Skip to main content
Log in

O-GlcNAc modification is essential for the regulation of autophagy in Drosophila melanogaster

  • Research Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

O-GlcNAcylation is a dynamic post-translational modification that takes place on ser/thr residues of nucleocytoplasmic proteins. O-GlcNAcylation regulates almost all cellular events as a nutrient sensor, a transcriptional and translational regulator, and a disease-related factor. Although the role of O-GlcNAcylation in insulin signaling and metabolism are well established, the relationship between O-GlcNAcylation and autophagy is largely unknown. Here, we manipulated O-GlcNAcylation in Drosophila and found that it regulates autophagy through Akt/dFOXO signaling. We demonstrate that O-GlcNAcylation and the levels of O-GlcNAc transferase (OGT) are increased during starvation. Furthermore, Atg proteins and autolysosomes are increased in OGT-reduced flies without fasting. Atg proteins and autophagosomes are reduced in OGT-overexpressing flies. Our results suggest that not only autophagy gene expression but also autophagic structures are regulated by OGT through Akt and dFOXO. These data imply that O-GlcNAcylation is important in modulating autophagy as well as insulin signaling in Drosophila.

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

Similar content being viewed by others

References

  1. Dias WB, Cheung WD, Hart GW (2012) O-GlcNAcylation of kinases. Biochem Biophys Res Commun 422:224–228

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  2. Ruan HB, Singh JP, Li MD, Wu J, Yang X (2013) Cracking the O-GlcNAc code in metabolism. Trends Endocrinol Metab 24:301–309

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Zachara NE, Hart GW (2004) O-GlcNAc a sensor of cellular state: the role of nucleocytoplasmic glycosylation in modulating cellular function in response to nutrition and stress. Biochim Biophys Acta 1673:13–28

    Article  CAS  PubMed  Google Scholar 

  4. Mehrpour M, Esclatine A, Beau I, Codogno P (2010) Autophagy in health and disease. 1. Regulation and significance of autophagy: an overview. Am J Physiol Cell Physiol 298:C776–C785

    Article  CAS  PubMed  Google Scholar 

  5. Mizushima N, Komatsu M (2011) Autophagy: renovation of cells and tissues. Cell 147:728–741

    Article  CAS  PubMed  Google Scholar 

  6. Rusten TE, Lindmo K, Juhasz G, Sass M, Seglen PO, Brech A, Stenmark H (2004) Programmed autophagy in the Drosophila fat body is induced by ecdysone through regulation of the PI3K pathway. Dev Cell 7:179–192

    Article  CAS  PubMed  Google Scholar 

  7. Fujitani Y, Ebato C, Uchida T, Kawamori R, Watada H (2009) Beta-cell autophagy: a novel mechanism regulating beta-cell function and mass: lessons from beta-cell-specific Atg7-deficient mice. Islets 1:151–153

    Article  PubMed  Google Scholar 

  8. Erickson JR, Pereira L, Wang L, Han G, Ferguson A, Dao K, Copeland RJ, Despa F, Hart GW, Ripplinger CM et al (2013) Diabetic hyperglycaemia activates CaMKII and arrhythmias by O-linked glycosylation. Nature 502:372–376

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Liu K, Paterson AJ, Chin E, Kudlow JE (2000) Glucose stimulates protein modification by O-linked GlcNAc in pancreatic beta cells: linkage of O-linked GlcNAc to beta cell death. Proc Natl Acad Sci USA 97:2820–2825

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Ma J, Hart GW (2013) Protein O-GlcNAcylation in diabetes and diabetic complications. Expert Rev Proteomics 10:365–380

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Marsh SA, Powell PC, Dell’italia LJ, Chatham JC (2013) Cardiac O-GlcNAcylation blunts autophagic signaling in the diabetic heart. Life Sci 92(11):648–656

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  12. Wang P, Lazarus BD, Forsythe ME, Love DC, Krause MW, Hanover JA (2012) O-GlcNAc cycling mutants modulate proteotoxicity in Caenorhabditis elegans models of human neurodegenerative diseases. Proc Natl Acad Sci USA 109:17669–17674

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Kumar A, Singh PK, Parihar R, Dwivedi V, Lakhotia SC, Ganesh S (2014) Decreased O-linked GlcNAcylation protects from cytotoxicity mediated by huntingtin exon1 protein fragment. J Biol Chem 289:13543–13553

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Park S, Park SH, Baek JY, Jy YJ, Kim KS, Roth J, Cho JW, Choe KM (2011) Protein O-GlcNAcylation regulates Drosophila growth through the insulin signaling pathway. Cell Mol Life Sci 68:3377–3384

    Article  CAS  PubMed  Google Scholar 

  15. Sinclair DA, Syrzycka M, Macauley MS, Rastgardani T, Komljenovic I, Vocadlo DJ, Brock HW, Honda BM (2009) Drosophila O-GlcNAc transferase (OGT) is encoded by the Polycomb group (PcG) gene, super sex combs (sxc). Proc Natl Acad Sci USA 106:13427–13432

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Gupta VK, Scheunemann L, Eisenberg T, Mertel S, Bhukel A, Koemans TS, Kramer JM, Liu KS, Schroeder S, Stunnenberg HG et al (2013) Restoring polyamines protects from age-induced memory impairment in an autophagy-dependent manner. Nat Neurosci 16:1453–1460

    Article  CAS  PubMed  Google Scholar 

  17. Mizushima N, Yoshimori T, Ohsumi Y (2011) The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol 27:107–132

    Article  CAS  PubMed  Google Scholar 

  18. Nezis IP, Shravage BV, Sagona AP, Lamark T, Bjorkoy G, Johansen T, Rusten TE, Brech A, Baehrecke EH, Stenmark H (2010) Autophagic degradation of dBruce controls DNA fragmentation in nurse cells during late Drosophila melanogaster oogenesis. J Cell Biol 190:523–531

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Wang B, Moya N, Niessen S, Hoover H, Mihaylova MM, Shaw RJ, Yates JR 3rd, Fischer WH, Thomas JB, Montminy M (2011) A hormone-dependent module regulating energy balance. Cell 145:596–606

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Sengupta A, Molkentin JD, Yutzey KE (2009) FoxO transcription factors promote autophagy in cardiomyocytes. J Biol Chem 284:28319–28331

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Bai Hua, Kang Ping, Hernandez Ana Maria, Tatar Marc (2013) Activin signaling targeted by insulin/dFOXO regulates aging and muscle proteostasis in Drosophila. PLoS Genet 9(11):e1003941

    Article  PubMed Central  PubMed  Google Scholar 

  22. Housley MP, Rodgers JT, Udeshi ND, Kelly TJ, Shabanowitz J, Hunt DF, Puigserver P, Hart GW (2008) O-GlcNAc regulates FoxO activation in response to glucose. J Biol Chem 283:16283–16292

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Juhasz G, Puskas LG, Komonyi O, Erdi B, Maroy P, Neufeld TP, Sass M (2007) Gene expression profiling identifies FKBP39 as an inhibitor of autophagy in larval Drosophila fat body. Cell Death Differ 14:1181–1190

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Villa-Cuesta E, Sage BT, Tatar M (2010) A role for Drosophila dFoxO and dFoxO 5′UTR internal ribosomal entry sites during fasting. PloS One 5(7):e11521

    Article  PubMed Central  PubMed  Google Scholar 

  25. Kim EY, Jeong EH, Park S, Jeong HJ, Edery I, Cho JW (2012) A role for O-GlcNAcylation in setting circadian clock speed. Genes Dev 26:490–502

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Scott RC, Schuldiner O, Neufeld TP (2004) Role and regulation of starvation-induced autophagy in the Drosophila fat body. Dev Cell 7:167–178

    Article  CAS  PubMed  Google Scholar 

  27. Juhasz G, Csikos G, Sinka R, Erdelyi M, Sass M (2003) The Drosophila homolog of Aut1 is essential for autophagy and development. FEBS Lett 543:154–158

    Article  CAS  PubMed  Google Scholar 

  28. Thummel CS (2001) Steroid-triggered death by autophagy. Bioessays 23:677–682

    Article  CAS  PubMed  Google Scholar 

  29. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S et al (2010) Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 90:1383–1435

    Article  CAS  PubMed  Google Scholar 

  30. Yuzwa SA, Macauley MS, Heinonen JE, Shan X, Dennis RJ, He Y, Whitworth GE, Stubbs KA, McEachern EJ, Davies GJ et al (2008) A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat Chem Biol 4:483–490

    Article  CAS  PubMed  Google Scholar 

  31. Puig O, Marr MT, Ruhf ML, Tjian R (2003) Control of cell number by Drosophila FOXO: downstream and feedback regulation of the insulin receptor pathway. Genes Dev 17:2006–2020

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Lee KS, Kwon OY, Lee JH, Kwon K, Min KJ, Jung SA, Kim AK, You KH, Tatar M, Yu K (2008) Drosophila short neuropeptide F signalling regulates growth by ERK-mediated insulin signalling. Nat Cell Biol 10(4):468–475

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We thank K. M. Choe, and K. Yu for fly stocks and DNA clones, and O. Puig and K. Yu for providing the anti dFOXO antibody. We are also grateful to other members of the Cho and Choe laboratories for helpful discussions. This work is partly supported by the Graduate School of Yonsei University. This work was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (NRF-2013R1A2A1A01008067) to J. W. C.

Conflict of interest

The authors declare that they have no conflicts of interest to declare.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Jürgen Roth or Jin Won Cho.

Additional information

J. Roth and J. W. Cho are co-corresponding authors.

Electronic supplementary material

Below is the link to the electronic supplementary material.

18_2015_1889_MOESM1_ESM.tif

Supplementary material 1 Figure S1. The effects of increased OGT or OGT knockdown on overall O-GlcNAc levels. (A) Larval lysates from UAS-OGT and Act>OGT in feeding and fasting were analyzed by immunoblot with RL2 antibody. (B) Larvae lysates used for immunoblotting from UAS-OGTi and Act>OGTi in feeding and fasting to detect overall O-GlcNAc levels (TIFF 7877 kb)

18_2015_1889_MOESM2_ESM.tif

Supplementary material 2 Figure S2. The effect of O-GlcNAcylation on protein levels of Atg8a and the number of lysosomes. (A) S2 cells were transfected with pMT-FLAG-dogt for 48 h and harvested for immunoblotting. Anti-RL2, anti-FLAG, anti-β-actin, and anti-Atg8a antibodies are used. (B) Quantification of the amount of Atg8a in (A) (*p<0.05, student’s t-test). (C) A morphometric evaluation of EM images (***p<0.001, student’s t-test). TEM reveals many lysosomes in OGT-overexpressing larval fat bodies (TIFF 10380 kb)

18_2015_1889_MOESM3_ESM.tif

Supplementary material 3 Figure S3. O-GlcNAcylated proteins of the autophagy pathway. (A) S2 cells were treated with 10μM PP242 for 4 h to induce autophagy. Representative western blots showing β-actin and Atg8a in control and PP242-treated S2 cells. Lysates were prepared and analyzed either directly by immunoblotting (Input) or following pull-down with sWGA-agarose to detect O-GlcNAc modification on dFOXO. (B) WT mice were treated with the O-GlcNAcase inhibitor, thiamet-G (TG) for 7 days to increase total O-GlcNAcylation. Total O-GlcNAcylation of control and thiamet-G injected mouse brain. (C) To demonstrate O-GlcNAcylation on Atg7, Atg7 was immunoprecipitated using specific O-GlcNAc antibody followed by Western blotting with anti-Atg7 antibody. Brain lysates were prepared and analyzed either directly by immunoblotting or following pull-down with sWGA agarose to detect O-GlcNAcylation on Atg7, LC3, and Atg5. β-actin was used as a loading control. (D) Control and PP242-treated S2 cell lysates were prepared and analyzed either directly by immunoblotting as input or following pull-down with sWGA agarose to detect O-GlcNAcylation on dAtg5 and Atg8a. β-actin was used as a loading control. (E) Whole cell lysate of GFP-mCherry-Atg8a-expressing larvae with feeding or fasting. Larval lysates were prepared and analyzed either directly by immunoblotting (input) or following pull-down with sWGA agarose to detect O-GlcNAcylation on Atg8a using mCherry and GFP antibodies. Upper arrow is mCherry-GFP-Atg8a and lower arrow is free mCherry. β-actin was used as a loading control (TIFF 173286 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, S., Lee, Y., Pak, J.W. et al. O-GlcNAc modification is essential for the regulation of autophagy in Drosophila melanogaster . Cell. Mol. Life Sci. 72, 3173–3183 (2015). https://doi.org/10.1007/s00018-015-1889-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-015-1889-z

Keywords

Navigation