Tau induces inflammasome activation and microgliosis through acetylating NLRP3

Lun Zhang , Yongkang Gai , Yushuang Liu , Dongli Meng , Yi Zeng , Yong Luo , Huiliang Zhang , Zhuoqun Wang , Mengzhe Yang , Yunfan Li , Yi Liu , Yiwen Lai , Jiayu Yang , Gang Wu , Yu Chen , Jingtan Zhu , Shaojun Liu , Tingting Yu , Ji Zeng , Jianzhi Wang , Dan Zhu , Xiaochuan Wang , Xiaoli Lan , Rong Liu

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1623

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1623 DOI: 10.1002/ctm2.1623
RESEARCH ARTICLE

Tau induces inflammasome activation and microgliosis through acetylating NLRP3

Author information +
History +
PDF

Abstract

Background: Alzheimer's disease (AD) and related Tauopathies are characterised by the pathologically hyperphosphorylated and aggregated microtubule-associated protein Tau, which is accompanied by neuroinflammation mediated by activated microglia. However, the role of Tau pathology in microglia activation or their causal relationship remains largely elusive.

Methods: The levels of nucleotide-binding oligomerisation domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) acetylation and inflammasome activation in multiple cell models with Tau proteins treatment, transgenic mice with Tauopathy, and AD patients were measured by Western blotting and enzyme-linked immunosorbent assay. In addition, the acetyltransferase activity of Tau and NLRP3 acetylation sites were confirmed using the test-tube acetylation assay, co-immunoprecipitation, immunofluorescence (IF) staining, mass spectrometry and molecular docking. The Tau-overexpressing mouse model was established by overexpression of human Tau proteins in mouse hippocampal CA1 neurons through the adeno-associated virus injection. The cognitive functions of Tau-overexpressing mice were assessed in various behavioural tests, and microglia activation was analysed by Iba-1 IF staining and [18F]-DPA-714 positron emission tomography/computed tomography imaging. A peptide that blocks the interaction between Tau and NLRP3 was synthesised to determine the in vitro and in vivo effects of Tau–NLRP3 interaction blockade on NLRP3 acetylation, inflammasome activation, microglia activation and cognitive function.

Results: Excessively elevated NLRP3 acetylation and inflammasome activation were observed in 3xTg-AD mice, microtubule-associated protein Tau P301S (PS19) mice and AD patients. It was further confirmed that mimics of ‘early’ phosphorylated-Tau proteins which increase at the initial stage of diseases with Tauopathy, including TauT181E, TauS199E, TauT217E and TauS262E, significantly promoted Tau–K18 domain acetyltransferase activity-dependent NLRP3 acetylation and inflammasome activation in HEK293T and BV-2 microglial cells. In addition, Tau protein could directly acetylate NLRP3 at the K21, K22 and K24 sites at its PYD domain and thereby induce inflammasome activation in vitro. Overexpression of human Tau proteins in mouse hippocampal CA1 neurons resulted in impaired cognitive function, Tau transmission to microglia and microgliosis with NLRP3 acetylation and inflammasome activation. As a targeted intervention, competitive binding of a designed Tau–NLRP3-binding blocking (TNB) peptide to block the interaction of Tau protein with NLRP3 inhibited the NLRP3 acetylation and downstream inflammasome activation in microglia, thereby alleviating microglia activation and cognitive impairment in mice.

Conclusions: In conclusion, our findings provide evidence for a novel role of Tau in the regulation of microglia activation through acetylating NLRP3, which has potential implications for early intervention and personalised treatment of AD and related Tauopathies.

Keywords

acetylation / Alzheimer's disease / microglia / NLRP3 inflammasome / Tauopathies

Cite this article

Download citation ▾
Lun Zhang, Yongkang Gai, Yushuang Liu, Dongli Meng, Yi Zeng, Yong Luo, Huiliang Zhang, Zhuoqun Wang, Mengzhe Yang, Yunfan Li, Yi Liu, Yiwen Lai, Jiayu Yang, Gang Wu, Yu Chen, Jingtan Zhu, Shaojun Liu, Tingting Yu, Ji Zeng, Jianzhi Wang, Dan Zhu, Xiaochuan Wang, Xiaoli Lan, Rong Liu. Tau induces inflammasome activation and microgliosis through acetylating NLRP3. Clinical and Translational Medicine, 2024, 14(3): e1623 DOI:10.1002/ctm2.1623

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci. 2016;17:22-35.

[2]

Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu Rev Neurosci. 2001;24:1121-1159.

[3]

Alonso AC, Zaidi T, Grundke-Iqbal I, Iqbal K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc Natl Acad Sci U S A. 1994;91:5562-5566.

[4]

Hoover BR, Reed MN, Su J, et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron. 2010;68:1067-1081.

[5]

Brundin P, Melki R, Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol. 2010;11:301-307.

[6]

Greenhalgh AD, David S, Bennett FC. Immune cell regulation of glia during CNS injury and disease. Nat Rev Neurosci. 2020;21:139-152.

[7]

Linnerbauer M, Wheeler MA, Quintana FJ. Astrocyte crosstalk in CNS inflammation. Neuron. 2020;108:608-622.

[8]

Patel DC, Tewari BP, Chaunsali L, Sontheimer H. Neuron–glia interactions in the pathophysiology of epilepsy. Nat Rev Neurosci. 2019;20:282-297.

[9]

Richetin K, Steullet P, Pachoud M, et al. Tau accumulation in astrocytes of the dentate gyrus induces neuronal dysfunction and memory deficits in Alzheimer's disease. Nat Neurosci. 2020;23:1567-1579.

[10]

Hu X, Yu G, Liao X, Xiao L. Interactions between astrocytes and oligodendroglia in myelin development and related brain diseases. Neurosci Bull. 2023;39:541-552.

[11]

Lloyd A, Miron V. The pro-remyelination properties of microglia in the central nervous system. Nat Rev Neurol. 2019;15:447-458.

[12]

Halle A, Hornung V, Petzold GC, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-β. Nat Immunol. 2008;9:857-865.

[13]

Bamberger ME, Harris ME, McDonald DR, Husemann J, Landreth GE. A cell surface receptor complex for fibrillar β-amyloid mediates microglial activation. J Neurosci. 2003;23:2665-2674.

[14]

Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674-678.

[15]

Gerhard A, Trender-Gerhard I, Turkheimer F, Quinn NP, Bhatia KP, Brooks DJ. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in progressive supranuclear palsy. Movement Disorders. 2006;21:89-93.

[16]

Ishizawa K, Dickson DW. Microglial activation parallels system degeneration in progressive supranuclear palsy and corticobasal degeneration. J Neuropathol Exp Neurol. 2001;60:647-657.

[17]

Terada T, Yokokura M, Obi T, et al. In vivo direct relation of tau pathology with neuroinflammation in early Alzheimer's disease. J Neurol. 2019;266:2186-2196.

[18]

Bolós M, Llorens-Martín M, Jurado-Arjona J, Hernández F, Rábano A, Avila J. Direct evidence of internalization of tau by microglia in vitro and in vivo. J Alzheimer's Disease. 2016;50:77-87.

[19]

Stancu I-C, Cremers N, Vanrusselt H, et al. Aggregated Tau activates NLRP3–ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol. 2019;137:599-617.

[20]

Brelstaff JH, Mason M, Katsinelos T, et al. Microglia become hypofunctional and release metalloproteases and tau seeds when phagocytosing live neurons with P301S tau aggregates. Sci Adv. 2021;7:eabg4980.

[21]

Wang Q, Xue X, Huang Z, Wang Y. Microglia share the burden. Neurosci Bull. 2022;38:695-698.

[22]

Bellucci A, Bugiani O, Ghetti B, Spillantini MG. Presence of reactive microglia and neuroinflammatory mediators in a case of frontotemporal dementia with P301S mutation. Neurodegenerative Diseases. 2011;8:221-229.

[23]

Dani M, Wood M, Mizoguchi R, et al. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer's disease. Brain. 2018;141:2740-2754.

[24]

Maphis N, Xu G, Kokiko-Cochran ON, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738-1755.

[25]

Asai H, Ikezu S, Tsunoda S, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18:1584-1593.

[26]

Ising C, Venegas C, Zhang S, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669-673.

[27]

Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17:157-172.

[28]

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. Nat Med. 2020;26:398-407.

[29]

Yang C-C, Chiu M-J, Chen T-F, Chang H-L, Liu B-H, Yang S-Y. Assay of plasma phosphorylated tau protein (threonine 181) and total tau protein in early-stage Alzheimer's disease. J Alzheimer's Disease. 2018;61:1323-1332.

[30]

Janelidze S, Berron D, Smith R, et al. Associations of plasma phospho-Tau217 levels with tau positron emission tomography in early Alzheimer disease. JAMA Neurol. 2021;78:149-156.

[31]

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

[32]

Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397-411.

[33]

Walsh JG, Muruve DA, Power C. Inflammasomes in the CNS. Nat Rev Neurosci. 2014;15:84-97.

[34]

Heneka MT, McManus RM, Latz E. Inflammasome signalling in brain function and neurodegenerative disease. Nat Rev Neurosci. 2018;19:610-621.

[35]

He W, Hu Z, Zhong Y, Wu C, Li J. The potential of NLRP3 inflammasome as a therapeutic target in neurological diseases. Mol Neurobiol. 2023;60:2520-2538.

[36]

Johnson AM, Lukens JR. The innate immune response in tauopathies. Eur J Immunol. 2023;53:2250266.

[37]

Odfalk KF, Bieniek KF, Hopp SC. Microglia: friend and foe in tauopathy. Prog Neurobiol. 2022;216:102306.

[38]

He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41:1012-1021.

[39]

Paik S, Kim JK, Silwal P, Sasakawa C, Jo E-K. An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol Immunol. 2021;18:1141-1160.

[40]

Xia J, Jiang S, Dong S, Liao Y, Zhou Y. The role of post-translational modifications in regulation of NLRP3 inflammasome activation. Int J Mol Sci. 2023;24:6126.

[41]

He M, Chiang H-H, Luo H, et al. An acetylation switch of the NLRP3 inflammasome regulates aging-associated chronic inflammation and insulin resistance. Cell Metab. 2020;31:580-591.e5.

[42]

Zhao K, Zhang Y, Xu X, et al. Acetylation is required for NLRP3 self-aggregation and full activation of the inflammasome . BioRXiv. 2019. 2019.12.31.891556.

[43]

Cohen TJ, Friedmann D, Hwang AW, Marmorstein R, Lee VM. The microtubule-associated tau protein has intrinsic acetyltransferase activity. Nat Struct Mol Biol. 2013;20:756-762.

[44]

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

[45]

Xie JZ, Zhang Y, Li SH, et al. P301S-hTau acetylates KEAP1 to trigger synaptic toxicity via inhibiting NRF2/ARE pathway: a novel mechanism underlying hTau-induced synaptic toxicities. Clin Transl Med. 2022;12:e1003.

[46]

Liu E, Zhou Q, Xie AJ, et al. Tau acetylates and stabilizes β-catenin thereby promoting cell survival. EMBO Rep. 2020;21:e48328.

[47]

Huiliang Z, Mengzhe Y, Xiaochuan W, et al. Zinc induces reactive astrogliosis through ERK-dependent activation of Stat3 and promotes synaptic degeneration. J Neurochem. 2021;159:1016-1027.

[48]

Hu WT, Liuyang ZY, Tian Y, et al. CIP2A deficiency promotes depression-like behaviors in mice through inhibition of dendritic arborization. EMBO Rep. 2022;23:e54911.

[49]

Chen J, Chen ZJ. PtdIns4P on dispersed trans-Golgi network mediates NLRP3 inflammasome activation. Nature. 2018;564:71-76.

[50]

Shi H, Murray A, Beutler B. Reconstruction of the mouse inflammasome system in HEK293T cells. Bio-Protocol. 2016;6:e1986.

[51]

Meng D, Yang M, Hu L, et al. Rifaximin protects against circadian rhythm disruption-induced cognitive impairment through preventing gut barrier damage and neuroinflammation. J Neurochem. 2022;163:406-418.

[52]

Kozakov D, Hall DR, Xia B, et al. The ClusPro web server for protein–protein docking. Nat Protoc. 2017;12:255-278.

[53]

Lépinoux-Chambaud C, Barreau K, Eyer J. The neurofilament-derived peptide NFL-TBS. 40–63 targets neural stem cells and affects their properties. Stem Cells Transl Med. 2016;5:901-913.

[54]

Zhou Y, Liu X, Ma S, et al. ChK1 activation induces reactive astrogliosis through CIP2A/PP2A/STAT3 pathway in Alzheimer's disease. FASEB J. 2022;36:e22209.

[55]

Hu W, Wang Z, Zhang H, et al. Chk1 inhibition ameliorates Alzheimer's disease pathogenesis and cognitive dysfunction through CIP2A/PP2A signaling. Neurotherapeutics. 2022;19:570-591.

[56]

Rodríguez-Chinchilla T, Quiroga-Varela A, Molinet-Dronda F, et al. [18F]-DPA-714 PET as a specific in vivo marker of early microglial activation in a rat model of progressive dopaminergic degeneration. Eur J Nucl Med Mol Imaging. 2020;47:2602-2612.

[57]

Oddo S, Caccamo A, Shepherd JD, et al. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron. 2003;39:409-421.

[58]

Pampuscenko K, Morkuniene R, Krasauskas L, Smirnovas V, Tomita T, Borutaite V. Distinct neurotoxic effects of extracellular tau species in primary neuronal-glial cultures. Mol Neurobiol. 2021;58:658-667.

[59]

Pampuscenko K, Morkuniene R, Sneideris T, et al. Extracellular tau induces microglial phagocytosis of living neurons in cell cultures. J Neurochem. 2020;154:316-329.

[60]

Furman JL, Vaquer-Alicea J, White CL, Cairns NJ, Nelson PT, Diamond MI. Widespread tau seeding activity at early Braak stages. Acta Neuropathol. 2017;133:91-100.

[61]

Wang C, Fan L, Khawaja RR, et al. Microglial NF-κB drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun. 2022;13:1969.

[62]

Gratuze M, Chen Y, Parhizkar S, et al. Activated microglia mitigate Aβ-associated tau seeding and spreading. J Exp Med. 2021;218:e20210542.

[63]

Liu GJ, Middleton RJ, Hatty CR, et al. The 18 kDa translocator protein, microglia and neuroinflammation. Brain Pathol. 2014;24:631-653.

[64]

Peyronneau M, Kuhnast B, Nguyen D, et al. [18F] DPA-714: effect of co-medications, age, sex, BMI and TSPO polymorphism on the human plasma input function. Eur J Nucl Med Mol Imaging. 2023;50:3251-3264.

[65]

Zhao X, Li S, Gaur U, Zheng W. Artemisinin improved neuronal functions in Alzheimer's disease animal model 3xtg mice and neuronal cells via stimulating the ERK/CREB signaling pathway. Aging Disease. 2020;11:801.

[66]

Jiang S, Maphis NM, Binder J, et al. Proteopathic tau primes and activates interleukin-1β via myeloid-cell-specific MyD88-and NLRP3-ASC-inflammasome pathway. Cell Rep. 2021;36:109720.

[67]

Morales I, Jiménez JM, Mancilla M, Maccioni RB. Tau oligomers and fibrils induce activation of microglial cells. J Alzheimer's Disease. 2013;37:849-856.

[68]

Van Zeller M, Dias D, Sebastiao AM, Valente CA. NLRP3 inflammasome: a starring role in amyloid-β-and tau-driven pathological events in Alzheimer's disease. J Alzheimer's Disease. 2021;83:939-961.

[69]

Hanslik KL, Ulland TK. The role of microglia and the Nlrp3 inflammasome in Alzheimer's disease. Front Neurol. 2020;11:570711.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

280

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/