed Fritsch RM, Schneider G, Saur D, Scheibel M, Schmid RM. Translational repression of MCL-1 couples stress-induced eIF2 alpha phosphorylation to mitochondrial apoptosis initiation. J Biol Chem. 2007;282(31):22551–62.CrossRefPubMed
48.
go back to reference Pradelli LA, Bénéteau M, Chauvin C, Jacquin MA, Marchetti S, Muñoz-Pinedo C, et al. Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation. Oncogene. 2010;29(11):1641–52.CrossRefPubMed Pradelli LA, Bénéteau M, Chauvin C, Jacquin MA, Marchetti S, Muñoz-Pinedo C, et al. Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation. Oncogene. 2010;29(11):1641–52.CrossRefPubMed
49.
go back to reference Palam LR, Baird TD, Wek RC. Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation. J Biol Chem. 2011;286(13):10939–49.CrossRefPubMedPubMedCentral Palam LR, Baird TD, Wek RC. Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation. J Biol Chem. 2011;286(13):10939–49.CrossRefPubMedPubMedCentral
50.
go back to reference Cagnetta A, Caffa I, Acharya C, Soncini D, Acharya P, Adamia S, et al. APO866 Increases Antitumor Activity of Cyclosporin-A by Inducing Mitochondrial and Endoplasmic Reticulum Stress in Leukemia Cells. Clin Cancer Res. 2015;21(17):3934–45.CrossRefPubMed Cagnetta A, Caffa I, Acharya C, Soncini D, Acharya P, Adamia S, et al. APO866 Increases Antitumor Activity of Cyclosporin-A by Inducing Mitochondrial and Endoplasmic Reticulum Stress in Leukemia Cells. Clin Cancer Res. 2015;21(17):3934–45.CrossRefPubMed
51.
go back to reference Fullwood MJ, Zhou W, Shenolikar S. Targeting phosphorylation of eukaryotic initiation factor-2α to treat human disease. Prog Mol Biol Transl Sci. 2012;106:75–106.CrossRefPubMed Fullwood MJ, Zhou W, Shenolikar S. Targeting phosphorylation of eukaryotic initiation factor-2α to treat human disease. Prog Mol Biol Transl Sci. 2012;106:75–106.CrossRefPubMed
52.
go back to reference Dowling RJO, Zakikhani M, Fantus IG, Pollak M, Sonenberg N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 2007;67(22):10804–12.CrossRefPubMed Dowling RJO, Zakikhani M, Fantus IG, Pollak M, Sonenberg N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 2007;67(22):10804–12.CrossRefPubMed
53.
go back to reference Larsson O, Morita M, Topisirovic I, Alain T, Blouin M-J, Pollak M, et al. Distinct perturbation of the translatome by the antidiabetic drug metformin. Proc Natl Acad Sci U S A. 2012;109(23):8977–82.CrossRefPubMedPubMedCentral Larsson O, Morita M, Topisirovic I, Alain T, Blouin M-J, Pollak M, et al. Distinct perturbation of the translatome by the antidiabetic drug metformin. Proc Natl Acad Sci U S A. 2012;109(23):8977–82.CrossRefPubMedPubMedCentral
Metadata
Title
EIF2A-dependent translational arrest protects leukemia cells from the energetic stress induced by NAMPT inhibition
Authors
Chiara Zucal
Vito G. D’Agostino
Antonio Casini
Barbara Mantelli
Natthakan Thongon
Debora Soncini
Irene Caffa
Michele Cea
Alberto Ballestrero
Alessandro Quattrone
Stefano Indraccolo
Alessio Nencioni
Alessandro Provenzani
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1845-1

Other articles of this Issue 1/2015

BMC Cancer 1/2015 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