Skip to main content

Advertisement

Log in

WIN induces apoptotic cell death in human colon cancer cells through a block of autophagic flux dependent on PPARγ down-regulation

  • Original Paper
  • Published:
Apoptosis Aims and scope Submit manuscript

Abstract

Cannabinoids have been reported to possess anti-tumorigenic activity in cancer models although their mechanism of action is not well understood. Here, we show that the synthetic cannabinoid WIN55,212-2 (WIN)-induced apoptosis in colon cancer cell lines is accompanied by endoplasmic reticulum stress induction. The formation of acidic vacuoles and the increase in LC3-II protein indicated the involvement of autophagic process which seemed to play a pro-survival role against the cytotoxic effects of the drug. However, the enhanced lysosomal membrane permeabilization (LMP) blocked the autophagic flux after the formation of autophagosomes as demonstrated by the accumulation of p62 and LC3, two markers of autophagic degradation. Data also provided evidence for a role for nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) in cannabinoid signalling. PPARγ expression, at both protein and mRNA levels, was significantly down-regulated after WIN treatment and its inhibition, either by specific antagonists or by down-regulation via gene silencing, induced effects on cell viability as well as on ER stress and autophagic markers similar to those obtained in the presence of WIN. Moreover, the observation that the increase in p62 level and the induction of LMP were also modified by PPARγ antagonists seemed to indicate that PPARγ down-regulation was crucial to determinate the block of autophagic flux, thus confirming the critical role of PPARγ in WIN action. In conclusion, at our knowledge, our results are the first to show that the reduction of PPARγ levels contributes to WIN-induced colon carcinoma cell death by blocking the pro-survival autophagic response of cells.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Abbreviations

ER:

Endoplasmic reticulum

PPARγ:

Peroxisome proliferator-activated receptor γ

CHOP:

CCAAT/enhancer binding protein homologous protein

GRP78:

Glucose-regulated protein 78/Binding immunoglobulin protein

TRB3:

Tribbles-related protein 3

LC3 (MAP1LC3):

Microtubule-associated protein 1 light chain 3

PARP:

Poly-(ADP-ribose) polymerase

WIN:

(WIN55,212-2) R-[2,3-dihydro-5-methyl-3[(4-morpholinyl)methyl] pyrrolo[1,2,3,-de]-1,4-benzoxazin-6-yl]-1-naphthalenyl methanone mesylate

MTT:

3-(4,5)-Dimethylthiahiazo(-z-y1)-3,5-diphenytetrazoliumromide

MDC:

Monodansylcadaverine

PI:

Propidium iodide

3-MA:

3-Methyladenine

AO:

Acridine orange

CCCP:

Carbonylcyanide m-chlorophenylhydrazone

LMP:

Lysosomal membrane permeabilization

Baf A1:

Bafilomycin A1

References

  1. Di Marzo V, Petrocellis LD (2006) Plant, synthetic, and endogenous cannabinoids in medicine. Annu Rev Med 57:553–574

    Article  PubMed  Google Scholar 

  2. Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham M, Mackie K, Martin BR, Mechoulam R, Pertwee RG (2002) International Union of Pharmacology. XXVII. Classification of cannabinoid receptor. Pharmacol Rev 54:161–202

    Article  CAS  PubMed  Google Scholar 

  3. Herkenham M, Lynn AB, Johnson MR, Melvin LS, De Costa BR, Rice KC (1991) Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci 11:563–583

    CAS  PubMed  Google Scholar 

  4. Van der Stelt M, Di Marzo V (2005) Cannabinoid receptors and their role in neuroprotection. NeuroMol Med 7:37–50

    Article  Google Scholar 

  5. Dainese E, Oddi S, Bari M, Maccarrone M (2007) Modulation of the endocannabinoid system by lipid rafts. Curr Med Chem 14:2702–2715

    Article  CAS  PubMed  Google Scholar 

  6. Guzmán M, Sánchez C, Galve-Roperh I (2002) Cannabinoids and cell fate. Pharmacol Ther 95:175–184

    Article  PubMed  Google Scholar 

  7. Qamri Z, Preet A, Leone G, Preet A, Barsky SH, Ganju RK (2009) Synthetic cannabinoid receptor agonists inhibit tumor growth and metastasis of breast cancer. Mol Cancer Ther 8:117–129

    Article  Google Scholar 

  8. Sarfaraz S, Afaq F, Adhami VM, Mukhtar H (2005) Cannabinoid receptor as a novel target for the treatment of prostate cancer. Cancer Res 65:1635–1641

    Article  CAS  PubMed  Google Scholar 

  9. Sarker KP, Biswas KK, Yamakuchi M, Lee KY, Hahiguchi T, Kracht M, Kitajima I, Maruyama I (2003) ASK1-p38 MAPK/JNK signaling cascade mediates anandamide-induced PC12 cell death. J Neurochem 85:50–61

    Article  CAS  PubMed  Google Scholar 

  10. Velasco G, Galve-Roperh I, Sánchez C, Blázquez C, Guzmán M (2004) Hypothesis: cannabinoid therapy for the treatment of gliomas? Neuropharmacol 47:315–323

    Article  CAS  Google Scholar 

  11. Guzmán M (2003) Cannabinoids: potential anticancer agents. Nat Rev Cancer 3:745–755

    Article  PubMed  Google Scholar 

  12. Salazar M, Carracedo A, Salanueva IJ, Hernández-Tiedra S, Lorente M, Egia A, Vázquez P, Blázquez C, Torres S, García S, Nowak J, Fimia GM, Piacentini M, Cecconi F, Pandolfi PP, González-Feria L, Iovanna JL, Guzmán M, Boya P, Velasco G (2009) Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest 119:1359–1372

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Yorimitsu T, Nail U, Yang Z, Klionsky DJ (2006) Endoplasmic reticulum stress triggers autophagy. J Biol Chem 281:30299–30304

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Yorimitsu T, Klionsky DJ (2007) Endoplasmic reticulum stress: a new pathway to induce autophagy. Autophagy 3(2):160–162

    CAS  PubMed  Google Scholar 

  15. Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, Ogawa S, Kaufman RJ, Kominami E, Momoi T (2007) ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ 14(2):230–239

    Article  CAS  PubMed  Google Scholar 

  16. Salazar M, Lorente M, García-Taboada E, Hernández-Tiedra S, Davila D, Francis SE, Guzmán M, Kiss-Toth E, Velasco G (2013) The pseudokinase tribbles homologue-3 plays a crucial role in cannabinoid anticancer action. Biochim Biophys Acta 1831(10):1573–1578

    Article  CAS  PubMed  Google Scholar 

  17. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K (2012) Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8:445–544

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Spiegelman BM (1998) PPAR-γ: adipogenic regulator and thiazolidinedione receptor. Diabetes 47:507–514

    Article  CAS  PubMed  Google Scholar 

  19. Tontonoz P, Hu E, Spiegelman BM (1994) Stimulation of adipogenesis in fibroblasts by PPAR γ2, a lipid-activated transcription factor. Cell 79:1147–1156

    Article  CAS  PubMed  Google Scholar 

  20. Han SW, Roman J (2008) Activated PPARgamma targets surface and intracellular signals that inhibit the proliferation of lung carcinoma cells. PPAR Res. doi:10.1155/2008/254108

    PubMed Central  PubMed  Google Scholar 

  21. Kota BP, Huang TH, Roufogalis BD (2005) An overview on biological mechanisms of PPARs. Pharmacol Res 51:85–94

    Article  CAS  PubMed  Google Scholar 

  22. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM (1995) The nuclear receptor superfamily: the second decade. Cell 83:835–839

    Article  CAS  PubMed  Google Scholar 

  23. Yang FG, Zhang ZW, Xin DQ, Shi CJ, Wu JP, Guo YL, Guan YF (2005) Peroxisome proliferator-activated receptor gamma ligands induce cell cycle arrest and apoptosis in human renal carcinoma cell lines. Acta Pharmacol Sin 26:753–761

    Article  CAS  PubMed  Google Scholar 

  24. Lee JJ, Drakaki A, Iliopoulos D, Struhl K (2011) MiR-27b targets PPARγ to inhibit growth, tumor progression and the inflammatory response in neuroblastoma cells. Oncogene. doi:10.1038/onc.2011.543

    Google Scholar 

  25. Giuliano M, Pellerito O, Portanova P, Calvaruso G, Santulli A, De Blasio A, Vento R, Tesoriere G (2009) Apoptosis induced in HepG2 cells by the synthetic cannabinoid WIN: involvement of the transcription factor PPARgamma. Biochimie 91:457–465

    Article  CAS  PubMed  Google Scholar 

  26. O’Sullivan SE (2007) Cannabinoids go nuclear: evidence for activation of peroxisome proliferator-activated receptors. Br J Pharmacol 152:576–582

    Article  PubMed Central  PubMed  Google Scholar 

  27. Rubino S, Portanova P, Girasolo A, Calvaruso G, Orecchio S, Stocco GC (2009) Synthetic, structural and biochemical studies of polynuclear platinum(II) complexes with heterocyclic ligands. Eur J Med Chem 44:1041–1048

    Article  CAS  PubMed  Google Scholar 

  28. Drago-Ferrante R, Santulli A, Di Fiore R, Giuliano M, Calvaruso G, Tesoriere G, Vento R (2008) Low doses of paclitaxel potently induce apoptosis in human retinoblastoma Y79 cells by up-regulating E2F1. Int J Oncol 33:677–687

    CAS  PubMed  Google Scholar 

  29. Zdolsek JM, Olsson GM, Brunk UT (1999) Photooxidative damage to lysosomes of cultured macrophages by acridine orange. Photochem Photobiol 51:67–76

    Article  Google Scholar 

  30. Antunes F, Cadenas E, Brunk UT (2001) Apoptosis induced by exposure to a low steady-state concentration of H2O2 is a consequence of lysosomal rupture. Biochem J 356:549–555

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Calvaruso G, Giuliano M, Portanova P, Pellerito O, Vento R, Tesoriere G (2007) Hsp72 controls bortezomib-induced HepG2 cell death via interaction with pro-apoptotic factors. Oncol Rep 18:447–450

    CAS  PubMed  Google Scholar 

  32. Tsukahara T, Haniu H, Matsuda Y (2013) PTB-associated splicing factor (PSF) is a PPARγ-binding protein and growth regulator of colon cancer cells. PLoS One 8(3):e58749. doi:10.1371/journal.pone.0058749

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Fu M, Zhang J, Lin Y, Zhu X, Zhao L, Ahmad M, Ehrengruber MU, Chen YE (2003) Early stimulation and late inhibition of peroxisome proliferator-activated receptor gamma (PPARgamma) gene expression by transforming growth factor beta in human aortic smooth muscle cells: role of early growth-response factor-1 (Egr-1), activator protein 1 (AP1) and Smads. Biochem J 370:1019–1025

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. He CQ, Ding NZ, Fan W (2008) YY1 repressing peroxisome proliferator-activated receptor delta promoter. Mol Cell Biochem 308:247–252

    Article  CAS  PubMed  Google Scholar 

  35. Huang Y, Yang X, Wu Y, Jing W, Cai X, Tang W, Liu L, Liu Y, Grottkau BE, Lin Y (2010) Gamma-secretase inhibitor induces adipogenesis of adipose-derived stem cells by regulation of Notch and PPAR-gamma. Cell Prolif 43:147–156

    Article  CAS  PubMed  Google Scholar 

  36. Ohoka N, Yoshii S, Hattori T, Onozaki K, Hayashi H (2005) TRB3, a novel ER stress-inducible gene, is induced via ATF4-CHOP pathway and is involved in cell death. EMBO J 24:243–255

    Article  Google Scholar 

  37. Carnero A (2010) The PKB/AKT pathway in cancer. Curr Pharm Des 16:34–44

    Article  CAS  PubMed  Google Scholar 

  38. Vara D, Salazar M, Olea-Herrero N, Guzmán M, Velasco G, Díaz-Laviada I (2011) Anti-tumoral action of cannabinoids on hepatocellular carcinoma: role of AMPK-dependent activation of autophagy. Cell Death Differ 18:1099–1111

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Biederbick A, Kern HF, Elsässer HP (1995) Monodansylcadaverine (MDC) is a specific in vivo marker for autophagic vacuoles. Eur J Cell Biol 6:3–14

    Google Scholar 

  40. Seglen PO, Gordon PB (1982) 3-Methyladenine: specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc Natl Acad Sci USA 79:1889–1892

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Kang JH, Chang YC, Maurizi MR (2012) 4-O-carboxymethyl ascochlorin causes ER stress and induced autophagy in human hepatocellular carcinoma cells. J Biol Chem 287:15661–15671

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Adiseshaiah PP, Clogston JD, McLeland CB, Rodriguez J, Potter TM, Neun BW, Skoczen SL, Shanmugavelandy SS, Kester M, Stern ST, McNeil SE (2013) Synergistic combination therapy with nanoliposomal C6-ceramide and vinblastine is associated with autophagy dysfunction in hepatocarcinoma and colorectal cancer models. Cancer Lett 337:254–265. doi:10.1016/j.canlet.2013.04.034

    Article  CAS  PubMed  Google Scholar 

  43. Sreevalsan S, Joseph S, Jutooru I, Chadalapaka G, Safe SH (2011) Induction of apoptosis by cannabinoids in prostate and colon cancer cells is phosphatase dependent. Anticancer Res 31:3799–3807

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Pellerito O, Calvaruso G, Portanova P, De Blasio A, Santulli A, Vento R, Tesoriere G, Giuliano M (2010) The synthetic cannabinoid WIN55,212-2 sensitizes hepatocellular carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by activating p8/CCAAT/enhancer binding protein homologous protein (CHOP)/death receptor 5 (DR5) axis. Mol Pharmacol 77:854–863

    Article  CAS  PubMed  Google Scholar 

  45. Vara D, Morell C, Rodríguez-Henche N, Diaz-Laviada I (2013) Involvement of PPARγ in the antitumoral action of cannabinoids on hepatocellular carcinoma. Cell Death Dis 4:e618. doi:10.1038/cddis.2013.141

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Park SH, Choi HJ, Yang H, Do KH, Kim J, Lee DW, Moon Y (2010) Endoplasmic reticulum stress-activated C/EBP homologous protein enhances nuclear factor-kappaB signals via repression of peroxisome proliferator-activated receptor gamma. J Biol Chem 285:35330–35339

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by University of Palermo [Grant ORPA07EZ5Z and ORPA06F3TB].

Conflict of interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michela Giuliano.

Additional information

O. Pellerito and A. Notaro have contributed equally to this study.

G. Calvaruso and M. Giuliano share senior co-authorship.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10495_2014_985_MOESM1_ESM.jpg

Supplementary Fig. 1. Complete depolarization of mitochondria was evaluated by means of carbonyl cyanide 3-chlorophenylhydrazone (CCCP). 50 μM CCCP was added to HT29 cells for 10 min at room temperature. Then, mitochondrial transmembrane potential (Δψm) dissipation was quantified by flow cytometry in the presence of the lipophilic dye DiOC6, as reported in “Materials and methods” section. Filled profile: untreated cells; continuous line: 50 μM CCCP (JPEG 106 kb)

10495_2014_985_MOESM2_ESM.jpg

Supplementary Fig. 2. Uptake of propidium iodide and cell cycle profile of WIN-treated HT29 cells. a Cell were treated for 36 h with 10 μM WIN. The percentage of dead cells was evaluated by cytofluorimetric analysis of the uptake of propidium iodide (PI), indicative of a loss of plasma membrane integrity, as reported in “Materials and methods” section. b Effects of WIN and/or GW9662 on cell cycle distribution of HT29 cells. Flow cytometric analysis was carried out on propidium iodide-stained cells treated for 16 h with 10 μM WIN or 36 h with 10 μM WIN and/or 60 μM GW9662. The percentage of cells in each phase of the cell cycle was calculated as reported in “Materials and methods” section using Expo32 software. **p<0.001 (JPEG 465 kb)

10495_2014_985_MOESM3_ESM.jpg

Supplementary Fig. 3. Representative western blots of different transcription factors involved in PPARγ expression in HT29 cells treated for 36 h in the presence of 10 μM WIN. Cell lysates were analysed via immunoblotting using specific antibodies as reported in “Materials and methods” section. Actin blots were included as a loading control (JPEG 216 kb)

10495_2014_985_MOESM4_ESM.jpg

Supplementary Fig. 4. Effects of PPARγ antagonist T007 in HT29 cells. a Table 1 Determination of mitochondrial transmembrane potential (Δψm) dissipation and DNA fragmentation in HT29 cells treated with 10 μM WIN and/or 25 μM T007. Δψm and DNA fragmentation were evalualted by means of flow cytometry after 16 h or 36 h of treatment, respectively, as reported in “Materials and methods” section. b Effects of WIN and/or T007 on HT29 cell viability. Cells were incubated for the indicated times with 10 μM WIN and/or 25 μM T007. Cell survival was estimated by MTT assay, as reported in “Materials and methods” section, and expressed as the percentage of control cells. c-d Representative western blots of PPARγ (c), CHOP, Beclin-1, p62 and LC3 (d) levels. After treatment for 36 h with 10 μM WIN and/or 25 μM T007, cell lysates were analysed via immunoblotting using specific antibodies as reported in “Materials and methods” section. Actin blots were included as a loading control. e Effects of T007 on WIN-induced lysosomal destabilization. HT29 cells were loaded with 5 μg/ml AO for 15 min before treatment with 10 μM WIN and/or 25 μM T007 for 5 h. Induction of lysosomal destabilization was evaluated under fluorescence microscopy. Control cells show punctate red staining of acidic organelles red fluorescence while in WIN and/or T007-treated cells a diffuse green fluorescence is observed. The data shown are from one of three experiments giving essentially the same results. **p<0.001 (JPEG 619 kb)

10495_2014_985_MOESM5_ESM.jpg

Supplementary Fig. 5. Effects of anti-Beclin-1 siRNA on autophagic markers and on HT29 cell viability after WIN treatment. a Cells were transfected with siRNA control (siScr) or specific anti-Beclin-1 siRNA (siBeclin-1) as reported in “Materials and methods” section and treated with 10 μM WIN for 16 h. Then, the level of Beclin-1 and LC3 were analysed by western blot. Actin blots were included as a loading control. b Effects of siBeclin-1 on viability of WIN-treated HT29 cells. Silenced cells were incubated for 16 h with 10 μM WIN, cell survival was estimated by MTT assay, as reported in “Materials and methods” section, and expressed as the percentage of control cells. *p<0.01; **p<0.001 (JPEG 304 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Pellerito, O., Notaro, A., Sabella, S. et al. WIN induces apoptotic cell death in human colon cancer cells through a block of autophagic flux dependent on PPARγ down-regulation. Apoptosis 19, 1029–1042 (2014). https://doi.org/10.1007/s10495-014-0985-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10495-014-0985-0

Keywords

Navigation