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:

Augmented O-GlcNAcylation exacerbates right ventricular dysfunction and remodeling via enhancement of hypertrophy, mitophagy, and fibrosis in mice exposed to long-term intermittent hypoxia

Abstract

Previously, we showed that augmented O-linked N-acetylglucosaminylation (O-GlcNAcylation) mitigates cardiac remodeling in O-GlcNAc transferase-transgenic (Ogt-Tg) mice exposed to acute (2-week) intermittent hypoxia (IH) by suppressing nuclear factor of activated T cells (NFAT) and nuclear factor kappa B (NF-κB) via the O-GlcNAcylation of glycogen synthase kinase 3 beta (GSK-3β) and NF-κB p65. Because this effect is time dependent, we exposed Ogt-Tg mice to IH for 4 weeks (IH4W) in the present study. O-GlcNAcylation was significantly enhanced in Ogt-Tg mice vs. wild-type (WT) mice exposed to normoxia and IH4W. Total O-GlcNAcylation levels were significantly increased in WT and Ogt-Tg mice after IH4W vs. normoxia. After IH4W, Ogt-Tg mice displayed significantly exacerbated signs of cardiac hypertrophy and fibrosis in the right ventricles (RVs) but not the left ventricles (LVs). Echocardiography revealed IH4W-induced right ventricular dysfunction. Phosphorylated GSK-3β levels were increased in Ogt-Tg mice vs. WT mice after IH4W, whereas phosphorylated NF-κB p65 levels were unaffected. Mitophagy, which is associated with cardiac dysfunction, was increased in the RVs of Ogt-Tg mice after IH4W. Furthermore, the levels of phosphorylated dynamin-related protein 1 (p-Drp1) were significantly increased, and the expression of mitofusin-2 (MFN2) was significantly decreased. In human embryonic kidney cells, mitochondrial uncoupler-induced mitochondrial dysfunction was accelerated in Ogt-overexpressing cells. In addition to increasing the levels of phosphorylated Smad2, IH4W promoted cardiac fibrosis in the RVs of Ogt-Tg mice. Thus, augmented O-GlcNAcylation may aggravate IH4W-induced right ventricular dysfunction and remodeling by promoting hypertrophy, mitophagy, and fibrosis via GSK-3β inactivation, an increased p-Drp-1/MFN2 ratio, and Smad2 activation, respectively.

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

Similar content being viewed by others

References

  1. 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.

    Article  CAS  Google Scholar 

  2. Hart GW, Housley MP, Slawson C. Cycling of O-linked beta-N-acetylglucosamine on nucleocytoplasmic proteins. Nature 2007;446:1017–22.

    Article  CAS  Google Scholar 

  3. Iyer SP, Hart GW. Dynamic nuclear and cytoplasmic glycosylation: enzymes of O-GlcNAc cycling. Biochemistry. 2003;42:2493–9.

    Article  CAS  Google Scholar 

  4. Jones SP, Zachara NE, Ngoh GA, Hill BG, Teshima Y, Bhatnagar A, et al. Cardioprotection by N-acetylglucosamine linkage to cellular proteins. Circulation. 2008;117:1172–82.

    Article  CAS  Google Scholar 

  5. Laczy B, Hill BG, Wang K, Paterson AJ, White CR, Xing D, et al. Protein O-GlcNAcylation: a new signaling paradigm for the cardiovascular system. Am J Physiol Heart Circ Physiol. 2009;296:H13–28.

    Article  CAS  Google Scholar 

  6. 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.

    Article  CAS  Google Scholar 

  7. Chen LM, Kuo WW, Yang JJ, Wang SG, Yeh YL, Tsai FJ, et al. Eccentric cardiac hypertrophy was induced by long-term intermittent hypoxia in rats. Exp Physiol. 2007;92:409–16.

    Article  CAS  Google Scholar 

  8. Pena E, Siques P, Brito J, Arribas SM, Boger R, Hannemann J, et al. Nox2 upregulation and p38alpha MAPK activation in right ventricular hypertrophy of rats exposed to long-term chronic intermittent hypobaric hypoxia. Int J Mol Sci. 2020;21:8576.

  9. Guan P, Sun ZM, Wang N, Zhou J, Luo LF, Zhao YS, et al. Resveratrol prevents chronic intermittent hypoxia-induced cardiac hypertrophy by targeting the PI3K/AKT/mTOR pathway. Life Sci. 2019;233:116748.

    Article  CAS  Google Scholar 

  10. Xie S, Deng Y, Pan YY, Ren J, Jin M, Wang Y, et al. Chronic intermittent hypoxia induces cardiac hypertrophy by impairing autophagy through the adenosine 5’-monophosphate-activated protein kinase pathway. Arch Biochem Biophys. 2016;606:41–52.

    Article  CAS  Google Scholar 

  11. 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:C1509–20.

    Article  CAS  Google Scholar 

  12. Genovese A, Chiariello M, Cacciapuoti AA, De Alfieri W, Latte S, Condorelli M. Inhibition of hypoxia-induced cardiac hypertrophy by verapamil in rats. Basic Res Cardiol. 1980;75:757–63.

    Article  CAS  Google Scholar 

  13. Jensen RV, Zachara NE, Nielsen PH, Kimose HH, Kristiansen SB, Botker HE. Impact of O-GlcNAc on cardioprotection by remote ischaemic preconditioning in non-diabetic and diabetic patients. Cardiovasc Res. 2013;97:369–78.

    Article  CAS  Google Scholar 

  14. 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:C178–87.

    Article  CAS  Google Scholar 

  15. Liu J, Marchase RB, Chatham JC. Increased O-GlcNAc levels during reperfusion lead to improved functional recovery and reduced calpain proteolysis. Am J Physiol Heart Circ Physiol. 2007;293:H1391–9.

    Article  CAS  Google Scholar 

  16. Liu J, Pang Y, Chang T, Bounelis P, Chatham JC, Marchase RB. Increased hexosamine biosynthesis and protein O-GlcNAc levels associated with myocardial protection against calcium paradox and ischemia. J Mol Cell Cardiol. 2006;40:303–12.

    Article  CAS  Google Scholar 

  17. Lunde IG, Aronsen JM, Kvaloy H, Qvigstad E, Sjaastad I, Tonnessen T, et al. Cardiac O-GlcNAc signaling is increased in hypertrophy and heart failure. Physiol Genomics. 2012;44:162–72.

    Article  CAS  Google Scholar 

  18. Wang D, Hu X, Lee SH, Chen F, Jiang K, Tu Z, et al. Diabetes exacerbates myocardial ischemia/reperfusion injury by down-regulation of microRNA and up-regulation of O-GlcNAcylation. JACC Basic Transl Sci. 2018;3:350–62.

    Article  Google Scholar 

  19. Nakagawa T, Furukawa Y, Hayashi T, Nomura A, Yokoe S, Moriwaki K, et al. Augmented O-GlcNAcylation attenuates intermittent hypoxia-induced cardiac remodeling through the suppression of NFAT and NF-kappaB activities in mice. Hypertens Res. 2019;42:1858–71.

    Article  CAS  Google Scholar 

  20. Moriwaki K, Asahi M. Augmented TME O-GlcNAcylation promotes tumor proliferation through the inhibition of p38 MAPK. Mol Cancer Res. 2017;15:1287–98.

    Article  CAS  Google Scholar 

  21. Nishioka S, Yoshioka T, Nomura A, Kato R, Miyamura M, Okada Y, et al. Celiprolol reduces oxidative stress and attenuates left ventricular remodeling induced by hypoxic stress in mice. Hypertens Res. 2013;36:934–9.

    Article  CAS  Google Scholar 

  22. Kato R, Nomura A, Sakamoto A, Yasuda Y, Amatani K, Nagai S, et al. Hydrogen gas attenuates embryonic gene expression and prevents left ventricular remodeling induced by intermittent hypoxia in cardiomyopathic hamsters. Am J Physiol Heart Circ Physiol. 2014;307:H1626–33.

    Article  CAS  Google Scholar 

  23. Yamashita C, Hayashi T, Mori T, Tazawa N, Kwak CJ, Nakano D, et al. Angiotensin II receptor blocker reduces oxidative stress and attenuates hypoxia-induced left ventricular remodeling in apolipoprotein E-knockout mice. Hypertens Res. 2007;30:1219–30.

    Article  CAS  Google Scholar 

  24. Hayashi T, James TN, Buckingham DC. Ultrastructure and immunohistochemistry of the coronary chemoreceptor in human and canine hearts. Am Heart J. 1995;129:946–59.

    Article  CAS  Google Scholar 

  25. Ogihara Y, Yamada N, Dohi K, Matsuda A, Tsuji A, Ota S, et al. Utility of right ventricular Tei-index for assessing disease severity and determining response to treatment in patients with pulmonary arterial hypertension. J Cardiol. 2014;63:149–53.

    Article  Google Scholar 

  26. Sun LY, Zhao H, Kang Y, Shen XD, Cai ZY, Shen JY, et al. Two-dimensional echocardiography in the assessment of long-term prognosis in patients with pulmonary arterial hypertension. PLoS One. 2014;9:e114443.

    Article  Google Scholar 

  27. Lanfranchi PA, Somers VK, Braghiroli A, Corra U, Eleuteri E, Giannuzzi P. Central sleep apnea in left ventricular dysfunction: prevalence and implications for arrhythmic risk. Circulation. 2003;107:727–32.

    Article  Google Scholar 

  28. Tugcu A, Guzel D, Yildirimturk O, Aytekin S. Evaluation of right ventricular systolic and diastolic function in patients with newly diagnosed obstructive sleep apnea syndrome without hypertension. Cardiology. 2009;113:184–92.

    Article  CAS  Google Scholar 

  29. Tugcu A, Yildirimturk O, Tayyareci Y, Demiroglu C, Aytekin S. Evaluation of subclinical right ventricular dysfunction in obstructive sleep apnea patients using velocity vector imaging. Circ J. 2010;74:312–9.

    Article  Google Scholar 

  30. Cho HJ, Heo W, Han JW, Lee YH, Park JM, Kang MJ, et al. Chronological change of right ventricle by chronic intermittent hypoxia in mice. Sleep. 2017;40.

  31. Smith KA, Waypa GB, Dudley VJ, Budinger GRS, Abdala-Valencia H, Bartom E, et al. Role of hypoxia-inducible factors in regulating right ventricular function and remodeling during chronic hypoxia-induced pulmonary hypertension. Am J Respir Cell Mol Biol. 2020;63:652–64.

    Article  CAS  Google Scholar 

  32. Bradley TD, Floras JS. Obstructive sleep apnoea and its cardiovascular consequences. Lancet. 2009;373:82–93.

    Article  Google Scholar 

  33. 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;266:4706–12.

    Article  CAS  Google Scholar 

  34. Gurel Z, Sieg KM, Shallow KD, Sorenson CM, Sheibani N. Retinal O-linked N-acetylglucosamine protein modifications: implications for postnatal retinal vascularization and the pathogenesis of diabetic retinopathy. Mol Vis. 2013;19:1047–59.

    CAS  Google Scholar 

  35. Park MJ, Kim DI, Lim SK, Choi JH, Han HJ, Yoon KC, et al. High glucose-induced O-GlcNAcylated carbohydrate response element-binding protein (ChREBP) mediates mesangial cell lipogenesis and fibrosis: the possible role in the development of diabetic nephropathy. J Biol Chem. 2014;289:13519–30.

    Article  CAS  Google Scholar 

  36. Yokoe S, Asahi M, Takeda T, Otsu K, Taniguchi N, Miyoshi E, et al. Inhibition of phospholamban phosphorylation by O-GlcNAcylation: implications for diabetic cardiomyopathy. Glycobiology. 2010;20:1217–26.

    Article  CAS  Google Scholar 

  37. Nomura A, Yokoe S, Tomoda K, Nakagawa T, Martin-Romero FJ, Asahi M. Fluctuation in O-GlcNAcylation inactivates STIM1 to reduce store-operated calcium ion entry via down-regulation of Ser(621) phosphorylation. J Biol Chem. 2020;295:17071–82.

    Article  CAS  Google Scholar 

  38. Pennanen C, Parra V, Lopez-Crisosto C, Morales PE, Del Campo A, Gutierrez T, et al. Mitochondrial fission is required for cardiomyocyte hypertrophy mediated by a Ca2+-calcineurin signaling pathway. J Cell Sci. 2014;127:2659–71.

    CAS  Google Scholar 

  39. Vasquez-Trincado C, Garcia-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA, et al. Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol. 2016;594:509–25.

    Article  CAS  Google Scholar 

  40. Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, et al. Fibroblast-specific TGF-beta-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest. 2017;127:3770–83.

    Article  Google Scholar 

  41. Saadat S, Noureddini M, Mahjoubin-Tehran M, Nazemi S, Shojaie L, Aschner M, et al. Pivotal role of TGF-beta/Smad signaling in cardiac fibrosis: non-coding RNAs as effectual players. Front Cardiovasc Med. 2020;7:588347.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to Yudai Yamaguchi, Yuhei Yamanaka, Maki Tamura (research assistants at the Department of Cardiovascular Pharmacotherapy and Toxicology, Faculty of Pharmacy) and Yumiko Okumura (Osaka Medical and Pharmaceutical University) for their expert technical assistance.

Funding

This study was partially supported by Grant-in-Aid for Scientific Research (C) no. 25460397 from the Japan Society for the Promotion of Science (MA) under the Ministry of Education, Science, Culture, Sports and Technology of Japan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michio Asahi.

Ethics declarations

Conflict of interest

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

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yokoe, S., Hayashi, T., Nakagawa, T. et al. Augmented O-GlcNAcylation exacerbates right ventricular dysfunction and remodeling via enhancement of hypertrophy, mitophagy, and fibrosis in mice exposed to long-term intermittent hypoxia. Hypertens Res 46, 667–678 (2023). https://doi.org/10.1038/s41440-022-01088-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41440-022-01088-8

Keywords

This article is cited by

Search

Quick links