Skip to main content

Advertisement

Log in

ApoE4 inhibition of VMAT2 in the locus coeruleus exacerbates Tau pathology in Alzheimer’s disease

  • Original Paper
  • Published:
Acta Neuropathologica Aims and scope Submit manuscript

Abstract

ApoE4 enhances Tau neurotoxicity and promotes the early onset of AD. Pretangle Tau in the noradrenergic locus coeruleus (LC) is the earliest detectable AD-like pathology in the human brain. However, a direct relationship between ApoE4 and Tau in the LC has not been identified. Here we show that ApoE4 selectively binds to the vesicular monoamine transporter 2 (VMAT2) and inhibits neurotransmitter uptake. The exclusion of norepinephrine (NE) from synaptic vesicles leads to its oxidation into the toxic metabolite 3,4-dihydroxyphenyl glycolaldehyde (DOPEGAL), which subsequently activates cleavage of Tau at N368 by asparagine endopeptidase (AEP) and triggers LC neurodegeneration. Our data reveal that ApoE4 boosts Tau neurotoxicity via VMAT2 inhibition, reduces hippocampal volume, and induces cognitive dysfunction in an AEP- and Tau N368-dependent manner, while conversely ApoE3 binds Tau and protects it from cleavage. Thus, ApoE4 exacerbates Tau neurotoxicity by increasing VMAT2 vesicle leakage and facilitating AEP-mediated Tau proteolytic cleavage in the LC via DOPEGAL.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Agosta F, Vossel KA, Miller BL, Migliaccio R, Bonasera SJ, Filippi M, Boxer AL, Karydas A, Possin KL, Gorno-Tempini ML (2009) Apolipoprotein E epsilon4 is associated with disease-specific effects on brain atrophy in Alzheimer’s disease and frontotemporal dementia. Proc Natl Acad Sci USA 106:2018–2022. https://doi.org/10.1073/pnas.0812697106

    Article  PubMed  PubMed Central  Google Scholar 

  2. Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT (1992) Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 42:631–639. https://doi.org/10.1212/wnl.42.3.631

    Article  CAS  PubMed  Google Scholar 

  3. Bales KR, Verina T, Dodel RC, Du Y, Altstiel L, Bender M, Hyslop P, Johnstone EM, Little SP, Cummins DJ et al (1997) Lack of apolipoprotein E dramatically reduces amyloid β-peptide deposition. Nat Genet 17:263–264. https://doi.org/10.1038/ng1197-263

    Article  CAS  PubMed  Google Scholar 

  4. Bernstein AI, Stout KA, Miller GW (2014) The vesicular monoamine transporter 2: an underexplored pharmacological target. Neurochem Int 73:89–97. https://doi.org/10.1016/j.neuint.2013.12.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Braak H, Thal DR, Ghebremedhin E, Del Tredici K (2011) Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol 70:960–969. https://doi.org/10.1097/NEN.0b013e318232a379

    Article  CAS  PubMed  Google Scholar 

  6. Braak H, Del Tredici K (2011) The pathological process underlying Alzheimer’s disease in individuals under thirty. Acta Neuropathol 121:171–181. https://doi.org/10.1007/s00401-010-0789-4

    Article  PubMed  Google Scholar 

  7. Braak H, Del Tredici K (2012) Where, when, and in what form does sporadic Alzheimer’s disease begin? Curr Opin Neurol 25:708–714. https://doi.org/10.1097/WCO.0b013e32835a3432

    Article  CAS  PubMed  Google Scholar 

  8. Brecht WJ, Harris FM, Chang S, Tesseur I, Yu GQ, Xu Q, Dee Fish J, Wyss-Coray T, Buttini M, Mucke L et al (2004) Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice. J Neurosci 24:2527–2534. https://doi.org/10.1523/JNEUROSCI.4315-03.2004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Burke WJ, Kristal BS, Yu BP, Li SW, Lin TS (1998) Norepinephrine transmitter metabolite generates free radicals and activates mitochondrial permeability transition: a mechanism for DOPEGAL-induced apoptosis. Brain Res 787:328–332. https://doi.org/10.1016/s0006-8993(97)01488-1

    Article  CAS  PubMed  Google Scholar 

  10. Burke WJ, Li SW, Chung HD, Ruggiero DA, Kristal BS, Johnson EM, Lampe P, Kumar VB, Franko M, Williams EA et al (2004) Neurotoxicity of MAO metabolites of catecholamine neurotransmitters: role in neurodegenerative diseases. Neurotoxicology 25:101–115. https://doi.org/10.1016/S0161-813X(03)00090-1

    Article  CAS  PubMed  Google Scholar 

  11. Burke WJ, Li SW, Schmitt CA, Xia P, Chung HD, Gillespie KN (1999) Accumulation of 3,4-dihydroxyphenylglycolaldehyde, the neurotoxic monoamine oxidase A metabolite of norepinephrine, in locus ceruleus cell bodies in Alzheimer’s disease: mechanism of neuron death. Brain Res 816:633–637. https://doi.org/10.1016/s0006-8993(98)01211-6

    Article  CAS  PubMed  Google Scholar 

  12. Burke WJ, Schmitt CA, Gillespie KN, Li SW (1996) Norepinephrine transmitter metabolite is a selective cell death messenger in differentiated rat pheochromocytoma cells. Brain Res 722:232–235. https://doi.org/10.1016/0006-8993(96)00129-1

    Article  CAS  PubMed  Google Scholar 

  13. Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, Fagan AM, Morris JC, Mawuenyega KG, Cruchaga C et al (2011) Human apoE isoforms differentially regulate brain amyloid-β peptide clearance. Sci Transl Med 3:89ra57. https://doi.org/10.1126/scitranslmed.3002156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Caudle WM, Richardson JR, Wang MZ, Taylor TN, Guillot TS, McCormack AL, Colebrooke RE, Di Monte DA, Emson PC, Miller GW (2007) Reduced vesicular storage of dopamine causes progressive nigrostriatal neurodegeneration. J Neurosci 27:8138–8148. https://doi.org/10.1523/JNEUROSCI.0319-07.2007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chalermpalanupap T, Schroeder JP, Rorabaugh JM, Liles LC, Lah JJ, Levey AI, Weinshenker D (2018) Locus coeruleus ablation exacerbates cognitive deficits, neuropathology, and lethality in P301S Tau transgenic mice. J Neurosci 38:74–92. https://doi.org/10.1523/JNEUROSCI.1483-17.2017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chartier-Harlin MC, Parfitt M, Legrain S, Perez-Tur J, Brousseau T, Evans A, Berr C, Vidal O, Roques P, Gourlet V et al (1994) Apolipoprotein E, epsilon 4 allele as a major risk factor for sporadic early and late-onset forms of Alzheimer’s disease: analysis of the 19q13.2 chromosomal region. Hum Mol Genet 3:569–574

    Article  CAS  Google Scholar 

  17. Deming Y, Li Z, Kapoor M, Harari O, Del-Aguila JL, Black K, Carrell D, Cai Y, Fernandez MV, Budde J et al (2017) Genome-wide association study identifies four novel loci associated with Alzheimer’s endophenotypes and disease modifiers. Acta Neuropathol 133:839–856. https://doi.org/10.1007/s00401-017-1685-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Djordjevic J, Roy Chowdhury S, Snow WM, Perez C, Cadonic C, Fernyhough P, Albensi BC (2020) Early onset of sex-dependent mitochondrial deficits in the cortex of 3xTg Alzheimer’s mice. Cells. https://doi.org/10.3390/cells9061541

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ehrenberg AJ, Nguy AK, Theofilas P, Dunlop S, Suemoto CK, Di Lorenzo Alho AT, Leite RP, Diehl Rodriguez R, Mejia MB, Rub U et al (2017) Quantifying the accretion of hyperphosphorylated tau in the locus coeruleus and dorsal raphe nucleus: the pathological building blocks of early Alzheimer’s disease. Neuropathol Appl Neurobiol 43:393–408. https://doi.org/10.1111/nan.12387

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Engelborghs S, Dermaut B, Marien P, Symons A, Vloeberghs E, Maertens K, Somers N, Goeman J, Rademakers R, Van den Broeck M et al (2006) Dose dependent effect of APOE epsilon4 on behavioral symptoms in frontal lobe dementia. Neurobiol Aging 27:285–292. https://doi.org/10.1016/j.neurobiolaging.2005.02.005

    Article  CAS  PubMed  Google Scholar 

  21. Ghosh A, Torraville SE, Mukherjee B, Walling SG, Martin GM, Harley CW, Yuan Q (2019) An experimental model of Braak’s pretangle proposal for the origin of Alzheimer’s disease: the role of locus coeruleus in early symptom development. Alzheimers Res Ther 11:59. https://doi.org/10.1186/s13195-019-0511-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gillespie AK, Jones EA, Lin YH, Karlsson MP, Kay K, Yoon SY, Tong LM, Nova P, Carr JS, Frank LM et al (2016) Apolipoprotein E4 causes age-dependent disruption of slow gamma oscillations during hippocampal sharp-wave ripples. Neuron 90:740–751. https://doi.org/10.1016/j.neuron.2016.04.009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gomes LA, Hipp SA, Rijal Upadhaya A, Balakrishnan K, Ospitalieri S, Koper MJ, Largo-Barrientos P, Uytterhoeven V, Reichwald J, Rabe S et al (2019) Aβ-induced acceleration of Alzheimer-related tau-pathology spreading and its association with prion protein. Acta Neuropathol 138:913–941. https://doi.org/10.1007/s00401-019-02053-5

    Article  CAS  PubMed  Google Scholar 

  24. Grudzien A, Shaw P, Weintraub S, Bigio E, Mash DC, Mesulam MM (2007) Locus coeruleus neurofibrillary degeneration in aging, mild cognitive impairment and early Alzheimer’s disease. Neurobiol Aging 28:327–335. https://doi.org/10.1016/j.neurobiolaging.2006.02.007

    Article  CAS  PubMed  Google Scholar 

  25. Guillot TS, Miller GW (2009) Protective actions of the vesicular monoamine transporter 2 (VMAT2) in monoaminergic neurons. Mol Neurobiol 39:149–170. https://doi.org/10.1007/s12035-009-8059-y

    Article  CAS  PubMed  Google Scholar 

  26. Harley CW (2007) Norepinephrine and the dentate gyrus. Prog Brain Res 163:299–318. https://doi.org/10.1016/S0079-6123(07)63018-0

    Article  CAS  PubMed  Google Scholar 

  27. Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Williams A et al (2009) Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet 41:1088–1093. https://doi.org/10.1038/ng.440

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Holtzman DM (2001) Role of apoe/Aβ interactions in the pathogenesis of Alzheimer’s disease and cerebral amyloid angiopathy. J Mol Neurosci 17:147–155. https://doi.org/10.1385/JMN:17:2:147

    Article  CAS  PubMed  Google Scholar 

  29. Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM (2013) Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J Neurosci 33:1024–1037. https://doi.org/10.1523/JNEUROSCI.2642-12.2013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Iba M, McBride JD, Guo JL, Zhang B, Trojanowski JQ, Lee VM (2015) Tau pathology spread in PS19 tau transgenic mice following locus coeruleus (LC) injections of synthetic tau fibrils is determined by the LC’s afferent and efferent connections. Acta Neuropathol 130:349–362. https://doi.org/10.1007/s00401-015-1458-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Irizarry MC, Rebeck GW, Cheung B, Bales K, Paul SM, Holzman D, Hyman BT (2000) Modulation of Aβ deposition in APP transgenic mice by an apolipoprotein E null background. Ann NY Acad Sci 920:171–178. https://doi.org/10.1111/j.1749-6632.2000.tb06919.x

    Article  CAS  PubMed  Google Scholar 

  32. Josephs KA, Whitwell JL, Ahmed Z, Shiung MM, Weigand SD, Knopman DS, Boeve BF, Parisi JE, Petersen RC, Dickson DW et al (2008) β-amyloid burden is not associated with rates of brain atrophy. Ann Neurol 63:204–212. https://doi.org/10.1002/ana.21223

    Article  PubMed  PubMed Central  Google Scholar 

  33. Kang SS, Liu X, Ahn EH, Xiang J, Manfredsson FP, Yang X, Luo HR, Liles LC, Weinshenker D, Ye K (2020) Norepinephrine metabolite DOPEGAL activates AEP and pathological Tau aggregation in locus coeruleus. J Clin Invest 130:422–437. https://doi.org/10.1172/JCI130513

    Article  CAS  PubMed  Google Scholar 

  34. Leoni V (2011) The effect of apolipoprotein E (ApoE) genotype on biomarkers of amyloidogenesis, tau pathology and neurodegeneration in Alzheimer’s disease. Clin Chem Lab Med 49:375–383. https://doi.org/10.1515/CCLM.2011.088

    Article  CAS  PubMed  Google Scholar 

  35. Liu X, Ye K, Weinshenker D (2015) Norepinephrine protects against amyloid-β toxicity via TrkB. J Alzheimers Dis 44:251–260. https://doi.org/10.3233/JAD-141062

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mahley RW (1988) Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science 240:622–630

    Article  CAS  Google Scholar 

  37. Mahley RW, Rall SC Jr (2000) Apolipoprotein E: far more than a lipid transport protein. Annu Rev Genomics Hum Genet 1:507–537. https://doi.org/10.1146/annurev.genom.1.1.507

    Article  CAS  PubMed  Google Scholar 

  38. Mishra A, Ferrari R, Heutink P, Hardy J, Pijnenburg Y, Posthuma D, International FTDGC (2017) Gene-based association studies report genetic links for clinical subtypes of frontotemporal dementia. Brain 140:1437–1446. https://doi.org/10.1093/brain/awx066

    Article  PubMed  Google Scholar 

  39. Nathan BP, Bellosta S, Sanan DA, Weisgraber KH, Mahley RW, Pitas RE (1994) Differential effects of apolipoproteins E3 and E4 on neuronal growth in vitro. Science 264:850–852. https://doi.org/10.1126/science.8171342

    Article  CAS  PubMed  Google Scholar 

  40. Payami H, Montee K, Grimslid H, Shattuc S, Kaye J (1996) Increased risk of familial late-onset Alzheimer’s disease in women. Neurology 46:126–129. https://doi.org/10.1212/wnl.46.1.126

    Article  CAS  PubMed  Google Scholar 

  41. Reitz C, Mayeux R (2010) Use of genetic variation as biomarkers for mild cognitive impairment and progression of mild cognitive impairment to dementia. J Alzheimers Dis 19:229–251. https://doi.org/10.3233/JAD-2010-1255

    Article  PubMed  PubMed Central  Google Scholar 

  42. Rorabaugh JM, Chalermpalanupap T, Botz-Zapp CA, Fu VM, Lembeck NA, Cohen RM, Weinshenker D (2017) Chemogenetic locus coeruleus activation restores reversal learning in a rat model of Alzheimer’s disease. Brain 140:3023–3038. https://doi.org/10.1093/brain/awx232

    Article  PubMed  PubMed Central  Google Scholar 

  43. Shi Y, Yamada K, Liddelow SA, Smith ST, Zhao L, Luo W, Tsai RM, Spina S, Grinberg LT, Rojas JC et al (2017) ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549:523–527. https://doi.org/10.1038/nature24016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Stevens M, van Duijn CM, de Knijff P, van Broeckhoven C, Heutink P, Oostra BA, Niermeijer MF, van Swieten JC (1997) Apolipoprotein E gene and sporadic frontal lobe dementia. Neurology 48:1526–1529. https://doi.org/10.1212/wnl.48.6.1526

    Article  CAS  PubMed  Google Scholar 

  45. Stratmann K, Heinsen H, Korf HW, Del Turco D, Ghebremedhin E, Seidel K, Bouzrou M, Grinberg LT, Bohl J, Wharton SB et al (2016) Precortical phase of Alzheimer’s disease (AD)-related tau cytoskeletal pathology. Brain Pathol 26:371–386. https://doi.org/10.1111/bpa.12289

    Article  CAS  PubMed  Google Scholar 

  46. Strittmatter WJ, Saunders AM, Goedert M, Weisgraber KH, Dong LM, Jakes R, Huang DY, Pericak-Vance M, Schmechel D, Roses AD (1994) Isoform-specific interactions of apolipoprotein E with microtubule-associated protein tau: implications for Alzheimer disease. Proc Natl Acad Sci USA 91:11183–11186. https://doi.org/10.1073/pnas.91.23.11183

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sudhof TC (2004) The synaptic vesicle cycle. Annu Rev Neurosci 27:509–547. https://doi.org/10.1146/annurev.neuro.26.041002.131412

    Article  CAS  PubMed  Google Scholar 

  48. Tai LM, Mehra S, Shete V, Estus S, Rebeck GW, Bu G, LaDu MJ (2014) Soluble apoE/Aβ complex: mechanism and therapeutic target for APOE4-induced AD risk. Mol Neurodegener 9:2. https://doi.org/10.1186/1750-1326-9-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Taylor TN, Alter SP, Wang M, Goldstein DS, Miller GW (2014) Reduced vesicular storage of catecholamines causes progressive degeneration in the locus ceruleus. Neuropharmacology 76 Pt A:97–105. https://doi.org/10.1016/j.neuropharm.2013.08.033

    Article  CAS  PubMed  Google Scholar 

  50. Wang ZH, Xia Y, Liu P, Liu X, Edgington-Mitchell L, Lei K, Yu SP, Wang XC, Ye K (2021) ApoE4 Activates C/EBPβ/δ-secretase with 27-hydroxycholesterol, driving the pathogenesis of Alzheimer’s disease. Prog Neurobiol. https://doi.org/10.1016/j.pneurobio.2021.102032

    Article  PubMed  Google Scholar 

  51. Williams DR, Holton JL, Strand C, Pittman A, de Silva R, Lees AJ, Revesz T (2007) Pathological tau burden and distribution distinguishes progressive supranuclear palsy-parkinsonism from Richardson’s syndrome. Brain 130:1566–1576. https://doi.org/10.1093/brain/awm104

    Article  PubMed  Google Scholar 

  52. Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y (2006) Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci 26:4985–4994. https://doi.org/10.1523/JNEUROSCI.5476-05.2006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhang Z, Obianyo O, Dall E, Du Y, Fu H, Liu X, Kang SS, Song M, Yu SP, Cabrele C et al (2017) Inhibition of delta-secretase improves cognitive functions in mouse models of Alzheimer’s disease. Nat Commun 8:14740. https://doi.org/10.1038/ncomms14740

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, Seyfried NT, Hu WT, Liu Z, Wang JZ et al (2014) Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat Med 20:1254–1262. https://doi.org/10.1038/nm.3700

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhang Z, Song M, Liu X, Su Kang S, Duong DM, Seyfried NT, Cao X, Cheng L, Sun YE, Ping YuS et al (2015) Delta-secretase cleaves amyloid precursor protein and regulates the pathogenesis in Alzheimer’s disease. Nat Commun 6:8762. https://doi.org/10.1038/ncomms9762

    Article  CAS  PubMed  Google Scholar 

  56. Zhao N, Liu CC, Van Ingelgom AJ, Linares C, Kurti A, Knight JA, Heckman MG, Diehl NN, Shinohara M, Martens YA et al (2018) APOE epsilon2 is associated with increased tau pathology in primary tauopathy. Nat Commun 9:4388. https://doi.org/10.1038/s41467-018-06783-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Emory Goizueta Alzheimer’s Disease Research Center for postmortem human AD and healthy control samples. This study was supported in part by the Rodent Behavioral Core (RBC), Viral Vector Core, and HPLC Bioanalytical Core, which are subsidized by the Emory University School of Medicine and are part of the Emory Integrated Core Facilities.

Funding

This work was supported by the NIH (R01AG051538; RF1 AG061175 to KY and DW). Additional support was provided by the Emory Neuroscience NINDS Core Facilities (P30NS055077). Further support was provided by the Georgia Clinical and Translational Science Alliance of the National Institutes of Health under Award Number UL1TR002378.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Keqiang Ye.

Ethics declarations

Conflict of interest

The authors have declared that no conflict of interests exists.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

401_2021_2315_MOESM1_ESM.tif

Supplementary Fig. 1. ApoE4 selectively interacts with VMAT2. a The binding between ApoE4 and VMAT2 was investigated in SH-SY5Y cells, which were co-transfected with GFP-APOE3, GFP-APOE4, and VMAT2 (3 more N number of Fig. 2A). Transfected cell lysates were immunoprecipitated with anti-GFP, and the precipitated proteins were analyzed by immunoblotting with anti-VMAT2. b ApoE TR mouse brain cell lysates were immunoprecipitated with anti-ApoE and the precipitated proteins were analyzed by immunoblotting with anti-VMAT2 (3 more N number of Fig. 2L). c The interaction between ApoE4 and VMAT2 was confirmed in LC of ApoE TR mice. The interacted proximity signals of anti-ApoE4 and anti-VMAT2 are shown in red, and the nuclei are shown in blue by Proximity ligation assay. Scale bar is 20 μm. (TIF 1539 KB)

401_2021_2315_MOESM2_ESM.tif

Supplementary Fig. 2. ApoE4 triggers AEP activation and Tau cleavage and phosphorylation. Primary neurons were cultured and infected with AAV-hTau, AAV-ApoE3 or ApoE4, and AAV-AEP or AEP C189S. a Western blot analysis showed that ApoE4 overexpression induced Tau phosphorylation and Tau N368 cleavage with AEP overexpression, but not inactive AEP C189S (2 more N number of Fig. 3A). b Western blot analysis showed that Tau and ApoE4-induced AEP activation, Tau phosphorylation, and Tau N368 cleavage were blocked by AEP inhibitor compound 11 (2 more N number of Fig. 3d). c Western blot analysis showed that non-cleavable Tau N255A/N368A suppressed the effects of Tau and ApoE4 on AEP activation, Tau phosphorylation, and Tau N368 cleavage (2 more N number of Fig. 3g). (TIF 1894 KB)

401_2021_2315_MOESM3_ESM.tif

Supplementary Fig. 3. ApoE3 but not ApoE4 selectively binds to Tau and protects it from AEP cleavage. SH-SY5Y cells were co-transfected with ApoE3/E4 and GST-Tau. a GST pull down and western blot analysis showed that ApoE3 but not ApoE4 bound to Tau and prevented AEP activation, Tau phosphorylation and Tau aggregation. b LDH assay indicating Tau and ApoE4 mediated cell death. c The activation of AEP was confirmed by the enzymatic assay. Data are shown as mean ± SEM. N=3 per group. *p<0.05, **p<0.01. HEK293 cells were co-transfected with GFP-ApoE3 and GST-Tau Fragments to investigate ApoE3’s binding domain on Tau. d GST pull down analysis showed that ApoE3 bound to Tau in the repeat motifs (fragment 256–368) including AEP cleavage sites. e Recombinant Tau (5 μg) was incubated with recombinant AEP (0.05 μg) and escalating doses of recombinant ApoE3/E4 (0.1, 0.5, and 1 μg). Western blot and densitometric quantification of cleaved Tau band showed that rTau cleavage by rAEP was attenuated by rApoE3. f HEK293 cells were co-transfected with ApoE3/ApoE4 and Tau. Western data and densitometric analysis of cleaved Tau band presented the protective effect of ApoE3 in Tau cleavage by AEP. Data are shown as mean ± SEM. N=3 per group. *p<0.05, **p<0.01. (TIF 1799 KB)

401_2021_2315_MOESM4_ESM.tif

Supplementary Fig. 4. AEP is required for mediating ApoE4-provoked AD pathologies in MAPT mice. AAV-ApoE3 or AAV-ApoE4 was injected into the hippocampus of MAPT/AEP WT or MAPT/AEP KO mice, and the pathologic events in the hippocampus for AD were measured by various analysis. a ELISA analysis showed that Aβ40 and Aβ42 in the hippocampus were not changed by ApoE4 in MAPT mice. b ELISA assay for IL-6, TNF-α, and IL-1β showed that inflammatory cytokines were increased in the hippocampus by ApoE4 in MAPT/AEP WT mice, but not MAPT/AEP KO mice. Data of ELISA assay in a and b are shown as mean ± SEM. N=6 per group. *p<0.05. c Representative images of Golgi staining demonstrating the reduction of spines in CA1 region of ApoE4-injected MAPT/AEP WT mice. Scale bar is 5 μm. d Quantification of dendritic spines in Golgi staining of CA1 region. Data are shown as mean ± SEM. N=6 per group. *p<0.05. e Representative images by electron microscopy demonstrating the reduction of synapses (red arrows) in CA1 region of ApoE4-injected MAPT/AEP WT mice. Scale bar is 2 μm. f Quantification of synapse number in electron microscopy images of CA1 region. Data are shown as mean ± SEM. N=6 per group. *p<0.05. (TIF 1341 KB)

401_2021_2315_MOESM5_ESM.tif

Supplementary Fig. 5. Tau is required for ApoE4-induced AEP activation and neuronal cell death. Primary neurons from MAPT or Tau–/– mice brains were infected with AAV-ApoE3 or AAV-ApoE4. a Western blot analysis showed that ApoE4 induced AEP activation, Tau aggregation, Tau phosphorylation, and Tau N368 cleavage in MAPT mice primary neurons. b LDH assay demonstrated that the cell death by ApoE4 was mediated in a Tau-dependent manner. c The activation of AEP was confirmed by enzymatic assay. d ELISA assay showed that the change of Aβ40 content by ApoE4 was not affected by Tau. Data of b–d are shown as mean ± SEM. N=3 per group. *p<0.05, **p<0.01. (TIF 576 KB)

401_2021_2315_MOESM6_ESM.tif

Supplementary Fig. 6. Tau is required for ApoE4-triggered AD pathologies and LC neuronal loss. AAV-ApoE3 or AAV-ApoE4 was injected into the hippocampus of Tau–/– or MAPT mice, and pathologic events in the hippocampus for AD were assessed. a ELISA analysis showed that Aβ40 and Aβ42 in the hippocampus were not changed by ApoE4 in MAPT mice. b ELISA assay for IL-6, TNF-α, and IL-1β showed that inflammatory cytokines were increased in the hippocampus by ApoE4 in MAPT mice but not Tau–/– mice. Data of ELISA assay in a and b are shown as mean ± SEM. N=6 per group. *p<0.05. c Representative images of Golgi staining demonstrating the reduction of spines in CA1 region of ApoE4-injected MAPT mice. Scale bar is 5 μm. d Quantification of the dendritic spines in Golgi staining of CA1 region. Data are shown as mean ± SEM. N=6 per group. *p<0.05. e Representative images by electron microscopy demonstrating the reduction of synapses (red arrows) in CA1 region of ApoE4-injected MAPT mice. Scale bar is 2 μm. f Quantification of synapse number in electron microscopy images of CA1 region. Data are shown as mean ± SEM. N=6 per group. *p<0.05. g Stereological cell counting of TH+ cells in the LC region of Fig. 5d showed LC neurodegeneration mediated by ApoE4 injection into the hippocampus of MAPT mice. h Quantification of Tau N368+ cells and T22+ cells in the LC of Fig. 5d to show the retrograde spread of Tau pathologies. Data are shown as mean ± SEM. N=6 per group. *p<0.05. (TIF 1424 KB)

401_2021_2315_MOESM7_ESM.tif

Supplementary Fig. 7. ApoE4 inhibits VMAT2 and increases DOPEGAL and triggers cognitive impairments in MAPT mice. AAV-ApoE3/ApoE4 and Lenti-Sh-VMAT2 were injected into the LC of MAPT or Tau–/– mice, and then mice were assessed for Tau pathology and memory dysfunction 6 months later. a Quantification of ApoE+ cells and VMAT2+ cells in the LC of Fig. 6A to verify virus infection. b Representative images of T22 (red) and Thioflavin S immunofluorescence co-staining verifying Tau aggregation and fibrillization. Scale bar is 100 μm. c Quantification of Thioflavin S/T22+ cells in the LC shows Tau fibrillization is mediated by ApoE4 overexpression and VMAT2 deficiency. d MAO-A enzymatic assay in the LC for AAV-ApoE3/ApoE4 and Lenti-Sh-VMAT2-injected MAPT or Tau–/– mice. e Measurement of DOPEGAL in the LC following ApoE4 overexpression and VMAT2 deficiency via HPLC analysis. Behavioral tests for Morris water maze (f–h) and fear conditioning test (i and j) demonstrating memory dysfunctions mediated by ApoE4 overexpression and VMAT2 deficiency in MAPT mice. Distance travelled to the platform (f), area under the curve for total distance travelled (g), and percent time spent in the quadrant (h) previously containing the platform during the probe trial in the Morris water maze. Percent time spent freezing during the cued fear (i) and contextual fear (j) tests following fear conditioning. All data were analyzed using two-way ANOVA and shown as mean ± SEM. N=6 per group. * <0.05, ** <0.01 (TIF 2458 KB)

401_2021_2315_MOESM8_ESM.tif

Supplementary Fig. 8. VMAT2 knockout elicits DOPEGAL production, AEP activation, and LC neuronal loss in an age-dependent manner. LC neurodegeneration and DOPEGAL concentrations in VMAT2 LO mice were examined by immunofluorescence staining, enzymatic analysis, and HPLC assay. a Representative images of staining in LC sections of VMAT2 LO mice by TH (red), VMAT2 (green), and DAPI (blue). Scale bar is 100 μm. b Quantification of TH + cells representing the slight but significant reduction of LC neurons at 12 months of age in VMAT2-LO mice. c and d Enzymatic assays in LC of age-dependent VMAT2 LO mice for AEP activity (c) and MAO-A activity (d). e Measurement of age-dependent increase of DOPEGAL in the LC of VMAT2-LO mice via HPLC analysis. Data of be were analyzed using two-way ANOVA and shown as mean ± SEM, N = 3 per group. *p<0.05, **p < 0.01. f Western blot analysis showed the change of endogenous protein expression for Tau and TH in LC tissue of VMAT2 LO mice. (TIF 1940 KB)

Supplementary Fig. 9. The full western bolts used in Figures are presented (TIF 4367 KB)

Supplementary Fig. 10. The full western bolts used in Supplementary Figures are presented (TIF 4103 KB)

Supplementary Fig. 11. The full western bolts used in Supplementary Figures are presented (TIF 3969 KB)

Supplementary Table 1. Information of postmortem human patients for LC sections (TIF 342 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kang, S.S., Ahn, E.H., Liu, X. et al. ApoE4 inhibition of VMAT2 in the locus coeruleus exacerbates Tau pathology in Alzheimer’s disease. Acta Neuropathol 142, 139–158 (2021). https://doi.org/10.1007/s00401-021-02315-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00401-021-02315-1

Keywords

Navigation