Browse > Article
http://dx.doi.org/10.5483/BMBRep.2018.51.6.077

Degradation or aggregation: the ramifications of post-translational modifications on tau  

Park, Seoyoung (Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine)
Lee, Jung Hoon (Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine)
Jeon, Jun Hyoung (Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine)
Lee, Min Jae (Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine)
Publication Information
BMB Reports / v.51, no.6, 2018 , pp. 265-273 More about this Journal
Abstract
Tau protein is encoded in the microtubule-associated protein tau (MAPT) gene and contributes to the stability of microtubules in axons. Despite of its basic isoelectric point and high solubility, tau is often found in intraneuronal filamentous inclusions such as paired helical filaments (PHFs), which are the primary constituent of neurofibrillary tangles (NFTs). This pathological feature is the nosological entity termed "tauopathies" which notably include Alzheimer's disease (AD). A proteinaceous signature of all tauopathies is hyperphosphorylation of the accumulated tau, which has been extensively studied as a major pharmacological target for AD therapy. However, in addition to phosphorylation events, tau undergoes a number of diverse posttranslational modifications (PTMs) which appear to be controlled by complex crosstalk. It remains to be elucidated which of the PTMs or their combinations have pro-aggregation or anti-aggregation properties. In this review, we outline the consequences of and communications between several key PTMs of tau, such as acetylation, phosphorylation, and ubiquitination, focusing on their roles in aggregation and degradation. We place emphasis on the structure of tau protofilaments from the human AD brain, which may be good targets to modulate etiological PTMs which cause tau aggregation.
Keywords
Acetylation; Crosstalk; Phosphorylation; Posttranslational modification; Tau; Tauopathies; Ubiquitination;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Kenessey A and Yen SH (1993) The extent of phosphorylation of fetal tau is comparable to that of PHF-tau from Alzheimer paired helical filaments. Brain Res 629, 40-46   DOI
2 Lee G, Thangavel R, Sharma VM et al (2004) Phosphorylation of tau by fyn: implications for Alzheimer's disease. J Neurosci 24, 2304-2312   DOI
3 Cook C, Carlomagno Y, Gendron TF et al (2014) Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum Mol Genet 23, 104-116   DOI
4 Ding H, Dolan PJ and Johnson GV (2008) Histone deacetylase 6 interacts with the microtubule-associated protein tau. J Neurochem 106, 2119-2130   DOI
5 Fan SJ, Huang FI, Liou JP, Yang CR (2018) The novel histone de acetylase 6 inhibitor, MPT0G211, ameliorates tau phosphorylation and cognitive deficits in an Alzheimer's disease model. Cell Death Dis 9, 655   DOI
6 Carlomagno Y, Chung DC, Yue M et al (2017) An acetylation-phosphorylation switch that regulates tau aggregation propensity and function. J Biol Chem 292, 15277-15286   DOI
7 Julien C, Tremblay C, Emond V et al (2009) Sirtuin 1 reduction parallels the accumulation of tau in Alzheimer disease. J Neuropathol Exp Neurol 68, 48-58   DOI
8 Gorsky MK, Burnouf S, Sofola-Adesakin O et al (2017) Pseudo-acetylation of multiple sites on human Tau proteins alters Tau phosphorylation and microtubule binding, and ameliorates amyloid beta toxicity. Sci Rep 7, 9984   DOI
9 Min SW, Chen X, Tracy TE et al (2015) Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med 21, 1154-1162   DOI
10 Tracy TE, Sohn PD, Minami SS et al (2016) Acetylated Tau Obstructs KIBRA-Mediated Signaling in Synaptic Plasticity and Promotes Tauopathy-Related Memory Loss. Neuron 90, 245-260   DOI
11 Irwin DJ, Cohen TJ, Grossman M et al (2013) Acetylated tau neuropathology in sporadic and hereditary tauopathies. Am J Pathol 183, 344-351   DOI
12 Trzeciakiewicz H, Tseng JH, Wander CM et al (2017) A Dual Pathogenic Mechanism Links Tau Acetylation to Sporadic Tauopathy. Sci Rep 7, 44102   DOI
13 Shimura H, Schwartz D, Gygi SP and Kosik KS (2004) CHIP-Hsc70 complex ubiquitinates phosphorylated tau and enhances cell survival. J Biol Chem 279, 4869-4876   DOI
14 Hatakeyama S, Matsumoto M, Kamura T et al (2004) U-box protein carboxyl terminus of Hsc70-interacting protein (CHIP) mediates poly-ubiquitylation preferentially on four-repeat Tau and is involved in neurodegeneration of tauopathy. J Neurochem 91, 299-307   DOI
15 Sahara N, Murayama M, Mizoroki T et al (2005) In vivo evidence of CHIP up-regulation attenuating tau aggregation. J Neurochem 94, 1254-1263   DOI
16 Wang Y, Garg S, Mandelkow EM and Mandelkow E (2010) Proteolytic processing of tau. Biochem Soc Trans 38, 955-961   DOI
17 Joshi V, Amanullah A, Upadhyay A, Mishra R, Kumar A and Mishra A (2016) A Decade of Boon or Burden: What Has the CHIP Ever Done for Cellular Protein Quality Control Mechanism Implicated in Neurodegeneration and Aging? Front Mol Neurosci 9, 93
18 Gupta R, Lan M, Mojsilovic-Petrovic J et al (2017) The Proline/Arginine Dipeptide from Hexanucleotide Repeat Expanded C9ORF72 Inhibits the Proteasome. eNeuro 4, e0249-16
19 Shin SK, Kim JH, Lee JH et al (2017) Docosahexaenoic acid-mediated protein aggregates may reduce proteasome activity and delay myotube degradation during muscle atrophy in vitro. Exp Mol Med 49, e287   DOI
20 Lee JH, Shin SK, Jiang Y et al (2015) Facilitated Tau Degradation by USP14 Aptamers via Enhanced Proteasome Activity. Sci Rep 5, 10757   DOI
21 Keck S, Nitsch R, Grune T and Ullrich O (2003) Proteasome inhibition by paired helical filament-tau in brains of patients with Alzheimer's disease. J Neurochem 85, 115-122   DOI
22 Keller JN, Hanni KB and Markesbery WR (2000) Impaired proteasome function in Alzheimer's disease. J Neurochem 75, 436-439
23 Lopez Salon M, Morelli L, Castano EM, Soto EF and Pasquini JM (2000) Defective ubiquitination of cerebral proteins in Alzheimer's disease. J Neurosci Res 62, 302-310   DOI
24 Cardozo C and Michaud C (2002) Proteasome-mediated degradation of tau proteins occurs independently of the chymotrypsin-like activity by a nonprocessive pathway. Arch Biochem Biophys 408, 103-110   DOI
25 Zhang JY, Liu SJ, Li HL and Wang JZ (2005) Microtubule-associated protein tau is a substrate of ATP/Mg(2+)-dependent proteasome protease system. J Neural Transm 112, 547-555   DOI
26 Babu JR, Geetha T and Wooten MW (2005) Sequestosome 1/p62 shuttles polyubiquitinated tau for proteasomal degradation. J Neurochem 94, 192-203   DOI
27 de Vrij FM, Fischer DF, van Leeuwen FW and Hol EM (2004) Protein quality control in Alzheimer's disease by the ubiquitin proteasome system. Prog Neurobiol 74, 249-270   DOI
28 Lee MJ, Lee JH and Rubinsztein DC (2013) Tau degradation: the ubiquitin-proteasome system versus the autophagy-lysosome system. Prog Neurobiol 105, 49-59   DOI
29 Petrucelli L, Dickson D, Kehoe K et al (2004) CHIP and Hsp70 regulate tau ubiquitination, degradation and aggregation. Hum Mol Genet 13, 703-714   DOI
30 Flach K, Ramminger E, Hilbrich I et al (2014) Axotrophin/MARCH7 acts as an E3 ubiquitin ligase and ubiquitinates tau protein in vitro impairing microtubule binding. Biochim Biophys Acta 1842, 1527-1538   DOI
31 David DC, Layfield R, Serpell L, Narain Y, Goedert M and Spillantini MG (2002) Proteasomal degradation of tau protein. J Neurochem 83, 176-185   DOI
32 Grune T, Botzen D, Engels M et al (2010) Tau protein degradation is catalyzed by the ATP/ubiquitin-independent 20S proteasome under normal cell conditions. Arch Biochem Biophys 500, 181-188   DOI
33 Mori H, Kondo J and Ihara Y (1987) Ubiquitin is a component of paired helical filaments in Alzheimer's disease. Science 235, 1641-1644   DOI
34 Perry G, Friedman R, Shaw G and Chau V (1987) Ubiquitin is detected in neurofibrillary tangles and senile plaque neurites of Alzheimer disease brains. Proc Natl Acad Sci U S A 84, 3033-3036   DOI
35 Wille H, Drewes G, Biernat J, Mandelkow EM and Mandelkow E (1992) Alzheimer-like paired helical filaments and antiparallel dimers formed from microtubule-associated protein tau in vitro. J Cell Biol 118, 573-584   DOI
36 Cripps D, Thomas SN, Jeng Y, Yang F, Davies P and Yang AJ (2006) Alzheimer disease-specific conformation of hyperphosphorylated paired helical filament-Tau is polyubiquitinated through Lys-48, Lys-11, and Lys-6 ubiquitin conjugation. J Biol Chem 281, 10825-10838   DOI
37 Morishima-Kawashima M, Hasegawa M, Takio K, Suzuki M, Titani K and Ihara Y (1993) Ubiquitin is conjugated with amino-terminally processed tau in paired helical filaments. Neuron 10, 1151-1160   DOI
38 Paine S, Bedford L, Thorpe JR et al (2009) Immunoreactivity to Lys63-linked polyubiquitin is a feature of neurodegeneration. Neurosci Lett 460, 205-208   DOI
39 Tan JM, Wong ES, Kirkpatrick DS et al (2008) Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases. Hum Mol Genet 17, 431-439   DOI
40 Tan Z, Sun X, Hou FS et al (2007) Mutant ubiquitin found in Alzheimer's disease causes neuritic beading of mitochondria in association with neuronal degeneration. Cell Death Differ 14, 1721-1732   DOI
41 Fitzpatrick AWP, Falcon B, He S et al (2017) Cryo-EM structures of tau filaments from Alzheimer's disease. Nature 547, 185-190   DOI
42 Giannakopoulos P, Herrmann FR, Bussiere T et al (2003) Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer's disease. Neurology 60, 1495-1500   DOI
43 Ethell DW (2010) An amyloid-notch hypothesis for Alzheimer's disease. Neuroscientist 16, 614-617   DOI
44 Hung SY and Fu WM (2017) Drug candidates in clinical trials for Alzheimer's disease. J Biomed Sci 24, 47   DOI
45 Wang Y and Mandelkow E (2016) Tau in physiology and pathology. Nat Rev Neurosci 17, 5-21   DOI
46 Min SW, Cho SH, Zhou Y et al (2010) Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67, 953-966   DOI
47 Roth SY, Denu JM and Allis CD (2001) Histone acetyltransferases. Annu Rev Biochem 70, 81-120   DOI
48 Liu X, Wang L, Zhao K et al (2008) The structural basis of protein acetylation by the p300/CBP transcriptional coactivator. Nature 451, 846-850   DOI
49 Marmorstein R (2001) Structure of histone acetyltransferases. J Mol Biol 311, 433-444   DOI
50 Cohen TJ, Guo JL, Hurtado DE et al (2011) The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun 2, 252   DOI
51 Dickey CA, Kamal A, Lundgren K et al (2007) The high-affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins. J Clin Invest 117, 648-658   DOI
52 Kim JH, Kim E, Choi WH et al (2016) Inhibitory RNA Aptamers of Tau Oligomerization and Their Neuroprotective Roles against Proteotoxic Stress. Mol Pharm 13, 2039-2048   DOI
53 Karabiyik C, Lee MJ and Rubinsztein DC (2017) Autophagy impairment in Parkinson's disease. Essays Biochem 61, 711-720   DOI
54 Cohen TJ, Friedmann D, Hwang AW, Marmorstein R and Lee VM (2013) The microtubule-associated tau protein has intrinsic acetyltransferase activity. Nat Struct Mol Biol 20, 756-762   DOI
55 Dickey CA, Dunmore J, Lu B et al (2006) HSP induction mediates selective clearance of tau phosphorylated at proline-directed Ser/Thr sites but not KXGS (MARK) sites. FASEB J 20, 753-755   DOI
56 Liu WK, Ksiezak-Reding H and Yen SH (1991) Abnormal tau proteins from Alzheimer's disease brains. Purification and amino acid analysis. J Biol Chem 266, 21723-21727
57 King MK, Pardo M, Cheng Y, Downey K, Jope RS and Beurel E (2014) Glycogen synthase kinase-3 inhibitors: Rescuers of cognitive impairments. Pharmacol Ther 141, 1-12   DOI
58 Braak E, Braak H and Mandelkow EM (1994) A sequence of cytoskeleton changes related to the formation of neurofibrillary tangles and neuropil threads. Acta Neuropathol 87, 554-567   DOI
59 Lucas JJ, Hernandez F, Gomez-Ramos P, Moran MA, Hen R and Avila J (2001) Decreased nuclear beta-catenin, tau hyperphosphorylation and neurodegeneration in GSK-3beta conditional transgenic mice. EMBO J 20, 27-39   DOI
60 Forlenza OV, Aprahamian I, de Paula VJ and Hajek T (2016) Lithium, a Therapy for AD: Current Evidence from Clinical Trials of Neurodegenerative Disorders. Curr Alzheimer Res 13, 879-886   DOI
61 Abraha A, Ghoshal N, Gamblin TC et al (2000) C-terminal inhibition of tau assembly in vitro and in Alzheimer's disease. J Cell Sci 113 Pt 21, 3737-3745
62 Morris M, Knudsen GM, Maeda S et al (2015) Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci 18, 1183-1189   DOI
63 Alonso AD, Zaidi T, Novak M, Barra HS, Grundke-Iqbal I and Iqbal K (2001) Interaction of tau isoforms with Alzheimer's disease abnormally hyperphosphorylated tau and in vitro phosphorylation into the disease-like protein. J Biol Chem 276, 37967-37973
64 Wolfe MS (2009) Tau mutations in neurodegenerative diseases. J Biol Chem 284, 6021-6025   DOI
65 Flores-Rodriguez P, Ontiveros-Torres MA, Cardenas-Aguayo MC et al (2015) The relationship between truncation and phosphorylation at the C-terminus of tau protein in the paired helical filaments of Alzheimer's disease. Front Neurosci 9, 33
66 Peeraer E, Bottelbergs A, Van Kolen K et al (2015) Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice. Neurobiol Dis 73, 83-95   DOI
67 Guo JL and Lee VM (2013) Neurofibrillary tangle-like tau pathology induced by synthetic tau fibrils in primary neurons over-expressing mutant tau. FEBS Lett 587, 717-723   DOI
68 Noble W, Hanger DP, Miller CC and Lovestone S (2013) The importance of tau phosphorylation for neurodegenerative diseases. Front Neurol 4, 83
69 Matsunaga S, Kishi T, Annas P, Basun H, Hampel H and Iwata N (2015) Lithium as a Treatment for Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis 48, 403-410   DOI
70 Yu Y, Run X, Liang Z et al (2009) Developmental regulation of tau phosphorylation, tau kinases, and tau phosphatases. J Neurochem 108, 1480-1494   DOI
71 Sontag JM and Sontag E (2014) Protein phosphatase 2A dysfunction in Alzheimer's disease. Front Mol Neurosci 7, 16
72 Liu F, Grundke-Iqbal I, Iqbal K and Gong CX (2005) Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation. Eur J Neurosci 22, 1942-1950   DOI
73 Luo Y, Ma B, Nussinov R and Wei G (2014) Structural Insight into Tau Protein's Paradox of Intrinsically Disordered Behavior, Self-Acetylation Activity, and Aggregation. J Phys Chem Lett 5, 3026-3031   DOI
74 Guo JL and Lee VM (2011) Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J Biol Chem 286, 15317-15331   DOI
75 Nubling G, Bader B, Levin J, Hildebrandt J, Kretzschmar H and Giese A (2012) Synergistic influence of phosphorylation and metal ions on tau oligomer formation and coaggregation with alpha-synuclein at the single molecule level. Mol Neurodegener 7, 35   DOI
76 Gong CX, Shaikh S, Wang JZ, Zaidi T, Grundke-Iqbal I and Iqbal K (1995) Phosphatase activity toward abnormally phosphorylated tau: decrease in Alzheimer disease brain. J Neurochem 65, 732-738
77 Gorsky MK, Burnouf S, Dols J, Mandelkow E and Partridge L (2016) Acetylation mimic of lysine 280 exacerbates human Tau neurotoxicity in vivo. Sci Rep 6, 22685   DOI
78 Irwin DJ, Cohen TJ, Grossman M et al (2012) Acetylated tau, a novel pathological signature in Alzheimer's disease and other tauopathies. Brain 135, 807-818   DOI
79 Santa-Maria I, Hernandez F, Martin CP, Avila J and Moreno FJ (2004) Quinones facilitate the self-assembly of the phosphorylated tubulin binding region of tau into fibrillar polymers. Biochemistry 43, 2888-2897   DOI
80 Choi TS, Lee J, Han JY et al (2018) Supramolecular Modulation of Structural Polymorphism in Pathogenic alpha-Synuclein Fibrils Using Copper(II) Coordination. Angew Chem Int Ed Engl 57, 3099-3103   DOI
81 Goedert M, Spillantini MG, Jakes R, Rutherford D and Crowther RA (1989) Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease. Neuron 3, 519-526   DOI
82 Mandelkow EM and Mandelkow E (1998) Tau in Alzheimer's disease. Trends Cell Biol 8, 425-427   DOI
83 Andreadis A (2006) Misregulation of tau alternative splicing in neurodegeneration and dementia. Prog Mol Subcell Biol 44, 89-107
84 Goedert M, Cohen ES, Jakes R and Cohen P (1992) p42 MAP kinase phosphorylation sites in microtubule-associated protein tau are dephosphorylated by protein phosphatase 2A1. Implications for Alzheimer's disease [corrected]. FEBS Lett 312, 95-99   DOI
85 Kyte J and Doolittle RF (1982) A simple method for displaying the hydropathic character of a protein. J Mol Biol 157, 105-132   DOI
86 Kopke E, Tung YC, Shaikh S, Alonso AC, Iqbal K and Grundke-Iqbal I (1993) Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J Biol Chem 268, 24374-24384
87 Perez M, Hernandez F, Lim F, Diaz-Nido J and Avila J (2003) Chronic lithium treatment decreases mutant tau protein aggregation in a transgenic mouse model. J Alzheimers Dis 5, 301-308   DOI
88 Noble W, Planel E, Zehr C et al (2005) Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci U S A 102, 6990-6995   DOI
89 Avila J (2006) Tau phosphorylation and aggregation in Alzheimer's disease pathology. FEBS Lett 580, 2922-2927   DOI
90 Gong CX, Singh TJ, Grundke-Iqbal I and Iqbal K (1993) Phosphoprotein phosphatase activities in Alzheimer disease brain. J Neurochem 61, 921-927   DOI
91 Choi WH, de Poot SA, Lee JH et al (2016) Open-gate mutants of the mammalian proteasome show enhanced ubiquitin-conjugate degradation. Nat Commun 7, 10963   DOI
92 Han DH, Na HK, Choi WH et al (2014) Direct cellular delivery of human proteasomes to delay tau aggregation. Nat Commun 5, 5633   DOI
93 Jiang Y, Lee J, Lee JH et al (2016) The arginylation branch of the N-end rule pathway positively regulates cellular autophagic flux and clearance of proteotoxic proteins. Autophagy 12, 2197-2212   DOI