Targeting glutamine metabolism in multiple myeloma enhances BIM binding to BCL-2 eliciting synthetic lethality to venetoclax

R Bajpai, S M Matulis, C Wei, A K Nooka, H E Von Hollen, S Lonial, L H Boise, M Shanmugam, R Bajpai, S M Matulis, C Wei, A K Nooka, H E Von Hollen, S Lonial, L H Boise, M Shanmugam

Abstract

Multiple myeloma (MM) is a plasma cell malignancy that is largely incurable due to development of resistance to therapy-elicited cell death. Nutrients are intricately connected to maintenance of cellular viability in part by inhibition of apoptosis. We were interested to determine if examination of metabolic regulation of BCL-2 proteins may provide insight on alternative routes to engage apoptosis. MM cells are reliant on glucose and glutamine and withdrawal of either nutrient is associated with varying levels of apoptosis. We and others have demonstrated that glucose maintains levels of key resistance-promoting BCL-2 family member, myeloid cell leukemic factor 1 (MCL-1). Cells continuing to survive in the absence of glucose or glutamine were found to maintain expression of MCL-1 but importantly induce pro-apoptotic BIM expression. One potential mechanism for continued survival despite induction of BIM could be due to binding and sequestration of BIM to alternate pro-survival BCL-2 members. Our investigation revealed that cells surviving glutamine withdrawal in particular, enhance expression and binding of BIM to BCL-2, consequently sensitizing these cells to the BH3 mimetic venetoclax. Glutamine deprivation-driven sensitization to venetoclax can be reversed by metabolic supplementation with TCA cycle intermediate α-ketoglutarate. Inhibition of glucose metabolism with the GLUT4 inhibitor ritonavir elicits variable cytotoxicity in MM that is marginally enhanced with venetoclax treatment, however, targeting glutamine metabolism with 6-diazo-5-oxo-l-norleucine uniformly sensitized MM cell lines and relapse/refractory patient samples to venetoclax. Our studies reveal a potent therapeutic strategy of metabolically driven synthetic lethality involving targeting glutamine metabolism for sensitization to venetoclax in MM.

Figures

Figure 1
Figure 1
MM cell lines and CD138+ MM primary patient cells are variably sensitive to glucose or glutamine deprivation. (a) MM cell lines and (b) patient cells were cultured in the presence of glucose (5 mm) and glutamine (2 mm) (Control) or in the absence of glucose (NG), absence of glutamine (NGlut) or absence of both nutrients (NG+NGlut) for 72 h (cell lines) or 48 h (patient cells). Cell viability was assessed by AnnexinV/DAPI staining and Flow cytometry. Data (a) is mean ± s.e.m. (n = 3). Control vs NG: P-value <0.001 (***) in all cell lines except U266 which is >0.05. Control vs NGlut: P-value <0.001 in all cell lines except U266 <0.1 (*) and L363 <0.01 (**).
Figure 2
Figure 2
Glucose or glutamine deprivation regulates expression of pro and anti-apoptotic BCL-2 proteins. MM cell lines cultured in the absence or presence of 5 mm glucose or 2 mm glutamine or both nutrients for 24 h were evaluated for expression of BCL-2 proteins. Cellular lysates were analyzed for expression of indicated proteins or GAPDH (loading control) by immunoblot analyses. One of two representative experiments is shown. NG, media without glucose; NGlut, media without glutamine.
Figure 3
Figure 3
Glucose or glutamine deprivation increase binding of pro-apoptotic BIM to BCL-2. L363 (a), KMS11 (b) and JJN3 cells (c) were cultured in the absence or presence of 5 mm glucose or 2 mm glutamine with or without (0.5 μm) ABT-199 for 24 h. Immunoprecipitates of MCL-1, BCL-2 and BCL-xL prepared from cellular lysates obtained from treated cells were evaluated for bound BIM, MCL-1, BCL-2 and BCL-xL by immunoblotting. Densitometric analysis of the BIM immunoblot was performed using IMAGE J software (imagej.nih.gov) and percent BIM bound to MCL-1, BCL-2 and BCL-xL presented in the corresponding stacked bar graph. NG, media without glucose; NGlut, media without glutamine. Representative blots from a minimum of three independent experiments are presented.
Figure 4
Figure 4
Glucose or glutamine deprivation sensitizes MM cell lines to the BH3 mimetic ABT-199. MM cell lines were cultured in the absence or presence of 5 mm glucose (a) or 2 mm glutamine (b) with or without the BH3 mimetic ABT-199 (0.5 μM) for 72 h. Cell viability was assessed by AnnexinV/DAPI staining and flow cytometry. NG, media without glucose; NGlut, media without glutamine. Data (a and b) are mean ± s.e.m. (n = 3). P-value of NG vs NG+ABT data is >0.05 (ns) in all cell lines except JJN3 and AMO1 where P-value is <0.001 (***). P-value of NGlut vs NGlut+ABT is <0.001 (***) in KMS18, L363 and U266, <0.01 (**) in MM.1S and JJN3, <0.1 (*) in KMS11 and >0.05 in RPMI-8226 and AMO1.
Figure 5
Figure 5
Metabolic supplementation with DMK reverses glutamine deprivation-elicited sensitivity to ABT-199. (a–d) MM cell lines AMO1 (a), KMS11 (b), MM.1S (c) and L363 (d) grown in absence or presence of glutamine treated with or without 0.5 μm ABT-199 were supplemented with indicated concentrations of dimethyl alpha ketoglutarate (DMK) for 18–24 h. Cells were harvested and evaluated for viability by AnnexinV/DAPI staining and flow cytometry. (e) Cellular lysates prepared from treated cells from d were evaluated for BIM, MCL-1, BCL-2 and BCL-xL protein expression with GAPDH evaluated as a loading control. (f) Cells from d were utilized for co-IP of MCL-1, BCL-xL and BCL-2 to evaluate BIM binding. MCL-1, BCL-xL and BCL-2 protein levels in IPs are also evaluated as loading controls. (g) L363 cells grown in presence or absence of DON treated with or without ABT-199 for 18 h were subjected to co-IP of MCL-1, BCL-xL and BCL-2 to evaluate BIM binding. Distribution of BIM binding is presented in bar graph to the right of f and g. One of two representative co-IP results is presented in f and g. NG, media without glucose; NGlut, media without glutamine. Data (a–d) is mean ± s.e.m. of n ≥ 3; (e–g) co-IPs and immunoblots are representative of two independent experiments. P-value of NGlut+ABT vs NGlut+ABT+DMK in a–d is <0.0001 in KMS11 (b), MM.1S (c) and L363 (d) indicated as ‘****’ and P<0.01 in AMO1 (a) indicated as ‘**’.
Figure 6
Figure 6
Evaluation of Ritonavir (targeting glucose transport) or DON targeting glutamine to ABT-199 sensitivity in MM. MM cell lines were cultured with 40 μm ritonavir (a) or 0.1 mm DON (c) with or without ABT-199 (0.5 μm) or in combination for 72 h. MM patient samples were cultured with ritonavir (20 μm) (b) or DON (1 mm or 0.1 mm), as indicated. (d) for 48 h with viability assessed by AnnexinV/DAPI staining. (e) Model showing the impact of glutamine deprivation on BCL-2 family members and the basis for synthetic lethality to ABT-199. This model is based on the L363 co-IP under glutamine deprivation. Data in a are mean ± s.e.m. (n = 3) and b are mean ± s.e.m. (n = 2). P-values of RIT vs RIT+ABT: >0.05 in RPMI-8226, KMS18, KMS11 and U266, <0.1 in AMO1, <0.01 in JJN3, <0.001 in L363 and MM.1S. P-value of DON vs DON+ABT is <0.001 in all cell lines except RPMI-8226. co-IP, co-immunoprecipitation.

References

    1. Palumbo A, Anderson K. Multiple myeloma. N Engl J Med. 2011;364:1046–1060.
    1. Barlogie B, Mitchell A, van Rhee F, Epstein J, Morgan GJ, Crowley J. Curing myeloma at last: defining criteria and providing the evidence. Blood. 2014;124:3043–3051.
    1. Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell. 2004;116:205–219.
    1. Cheng EH, Wei MC, Weiler S, Flavell RA, Mak TW, Lindsten T, et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol Cell. 2001;8:705–711.
    1. Ni Chonghaile T, Letai A. Mimicking the BH3 domain to kill cancer cells. Oncogene. 2008;27(Suppl 1):S149–S157.
    1. Podar K, Gouill SL, Zhang J, Opferman JT, Zorn E, Tai YT, et al. A pivotal role for Mcl-1 in Bortezomib-induced apoptosis. Oncogene. 2008;27:721–731.
    1. Mills JR, Hippo Y, Robert F, Chen SM, Malina A, Lin CJ, et al. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci USA. 2008;105:10853–10858.
    1. Eguchi T, Itadani H, Shimomura T, Kawanishi N, Hirai H, Kotani H. Expression levels of p18INK4C modify the cellular efficacy of cyclin-dependent kinase inhibitors via regulation of Mcl-1 expression in tumor cell lines. Mol Cancer Ther. 2009;8:1460–1472.
    1. Nguyen M, Marcellus RC, Roulston A, Watson M, Serfass L, Murthy Madiraju SR, et al. Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci USA. 2007;104:19512–19517.
    1. Pradelli LA, Beneteau M, Chauvin C, Jacquin MA, Marchetti S, Munoz-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. 2009;29:1641–1652.
    1. Liu Z, Xu J, He J, Zheng Y, Li H, Lu Y, et al. A critical role of autocrine sonic hedgehog signaling in human CD138+ myeloma cell survival and drug resistance. Blood. 2014;124:2061–2071.
    1. Pepper C, Hoy T, Bentley DP. Bcl-2/Bax ratios in chronic lymphocytic leukaemia and their correlation with in vitro apoptosis and clinical resistance. Br J Cancer. 1997;76:935–938.
    1. Ni Chonghaile T, Sarosiek KA, Vo TT, Ryan JA, Tammareddi A, Moore Vdel G, et al. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science. 2011;334:1129–1133.
    1. Bartel TB, Haessler J, Brown TL, Shaughnessy JD, Jr, van Rhee F, Anaissie E, et al. F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma. Blood. 2009;114:2068–2076.
    1. Roberts RS, Hsu HW, Lin KD, Yang TJ. Amino acid metabolism of myeloma cells in culture. J Cell Sci. 1976;21:609–615.
    1. Dalva-Aydemir S, Bajpai R, Martinez M, Adekola KU, Kandela I, Wei C, et al. Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin. Clin Cancer Res. 2015;21:1161–1171.
    1. Graham NA, Tahmasian M, Kohli B, Komisopoulou E, Zhu M, Vivanco I, et al. Glucose deprivation activates a metabolic and signaling amplification loop leading to cell death. Mol Syst Biol. 2012;8:589.
    1. Coloff JL, Mason EF, Altman BJ, Gerriets VA, Liu T, Nichols AN, et al. Akt requires glucose metabolism to suppress puma expression and prevent apoptosis of leukemic T cells. J Biol Chem. 2011;286:5921–5933.
    1. Zhao Y, Altman BJ, Coloff JL, Herman CE, Jacobs SR, Wieman HL, et al. Glycogen synthase kinase 3alpha and 3beta mediate a glucose-sensitive antiapoptotic signaling pathway to stabilize Mcl-1. Mol Cell Biol. 2007;27:4328–4339.
    1. Danial NN, Gramm CF, Scorrano L, Zhang CY, Krauss S, Ranger AM, et al. BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature. 2003;424:952–956.
    1. Rathmell JC, Fox CJ, Plas DR, Hammerman PS, Cinalli RM, Thompson CB. Akt-directed glucose metabolism can prevent Bax conformation change and promote growth factor-independent survival. Mol Cell Biol. 2003;23:7315–7328.
    1. Zhang B, Gojo I, Fenton RG. Myeloid cell factor-1 is a critical survival factor for multiple myeloma. Blood. 2002;99:1885–1893.
    1. Derenne S, Monia B, Dean NM, Taylor JK, Rapp MJ, Harousseau JL, et al. Antisense strategy shows that Mcl-1 rather than Bcl-2 or Bcl-x(L) is an essential survival protein of human myeloma cells. Blood. 2002;100:194–199.
    1. Morales AA, Kurtoglu M, Matulis SM, Liu J, Siefker D, Gutman DM, et al. Distribution of Bim determines Mcl-1 dependence or codependence with Bcl-xL/Bcl-2 in Mcl-1-expressing myeloma cells. Blood. 2011;118:1329–1339.
    1. McBrayer SK, Cheng JC, Singhal S, Krett NL, Rosen ST, Shanmugam M. Multiple myeloma exhibits novel dependence on GLUT4, GLUT8, and GLUT11: implications for glucose transporter-directed therapy. Blood. 2012;119:4686–4697.
    1. Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013;19:202–208.
    1. Wensveen FM, van Gisbergen KP, Derks IA, Gerlach C, Schumacher TN, van Lier RA, et al. Apoptosis threshold set by Noxa and Mcl-1 after T cell activation regulates competitive selection of high-affinity clones. Immunity. 2010;32:754–765.
    1. Nakajima W, Hicks MA, Tanaka N, Krystal GW, Harada H. Noxa determines localization and stability of MCL-1 and consequently ABT-737 sensitivity in small cell lung cancer. Cell Death Dis. 2014;5:e1052.
    1. Vyas AK, Koster JC, Tzekov A, Hruz PW. Effects of the HIV protease inhibitor ritonavir on GLUT4 knock-out mice. J Biol Chem. 2010;285:36395–36400.
    1. Shapiro RA, Clark VM, Curthoys NP. Inactivation of rat renal phosphate-dependent glutaminase with 6-diazo-5-oxo-L-norleucine. Evidence for interaction at the glutamine binding site. J Biol Chem. 1979;254:2835–2838.
    1. Brown G, Singer A, Proudfoot M, Skarina T, Kim Y, Chang C, et al. Functional and structural characterization of four glutaminases from Escherichia coli and Bacillus subtilis. Biochemistry. 2008;47:5724–5735.
    1. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2:183–192.
    1. Lessene G, Czabotar PE, Colman PM. BCL-2 family antagonists for cancer therapy. Nat Rev Drug Discov. 2008;7:989–1000.
    1. van Delft MF, Wei AH, Mason KD, Vandenberg CJ, Chen L, Czabotar PE, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell. 2006;10:389–399.
    1. Wilson WH, O'Connor OA, Czuczman MS, LaCasce AS, Gerecitano JF, Leonard JP, et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol. 2010;11:1149–1159.
    1. Schoenwaelder SM, Jarman KE, Gardiner EE, Hua M, Qiao J, White MJ, et al. Bcl-xL-inhibitory BH3 mimetics can induce a transient thrombocytopathy that undermines the hemostatic function of platelets. Blood. 2011;118:1663–1674.
    1. Touzeau C, Dousset C, Le Gouill S, Sampath D, Leverson JD, Souers AJ, et al. The Bcl-2 specific BH3 mimetic ABT-199: a promising targeted therapy for t(11;14) multiple myeloma. Leukemia. 2014;28:210–212.
    1. Olsen RR, Mary-Sinclair MN, Yin Z, Freeman KW. Antagonizing Bcl-2 family members sensitizes neuroblastoma and Ewing's sarcoma to an inhibitor of glutamine metabolism. PLoS One. 2015;10:e0116998.
    1. Leverson JD, Zhang H, Chen J, Tahir SK, Phillips DC, Xue J, et al. Potent and selective small-molecule MCL-1 inhibitors demonstrate on-target cancer cell killing activity as single agents and in combination with ABT-263 (navitoclax). Cell Death Dis. 2015;6:e1590.
    1. Thomas RL, Roberts DJ, Kubli DA, Lee Y, Quinsay MN, Owens JB, et al. Loss of MCL-1 leads to impaired autophagy and rapid development of heart failure. Genes Dev. 2013;27:1365–1377.
    1. Tuffy LP, Concannon CG, D'Orsi B, King MA, Woods I, Huber HJ, et al. Characterization of Puma-dependent and Puma-independent neuronal cell death pathways following prolonged proteasomal inhibition. Mol Cell Biol. 2010;30:5484–5501.
    1. Puthalakath H, Huang DC, O'Reilly LA, King SM, Strasser A. The proapoptotic activity of the Bcl-2 family member Bim is regulated by interaction with the dynein motor complex. Mol Cell. 1999;3:287–296.
    1. Lei K, Davis RJ. JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc Natl Acad Sci USA. 2003;100:2432–2437.
    1. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–105.
    1. Fabrizio Accardi MC, Bolzoni M, Storti P, Todoerti K, Agnelli L, Ferrari M, et al. Ammonium production and glutamine-addiction of myeloma cells: new attractive targets in multiple myeloma. Blood. 2014;124:2067.
    1. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901.
    1. Prideaux SM, Conway O'Brien E, Chevassut TJ. The genetic architecture of multiple myeloma. Adv Hematol. 2014;2014:864058.
    1. Shanmugam M, McBrayer SK, Qian J, Raikoff K, Avram MJ, Singhal S, et al. Targeting glucose consumption and autophagy in myeloma with the novel nucleoside analogue 8-aminoadenosine. J Biol Chem. 2009;284:26816–26830.
    1. Vo TT, Ryan J, Carrasco R, Neuberg D, Rossi DJ, Stone RM, et al. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell. 2012;151:344–355.
    1. Boise LH, Minn AJ, Noel PJ, June CH, Accavitti MA, Lindsten T, et al. CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL. Immunity. 1995;3:87–98.
    1. Mishra RK, Wei C, Hresko RC, Bajpai R, Heitmeier M, Matulis SM, et al. In silico modeling-based identification of glucose transporter 4 (GLUT4)-selective inhibitors for cancer therapy. J Biol Chem. 2015;290:14441–14453.

Source: PubMed

3
Předplatit