Suppression of mTOR pathway and induction of autophagy-dependent cell death by cabergoline

Shao Jian Lin, Zhi Gen Leng, Yu Hang Guo, Lin Cai, Yu Cai, Ning Li, Han Bing Shang, Wei-Dong Le, Wei Guo Zhao, Zhe Bao Wu, Shao Jian Lin, Zhi Gen Leng, Yu Hang Guo, Lin Cai, Yu Cai, Ning Li, Han Bing Shang, Wei-Dong Le, Wei Guo Zhao, Zhe Bao Wu

Abstract

Cabergoline (CAB), the first-line drug for treatment of prolactinomas, is effective in suppressing prolactin hypersecretion, reducing tumor size, and restoring gonadal function. However, mechanisms for CAB-mediated tumor shrinkage are largely unknown. Here we report a novel cytotoxic mechanism for CAB. CAB induced formation of autophagosome in rat pituitary tumor MMQ and GH3 cells at the early stage through inhibiting mTOR pathway, resulting in higher conversion rates of LC3-I to LC3-II, GFP-LC3 aggregation, and increased autophagosome formation. Interestingly, CAB treatment augmented lysosome acidification and resulted in impaired proteolytic degradation within autolysosomes. This blocked the autophagic flux, leading to the accumulation of p62 aggregation and undigested autolysosomes. Knockdown of ATG7, ATG5, or Becn1, could significantly rescue the CAB-mediated cell death of MMQ cells (p < 0.05). CAB-induced autophagy and blockade of autophagy flux participated in antitumoral action in vivo. In conclusion, our study provides evidence that CAB concomitantly induces autophagy and inhibits the autophagic flux, leading to autophagy-dependent cell death. These findings elucidate novel mechanisms for CAB action.

Keywords: autophagic cell death; autophagic flux; autophagy; cabergoline; prolactinoma.

Conflict of interest statement

CONFLICTS OF INTEREST

No potential conflicts of interest were disclosed.

Figures

Figure 1. CAB induces both apoptosis and…
Figure 1. CAB induces both apoptosis and non-apoptosis cell death
A–B. Cell survival was determined by the MTS assay. C. MMQ and GH3 cells were treated with CAB as indicated for apoptosis assay by Annexin V-FTIC and PI double staining. D–E. MMQ and GH3 cells were treated with CAB as indicated, and total proteins analyzed by Western blot using antibodies against caspase-3, PARP and GAPDH. F. MMQ and GH3 cells were treated with CAB in the presence or absence of Z-VAD-FMK at different concentrations for 48 hours, and cell survival was determined by the MTS. G. Transmission electron microscopy (TEM) images with enlargements; boxes showed autophagic vacuoles in MMQ cells treated with CAB for 48 h, and a cell death morphology accompanied by large-scale autophagic vacuoles in the cytoplasm with the absence of chromatin condensation.
Figure 2. CAB suppresses the AKT/mTOR pathway
Figure 2. CAB suppresses the AKT/mTOR pathway
A. CAB-treated GH3 and MMQ cellular lysates were blotted with antibodies recognizing p-mTOR (Ser 2448), mTOR, p-4EBP1 (Thr37/46), 4EBP1, p-p70s6k(Thr389) and p70s6k. A mouse anti-GAPDH antibody was utilized as a loading control. Blots are representative of three independent experiments. B. Immunoblots analysis of p-mTOR, p-4EBP1 and GAPDH in MMQ cells treated CAB with or without rapamycin. C. MMQ cells were treated with CAB in the presence or absence of rapamycin (100 nM) for 24 hours, and cell survival was determined by the MTS.
Figure 3. CAB induces autophagy
Figure 3. CAB induces autophagy
A. Immunoblot analysis of LC3-I and LC3-II in GH3 and MMQ cells with or without CAB treatment at different time points. B. CAB treatment at 24 hours induced punctuative distribution of membrane-associated lipidated LC3-II in GH3-GFP-LC3 (left panels) and MMQ-GFP-LC3 (right panels) cells, observed with a confocal microscope. Scale bar represents 10 μm. Histogram shows the LC3 puncta from multiple experiments counting a total of 50 cells (mean ± SD). C. Electron micrographs of GH3 and MMQ cells with or without 24 h CAB treatment. Enlarged images of CAB treated cell (bottom parts) indicate autolysosomes (right). Histogram shows the autolysosome structures from multiple experiments counting a total of 50 cells (mean ± SD).
Figure 4. CAB blocks autophagic flux
Figure 4. CAB blocks autophagic flux
A. Immunoblotof p62 in MMQ (left panel) or GH3 (right panel) cells with or without CAB treatment at different time points. B. Analysis of p62 mRNA levels in MMQ (left panel) or GH3 (right panel) cells with or without CAB treatment at different time points.
Figure 5. CAB impairs lysosomal degradation within…
Figure 5. CAB impairs lysosomal degradation within autolysosomes
A. CAB-treated MMQ (GFP-LC3) cells stained with Lysotracker. Scale bar: 5 μm. B. Following CAB treatment for 24 h, cells were loaded with 1 μM Lysosensor Green and examined with confocal microscopy. Scale bar: 20 μm. C. Samples from B were analyzed by flow cytometry. D. MMQ cells treated with DMSO, CAB, BafA1, or CAB+BafA1were loaded with 1 μM Lysosensor Green for 24 h and analyzed by flow cytometry. E. MMQ cells treated with DMSO, CAB, BafA1, or CAB+BafA1 assayed for cell proliferation in 24 h. F. Immunoblots analysis of PARP in MMQ cells treated with DMSO, CAB, BafA1, or CAB+BafA1 for 6 h and 24 h. ***p < 0.001.
Figure 6. Silencing of autophagic key genes…
Figure 6. Silencing of autophagic key genes rescues CAB-induced cell death
A–D. MMQ cells were transfected with control (Ctrl) or with ATG7 siRNA for three days before treating with CAB or DMSO, followed by immunoblot analysis for LC3, p62, PARP and β-tubulin (A) Histogram shows the average number of autolysosome seen in electron microscope (B), cell proliferation assay (C), and Annexin-V/PI staining assay (D). E–F. MMQ cells were transfected with control (Ctrl) or with ATG5 or Becn1 siRNA for three days before treating with CAB or DMSO, followed by cell proliferation assay. At the end of incubation, the cell survival rates were determined by CellTiter-Glo®luminescent cell viability assay and the cell survival rate was calculated. Results were reported as the mean ± SD of three independent experiments performed in four replicates. **p < 0.01, ***p < 0.001.
Figure 7. The growth-suppressive effect of CAB…
Figure 7. The growth-suppressive effect of CAB on MMQ cells in vivo
A–C. CAB treatment inhibited the in vivo tumor growth of MMQ cells. The representative images for xenograft tumor on the nude mouse are shown in Figure 7A, and the tumor weight and tumor growth curve is shown in Figure 7B and 7C, respectively (n = 5). D. Immunoblot analyses showing p-4EBP1, p62, LC3 and β-tubulin in tumor samples. The protein on behalf of the average of the five tumor tissues.

References

    1. Colao A, Savastano S. Medical treatment of prolactinomas. Nature reviews Endocrinology. 2011;7:267–278.
    1. Bevan JS, Webster J, Burke CW, Scanlon MF. Dopamine agonists and pituitary tumor shrinkage. Endocrine reviews. 1992;13:220–240.
    1. Gillam MP, Molitch ME, Lombardi G, Colao A. Advances in the treatment of prolactinomas. Endocrine reviews. 2006;27:485–534.
    1. Wu ZB, Yu CJ, Su ZP, Zhuge QC, Wu JS, Zheng WM. Bromocriptine treatment of invasive giant prolactinomas involving the cavernous sinus: results of a long-term follow up. J Neurosurg. 2006;104:54–61.
    1. Casanueva FF, Molitch ME, Schlechte JA, Abs R, Bonert V, Bronstein MD, Brue T, Cappabianca P, Colao A, Fahlbusch R, Fideleff H, Hadani M, Kelly P, Kleinberg D, Laws E, Marek J, et al. Guidelines of the Pituitary Society for the diagnosis and management of prolactinomas. Clin Endocrinol (Oxf) 2006;65:265–273.
    1. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, Wass JA. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011;96:273–288.
    1. Molitch ME. Medical management of prolactin-secreting pituitary adenomas. Pituitary. 2002;5:55–65.
    1. Colao A, Di Sarno A, Sarnacchiaro F, Ferone D, Di Renzo G, Merola B, Annunziato L, Lombardi G. Prolactinomas resistant to standard dopamine agonists respond to chronic cabergoline treatment. J Clin Endocrinol Metab. 1997;82:876–883.
    1. Colao A, Lombardi G, Annunziato L. Cabergoline. Expert Opin Pharmacother. 2000;1:555–574.
    1. Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217.
    1. An JJ, Cho SR, Jeong DW, Park KW, Ahn YS, Baik JH. Anti-proliferative effects and cell death mediated by two isoforms of dopamine D2 receptors in pituitary tumor cells. Molecular and cellular endocrinology. 2003;206:49–62.
    1. Radl DB, Ferraris J, Boti V, Seilicovich A, Sarkar DK, Pisera D. Dopamine-induced apoptosis of lactotropes is mediated by the short isoform of D2 receptor. PLoS One. 2011;6:e18097.
    1. Al-Azzawi H, Yacqub-Usman K, Richardson A, Hofland LJ, Clayton RN, Farrell WE. Reversal of endogenous dopamine receptor silencing in pituitary cells augments receptor-mediated apoptosis. Endocrinology. 2011;152:364–373.
    1. Junn E, Mouradian MM. Apoptotic signaling in dopamine-induced cell death: the role of oxidative stress, p38 mitogen-activated protein kinase, cytochrome c and caspases. J Neurochem. 2001;78:374–383.
    1. Li Q, Su Z, Liu J, Cai L, Lu J, Lin S, Xiong Z, Li W, Zheng W, Wu J, Zhuge Q, Wu Z. Dopamine receptor D2S gene transfer improves the sensitivity of GH3 rat pituitary adenoma cells to bromocriptine. Molecular and cellular endocrinology. 2014;382:377–384.
    1. Rowther FB, Richardson A, Clayton RN, Farrell WE. Bromocriptine and dopamine mediate independent and synergistic apoptotic pathways in pituitary cells. Neuroendocrinology. 2010;91:256–267.
    1. De Duve C, Wattiaux R. Functions of lysosomes. Annu Rev Physiol. 1966;28:435–492.
    1. Weckman A, Di Ieva A, Rotondo F, Syro LV, Ortiz LD, Kovacs K, Cusimano MD. Autophagy in the endocrine glands. Journal of molecular endocrinology. 2014;52:R151–163.
    1. Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science. 2000;290:1717–1721.
    1. Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075.
    1. Hale AN, Ledbetter DJ, Gawriluk TR, Rucker EB., 3rd Autophagy: regulation and role in development. Autophagy. 2013;9:951–972.
    1. Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science. 2004;306:990–995.
    1. Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol. 2008;9:1004–1010.
    1. Radl DB, Zarate S, Jaita G, Ferraris J, Zaldivar V, Eijo G, Seilicovich A, Pisera D. Apoptosis of lactotrophs induced by D2 receptor activation is estrogen dependent. Neuroendocrinology. 2008;88:43–52.
    1. Kim YC, Guan KL. mTOR: a pharmacologic target for autophagy regulation. The Journal of clinical investigation. 2015;125:25–32.
    1. Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell. 2010;140:313–326.
    1. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, Johansen T. Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol. 2009;452:181–197.
    1. Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy. 2007;3:542–545.
    1. Levine B, Yuan J. Autophagy in cell death: an innocent convict? J Clin Invest. 2005;115:2679–2688.
    1. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122:927–939.
    1. Lenardo MJ, McPhee CK, Yu L. Autophagic cell death. Methods Enzymol. 2009;453:17–31.
    1. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA, Ahn HJ, Ait-Mohamed O, Ait-Si-Ali S, Akematsu T, Akira S, Al-Younes HM, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012;8:445–544.
    1. Tang Y, Hamed HA, Cruickshanks N, Fisher PB, Grant S, Dent P. Obatoclax and lapatinib interact to induce toxic autophagy through NOXA. Molecular pharmacology. 2012;81:527–540.
    1. Rong Y, McPhee CK, Deng S, Huang L, Chen L, Liu M, Tracy K, Baehrecke EH, Yu L, Lenardo MJ. Spinster is required for autophagic lysosome reformation and mTOR reactivation following starvation. Proc Natl Acad Sci U S A. 2011;108:7826–7831.
    1. Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB, Tsujimoto Y. Role of Bcl-2 family proteins in a non-apoptotic programmed cell death dependent on autophagy genes. Nat Cell Biol. 2004;6:1221–1228.
    1. Yu L, Alva A, Su H, Dutt P, Freundt E, Welsh S, Baehrecke EH, Lenardo MJ. Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science. 2004;304:1500–1502.
    1. Espert L, Denizot M, Grimaldi M, Robert-Hebmann V, Gay B, Varbanov M, Codogno P, Biard-Piechaczyk M. Autophagy is involved in T cell death after binding of HIV-1 envelope proteins to CXCR4. J Clin Invest. 2006;116:2161–2172.
    1. Turcotte S, Chan DA, Sutphin PD, Hay MP, Denny WA, Giaccia AJ. A molecule targeting VHL-deficient renal cell carcinoma that induces autophagy. Cancer Cell. 2008;14:90–102.
    1. Shi S, Wang Q, Xu J, Jang JH, Padilla MT, Nyunoya T, Xing C, Zhang L, Lin Y. Synergistic anticancer effect of cisplatin and Chal-24 combination through IAP and c-FLIPL degradation, Ripoptosome formation and autophagy-mediated apoptosis. Oncotarget. 2015;6:1640–1651.
    1. Hsueh YS, Chang HH, Chiang NJ, Yen CC, Li CF, Chen LT. MTOR inhibition enhances NVP-AUY922-induced autophagy-mediated KIT degradation and cytotoxicity in imatinib-resistant gastrointestinal stromal tumors. Oncotarget. 2014;5:11723–11736.
    1. Gump JM, Staskiewicz L, Morgan MJ, Bamberg A, Riches DW, Thorburn A. Autophagy variation within a cell population determines cell fate through selective degradation of Fap-1. Nature cell biology. 2014;16:47–54.
    1. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gelinas C, Fan Y, Nelson DA, Jin S, White E. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell. 2006;10:51–64.
    1. Ge YY, Shi Q, Zheng ZY, Gong J, Zeng C, Yang J, Zhuang SM. MicroRNA-100 promotes the autophagy of hepatocellular carcinoma cells by inhibiting the expression of mTOR and IGF-1R. Oncotarget. 2014;5:6218–6228.
    1. Gonzalez P, Mader I, Tchoghandjian A, Enzenmuller S, Cristofanon S, Basit F, Debatin KM, Fulda S. Impairment of lysosomal integrity by B10, a glycosylated derivative of betulinic acid, leads to lysosomal cell death and converts autophagy into a detrimental process. Cell Death Differ. 2012;19:1337–1346.
    1. Rozman-Pungercar J, Kopitar-Jerala N, Bogyo M, Turk D, Vasiljeva O, Stefe I, Vandenabeele P, Bromme D, Puizdar V, Fonovic M, Trstenjak-Prebanda M, Dolenc I, Turk V, Turk B. Inhibition of papain-like cysteine proteases and legumain by caspase-specific inhibitors: when reaction mechanism is more important than specificity. Cell death and differentiation. 2003;10:881–888.
    1. Zheng X, Chu F, Mirkin BL, Sudha T, Mousa SA, Rebbaa A. Role of the proteolytic hierarchy between cathepsin L, cathepsin D and caspase-3 in regulation of cellular susceptibility to apoptosis and autophagy. Biochimica et biophysica acta. 2008;1783:2294–2300.
    1. Martin DN, Baehrecke EH. Caspases function in autophagic programmed cell death in Drosophila. Development. 2004;131:275–284.
    1. Akdemir F, Farkas R, Chen P, Juhasz G, Medved'ova L, Sass M, Wang L, Wang X, Chittaranjan S, Gorski SM, Rodriguez A, Abrams JM. Autophagy occurs upstream or parallel to the apoptosome during histolytic cell death. Development. 2006;133:1457–1465.

Source: PubMed

3
Subscribe