Metformin induces apoptosis through AMPK-dependent inhibition of UPR signaling in ALL lymphoblasts

Gilles M Leclerc, Guy J Leclerc, Jeffim N Kuznetsov, Joanna DeSalvo, Julio C Barredo, Gilles M Leclerc, Guy J Leclerc, Jeffim N Kuznetsov, Joanna DeSalvo, Julio C Barredo

Abstract

The outcome of patients with resistant phenotypes of acute lymphoblastic leukemia (ALL) or those who relapse remains poor. We investigated the mechanism of cell death induced by metformin in Bp- and T-ALL cell models and primary cells, and show that metformin effectively induces apoptosis in ALL cells. Metformin activated AMPK, down-regulated the unfolded protein response (UPR) demonstrated by significant decrease in the main UPR regulator GRP78, and led to UPR-mediated cell death via up-regulation of the ER stress/UPR cell death mediators IRE1α and CHOP. Using shRNA, we demonstrate that metformin-induced apoptosis is AMPK-dependent since AMPK knock-down rescued ALL cells, which correlated with down-regulation of IRE1α and CHOP and restoration of the UPR/GRP78 function. Additionally rapamycin, a known inhibitor of mTOR-dependent protein synthesis, rescued cells from metformin-induced apoptosis and down-regulated CHOP expression. Finally, metformin induced PIM-2 kinase activity and co-treatment of ALL cells with a PIM-1/2 kinase inhibitor plus metformin synergistically increased cell death, suggesting a buffering role for PIM-2 in metformin's cytotoxicity. Similar synergism was seen with agents targeting Akt in combination with metformin, supporting our original postulate that AMPK and Akt exert opposite regulatory roles on UPR activity in ALL. Taken together, our data indicate that metformin induces ALL cell death by triggering ER and proteotoxic stress and simultaneously down-regulating the physiologic UPR response responsible for effectively buffering proteotoxic stress. Our findings provide evidence for a role of metformin in ALL therapy and support strategies targeting synthetic lethal interactions with Akt and PIM kinases as suitable for future consideration for clinical translation in ALL.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Metformin induces cell growth arrest…
Figure 1. Metformin induces cell growth arrest and apoptosis in ALL cell lines.
Growth inhibition (A) and apoptosis (B) in CCRF-CEM and NALM6 cells treated with metformin (MET, 1, 2, and 5 mM) for 48 h. Apoptosis (C) in T-ALL (CCRF-CEM, Jurkat, and primary T-ALL) and Bp-ALL (NALM6, REH, and primary Bp-ALL) cells treated with metformin (MET, 5 mM) for 48 h. Growth inhibition was expressed relative to control values (mean ±SEM, n = 3). * and # denote p < 0.001 and p < 0.0001 for metformin vs. CTRL, respectively.
Figure 2. Metformin activates AMPK, Akt, and…
Figure 2. Metformin activates AMPK, Akt, and UPR signaling pathway proteins in ALL primary and cell line models.
A) Western blot analysis of proteins associated with AMPK, and Akt/mTOR signaling pathways in CCRF-CEM and NALM6 cells treated with metformin (MET, 2.5 and 5.0 mM) for 48 h. The density value of each band was normalized to β-actin level and expressed relative to control (shown as fold induction). B) Immunoblotting of UPR signaling factors in CCRF-CEM and NALM6 cells treated with metformin (MET, 2.5 and 5.0 mM) for 48 h. C) Western blots of AMPK, Akt/mTOR and UPR signaling proteins in representative sample of primary T- and Bp-ALL cells treated with metformin (MET, 10 mM) for 24 h. D) Western blot analysis of GRP78 expression in CCRF-CEM and NALM6 cells treated with metformin (MET, 10mM) and tunicamycin (TUN, 2.5 µg/ml for NALM6; 5.0 µg/ml for CCRF-CEM), either alone or in combination for 48 h.
Figure 3. Inhibition of mTOR-dependent protein synthesis…
Figure 3. Inhibition of mTOR-dependent protein synthesis reverses metformin-induced cell death.
A) Apoptosis in CCRF-CEM cells expressing either scramble shRNA (shCTRL) or shRNA against AMPKα1 (shAMPK) treated with metformin (MET, 10 mM) for 48 h. B) Immunoblotting of AMPK, Akt/mTOR, and ER stress/UPR signaling pathway proteins in the cells described in (A). C) Cell death (upper panel) in CCRF-CEM and NALM6 cells treated with metformin (MET, 5 and 10 mM) and rapamycin (RAPA; 0.1 µg/mL), either alone or in combination for 48 h. A statistical value of p < 0.01 was obtained for MET + RAPA vs. either control or each agent alone. The cell death was expressed as a percentage (%) of cells in the population (mean ±SEM, n = 3). Western blot analysis (lower panel) of p-4EBP1 (T70), IRE1α, and CHOP expression in the CCRF-CEM and NALM6 cells treated with metformin plus rapamycin described in the upper panel.
Figure 4. Metformin induces expression of PIM-2…
Figure 4. Metformin induces expression of PIM-2 in ALL cells.
Western blot analysis of PIM-2, p-BAD (S112) and p-AMPK (T172) expression in CCRF-CEM and NALM6 cells treated with metformin (MET, 5 and 10 mM) for 72 h at 37°C.
Figure 5. Inhibition of PIM-2 and Akt…
Figure 5. Inhibition of PIM-2 and Akt kinases synergistically sensitizes ALL cells to metformin.
A) Cell death in CCRF-CEM and NALM6 cells treated with metformin (MET, 4 mM) and the PIM-1/2 kinase inhibitor V (PKI; 80 µM), either alone or in combination for 72 h at 37°C. The CI values of 0.27 and 0.28 indicate synergism. B) Immunoblotting of p-ACC (S79), GRP78, and PIM-2 expression in the CCRF-CEM and NALM6 cells treated with MET plus PKI described in (A). C) Cell death in NALM6 cells treated with metformin (MET, 5.0 mM) and Akt inhibitor X (AIX; 5 µM) or perifosine (PER; 6 µM), either alone or in combination for 72 h at 37°C. CI values of 0.19 (MET + AIX) and 0.21 (MET + PER) indicate synergism. The cell death values were expressed as a percentage (%) of cells in the population (mean ±SEM, n = 3). D) Immunoblotting of AMPK/ACC, Akt/mTOR, and UPR signaling pathway proteins in the NALM6 cells treated with MET (5.0 mM) plus AIX (5 µM) described in (C).
Figure 6. Proposed mechanism of action for…
Figure 6. Proposed mechanism of action for metformin in ALL cells.
Metformin induces metabolic stress by decreasing the ATP: AMP ratio, which leads to activation of AMPK, and increased level of unfolded/misfolded proteins in the ER lumen. The inability of ALL cells to engage the UPR caused by AMPK-dependent down-regulation of GRP78 leads to ER stress/UPR mediated cell death. The survival PIM and Akt kinases are expressed as compensatory survival mechanisms in response to metformin’s cytotoxicity to down-regulate AMPK allowing cells to effectively engage UPR and process the ER stress.

References

    1. Siegel R, Naishadham D, Jemal A (2012) Cancer statistics, 2012. CA Cancer J Clin 62: 10-29. doi:. PubMed: .
    1. Pui CH, Mullighan CG, Evans WE, Relling MV (2012) Pediatric acute lymphoblastic leukemia: where are we going and how do we get there? Blood 120: 1165-1174. doi:. PubMed: .
    1. Hardie DG (2011) AMP-activated protein kinase: a cellular energy sensor with a key role in metabolic disorders and in cancer. Biochem Soc Trans 39: 1-13. doi:. PubMed: .
    1. Sengupta TK, Leclerc GM, Hsieh-Kinser TT, Leclerc GJ, Singh I et al. (2007) Cytotoxic effect of 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR) on childhood acute lymphoblastic leukemia (ALL) cells: implication for targeted therapy. Mol Cancer 6: 46. doi:. PubMed: .
    1. Carling D, Mayer FV, Sanders MJ, Gamblin SJ (2011) AMP-activated protein kinase: nature’s energy sensor. Nat Chem Biol 7: 512-518. doi:. PubMed: .
    1. Fay JR, Steele V, Crowell JA (2009) Energy homeostasis and cancer prevention: the AMP-activated protein kinase. Cancer Prev Res (Phila) 2: 301-309. doi:. PubMed: .
    1. Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L et al. (2003) Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J Biol 2: 28. doi:. PubMed: .
    1. Steinberg GR, Kemp BE (2009) AMPK in Health and Disease. Physiol Rev 89: 1025-1078. doi:. PubMed: .
    1. Bolen S, Feldman L, Vassy J, Wilson L, Yeh HC et al. (2007) Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 147: 386-399. doi:. PubMed: .
    1. Ouyang J, Parakhia RA, Ochs RS (2011) Metformin activates AMP kinase through inhibition of AMP deaminase. J Biol Chem 286: 1-11. doi:. PubMed: .
    1. Owen MR, Doran E, Halestrap AP (2000) Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J 348 3: 607-614. doi:. PubMed: .
    1. Kourelis TV, Siegel RD (2012) Metformin and cancer: new applications for an old drug. Med Oncol 29: 1314-1327. doi:. PubMed: .
    1. Ben Sahra I, Le Marchand-Brustel Y, Tanti JF, Bost F (2010) Metformin in cancer therapy: a new perspective for an old antidiabetic drug? Mol Cancer Ther 9: 1092-1099. doi:. PubMed: .
    1. Pollak M (2010) Metformin and other biguanides in oncology: advancing the research agenda. Cancer Prev Res (Phila) 3: 1060-1065. doi:. PubMed: .
    1. Kuznetsov JN, Leclerc GJ, Leclerc GM, Barredo JC (2011) AMPK and Akt Determine Apoptotic Cell Death following Perturbations of One-Carbon Metabolism by Regulating ER Stress in Acute Lymphoblastic Leukemia. Mol Cancer Ther 10: 437-447. doi:. PubMed: .
    1. Terai K, Hiramoto Y, Masaki M, Sugiyama S, Kuroda T et al. (2005) AMP-activated protein kinase protects cardiomyocytes against hypoxic injury through attenuation of endoplasmic reticulum stress. Mol Cell Biol 25: 9554-9575. doi:. PubMed: .
    1. Yeh CH, Chen TP, Wang YC, Lin YM, Fang SW (2010) AMP-activated protein kinase activation during cardioplegia-induced hypoxia/reoxygenation injury attenuates cardiomyocytic apoptosis via reduction of endoplasmic reticulum stress. Mediat Inflamm, 2010: 2010: . PubMed: 21318153
    1. Dong Y, Zhang M, Wang S, Liang B, Zhao Z et al. (2010) Activation of AMP-activated protein kinase inhibits oxidized LDL-triggered endoplasmic reticulum stress in vivo. Diabetes 59: 1386-1396. doi:. PubMed: .
    1. DeSalvo J, Kuznetsov JN, Du J, Leclerc GM, Leclerc GJ et al. (2012) Inhibition of Akt potentiates 2-DG-induced apoptosis via downregulation of UPR in acute lymphoblastic leukemia. Mol Cancer Res 10: 969-978. doi:. PubMed: .
    1. Woehlbier U, Hetz C (2011) Modulating stress responses by the UPRosome: a matter of life and death. Trends Biochem Sci 36: 329-337. doi:. PubMed: .
    1. Kim I, Xu W, Reed JC (2008) Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat Rev Drug Discov 7: 1013-1030. doi:. PubMed: .
    1. Boyce M, Yuan J (2006) Cellular response to endoplasmic reticulum stress: a matter of life or death. Cell Death Differ 13: 363-373. doi:. PubMed: .
    1. Hu P, Han Z, Couvillon AD, Exton JH (2004) Critical role of endogenous Akt/IAPs and MEK1/ERK pathways in counteracting endoplasmic reticulum stress-induced cell death. J Biol Chem 279: 49420-49429. doi:. PubMed: .
    1. Pfaffenbach KT, Lee AS (2011) The critical role of GRP78 in physiologic and pathologic stress. Curr Opin Cell Biol 23: 150-156. doi:. PubMed: .
    1. Shore GC, Papa FR, Oakes SA (2011) Signaling cell death from the endoplasmic reticulum stress response. Curr Opin Cell Biol 23: 143-149. doi:. PubMed: .
    1. Kim I, Shu CW, Xu W, Shiau CW, Grant D et al. (2009) Chemical biology investigation of cell death pathways activated by endoplasmic reticulum stress reveals cytoprotective modulators of ASK1. J Biol Chem 284: 1593-1603. PubMed: .
    1. Rutkowski DT, Kaufman RJ (2007) That which does not kill me makes me stronger: adapting to chronic ER stress. Trends Biochem Sci 32: 469-476. doi:. PubMed: .
    1. Yecies JL, Manning BD (2011) Transcriptional control of cellular metabolism by mTOR signaling. Cancer Res 71: 2815-2820. doi:. PubMed: .
    1. Korets SB, Czok S, Blank SV, Curtin JP, Schneider RJ (2011) Targeting the mTOR/4E-BP pathway in endometrial cancer. Clin Cancer Res 17: 7518-7528. doi:. PubMed: .
    1. Tamburini J, Green AS, Bardet V, Chapuis N, Park S et al. (2009) Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia. Blood 114: 1618-1627. doi:. PubMed: .
    1. Beharry Z, Mahajan S, Zemskova M, Lin YW, Tholanikunnel BG et al. (2011) The Pim protein kinases regulate energy metabolism and cell growth. Proc Natl Acad Sci U S A 108: 528-533. doi:. PubMed: .
    1. Amaravadi R, Thompson CB (2005) The survival kinases Akt and Pim as potential pharmacological targets. J Clin Invest 115: 2618-2624. doi:. PubMed: .
    1. Mikkers H, Nawijn M, Allen J, Brouwers C, Verhoeven E et al. (2004) Mice deficient for all PIM kinases display reduced body size and impaired responses to hematopoietic growth factors. Mol Cell Biol 24: 6104-6115. doi:. PubMed: .
    1. Cohen AM, Grinblat B, Bessler H, Kristt D, Kremer A et al. (2004) Increased expression of the hPim-2 gene in human chronic lymphocytic leukemia and non-Hodgkin lymphoma. Leuk Lymphoma 45: 951-955. doi:. PubMed: .
    1. Nawijn MC, Alendar A, Berns A (2011) For better or for worse: the role of Pim oncogenes in tumorigenesis. Nat Rev Cancer 11: 23-34. doi:. PubMed: .
    1. Leclerc GM, Leclerc GJ, Fu G, Barredo JC (2010) AMPK-induced activation of Akt by AICAR is mediated by IGF-1R dependent and independent mechanisms in acute lymphoblastic leukemia. J Mol Signal 5: 15. doi:. PubMed: .
    1. Chou TC (2006) Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 58: 621-681. doi:. PubMed: .
    1. Thériault JR, Palmer HJ, Pittman DD (2011) Inhibition of the Unfolded Protein Response by metformin in renal proximal tubular epithelial cells. Biochem Biophys Res Commun 409: 500-505. doi:. PubMed: .
    1. Saito S, Furuno A, Sakurai J, Sakamoto A, Park HR et al. (2009) Chemical genomics identifies the unfolded protein response as a target for selective cancer cell killing during glucose deprivation. Cancer Res 69: 4225-4234. doi:. PubMed: .
    1. Oyadomari S, Mori M (2004) Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ 11: 381-389. doi:. PubMed: .
    1. Schönthal AH (2013) Pharmacological targeting of endoplasmic reticulum stress signaling in cancer. Biochem Pharmacol 85: 653-666. doi:. PubMed: .
    1. Benjamin D, Colombi M, Moroni C, Hall MN (2011) Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov 10: 868-880. doi:. PubMed: .
    1. Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LG et al. (2003) LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol 13: 2004-2008. doi:. PubMed: .
    1. Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J et al. (2005) Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab 2: 9-19. doi:. PubMed: .
    1. Hammerman PS, Fox CJ, Birnbaum MJ, Thompson CB (2005) Pim and Akt oncogenes are independent regulators of hematopoietic cell growth and survival. Blood 105: 4477-4483. doi:. PubMed: .
    1. Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ (1996) Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X. Cell Volumes L: 87: 619-628
    1. El-Mir MY, Nogueira V, Fontaine E, Avéret N, Rigoulet M et al. (2000) Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J Biol Chem 275: 223-228. doi:. PubMed: .
    1. Xu C, Bailly-Maitre B, Reed JC (2005) Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest 115: 2656-2664. doi:. PubMed: .
    1. Matsuo J, Tsukumo Y, Saito S, Tsukahara S, Sakurai J et al. (2012) Hyperactivation of 4E-binding protein 1 as a mediator of biguanide-induced cytotoxicity during glucose deprivation. Mol Cancer Ther 11: 1082-1091. doi:. PubMed: .
    1. Grimaldi C, Chiarini F, Tabellini G, Ricci F, Tazzari PL et al. (2012) AMP-dependent kinase/mammalian target of rapamycin complex 1 signaling in T-cell acute lymphoblastic leukemia: therapeutic implications. Leukemia 26: 91-100. doi:. PubMed: .
    1. Kovacic S, Soltys CL, Barr AJ, Shiojima I, Walsh K et al. (2003) Akt activity negatively regulates phosphorylation of AMP-activated protein kinase in the heart. J Biol Chem 278: 39422-39427. doi:. PubMed: .

Source: PubMed

3
Subscribe