MYC regulates the unfolded protein response and glucose and glutamine uptake in endocrine resistant breast cancer

Ayesha N Shajahan-Haq, Katherine L Cook, Jessica L Schwartz-Roberts, Ahreej E Eltayeb, Diane M Demas, Anni M Warri, Caroline O B Facey, Leena A Hilakivi-Clarke, Robert Clarke, Ayesha N Shajahan-Haq, Katherine L Cook, Jessica L Schwartz-Roberts, Ahreej E Eltayeb, Diane M Demas, Anni M Warri, Caroline O B Facey, Leena A Hilakivi-Clarke, Robert Clarke

Abstract

Background: About 70% of all breast cancers are estrogen receptor alpha positive (ER+) and are treated with antiestrogens. However, 50% of ER + tumors develop resistance to these drugs (endocrine resistance). In endocrine resistant cells, an adaptive pathway called the unfolded protein response (UPR) is elevated that allows cells to tolerate stress more efficiently than in sensitive cells. While the precise mechanism remains unclear, the UPR can trigger both pro-survival and pro-death outcomes that depend on the nature and magnitude of the stress. In this study, we identified MYC, an oncoprotein that is upregulated in endocrine resistant breast cancer, as a regulator of the UPR in glucose-deprived conditions.

Methods: ER+ human breast cancer cell lines (LCC1, LCC1, LY2 and LCC9) and rat mammary tumors were used to confirm upregulation of MYC in endocrine resistance. To evaluate functional relevance of proteins, siRNA-mediated inhibition or small molecule inhibitors were used. Cell density/number was evaluated with crystal violet assay; cell cycle and apoptosis were measured by flow cytometry. Relative quantification of glutamine metabolites were determined by mass spectrometry. Signaling molecules of the UPR, apoptosis or autophagy pathways were investigated by western blotting.

Results: Increased MYC function in resistant cells correlated with increased dependency on glutamine and glucose for survival. Inhibition of MYC reduced cell growth and uptake of both glucose and glutamine in resistant cells. Interestingly, in glucose-deprived conditions, glutamine induced apoptosis and necrosis, arrested autophagy, and triggered the unfolded protein response (UPR) though GRP78-IRE1α with two possible outcomes: (i) inhibition of cell growth by JNK activation in most cells and, (ii) promotion of cell growth by spliced XBP1 in the minority of cells. These disparate effects are regulated, at different signaling junctions, by MYC more robustly in resistant cells.

Conclusions: Endocrine resistant cells overexpress MYC and are better adapted to withstand periods of glucose deprivation and can use glutamine in the short term to maintain adequate metabolism to support cell survival. Our findings reveal a unique role for MYC in regulating cell fate through the UPR, and suggest that targeting glutamine metabolism may be a novel strategy in endocrine resistant breast cancer.

Figures

Figure 1
Figure 1
MYC expression is elevated in antiestrogen resistant breast cancerin vitroandin vivo.A, Basal MYC-luciferase activity is 4.24-fold (SE = 0.10) higher in LY2 and LCC2 (estrogen independent but responsive; antiestrogen resistant) and 6.67-fold (SE = 0.09) higher in LCC9 (estrogen independent and non-responsive; antiestrogen resistant) compared with LCC1 (estrogen independent but responsive; antiestrogen sensitive); see Experimental Procedures. ANOVA, p < 0.001; *p < 0.05 for MYC promoter activation in indicated cells compared with LCC1 cells. B, Western blot shows increased expression of MYC protein in LCC9 cells compared to LCC1 cells while MAX protein levels did not change; actin was used as a loading control. C, Immunohistochemical (IHC) MYC staining show increased protein levels (brown) in LCC9 compared with LCC1 xenografts; for negative controls, antibody diluents without MYC antibody were used. D, DMBA-induced rat mammary gland tumors with acquired resistance to TAM show increased levels MYC protein levels (brown) compared to sensitive (or de novo resistant) tumors.
Figure 2
Figure 2
MYC promotes survival in antiestrogen resistant cells.A, Western blot, reduced MYC in LCC9 cells at 48 h with MYC siRNA compared to control siRNA. Actin is a loading control. B, Quantitation of MYC in in LCC9 cell show 60% reduction in MYC siRNA transfected cells compared with control siRNA. C, MYC siRNA interacted additively (RI = 1.11) with ICI in inhibiting cell number in LCC1 but not in LCC9 cells. Bars,mean ± SE of relative cell number (normalized to vehicle controls) for a representative experiment performed in sextuplicate. ANOVA, p < 0.001; *p < 0.05 for treatment versus control for respective cell lines. ^, p < 0.05 for LCC1 versus LCC9 cells with MYC siRNA + ICI. D, LCC9 cells showed increased sensitivity to 10058-F4compared with LCC1 cells at 48 h. Points, mean of cell number; bars, ±SE. E, 10058-F4 or ICI alone or the combination for 48 h inhibit cell number in LCC1. In LCC9 cells, RI = 1.51, suggest a modest synergistic interaction between ICI and 10058-F4; ANOVA, p < 0.001; *p < 0.05 for treatment versus control for respective cell lines. ^, p < 0.05 for ICI + 10058-F4 versus 10058-F4. F, Western blot show decrease in MYC, MAX, BCL2 and an increase in cleaved CASP7 , with 10058-F4 (MI: MYC inhibitor) or ICI + 10058-F4 (C: combination) compared with vehicle (V) alone or with ICI alone (I) treatment (48 h). G, Annexin V-FITC (apoptosis) in LCC1 and LCC9 cells with vehicle, ICI , 10058-F4 , or ICI + 10058-F4 (combination). ANOVA, p < 0.001; *p < 0.05 for indicated treatment versus vehicle control for respective cells lines. Paclitaxel, a positive control for apoptosis. H, Dot plots showcells positive for annexin-V-FITC (x-axis) and propidium iodide (PI; y-axis).
Figure 3
Figure 3
Combination of MYC inhibitor and antiestrogen increased G1 cell cycle arrest in endocrine resistant cells.A, Top, ICI (100 nM), 10058-F4 (25 μM), or the combination significantly increased percentage of cells in G1 arrest and reduced percentage of cells in S phase in LCC1 (p < 0.001). Bottom, Representative cell count plots for propidium iodide (PI) in LCC1 cells are shown. B, Top, Only the combination of ICI and 10058-F4 induced significant increase in G1 arrest in LCC9 cells (p < 0.001). Bottom, Representative cell count plots for PI in LCC9 cells are shown. Graphs represent data that are presented as the mean ± SE for three independent experiments. ANOVA, p < 0.001; *p < 0.001 for indicated treatment versus vehicle control.
Figure 4
Figure 4
Increased dependence on glutamine and glucose in antiestrogen resistant cells.A, Schematic for glutamine metabolism: glutamine is converted to glutamate by the mitochondrial enzyme, glutaminase (GLS); the reverse reaction is catalyzed by glutamate-ammonia ligase (GLUL). Glutamine is an essential substrate for the biosynthesis of proline. B-D, Relative quantification of glutamine, glutamate, and proline by UPLC-QqQLIT showed a significant increase in glutamate (p = 0.002) and proline levels (p = 0.032) in LCC9 cells when compared with LCC1 control cells; six biological replicates from each cell line was used and levels of each respective metabolite was normalized to total protein levels in each sample. E, Uptake of glucose is significantly increased in LCC9 cells compared with LCC1 cells under basal conditions (p < 0.05). Relative cellular metabolites and glucose uptake were compared using Student’s t test F-G, Inhibition of MYC using siRNA significantly deceased (F) glutamine (p < 0.05) and (G) glucose uptake (p = 0.011) in LCC9 compared with LCC1 cells. ANOVA, p < 0.001. H, Inhibition of MYC with siRNA decreased protein levels of transporters of glutamine (ASCT2/SLC1A5), glutamate (EAAT2/SLC1A2) and glucose (GLUT1/SLC2A1) in LCC9 cells. Western blot shown is representative of three independent experiments.
Figure 5
Figure 5
Glutamine and glucose metabolism is increased in antiestrogen resistant cells.A-B, LCC9 cells were significantly more sensitive to (A) compound-968, an inhibitor of GLS/GAC, and to (B) STF-31, an inhibitor of GLUT-1. Bars represent the mean ± SE of relative number (normalized to vehicle control) for a single representative experiment performed in sextuplicate. ANOVA, p ≤ 0.001; *p < 0.05 for LCC9 versus LCC1 for indicated concentrations. C, Cells were treated with compound-968 (20 μM), STF-31 (5 μM), ICI (100 nM), or the indicated combinations for 48 h. Bars represent the mean ± SE of relative cell number (normalized to vehicle controls) for a single representative experiment performed in sextuplicate. ANOVA, p < 0.001; *p < 0.05 for LCC9 versus LCC1 for indicated treatments. D, Knockdown of GLS levels with siRNA in LCC9 cells showed significant decrease in cell number within 24 h compared with that in LCC1 cells. ANOVA, p = 0.03; *p ≤ 0.05 for LCC9 GLS siRNA compared with LCC1 GLS siRNA. E, Western blot showing decreased levels of GLS in both cell lines; actin was used as a protein loading control. F, Right, LCC9 ells were treated with 10058-F4 (25 μM), or vehicle for 48 h; left, transfected with MYC or control siRNA for 48 h. Knockdown of MYC increased GLS/GAC levels and decreased GLUL levels. G, siRNA mediated MYC knockdown showed increase in GLS and a decrease in GLUL levels in LCC2 and LY2 cells.
Figure 6
Figure 6
MYC expression increases sensitivity to glucose and glutamine deprivation.A-B, Overexpression of MYC in LCC1 cells significantly increased (p < 0.01) (A) and knockdown of MYC in LCC9 cells (B) significantly decreased cell number in the absence of glucose and glutamine (p ≤ 0.001). (C-F) LCC1 and LCC9 cells were grown in complete (12 mM glucose; 2 mM glutamine), incomplete (no glucose; no glutamine), glucose only (12 mM glucose; no glutamine), and glutamine-only (2 mM glutamine; no glucose) for 72 h. Changes in cell growth rates were determined by normalizing cell numbers measurements at 24 h, 48 h, and 72 h to cell numbers measurements at 0 h. At 72 h, LCC9 cells showed significantly higher growth rate compared to LCC1 in complete media. However, growth rate was significantly reduced for LCC9 in incomplete media when compared with LCC1 cells. In glucose-only media (at 72 h), LCC1 and LCC9 cells did not show an increase in cell growth. In glutamine-only media, LCC9 cells showed a significant decrease in cell number relative to LCC1 cells. Dashed line denotes change in scales between the graphs. Bars represent the mean ± SE of relative number (normalized to vehicle control) for a single representative experiment performed in sextuplicate. G, Knockdown of MYC in LCC9 cells reduced sensitivity to incomplete media, as seen in B, and also reduced inhibition of cell number in the presence of 2 mM glutamine in glucose-deprived conditions. ANOVA, p < 0.001; p ≤ 0.01 for LCC9-MYC siRNA versus LCC9-control siRNA for indicated treatment. Bars represent the mean ± SE of relative number (normalized to vehicle control) for a single representative experiment performed in sextuplicate.
Figure 7
Figure 7
Glutamine induces apoptosis and arrests autophagy via the UPR in glucose-deprived conditions.A, Significantly higher levels of apoptosis were seen in LCC9 compared with LCC1 cells following treatment with 2 or 4 mM glutamine at 48 h. ANOVA, p < 0.05; *p < 0.05 for LCC9 versus LCC1 for indicated treatment. B, Time-course, 0, 24 and 48 h, analysis of the autophagosome-associated proteins LC3II (marker for autophagosome formation or enlargement) and p62/SQSTM1 (marker for autophagosome activity, degradation of cargo). Increased formation of autophagosomes but arrested cargo degradation was seen within 24 h in both LCC1 and LCC9 cells in glutamine only media (and in no-glucose + no glutamine) conditions at 24 and 48 h but not in glucose-only (or in glucose + glutamine) media. C, In presence of 2 or 4 mM glutamine at 48 h, LCC9 cells showed increased levels of MYC and MAX and LC3II but no change in SQSTM1/p62. D, Cellular levels of total reactive species (RS) was significantly elevated in LCC9 compared to LCC1 cells in incomplete media (ANOVA, p < 0.001; *p < 0.05 for LCC9 versus LCC1 with no glucose + no glutamine).
Figure 8
Figure 8
Glutamine in glucose-deprived conditions activates the UPR.A, Cells were plated at 70% confluence. 24 hr later, media was changed to 0, 2, or 4 mM glutamine alone or in presence of 12 mM glucose. Western blot analysis showed Increased levels of GRP78, IRE1a, phospho-JNK, CHOP and decreased levels of BCL2 were present in LCC1 (right) and LCC9 (left) cells in glutamine-only conditions. MYC protein levels were highest when both glucose and glutamine are present; MYC is undetectable when these metabolites are absent in the media. MYC expression in the presence of glutamine-only, but not in presence of glucose-only, conditions correlated with increased expression of UPR proteins. B, Knockdown of MYC for 24 h was followed by media change to either glucose + glutamine, glucose-only, glutamine-only or no glucose + no glutamine conditions for another 48 h. Western blots analysis showed that a decrease in MYC protein levels correlated with an increase in the UPR proteins IRE1α and phospho-JNK(Thr183/Ty4185), and the autophagosome formation marker LC3II, and the autophagosome cargo degradation marker p62/SQSTM1. GRP78 was also increased in glucose + glutamine, glucose-only and no glucose + no glutamine conditions but robust expression of GRP78 in glutamine-only conditions was not affected by MYC siRNA. Total levels of JNK did not change.
Figure 9
Figure 9
UPR in glutamine-only conditions can lead to both pro-survival and pro-death outcomes. Effect of transfection of siRNA targeting GRP78, IRE1α, XBP1(s), and MYC for 24 h; or JNK inhibition with a small molecule inhibitor (SP600125) on growth in either glucose + glutamine or glutamine-alone media. Western blot (48 h); A-C, GRP78. D-F, IRE1α. G-I, JNK. J-L, XBP1. M-O, MYC. Inhibition of GRP78 did not significantly further affect cell numbers in glutamine-only conditions in both LCC1 and LCC9 cell lines, A. Western blot analysis of total GRP78 protein are shown in both cell lines in different conditions, B-C. Knockdown of IRE1α, D-F and XBP1, J-L, significantly increased inhibition of cell growth in glutamine-only conditions in both cell lines. However, inhibition of JNK with SP600125 significantly decreased the inhibition of cell growth in glutamine-only conditions, G-I. Also, knockdown of MYC, M-O, significantly decreased inhibition of cell growth in glutamine-only conditions. Overall, MYC may have facilitate an IRE1α-XBP1 pathway to promote cell survival during glutamine-only conditions, and an IRE1α-phospho-JNK pathway to promote cell death in this condition. ANOVA, p ≤ 0.001; *p < 0.05 for respective cell lines transfected with indicated siRNA (or treated with SP600125, for JNK) compared with control siRNA (or vehicle alone, for JNK) in glutamine-only conditions.
Figure 10
Figure 10
MYC confers metabolic flexibility in antiestrogen resistant cells.A, Rate of cell growth was significantly reduced in LCC9Gln cells compared with LCC9 control cells (p ≤ 0.001). Cell numbers at 72 h were compared using Student’s t test. B, MYC, MAX and GLUL protein levels were reduced, while GLS/GAC was increased, in LCC9Gln cells compared with control. C, Schematic diagram illustrating the role of MYC in regulating glutamine metabolism in complete (right; basal; with glucose and glutamine) and in glutamine-only conditions (left; glutamine but no glucose). MYC regulates glutamine, glutamate, and glucose uptake through transporters, ASCT2, EAAT2 and GLUT1, respectively, under normal conditions. In glucose-deprived conditions, glutamine metabolism triggers the UPR and induces cell death (inducing apoptosis and arresting autophagy) via a MYC-regulated IRE1α-JNK-CHOP in the short-term (72 h), and also promotes cell survival, through a IRE1α-XBP1(s); the surviving cells grow at a slower rate of cell proliferation (A), at >72 h. Dashed line denotes presence of intermediate metabolites/proteins that are not addressed in this study.

References

    1. Clarke R, Skaar T, Leonessa F, Brankin B, James M, Brunner N, Lippman ME. Acquisition of an antiestrogen-resistant phenotype in breast cancer: role of cellular and molecular mechanisms. Cancer Treat Res. 1996;87:263–283. doi: 10.1007/978-1-4613-1267-3_11.
    1. Clarke R, Liu MC, Bouker KB, Gu Z, Lee RY, Zhu Y, Skaar TC, Gomez B, O’Brien K, Wang Y, Hilakivi-Clarke LA. Antiestrogen resistance in breast cancer and the role of estrogen receptor signaling. Oncogene. 2003;22:7316–7339. doi: 10.1038/sj.onc.1206937.
    1. Amati B, Alevizopoulos K, Vlach J. Myc and the cell cycle. Front Biosci. 1998;3:d250–d268.
    1. Chen Y, Olopade OI. MYC in breast tumor progression. Expert Rev Anticancer Ther. 2008;8:1689–1698. doi: 10.1586/14737140.8.10.1689.
    1. Dang CV. MYC on the path to cancer. Cell. 2012;149:22–35. doi: 10.1016/j.cell.2012.03.003.
    1. Planas-Silva MD, Bruggeman RD, Grenko RT, Smith JS. Overexpression of c-Myc and Bcl-2 during progression and distant metastasis of hormone-treated breast cancer. Exp Mol Pathol. 2007;82:85–90. doi: 10.1016/j.yexmp.2006.09.001.
    1. Blancato J, Singh B, Liu A, Liao DJ, Dickson RB. Correlation of amplification and overexpression of the c-myc oncogene in high-grade breast cancer: FISH, in situ hybridisation and immunohistochemical analyses. Br J Cancer. 2004;90:1612–1619. doi: 10.1038/sj.bjc.6601703.
    1. Deming SL, Nass SJ, Dickson RB, Trock BJ. C-myc amplification in breast cancer: a meta-analysis of its occurrence and prognostic relevance. Br J Cancer. 2000;83:1688–1695. doi: 10.1054/bjoc.2000.1522.
    1. McNeil CM, Sergio CM, Anderson LR, Inman CK, Eggleton SA, Murphy NC, Millar EK, Crea P, Kench JG, Alles MC, Gardiner-Garden M, Ormandy CJ, Butt AJ, Henshall SM, Musgrove EA, Sutherland RL. c-Myc overexpression and endocrine resistance in breast cancer. J Steroid Biochem Mol Biol. 2006;102:147–155. doi: 10.1016/j.jsbmb.2006.09.028.
    1. Miller TW, Balko JM, Ghazoui Z, Dunbier A, Anderson H, Dowsett M, Gonzalez-Angulo AM, Mills GB, Miller WR, Wu H, Shyr Y, Arteaga CL. A gene expression signature from human breast cancer cells with acquired hormone independence identifies MYC as a mediator of antiestrogen resistance. Clin Cancer Res. 2011;17:2024–2034. doi: 10.1158/1078-0432.CCR-10-2567.
    1. Dang CV, Lewis BC. Role of oncogenic transcription factor c-Myc in cell cycle regulation, apoptosis and metabolism. J Biomed Sci. 1997;4:269–278. doi: 10.1007/BF02258350.
    1. Nair SK, Burley SK. X-ray structures of Myc-Max and Mad-Max recognizing DNA. Molecular bases of regulation by proto-oncogenic transcription factors. Cell. 2003;112:193–205. doi: 10.1016/S0092-8674(02)01284-9.
    1. Dang CV. Therapeutic targeting of Myc-reprogrammed cancer cell metabolism. Cold Spring Harb Symp Quant Biol. 2011;76:369–374. doi: 10.1101/sqb.2011.76.011296.
    1. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, Zeller KI, De Marzo AM, Van Eyk JE, Mendell JT, Dang CV. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458:762–765. doi: 10.1038/nature07823.
    1. Teicher BA, Linehan WM, Helman LJ. Targeting cancer metabolism. Clin Cancer Res. 2012;18:5537–5545. doi: 10.1158/1078-0432.CCR-12-2587.
    1. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell. 2012;21:297–308. doi: 10.1016/j.ccr.2012.02.014.
    1. Brunner N, Boulay V, Fojo A, Freter CE, Lippman ME, Clarke R. Acquisition of hormone-independent growth in MCF-7 cells is accompanied by increased expression of estrogen-regulated genes but without detectable DNA amplifications. Cancer Res. 1993;53:283–290.
    1. Brunner N, Boysen B, Jirus S, Skaar TC, Holst-Hansen C, Lippman J, Frandsen T, Spang-Thomsen M, Fuqua SA, Clarke R. MCF7/LCC9: an antiestrogen-resistant MCF-7 variant in which acquired resistance to the steroidal antiestrogen ICI 182,780 confers an early cross-resistance to the nonsteroidal antiestrogen tamoxifen. Cancer Res. 1997;57:3486–3493.
    1. Shajahan AN, Wang A, Decker M, Minshall RD, Liu MC, Clarke R. Caveolin-1 tyrosine phosphorylation enhances paclitaxel-mediated cytotoxicity. J Biol Chem. 2007;282:5934–5943. doi: 10.1074/jbc.M608857200.
    1. Shajahan AN, Dobbin ZC, Hickman FE, Dakshanamurthy S, Clarke R. Tyrosine-phosphorylated caveolin-1 (Tyr-14) increases sensitivity to paclitaxel by inhibiting BCL2 and BCLxL proteins via c-Jun N-terminal kinase (JNK) J Biol Chem. 2012;287:17682–17692. doi: 10.1074/jbc.M111.304022.
    1. Vindelov LL, Christensen IJ, Nissen NI. A detergent-trypsin method for the preparation of nuclei for flow cytometric DNA analysis. Cytometry. 1983;3:323–327. doi: 10.1002/cyto.990030503.
    1. Ricci MS, Jin Z, Dews M, Yu D, Thomas-Tikhonenko A, Dicker DT, El-Deiry WS. Direct repression of FLIP expression by c-myc is a major determinant of TRAIL sensitivity. Mol Cell Biol. 2004;24:8541–8555. doi: 10.1128/MCB.24.19.8541-8555.2004.
    1. Cook KL, Shajahan AN, Warri A, Jin L, Hilakivi-Clarke LA, Clarke R. Glucose-regulated protein 78 controls cross-talk between apoptosis and autophagy to determine antiestrogen responsiveness. Cancer Res. 2012;72:3337–3349. doi: 10.1158/0008-5472.CAN-12-0269.
    1. Sheikh KD, Khanna S, Byers SW, Fornace A, Jr, Cheema AK. Small molecule metabolite extraction strategy for improving LC/MS detection of cancer cell metabolome. J Biomol Tech. 2011;22:1–4.
    1. Romanelli S, Perego P, Pratesi G, Carenini N, Tortoreto M, Zunino F. In vitro and in vivo interaction between cisplatin and topotecan in ovarian carcinoma systems. Cancer Chemother Pharmacol. 1998;41:385–390. doi: 10.1007/s002800050755.
    1. Musgrove EA, Sergio CM, Loi S, Inman CK, Anderson LR, Alles MC, Pinese M, Caldon CE, Schutte J, Gardiner-Garden M, Ormandy CJ, McArthur G, Butt AJ, Sutherland RL. Identification of functional networks of estrogen- and c-Myc-responsive genes and their relationship to response to tamoxifen therapy in breast cancer. PLoS One. 2008;3:e2987. doi: 10.1371/journal.pone.0002987.
    1. Cook KL, Clarke PA, Parmar J, Hu R, Schwartz-Roberts JL, Abu-Asab M, Wärri A, Baumann WT, Clarke R. Knockdown of estrogen receptor-alpha induces autophagy and inhibits antiestrogen-mediated unfolded protein response activation, promoting ROS-induced breast cancer cell death. FASEB J. 2014;72:3337–3349.
    1. Guo J, Parise RA, Joseph E, Egorin MJ, Lazo JS, Prochownik EV, Eiseman JL. Efficacy, pharmacokinetics, tisssue distribution, and metabolism of the Myc-Max disruptor, 10058–F4 [Z, E]-5-[4-ethylbenzylidine]-2-thioxothiazolidin-4-one, in mice. Cancer Chemother Pharmacol. 2009;63:615–625. doi: 10.1007/s00280-008-0774-y.
    1. Crawford AC, Riggins RB, Shajahan AN, Zwart A, Clarke R. Co-inhibition of BCL-W and BCL2 restores antiestrogen sensitivity through BECN1 and promotes an autophagy-associated necrosis. PLoS One. 2010;5:e8604. doi: 10.1371/journal.pone.0008604.
    1. Elgadi KM, Meguid RA, Qian M, Souba WW, Abcouwer SF. Cloning and analysis of unique human glutaminase isoforms generated by tissue-specific alternative splicing. Physiol Genomics. 1999;1:51–62.
    1. Johansen T, Lamark T. Selective autophagy mediated by autophagic adapter proteins. Autophagy. 2011;7:279–296. doi: 10.4161/auto.7.3.14487.
    1. Lamark T, Kirkin V, Dikic I, Johansen T. NBR1 and p62 as cargo receptors for selective autophagy of ubiquitinated targets. Cell Cycle. 2009;8:1986–1990. doi: 10.4161/cc.8.13.8892.
    1. Reggiori F, Klionsky DJ. Autophagy in the eukaryotic cell. Eukaryot Cell. 2002;1:11–21. doi: 10.1128/EC.01.1.11-21.2002.
    1. Schwartz-Roberts JL, Shajahan AN, Cook KL, Warri A, Abu-Asab M, Clarke R. GX15-070 (obatoclax) induces apoptosis and inhibits cathepsin D- and L-mediated autophagosomal lysis in antiestrogen-resistant breast cancer cells. Mol Cancer Ther. 2013;12:448–459. doi: 10.1158/1535-7163.MCT-12-0617.
    1. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 2007;26:1749–1760. doi: 10.1038/sj.emboj.7601623.
    1. Tripathi DN, Chowdhury R, Trudel LJ, Tee AR, Slack RS, Walker CL, Wogan GN. Reactive nitrogen species regulate autophagy through ATM-AMPK-TSC2-mediated suppression of mTORC1. Proc Natl Acad Sci U S A. 2013;110:E2950–E2957. doi: 10.1073/pnas.1307736110.
    1. Clarke R, Cook KL, Hu R, Facey CO, Tavassoly I, Schwartz JL, Baumann WT, Tyson JJ, Xuan J, Wang Y, Warri A, Shajahan AN. Endoplasmic reticulum stress, the unfolded protein response, autophagy, and the integrated regulation of breast cancer cell fate. Cancer Res. 2012;72:1321–1331. doi: 10.1158/1538-7445.AM2012-1321.
    1. de la Cadena SG, Hernandez-Fonseca K, Camacho-Arroyo I, Massieu L. Glucose deprivation induces reticulum stress by the PERK pathway and caspase-7- and calpain-mediated caspase-12 activation. Apoptosis. 2013;19:414–427. doi: 10.1007/s10495-013-0930-7.
    1. Haga N, Saito S, Tsukumo Y, Sakurai J, Furuno A, Tsuruo T, Tomida A. Mitochondria regulate the unfolded protein response leading to cancer cell survival under glucose deprivation conditions. Cancer Sci. 2010;101:1125–1132. doi: 10.1111/j.1349-7006.2010.01525.x.
    1. Davies MP, Barraclough DL, Stewart C, Joyce KA, Eccles RM, Barraclough R, Rudland PS, Sibson DR. Expression and splicing of the unfolded protein response gene XBP-1 are significantly associated with clinical outcome of endocrine-treated breast cancer. Int J Cancer. 2008;123:85–88. doi: 10.1002/ijc.23479.
    1. Gomez BP, Riggins RB, Shajahan AN, Klimach U, Wang A, Crawford AC, Zhu Y, Zwart A, Wang M, Clarke R. Human X-box binding protein-1 confers both estrogen independence and antiestrogen resistance in breast cancer cell lines. FASEB J. 2007;21:4013–4027. doi: 10.1096/fj.06-7990com.
    1. Bennett BL, Sasaki DT, Murray BW, O’Leary EC, Sakata ST, Xu W, Leisten JC, Motiwala A, Pierce S, Satoh Y, Bhagwat SS, Manning AM, Anderson DW. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc Natl Acad Sci U S A. 2001;98:13681–13686. doi: 10.1073/pnas.251194298.
    1. Chambliss KL, Yuhanna IS, Mineo C, Liu P, German Z, Sherman TS, Mendelsohn ME, Anderson RG, Shaul PW. Estrogen receptor alpha and endothelial nitric oxide synthase are organized into a functional signaling module in caveolae. Circ Res. 2000;87:E44–E52. doi: 10.1161/01.RES.87.11.e44.
    1. Chen X, Iliopoulos D, Zhang Q, Tang Q, Greenblatt MB, Hatziapostolou M, Lim E, Tam WL, Ni M, Chen Y, Mai J, Shen H, Hu DZ, Adoro S, Hu B, Song M, Tan C, Landis MD, Ferrari M, Shin SJ, Brown M, Chang JC, Liu XS, Glimcher LH. XBP1 promotes triple-negative breast cancer by controlling the HIF1alpha pathway. Nature. 2014;508:103–107. doi: 10.1038/nature13119.
    1. Clarke R, Shajahan AN, Riggins RB, Cho Y, Crawford A, Xuan J, Zhang B, Facey C, Aiyer H, Cook K, Hickman FE, Tavassoly I, Verdugo A, Chen C, Zwart A, Wärri A, Hilakivi-Clarke LA. Gene network signaling in hormone responsiveness modifies apoptosis and autophagy in breast cancer cells. J Steroid Biochem Mol Biol. 2009;114:8–20. doi: 10.1016/j.jsbmb.2008.12.023.
    1. Zhu Y, Singh B, Hewitt S, Liu A, Gomez B, Wang A, Clarke R. Expression patterns among interferon regulatory factor-1, human X-box binding protein-1, nuclear factor kappa B, nucleophosmin, estrogen receptor-alpha and progesterone receptor proteins in breast cancer tissue microarrays. Int J Oncol. 2006;28:67–76.
    1. Benetatos L, Vartholomatos G, Hatzimichael E. Polycomb group proteins and MYC: the cancer connection. Cell Mol Life Sci. 2014;71:257–269. doi: 10.1007/s00018-013-1426-x.
    1. Dang CV, Hamaker M, Sun P, Le A, Gao P. Therapeutic targeting of cancer cell metabolism. J Mol Med (Berl) 2011;89:205–212. doi: 10.1007/s00109-011-0730-x.
    1. Zhao Y, Butler EB, Tan M. Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis. 2013;4:e532. doi: 10.1038/cddis.2013.60.
    1. Terunuma A, Putluri N, Mishra P, Mathe EA, Dorsey TH, Yi M, Wallace TA, Issaq HJ, Zhou M, Killian JK, Stevenson HS, Karoly ED, Chan K, Samanta S, Prieto D, Hsu TY, Kurley SJ, Putluri V, Sonavane R, Edelman DC, Wulff J, Starks AM, Yang Y, Kittles RA, Yfantis HG, Lee DH, Ioffe OB, Schiff R, Stephens RM, Meltzer PS, et al. MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. J Clin Invest. 2014;124:398–412. doi: 10.1172/JCI71180.
    1. Dang CV. MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harb Perspect Med. 2013;3:a014217. doi: 10.1101/cshperspect.a014217.
    1. DeBerardinis RJ, Cheng T. Q’s next: the diverse functions of glutamine in metabolism, cell biology and cancer. Oncogene. 2010;29:313–324. doi: 10.1038/onc.2009.358.
    1. Wise DR, Thompson CB. Glutamine addiction: a new therapeutic target in cancer. Trends Biochem Sci. 2010;35:427–433. doi: 10.1016/j.tibs.2010.05.003.
    1. Morrish F, Isern N, Sadilek M, Jeffrey M, Hockenbery DM. c-Myc activates multiple metabolic networks to generate substrates for cell-cycle entry. Oncogene. 2009;28:2485–2491. doi: 10.1038/onc.2009.112.
    1. Gaglio D, Soldati C, Vanoni M, Alberghina L, Chiaradonna F. Glutamine deprivation induces abortive s-phase rescued by deoxyribonucleotides in k-ras transformed fibroblasts. PLoS One. 2009;4:e4715. doi: 10.1371/journal.pone.0004715.
    1. Liu W, Le A, Hancock C, Lane AN, Dang CV, Fan TW, Phang JM. Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC. Proc Natl Acad Sci U S A. 2012;109:8983–8988. doi: 10.1073/pnas.1203244109.
    1. Sehgal V, Ram PT. Network motifs in JNK signaling. Genes Cancer. 2013;4:409–413. doi: 10.1177/1947601913507577.
    1. Noguchi K, Kitanaka C, Yamana H, Kokubu A, Mochizuki T, Kuchino Y. Regulation of c-Myc through phosphorylation at Ser-62 and Ser-71 by c-Jun N-terminal kinase. J Biol Chem. 1999;274:32580–32587. doi: 10.1074/jbc.274.46.32580.
    1. Alarcon-Vargas D, Ronai Z. c-Jun-NH2 kinase (JNK) contributes to the regulation of c-Myc protein stability. J Biol Chem. 2004;279:5008–5016. doi: 10.1074/jbc.M312054200.
    1. Toh PP, Luo S, Menzies FM, Rasko T, Wanker EE, Rubinsztein DC. Myc inhibition impairs autophagosome formation. Hum Mol Genet. 2013;22:5237–5248. doi: 10.1093/hmg/ddt381.
    1. Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review. Cancer Res. 1989;49:6449–6465.
    1. Millon SR, Ostrander JH, Brown JQ, Raheja A, Seewaldt VL, Ramanujam N. Uptake of 2-NBDG as a method to monitor therapy response in breast cancer cell lines. Breast Cancer Res Treat. 2011;126:55–62. doi: 10.1007/s10549-010-0884-1.
    1. Wirth M, Stojanovic N, Christian J, Paul MC, Stauber RH, Schmid RM, Häcker G, Krämer OH, Saur D, Schneider G. MYC and EGR1 synergize to trigger tumor cell death by controlling NOXA and BIM transcription upon treatment with the proteasome inhibitor bortezomib. Nucleic Acids Res. 2014;42:10433–10447. doi: 10.1093/nar/gku763.
    1. Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev. 2009;89:381–410. doi: 10.1152/physrev.00016.2008.
    1. Holroyde CP, Skutches CL, Boden G, Reichard GA. Glucose metabolism in cachectic patients with colorectal cancer. Cancer Res. 1984;44:5910–5913.
    1. Ko YH, Lin Z, Flomenberg N, Pestell RG, Howell A, Sotgia F, Lisanti MP, Martinez-Outschoorn UE. Glutamine fuels a vicious cycle of autophagy in the tumor stroma and oxidative mitochondrial metabolism in epithelial cancer cells: implications for preventing chemotherapy resistance. Cancer Biol Ther. 2011;12:1085–1097. doi: 10.4161/cbt.12.12.18671.

Source: PubMed

3
購読する