Abstract
Cells produce metabolic intermediates through catalytic reactions, mainly via post-translational modifications. The modification of proteins by O-linked N-acetylglucosamine, known as O-GlcNAcylation, is one of the most common post-translational modifications. As O-GlcNAcylation and phosphorylation can occur at serine or threonine residues, it is crucial that the interplay between these two modifications is vital to bioenergetic and biosynthetic demand. Although emerging recognition linking O-GlcNAc modification and phosphorylation to protein functions has been obtained, the issue of how altered O-GlcNAcylation or phosphorylation regulates each other in the metabolic system remains uncertain. The combination of cell biological and proteomic approaches over the recent few years has not only highlighted the interactions between O-GlcNAcylation and phosphorylation in protein function but also prompted us to elucidate the underlying mechanisms behind this crosstalk controlling metabolic homeostasis. The purpose of this review is to summarize recent advances in the O-GlcNAcylation/phosphorylation regulation of the metabolic process. An extensive exploration of this interplay has significant implications for metabolic control systems, including glucose, lipid, and nucleotide metabolism, where dysregulation in O-GlcNAcylation and phosphorylation of metabolic syndrome is essential.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout





Similar content being viewed by others
References
Judge A, Dodd MS. Metabolism. Essays Biochem. 2020;64:607–47.
Ye J, Medzhitov R. Control strategies in systemic metabolism. Nat Metab. 2019;1:947–57.
Chen L, Chen X-W, Huang X, Song B-L, Wang Y, Wang Y. Regulation of glucose and lipid metabolism in health and disease. Sci China Life Sci. 2019;62:1420–58.
Knaus LS, Basilico B, Malzl D, Gerykova Bujalkova M, Smogavec M, Schwarz LA, et al. Large neutral amino acid levels tune perinatal neuronal excitability and survival. Cell. 2023;186:1950–67.e25.
Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer. 2011;11:85–95.
Finley LWS. What is cancer metabolism? Cell. 2023;186:1670–88.
Wu X, Xu M, Geng M, Chen S, Little PJ, Xu S, et al. Targeting protein modifications in metabolic diseases: molecular mechanisms and targeted therapies. Sig Transduct Target Ther. 2023;8:220.
DeBerardinis RJ, Keshari KR. Metabolic analysis as a driver for discovery, diagnosis, and therapy. Cell. 2022;185:2678–89.
Torres CR, Hart GW. Topography and polypeptide distribution of terminal N-acetylglucosamine residues on the surfaces of intact lymphocytes. Evidence for O-linked GlcNAc. J Biol Chem. 1984;259:3308–17.
Holt GD, Snow CM, Senior A, Haltiwanger RS, Gerace L, Hart GW. Nuclear pore complex glycoproteins contain cytoplasmically disposed O-linked N-acetylglucosamine. J Cell Biol. 1987;104:1157–64.
Hart GW, Housley MP, Slawson C. Cycling of O-linked β-N-acetylglucosamine on nucleocytoplasmic proteins. Nature. 2007;446:1017–22.
Xu Z, Zhang Y, Ocansey DKW, Wang B, Mao F. Glycosylation in cervical cancer: new insights and clinical implications. Front Oncol. 2021;11:706862.
Zhang Y, Wang L, Ocansey DKW, Wang B, Wang L, Xu Z. Mucin-type O-glycans: barrier, microbiota, and immune anchors in inflammatory bowel disease. J Inflamm Res. 2021;14:5939–53.
Ma J, Wu C, Hart GW. Analytical and biochemical perspectives of protein O-GlcNAcylation. Chem Rev. 2021;121:1513–81.
Müller R, Jenny A, Stanley P. The EGF repeat-specific O-GlcNAc-transferase Eogt interacts with notch signaling and pyrimidine metabolism pathways in Drosophila. PLoS One. 2013;8:e62835.
Sawaguchi S, Varshney S, Ogawa M, Sakaidani Y, Yagi H, Takeshita K, et al. O-GlcNAc on NOTCH1 EGF repeats regulates ligand-induced Notch signaling and vascular development in mammals. Elife. 2017;6:e24419.
Wu C, Li J, Lu L, Li M, Yuan Y, Li J. OGT and OGA: sweet guardians of the genome. J Biol Chem. 2024;300:107141.
Akella NM, Ciraku L, Reginato MJ. Fueling the fire: emerging role of the hexosamine biosynthetic pathway in cancer. BMC Biol. 2019;17:52.
Ma X, Liu H, Li J, Wang Y, Ding Y-H, Shen H, et al. Polη O-GlcNAcylation governs genome integrity during translesion DNA synthesis. Nat Commun. 2017;8:1941.
Xu Z, Isaji T, Fukuda T, Wang Y, Gu J. O-GlcNAcylation regulates integrin-mediated cell adhesion and migration via formation of focal adhesion complexes. J Biol Chem. 2019;294:3117–24.
Zhao J, Hua J, Zhan Y, Chen C, Liu Y, Yang L, et al. O-GlcNAcylation stimulates the deubiquitination activity of USP16 and regulates cell cycle progression. J Biol Chem. 2024;300:107150.
Zhou Z, Zheng X, Zhao J, Yuan A, Lv Z, Shao G, et al. ULK1-dependent phosphorylation of PKM2 antagonizes O-GlcNAcylation and regulates the Warburg effect in breast cancer. Oncogene. 2024;43:1–10.
Yang X, Qian K. Protein O-GlcNAcylation: emerging mechanisms and functions. Nat Rev Mol Cell Biol. 2017;18:452–65.
Stanley P. Genetics of glycosylation in mammalian development and disease. Nat Rev Genet. 2024. https://doi.org/10.1038/s41576-024-00725-x.
Zeidan Q, Hart GW. The intersections between O-GlcNAcylation and phosphorylation: implications for multiple signaling pathways. J Cell Sci. 2010;123:13–22.
Bilbrough T, Piemontese E, Seitz O. Dissecting the role of protein phosphorylation: a chemical biology toolbox. Chem Soc Rev. 2022;51:5691–730.
Castelo-Soccio L, Kim H, Gadina M, Schwartzberg PL, Laurence A, O’Shea JJ. Protein kinases: drug targets for immunological disorders. Nat Rev Immunol. 2023;23:787–806.
Olsen JV, Blagoev B, Gnad F, Macek B, Kumar C, Mortensen P, et al. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell. 2006;127:635–48.
Nishi H, Shaytan A, Panchenko AR. Physicochemical mechanisms of protein regulation by phosphorylation. Front Genet. 2014;5:270.
Chen T, Xie S, Cheng J, Zhao Q, Wu H, Jiang P, et al. AKT1 phosphorylation of cytoplasmic ME2 induces a metabolic switch to glycolysis for tumorigenesis. Nat Commun. 2024;15:686.
Cai Z, Li C-F, Han F, Liu C, Zhang A, Hsu C-C, et al. Phosphorylation of PDHA by AMPK drives TCA cycle to promote cancer metastasis. Mol Cell. 2020;80:263–278.e7.
Humphrey SJ, James DE, Mann M. Protein phosphorylation: a major switch mechanism for metabolic regulation. Trends Endocrinol Metab. 2015;26:676–87.
Tan W, Jiang P, Zhang W, Hu Z, Lin S, Chen L, et al. Posttranscriptional regulation of de novo lipogenesis by glucose-induced O-GlcNAcylation. Mol Cell. 2021;81:1890–1904.e7.
Zhao M, Ren K, Xiong X, Xin Y, Zou Y, Maynard JC, et al. Epithelial STAT6 O-GlcNAcylation drives a concerted anti-helminth alarmin response dependent on tuft cell hyperplasia and Gasdermin C. Immunity. 2022;55:1327.
Wang Q, Zhang B, Stutz B, Liu Z-W, Horvath TL, Yang X. Ventromedial hypothalamic OGT drives adipose tissue lipolysis and curbs obesity. Sci Adv. 2022;8:eabn8092.
Chen L, Zhou Q, Zhang P, Tan W, Li Y, Xu Z, et al. Direct stimulation of de novo nucleotide synthesis by O-GlcNAcylation. Nat Chem Biol. 2023. https://doi.org/10.1038/s41589-023-01354-x.
Nelson ZM, Leonard GD, Fehl C. Tools for investigating O-GlcNAc in signaling and other fundamental biological pathways. J Biol Chem. 2024;300:105615.
Swamy M, Pathak S, Grzes KM, Damerow S, Sinclair LV, van Aalten DMF, et al. Glucose and glutamine fuel protein O-GlcNAcylation to control T cell self-renewal and malignancy. Nat Immunol. 2016;17:712–20.
Yang X, Ongusaha PP, Miles PD, Havstad JC, Zhang F, So WV, et al. Phosphoinositide signalling links O-GlcNAc transferase to insulin resistance. Nature. 2008;451:964–9.
Wong Y-K, Wang J, Lim TK, Lin Q, Yap CT, Shen H-M. O-GlcNAcylation promotes fatty acid synthase activity under nutritional stress as a pro-survival mechanism in cancer cells. Proteomics. 2022;22:e2100175.
Yang Y, Fu M, Li M-D, Zhang K, Zhang B, Wang S, et al. O-GlcNAc transferase inhibits visceral fat lipolysis and promotes diet-induced obesity. Nat Commun. 2020;11:181.
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.
Hart GW, Slawson C, Ramirez-Correa G, Lagerlof O. Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. Annu Rev Biochem. 2011;80:825–58.
Ma J, Hou C, Wu C. Demystifying the O-GlcNAc code: a systems view. Chem Rev. 2022;122:15822–64.
Sam van der L, Ac L, Ajr H. Crosstalk between phosphorylation and O-GlcNAcylation: friend or foe. FEBS J. 2018;285. https://doi.org/10.1111/febs.14491.
Chatham JC, Zhang J, Wende AR. Role of O-Linked N-acetylglucosamine protein modification in cellular (patho)physiology. Physiol Rev. 2021;101:427–93.
Wang Z, Li M, Jiang H, Luo S, Shao F, Xia Y, et al. Fructose-1,6-bisphosphatase 1 functions as a protein phosphatase to dephosphorylate histone H3 and suppresses PPARα-regulated gene transcription and tumour growth. Nat Cell Biol. 2022;24:1655–65.
Chen Y-F, Zhu J-J, Li J, Ye X-S. O-GlcNAcylation increases PYGL activity by promoting phosphorylation. Glycobiology. 2022;32:101–9.
Wang Z, Gucek M, Hart GW. Cross-talk between GlcNAcylation and phosphorylation: site-specific phosphorylation dynamics in response to globally elevated O-GlcNAc. Proc Natl Acad Sci USA. 2008;105:13793–8.
Yang Y, Li X, Luan HH, Zhang B, Zhang K, Nam JH, et al. OGT suppresses S6K1-mediated macrophage inflammation and metabolic disturbance. Proc Natl Acad Sci USA. 2020;117:16616–25.
Hb R, Jp S, Md L, J W, X Y. Cracking the O-GlcNAc code in metabolism. Trends Endocrinol Metab. TEM 2013;24. https://doi.org/10.1016/j.tem.2013.02.002.
Du D, Liu C, Qin M, Zhang X, Xi T, Yuan S, et al. Reprogramming of glucose, fatty acid and amino acid metabolism for cancer. Acta Pharm Sin B. 2022;12:558–80.
Gu L, Zhu Y, Watari K, Lee M, Liu J, Perez S, et al. Fructose-1,6-bisphosphatase is a nonenzymatic safety valve that curtails AKT activation to prevent insulin hyperresponsiveness. Cell Metab. 2023;35:1009–1021.e9.
Shi L, Pan H, Liu Z, Xie J, Han W. Roles of PFKFB3 in cancer. Signal Transduct Target Ther. 2017;2:17044.
Hamanaka RB, Mutlu GM. PFKFB3, a direct target of p63, is required for proliferation and inhibits differentiation in epidermal keratinocytes. J Investig Dermatol. 2017;137:1267–76.
Li F-L, Liu J-P, Bao R-X, Yan G, Feng X, Xu Y-P, et al. Acetylation accumulates PFKFB3 in cytoplasm to promote glycolysis and protects cells from cisplatin-induced apoptosis. Nat Commun. 2018;9:508.
Hawkins LJ, Wang X, Xue X, Wang H, Storey KB. Phosphoproteomic analysis of X enopus laevis reveals expression and phosphorylation of hypoxia-inducible PFKFB3 during dehydration. iScience. 2020;23:101598.
Yi W, Clark PM, Mason DE, Keenan MC, Hill C, Goddard WA, et al. Phosphofructokinase 1 glycosylation regulates cell growth and metabolism. Science. 2012;337:975–80.
Lei Y, Chen T, Li Y, Shang M, Zhang Y, Jin Y, et al. O-GlcNAcylation of PFKFB3 is required for tumor cell proliferation under hypoxia. Oncogenesis. 2020;9:21.
Olzmann JA, Carvalho P. Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol. 2019;20:137–55.
Chen J, Yang S, Li Y, Ziwen X, Zhang P, Song Q, et al. De novo nucleotide biosynthetic pathway and cancer. Genes Dis. 2023;10:2331–8.
Li J, Shao J, Zeng Z, He Y, Tang C, Park SH, et al. Mechanosensitive turnover of phosphoribosyl pyrophosphate synthetases regulates nucleotide metabolism. Cell Death Differ. 2022;29:206–17.
Qian X, Li X, Tan L, Lee J-H, Xia Y, Cai Q, et al. Conversion of PRPS Hexamer to Monomer by AMPK-Mediated Phosphorylation Inhibits Nucleotide Synthesis in Response to Energy Stress. Cancer Discov. 2018;8:94–107.
Tarrant MK, Rho H-S, Xie Z, Jiang YL, Gross C, Culhane JC, et al. Regulation of CK2 by phosphorylation and O-GlcNAcylation revealed by semisynthesis. Nat Chem Biol. 2012;8:262–9.
Deng H, Rao X, Zhang S, Chen L, Zong Y, Zhou R, et al. Protein kinase CK2: An emerging regulator of cellular metabolism. Biofactors. 2023. https://doi.org/10.1002/biof.2032.
Uhle S, Medalia O, Waldron R, Dumdey R, Henklein P, Bech-Otschir D, et al. Protein kinase CK2 and protein kinase D are associated with the COP9 signalosome. EMBO J. 2003;22:1302–12.
Su Y, Luo Y, Zhang P, Lin H, Pu W, Zhang H, et al. Glucose-induced CRL4COP1-p53 axis amplifies glycometabolism to drive tumorigenesis. Mol Cell. 2023;83:2316–2331.e7.
Li Z, Zhang H. Reprogramming of glucose, fatty acid and amino acid metabolism for cancer progression. Cell Mol Life Sci. 2016;73:377–92.
Yoon I, Nam M, Kim HK, Moon H-S, Kim S, Jang J, et al. Glucose-dependent control of leucine metabolism by leucyl-tRNA synthetase 1. Science. 2020;367:205–10.
Kim K, Yoo HC, Kim BG, Kim S, Sung Y, Yoon I, et al. O-GlcNAc modification of leucyl-tRNA synthetase 1 integrates leucine and glucose availability to regulate mTORC1 and the metabolic fate of leucine. Nat Commun. 2022;13:2904.
Mossmann D, Park S, Hall MN. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer. 2018;18:744–57.
Hardie DG. AMPK and Raptor: matching cell growth to energy supply. Mol Cell. 2008;30:263–5.
Xu C, Pan X, Wang D, Guan Y, Yang W, Chen X, et al. O-GlcNAcylation of Raptor transduces glucose signals to mTORC1. Mol Cell. 2023;83:3027–3040.e11.
Kim KH, Lee M-S. Autophagy-a key player in cellular and body metabolism. Nat Rev Endocrinol. 2014;10:322–37.
Kimmelman AC, White E. Autophagy and tumor metabolism. Cell Metab. 2017;25:1037–43.
Zachari M, Ganley IG. The mammalian ULK1 complex and autophagy initiation. Essays Biochem. 2017;61:585–96.
Kim J, Kundu M, Viollet B, Guan K-L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:132–41.
Ruan H-B, Ma Y, Torres S, Zhang B, Feriod C, Heck RM, et al. Calcium-dependent O-GlcNAc signaling drives liver autophagy in adaptation to starvation. Genes Dev. 2017;31:1655–65.
Shi Y, Yan S, Shao G-C, Wang J, Jian Y-P, Liu B, et al. O-GlcNAcylation stabilizes the autophagy-initiating kinase ULK1 by inhibiting chaperone-mediated autophagy upon HPV infection. J Biol Chem. 2022;298:102341.
Pyo KE, Kim CR, Lee M, Kim J-S, Kim KI, Baek SH. ULK1 O-GlcNAcylation is crucial for activating VPS34 via ATG14L during autophagy initiation. Cell Rep. 2018;25:2878–2890.e4.
Ding Z, Pan Y, Shang T, Jiang T, Lin Y, Yang C, et al. URI alleviates tyrosine kinase inhibitors-induced ferroptosis by reprogramming lipid metabolism in p53 wild-type liver cancers. Nat Commun. 2023;14:6269.
Gstaiger M, Luke B, Hess D, Oakeley EJ, Wirbelauer C, Blondel M, et al. Control of nutrient-sensitive transcription programs by the unconventional prefoldin URI. Science. 2003;302:1208–12.
Burén S, Gomes AL, Teijeiro A, Fawal M-A, Yilmaz M, Tummala KS, et al. Regulation of OGT by URI in response to glucose confers c-MYC-dependent survival mechanisms. Cancer Cell. 2016;30:290–307.
Fu Y, Ning L, Feng J, Yu X, Guan F, Li X. Dynamic regulation of O-GlcNAcylation and phosphorylation on STAT3 under hypoxia-induced EMT. Cell Signal. 2022;93:110277.
Freund P, Kerenyi MA, Hager M, Wagner T, Wingelhofer B, Pham HTT, et al. O-GlcNAcylation of STAT5 controls tyrosine phosphorylation and oncogenic transcription in STAT5-dependent malignancies. Leukemia. 2017;31:2132–42.
Han JM, Jeong SJ, Park MC, Kim G, Kwon NH, Kim HK, et al. Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell. 2012;149:410–24.
Liu Q, Li J, Zhang W, Xiao C, Zhang S, Nian C, et al. Glycogen accumulation and phase separation drives liver tumor initiation. Cell. 2021;184:5559–5576.e19.
Zois CE, Harris AL. Glycogen metabolism has a key role in the cancer microenvironment and provides new targets for cancer therapy. J Mol Med (Berl). 2016;94:137–54.
Zannini L, Delia D, Buscemi G. CHK2 kinase in the DNA damage response and beyond. J Mol Cell Biol. 2014;6:442–57.
Guo R, Wang S-S, Jiang X-Y, Zhang Y, Guo Y, Cui H-Y, et al. CHK2 promotes metabolic stress-induced autophagy through ULK1 phosphorylation. Antioxidants. 2022;11:1166.
Chong ACN, Vandana JJ, Jeng G, Li G, Meng Z, Duan X, et al. Checkpoint kinase 2 controls insulin secretion and glucose homeostasis. Nat Chem Biol. 2023. https://doi.org/10.1038/s41589-023-01466-4.
Lulli M, Del Coco L, Mello T, Sukowati C, Madiai S, Gragnani L, et al. DNA damage response protein CHK2 regulates metabolism in liver cancer. Cancer Res. 2021;81:2861–73.
Li C, Deng C, Wang S, Dong X, Dai B, Guo W, et al. A novel role for the ROS-ATM-Chk2 axis mediated metabolic and cell cycle reprogramming in the M1 macrophage polarization. Redox Biol. 2024;70:103059.
Xiang J, Chen C, Liu R, Gou D, Chang L, Deng H, et al. Gluconeogenic enzyme PCK1 deficiency promotes CHK2 O-GlcNAcylation and hepatocellular carcinoma growth upon glucose deprivation. J Clin Invest. 2021;131:e144703.
Thompson LL, Guppy BJ, Sawchuk L, Davie JR, McManus KJ. Regulation of chromatin structure via histone post-translational modification and the link to carcinogenesis. Cancer Metastasis Rev. 2013;32:363–76.
Sakabe K, Wang Z, Hart GW. Beta-N-acetylglucosamine (O-GlcNAc) is part of the histone code. Proc Natl Acad Sci USA. 2010;107:19915–20.
Zou Y, Pei J, Long H, Lan L, Dong K, Wang T, et al. H4S47 O-GlcNAcylation regulates the activation of mammalian replication origins. Nat Struct Mol Biol. 2023;30:800–11.
Lee JB, Pyo K-H, Kim HR. Role and Function of O-GlcNAcylation in Cancer. Cancers. 2021;13:5365.
Singh JP, Zhang K, Wu J, Yang X. O-GlcNAc signaling in cancer metabolism and epigenetics. Cancer Lett. 2015;356:244–50.
Xu S, Suttapitugsakul S, Tong M, Wu R. Systematic analysis of the impact of phosphorylation and O-GlcNAcylation on protein subcellular localization. Cell Rep. 2023;42:112796.
Hu P, Shimoji S, Hart GW. Site-specific interplay between O-GlcNAcylation and phosphorylation in cellular regulation. FEBS Lett. 2010;584:2526–38.
Iakoucheva LM, Radivojac P, Brown CJ, O’Connor TR, Sikes JG, Obradovic Z, et al. The importance of intrinsic disorder for protein phosphorylation. Nucleic Acids Res. 2004;32:1037–49.
Yan W, Cao M, Ruan X, Jiang L, Lee S, Lemanek A, et al. Cancer-cell-secreted miR-122 suppresses O-GlcNAcylation to promote skeletal muscle proteolysis. Nat Cell Biol. 2022;24:793–804.
Kaasik K, Kivimäe S, Allen JJ, Chalkley RJ, Huang Y, Baer K, et al. Glucose sensor O-GlcNAcylation coordinates with phosphorylation to regulate circadian clock. Cell Metab. 2013;17:291–302.
Sager RA, Woodford MR, Backe SJ, Makedon AM, Baker-Williams AJ, DiGregorio BT, et al. Post-translational regulation of FNIP1 creates a rheostat for the molecular chaperone Hsp90. Cell Rep. 2019;26:1344–1356.e5.
Yang WH, Kim JE, Nam HW, Ju JW, Kim HS, Kim YS, et al. Modification of p53 with O-linked N-acetylglucosamine regulates p53 activity and stability. Nat Cell Biol. 2006;8:1074–83.
Xu C, Liu GD, Feng L, Zhang CH, Wang F. Identification of O-GlcNAcylation modification in diabetic retinopathy and crosstalk with phosphorylation of STAT3 in retina vascular endothelium cells. Cell Physiol Biochem. 2018;49:1389–402.
Shi Y. Serine/threonine phosphatases: mechanism through structure. Cell. 2009;139:468–84.
Matsuno M, Yokoe S, Nagatsuka T, Morihara H, Moriwaki K, Asahi M. O-GlcNAcylation-induced GSK-3β activation deteriorates pressure overload-induced heart failure via lack of compensatory cardiac hypertrophy in mice. Front Endocrinol. 2023;14:1122125.
Zhao Y, Tang Z, Shen A, Tao T, Wan C, Zhu X, et al. The role of PTP1B O-GlcNAcylation in hepatic insulin resistance. Int J Mol Sci. 2015;16:22856–69.
Zhou J, Huynh QK, Hoffman RT, Crook ED, Daniels MC, Gulve EA, et al. Regulation of glutamine:fructose-6-phosphate amidotransferase by cAMP-dependent protein kinase. Diabetes. 1998;47:1836–40.
Chang Q, Su K, Baker JR, Yang X, Paterson AJ, Kudlow JE. Phosphorylation of human glutamine:fructose-6-phosphate amidotransferase by cAMP-dependent protein kinase at serine 205 blocks the enzyme activity. J Biol Chem. 2000;275:21981–7.
Li Y, Roux C, Lazereg S, LeCaer J-P, Laprévote O, Badet B, et al. Identification of a novel serine phosphorylation site in human glutamine:fructose-6-phosphate amidotransferase isoform 1. Biochemistry. 2007;46:13163–9.
Zibrova D, Vandermoere F, Göransson O, Peggie M, Mariño KV, Knierim A, et al. GFAT1 phosphorylation by AMPK promotes VEGF-induced angiogenesis. Biochem J. 2017;474:983–1001.
Gélinas R, Mailleux F, Dontaine J, Bultot L, Demeulder B, Ginion A, et al. AMPK activation counteracts cardiac hypertrophy by reducing O-GlcNAcylation. Nat Commun. 2018;9:374.
Eguchi S, Oshiro N, Miyamoto T, Yoshino K-I, Okamoto S, Ono T, et al. AMP-activated protein kinase phosphorylates glutamine: fructose-6-phosphate amidotransferase 1 at Ser243 to modulate its enzymatic activity. Genes Cells. 2009;14:179–89.
Qin W, Lv P, Fan X, Quan B, Zhu Y, Qin K, et al. Quantitative time-resolved chemoproteomics reveals that stable O-GlcNAc regulates box C/D snoRNP biogenesis. Proc Natl Acad Sci USA. 2017;114:E6749–58.
Ruegenberg S, Horn M, Pichlo C, Allmeroth K, Baumann U, Denzel MS. Loss of GFAT-1 feedback regulation activates the hexosamine pathway that modulates protein homeostasis. Nat Commun. 2020;11:687.
Li Z, Li X, Nai S, Geng Q, Liao J, Xu X, et al. Checkpoint kinase 1-induced phosphorylation of O-linked β-N-acetylglucosamine transferase regulates the intermediate filament network during cytokinesis. J Biol Chem. 2017;292:19548–55.
Bullen JW, Balsbaugh JL, Chanda D, Shabanowitz J, Hunt DF, Neumann D, et al. Cross-talk between two essential nutrient-sensitive enzymes: O-GlcNAc transferase (OGT) and AMP-activated protein kinase (AMPK). J Biol Chem. 2014;289:10592–606.
Wang Y, Shu H, Liu J, Jin X, Wang L, Qu Y, et al. EGF promotes PKM2 O-GlcNAcylation by stimulating O-GlcNAc transferase phosphorylation at Y976 and their subsequent association. J Biol Chem. 2022;298:102340.
Latorre-Muro P, O’Malley KE, Bennett CF, Perry EA, Balsa E, Tavares CDJ, et al. A cold-stress-inducible PERK/OGT axis controls TOM70-assisted mitochondrial protein import and cristae formation. Cell Metab. 2021;33:598–614.e7.
Xu Q, Yang C, Du Y, Chen Y, Liu H, Deng M, et al. AMPK regulates histone H2B O-GlcNAcylation. Nucleic Acids Res. 2014;42:5594–604.
Popovic D, Vucic D, Dikic I. Ubiquitination in disease pathogenesis and treatment. Nat Med. 2014;20:1242–53.
Sheng X, Xia Z, Yang H, Hu R. The ubiquitin codes in cellular stress responses. Protein Cell. 2024;15:157–90.
Dikic I, Schulman BA. An expanded lexicon for the ubiquitin code. Nat Rev Mol Cell Biol. 2023;24:273–87.
Koo S-Y, Park E-J, Noh H-J, Jo S-M, Ko B-K, Shin H-J, et al. Ubiquitination links DNA damage and repair signaling to cancer metabolism. Int J Mol Sci. 2023;24:8441.
Ruan H-B, Nie Y, Yang X. Regulation of protein degradation by O-GlcNAcylation: crosstalk with ubiquitination. Mol Cell Proteom. 2013;12:3489–97.
Baba M, Hong S-B, Sharma N, Warren MB, Nickerson ML, Iwamatsu A, et al. Folliculin encoded by the BHD gene interacts with a binding protein, FNIP1, and AMPK, and is involved in AMPK and mTOR signaling. Proc Natl Acad Sci USA. 2006;103:15552–7.
Zalk R, Lehnart SE, Marks AR. Modulation of the ryanodine receptor and intracellular calcium. Annu Rev Biochem. 2007;76:367–85.
Menet JS, Pescatore S, Rosbash M. CLOCK:BMAL1 is a pioneer-like transcription factor. Genes Dev. 2014;28:8–13.
Bass J, Takahashi JS. Circadian integration of metabolism and energetics. Science. 2010;330:1349–54.
Li M-D, Ruan H-B, Hughes ME, Lee J-S, Singh JP, Jones SP, et al. O-GlcNAc signaling entrains the circadian clock by inhibiting BMAL1/CLOCK ubiquitination. Cell Metab. 2013;17:303–10.
Chatham JC, Patel RP. Protein glycosylation in cardiovascular health and disease. Nat Rev Cardiol. 2024. https://doi.org/10.1038/s41569-024-00998-z.
Song W, Isaji T, Nakano M, Liang C, Fukuda T, Gu J. O-GlcNAcylation regulates β1,4-GlcNAc-branched N-glycan biosynthesis via the OGT/SLC35A3/GnT-IV axis. FASEB J. 2022;36:e22149.
Alam SMD, Tsukamoto Y, Ogawa M, Senoo Y, Ikeda K, Tashima Y, et al. N-Glycans on EGF domain-specific O-GlcNAc transferase (EOGT) facilitate EOGT maturation and peripheral endoplasmic reticulum localization. J Biol Chem. 2020;295:8560–74.
Lv Z, Ma G, Zhong Z, Xie X, Li B, Long D. O-GlcNAcylation of RAB10 promotes hepatocellular carcinoma progression. Carcinogenesis. 2023;44:785–94.
Zhang Y, Sun C, Ma L, Xiao G, Gu Y, Yu WO. GlcNAcylation promotes malignancy and cisplatin resistance of lung cancer by stabilising NRF2. Clin Transl Med. 2024;14:e70037.
Wang X, Liu M, Chu Y, Liu Y, Cao X, Zhang H, et al. O-GlcNAcylation of ZEB1 facilitated mesenchymal pancreatic cancer cell ferroptosis. Int J Biol Sci. 2022;18:4135–50.
Jiang M, Qiu Z, Zhang S, Fan X, Cai X, Xu B, et al. Elevated O-GlcNAcylation promotes gastric cancer cells proliferation by modulating cell cycle related proteins and ERK 1/2 signaling. Oncotarget. 2016;7:61390–402.
Kronlage M, Dewenter M, Grosso J, Fleming T, Oehl U, Lehmann LH, et al. O-GlcNAcylation of Histone Deacetylase 4 Protects the Diabetic Heart From Failure. Circulation. 2019;140:580–94.
Lei Y, Liu Q, Chen B, Wu F, Li Y, Dong X, et al. Protein O-GlcNAcylation coupled to Hippo signaling drives vascular dysfunction in diabetic retinopathy. Nat Commun. 2024;15:9334.
Shafi R, Iyer SP, Ellies LG, O’Donnell N, Marek KW, Chui D, et al. The O-GlcNAc transferase gene resides on the X chromosome and is essential for embryonic stem cell viability and mouse ontogeny. Proc Natl Acad Sci USA. 2000;97:5735–9.
Deng Y, Li B, Liu Y, Iqbal K, Grundke-Iqbal I, Gong C-X. Dysregulation of insulin signaling, glucose transporters, O-GlcNAcylation, and phosphorylation of tau and neurofilaments in the brain: implication for Alzheimer’s disease. Am J Pathol. 2009;175:2089–98.
Pratt MR, Vocadlo DJ. Understanding and exploiting the roles of O-GlcNAc in neurodegenerative diseases. J Biol Chem. 2023;299. https://doi.org/10.1016/j.jbc.2023.105411.
Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong C-X. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer’s disease. Proc Natl Acad Sci USA. 2004;101:10804–9.
Yuzwa SA, Macauley MS, Heinonen JE, Shan X, Dennis RJ, He Y, et al. A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat Chem Biol. 2008;4:483–90.
Chen J, Zhao B, Dong H, Li T, Cheng X, Gong W, et al. Inhibition of O-GlcNAc transferase activates type I interferon-dependent antitumor immunity by bridging cGAS-STING pathway. eLife. 2024;13:RP94849.
Zhu Q, Wang H, Chai S, Xu L, Lin B, Yi W, et al. O-GlcNAcylation promotes tumor immune evasion by inhibiting PD-L1 lysosomal degradation. Proc Natl Acad Sci USA. 2023;120:e2216796120.
Shcherbinin S, Kielbasa W, Dubois S, Lowe SL, Phipps KM, Tseng J, et al. Brain target occupancy of LY3372689, an inhibitor of the O‐GlcNAcase (OGA) enzyme: Translation from rat to human: Neuroimaging / evaluating treatments. Alzheimer’s Dement. 2020;16:e040558.
Selnick HG, Hess JF, Tang C, Liu K, Schachter JB, Ballard JE, et al. Discovery of MK-8719, a potent O-GlcNAcase inhibitor as a potential treatment for tauopathies. J Med Chem. 2019;62:10062–97.
Permanne B, Sand A, Ousson S, Nény M, Hantson J, Schubert R, et al. O-GlcNAcase inhibitor ASN90 is a multimodal drug candidate for tau and α-synuclein proteinopathies. ACS Chem Neurosci. 2022;13:1296–314.
He P, Bian A, Miao Y, Jin W, Chen H, He J, et al. Discovery of a highly potent and orally bioavailable STAT3 dual phosphorylation inhibitor for pancreatic cancer treatment. J Med Chem. 2022;65:15487–511.
Ma J, Li Y, Hou C, Wu C. O-GlcNAcAtlas: a database of experimentally identified O-GlcNAc sites and proteins. Glycobiology. 2021;31:719–23.
Hu F, Li W, Li Y, Hou C, Ma J, Jia C. O-GlcNAcPRED-DL: prediction of protein O-GlcNAcylation sites based on an ensemble model of deep learning. J Proteome Res. 2023. https://doi.org/10.1021/acs.jproteome.3c00458.
Ma J, Hou C, Li Y, Chen S, Wu C. OGT protein interaction network (OGT-PIN): a curated database of experimentally identified interaction proteins of OGT. Int J Mol Sci. 2021;22:9620.
Wulff-Fuentes E, Berendt RR, Massman L, Danner L, Malard F, Vora J, et al. The human O-GlcNAcome database and meta-analysis. Sci Data. 2021;8:25.
Comer FI, Vosseller K, Wells L, Accavitti MA, Hart GW. Characterization of a mouse monoclonal antibody specific for O-linked N-acetylglucosamine. Anal Biochem. 2001;293:169–77.
Pathak S, Borodkin VS, Albarbarawi O, Campbell DG, Ibrahim A, van Aalten DM. O-GlcNAcylation of TAB1 modulates TAK1-mediated cytokine release. EMBO J. 2012;31:1394–404.
Kamemura K, Hayes BK, Comer FI, Hart GW. Dynamic interplay between O-glycosylation and O-phosphorylation of nucleocytoplasmic proteins: alternative glycosylation/phosphorylation of THR-58, a known mutational hot spot of c-Myc in lymphomas, is regulated by mitogens. J Biol Chem. 2002;277:19229–35.
Yuzwa SA, Yadav AK, Skorobogatko Y, Clark T, Vosseller K, Vocadlo DJ. Mapping O-GlcNAc modification sites on tau and generation of a site-specific O-GlcNAc tau antibody. Amino Acids. 2011;40:857–68.
Hirosawa M, Hayakawa K, Yoneda C, Arai D, Shiota H, Suzuki T, et al. Novel O-GlcNAcylation on Ser(40) of canonical H2A isoforms specific to viviparity. Sci Rep. 2016;6:31785.
Fehl C, Hanover JA. Tools, tactics and objectives to interrogate cellular roles of O-GlcNAc in disease. Nat Chem Biol. 2022;18:8–17.
Saha A, Bello D, Fernández-Tejada A. Advances in chemical probing of protein O-GlcNAc glycosylation: structural role and molecular mechanisms. Chem Soc Rev. 2021;50:10451–85.
Yu S-H, Boyce M, Wands AM, Bond MR, Bertozzi CR, Kohler JJ. Metabolic labeling enables selective photocrosslinking of O-GlcNAc-modified proteins to their binding partners. Proc Natl Acad Sci USA. 2012;109:4834–9.
Ge Y, Lu H, Yang B, Woo CM. Small molecule-activated O -GlcNAcase for spatiotemporal removal of O -GlcNAc in live cells. ACS Chem Biol. 2023;18:193–201.
He J, Fan Z, Tian Y, Yang W, Zhou Y, Zhu Q, et al. Spatiotemporal activation of protein O-GlcNAcylation in living cells. J Am Chem Soc. 2022;144:4289–93.
Zhu Y, Hart GW. Dual-specificity RNA aptamers enable manipulation of target-specific O-GlcNAcylation and unveil functions of O-GlcNAc on β-catenin. Cell. 2022;S0092-8674:01529–0152.
Acknowledgements
We would like to thank the members of the Qian and Xu laboratories for helpful discussions. Thanks to Jiajia Song for her advice and encouragement.
Funding
This work has been supported by National Natural Science Foundation of China (grant No. 32000903), and the Scientific Research Foundation of Jiangsu University for Senior Professional Talents (20JDG48).
Author information
Authors and Affiliations
Contributions
QJZ and ZWX conceived and wrote the first draft of both text and figures. SSZ, WHL, and HQ conceptualized, wrote, revised, and edited the manuscript.
Corresponding authors
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.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Zhao, Q., Zhou, S., Lou, W. et al. Crosstalk between O-GlcNAcylation and phosphorylation in metabolism: regulation and mechanism. Cell Death Differ 32, 1181–1199 (2025). https://doi.org/10.1038/s41418-025-01473-z
Received:
Revised:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41418-025-01473-z