Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2017

Open Access 01-12-2017 | Research article

Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice

Authors: Nicholas B. Hastings, Xiaohai Wang, Lixin Song, Brent D. Butts, Diane Grotz, Richard Hargreaves, J. Fred Hess, Kwok-Lam Karen Hong, Cathy Ruey-Ruey Huang, Lynn Hyde, Maureen Laverty, Julie Lee, Diane Levitan, Sherry X. Lu, Maureen Maguire, Veeravan Mahadomrongkul, Ernest J. McEachern, Xuesong Ouyang, Thomas W. Rosahl, Harold Selnick, Michaela Stanton, Giuseppe Terracina, David J. Vocadlo, Ganfeng Wang, Joseph L. Duffy, Eric M. Parker, Lili Zhang

Published in: Molecular Neurodegeneration | Issue 1/2017

Login to get access

Abstract

Background

Hyperphosphorylation of microtubule-associated protein tau is a distinct feature of neurofibrillary tangles (NFTs) that are the hallmark of neurodegenerative tauopathies. O-GlcNAcylation is a lesser known post-translational modification of tau that involves the addition of N-acetylglucosamine onto serine and threonine residues. Inhibition of O-GlcNAcase (OGA), the enzyme responsible for the removal of O-GlcNAc modification, has been shown to reduce tau pathology in several transgenic models. Clarifying the underlying mechanism by which OGA inhibition leads to the reduction of pathological tau and identifying translatable measures to guide human dosing and efficacy determination would significantly facilitate the clinical development of OGA inhibitors for the treatment of tauopathies.

Methods

Genetic and pharmacological approaches are used to evaluate the pharmacodynamic response of OGA inhibition. A panel of quantitative biochemical assays is established to assess the effect of OGA inhibition on pathological tau reduction. A “click” chemistry labeling method is developed for the detection of O-GlcNAcylated tau.

Results

Substantial (>80%) OGA inhibition is required to observe a measurable increase in O-GlcNAcylated proteins in the brain. Sustained and substantial OGA inhibition via chronic treatment with Thiamet G leads to a significant reduction of aggregated tau and several phosphorylated tau species in the insoluble fraction of rTg4510 mouse brain and total tau in cerebrospinal fluid (CSF). O-GlcNAcylated tau is elevated by Thiamet G treatment and is found primarily in the soluble 55 kD tau species, but not in the insoluble 64 kD tau species thought as the pathological entity.

Conclusion

The present study demonstrates that chronic inhibition of OGA reduces pathological tau in the brain and total tau in the CSF of rTg4510 mice, most likely by directly increasing O-GlcNAcylation of tau and thereby maintaining tau in the soluble, non-toxic form by reducing tau aggregation and the accompanying panoply of deleterious post-translational modifications. These results clarify some conflicting observations regarding the effects and mechanism of OGA inhibition on tau pathology, provide pharmacodynamic tools to guide human dosing and identify CSF total tau as a potential translational biomarker. Therefore, this study provides additional support to develop OGA inhibitors as a treatment for Alzheimer’s disease and other neurodegenerative tauopathies.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Ann Rev Neurosci. 2001;24:1121–59.CrossRefPubMed Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Ann Rev Neurosci. 2001;24:1121–59.CrossRefPubMed
2.
3.
go back to reference Goedert M, Jakes R. Mutations causing neurodegenerative tauopathies. Biochim Biophys Acta. 1739;2005:240–50. Goedert M, Jakes R. Mutations causing neurodegenerative tauopathies. Biochim Biophys Acta. 1739;2005:240–50.
4.
go back to reference Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, Pickering-Brown S, Chakraverty S, Isaacs A, Grover A, et al. Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17. Nature. 1998;393:702–5.CrossRefPubMed Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, Pickering-Brown S, Chakraverty S, Isaacs A, Grover A, et al. Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17. Nature. 1998;393:702–5.CrossRefPubMed
5.
go back to reference 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
7.
go back to reference Yuzwa SA, Shan X, Macauley MS, Clark T, Skorobogatko Y, Vosseller K, Vocadlo DJ. Increasing O-GlcNAc slows neurodegeneration and stabilizes tau against aggregation. Nat Chem Biol. 2012;8:393–9.CrossRefPubMed Yuzwa SA, Shan X, Macauley MS, Clark T, Skorobogatko Y, Vosseller K, Vocadlo DJ. Increasing O-GlcNAc slows neurodegeneration and stabilizes tau against aggregation. Nat Chem Biol. 2012;8:393–9.CrossRefPubMed
8.
go back to reference Borghgraef P, Menuet C, Theunis C, Louis J, Devijver H, Maurin H, Smet-Nocca C, Lippens G, Hilaire G, Gijsen H, et al. Increasing brain protein O-GlcNAc-ylation mitigates breathing defects and mortality of tau.P301L mice. PLoS One. 2013;8:e8442.CrossRef Borghgraef P, Menuet C, Theunis C, Louis J, Devijver H, Maurin H, Smet-Nocca C, Lippens G, Hilaire G, Gijsen H, et al. Increasing brain protein O-GlcNAc-ylation mitigates breathing defects and mortality of tau.P301L mice. PLoS One. 2013;8:e8442.CrossRef
9.
go back to reference Graham DL, Gray AJ, Joyce JA, Yu D, O’Moore J, Carlson GA, Shearman MS, Dellovade TL, Hering H. Increased O-GlcNAcylation reduces pathological tau without affecting its normal phosphorylation in a mouse model of tauopathy. Neuropharmacology. 2014;79:307–13.CrossRefPubMed Graham DL, Gray AJ, Joyce JA, Yu D, O’Moore J, Carlson GA, Shearman MS, Dellovade TL, Hering H. Increased O-GlcNAcylation reduces pathological tau without affecting its normal phosphorylation in a mouse model of tauopathy. Neuropharmacology. 2014;79:307–13.CrossRefPubMed
10.
go back to reference Khanna MR, Kovalevich J, Lee VM, Trojanowski JQ, Brunden KR. Therapeutic strategies for the treatment of tauopathies: hopes and challenges. Alzheimers Dement. 2016;12:1051–65.CrossRefPubMed Khanna MR, Kovalevich J, Lee VM, Trojanowski JQ, Brunden KR. Therapeutic strategies for the treatment of tauopathies: hopes and challenges. Alzheimers Dement. 2016;12:1051–65.CrossRefPubMed
11.
go back to reference SantaCruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McGowan E, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81.CrossRefPubMedPubMedCentral SantaCruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McGowan E, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81.CrossRefPubMedPubMedCentral
12.
go back to reference Barten DM, Cadelina GW, Hoque N, DeCarr LB, Guss VL, Yang L, Sankaranarayanan S, Wes PD, Flynn ME, Meredith JE, et al. Tau transgenic mice as models for cerebrospinal fluid tau biomarkers. J Alzheimers Dis. 2011;Suppl 2:127–41. Barten DM, Cadelina GW, Hoque N, DeCarr LB, Guss VL, Yang L, Sankaranarayanan S, Wes PD, Flynn ME, Meredith JE, et al. Tau transgenic mice as models for cerebrospinal fluid tau biomarkers. J Alzheimers Dis. 2011;Suppl 2:127–41.
13.
go back to reference Song L, Lu S, Ouyang X, Melchor J, Lee J, Terracina G, Wang X, Hyde L, Hess J, Parker E, Zhang L. Analysis of tau post-translational modifications in rTg4510 mice, a model of tau pathology. Mol Neurodegener. 2015;10:14.CrossRefPubMedPubMedCentral Song L, Lu S, Ouyang X, Melchor J, Lee J, Terracina G, Wang X, Hyde L, Hess J, Parker E, Zhang L. Analysis of tau post-translational modifications in rTg4510 mice, a model of tau pathology. Mol Neurodegener. 2015;10:14.CrossRefPubMedPubMedCentral
14.
go back to reference Ramsden M, Kotilinek L, Forster C, Paulson J, McGowan E, SantaCruz K, Guimaraes A, Yue M, Lewis J, Carlson G, et al. Age-dependent neurofibrillary tangle formation, neuron loss, and memory impairment in a mouse model of human tauopathy (P301L). J Neurosci. 2005;25:10637–47.CrossRefPubMed Ramsden M, Kotilinek L, Forster C, Paulson J, McGowan E, SantaCruz K, Guimaraes A, Yue M, Lewis J, Carlson G, et al. Age-dependent neurofibrillary tangle formation, neuron loss, and memory impairment in a mouse model of human tauopathy (P301L). J Neurosci. 2005;25:10637–47.CrossRefPubMed
15.
go back to reference Yuzwa SA, Macauley MS, Heinonen JE, Shan X, Dennis RJ, He Y, Whitworth GE, Stubbs KA, McEachern EJ, Davies GJ, Vocadlo DJ. A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat Chem Biol. 2008;4:483–90.CrossRefPubMed Yuzwa SA, Macauley MS, Heinonen JE, Shan X, Dennis RJ, He Y, Whitworth GE, Stubbs KA, McEachern EJ, Davies GJ, Vocadlo DJ. A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat Chem Biol. 2008;4:483–90.CrossRefPubMed
16.
go back to reference Yang YR, Song M, Lee H, Jeon Y, Choi EJ, Jang HJ, Moon HY, Byun HY, Kim EK, Kim DH, et al. O-GlcNAcase is essential for embryonic development and maintenance of genomic stability. Aging Cell. 2012;11:439–48.CrossRefPubMed Yang YR, Song M, Lee H, Jeon Y, Choi EJ, Jang HJ, Moon HY, Byun HY, Kim EK, Kim DH, et al. O-GlcNAcase is essential for embryonic development and maintenance of genomic stability. Aging Cell. 2012;11:439–48.CrossRefPubMed
17.
go back to reference Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med. 2012;2:a006247.CrossRefPubMedPubMedCentral Mandelkow EM, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med. 2012;2:a006247.CrossRefPubMedPubMedCentral
18.
go back to reference Arnold CS, Johnson GV, Cole RN, Dong DL, Lee M, Hart GW. The microtubule-associated protein tau is extensively modified with O-linked N-acetylglucosamine. J Biol Chem. 1996;271:26741–4. Arnold CS, Johnson GV, Cole RN, Dong DL, Lee M, Hart GW. The microtubule-associated protein tau is extensively modified with O-linked N-acetylglucosamine. J Biol Chem. 1996;271:26741–4.
19.
go back to reference Morris M, Knudsen GM, Maeda S, Trinidad JC, Ioanoviciu A, Burlingame AL, Mucke L. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci. 2015;18:1183–9.CrossRefPubMed Morris M, Knudsen GM, Maeda S, Trinidad JC, Ioanoviciu A, Burlingame AL, Mucke L. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci. 2015;18:1183–9.CrossRefPubMed
20.
go back to reference Yuzwa SA, Shan X, Jones BA, Zhao G, Woodward ML, Li X, Zhu Y, McEachern EJ, Silverman MA, Watson NV, et al. Pharmacological inhibition of O-GlcNAcase (OGA) prevents cognitive decline and amyloid plaque formation in bigenic tau/APP mutant mice. Mol Neurodegener. 2014;9:42.CrossRefPubMedPubMedCentral Yuzwa SA, Shan X, Jones BA, Zhao G, Woodward ML, Li X, Zhu Y, McEachern EJ, Silverman MA, Watson NV, et al. Pharmacological inhibition of O-GlcNAcase (OGA) prevents cognitive decline and amyloid plaque formation in bigenic tau/APP mutant mice. Mol Neurodegener. 2014;9:42.CrossRefPubMedPubMedCentral
21.
go back to reference Yu Y, Zhang L, Li X, Run X, Liang Z, Li Y, Liu Y, Lee MH, Grundke-Iqbal I, Iqbal K, et al. Differential effects of an O-GlcNAcase inhibitor on tau phosphorylation. PlosOne. 2012;7:e35277.CrossRef Yu Y, Zhang L, Li X, Run X, Liang Z, Li Y, Liu Y, Lee MH, Grundke-Iqbal I, Iqbal K, et al. Differential effects of an O-GlcNAcase inhibitor on tau phosphorylation. PlosOne. 2012;7:e35277.CrossRef
22.
go back to reference Yuzwa SA, Cheung AH, Okon M, McIntosh LP, Vocadlo DJ. O-GlcNAc modification of tau directly inhibits its aggregation without perturbing the conformational properties of tau monomers. J Mol Biol. 2014;426:1736–52.CrossRefPubMed Yuzwa SA, Cheung AH, Okon M, McIntosh LP, Vocadlo DJ. O-GlcNAc modification of tau directly inhibits its aggregation without perturbing the conformational properties of tau monomers. J Mol Biol. 2014;426:1736–52.CrossRefPubMed
23.
go back to reference Hatcher NG, Miller RA, Ke Z, Lee J, Cardasis HL, Terracina G, Zhang L, Marcus J, Wang X, Toolan DM, et al. Discovery proteomics to targeted biomarker assays: identification of tau post-translational modifications that track O-GlcNAcase inhibition by Thiamet G. J Am Soc Mass Spectrom. 2015;26(Supplement 1):52. Hatcher NG, Miller RA, Ke Z, Lee J, Cardasis HL, Terracina G, Zhang L, Marcus J, Wang X, Toolan DM, et al. Discovery proteomics to targeted biomarker assays: identification of tau post-translational modifications that track O-GlcNAcase inhibition by Thiamet G. J Am Soc Mass Spectrom. 2015;26(Supplement 1):52.
24.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
25.
go back to reference Marotta NP, Lin YH, Lewis YE, Ambroso MR, Zaro BW, Roth MT, Arnold DB, Langen R, Pratt MR. O-GlcNAc modification blocks the aggregation and toxicity of the protein α-synuclein associated with Parkinson's disease. Nat Chem. 2015;7:913–20.CrossRefPubMedPubMedCentral Marotta NP, Lin YH, Lewis YE, Ambroso MR, Zaro BW, Roth MT, Arnold DB, Langen R, Pratt MR. O-GlcNAc modification blocks the aggregation and toxicity of the protein α-synuclein associated with Parkinson's disease. Nat Chem. 2015;7:913–20.CrossRefPubMedPubMedCentral
26.
go back to reference Gambetta MC, Müller J. O-GlcNAcylation prevents aggregation of the Polycomb group repressor polyhomeotic. Dev Cell. 2014;31:629–39.CrossRefPubMed Gambetta MC, Müller J. O-GlcNAcylation prevents aggregation of the Polycomb group repressor polyhomeotic. Dev Cell. 2014;31:629–39.CrossRefPubMed
27.
go back to reference Blennow K, Hampel H, Weiner M, Zetterberg H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol. 2010;6:131–44.CrossRefPubMed Blennow K, Hampel H, Weiner M, Zetterberg H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol. 2010;6:131–44.CrossRefPubMed
28.
go back to reference Sean M, Smith SM, Struyk A, Jonathan D, Declercq R, Marcus J, Toolan D, Wang X, Schachter JB, Cosden M, Pearson M, et al. Early clinical results and preclinical validation of the O-GlcNAcase (OGA) inhibitor MK-8719 as a novel therapeutic for the treatment of tauopathies. Alzheimers Dement. 2016;16:P261. Sean M, Smith SM, Struyk A, Jonathan D, Declercq R, Marcus J, Toolan D, Wang X, Schachter JB, Cosden M, Pearson M, et al. Early clinical results and preclinical validation of the O-GlcNAcase (OGA) inhibitor MK-8719 as a novel therapeutic for the treatment of tauopathies. Alzheimers Dement. 2016;16:P261.
29.
go back to reference Seibler J, Kleinridders A, Küter-Luks B, Niehaves S, Brüning JC, Schwenk F. Reversible gene knockdown in mice using a tight, inducible shRNA expression system. Nucleic Acid Res. 2007;35:e54.CrossRefPubMedPubMedCentral Seibler J, Kleinridders A, Küter-Luks B, Niehaves S, Brüning JC, Schwenk F. Reversible gene knockdown in mice using a tight, inducible shRNA expression system. Nucleic Acid Res. 2007;35:e54.CrossRefPubMedPubMedCentral
30.
go back to reference Liu K, McEachern EJ, Mu C, Selnick HG, Vocadlo DJ, Wang Y, Wei Z, Zhou Y, Zhu Y. Selective glycosidase inhibitors and uses thereof, vol. WO2012061972 A1; 2012. Liu K, McEachern EJ, Mu C, Selnick HG, Vocadlo DJ, Wang Y, Wei Z, Zhou Y, Zhu Y. Selective glycosidase inhibitors and uses thereof, vol. WO2012061972 A1; 2012.
31.
go back to reference Chalkley RJ, Thalhammer A, Schoepfer R, Burlingame AL. Identification of protein O-GlcNAcylation sites using electron transfer dissociation mass spectrometry on native peptides. Proc Natl Acad Sci. 2009;106:8894–9.CrossRefPubMedPubMedCentral Chalkley RJ, Thalhammer A, Schoepfer R, Burlingame AL. Identification of protein O-GlcNAcylation sites using electron transfer dissociation mass spectrometry on native peptides. Proc Natl Acad Sci. 2009;106:8894–9.CrossRefPubMedPubMedCentral
32.
go back to reference Di Domenico F, Owen JB, Sultana R, Sowell RA, Perluigi M, Cini C, Cai J, Pierce WM, Butterfield DA. The wheat germ agglutinin-fractionated proteome of subjects with Alzheimer's disease and mild cognitive impairment hippocampus and inferior parietal lobule: implications for disease pathogenesis and progression. J Neurosci Res. 2010;88:3566–77.CrossRefPubMed Di Domenico F, Owen JB, Sultana R, Sowell RA, Perluigi M, Cini C, Cai J, Pierce WM, Butterfield DA. The wheat germ agglutinin-fractionated proteome of subjects with Alzheimer's disease and mild cognitive impairment hippocampus and inferior parietal lobule: implications for disease pathogenesis and progression. J Neurosci Res. 2010;88:3566–77.CrossRefPubMed
33.
go back to reference Snow CM, Senior A, Gerace L. Monoclonal antibodies identify a Group of Nuclear Pore Complex Glycoproteins. J Cell Biol. 1987;104:1143–56.CrossRefPubMed Snow CM, Senior A, Gerace L. Monoclonal antibodies identify a Group of Nuclear Pore Complex Glycoproteins. J Cell Biol. 1987;104:1143–56.CrossRefPubMed
35.
go back to reference Wessel D, Flügge UI. A method for the quantitative recovery of protein in dilute solution in the presence of detergents and lipids. Anal Biochem. 1984;138:141–3.CrossRefPubMed Wessel D, Flügge UI. A method for the quantitative recovery of protein in dilute solution in the presence of detergents and lipids. Anal Biochem. 1984;138:141–3.CrossRefPubMed
36.
go back to reference Freeze H, Kranz C. Endoglycosidase and glycoamidase release of N-linked glycans. Curr Protoc Mol Biol. 2010;17.14A:1–25. Freeze H, Kranz C. Endoglycosidase and glycoamidase release of N-linked glycans. Curr Protoc Mol Biol. 2010;17.14A:1–25.
37.
go back to reference Seibler J, Küter-Luks B, Kern H, Streu S, Plum L, Mauer J, Kühn R, Brüning JC, Schwenk F. Single copy shRNA configuration for ubiquitous gene knockdown in mice. Nucleic Acid Res. 2005;33:e67.CrossRefPubMedPubMedCentral Seibler J, Küter-Luks B, Kern H, Streu S, Plum L, Mauer J, Kühn R, Brüning JC, Schwenk F. Single copy shRNA configuration for ubiquitous gene knockdown in mice. Nucleic Acid Res. 2005;33:e67.CrossRefPubMedPubMedCentral
Metadata
Title
Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice
Authors
Nicholas B. Hastings
Xiaohai Wang
Lixin Song
Brent D. Butts
Diane Grotz
Richard Hargreaves
J. Fred Hess
Kwok-Lam Karen Hong
Cathy Ruey-Ruey Huang
Lynn Hyde
Maureen Laverty
Julie Lee
Diane Levitan
Sherry X. Lu
Maureen Maguire
Veeravan Mahadomrongkul
Ernest J. McEachern
Xuesong Ouyang
Thomas W. Rosahl
Harold Selnick
Michaela Stanton
Giuseppe Terracina
David J. Vocadlo
Ganfeng Wang
Joseph L. Duffy
Eric M. Parker
Lili Zhang
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2017
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-017-0181-0

Other articles of this Issue 1/2017

Molecular Neurodegeneration 1/2017 Go to the issue