Pharmacologic screens reveal metformin that suppresses GRP78-dependent autophagy to enhance the anti-myeloma effect of bortezomib

S Jagannathan, M A Y Abdel-Malek, E Malek, N Vad, T Latif, K C Anderson, J J Driscoll, S Jagannathan, M A Y Abdel-Malek, E Malek, N Vad, T Latif, K C Anderson, J J Driscoll

Abstract

Although the therapeutic benefit of proteasome inhibition in multiple myeloma remains unchallenged, drug resistance inevitably emerges through mechanisms that remain elusive. Bortezomib provokes unwanted protein accumulation and the endoplasmic reticulum stress to activate the unfolded protein response (UPR) and autophagy as compensatory mechanisms that restore protein homeostasis. High-throughput screens to detect pharmacologics that modulated autophagy to enhance the anti-myeloma effect of bortezomib revealed metformin, a widely used antidiabetic agent with proven efficacy and limited adverse effects. Metformin co-treatment with bortezomib suppressed induction of the critical UPR effector glucose-regulated protein 78 (GRP78) to impair autophagosome formation and enhance apoptosis. Gene expression profiling of newly diagnosed myeloma patient tumors further correlated the hyperexpression of GRP78-encoding HSPA5 with reduced clinical response to bortezomib. The effect of bortezomib was enhanced with metformin co-treatment using myeloma patient tumor cells and the chemoresistant, stem cell-like side population that may contribute to disease recurrence. The relevance of the findings was confirmed in vivo as shown by metformin co-treatment with bortezomib that delayed the growth of myeloma xenotransplants. Taken together, our results suggest that metformin suppresses GRP78, a key driver of bortezomib-induced autophagy, and support the pharmacologic repositioning of metformin to enhance the anti-myeloma benefit of bortezomib.

Figures

Figure 1
Figure 1
Effect of proteasome inhibitors on myeloma SP and MP viability, proliferation and induction of apoptosis. (a) Representative flow cytometric dot plots of SP analysis. Dot plots show RPMI8226 cells incubated in Hoechst 33342 alone (left), Hoechst 33342 accumulation in the presence of 100 μM verapamil (middle) and Hoechst 33342 accumulation in the presence of 50 μM reserpine (right). Abscissa is Hoechst red fluorescence intensity and ordinate is Hoechst blue fluorescence intensity with the gate representing the SP fraction. (b) Effect of bortezomib, carfilzomib or ixazomib at indicated concentrations on SP and MP viability. SP and MP were isolated from RPMI8226 cells and viability determined using the XTT assay. Error bars represent s.d. values determined from triplicate measurements. (c) Effect of bortezomib, carfilzomib and ixazomib on SP or MP cells isolated from MM patients. Cells were incubated with drugs as indicated and viability determined using the XTT assay. Error bars represent s.d. values determined from triplicate measurements. (d) Effect of bortezomib on SP and MP cell proliferation. Cells were incubated with bortezomib at indicated concentrations for 8 h and BrdU then added for 2 h. BrdU incorporation was determined relative to untreated MP cells. Error bars represent s.d. values determined from triplicate measurements. (e) Effect of bortezomib on the generation of annexin-positive SP and MP cells. Cells were incubated with bortezomib as indicated for 24 h and the percentage of annexin-positive cells determined relative to untreated SP or MP cells. Shown is the percentage increase in annexin-V-positive cells as determined by flow cytometry. Error bars represent s.d. values from triplicate measurements.
Figure 2
Figure 2
Effect of metformin co-treatment with bortezomib on myeloma SP and MP cell viability, proliferation and induction of apoptosis. (a) Dose-dependent effect of bortezomib alone or combined with metformin (500 μM) on the viability of SP and MP isolated from RPMI8226 cells. SP and MP cells were incubated with bortezomib alone or combined with metformin (500 μM). Cell viability was determined using the XTT assay. Error bars represent s.d. values determined from triplicate measurements. (b) Dose-dependent effect of bortezomib added at the indicated concentrations alone or combined with metformin (500 μM) on myeloma patient's SP and MP cell viability. (c) Dose-dependent effect of bortezomib added at indicated concentrations on colony formation. Values were normalized relative to the number of colonies formed from untreated SP. (d) Dose-dependent effect of bortezomib added at indicated concentrations alone or combined with metformin on colony formation. Values were normalized relative to the number of colonies detected with untreated SP cells. (e) Effect of bortezomib on SP and MP cell proliferation. Cells were incubated with bortezomib for 8 h at indicated concentrations and BrdU then added for 2 h. BrdU incorporation was determined relative to untreated MP cells. Error bars represent s.d. values determined from triplicate measurements. (f) Effect of bortezomib on the percentage of annexin-positive SP and MP cells. Cells were incubated with bortezomib at indicated concentrations for 24 h. The relative percentage increase in annexin-positive cells was determined relative to untreated SP or MP cells. Shown is the percentage increase in annexin-positive cells determined by flow cytometry. Error bars represent s.d. values determined from triplicate measurements.
Figure 3
Figure 3
Effect of metformin on GRP78 and bortezomib-induced autophagy. (a) Western blot to determine the effect of bortezomib treatment on GRP78 levels in RPMI8226 cell lysates. Cells were treated with bortezomib at indicated concentrations for 24 h, lysates prepared and probed using a GRP78-specific antibody. (b) Western blot to determine the effect of metformin co-treatment with bortezomib on GRP78 levels in RPMI8226 cells. Cells were treated with bortezomib and metformin at indicated concentrations for 24 h; lysates prepared and probed using an antibody specific to GRP78. (c) Western blot to determine the effect of metformin co-treatment with bortezomib on GRP78 levels in SP and MP cells isolated from RPMI8226. Cells were treated with bortezomib and metformin at indicated concentrations for 24 h; lysates prepared and probed using an antibody specific to GRP78. (d) Effect of metformin co-treatment with bortezomib on the conversion of LC3B-I to LC3B-II in RPMI8226 cells. Cells were treated with bortezomib (5 nM), metformin (500 μM) for 24 h. Bafilomycin A1 (100 nM), a specific inhibitor of the vacuolar type H+-ATPase was then added for the final 4 h of incubation to inhibit autophagy flux and to prevent autophagosome recycling and lead to LC3B-II accumulation. Lysates were prepared and probed by western blot to detect the relative levels of LC3B-I and LC3B-II. (e) Effect of bortezomib (5 nM) and metformin (500 μM) alone and combined on the levels of GRP78 and autophagosomes in RPMI8226 cells. Cells were treated with drugs for 16 h and GRP78 was then visualized by immunohistochemistry staining and confocal microscopy. Autophagosomes were detected using the cytologic identification (cyto-ID) autophagy detection kit (Enzo Life Sciences, Farmingdale, NY, USA) and visualized using a Zeiss LSM710 confocal microscope (Zeiss Microscopy, Thornwood, NY, USA). Settings for fluorescein isothiocyanate detection were EX488nm and EM550nm. (f) Effect of HSPA5-specific shRNA on autophagosome formation. RPMI8226 cells were transfected with control or HSPA5-specific shRNA and transfectants treated as indicated. Autophagosomes were detected by dye-based staining and confocal microscopy. (g) Autophagosomes were detected by the dye-based method and the relative amount of green fluorescence was determined using ImageJ software (National Institutes of Health, Bethesda, MD, USA). Statistically significant differences in fluorescence are indicated by *. (h) Effect of bortezomib (5 nM) and metformin (500 μM) alone and combined on autophagosome formation in SP cells. SP were treated with drugs as indicated for 16 h and autophagosome formation visualized by dye-based staining and confocal microscopy. Shown are representative images observed in multiple experiments.
Figure 4
Figure 4
Effect of bortezomib and metformin on tumor volume (TV) and overall survival. MM.1S cells (3 × 106/injection) were subcutaneously placed in the flanks of 5-week-old nude NCr nu/nu female mice. Mice were then randomly distributed into four groups (10/group) and after the formation of palpable tumors treatment was commenced. Groups received intravenous injection of either vehicle (phosphate-buffered saline containing 10% dimethyl sulfoxide (DMSO)) or bortezomib (0.5 mg/kg in phosphate-buffered saline, 10% DMSO) administered with or without metformin (600 μg/ml diluted in drinking water that contained 5% dextrose). Metformin was administered throughout treatment and vehicle or bortezomib administered on days 1–4. TVmeasurements were made using a vernier caliper and calculated using the formula: TV=0.5(a × b2) where a is the long diameter and b the short diameter. Mice were euthanized when tumors reached 2 cm3, became ulcerated or elicited neurologic or musculoskeletal complications that limited mobility and feeding. TV was evaluated from the first day of treatment. Shown is the average of replicate measurements. (b) Kaplan–Meier survival curves after treated with either vehicle, metformin, bortezomib or both.

References

    1. 1Kyle RA, Rajkumar SV. Multiple myeloma. Blood 2008; 111: 2962–2972.
    1. 2Palumbo A, Anderson K. Multiple myeloma. N Engl J Med 2011; 364: 1046–1060.
    1. 3Obeng EA, Carlson LM, Gutman DM, Harrington WJ Jr, Lee KP, Boise LH et al. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood 2006; 107: 4907–4916.
    1. 4Chen Y, Brandizzi F. IRE1: ER stress sensor and cell fate executor. Trends Cell Biol 2013; 23: 547–555.
    1. 5Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol 2012; 13: 89–102.
    1. 6Cao SS, Kaufman RJ. Unfolded protein response. Curr Biol 2012; 22: R622–R626.
    1. 7Saito S, Furuno A, Sakurai J, Sakamoto A, Park HR, Shin-Ya K et al. Chemical genomics identifies the unfolded protein response as a target for selective cancer cell killing during glucose deprivation. Cancer Res 2009; 69: 4225–4234.
    1. 8Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H et al. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ 2007; 14: 230–239.
    1. 9Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 2006; 26: 9220–9231.
    1. 10Dong D, Stapleton C, Luo B, Xiong S, Ye W, Zhang Y et al. A critical role for GRP78/BiP in the tumor microenvironment for neovascularization during tumor growth and metastasis. Cancer Res 2011; 71: 2848–2857.
    1. 11Fu Y, Lee AS. Glucose regulated proteins in cancer progression, drug resistance and immunotherapy. Cancer Biol Ther 2006; 5: 741–744.
    1. 12Li J, Ni M, Lee B, Barron E, Hinton DR, Lee AS. The unfolded protein response regulator GRP78/BiP is required for endoplasmic reticulum integrity and stress-induced autophagy in mammalian cells. Cell Death Differ 2008; 15: 1460–1471.
    1. 13Abdel Malek M, Jagannathan S, Malek E, Sayed DM, Elgammal SA, Driscoll JJ. Molecular chaperone GRP78 enhances aggresome delivery to autophagosomes to promote drug resistance in multiple myeloma. Oncotarget 2015; 6: 3098–3110, advanced online publication.
    1. 14Lee AS. GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res 2007; 67: 3496–3499.
    1. 15Wang Q, He Z, Zhang J, Wang Y, Wang T, Tong S et al. Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance. Cancer Detect Prev 2005; 29: 544–551.
    1. 16Lee HK, Xiang C, Cazacu S, Finniss S, Kazimirsky G, Lemke N et al. GRP78 is overexpressed in glioblastomas and regulates glioma cell growth and apoptosis. Neuro Oncol 2008; 10: 236–243.
    1. 17Zhang J, Jiang Y, Jia Z, Li Q, Gong W, Wang L et al. Association of elevated GRP78 expression with increased lymph node metastasis and poor prognosis in patients with gastric cancer. Clin Exp Metastas 2006; 23: 401–410.
    1. 18Dong D, Ni M, Li J, Xiong S, Ye W, Virrey JJ et al. Critical role of the stress chaperone GRP78/BiP in tumor proliferation, survival, and tumor angiogenesis in transgene-induced mammary tumor development. Cancer Res 2008; 68: 498–505.
    1. 19Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov 2009; 8: 627–644.
    1. 20Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila) 2010; 3: 1451–1461.
    1. 21Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ 2005; 330: 1304–1305.
    1. 22Dowling RJ, 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: 10804–10812.
    1. 23Leclerc GM, Leclerc GJ, Kuznetsov JM, DeSalvo J, Barredo JC. Metformin induces apoptosis through AMPK-dependent inhibition of UPR signaling in ALL lymphoblasts. PLoS One 2013; 8: e74420.
    1. 24DeSalvo J, Kuznetsov JN, Du J, Leclerc GM, Leclerc GJ, Lampidis TJ et al. Inhibition of Akt potentiates 2-DG-induced apoptosis via downregulation of UPR in acute lymphoblastic leukemia. Mol Cancer Res 2012; 10: 969–978.
    1. 25Jakubikova J, Adamia S, Kost-Alimova M, Klippel S, Cervi D, Daley JF et al. Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications. Blood 2011; 117: 4409–4419.
    1. 26Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 1996; 183: 1797–1806.
    1. 27Kim KH, Cheong HJ, Kim SJ, Kim SH, Yoon J, Kim HJ et al. Side population of multiple myeloma and multiple myeloma stem cell. Blood, American Society of Hematology Annual Meeting 2014; 124: 5786.
    1. 28Scudiero DA. Evaluation of a soluble tetrazolium/formazan assay for cell growth and drug sensitivity in culture using human and other tumor cell lines. Cancer Res 1988; 48: 4827–4833.
    1. 29Fukuda K, Saikawa Y, Ohashi M, Kumagai K, Kitajima M, Okano H et al. Tumor initiating potential of side population cells in human gastric cancer. Int J Oncol 2009; 34: 1201–1207.
    1. 30Huang D, Gao Q, Guo L, Zhang C, Jiang W, Li H et al. Isolation and identification of cancer stem-like cells in esophageal carcinoma cell lines. Stem Cells Dev 2009; 18: 465–473.
    1. 31Loebinger MR, Giangreco A, Groot KR, Prichard L, Allen K, Simpson C et al. Squamous cell cancers contain a side population of stem-like cells that are made chemosensitive by ABC transporter blockade. Br J Cancer 2008; 98: 380–387.
    1. 32Kee N, Sivalingam S, Boonstra R, Wojtowicz JM. Utility of Ki-67 and BrdU as proliferative markers of adult neurogenesis. J Neurosci Methods 2002; 115: 97–105.
    1. 33Hideshima T, Ikeda H, Chauhan D, Okawa Y, Raje N, Podar K et al. Bortezomib induces canonical nuclear factor-kappaB activation in multiple myeloma cells. Blood 2009; 114: 1046–1052.
    1. 34Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab 2005; 1: 15–25.
    1. 35Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 2001; 108: 1167–1174.
    1. 36Sahra IB, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P et al. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene 2008; 27: 3576–3586.
    1. 37Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc 2006; 1: 2315–2319.
    1. 38Richardson PG, Mitsiades C, Hideshima T, Anderson KC. Bortezomib: Proteasome inhibition as an effective anticancer therapy. Future Oncol 2005; 1: 161–171.
    1. 39Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell 2007; 29: 1261–1274.
    1. 40Chen K-F, Yeh P-Y, Yeh K-H, Lu Y-S, Huang S-Y, Cheng A-L. Down-regulation of phospho-Akt is a major molecular determinant of bortezomib-induced apoptosis in hepatocellular carcinoma cells. Cancer Res 2008; 68: 6698–6707.
    1. 41Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 2006; 26: 9220–9231.
    1. 42Ding WX, Ni HM, Gao W, Hou YF, Melan MA, Chen X et al. Differential effects of endoplasmic reticulum stress-induced autophagy on cell survival. J Biol Chem 2007; 282: 4702–4710.
    1. 43Pyo JO, Jang MH, Kwon YK, Lee HJ, Jun JI, Woo HN et al. Essential roles of Atg5 and FADD in autophagic cell death: dissection of autophagic cell death into vacuole formation and cell death. J Biol Chem 2005; 280: 20722–20729.
    1. 44Tomic T, Botton T, Cerezo M, Robert G, Luciano F, Puissant A et al. Metformin inhibits melanoma development through autophagy and apoptosis mechanisms. Cell Death Dis 2011; 2: e199.
    1. 45Jain MV, Paczulla AM, Klonisch T, Dimgba FN, Rao SB, Roberg K et al. Interconnections between apoptotic, autophagic and necrotic pathways: implications for cancer therapy development. J Cell Mol Med 2013; 17: 12–29.
    1. 46Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol 2007; 8: 741–752.
    1. 47Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care 2009; 32: 1620–1625.
    1. 48Badros A, Goloubeva O, Dalal JS, Can I, Thompson J, Rapoport AP et al. Neurotoxicity of bortezomib therapy in multiple myeloma: a single-center experience and review of the literature. Cancer 2007; 110: 1042–1049.
    1. 49Pentikainen PJ, Neuvonen PJ, Penttila A. Pharmacokinetics of metformin after intravenous and oral administration to man. Eur J Clin Pharmacol 1979; 16: 195–202.
    1. 50Quinn BJ, Kitagawa H, Memmott RM, Gillis JJ, Dennis PA. Repositioning metformin for cancer prevention and treatment. Trends Endocrinol Metab 2013; 24: 469–480.

Source: PubMed

3
Abonnere