Clusterin regulates β-amyloid toxicity via Dickkopf-1-driven induction of the wnt-PCP-JNK pathway

R Killick, E M Ribe, R Al-Shawi, B Malik, C Hooper, C Fernandes, R Dobson, P M Nolan, A Lourdusamy, S Furney, K Lin, G Breen, R Wroe, A W M To, K Leroy, M Causevic, A Usardi, M Robinson, W Noble, R Williamson, K Lunnon, S Kellie, C H Reynolds, C Bazenet, A Hodges, J-P Brion, J Stephenson, J Paul Simons, Simon Lovestone, R Killick, E M Ribe, R Al-Shawi, B Malik, C Hooper, C Fernandes, R Dobson, P M Nolan, A Lourdusamy, S Furney, K Lin, G Breen, R Wroe, A W M To, K Leroy, M Causevic, A Usardi, M Robinson, W Noble, R Williamson, K Lunnon, S Kellie, C H Reynolds, C Bazenet, A Hodges, J-P Brion, J Stephenson, J Paul Simons, Simon Lovestone

Abstract

Although the mechanism of Aβ action in the pathogenesis of Alzheimer's disease (AD) has remained elusive, it is known to increase the expression of the antagonist of canonical wnt signalling, Dickkopf-1 (Dkk1), whereas the silencing of Dkk1 blocks Aβ neurotoxicity. We asked if clusterin, known to be regulated by wnt, is part of an Aβ/Dkk1 neurotoxic pathway. Knockdown of clusterin in primary neurons reduced Aβ toxicity and DKK1 upregulation and, conversely, Aβ increased intracellular clusterin and decreased clusterin protein secretion, resulting in the p53-dependent induction of DKK1. To further elucidate how the clusterin-dependent induction of Dkk1 by Aβ mediates neurotoxicity, we measured the effects of Aβ and Dkk1 protein on whole-genome expression in primary neurons, finding a common pathway suggestive of activation of wnt-planar cell polarity (PCP)-c-Jun N-terminal kinase (JNK) signalling leading to the induction of genes including EGR1 (early growth response-1), NAB2 (Ngfi-A-binding protein-2) and KLF10 (Krüppel-like factor-10) that, when individually silenced, protected against Aβ neurotoxicity and/or tau phosphorylation. Neuronal overexpression of Dkk1 in transgenic mice mimicked this Aβ-induced pathway and resulted in age-dependent increases in tau phosphorylation in hippocampus and cognitive impairment. Furthermore, we show that this Dkk1/wnt-PCP-JNK pathway is active in an Aβ-based mouse model of AD and in AD brain, but not in a tau-based mouse model or in frontotemporal dementia brain. Thus, we have identified a pathway whereby Aβ induces a clusterin/p53/Dkk1/wnt-PCP-JNK pathway, which drives the upregulation of several genes that mediate the development of AD-like neuropathologies, thereby providing new mechanistic insights into the action of Aβ in neurodegenerative diseases.

Figures

Figure 1
Figure 1
Aβ induction of Dickkopf-1 (Dkk1) is clusterin dependent. (a) Rat primary cortical neurons were treated with Pen1 small interfering RNA (siRNA) to CLU overnight and subsequently with 20 μM Aβ25-35 for 24 and 48 h and cell survival determined by the nuclear morphology assay. Cont, Control. (b) Neurons were treated as in (a), RNA collected after 3 h of Aβ treatment and qRT-PCR performed (detailed in Supplementary Methods). (c) Neurons were treated for 4 h with 20 μM Aβ25-35 and culture media and total cell lysates were collected and immunoblotted for clusterin. Clusterin in cell lysates was normalised to β-actin levels. (d) Neurons were treated with 20 μM Aβ25-35 for the times indicated and immunoblotted as in (c). Error bars in (c) and (d) show s.d. qRT-PCR, quantitative reverse transcription polymerase chain reaction.
Figure 2
Figure 2
Aβ-induced gene expression is dependent on p53 and is necessary and sufficient for neurotoxicity. (a) Rat cortical neurons were treated with 10 μM pifithrin-α for 18 h as indicated and subsequently with Aβ1-42(olig) (3 μM, 3 h). The expression levels of DKK1 (Dickkopf-1), EGR1 (early growth response-1) and FOS (FBJ murine osteosarcoma viral oncogene homologue) were determined by qRT-PCR. (b) Neurons were treated as in (b) using 10 μM PRIMA-1 and then with Aβ and qRT-PCR performed. (c) Rat neurons were treated o/n at 7 d.i.c. with control or Pen1 small interfering RNA (siRNA) to DKK1 (160 nM), and then with 3 μM Aβ1-42(olig) for 24 h and cytotoxicity assayed by the live/dead assay. Healthy cells are labelled green and dead cells are red. Scale bar=10 μM. (d) Neurons were treated as in (c) and cell survival determined at 24, 48 and 72 h by the nuclear morphology assay. d.i.c., days in culture; o/n, over night; qRT-PCR, quantitative reverse transcription polymerase chain reaction.
Figure 3
Figure 3
EGR1 (early growth response-1), KLF10 (Krüppel-like factor-10) and NAB2 (Ngfi-A-binding protein-2) mediate neurotoxicity and tau phosphorylation. (a) Neurons were treated o/n with Pen1 small interfering RNAs (siRNAs) to EGR1, FOS (FBJ murine osteosarcoma viral oncogene homologue), KLF10, NAB2, CCND1 (cyclin D1) and then with 3 μM Aβ1-42(olig) for 24 h and cytotoxicity assayed by the live/dead assay. Protective effects of siRNAs targeting EGR1 and KLF10 are shown. (b) Neurons were treated as in (a) and cell survival determined by the nuclear morphology assay up to 72 h. Significance values (not shown) for the effect of EGR1 and KLF10 siRNA on cell survival at each time point were ≤0.01. (c) Neurons were treated as in (a) and cell survival measured by lactate dehydrogenase (LDH) release. (d) Neurons were treated as in (a) and subsequently with 3 μM Aβ1-42(olig) for 4 h. Total lysates were collected and immunoblotted for phospho-tau using PHF-1. Immunoreactivity values for PHF-1 were normalised to total tau values; densitometric values are shown in the right. (e) Neurons were treated with Pen1-siCLU or control Pen1 siRNA and subsequently with 3 μM Aβ1-42(olig) for 3 h, RNA collected and qRT-PCR performed for DKK1 and the five Aβ/Dkk1 target genes. o/n, over night; qRT-PCR, quantitative reverse transcription polymerase chain reaction.
Figure 4
Figure 4
Aβ induced gene expression in amyloidopathy, but not tauopathy, brain. (a–d) Expression levels of the five common Aβ/Dickkopf-1 (Dkk1)-responsive genes were measured by qRT-PCR in total RNA from: cortex of (a) 12-month-old Tg2576 mice (n=9) and their non-TG littermate controls (n=7) and (b) 12-month-old hTau mice (n=5) and their non-TG littermate controls (n=5); and from hippocampi of (c) Alzheimer's disease (AD; n=10) and age-matched controls (n=9) and (d) frontotemporal dementia (FTD; n=9) and age-matched controls (n=9). TG, transgenic. Mouse data were normalised to HPRT and human data to GAPDH by the 2-ΔΔCT method. Significance was determined by one-way analysis of variance (ANOVA) and post hoc t-tests. Data are represented as normalised fold increases over control. Significance values of gene changes in human samples are given in table (e). Outlier values (between 1.5 and 3 times the interquartile range) and extreme values (>3 times the range) are shown as circles and filled circles, respectively. (e) Dkk1-responsive genes examined in AD hippocampus. Nonsignificant and omitted genes are shown in grey. (f) Human brain transcriptome data sets were mined with the top 50 most significant Dkk1-responsive genes. Significance of Dkk1 gene enrichment in AD in the six brain regions examined by Liang et al. are shown. Significance was determined by asymptotic globaltest (see Supplementary Information for full description). (g) KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis of human homologues of rat Dkk1-responsive genes. The pathway identifier, name and P-values after correcting for multiple testing by the method of Benjamini are shown. Pathways in bold have been associated with disease in AD brain expression data by Huang et al. qRT-PCR, quantitative reverse transcription polymerase chain reaction.
Figure 5
Figure 5
Transgenic overexpression of Dickkopf-1 (DKK1) induces ‘Aβ neurotoxicity pathway genes', tau phosphorylation and cognitive deficits. (a) DKK1 expression was determined by qRT-PCR (left bar graph) and enzyme-linked immunosorbent assay (ELISA; (right bar graph) in temporal cortex of neonatal DKK1 transgenic (TG) mice (n=6) and their non-transgenic (non-TG) littermates (n=5). (b) Neonatal expression of the five Aβ/Dkk1 genes determined by qRT-PCR. (c) Immunoblots of hippocampal lysates from 18- to 24-month-old DKK1 TG (n=7) and non-TG (n=7) mice using antibodies AT8 and PHF-1. Phosphoimmunoreactivity values were normalised to total tau values, within blot (bar charts, right). (d) The 14–16-month-old Dkk1 TG (n=17) and non-TG littermates (n=16) were subjected to contextual fear conditioning. Time spent freezing upon placement in the conditioning apparatus at baseline and at 24 h after training are shown. qRT-PCR, quantitative reverse transcription polymerase chain reaction.
Figure 6
Figure 6
The Aβ neurotoxicity pathway is the wnt–planar cell polarity (PCP) pathway. (a) Neurons were treated for 2 h with full-length, N-Terminal CRD or C-terminal CRD containing conditioned media (CM) and qRT-PCR performed. (b) Neurons were treated as in (c) with recombinant Dickkopf-1 (Dkk1), Dkk2, Dkk3 and Dkk4 proteins at 800 ng ml−1 and expression measured. (c) Neuronal lysates were immunoblotted for phospho-Thr183/Tyr185-SAPK/JNK and total SAPK/JNK following 3 h of treatments with 3 μM Aβ1-42(olig) or 800 ng ml−1 Dkk1. (d) Nuclear fractions were prepared from neurons treated as in (e) and immunoblotted for phospho-Serine63-c-Jun. Equal loading was determined using anti-H2A.X. (e) Neurons were treated as in (c), fixed and stained for phospho Ser63-c-Jun. Scale bar=10 μm. (f) Neurons were pre-treated with the c-Jun N-terminal kinase (JNK) inhibitors SP600125 and BI-87G3, each at 10 μM for 3 h, and then with 800 ng ml−1 recombinant Dkk1 for 2 h and qRT-PCR was performed. (g) Human brain transcriptomic data were mined with the wnt–PCP pathway component genes. Significance of differences in Alzheimer's disease (AD) in the six brain regions examined by Liang et al. are shown. CRD, cysteine rich domain; qRT-PCR, quantitative reverse transcription polymerase chain reaction; SAPK, stress activated protein kinase.

References

    1. Glenner GG, Wong CW. Alzheimer's disease and Down's syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem Biophys Res Commun. 1984;122:1131–1135.
    1. Brion JP, Flament-Durand J, Dustin P. Alzheimer's disease and tau proteins. Lancet. 1986;2:1098.
    1. Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends Pharmacol Sci. 1991;12:383–388.
    1. Hardy J. The amyloid hypothesis for Alzheimer's disease: a critical reappraisal. J Neurochem. 2009;110:1129–1134.
    1. Kremer A, Louis JV, Jaworski T, F VanLeuven. GSK3 and Alzheimer's disease: facts and fiction. Front Mol Neurosci. 2011;4:17.
    1. Kang DE, Soriano S, Xia X, Eberhart CG, De Strooper B, Zheng H, et al. Presenilin couples the paired phosphorylation of beta-catenin independent of axin: implications for beta-catenin activation in tumorigenesis. Cell. 2002;110:751–762.
    1. Alvarez AR, Godoy JA, Mullendorff K, Olivares GH, Bronfman M, Inestrosa NC. Wnt-3a overcomes beta-amyloid toxicity in rat hippocampal neurons. Exp Cell Res. 2004;297:186–196.
    1. Caricasole A, Copani A, Caraci F, Aronica E, Rozemuller AJ, Caruso A, et al. Induction of Dickkopf-1, a negative modulator of the Wnt pathway, is associated with neuronal degeneration in Alzheimer's brain. J Neurosci. 2004;24:6021–6027.
    1. Toledo EM, Inestrosa NC.Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer's disease Mol Psychiatry 201015272–285.,228.
    1. De Ferrari GV, Papassotiropoulos A, Biechele T, Wavrant De-Vrieze F, Avila ME, Major MB, et al. Common genetic variation within the low-density lipoprotein receptor-related protein 6 and late-onset Alzheimer's disease. Proc Natl Acad Sci USA. 2007;104:9434–9439.
    1. Mudher A, Chapman S, Richardson J, Asuni A, Gibb G, Pollard C, et al. Dishevelled regulates the metabolism of amyloid precursor protein via protein kinase C/mitogen-activated protein kinase and c-Jun terminal kinase. J Neurosci. 2001;21:4987–4995.
    1. Killick R, Pollard CC, Asuni AA, Mudher AK, Richardson JC, Rupniak HT, et al. Presenilin 1 independently regulates beta-catenin stability and transcriptional activity. J Biol Chem. 2001;276:48554–48561.
    1. Rosen EY, Wexler EM, Versano R, Coppola G, Gao F, Winden KD, et al. Functional genomic analyses identify pathways dysregulated by progranulin deficiency, implicating Wnt signaling. Neuron. 2011;71:1030–1042.
    1. Chen S, Guttridge DC, You Z, Zhang Z, Fribley A, Mayo MW, et al. Wnt-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor-mediated transcription. J Cell Biol. 2001;152:87–96.
    1. Purro SA, Dickins EM, Salinas PC. The Secreted Wnt antagonist Dickkopf-1 is required for amyloid beta-mediated synaptic loss. J Neurosci. 2012;32:3492–3498.
    1. Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nat Genet. 2009;41:1094–1099.
    1. Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat Genet. 2009;41:1088–1093.
    1. Koch-Brandt C, Morgans C. Clusterin: a role in cell survival in the face of apoptosis. Prog Mol Subcell Biol. 1996;16:130–149.
    1. Rosi MC, Luccarini I, Grossi C, Fiorentini A, Spillantini MG, Prisco A, et al. Increased Dickkopf-1 expression in transgenic mouse models of neurodegenerative disease. J Neurochem. 2010;112:1539–1551.
    1. Thambisetty M, Simmons A, Velayudhan L, Hye A, Campbell J, Zhang Y, et al. Association of plasma clusterin concentration with severity, pathology, and progression in Alzheimer disease. Arch Gen Psychiatry. 2010;67:739–748.
    1. Schrijvers EM, Koudstaal PJ, Hofman A, Breteler MM. Plasma clusterin and the risk of Alzheimer disease. JAMA. 2011;305:1322–1326.
    1. Schepeler T, Mansilla F, Christensen LL, Orntoft TF, Andersen CL. Clusterin expression can be modulated by changes in TCF1-mediated Wnt signaling. J Mol Signal. 2007;2:6.
    1. Gonzalez-Sancho JM, Aguilera O, Garcia JM, Pendas-Franco N, Pena C, Cal S, et al. The Wnt antagonist DICKKOPF-1 gene is a downstream target of beta-catenin/TCF and is downregulated in human colon cancer. Oncogene. 2005;24:1098–1103.
    1. Theuns J, Remacle J, Killick R, Corsmit E, Vennekens K, Huylebroeck D, et al. Alzheimer-associated C allele of the promoter polymorphism -22C>T causes a critical neuron-specific decrease of presenilin 1 expression. Hum Mol Genet. 2003;12:869–877.
    1. Tizon B, Ribe EM, Mi W, Troy CM, Levy E. Cystatin C protects neuronal cells from amyloid-beta-induced toxicity. J Alzheimers Dis. 2010;19:885–894.
    1. Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS, Khvorova A. Rational siRNA design for RNA interference. Nat Biotechnol. 2004;22:326–330.
    1. Davidson TJ, Harel S, Arboleda VA, Prunell GF, Shelanski ML, Greene LA, et al. Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation. J Neurosci. 2004;24:10040–10046.
    1. Killick R, Scales G, Leroy K, Causevic M, Hooper C, Irvine EE, et al. Deletion of Irs2 reduces amyloid deposition and rescues behavioural deficits in APP transgenic mice. Biochem Biophys Res Commun. 2009;386:257–262.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–408.
    1. Trougakos IP, So A, Jansen B, Gleave ME, Gonos ES. Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 2004;64:1834–1842.
    1. Bertram L, Tanzi RE. Alzheimer disease: new light on an old CLU. Nat Rev Neurol. 2010;6:11–13.
    1. Hooper C, Killick R, Fernandes C, Sugden D, Lovestone S. Transcriptomic profiles of Wnt3a and insulin in primary cultured rat cortical neurones. J Neurochem. 2011;118:512–520.
    1. Millucci L, Ghezzi L, Bernardini G, Santucci A. Conformations and biological activities of amyloid beta peptide 25-35. Curr Protein Pept Sci. 2009;11:54–67.
    1. Abeti R, Abramov AY, Duchen MR. Beta-amyloid activates PARP causing astrocytic metabolic failure and neuronal death. Brain. 2011;134 (Pt 6:1658–1672.
    1. Lu Y, Li T, Qureshi HY, Han D, Paudel HK. Early growth response 1 (Egr-1) regulates phosphorylation of microtubule-associated protein tau in mammalian brain. J Biol Chem. 2011;286:20569–20581.
    1. Kumbrink J, Kirsch KH, JP Johnson. EGR1, EGR2, and EGR3 activate the expression of their coregulator NAB2 establishing a negative feedback loop in cells of neuroectodermal and epithelial origin. J Cell Biochem. 2010;111:207–217.
    1. Svaren J, Sevetson BR, Golda T, Stanton JJ, Swirnoff AH, Milbrandt J. Novel mutants of NAB corepressors enhance activation by Egr transactivators. EMBO J. 1998;17:6010–6019.
    1. Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, et al. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99–102.
    1. Andorfer C, Kress Y, Espinoza M, de Silva R, Tucker KL, Barde YA, et al. Hyperphosphorylation and aggregation of tau in mice expressing normal human tau isoforms. J Neurochem. 2003;86:582–590.
    1. Liang WS, Dunckley T, Beach TG, Grover A, Mastroeni D, Ramsey K, et al. Altered neuronal gene expression in brain regions differentially affected by Alzheimer's disease: a reference data set. Physiol Genomics. 2008;33:240–256.
    1. Huang Y, Sun X, Hu G. An integrated genetics approach for identifying protein signal pathways of Alzheimer's disease. Comput Methods Biomech Biomed Engin. 2011;14:371–378.
    1. Webster JA, Gibbs JR, Clarke J, Ray M, Zhang W, Holmans P, et al. Genetic control of human brain transcript expression in Alzheimer disease. Am J Hum Genet. 2009;84:445–458.
    1. Lockstone HE, Harris LW, Swatton JE, Wayland MT, Holland AJ, Bahn S. Gene expression profiling in the adult Down syndrome brain. Genomics. 2007;90:647–660.
    1. Ideker T, Ozier O, Schwikowski B, Siegel AF. Discovering regulatory and signalling circuits in molecular interaction networks. Bioinformatics. 2002;18 (Suppl 1:S233–S240.
    1. Bertrand J, Plouffe V, Senechal P, Leclerc N. The pattern of human tau phosphorylation is the result of priming and feedback events in primary hippocampal neurons. Neuroscience. 2010;168:323–334.
    1. Korol O, Gupta RW, Mercola M. A novel activity of the Dickkopf-1 amino terminal domain promotes axial and heart development independently of canonical Wnt inhibition. Dev Biol. 2008;324:131–138.
    1. Krupnik VE, Sharp JD, Jiang C, Robison K, Chickering TW, Amaravadi L, et al. Functional and structural diversity of the human Dickkopf gene family. Gene. 1999;238:301–313.
    1. Bafico A, Liu G, Yaniv A, Gazit A, Aaronson SA. Novel mechanism of Wnt signalling inhibition mediated by Dickkopf-1 interaction with LRP6/Arrow. Nat Cell Biol. 2001;3:683–686.
    1. Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A, et al. LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature. 2001;411:321–325.
    1. Semenov MV, Tamai K, Brott BK, Kuhl M, Sokol S, He X. Head inducer Dickkopf-1 is a ligand for Wnt coreceptor LRP6. Curr Biol. 2001;11:951–961.
    1. Caneparo L, Huang YL, Staudt N, Tada M, Ahrendt R, Kazanskaya O, et al. Dickkopf-1 regulates gastrulation movements by coordinated modulation of Wnt/beta catenin and Wnt/PCP activities, through interaction with the Dally-like homolog Knypek. Genes Dev. 2007;21:465–480.
    1. Boutros M, Paricio N, Strutt DI, Mlodzik M. Dishevelled activates JNK and discriminates between JNK pathways in planar polarity and wingless signaling. Cell. 1998;94:109–118.
    1. Yamanaka H, Moriguchi T, Masuyama N, Kusakabe M, Hanafusa H, Takada R, et al. JNK functions in the non-canonical Wnt pathway to regulate convergent extension movements in vertebrates. EMBO Rep. 2002;3:69–75.
    1. Pandur P, Lasche M, Eisenberg LM, Kuhl M. Wnt-11 activation of a non-canonical Wnt signalling pathway is required for cardiogenesis. Nature. 2002;418:636–641.
    1. Checler F, Dunys J, Pardossi-Piquard R, Alves da Costa C. p53 is regulated by and regulates members of the gamma-secretase complex. Neurodegener Dis. 2010;7:50–55.
    1. MacGibbon GA, Lawlor PA, Walton M, Sirimanne E, Faull RL, Synek B, et al. Expression of Fos, Jun, and Krox family proteins in Alzheimer's disease. Exp Neurol. 1997;147:316–332.
    1. Gomez Ravetti M, Rosso OA, Berretta R, Moscato P. Uncovering molecular biomarkers that correlate cognitive decline with the changes of hippocampus' gene expression profiles in Alzheimer's disease. PLoS One. 2010;5:e10153.
    1. Criswell T, Beman M, Araki S, Leskov K, Cataldo E, Mayo LD, et al. Delayed activation of insulin-like growth factor-1 receptor/Src/MAPK/Egr-1 signaling regulates clusterin expression, a pro-survival factor. J Biol Chem. 2005;280:14212–14221.
    1. Tolias KF, Duman JG, Um K. Control of synapse development and plasticity by Rho GTPase regulatory proteins. Prog Neurobiol. 2011;94:133–148.
    1. Montcouquiol M, Crenshaw EB, Kelley MW. Noncanonical Wnt signaling and neural polarity. Annu Rev Neurosci. 2006;29:363–386.

Source: PubMed

3
Prenumerera