Korean Diabetes J.  2010 Aug;34(4):211-219. 10.4093/kdj.2010.34.4.211.

O-GlcNAc Modification: Friend or Foe in Diabetic Cardiovascular Disease

Affiliations
  • 1Department of Medical Sciences, Kyungpook National University School of Medicine, Daegu, Korea.
  • 2Department of Fundamental Medical and Pharmaceutical Sciences, Catholic University of Daegu CU Leaders' College, Gyeongsan, Korea. syjeoung@cu.ac.kr

Abstract

O-Linked beta-N-acetyl glucosaminylation (O-GlcNAcylation) is a dynamic post-translational modification that occurs on serine and threonine residues of cytosolic and nuclear proteins in all cell types, including those involved in the cardiovascular system. O-GlcNAcylation is thought to act in a manner analogous to protein phosphorylation. O-GlcNAcylation rapidly cycles on/off proteins in a time scale similar to that for phosphorylation/dephosphorylation of proteins. Several studies indicate that O-GlcNAc might induce nuclear localization of some transcription factors and may affect their DNA binding activities. However, at the cellular level, it has been shown that O-GlcNAc levels increase in response to stress and augmentation of this response suppresses cell survival. Increased levels of O-GlcNAc have been implicated as a pathogenic contributor to glucose toxicity and insulin resistance, which are major hallmarks of type 2 diabetes and diabetes-related cardiovascular complications. Thus, O-GlcNAc and its metabolic functions are not yet well-understood; focusing on the role of O-GlcNAc in the cardiovascular system is a viable target for biomedical investigation. In this review, we summarize our current understanding of the role of O-GlcNAc on the regulation of cell function and survival in the cardiovascular system.

Keyword

Acetylglucosaminidase; Diabetes mellitus, type 2; Glycosyltransferase; Vascular diseases

MeSH Terms

Acetylglucosaminidase
Cardiovascular Diseases
Cardiovascular System
Cell Survival
Cytosol
Diabetes Mellitus, Type 2
DNA
Friends
Glucose
Humans
Insulin Resistance
Nuclear Proteins
Phosphorylation
Protein Processing, Post-Translational
Proteins
Serine
Threonine
Transcription Factors
Vascular Diseases
Acetylglucosaminidase
DNA
Glucose
Nuclear Proteins
Proteins
Serine
Threonine
Transcription Factors

Figure

  • Fig. 1 Interplay between O-GlcNAcylation and protein phosphorylation. (A) O-GlcNAc modification is strongly dependent on the concentration of UDP-GlcNAc produced by the hexosamine biosynthetic pathway (HBP). OGT uses UDP-GlcNAc as a substrate for GlcNAcylation on protein serine and threonine residues. O-GlcNAcase (OGA) removes the O-GlcNAc moiety from O-GlcNAc-modified proteins. PUGNAC inhibits the activity of O-GlcNAcase. (B) O-GlcNAc modification is analogous to protein phosphorylation/dephosphorylation. Interplay between GlcNAcylation and phosphorylation can affect the activities or stabilities of the proteins, and both processes can occur on the same protein at proximal sites.

  • Fig. 2 A fraction (2-5%) of the glucose entering a cell is directed into the hexosamine biosynthesis pathway (HBP) pathway. GFAT (glutamine: fructose-6-phosphate amidotransferase) uses glutamine to convert fructose-6-phosphate into glucosamine-6-phosphate, which is then used for the synthesis of UDP-GlcNAc in the cell. Activation of the HBP pathway acts through Sp1 sites to increase plasminogen activator inhibitor-1 (PAI-1) and tumor growth factor-alpha expressions. At the same time, O-GlcNAc modification of endothelial nitric oxide synthase (eNOS) decreases its activity and nitric oxide (NO) production.

  • Fig. 3 Hyperglycemia-induced activation of O-linked N-acetyl glucosamine activates GPX1 and its binding to c-Abl and Arg kinases and protects the cell via the antioxidant response. Along with an increase in the antioxidant response, increased O-GlcNAcylation activates p38 MAPK phosphorylation with increased glucose transport. Increased O-GlcNAc formation also increases the level of mitochondrial Bcl2, which inhibits apoptosis and protects the cell during stress.


Reference

1. Copeland RJ, Bullen JW, Hart GW. Cross-talk between GlcNAcylation and phosphorylation: roles in insulin resistance and glucose toxicity. Am J Physiol Endocrinol Metab. 2008. 295:E17–E28.
2. Wells L, Vosseller K, Hart GW. Glycosylation of nucleocytoplasmic proteins: signal transduction and O-GlcNAc. Science. 2001. 291:2376–2378.
3. Guinez C, Morelle W, Michalski JC, Lefebvre T. O-GlcNAc glycosylation: a signal for the nuclear transport of cytosolic proteins? Int J Biochem Cell Biol. 2005. 37:765–774.
4. Zachara NE, Hart GW. 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. 2004. 1673:13–28.
5. Butkinaree C, Park K, Hart GW. O-linked β-N-acetyl glucosamine (O-GlcNAc): extensive crosstalk with phosphorylation to regulate signaling and transcription in response to nutrients and stress. Biochim Biophys Acta. 2010. 1800:96–106.
6. Bouche C, Serdy S, Kahn CR, Goldfine AB. The cellular fate of glucose and its relevance in type-2 diabetes. Endocr Rev. 2004. 25807–25830.
7. Hanover JA. Glycan-dependent signaling: O-linked N-acetyl glucosamine. FASEB J. 2001. 15:1865–1876.
8. Buse MG. Hexosamines, insulin resistance, and the complications of diabetes; current status. Am J Physiol Endocrinol Metab. 2006. 290:E1–E8.
9. Kornfeld R. Studies on L-glutamine D-fructose 6-phosphate amidotransferase. I. Feedback inhibition by uridine diphosphate-N-acetylglucosamine. J Biol Chem. 1967. 242:3135–3141.
10. Marshall S, Nadeau O, Yamasaki K. Dynamic actions of glucose and glucosamine on Hexosamine biosynthesis in isolated adipocytes: differential effects on glucosamine 6-phosphate, UDP-N-acetyl glucosamine and ATP levels. J Biol Chem. 2004. 279:35313–35319.
11. Haltiwanger RS, Holt GD, Hart GW. Enzymatic addition of O-GlcNAc to nuclear and cytosolic proteins. Identification of a uridine diphospho-N-acetyl glucosamine: peptide beta-Nacetylglucosaminyl transferase. J Biol Chem. 1990. 265:2563–2568.
12. Shafi R, Lyer SP, Ellies LG, O'Donnell N, Marek KW, Chui D, Hart GW, Marth JD. 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 U S A. 2000. 97:5735–5739.
13. 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 U S A. 2008. 105:13793–13798.
14. Defronzo RA, Ferrannini E. Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia and atherosclerotic cardiovascular disease. Diabetes care. 1991. 14:173–194.
15. Marshall S, Bacote V, Traxinger RR. Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of Hexosamine biosynthesis in the induction of insulin resistance. J Biol Chem. 1991. 266:4706–4712.
16. Marshall S. Role of insulin, adipocyte hormones, and nutrient-sensing pathways in regulating fuel metabolism and energy homeostasis: a nutritional prospective of diabetes, obesity and cancer. Sci STKE. 2006. 2006:re7.
17. Rajapakse AG, Ming XF, Carvas JM, Yang Z. The hexosamine biosynthesis inhibitor azaserine prevents endothelial inflammation and dysfunction under hyperglycemic condition through antioxidant effects. Am J Physiol Heart Circ Physiol. 2009. 296:H815–H822.
18. Werstuck GH, Khan MI, Femia G, Kim AJ, Tedesco V, Trigatti B, Shi Y. Glucosamine-induced endoplasmic reticulum dysfunction is associated with accelerated atherosclerosis in a hyperglycemic mouse model. Diabetes. 2006. 55:93–101.
19. Vosseller K, Wells L, Lane MD, Hart GW. Elevated nucleocytoplasmic glycosylation by O-GlcNAc results in insulin resistance associated with defects in AKT activation in 3T3-L1 adipocytes. Proc Natl Acad Sci U S A. 2002. 99:5313–5318.
20. Cooksey RC, Hebert LF Jr, Zhu JH, Wofford P, Garrey WT, McClain DA. Mechanism of Hexosamine-induced insulin resistance in transgenic mice over-expressing glutamine: fructose-6-phosphate amidotransferase: decreased glucose transporter GLUT4 translocation and reversal by treatment with thiazolidinedione. Endocrinology. 1999. 140:1151–1157.
21. Brozinick JT Jr, Berkemeier BA, Elmendorf JS. Acting on GLUT4: membrane and cytoskeletal components of insulin action. Curr Diabetes Rev. 2007. 3:111–122.
22. Bhonagiri P, Pattar GR, Horvath EM, Habegger KM, McCarthy AM, Elmendof JS. Hexosamine biosynthesis pathway flux contributes to insulin resistance via altering membrane phosphatidylinositol 4, 5-bisphosphate and cortical filamentous actin. Endocrinology. 2009. 150:1636–1645.
23. Gandy JC, Rountree AE, Bijur GN. Akt is dynamically modified with O-GlcNAc following treatments with PUGNAC and insulin-like growth factor-1. FEBS Lett. 2006. 580:3051–3058.
24. Yang X, Ongusaha PP, Miles PD, Havstad JC, Zhang F, So WV, Kudlow JE, Michell RH, Olefsky JM, Field SJ, Evans RM. Phosphoinositide signaling links O-GlcNAc transferase to insulin resistance. Nature. 2008. 451:964–969.
25. Schalkwijk CG, Stehouwer CD. Vascular complications in diabetes mellitus: the role of endothelial dysfunction. Clin Sci. 2005. 109:143–159.
26. Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, Szado C, Brownlee M. Inhibition of GAPDH activity by poly (ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest. 2003. 112:1049–1057.
27. Nerlich AG, Sauer U, Kolm-Lilly V, Wagner E, Koch M, Schleicher ED. Expression of glutamine: fructose 6-phosphate amidotransferase in human tissues; evidence for high variability and distinct regulation in diabetes. Diabetes. 1998. 47:170–178.
28. Hall JL, Chatham JC, Eldar-Finkelman H, Gibbons GH. Upregulation of glucose metabolism during intimal lesion formation is coupled to the inhibition of vascular smooth muscle cell apoptosis: role of GSK3beta. Diabetes. 2001. 50:1171–1179.
29. Akimoto Y, Kreppel LK, Hirano H, Hart GW. Hyperglycemia and the O-GlcNAc transferase in rat aortic smooth muscle cells; elevated expression and altered patterns of O-GlcNAcylation. Arch Biochem Biophys. 2001. 389:166–175.
30. McClain DA, Paterson AJ, Roos MD, Wei X, Kudlow JE. Glucose and glucosamine regulate growth factor gene expression in vascular smooth muscle cells. Proc Natl Acad Sci U S A. 1992. 89:8150–8154.
31. Slawson C, Zachara NE, Vosseller K, Cheung WD, Lane MD, Hart GW. Perturbations in O-linked beta-N-acetyl glucosamine protein modification cause severe defects in mitotic progression and cytokinesis. J Biol Chem. 2005. 280:32944–32956.
32. Williams B, Schrier RW. Characterization of glucose-induced in situ protein kinase C activity in cultured vascular smooth muscle cells. Diabetes. 1992. 41:1464–1472.
33. Hattori Y, Hattori S, Sato N, Kasai K. High-glucose-induced nuclear factor kB activation in vascular smooth muscle cells. Cardiovasc Res. 2000. 46:188–197.
34. Yang WH, Park SY, Nam HW, Kim do H, Kang JG, Kang ES, Kim YS, Lee HC, Kim KS, Cho JW. NF-kB activation is associated with its O-GlcNAcylation state under hyperglycemic conditions. Proc Natl Acad Sci U S A. 2008. 105:17345–17350.
35. Fulop N, Marchase RB, Chatham JC. Role of protein O-linked N-acetyl glucosamine in mediating cell function and survival in the cardiovascular system. Cardiovasc Res. 2007. 73:288–297.
36. Lima VV, Giachini FR, Carneiro FS, Carneiro ZN, Saleh MA, Pollock DM, Fortes ZB, Carvalho MH, Ergul A, Webb RC, Tostes RC. O-GlcNAcylation contributes to augmented vascular reactivity induced by endothelin 1. Hypertension. 2010. 55:180–188.
37. Hilgers RH, Webb RC. Molecular aspects of arterial smooth muscle contraction: focus on Rho. Exp Biol Med. 2005. 230:829–835.
38. Clark RJ, McDonough PM, Swanson E, Trost SU, Suzuki M, Fukuda M, Dillmann WH. Diabetes and the accompanying hyperglycemia impairs cardiomyocyte calcium cycling through increased nuclear O-GlcNAcylation. J Biol Chem. 2003. 278:44230–44237.
39. Song M, Kim HS, Park JM, Kim SH, Kim IH, Ryu SH, Suh PG. O-GlcNAc transferase is activated by CAMKIV-dependent phosphorylation under potassium chloride-induced depolarization in NG-108-15 cells. Cell Signal. 2008. 20:94–104.
40. Hirano K. Current topics in the regulatory mechanism underlying the Ca2+ sensitization of the contractile apparatus in vascular smooth muscle. J Pharmacol Sci. 2007. 104:109–115.
41. Cheung WD, Sakabe K, Housley MP, Dias WB, Hart GW. O-linked Beta-N-acetyl glucosaminyl transferease substrate specificity is regulated by myosin phosphatase targeting and other interacting proteins. J Biol Chem. 2008. 283:33935–33941.
42. Calles-Escandon J, Cipolla M. Diabetes and endothelial dysfunction: a clinical perspective. Endocr Rev. 2001. 22:36–52.
43. Makimattila S, Virkamaki A, Groop PH, Cockcroft J, Utriainen T, Fagerudd J, Yki-Jarvinen H. Chronic hyperglycemia impairs endothelial function and insulin sensitivity via different mechanisms in insulin dependent diabetes mellitus. Circulation. 1996. 94:1276–1282.
44. Yetik-Anacak G, Catravas JD. Nitric oxide and the endothelium: history and impact on cardiovascular disease. Vascul Pharmacol. 2006. 45:268–276.
45. Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, Brownlee M. Hyperglycemia inhibits endothelial nitric oxide synthase activity by post-translational modification at the AKT site. J Clin Invest. 2001. 108:1341–1348.
46. Federici M, Menghini R, Mauriello A, Hribal ML, Ferrelli F, Lauro D, Sbraccia P, Spagnoli LG, Sesti G, Lauro R. Insulin-dependent activation of endothelial nitric oxide synthase is impaired by O-linked glycosylation modification of signaling proteins in human coronary endothelial cells. Circulation. 2002. 106:466–472.
47. Du XL, Edelstein D, Rossetti L, Fantus IG, Goldberg H, Ziyadeh F, Wu J, Brownlee M. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc Natl Acad Sci U S A. 2000. 97:12222–12226.
48. Yao D, Taguchi T, Matsumura T, Pestell R, Edelstein D, Giardino I, Suske G, Rabbani N, Thornalley PJ, Sarthy VP, Hammes HP, Brownlee M. High glucose increases angiopoietin-2 transcription in micro vascular endothelial cells through methylglyoxal modification of mSin3A. J Biol Chem. 2007. 282:31038–31045.
49. Hata Y, Duh E, Zhang K, Robinson GS, Aiello LP. Transcription factors Sp1 and Sp3 alter vascular endothelial growth factor receptor expression through a novel recognition sequence. J Biol Chem. 1998. 273:19294–19303.
50. Fath-Ordoubadi F, Beatt KJ. Glucose-insulin-potassium therapy for treatment of acute myocardial infarction: an overview of randomized placebo-controlled trials. Circulation. 1997. 96:1152–1156.
51. Sohn KC, Lee KY, Park JE, Do SI. OGT functions as a catalytic chaperone under heat stress response: a unique defense role of OGT in hyperthermia. Biochem Biophys Res Commun. 2004. 322:1045–1051.
52. Zachara NE, O'Donnell N, Cheung WD, Mercer JJ, Marth JD, Hart GW. Dynamic O-GlcNAc modification of nucleocytoplasmic proteins in response to stress. A survival response of mammalian cells. J Biol Chem. 2004. 279:30133–30142.
53. Lim KH, Chang HI. O-linked N-acetyl glucosamine suppresses thermal aggregation of Sp1. FEBS Lett. 2006. 580:4645–4652.
54. Xing D, Feng W, Not LG, Miller AP, Zhang Y, Chen YF, Majid-Hassan E, Chatham JC, Oparil S. Increased protein O-GlcNAc modification inhibits inflammatory and neointimal responses to acute endoluinal arterial injury. Am J Physiol Heart Circ Physiol. 2008. 295:H335–H342.
55. Zou L, Yang S, Champattanachai V, Shunhua HU, Chaudry IH, Marchase RB, Chatham JC. Glucosamine improves cardiac function following trauma-hemorrhage by increased protein O-GlcNAcylation and attenuation of NF-kB signaling. Am J Physiol Heart Circ Physiol. 2009. 296:H515–H523.
56. Champattanachai V, Marchase RB, Chatham JC. Glucosamine protects neonatal cardiomyocytes from ischemia-reperfusion injury via increased protein-associated O-GlcNAc. Am J Physiol Cell Physiol. 2007. 292:C178–C187.
57. Schaffer SW, Croft CB, Solodushko V. Cardioprotective effect of chronic hyperglycemia: effect on hypoxia-induced apoptosis and necrosis. Am J Physiol Heart Circ Physiol. 2000. 278:H1948–H1954.
58. Ngoh GA, Jones SP. O-GlcNAc signaling attenuates mitochondrial membrane permeability transition. FASEB J. 2008. 22:1130.8. [Meeting Abstract].
59. Champattanachai V, Marchase RB, Chatham JC. Glucosamine protects neonatal cardiomyocytes from ischemia-reperfusion injury via increased protein O-GlcNAc and increased mitochondrial Bcl-2. Am J Physiol Cell Physiol. 2008. 294:C1509–C1520.
Full Text Links
  • KDJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr