Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

O-GlcNAc transferase regulates glioblastoma acetate metabolism via regulation of CDK5-dependent ACSS2 phosphorylation

Abstract

Glioblastomas (GBMs) preferentially generate acetyl-CoA from acetate as a fuel source to promote tumor growth. O-GlcNAcylation has been shown to be elevated by increasing O-GlcNAc transferase (OGT) in many cancers and reduced O-GlcNAcylation can block cancer growth. Here, we identify a novel mechanism whereby OGT regulates acetate-dependent acetyl-CoA and lipid production by regulating phosphorylation of acetyl-CoA synthetase 2 (ACSS2) by cyclin-dependent kinase 5 (CDK5). OGT is required and sufficient for GBM cell growth and regulates acetate conversion to acetyl-CoA and lipids. Elevating O-GlcNAcylation in GBM cells increases phosphorylation of ACSS2 on Ser-267 in a CDK5-dependent manner. Importantly, we show that ACSS2 Ser-267 phosphorylation regulates its stability by reducing polyubiquitination and degradation. ACSS2 Ser-267 is critical for OGT-mediated GBM growth as overexpression of ACSS2 Ser-267 phospho-mimetic rescues growth in vitro and in vivo. Importantly, we show that pharmacologically targeting OGT and CDK5 reduces GBM growth ex vivo. Thus, the OGT/CDK5/ACSS2 pathway may be a way to target altered metabolic dependencies in brain tumors.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: OGT and O-GlcNAcylation levels are elevated in human glioblastoma.
Fig. 2: OGT is required for glioblastoma growth in vitro and in vivo.
Fig. 3: OGT promotes acetate and lipid accumulation in glioblastoma cells.
Fig. 4: Ser267-ACSS2 phosphorylation is mediated by O-GlcNAcylation in a CDK5-dependent manner.
Fig. 5: Phosphorylation of Ser267 enhances stability of ACSS2 and is required for GBM growth.
Fig. 6: CDK5 is a critical regulator of GBM growth and requires ACSS2 phosphorylation.
Fig. 7: Phosphorylation of S267-ACSS2 is required for OGT-mediated GBM growth.
Fig. 8: Targeting OGT or CDKs blocks GBM growth ex vivo.

Similar content being viewed by others

References

  1. Dunn GP, Rinne ML, Wykosky J, Genovese G, Quayle SN, Dunn IF, et al. Emerging insights into the molecular and cellular basis of glioblastoma. Genes Dev. 2012;26:756–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Schug ZT, Vande Voorde J, Gottlieb E. The metabolic fate of acetate in cancer. Nat Rev Cancer. 2016;16:708–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Comerford SA, Huang Z, Du X, Wang Y, Cai L, Witkiewicz AK, et al. Acetate dependence of tumors. Cell. 2014;159:1591–602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Mashimo T, Pichumani K, Vemireddy V, Hatanpaa KJ, Singh DK, Sirasanagandla S, et al. Acetate is a bioenergetic substrate for human glioblastoma and brain metastases. Cell. 2014;159:1603–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lewis CA, Brault C, Peck B, Bensaad K, Griffiths B, Mitter R, et al. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene. 2015;34:5128–40.

    Article  CAS  PubMed  Google Scholar 

  6. Yoshii Y, Furukawa T, Yoshii H, Mori T, Kiyono Y, Waki A, et al. Cytosolic acetyl-CoA synthetase affected tumor cell survival under hypoxia: the possible function in tumor acetyl-CoA/acetate metabolism. Cancer Sci. 2009;100:821–7.

    Article  CAS  PubMed  Google Scholar 

  7. Zachara NE, Hart GW. O-GlcNAc modification: a nutritional sensor that modulates proteasome function. Trends Cell Biol. 2004;14:218–21.

    Article  CAS  PubMed  Google Scholar 

  8. Gao Y, Wells L, Comer FI, Parker GJ, Hart GW. Dynamic O-glycosylation of nuclear and cytosolic proteins: cloning and characterization of a neutral, cytosolic beta-N-acetylglucosaminidase from human brain. J Biol Chem. 2001;276:9838–45.

    Article  CAS  PubMed  Google Scholar 

  9. Bond MR, Hanover JA. A little sugar goes a long way: the cell biology of O-GlcNAc. J Cell Biol. 2015;208:869–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ferrer CM, Sodi VL, Reginato MJ. O-GlcNAcylation in Cancer Biology: Linking Metabolism and Signaling. J Mol Biol. 2016;428:3282–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Caldwell SA, Jackson SR, Shahriari KS, Lynch TP, Sethi G, Walker S, et al. Nutrient sensor O-GlcNAc transferase regulates breast cancer tumorigenesis through targeting of the oncogenic transcription factor FoxM1. Oncogene. 2010;29:2831–42.

    Article  CAS  PubMed  Google Scholar 

  12. Lynch TP, Ferrer CM, Jackson SR, Shahriari KS, Vosseller K, Reginato MJ. Critical role of O-Linked β-N-acetylglucosamine transferase in prostate cancer invasion, angiogenesis, and metastasis. J Biol Chem. 2012;287:11070–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sodi VL, Bacigalupa ZA, Ferrer CM, Lee JV, Gocal WA, Mukhopadhyay D, et al. Nutrient sensor O-GlcNAc transferase controls cancer lipid metabolism via SREBP-1 regulation. Oncogene. 2018;37:924–34.

    Article  CAS  PubMed  Google Scholar 

  14. Pozo K, Bibb JA. The emerging role of Cdk5 in cancer. Trends Cancer. 2016;2:606–18.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Antoniou X, Gassmann M, Ogunshola OO. Cdk5 interacts with Hif-1alpha in neurons: a new hypoxic signalling mechanism? Brain Res. 2011;1381:1–10.

    Article  CAS  PubMed  Google Scholar 

  16. Fu AK, Fu WY, Ng AK, Chien WW, Ng YP, Wang JH, et al. Cyclin-dependent kinase 5 phosphorylates signal transducer and activator of transcription 3 and regulates its transcriptional activity. Proc Natl Acad Sci USA. 2004;101:6728–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Futatsugi A, Utreras E, Rudrabhatla P, Jaffe H, Pant HC, Kulkarni AB. Cyclin-dependent kinase 5 regulates E2F transcription factor through phosphorylation of Rb protein in neurons. Cell Cycle. 2012;11:1603–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lindqvist J, Imanishi SY, Torvaldson E, Malinen M, Remes M, Orn F, et al. Cyclin-dependent kinase 5 acts as a critical determinant of AKT-dependent proliferation and regulates differential gene expression by the androgen receptor in prostate cancer cells. Mol Biol Cell. 2015;26:1971–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sharma S, Zhang T, Michowski W, Rebecca VW, Xiao M, Ferretti R, et al. Targeting the cyclin-dependent kinase 5 in metastatic melanoma. Proc Natl Acad Sci USA. 2020;117:8001–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lenjisa JL, Tadesse S, Khair NZ, Kumarasiri M, Yu M, Albrecht H, et al. CDK5 in oncology: recent advances and future prospects. Future Med Chem. 2017;9:1939–62.

    Article  CAS  PubMed  Google Scholar 

  21. Yushan R, Wenjie C, Suning H, Yiwu D, Tengfei Z, Madushi WM, et al. Insights into the clinical value of cyclin-dependent kinase 5 in glioma: a retrospective study. World J Surgical Oncol. 2015;13:223.

    Article  Google Scholar 

  22. Catania A, Urban S, Yan E, Hao C, Barron G, Allalunis-Turner J. Expression and localization of cyclin-dependent kinase 5 in apoptotic human glioma cells. Neuro Oncol. 2001;3:89–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu R, Tian B, Gearing M, Hunter S, Ye K, Mao Z. Cdk5-mediated regulation of the PIKE-A-Akt pathway and glioblastoma cell invasion. Proc Natl Acad Sci USA. 2008;105:7570–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gloster TM, Zandberg WF, Heinonen JE, Shen DL, Deng L, Vocadlo DJ. Hijacking a biosynthetic pathway yields a glycosyltransferase inhibitor within cells. Nat Chem Biol. 2011;7:174–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ferrer CM, Lynch TP, Sodi VL, Falcone JN, Schwab LP, Peacock DL, et al. O-GlcNAcylation regulates cancer metabolism and survival stress signaling via regulation of the HIF-1 pathway. Mol Cell. 2014;54:820–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sodi VL, Khaku S, Krutilina R, Schwab LP, Vocadlo DJ, Seagroves TN, et al. mTOR/MYC Axis Regulates O-GlcNAc Transferase Expression and O-GlcNAcylation in Breast Cancer. Mol Cancer Res. 2015;13:923–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schug ZT, Peck B, Jones DT, Zhang Q, Grosskurth S, Alam IS, et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell. 2015;27:57–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kreppel LK, Blomberg MA, Hart GW. Dynamic glycosylation of nuclear and cytosolic proteins. Cloning and characterization of a unique O-GlcNAc transferase with multiple tetratricopeptide repeats. J Biol Chem. 1997;272:9308–15.

    Article  CAS  PubMed  Google Scholar 

  29. Obenauer JC, Cantley LC, Yaffe MB. Scansite 2.0: Proteome-wide prediction of cell signaling interactions using short sequence motifs. Nucleic Acids Res. 2003;31:3635–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hornbeck PV, Zhang B, Murray B, Kornhauser JM, Latham V, Skrzypek E. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res 2015;43:D512–20.

    Article  CAS  PubMed  Google Scholar 

  31. Lin H, Chen MC, Chiu CY, Song YM, Lin SY. Cdk5 regulates STAT3 activation and cell proliferation in medullary thyroid carcinoma cells. J Biol Chem. 2007;282:2776–84.

    Article  CAS  PubMed  Google Scholar 

  32. Piedrahita D, Hernandez I, Lopez-Tobon A, Fedorov D, Obara B, Manjunath BS, et al. Silencing of CDK5 reduces neurofibrillary tangles in transgenic alzheimer’s mice. J Neurosci. 2010;30:13966–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Parry D, Guzi T, Shanahan F, Davis N, Prabhavalkar D, Wiswell D, et al. Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor. Mol Cancer Ther. 2010;9:2344–53.

    Article  CAS  PubMed  Google Scholar 

  34. Jackson JG, O’Donnell JC, Takano H, Coulter DA, Robinson MB. Neuronal activity and glutamate uptake decrease mitochondrial mobility in astrocytes and position mitochondria near glutamate transporters. J Neurosci. 2014;34:1613–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ning X, Tao T, Shen J, Ji Y, Xie L, Wang H, et al. The O-GlcNAc Modification of CDK5 Involved in Neuronal Apoptosis Following In Vitro Intracerebral Hemorrhage. Cell Mol Neurobiol. 2017;37:527–36.

    Article  CAS  PubMed  Google Scholar 

  36. Chau V, Tobias JW, Bachmair A, Marriott D, Ecker DJ, Gonda DK, et al. A multiubiquitin chain is confined to specific lysine in a targeted short-lived protein. Science. 1989;243:1576–83.

    Article  CAS  PubMed  Google Scholar 

  37. Li X, Yu W, Qian X, Xia Y, Zheng Y, Lee JH, et al. Nucleus-Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy. Mol Cell. 2017;66:684–97 e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Parry D, Guzi T, Shanahan F, Davis N, Prabhavalkar D, Wiswell D, et al. Dinaciclib (SCH 727965), a novel and potent cyclin-dependent kinase inhibitor. Mol Cancer Ther. 2010;9:2344–53.

    Article  CAS  PubMed  Google Scholar 

  39. Magistretti PJ, Allaman I. A cellular perspective on brain energy metabolism and functional imaging. Neuron. 2015;86:883–901.

    Article  CAS  PubMed  Google Scholar 

  40. Schild T, Low V, Blenis J, Gomes AP. Unique metabolic adaptations dictate distal organ-specific metastatic colonization. Cancer Cell. 2018;33:347–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ubeda M, Kemp DM, Habener JF. Glucose-induced expression of the cyclin-dependent protein kinase 5 activator p35 involved in Alzheimer’s disease regulates insulin gene transcription in pancreatic beta-cells. Endocrinology. 2004;145:3023–31.

    Article  CAS  PubMed  Google Scholar 

  42. Kumar SK, LaPlant B, Chng WJ, Zonder J, Callander N, Fonseca R, et al. Dinaciclib, a novel CDK inhibitor, demonstrates encouraging single-agent activity in patients with relapsed multiple myeloma. Blood. 2015;125:443–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Feldmann G, Mishra A, Bisht S, Karikari C, Garrido-Laguna I, Rasheed Z, et al. Cyclin-dependent kinase inhibitor Dinaciclib (SCH727965) inhibits pancreatic cancer growth and progression in murine xenograft models. Cancer Biol Ther. 2011;12:598–609.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen XX, Xie FF, Zhu XJ, Lin F, Pan SS, Gong LH, et al. Cyclin-dependent kinase inhibitor dinaciclib potently synergizes with cisplatin in preclinical models of ovarian cancer. Oncotarget. 2015;6:14926–39.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Bulusu V, Tumanov S, Michalopoulou E, van den Broek NJ, MacKay G, Nixon C, et al. Acetate recapturing by nuclear acetyl-CoA synthetase 2 prevents loss of histone acetylation during oxygen and serum limitation. Cell Rep. 2017;18:647–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ferrer CM, Lynch TP, Sodi VL, Falcone JN, Schwab LP, Peacock DL, et al. O-GlcNAcylation regulates cancer metabolism and survival stress signaling via regulation of the HIF-1 pathway. Mol Cell. 2014;54:820–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jin X, Demere Z, Nair K, Ali A, Ferraro GB, Natoli T, et al. A metastasis map of human cancer cell lines. Nature 2020;588:331–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ferraro GB, Ali A, Luengo A, Kodack DP, Deik A, Abbott KL, et al. Fatty Acid Synthesis Is Required for Breast Cancer Brain Metastasis. Nat Cancer. 2021;2:414–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hothi P, Martins TJ, Chen L, Deleyrolle L, Yoon JG, Reynolds B, et al. High-throughput chemical screens identify disulfiram as an inhibitor of human glioblastoma stem cells. Oncotarget. 2012;3:1124–36.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi B, et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 2017;19:649–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26:1367–72.

    Article  CAS  PubMed  Google Scholar 

  52. Zhao S, Torres A, Henry RA, Trefely S, Wallace M, Lee JV, et al. ATP-Citrate Lyase Controls a Glucose-to-Acetate Metabolic Switch. Cell Rep. 2016;17:1037–52.

  53. Trefely S, Ashwell P, Snyder NW. FluxFix: automatic isotopologue normalization for metabolic tracer analysis. BMC Bioinforma. 2016;17:485.

  54. Ciraku L, Moeller RA, Esquea EM, Gocal WA, Hartsough EJ, Simone NL, et al. An Ex Vivo Brain Slice Model to Study and Target Breast Cancer Brain Metastatic Tumor Growth. J Vis Exp. 2021;175.

  55. Thompson JW, Griffin ME, Hsieh-Wilson LC. Methods for the Detection, Study, and Dynamic Profiling of O-GlcNAc Glycosylation. Methods Enzymol. 2018;598:101–35.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by PA CURE grant (to MJR, NWS and JGJ), UO1CA244303 (to MJR), Drexel University Dean’s Fellowship award (to ZAB and LC). KEW acknowledges R01CA228339. LTI is supported by 2-T32-CA-115299-14. The authors thank Chaitali Bhadiadra for technical assistance, Dr. Valerie Sodi and Dr. Edward Hartsough for helpful discussions. In memoriam Christos D. Katsetos.

Author information

Authors and Affiliations

Authors

Contributions

LC and ZAB performed most of the experimental work; GL, JJ, RAM, and RHL helped with experimental work; CMF helped establish intracranial mouse model; SF, MTD, LTI, KEW and NWS performed acetate labeling and analyzed data. WAG, LD and CDK helped with IHC and CDK performed pathological analysis; WS provided GBM tissue array; JGJ, LC, RAM helped establish ex vivo brain slice model. LC, ZAB and MJR participated in study conception and design as well as data analysis and interpretation; LC, ZAB and MJR drafted the manuscript; All co-authors reviewed the final manuscript version.

Corresponding author

Correspondence to Mauricio J. Reginato.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ciraku, L., Bacigalupa, Z.A., Ju, J. et al. O-GlcNAc transferase regulates glioblastoma acetate metabolism via regulation of CDK5-dependent ACSS2 phosphorylation. Oncogene 41, 2122–2136 (2022). https://doi.org/10.1038/s41388-022-02237-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02237-6

This article is cited by

Search

Quick links