Comprehensive review on Alzheimer’s disease: From the posttranslational modifications of Tau to corresponding treatments

Xin Li , Zhisheng Ba , Juan Huang , Jianhua Chen , Jinyu Jiang , Nanqu Huang , Yong Luo

Ibrain ›› 2024, Vol. 10 ›› Issue (4) : 427 -438.

PDF
Ibrain ›› 2024, Vol. 10 ›› Issue (4) : 427 -438. DOI: 10.1002/ibra.12176
REVIEW

Comprehensive review on Alzheimer’s disease: From the posttranslational modifications of Tau to corresponding treatments

Author information +
History +
PDF

Abstract

Alzheimer’s disease (AD) is a neurodegenerative disease, which is mainly characterized by the abnormal deposition of β-amyloid peptide (Aβ) and Tau. Since Tau aggregation is more closely associated with synaptic loss, neurodegeneration, and cognitive decline than Aβ, the correlation between Tau and cognitive function in AD has gradually gained attention. The posttranslational modifications (PTMs) of Tau are key factors contributing to its pathological changes, which include phosphorylation, acetylation, ubiquitination, glycosylation, glycation, small ubiquitin-like modifier mediated modification (SUMOylation), methylation, succinylation, etc. These modifications change the structure of Tau, regulating Tau microtubule interactions, localization, degradation, and aggregation, thereby affecting its propensity to aggregate and leading to neuronal injury and cognitive impairments. Among numerous PTMs, drug development based on phosphorylation, acetylation, ubiquitination, and SUMOylation primarily involves enzymatic reactions, affecting either the phosphorylation or degradation processes of Tau. Meanwhile, methylation, glycosylation, and succinylation are associated with maintaining the structural stability of Tau. Current research is more extensive on phosphorylation, acetylation, ubiquitination, and methylation, with related drugs already developed, particularly focusing on phosphorylation and ubiquitination. In contrast, there is less research on SUMOylation, glycosylation, and succinylation, requiring further basic research, with the potential to become novel drug targets. In conclusion, this review summarized the latest research on PTMs of Tau and related drugs, highlighting the potential of targeting specific PTMs for developing novel therapeutic strategies in AD.

Keywords

Alzheimer’s disease / post-translational modifications / Tau / treatments

Cite this article

Download citation ▾
Xin Li, Zhisheng Ba, Juan Huang, Jianhua Chen, Jinyu Jiang, Nanqu Huang, Yong Luo. Comprehensive review on Alzheimer’s disease: From the posttranslational modifications of Tau to corresponding treatments. Ibrain, 2024, 10(4): 427-438 DOI:10.1002/ibra.12176

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Breijyeh Z, Karaman R. Comprehensive review on Alzheimer’s disease: causes and treatment. Molecules. 2020;25(24):5789.

[2]

Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer’s disease: challenges, successes and future. Signal Transduct Target Ther. 2023;8(1):248.

[3]

Arnsten AFT, Datta D, Del Tredici K, Braak H. Hypothesis: tau pathology is an initiating factor in sporadic Alzheimer’s disease. Alzheimer’s Dement. 2021;17(1):115-124.

[4]

Dai C, Tung YC, Liu F, Gong CX, Iqbal K. Tau passive immunization inhibits not only tau but also Aβ pathology. Alzheimer’s Res Ther. 2017;9(1):1.

[5]

Ittner A, Asih PR, Tan ARP, et al. Reduction of advanced tau-mediated memory deficits by the MAP kinase p38γ. Acta Neuropathol. 2020;140(3):279-294.

[6]

Tapia-Rojas C, Cabezas-Opazo F, Deaton CA, Vergara EH, Johnson GVW, Quintanilla RA. It’s all about tau. Prog Neurobiol. 2019;175:54-76.

[7]

Nelson PT, Alafuzoff I, Bigio EH, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012;71(5):362-381.

[8]

Naseri NN, Wang H, Guo J, Sharma M, Luo W. The complexity of tau in Alzheimer’s disease. Neurosci Lett. 2019;705:183-194.

[9]

Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: a pathological or physiological phenomenon? Acta Neuropathol Commun. 2021;9(1):149.

[10]

Luo Y, Ma B, Nussinov R, Wei G. Structural insight into tau protein’s paradox of intrinsically disordered behavior, self-acetylation activity, and aggregation. J Phys Chem Lett. 2014;5(17):3026-3031.

[11]

Alquezar C, Arya S, Kao AW. Tau post-translational modifications: dynamic transformers of tau function, degradation, and aggregation. Front Neurol. 2020;11:595532.

[12]

Barber KW, Rinehart J. The ABCs of PTMs. Nat Chem Biol. 2018;14(3):188-192.

[13]

Uversky VN. Intrinsically disordered proteins and their (disordered) proteomes in neurodegenerative disorders. Front Aging Neurosci. 2015;7:18.

[14]

Wesseling H, Mair W, Kumar M, et al. Tau PTM profiles identify patient heterogeneity and stages of Alzheimer’s disease. Cell. 2020;183(6):1699-1713.e13.

[15]

Trushina NI, Bakota L, Mulkidjanian AY, Brandt R. The evolution of tau phosphorylation and interactions. Front Aging Neurosci. 2019;11:256.

[16]

Köpke E, Tung YC, Shaikh S, Alonso AC, Iqbal K, Grundke-Iqbal I. Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J Biol Chem. 1993;268(32):24374-24384.

[17]

Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol. 2017;133(5):665-704.

[18]

Stefanoska K, Gajwani M, Tan ARP, et al. Alzheimer’s disease: ablating single master site abolishes tau hyperphosphorylation. Sci Adv. 2022;8(27):eabl8809.

[19]

Man VH, He X, Gao J, Wang J. Phosphorylation of tau R2 repeat destabilizes its binding to microtubules: a molecular dynamics simulation study. ACS Chem Neurosci. 2023;14(3):458-467.

[20]

Barthélemy NR, Li Y, Joseph-Mathurin N, et al. A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nature Med. 2020;26(3):398-407.

[21]

Suárez-Calvet M, Karikari TK, Ashton NJ, et al. Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Aβ pathology are detected. EMBO Mol Med. 2020;12(12):e12921.

[22]

Janelidze S, Stomrud E, Smith R, et al. Cerebrospinal fluid p-tau217 performs better than p-tau181 as a biomarker of Alzheimer’s disease. Nat Commun. 2020;11(1):1683.

[23]

Moscoso A, Karikari TK, Grothe MJ, et al. CSF biomarkers and plasma p-tau181 as predictors of longitudinal tau accumulation: implications for clinical trial design. Alzheimer’s Dement. 2022;18(12):2614-2626.

[24]

Alcolea D, Beeri MS, Rojas JC, Gardner RC, Lleó A. Blood biomarkers in neurodegenerative diseases: implications for the clinical neurologist. Neurology. 2023;101(4):172-180.

[25]

Avila J. Tau kinases and phosphatases. J Cell Mol Med. 2008;12(1):258-259.

[26]

Hetman M, Cavanaugh JE, Kimelman D, Xia Z. Role of glycogen synthase Kinase-3β in neuronal apoptosis induced by trophic withdrawal. J Neurosci. 2000;20(7):2567-2574.

[27]

Domínguez JM, Fuertes A, Orozco L, del Monte-Millán M, Delgado E, Medina M. Evidence for irreversible inhibition of glycogen synthase kinase-3β by tideglusib. J Biol Chem. 2012;287(2):893-904.

[28]

Lovestone S, Boada M, Dubois B, et al. A phase II trial of tideglusib in Alzheimer’s disease. J Alzheimer’s Dis. 2015;45(1):75-88.

[29]

Terao I, Honyashiki M, Inoue T. Comparative efficacy of lithium and aducanumab for cognitive decline in patients with mild cognitive impairment or Alzheimer’s disease: a systematic review and network meta-analysis. Ageing Res Rev. 2022;81:101709.

[30]

Matsunaga S, Kishi T, Annas P, Basun H, Hampel H, Iwata N. Lithium as a treatment for Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimer’s Dis. 2015;48(2):403-410.

[31]

Lahmy V, Meunier J, Malmström S, et al. Blockade of tau hyperphosphorylation and Aβ 1–42 generation by the aminotetrahydrofuran derivative ANAVEX2-73, a mixed muscarinic and σ1 receptor agonist, in a nontransgenic mouse model of Alzheimer’s disease. Neuropsychopharmacology. 2013;38(9):1706-1723.

[32]

Li X. Effects of gensenoside Rg1 on tau protein phosphorylation induced by okadaic acid in rat brain slices. J Chin Integr Med. 2010;8(10):955-960.

[33]

Zhang Y, Zhang Z, Wang H, et al. Neuroprotective effect of ginsenoside Rg1 prevents cognitive impairment induced by isoflurane anesthesia in aged rats via antioxidant, anti-inflammatory and anti-apoptotic effects mediated by the PI3K/AKT/GSK-3β pathway. Mol Med Rep. 2016;14(3):2778-2784.

[34]

Ni YN, Wang YM, Kong L, et al. Effect of osthole on tau hyperphosphorylation and PI3K/Akt/Gsk3β signaling pathway in the brain of AD mice. Chin New Drugs J. 2019;28(23):2865-2871.

[35]

Liu F, Grundke-Iqbal I, Iqbal K, Gong CX. Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation. Eur J Neurosci. 2005;22(8):1942-1950.

[36]

Cardoso BR, Roberts BR, Malpas CB, et al. Supranutritional sodium selenate supplementation delivers selenium to the central nervous system: results from a randomized controlled pilot trial in Alzheimer’s disease. Neurotherapeutics. 2019;16(1):192-202.

[37]

Eteläinen TS, Silva MC, Uhari-Väänänen JK, et al. A prolyl oligopeptidase inhibitor reduces tau pathology in cellular models and in mice with tauopathy. Sci Transl Med. 2023;15(691):eabq2915.

[38]

Reisberg B, Doody R, Stöffler A, Schmitt F, Ferris S, Möbius HJ. Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med. 2003;348(14):1333-1341.

[39]

St-Cyr Giguère F, Attiori Essis S, Chagniel L, Germain M, Cyr M, Massicotte G. The sphingosine-1-phosphate receptor 1 agonist SEW2871 reduces Tau-Ser262 phosphorylation in rat hippocampal slices. Brain Res. 2017;1658:51-59.

[40]

Wang HY, Pei Z, Lee KC, et al. PTI-125 reduces biomarkers of Alzheimer’s disease in patients. J Prev Alzheimers Dis. 2020;7(4):256-264.

[41]

Sun X, Li L, Dong QX, et al. Rutin prevents tau pathology and neuroinflammation in a mouse model of Alzheimer’s disease. J Neuroinflammation. 2021;18(1):131.

[42]

Shen Q, Luo HB, Xie WZ. Banqiao Codonopsis Pilosula improves cognitive dysfunction in AD model rats by PP2A signaling pathway. Chin Pharmacol Bull. 2019;35:1232-1239.

[43]

Haj-Yahya M, Lashuel HA. Protein semisynthesis provides access to tau disease-associated post-translational modifications (PTMs) and paves the way to deciphering the tau PTM code in health and diseased states. J Am Chem Soc. 2018;140(21):6611-6621.

[44]

Zhou Q, Li S, Li M, et al. Human tau accumulation promotes glycogen synthase kinase-3βacetylation and thus upregulates the kinase: a vicious cycle in Alzheimer neurodegeneration. EBioMedicine. 2022;78:103970.

[45]

Liu Q, Wang X, Hu Y, et al. Acetylated tau exacerbates learning and memory impairment by disturbing with mitochondrial homeostasis. Redox Biol. 2023;62:102697.

[46]

Carlomagno Y, Chung DC, Yue M, et al. An acetylation-phosphorylation switch that regulates tau aggregation propensity and function. J Biol Chem. 2017;292(37):15277-15286.

[47]

Min SW, Chen X, Tracy TE, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nature Med. 2015;21(10):1154-1162.

[48]

Zhang L, Liu C, Wu J, et al. Tubastatin A/ACY-1215 improves cognition in Alzheimer’s disease transgenic Mice1. J Alzheimer’s Dis. 2014;41(4):1193-1205.

[49]

Choi H, Kim HJ, Yang J, et al. Acetylation changes tau interactome to degrade tau in Alzheimer’s disease animal and organoid models. Aging cell. 2020;19(1):e13081.

[50]

Abreha MH, Dammer EB, Ping L, et al. Quantitative analysis of the brain ubiquitylome in Alzheimer’s disease. Proteomics. 2018;18(20):e1800108.

[51]

Yan Y, Wang X, Chaput D, et al. X-linked ubiquitin-specific peptidase 11 increases tauopathy vulnerability in women. Cell. 2022;185(21):3913-3930.e19.

[52]

Arakhamia T, Lee CE, Carlomagno Y, et al. Posttranslational modifications mediate the structural diversity of tauopathy strains. Cell. 2020;180(4):633-644.e12.

[53]

Kim JH, Lee J, Choi WH, et al. CHIP-mediated hyperubiquitylation of tau promotes its self-assembly into the insoluble tau filaments. Chem Sci. 2021;12(15):5599-5610.

[54]

Myeku N, Clelland CL, Emrani S, et al. Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nature Med. 2016;22(1):46-53.

[55]

Puangmalai N, Sengupta U, Bhatt N, et al. Lysine 63-linked ubiquitination of tau oligomers contributes to the pathogenesis of Alzheimer’s disease. J Biol Chem. 2022;298(4):101766.

[56]

Wang P, Joberty G, Buist A, et al. Tau interactome mapping based identification of Otub1 as tau deubiquitinase involved in accumulation of pathological tau forms in vitro and in vivo. Acta Neuropathol. 2017;133(5):731-749.

[57]

Chu TT, Gao N, Li QQ, et al. Specific knockdown of endogenous tau protein by peptide-directed ubiquitin-proteasome degradation. Cell Chemical Biology. 2016;23(4):453-461.

[58]

Wang W, Zhou Q, Jiang T, et al. A novel small-molecule PROTAC selectively promotes tau clearance to improve cognitive functions in Alzheimer-like models. Theranostics. 2021;11(11):5279-5295.

[59]

Lu M, Liu T, Jiao Q, et al. Discovery of a Keap1-dependent peptide PROTAC to knockdown tau by ubiquitination-proteasome degradation pathway. Eur J Med Chem. 2018;146:251-259.

[60]

Huseby CJ, Hoffman CN, Cooper GL, et al. Quantification of tau protein lysine methylation in aging and Alzheimer’s disease. J Alzheimer’s Dis. 2019;71(3):979-991.

[61]

Bichmann M, Prat Oriol N, Ercan-Herbst E, et al. SETD7-mediated monomethylation is enriched on soluble tau in Alzheimer’s disease. Mol Neurodegener. 2021;16(1):46.

[62]

Cavallaro RA, Nicolia V, Fiorenza MT, Scarpa S, Fuso A. S-Adenosylmethionine and superoxide dismutase 1 synergistically counteract Alzheimer’s disease features progression in TgCRND8 mice. Antioxidants (Basel, Switzerland). 2017;6(4):76.

[63]

Balmik AA, Chinnathambi S. Methylation as a key regulator of tau aggregation and neuronal health in Alzheimer’s disease. Cell Commun Signaling. 2021;19(1):51.

[64]

Shams H, Matsunaga A, Ma Q, Mofrad MRK, Didonna A. Methylation at a conserved lysine residue modulates tau assembly and cellular functions. Mol Cell Neurosci. 2022;120:103707.

[65]

Berletch JB, Liu C, Love WK, Andrews LG, Katiyar SK, Tollefsbol TO. Epigenetic and genetic mechanisms contribute to telomerase inhibition by EGCG. J Cell Biochem. 2008;103(2):509-519.

[66]

Kato K, Long NK, Makita H, et al. Effects of green tea polyphenol on methylation status of RECK gene and cancer cell invasion in oral squamous cell carcinoma cells. Br J Cancer. 2008;99(4):647-654.

[67]

Mukherjee N, Kumar AP, Ghosh R. DNA methylation and flavonoids in genitourinary cancers. Curr Pharmacol Rep. 2015;1(2):112-120.

[68]

Zhou XW, Gustafsson , Tanila H, et al. Tau hyperphosphorylation correlates with reduced methylation of protein phosphatase 2A. Neurobiol Dis. 2008;31(3):386-394.

[69]

Zheng M, Zou C, Li M, Huang G, Gao Y, Liu H. Folic acid reduces tau phosphorylation by regulating PP2A methylation in Streptozotocin-induced diabetic mice. Int J Mol Sci. 2017;18(4):861.

[70]

Fontecave M, Atta M, Mulliez E. S-adenosylmethionine: nothing goes to waste. Trends Biochem Sci. 2004;29(5):243-249.

[71]

Matsuzaki S, Lee L, Knock E, et al. SUMO1 affects synaptic function, spine density and memory. Sci Rep. 2015;5:10730.

[72]

Luo HB, Xia YY, Shu XJ, et al. SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(46):16586-16591.

[73]

Mandel N, Agarwal N. Role of SUMOylation in neurodegenerative diseases. Cells. 2022;11(21):3395.

[74]

Qin M, Li H, Bao J, et al. SET SUMOylation promotes its cytoplasmic retention and induces tau pathology and cognitive impairments. Acta Neuropathol Commun. 2019;7(1):21.

[75]

Lee EJ, Hyun SH, Chun J, et al. Regulation of glycogen synthase kinase 3 functions by modification of the small ubiquitin-like modifier. Open Biochem J. 2008;2:67-76.

[76]

Gatta E, Lefebvre T, Gaetani S, et al. Evidence for an imbalance between tau O-GlcNAcylation and phosphorylation in the hippocampus of a mouse model of Alzheimer’s disease. Pharmacol Res. 2016;105:186-197.

[77]

Lim S, Haque M, Nam G, Ryoo N, Rhim H, Kim Y. Monitoring of intracellular tau aggregation regulated by OGA/OGT inhibitors. Int J Mol Sci. 2015;16(9):20212-20224.

[78]

Lu S, Yin X, Wang J, et al. SIRT1 regulates O-GlcNAcylation of tau through OGT. Aging. 2020;12(8):7042-7055.

[79]

Losev Y, Paul A, Frenkel-Pinter M, et al. Novel model of secreted human tau protein reveals the impact of the abnormal N-glycosylation of tau on its aggregation propensity. Sci Rep. 2019;9(1):2254.

[80]

Losev Y, Frenkel-Pinter M, Abu-Hussien M, et al. Differential effects of putative N-glycosylation sites in human tau on Alzheimer’s disease-related neurodegeneration. Cell Mol Life Sci. 2021;78(5):2231-2245.

[81]

Haukedal H, Freude KK. Implications of glycosylation in Alzheimer’s disease. Front Neurosci. 2021;14:625348.

[82]

Yuzwa SA, Macauley MS, Heinonen JE, et al. A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat Chem Biol. 2008;4(8):483-490.

[83]

Hastings NB, Wang X, Song L, et al. Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice. Mol Neurodegener. 2017;12(1):39.

[84]

Zhang Z, Tan M, Xie Z, Dai L, Chen Y, Zhao Y. Identification of lysine succinylation as a new post-translational modification. Nat Chem Biol. 2011;7(1):58-63.

[85]

von Bergen M, Friedhoff P, Biernat J, Heberle J, Mandelkow EM, Mandelkow E. Assembly of τ protein into Alzheimer paired helical filaments depends on a local sequence motif (306VQIVYK311) forming β structure. Proc Natl Acad Sci. 2000;97(10):5129-5134.

[86]

Yang Y, Tapias V, Acosta D, et al. Altered succinylation of mitochondrial proteins, APP and tau in Alzheimer’s disease. Nat Commun. 2022;13(1):159.

[87]

Yang Y, Gibson GE. Succinylation links metabolism to protein functions. Neurochem Res. 2019;44(10):2346-2359.

RIGHTS & PERMISSIONS

2024 The Author(s). Ibrain published by Affiliated Hospital of Zunyi Medical University (AHZMU) and Wiley-VCH GmbH.

AI Summary AI Mindmap
PDF

561

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/