ios press jnk plays a key role in tau hyperphosphorylation ... · jnk plays a key role in tau...

15
Uncorrected Author Proof Journal of Alzheimer’s Disease 25 (2011) 1–15 DOI 10.3233/JAD-2011-110320 IOS Press 1 JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s Disease Models 1 2 3 Cristina Ploia a,1 , Xanthi Antoniou a,1 , Alessandra Sclip a , Valentina Grande a,d , Daniele Cardinetti a , Alessio Colombo a,2 , Nadia Canu b, , Luisa Benussi c , Roberta Ghidoni c , Gianluigi Forloni a and Tiziana Borsello a,4 5 6 a Istituto di Ricerche Farmacologiche “Mario Negri”, Milano, Italy 7 b Dipartimento di Neuroscienze, Universit ` a di Tor Vergata, Rome, Italy and Istituto di Neurobiologia e Medicina Molecolare, CNR, Rome, Italy 8 9 c Proteomics Unit NeuroBioGen Lab-Memory Clinic, IRCCS Centro S. Giovanni di Dio-Fatebenefratelli, Brescia, Italy 10 11 d NICO, Scientific Institute of the Cavalieri-Ottolenghi Foundation Neuroscience Institute of Turin, Orbassano (To), Italy 12 13 Accepted 9 April 2011 Abstract. Alzheimer’s disease (AD) is a major clinical concern, and the search for new molecules to combat disease progression remains important. One of the major hallmarks in AD pathogenesis is the hyperphosphorylation of tau and subsequent formation of neurofibrillary tangles. Several kinases are involved in this process. Amongst them, c-Jun N-terminal kinases (JNKs) are activated in AD brains and are also associated with the development of amyloid plaques. This study was designed to investigate the contribution of JNK in tau hyperphosphorylation and whether it may represent a potential therapeutic target for the fight against AD. The specific inhibition of JNK by the cell permeable peptide D-JNKI-1 led to a reduction of p-tau at S202/T205 and S422, two established target sites of JNK, in rat neuronal cultures and in human AD fibroblasts cultures. Similarly, D-JNKI-1 reduced p-tau at S202/T205 in an in vivo model of AD (TgCRND8 mice). Our findings support the fundamental role of JNK in the regulation of tau hyperphosphorylation and subsequently in AD pathogenesis. 14 15 16 17 18 19 20 21 22 Keywords: Alzheimer’s disease, D-JNKI-1, JNK mitogen-activated protein kinases, phosphorylation, tau protein 23 INTRODUCTION 24 c-Jun N terminal kinases (JNKs) belong to the fam- 25 ily of serine and threonine mitogen-activated protein 1 These two authors contributed equally to this work. 2 Present Address: DZNE - German Center for Neurodegenerative Diseases, Schillerstrasse 44, 80336 Munich, Germany. Correspondence to: Tiziana Borsello, Neuronal death and Neu- roprotection Unit, Neuroscience Department, Istituto di Ricerche Farmacologiche “Mario Negri”, Via La Masa 19, 20156 Milano, Italy. Tel.: +39 02 39014469/39014592; Fax: +39 02 3546277; E-mail: [email protected]. kinases (MAPKs) and are involved in numerous cel- 26 lular processes, such as proliferation, differentiation, 27 development, inflammation, and apoptosis [1]. In the 28 central nervous system (CNS) the JNK signaling path- 29 way is important in controlling brain functions under 30 both normal and pathological conditions [2]. 31 In particular, in Alzheimer’s disease (AD), activa- 32 tion of JNK [3–6] as well as p-c-jun [7, 8], correlate 33 with disease progression. Furthermore, JNK is acti- 34 vated by amyloid- (A) fragments [9, 10] and causes 35 phosphorylation of both amyloid- protein precursor 36 ISSN 1387-2877/11/$27.50 © 2011 – IOS Press and the authors. All rights reserved

Upload: ngokien

Post on 09-Nov-2018

226 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

Journal of Alzheimer’s Disease 25 (2011) 1–15DOI 10.3233/JAD-2011-110320IOS Press

1

JNK Plays a Key Role in TauHyperphosphorylation in Alzheimer’sDisease Models

1

2

3

Cristina Ploiaa,1, Xanthi Antonioua,1, Alessandra Sclipa, Valentina Grandea,d, Daniele Cardinettia,Alessio Colomboa,2, Nadia Canub,, Luisa Benussic, Roberta Ghidonic, Gianluigi Forlonia andTiziana Borselloa,∗

4

5

6

aIstituto di Ricerche Farmacologiche “Mario Negri”, Milano, Italy7

bDipartimento di Neuroscienze, Universita di Tor Vergata, Rome, Italy and Istituto di Neurobiologiae Medicina Molecolare, CNR, Rome, Italy

8

9

cProteomics Unit NeuroBioGen Lab-Memory Clinic, IRCCS Centro S. Giovanni di Dio-Fatebenefratelli,Brescia, Italy

10

11

dNICO, Scientific Institute of the Cavalieri-Ottolenghi Foundation Neuroscience Institute of Turin,Orbassano (To), Italy

12

13

Accepted 9 April 2011

Abstract. Alzheimer’s disease (AD) is a major clinical concern, and the search for new molecules to combat disease progressionremains important. One of the major hallmarks in AD pathogenesis is the hyperphosphorylation of tau and subsequent formationof neurofibrillary tangles. Several kinases are involved in this process. Amongst them, c-Jun N-terminal kinases (JNKs) areactivated in AD brains and are also associated with the development of amyloid plaques. This study was designed to investigatethe contribution of JNK in tau hyperphosphorylation and whether it may represent a potential therapeutic target for the fightagainst AD. The specific inhibition of JNK by the cell permeable peptide D-JNKI-1 led to a reduction of p-tau at S202/T205 andS422, two established target sites of JNK, in rat neuronal cultures and in human AD fibroblasts cultures. Similarly, D-JNKI-1reduced p-tau at S202/T205 in an in vivo model of AD (TgCRND8 mice). Our findings support the fundamental role of JNK inthe regulation of tau hyperphosphorylation and subsequently in AD pathogenesis.

14

15

16

17

18

19

20

21

22

Keywords: Alzheimer’s disease, D-JNKI-1, JNK mitogen-activated protein kinases, phosphorylation, tau protein23

INTRODUCTION24

c-Jun N terminal kinases (JNKs) belong to the fam-25

ily of serine and threonine mitogen-activated protein1 These two authors contributed equally to this work.2 Present Address: DZNE - German Center for Neurodegenerative

Diseases, Schillerstrasse 44, 80336 Munich, Germany.∗Correspondence to: Tiziana Borsello, Neuronal death and Neu-

roprotection Unit, Neuroscience Department, Istituto di RicercheFarmacologiche “Mario Negri”, Via La Masa 19, 20156 Milano,Italy. Tel.: +39 02 39014469/39014592; Fax: +39 02 3546277;E-mail: [email protected].

kinases (MAPKs) and are involved in numerous cel- 26

lular processes, such as proliferation, differentiation, 27

development, inflammation, and apoptosis [1]. In the 28

central nervous system (CNS) the JNK signaling path- 29

way is important in controlling brain functions under 30

both normal and pathological conditions [2]. 31

In particular, in Alzheimer’s disease (AD), activa- 32

tion of JNK [3–6] as well as p-c-jun [7, 8], correlate 33

with disease progression. Furthermore, JNK is acti- 34

vated by amyloid-� (A�) fragments [9, 10] and causes 35

phosphorylation of both amyloid-� protein precursor 36

ISSN 1387-2877/11/$27.50 © 2011 – IOS Press and the authors. All rights reserved

Page 2: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

2 C. Ploia et al. / JNK and Tau Phosphorylation in AD

(A�PP), �−C-terminal fragment (�−CTF) [11–14],37

and tau [4, 15–18].38

The amyloid-cascade hypothesis of AD considers39

the production of A� a key event in AD pathol-40

ogy [19], however another school of thought sustains41

that the neurofibrillary tangles of hyperphosphorylated42

tau, as opposed to amyloid deposits, are more influ-43

ential to dementia associated with AD [20]. In line44

with the above, the decrease of tau phosphorylation45

has been correlated to improvement of memory and46

to reduction of neurodegeneration in transgenic mice47

over-expressing mutant human tau (P301 L) [21] and48

in double transgenic mice co-expressing human mutant49

A�PP and tau [22]. Thus hyperphosphorylated tau may50

be a primary cause of the disorder and therefore under-51

standing its regulation is important for the development52

of therapeutic strategies.53

Tau can be phosphorylated at multiple sites by an54

array of kinases, and the phosphorylation events that55

lead to AD have been extensively studied [23–25].56

In particular JNK is known to phosphorylate tau at57

S202/T205 and S422 [26, 27]. These two phosphoryla-58

tion sites are strictly associated with AD pathogenesis.59

Luna-Munoz and colleagues [25] reported that in AD60

brains, phosphorylation at the S202/T205 residues of61

tau, recognized by the AT8 antibody, occurs at an early62

stage of the disease, while phosphorylation at S42263

precedes and correlates with tangle formation [28].64

To study the involvement of the JNK pathway on65

tau hyperphosphorylation we analyzed the action of66

JNK at S202/T205, S422 using three different models:67

differentiated cortical neurons, as an in vitro control68

model; transgenic AD mice (TgCRND8), to assess the69

contribution of JNK in tau phosphorylation in an vivo70

model; and finally human AD fibroblasts, which are71

easily accessible from patients and are a good model72

to validate potential therapeutic strategies. The latter73

also offer an excellent way to study the mechanisms74

involved in the development of AD [29].75

To achieve specific inhibition of JNK in vitro and76

in vivo, we used a cell permeable JNK inhibitor pep-77

tide (D-JNKI-1), which prevents JNK action without78

interfering with its activation/phosphorylation [30, 31]79

in low concentrations (2 �M) and modulates the enzy-80

matic activity in high concentrations (4 and 6 �M), as81

we have previously demonstrated [32]. We analyzed82

the effect of D-JNKI-1 in these models and we proved83

that JNK plays a key role in tau hyperphosphorylation84

on S202/T205 and S422. A better understanding of85

the JNK pathway activation/inhibition is of great ther-86

apeutic interest and may have important implications87

for the fight against AD.

MATERIALS AND METHODS 88

Cortical neuronal cultures 89

Primary neuronal cultures were obtained from the 90

cerebral cortex of two days postnatal rats, incubated 91

with 200 U of papain (P3125, Sigma Aldrich, St Louis, 92

USA) (30’−34◦C), then with trypsin inhibitor (T- 93

9253, Sigma Aldrich) (45’ −34◦C), and subsequently 94

mechanically dissociated. Neurons were plated in 95

35 mm dishes (∼7 × 105 cells/dish) pre-coated with 96

25 �g/ml poly-D-lysine (P6407, Sigma Aldrich). 97

Plating medium was B27/neurobasal-A (17504- 98

044, 10888, Gibco-Invitrogen, Paisley, Scotland, 99

UK) supplemented with 0.5 mM glutamine (25030, 100

Gibco-Invitrogen), 100 U/ml penicillin, and 100 �g/ml 101

streptomycin (15140-122, Gibco-Invitrogen). To avoid 102

proliferation of glial cells and in order to achieve 95% 103

purity in neuronal cultures, AraC (10 �M), (C6645, 104

Sigma Aldrich) was added to the media at 2 days from 105

plating date (2DIV). 106

The experiments were performed 12 days from plat- 107

ing date (12DIV), at which time neurons are considered 108

differentiated [33–35]. All experiments were repeated 109

using at least six independent culture preparations. 110

Neurons were treated with D-JNKI-1 (2-4-6 �M) 111

(Istituto di Ricerche Farmacologiche “Mario Negri”, 112

Milano, Italy) for 24 h. This D-retro-inverso inhibitor 113

of JNK is composed exclusively of D- instead of L- 114

amino acids and is synthesized in a reverse order to 115

preserve functionality. The inactive peptide D-TAT was 116

used as a control to prove the specificity of D-JNKI- 117

1, as previously shown by Borsello et al. [30]. All 118

experimental procedures on animals were performed 119

in accordance with the European Communities Coun- 120

cil Directive of 24 November 1986 (86/609/EEC) and 121

all efforts were made to minimize animal suffering. 122

Subjects and human fibroblasts cultures 123

Patients included in this study were recruited and 124

evaluated at the Memory Clinic of IRCCS “Centro 125

San Giovanni di Dio-Fatebenefratelli”, Brescia, Italy. 126

Written informed consent was obtained from all sub- 127

jects or, where appropriate, their caregivers, following 128

the procedures approved by the hospital local ethical 129

committee. Fibroblast primary cultures were derived 130

from the dermal biopsy of AD and control subjects 131

(NA) as previously described [36]. Global cognition 132

was investigated by Mini-Mental State Examination 133

(MMSE). Demographic features of fibroblasts donors: 134

AD patients, n = 7, mean age = 72.2 ± 6.94 years, 135

Page 3: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 3

gender = 80% female, MMSE = 17.4 ± 5.9; NA con-136

trols subjects, n = 6, mean age = 62.6 ± 4.28 years,137

gender = 60% female, MMSE = 28.8 ± 1.1. Fibrob-138

lasts from unrelated healthy controls (NA) were139

obtained from Fatebenefratelli Biological Repository140

(F-BR) of IRCCS “Centro San Giovanni di Dio-141

Fatebenefratelli”, Brescia, Italy. Cells were cultured142

at 37◦C in 5% CO2/95% air in Eagle’s Mini-143

mum Essential Medium (31095, Gibco-Invitrogen),144

10% FBS (SH30070.03, Hyclone-Thermo Scientific,145

Rockford, USA), 100 U/ml P/S (15140-122, Gibco-146

Invitrogen), 1% v/v Non Essential Amminoacids147

(M7145, Sigma Aldrich), 2 mM L-Glutamine (25030,148

Gibco-Invitrogen). The different fibroblast cell cul-149

tures were plated and analyzed at the same passage150

number. Fibroblasts were plated in 35 mm dishes, �-151

Dishes (80826, Ibidi, Martinsried, Germany) or 96 well152

plate. 70–80% confluent cultures were treated with153

D-JNKI-1 (10–150 �M) (Istituto di Ricerche Farma-154

cologiche “Mario Negri”, Milano, Italy) for 24 h.155

Cell peptide uptake156

2 � M of FITC-labeled D-JNKI-1 (Istituto di157

Ricerche Farmacologiche “Mario Negri”) was added158

to the culture medium of cortical neurons for 24 h at159

37◦C. 50 �M of FITC-labeled D-JNKI-1 was added to160

the culture medium of fibroblasts (70–80% confluent)161

for 24 h at 37◦C. Cells were rinsed three times in PBS162

and fed with fresh medium. Free FITC was used as163

a control for background signals. Peptide internaliza-164

tion was monitored by fluorescence in live cells using165

a confocal laser scanning microscope. Excitation was166

performed at 488 nm.167

Real-time reverse transcription-PCR168

Total RNA was isolated from cortical neurons using169

RNeasy Mini kit (Qiagen GmbH, D-40724 Hilden)170

and then treated with DNase (Dnase I, Amplifica-171

tion Grade, Invitrogen, Carlsbad, CA 92008 USA).172

Extracted RNA was quantified using Agilent RNA173

6000 Nano Kit and Agilent Bioanalyzer 2100 (Agi-174

lent Technologies, Hewlett, Packard – Str.8, 76337175

Walbronn, Germany). Primer and probe sequences176

were designed using Beacon Designer 3.0 software177

(Premier Biosoft International, Palo Alto, CA) accord-178

ing to specification for SYBR-green [37]. Sequences179

for the forward primer and reverse primer for tau and180

for �-actin from Zhao et al. 2007 [38]. �-actin mRNA181

expression, which is not modulated by the peptide as182

previously demonstrated [13], was used as an endoge-183

nous control. Real-time PCR reaction was performed 184

in a final volume of 25 �l containing 45 ng of cDNA, 185

200 nM of both primers, 12.5 �l of SYBR-green PCR 186

Master Mix containing: SYBR-green I dye, Ampli- 187

Taq Gold DNA Polymerase, dNTPs, Passive Reference 188

RoxTM dye, using Applied Biosystems 7300 Real- 189

Time PCR System (Applied Biosystems, Warrington, 190

UK). The amplification protocol included 2 min at 191

50◦C, 10 min at 95◦C, followed by 60 cycles of 15 s 192

at 95◦C for denaturation and 30 s at 60◦C for anneal- 193

ing and extension. The relative expression of tau was 194

calculated by comparative method (Comparative CT 195

method - User Bulletin #2, Applied Biosystems). 196

TgCRND8 mice 197

Transgenic hemizygous TgCRND8 mice carry a 198

mutated human A�PP gene (A�PP-Swedish & Indi- 199

ana mutations on 129 SV strain) and show age-related 200

amyloid plaque accumulation at around 4/5 months of 201

age and tangles formation at the age of 7 months [39]. 202

A post-symptomatic treatment with D-JNKI-1 (IP 203

injection-22 mg/kg) was performed on TgCRND8 204

mice for 4 months (every 21 days), starting at the age 205

of 4 months. Mice were sacrificed 3 weeks after the 206

last injection. In these conditions the treatment has no 207

side effects. Mice were divided in 4 groups: wild type 208

treated with vehicle, wild type treated with D-JNKI-1, 209

transgenic animals treated with vehicle and transgenic 210

mice treated with D-JNKI-1. At least seven animals 211

were analyzed for each group. 212

Immunofluorescence analysis 213

Cortical neurons and fibroblasts were plated on 214

�-Dishes (80826, Ibidi, Martinsried, Germany) and 215

fixed in 4% paraformaldehyde, 5% sucrose-PBS for 216

30 min on ice. Nonspecific antibody binding sites were 217

blocked for 1 h with 10% normal goat serum in PBS 218

and triton 0.3% at room temperature. Primary antibod- 219

ies specific for PHF-tau clone AT8 (MN1020, Thermo 220

Scientific, Rockford, USA) (1 : 100) was diluted in 221

PBS-Triton 0.1%, NGS 1% and used overnight at 4◦C. 222

Cortical neurons were rinsed in PBS and exposed to 223

the secondary Alexa 546 antibody (A11030, Invitro- 224

gen, Molecular Probes, Eugene, Oregon, USA) for 225

1 h at room temperature. For fibroblasts TSA Fluo- 226

rescence Systems was used for signal amplification 227

(Cyanine 5 System, NEL705A001 KT, PerkinElmer, 228

Boston, MA). Hoechst reagent (33258, Invitrogen) 229

(1 : 500) in PBS was used to stain the nuclei. Stain- 230

ing for F-actin cytoskeleton was performed with 231

Page 4: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

4 C. Ploia et al. / JNK and Tau Phosphorylation in AD

Rhodamine-Phalloidin (R415, Invitrogen, Molecular232

Probes) (1 : 40 in PBS) for 30 min at room temperature.233

Cytotoxicity Assay234

Toxicity following D-JNKI-1 treatment was evalu-235

ated by a Lactate dehydrogenase (LDH) assay. LDH236

released in the medium was measured using the Cyto-237

tox 96 non radioactive cytotoxicity assay kit (G1780,238

Promega, WI, USA). All cytotoxicity assays were per-239

formed in triplicates.240

Cellular lysis241

Total protein extracts were obtained by washing242

cells twice in ice-cold PBS and lysed (20’ −4◦C) in 1%243

Triton x-100 lysis buffer supplemented with proteases244

(11873580001, Roche) and phosphatases (PhosStop245

04906845001, Roche) inhibitors [31].246

Western blot analysis247

Protein concentrations were quantified using Brad-248

ford Assay (Bio-Rad Protein Assay 500-0006,249

Munchen, Germany) and 20 �g of whole cell pro-250

teins or brain homogenates were separated by 8–10%251

SDS polyacrylamide gel. PVDF membranes were252

blocked in Tris-buffered saline (5% no fat milk pow-253

der, 0.1% Tween20) (1 h, room temperature). Primary254

antibodies were diluted in the same buffer (incuba-255

tion overnight, 4◦C) using: anti PHF-tau clone AT8256

(MN1020, Thermo Scientific, Rockford, USA), p-257

tau pSer422 (T 7944, Sigma), anti-tau clone tau-5258

(MAB361, Millipore, Billerica, Ma), p-c-Jun Ser73259

(9164, Cell Signaling, Danvers, MA), c-Jun (9165,260

Cell Signaling), P-JNK (G-7) (sc-6254, Santa Cruz261

Biotechnology, Santa Cruz, California), JNK (9252,262

Cell Signaling). All blots were normalized to Actin263

(MAB1501, Chemicon-Millipore, Billerica, Ma) and264

at least three independent experiments were per-265

formed. Western blots were quantified by densitometry266

using Quantity One software (Biorad).267

Statistical analysis268

Quantitative data were analyzed by paired Student’s269

t-test with two-tailed distribution or One Way-, Two270

Way- ANOVA (Tukey’s post hoc test). A p value of271

<0.05 was considered significant.

RESULTS 272

D-JNKI-1 treatment prevents c-Jun and tau 273

phosphorylation in rat cortical neurons without 274

initiating cell death processes 275

The efficacy of D-JNKI-1 was tested by assessing 276

the uptake of FITC-labeled D-JNKI-1 in adult corti- 277

cal neurons in control conditions. Briefly, cells were 278

exposed to 2 � M of FITC-labeled D-JNKI-1 for 24 h 279

at 37◦C. As shown in Fig. 1A, FITC- D-JNKI-1 pen- 280

etrated neurons and accumulated into the cell bodies. 281

In order to ascertain that the concentrations used were 282

not toxic we measured LDH leakage in the neuronal 283

media, an established index of neuronal death. Treat- 284

ment with 2, 4, and 6 �M D-JNKI-1 for 24 h did not 285

induce toxicity (Fig. 1B) as previously demonstrated 286

[13]. 287

The basal activity of JNK signaling pathway is high 288

in neurons. The efficiency of D-JNKI-1 to prevent 289

phosphorylation of c-Jun, a known JBD dependent 290

target of JNK, was assessed by Western blotting. As 291

expected, treatment with D-JNKI-1 led to inhibition of 292

c-Jun phosphorylation compared to control conditions 293

(Fig. 1C), [13, 30, 32, 40]. Densitometry quantifica- 294

tion confirmed a 28%, 90% and 95% decrease of c-Jun 295

phosphorylation following treatment with increasing 296

concentrations of D-JNKI-1 (2-4-6 �M respectively, 297

Fig. 1C,D), (p < 0.01 and p < 0.001, One Way ANOVA- 298

Tukey’s post-hoc Test) (± S.E.M.). We subsequently 299

analyzed the effect of D-JNKI-1 on activation of JNK. 300

As already demonstrated by Repici et al., [32] at 4 301

and 6 �M, D-JNKI-1 interferes with the two upstream 302

activators, MKK7 and MKK4, inducing a reduction of 303

the p-JNK/JNK ratio. We could indeed show that D- 304

JNKI-1 reduced the p-JNK/JNK ratio by 63 and 74% 305

at 4 and 6 �M respectively (Fig. 1C-E) (p < 0.01, One 306

way ANOVA- Tukey’s Test) (±S.E.M.). The control 307

TAT-peptide did not prevent JNK activity as previously 308

demonstrated (data not shown) [30, 31]. 309

We then analyzed the effect of D-JNKI-1 on tau 310

phosphorylation. p-tau and total tau levels were deter- 311

mined by Western blotting (Fig. 2). Treatment with 312

4 and 6 �M D-JNKI-1 for 24 h reduced p-tau (clone 313

AT8)/tau (clone tau-5) ratio by 40% and 75% respec- 314

tively (p < 0.05 and p < 0.01, One way ANOVA- 315

Tukey’s post-hoc test), (±S.E.M.), (Fig. 2A-B), while 316

the p-tau (clone AT8)/Actin ratio decreased by 70% 317

and 90% respectively (p < 0.01, One way ANOVA- 318

Tukey’s post-hoc test) (± S.E.M.), (Fig. 2A–C). Sim- 319

ilarly, treatment with 4 and 6 �M D-JNKI-1 reduced 320

p-tau S422/tau (clone tau-5) ratio by 54% and 51% 321

Page 5: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 5

B

C

D E

A

Fig. 1. D-JNKI-1 penetrates neurons successfully, it is not toxic and inhibits c-Jun and JNK phosphorylation in a concentration dependentmanner. A) Exposure of neurons to 2 �M FITC-labeled D-JNKI-1 for 24 h leads to accumulation of the peptide within the cell bodies (leftpanel: Bright Field images with FITC-D-JNKI-1, right panel FITC-D-JNKI-1 only, scale bar 50 �m). B) Incubation with D-JNKI-1 (2-4-6 �Mfor 24 h) is not toxic to neurons as indicated by the absence of LDH leakage (p > 0.05, One Way ANOVA). C) Representative Western blotsshowing reduction of p-c-Jun and p-JNK in rat cortical neurons treated with D-JNKI-1 2-4-6 �M for 24 h. D) Densitometric quantificationof p-c-Jun/c-Jun ratio revealed a 28%, 90% and 95% decrease after treatment with increasing concentrations of D-JNKI-1 (2-4-6 �M), andE) a reduction of p-JNK/JNK ratio by 63% and 74% at 4 and 6 �M. Loading control: Actin. (One Way ANOVA – Tukey’s Test – *p < 0.05,**p < 0.01) (± S.E.M.).

Page 6: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

6 C. Ploia et al. / JNK and Tau Phosphorylation in AD

A

B C

D E

F G

Fig. 2. JNK inhibition reduces tau phosphorylation in cortical neuronal cultures. A) Representative Western blots showing reduction of p-tauAT8, p-tau S422, and tau protein levels following treatment of neurons with increasing concentrations of D-JNKI-1 for 24 h. B) Densitometricquantification of p-tau AT8/Tau-5 ratio revealed a decrease (40–75%) after treatment with D-JNKI-1 (4–6 �M) and C) densitometric quantificationof p-tau AT8/Actin ratio revealed a 70–90% decrease after treatment with D-JNKI-1 (4–6 �M). D) D-JNKI-1 (4–6 �M) reduces p-tau S422/Tau-5ratio by 54–51% respectively and E) D-JNKI-1 (4–6 �M) reduces p-tau S422/Actin ratio by 72–93%. F) D-JNKI-1 treatment (4–6 �M) reducestotal levels of tau as revealed by the reduction of the Tau-5/Actin ratio (40–80%). Loading control: Actin. (One Way ANOVA – Tukey’s Test –*p < 0.05, **p < 0.01) (± S.E.M.). G) Real Time PCR revealed that tau mRNA levels are decreased by 30% after treatment with 4 �M D-JNKI-1(Student’s t-test - *p < 0.01) (± S.E.M.).

respectively (p < 0.01, One way ANOVA- Tukey’s322

Test) (±S.E.M.), (Fig. 2A–D). The p-tau S422/Actin323

ratio was also decreased by 72% and 93% following324

treatment with increasing concentrations of D-JNKI-325

1 (p < 0.01, One way ANOVA- Tukey’s post-hoc test)326

(±S.E.M.), (Fig. 2A–E).327

Interestingly, D-JNKI-1 treatment led to an over- 328

all reduction of tau protein levels, as shown by the 329

decrease of tau (clone tau-5)/Actin ratio by 40% 330

and 80% (4 �M and 6 �M D-JNKI-1) (p < 0.01, One 331

Way ANOVA- Tukey’s post-hoc test) (±S.E.M.), 332

(Fig. 2A–F). The effect of D-JNKI-1 on tau was fur- 333

Page 7: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 7

ther assessed by real-time reverse transcription PCR334

(RT-PCR). Treatment with D-JNKI-1 (4 �M) reduced335

tau mRNA levels by 30% (p < 0.01, Student’s t-test)336

(±S.E.M.), (Fig. 2G). Thus, JNK regulates tau at the337

transcriptional level.338

Immunostaining confirmed the effect of D-JNKI-1339

on p-tau AT8 levels. Following D-JNKI-1 treatment340

(4 �M- 24 h) we observed a clear decrease in tau phos-341

phorylation. P-tau was diffusely distributed along the342

axons in control conditions and D-JNKI-1 induced the343

formation of p-tau spots along the neurites (Fig. 3-344

A). The effect of D-JNKI-1 on p-tau AT8 was specific345

and did not affect the neuronal cytoskeleton as demon-346

strated by phalloidin staining, a marker of the F-actin347

cytoskeleton (Fig. 3B).348

Inhibition of the JNK pathway prevents tau349

phosphorylation in transgenic AD mice350

(TgCRND8)351

TgCRND8 mice are characterized by numerous A�352

deposits (3–5 months of age) in the hippocampus,353

cortex and other brain areas, as well as hyperphospho- 354

rylated tau (7–12 months of age) and impairment in 355

learning and memory functions [16, 39, 41]. Moreover, 356

in the neocortex and hippocampus of this transgenic 357

mouse model (7 and 12 months of age) activity of JNK 358

is increased in respect to Wt controls, and p-JNK is 359

localized within amyloid plaques [16]. 360

Herein we investigated if a chronic treatment (4 361

months) with D-JNKI-1 (22 mg/kg) prevents tau phos- 362

phorylation in TgCRND8 mice. Previous reports have 363

extensively demonstrated that in adult mice the peptide 364

is able to cross the blood-brain barrier, penetrate the 365

cerebral cortex within 1 h and 30 min after an intraperi- 366

toneal injection [30–32, 40], and remain stable in the 367

brain parenchyma for 3 weeks. Consequently, D-JNKI- 368

1 was injected in TgCRND8 mice and wt 129 sv mice 369

every 21 days intraperitoneally, starting from 4 to reach 370

8 months of age. 371

C-Jun was activated in TG-CTR mice compared to 372

WT-CTR mice both in the cortex (Fig. 4A,B) and hip- 373

pocampus (Fig. 4E,F) demonstrating a role of JNK 374

pathway in AD pathology (p < 0.05, two-way ANOVA, 375

A

B

Fig. 3. D-JNKI-1 regulates p-tau AT8 distribution. Representative immunostaining images show that A) treatment with 4 �M D-JNKI-1 for 24 hleads to a decrease in AT8 immunoreactivity (right panel: green p-tau AT8 – blue Hoechst, scale bar 50 �m). B) Neurons were stained for F-actin(phalloidin). Representative photomicrographs show that D-JNKI-1 does not affect the F-actin cytoskeleton and the neuronal architecture. Allexperiments were repeated at least six times (CTR = control conditions).

Page 8: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

8 C. Ploia et al. / JNK and Tau Phosphorylation in AD

CEREBRAL CORTEX

HIPPOCAMPUS

A B

C D

E F

G

HI

Fig. 4. D-JNKI-1 treatment inhibits p-c-Jun expression and reduces p-tau AT8 in the cortex and hippocampus of TgCRND8 mice. CerebralCortex: A) Representative Western blots of p-c-Jun, c-Jun, p-tau AT8, and Tau-5 from the cortex of TG-CTR, TG-D-JNKI-1, WT-CTR, andWT-D-JNKI-1 mice. B) In TG-CTR mice c-Jun activity is upregulated in comparison to WT-CTR. D-JNKI-1 reduced the p-c-Jun/c-Jun ratioby 60%. C) In TG-CTR mice the p-tau AT8/Tau5 ratio is increased in comparison to WT-CTR mice. Chronic treatment with D-JNKI-1 reducesp-tau AT8/Tau5 ratio in TG mice to WT-CTR levels. D) Total levels of tau remain unchanged following treatment with D-JNKI-1 as shown by theTau-5/Actin ratio (Two Way ANOVA – Tukey’s Test – *p < 0.05) (±S.E.M.). Hippocampus: E) Representative Western blots of p-c-Jun, c-Jun,p-tau AT8, and Tau-5 of TG-CTR, TG-D-JNKI-1, WT-CTR, and WT-D-JNKI-1. F) In TG-CTR mice c-Jun activity is upregulated in comparisonto WT-CTR. D-JNKI-1 reduces the p-c-Jun/c-Jun ratio by 60%. G) p-tau/tau is significantly increased in Tg-CTR compared to WT-CTR mice.D-JNKI-1 chronic treatment leads to an 85% reduction of p-tau AT8/Tau-5 ratio. (Two Way ANOVA – Tukey’s Test – *p < 0.05, **p < 0.01)(±S.E.M.). H) Representative Western blots of p-tau S422 in TG-CTR and WT-CTR mice. I) Densitometry analysis reveals no changes at p-tauS422 levels between TG-CTR and WT-CTR. Loading control: Actin. (Student’s t-test) (±S.E.M.). (TG-CTR = not treated TgCRND8 mice,TG-DJNKI-1 = D-JNKI-1 treated TgCRND8 mice, WT-CTR = not treated Wt mice, WT-DJ = D-JNKI-1 treated Wt mice).

Page 9: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 9

A

B

C

Fig. 5. Lack of toxicity and successful penetration by D-JNKI-1 in human fibroblasts. A) Exposure of fibroblasts to 50 � M FITC-labeledD-JNKI-1 for 24 h leads to accumulation of the peptide within the cell (left panel: Bright Field images with FITC-D-JNKI-1, right panel FITC-D-JNKI-1 only, scale bar 50 �m). B) Incubation with D-JNKI-1 is not toxic as indicated by the absence of LDH leakage (One way ANOVA).C) Cells were stained for F-actin (phalloidin). Representative photomicrographs show that D-JNKI-1 does not affect the F-actin cytoskeletonof fibroblasts from healthy and/or AD subjects. (CTR = control conditions, NA = not affected subject, AD = AD subject).

Page 10: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

10 C. Ploia et al. / JNK and Tau Phosphorylation in AD

Tukey’s post-hoc test). We subsequently assessed the376

efficiency of D-JNKI-1 treatment in TgCRND8 mice377

by analyzing the effect of the peptide on the phospho-378

rylation of c-Jun. D-JNKI-1 reduced the p-c-Jun/c-Jun379

ratio by 60% both in the cortex (Fig. 4A,B) and the380

hippocampus (Fig. 4E,F) (p < 0.05, two-way ANOVA,381

Tukey’s post-hoc test).382

Levels of p-tau (clone AT8) were examined by West-383

ern blotting in the cortex and in the hippocampus of384

Tg-D-JNKI-1 mice in comparison to untreated and to385

WT (treated and untreated) littermates. Total tau levels386

were detected by the anti-tau clone tau-5 previously387

used in the same model by Bellucci et al. [16]. As 388

opposed to the data we obtained in vitro, D-JNKI-1 389

had no effect on total tau expression as indicated by 390

tau (clone tau-5)/Actin ratio in cortex (Fig. 4A–D). 391

As expected [16], p-tau (clone AT8)/tau (clone tau- 392

5) was significantly increased in the cortex of TG-CTR 393

compared to WT-CTR mice (85%, p < 0.01, two-way 394

ANOVA, Tukey’s post-hoc test) (Fig. 4A–C). Con- 395

versely, p-tau AT8 levels in TG-D-JNKI-1 mice were 396

comparable to those detected in WT-CTR animals. 397

More specifically, in cortex homogenates of TG-D- 398

JNKI-1 mice the p-tau AT8/tau (clone tau-5) ratio 399

A

B C D

E F

Fig. 6. D-JNKI-1 reduces tau phosphorylation in human fibroblasts. A) D-JNKI-1 reduces AT8 staining in fibroblasts from AD patients, butnot in NA subjects (green p-tau AT8 – blue Hoechst, scale bar 50 �m) (CTR = control conditions, NA = not affected subject, AD = AD subject).B) Representative Western blots of p-c-Jun, c-Jun, p-tau AT8, p-tau S422, and Tau-5 in human AD fibroblasts untreated (CTR) and treatedwith increasing concentrations of D-JNKI-1 (10–50 �M) for 24 h. C) Treatment with 50 �M D-JNKI-1 reduces p-c-Jun/c-Jun ratio by 23%.D) Treatment with 50 �M D-JNKI-1 reduces p-tau AT8/Tau-5 ratio by 50%, and E) D-JNKI-1 (50 �M) also leads to a 38% reduction of p-tauS422/Tau-5 ratio. F) Total tau is not affected by treatment with D-JNKI-1. Loading control: Actin. (One Way ANOVA – Tukey’s Test – *p < 0.05)(± S.E.M.).

Page 11: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 11

was significantly lower in comparison to TG–CTR400

mice (–85%, p < 0.05, two-way ANOVA, Tukey’s post-401

hoc test) (Fig. 4A–C). Likewise, in the hippocampus402

the p-tau AT8/tau (clone tau-5) ratio was significantly403

increased in TG-CTR mice compared to WT-CTR404

mice (95%, p < 0.01, two-way ANOVA, Tukey’s post-405

hoc test) and decreased in TG-D-JNKI-1 compared406

to untreated TG-CTR mice (–85% p < 0.05, two-way407

ANOVA, Tukey’s post-hoc test) (Fig. 4E–G).408

We then examined the phosphorylation of the S422409

tau site in Tg-CTR compared to WT-CTR mice. Since410

we could not observe changes in tau phosphorylation411

at S422 between Tg and Wt animals (Fig. 4H,I) we did412

not process D-JNKI-1 treated mice (p > 0.05, Student’s413

t-test).414

Therapeutic efficacy of D-JNKI-1 was tested in415

primary fibroblasts from AD patients416

The effect of JNK inhibition on tau phosphoryla-417

tion was tested in human fibroblasts from AD patients418

and in not affected subjects (NA). We first tested pen-419

etration and toxicity of D-JNKI-1 on these cells that420

are defined as a resistant cell line to cell permeable421

peptides (CPPs) [42].422

Exposure to 50 �M D-JNKI-1 resulted in 100% pen-423

etration after 24 h (Fig. 5A). Toxicity was assessed424

by measuring LDH release in the culture media: no425

signs of toxicity were detected even at doses as high426

as 150 �M (Fig. 5B). Treatment of fibroblasts with427

D-JNKI-1 did not affect the cytoskeletal structure of428

fibroblasts, as demonstrated by phalloidin staining429

(Fig. 5C).430

The inhibitory effect of D-JNKI-1 on the p-c-Jun/c-431

Jun ratio in this cellular model was tested by Western432

blotting. 50 �M D-JNKI-1 reduced p-c-Jun/c-Jun ratio433

by 23% after 24 h (p < 0.05, One Way ANOVA-434

Tukey’s post-hoc test) (±S.E.M.) (Fig. 6B,C).435

We then evaluated the effect of D-JNKI-1 treatment436

on p-tau AT8 and tau expression and distribution. It437

is known that in cycling cells, like fibroblasts, tau is438

also localized in the nucleus and along mitotic chro-439

mosomes to regulate microtubules dynamic instability440

required for the rapid mitotic events [43]. Interest-441

ingly, p-tau AT8 distribution was quite different in NA442

healthy subjects and AD patients, suggesting a mislo-443

cation of the protein in pathological conditions. In NA444

fibroblasts p-tau AT8 staining was both nuclear and445

cytoplasmic [43], while in AD fibroblasts the phos-446

phorylated protein was mainly cytoplasmic (Fig. 6A).447

D-JNKI-1 treatment reduced p-tau AT8 in fibroblasts448

from AD patients. On the other hand the strong reduc-449

tion of p-tau AT8, observed in AD patients, was less 450

evident in D-JNKI-1 treated fibroblasts from NA sub- 451

jects suggesting a major effect of D-JNKI-1 peptide on 452

p-tau AT8 cytoplasmic pool (Fig. 6A). 453

To better investigate the effect of D-JNKI-1 on AD 454

fibroblasts we quantified the p-tau/tau ratio by Western 455

blotting. Treatment with D-JNKI-1, 50 �M, reduced 456

the p-tau (clone AT8)/tau (clone tau-5) ratio by 50% 457

(Fig. 6B–D) (p < 0.05, One Way ANOVA- Tukey’s 458

Test) (±S.E.M.). Moreover p-tau S422/tau (clone tau- 459

5) ratio was reduced by 38% (Fig. 6B-E) (p < 0.05, One 460

way ANOVA- Tukey’s Test) (±S.E.M.) (p < 0.05 and 461

p < 0.01, One Way ANOVA- Tukey’s Test) (±S.E.M.). 462

D-JNKI-1 did not affect tau (clone tau-5)/Actin ratio 463

indicating that the peptide does not have any effects on 464

total tau expression (Fig. 4B,F). 465

DISCUSSION 466

Neurofibrillary tangles composed of hyperphospho- 467

rylated tau are a major hallmark of AD. In the present 468

study we investigated the phosphorylation of tau medi- 469

ated by JNK at S202/T205 [44], recognized by the AT8 470

antibody, and S422 [26, 27]. As previously reported, 471

phosphorylation at S202/T205 is an early event in tau 472

hyperphosphorylation [45–47] and is associated with 473

an early stage of the pathology in human AD brains and 474

tauopathies [23, 25, 47]. Similarly, phosphorylation at 475

S422 is seen in AD brains but not in normal brains 476

[48]. However, this phosphorylation is not an early 477

event in tau pathology since staining for p-tau S422 is 478

rare in pretangle neurons [49]. Hyperphosphorylation 479

of tau has been linked extensively to neurodegenera- 480

tion [50, 51], and it involves a wide range of kinases. 481

Some of the kinases responsible of tau phosphory- 482

lation, Cdk-5, GSK-3�, and JNK [27, 52–57] are 483

also responsible for A�PP phosphorylation on T668 484

[11, 58–60], which is of crucial importance in �- 485

cleavage and consequent production of A� oligomers 486

[61]. These kinases lie at the intersection between 487

the two major histopathological hallmarks of AD a 488

feature that renders them potential targets to inhibit 489

the post-transcriptional modifications of two important 490

AD proteins. 491

Among the kinases responsible of both tau and 492

A�PP phosphorylation JNK is particularly intrigu- 493

ing. Increased levels of JNK have been observed in 494

AD brains while p-JNK co-localizes with p-tau in 495

neurons of AD patients [4, 62]. Our aim was to inves- 496

tigate the role of JNK in tau phosphorylation. To the 497

best of our knowledge almost all works reported have 498

Page 12: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

12 C. Ploia et al. / JNK and Tau Phosphorylation in AD

investigated tau phosphorylation using either in vitro499

models like cell free systems for kinase reactions, tran-500

siently transfected over-expressing cell systems [63,501

64], or transgenic mouse models where kinases have502

been inducibly/conditionally expressed (e.g., [65, 66]).503

Due to the importance of tau phosphorylation in AD504

neurodegeneration further and more in depth studies505

are needed to better understand the mechanisms that506

regulate this process. For this purpose we used the507

most specific JNK inhibitor peptide D-JNKI-1 [30]508

and investigated the effect of JNK inhibition in both509

physiological conditions as well as in two AD mod-510

els. The D-JNKI-1 peptide has been successfully used511

and inhibits phosphorylation of A�PP in vitro [13, 14],512

as well as in vivo (Sclip et al., submitted manuscript).513

We demonstrated that, in all three models, JNK inhi-514

bition strongly reduces tau hyperphosphorylation at515

S202/T205.516

The effect of the D-JNKI-1 inhibitor on tau hyper-517

phosphorylation was initially tested in rat cortical518

neurons. Our findings demonstrate that in physiologi-519

cal conditions JNK is involved in tau phosphorylation520

both at S202/T205 and S422. Of note, D-JNKI-1 also521

exerted an effect on total tau protein as well as mRNA522

levels. Although this issue needs further investigation,523

it should be noted that this effect is restricted to control524

cortical neurons and it is absent in the two other models525

studied (TgCRND8 mice and human fibroblasts).526

Subsequently we studied phosphorylation of tau by527

JNK in an in vivo AD mouse model (TgCRND8) fol-528

lowing chronic treatment with D-JNKI-1. This model529

offers a powerful tool to study the early events occur-530

ring in AD pathology and to analyze the intracellular531

mechanisms of AD neurodegeneration in vivo. The532

TgCRND8 mice over-express the A�PP with both533

Indiana and Swedish mutations and develop the AD534

phenotype very quickly [39]. In agreement with others535

[16, 39], we could show that TgCRND8 brains pre-536

sented hyperphosphorylation of tau in the cortex and537

hippocampus at 7 months of age and after the for-538

mation of plaques (plaques at 3–5 months). Of note,539

chronic treatment with D-JNKI-1 completely reverted540

the pathological phosphorylation of tau (clone AT8)541

in the cortex and hippocampus of TgCRND8 mice,542

and importantly it did not interfere with the total lev-543

els of tau. Although one could argue that the effect544

of JNK inhibition on tau phosphorylation observed545

in TgCRND8 mice could result from altered A�PP546

processing the results from the in vitro data would sug-547

gest that the effect is more direct. Further experiments,548

beyond the scope of this work would be required to549

confirm a direct interaction between JNK and tau.550

Interestingly, we did not observe alterations of p-tau 551

S422 levels in the TgCRND8 mice compared to Wt 552

mice. Overall the results obtained on TgCRND8 brains 553

demonstrated the importance of the JNK pathway in 554

the early stage of tau hyperphosphorylation in vivo. 555

Finally we analyzed the effect of JNK inhibition in 556

human AD fibroblasts, a model that is commonly used 557

to elucidate the primary pathophysiological mecha- 558

nisms leading to AD and to avoid the variability that 559

often derives from post-mortem studies. Treatment 560

with D-JNKI-1 (50 �M) led to a clear reduction of 561

the p-tau (clone AT8)/tau (Clone tau-5) and p-tau 562

S422/tau (Clone tau-5) ratio in all AD human fibrob- 563

lasts. Notably, we could show that D-JNKI-1 reduced 564

cytoplasmic AT8 staining in AD subjects but not in 565

healthy control (not affected) fibroblasts. Such findings 566

are indicative of a differential role of JNK in patholog- 567

ical and control conditions suggesting the involvement 568

of a different array of kinases acting on tau protein or 569

alternatively, a differential spatial distribution of p-tau 570

pool in the molecular cascade leading to AD. Whether 571

those differences are confined to the peripheral tissue 572

is worth further investigation. Nevertheless our data 573

demonstrate the similarity between human fibroblasts 574

and mouse AD pathology and support the use of this 575

peripheral tissue for the study of AD. 576

Altogether our findings underline the important 577

contribution of JNK on tau physiological but most 578

importantly pathological phosphorylation. The reduc- 579

tion of tau phosphorylation has been proposed as a 580

therapeutic approach for AD [67]. Treatment with D- 581

JNKI-1 clearly demonstrates the importance of the 582

JNK pathway on tau pathological phosphorylation and 583

indicates JNK as an innovative therapeutic target in AD 584

and other neurodegenerative disorders. 585

ACKNOWLEDGMENTS 586

This study was supported by the Marie Curie 587

Industry-Academia Partnerships and Pathways (IAPP) 588

cPADS; San Paolo 2008-2437; CARIPLO 2009-2425. 589

Ricerca Corrente, Italian Ministry of Health; Fon- 590

dazione CARIPLO 2009-2633. There are no actual or 591

potential conflicts of interest. We thank Dr. Feligioni 592

for his scientific advice. 593

Written informed consent was obtained from all 594

subjects or, where appropriate, their caregivers, fol- 595

lowing the procedures approved by the hospital local 596

ethical committee. All experimental procedures on ani- 597

mals were performed in accordance with the European 598

Communities Council Directive of 24 November 1986 599

Page 13: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 13

(86/609/EEC) and all efforts were made to minimize600

animal suffering.601

Authors’ disclosures available online (http://www.j-602

alz.com/disclosures/view.php?id=846).603

REFERENCES604

[1] Weston CR, Davis RJ (2007) The JNK signal transduction605

pathway. Curr Opin Cell Biol 19, 142-149.606

[2] Borsello T, Forloni G (2007) JNK signalling: a possible target607

to prevent neurodegeneration. Curr Pharm Des 13, 1875-608

1886.609

[3] Zhu X, Castellani RJ, Takeda A, Nunomura A, Atwood CS,610

Perry G, Smith MA (2001) Differential activation of neuronal611

ERK, JNK/SAPK and p38 in Alzheimer disease: the ‘two hit’612

hypothesis. Mech Ageing Dev 123, 39-46.613

[4] Zhu X, Raina AK, Rottkamp CA, Aliev G, Perry G, Boux H,614

Smith MA (2001) Activation and redistribution of c-jun N-615

terminal kinase/stress activated protein kinase in degenerating616

neurons in Alzheimer’s disease. J Neurochem 76, 435-441.617

[5] Shoji M, Iwakami N, Takeuchi S, Waragai M, Suzuki M,618

Kanazawa I, Lippa CF, Ono S, Okazawa H (2000) JNK619

activation is associated with intracellular beta-amyloid accu-620

mulation. Brain Res Mol Brain Res 85, 221-233.621

[6] Sato S, Tatebayashi Y, Akagi T, Chui DH, Murayama M,622

Miyasaka T, Planel E, Tanemura K, Sun X, Hashikawa623

T, Yoshioka K, Ishiguro K, Takashima A (2002) Aberrant624

tau phosphorylation by glycogen synthase kinase-3 beta and625

JNK3 induces oligomeric tau fibrils in COS-7 cells. J Biol626

Chem 277, 42060-42065.627

[7] Thakur A, Wang X, Siedlak SL, Perry G, Smith MA, Zhu X628

(2007) c-Jun phosphorylation in Alzheimer disease. J Neu-629

rosci Res 85, 1668-1673.630

[8] Zhu X, Ogawa O, Wang Y, Perry G, Smith MA (2003)631

JKK1, an upstream activator of JNK/SAPK, is activated in632

Alzheimer’s disease. J Neurochem 85, 87-93.633

[9] Morishima Y, Gotoh Y, Zieg J, Barrett T, Takano H, Flavell R,634

Davis RJ, Shirasaki Y, Greenberg ME (2001) Beta-amyloid635

induces neuronal apoptosis via a mechanism that involves the636

c-Jun N-terminal kinase pathway and the induction of Fas637

ligand. J Neurosci 21, 7551-7560.638

[10] Minogue AM, Schmid AW, Fogarty MP, Moore AC, Campbell639

VA, Herron CE, Lynch MA (2003) Activation of the c-Jun640

N-terminal kinase signaling cascade mediates the effect of641

amyloid-beta on long term potentiation and cell death in hip-642

pocampus: a role for interleukin-1 beta? J Biol Chem 278,643

27971-27980.644

[11] Muresan Z, Muresan V (2005) c-Jun NH2-terminal kinase-645

interacting protein-3 facilitates phosphorylation and controls646

localization of amyloid-beta precursor protein. J Neurosci 25,647

3741-3751.648

[12] Vingtdeux V, Hamdane M, Gompel M, Begard S, Drobecq649

H, Ghestem A, Grosjean ME, Kostanjevecki V, Grognet P,650

Vanmechelen E, Buee L, Delacourte A, Sergeant N (2005)651

Phosphorylation of amyloid precursor carboxy-terminal652

fragments enhances their processing by a gamma-secretase-653

dependent mechanism. Neurobiol Dis 20, 625-637.654

[13] Colombo A, Repici M, Pesaresi M, Santambrogio S, Forloni655

G, Borsello T (2007) The TAT-JNK inhibitor peptide inter-656

feres with beta amyloid protein stability. Cell Death Differ657

14, 1845-1848.658

[14] Colombo A, Bastone A, Ploia C, Sclip A, Salmona M,659

Forloni G, Borsello T (2009) JNK regulates APP cleavage and660

degradation in a model of Alzheimer’s disease. Neurobiol Dis 661

33, 518-525. 662

[15] Ferrer I, Blanco R, Carmona M, Puig B (2001) Phospho- 663

rylated mitogen-activated protein kinase (MAPK/ERK-P), 664

protein kinase of 38 kDa (p38-P), stress-activated protein 665

kinase (SAPK/JNK-P), and calcium/calmodulin-dependent 666

kinase II (CaM kinase II) are differentially expressed in tau 667

deposits in neurons and glial cells in tauopathies. J Neural 668

Transm 108, 1397-1415. 669

[16] Bellucci A, Rosi MC, Grossi C, Fiorentini A, Luccarini I, 670

Casamenti F (2007) Abnormal processing of tau in the brain 671

of aged TgCRND8 mice. Neurobiol Dis 27, 328-338. 672

[17] Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, Gant 673

DJ, Chen PP, Hudspeth B, Chen C, Zhao Y, Vinters HV, 674

Frautschy SA, Cole GM (2009) Beta-amyloid oligomers 675

induce phosphorylation of tau and inactivation of insulin 676

receptor substrate via c-Jun N-terminal kinase signaling: sup- 677

pression by omega-3 fatty acids and curcumin. J Neurosci 29, 678

9078-9089. 679

[18] Vogel J, Anand VS, Ludwig B, Nawoschik S, Dunlop J, 680

Braithwaite SP (2009) The JNK pathway amplifies and drives 681

subcellular changes in tau phosphorylation. Neuropharmacol- 682

ogy 57, 539-550. 683

[19] Hardy J, Selkoe DJ (2002) The amyloid hypothesis of 684

Alzheimer’s disease: progress and problems on the road to 685

therapeutics. Science 297, 353-356. 686

[20] Lahiri DK, Farlow MR, Sambamurti K, Greig NH, Giacobini 687

E, Schneider LS (2003) A critical analysis of new molecular 688

targets and strategies for drug developments in Alzheimer’s 689

disease. Curr Drug Targets 4, 97-112. 690

[21] Noble W, Planel E, Zehr C, Olm V, Meyerson J, Suleman 691

F, Gaynor K, Wang L, LaFrancois J, Feinstein B, Burns M, 692

Krishnamurthy P, Wen Y, Bhat R, Lewis J, Dickson D, Duff 693

K (2005) Inhibition of glycogen synthase kinase-3 by lithium 694

correlates with reduced tauopathy and degeneration in vivo. 695

Proc Natl Acad Sci U S A 102, 6990-6995. 696

[22] Sereno L, Coma M, Rodriguez M, Sanchez-Ferrer P, Sanchez 697

MB, Gich I, Agullo JM, Perez M, Avila J, Guardia-Laguarta 698

C, Clarimon J, Lleo A, Gomez-Isla T (2009) A novel GSK- 699

3 beta inhibitor reduces Alzheimer’s pathology and rescues 700

neuronal loss in vivo. Neurobiol Dis 35, 359-367. 701

[23] Su JH, Cummings BJ, Cotman CW (1994) Early phosphory- 702

lation of tau in Alzheimer’s disease occurs at Ser-202 and is 703

preferentially located within neurites. Neuroreport 5, 2358- 704

2362. 705

[24] Augustinack JC, Schneider A, Mandelkow EM, Hyman BT 706

(2002) Specific tau phosphorylation sites correlate with sever- 707

ity of neuronal cytopathology in Alzheimer’s disease. Acta 708

Neuropathol 103, 26-35. 709

[25] Luna-Munoz J, Chavez-Macias L, Garcia-Sierra F, Mena 710

R (2007) Earliest stages of tau conformational changes are 711

related to the appearance of a sequence of specific phospho- 712

dependent tau epitopes in Alzheimer’s disease. J Alzheimers 713

Dis 12, 365-375. 714

[26] Yoshida H, Hastie CJ, McLauchlan H, Cohen P, Goedert M 715

(2004) Phosphorylation of microtubule-associated protein tau 716

by isoforms of c-Jun N-terminal kinase (JNK). J Neurochem 717

90, 352-358. 718

[27] Reynolds CH, Utton MA, Gibb GM, Yates A, Anderton 719

BH (1997) Stress-activated protein kinase/c-jun N-terminal 720

kinase phosphorylates tau protein. J Neurochem 68, 1736- 721

1744. 722

[28] Grueninger F, Bohrmann B, Czech C, Ballard TM, Frey JR, 723

Weidensteiner C, von Kienlin M, Ozmen L, Phosphorylation 724

of Tau at S422 is enhanced by Abeta in TauPS2APP triple 725

transgenic mice. Neurobiol Dis 37, 294-306. 726

Page 14: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

14 C. Ploia et al. / JNK and Tau Phosphorylation in AD

[29] Malaplate-Armand C, Desbene C, Pillot T, Olivier JL (2009)727

[Biomarkers for early diagnosis of Alzheimer’s disease: cur-728

rent update and future directions]. Rev Neurol (Paris) 165,729

511-520.730

[30] Borsello T, Clarke PG, Hirt L, Vercelli A, Repici M,731

Schorderet DF, Bogousslavsky J, Bonny C (2003) A peptide732

inhibitor of c-Jun N-terminal kinase protects against excito-733

toxicity and cerebral ischemia. Nat Med 9, 1180-1186.734

[31] Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF735

(2001) Cell-permeable peptide inhibitors of JNK: novel736

blockers of beta-cell death. Diabetes 50, 77-82.737

[32] Repici M, Mare L, Colombo A, Ploia C, Sclip A, Bonny738

C, Nicod P, Salmona M, Borsello T (2009) c-Jun N-739

terminal kinase binding domain-dependent phosphorylation740

of mitogen-activated protein kinase kinase 4 and mitogen-741

activated protein kinase kinase 7 and balancing cross-talk742

between c-Jun N-terminal kinase and extracellular signal-743

regulated kinase pathways in cortical neurons. Neuroscience744

159, 94-103.745

[33] Antoniou X, Sclip A, Ploia C, Colombo A, Moroy G, Borsello746

T, JNK contributes to Hif-1 alpha regulation in hypoxic neu-747

rons. Molecules 15, 114-127.748

[34] Lesuisse C, Martin LJ (2002) Immature and mature corti-749

cal neurons engage different apoptotic mechanisms involving750

caspase-3 and the mitogen-activated protein kinase pathway.751

J Cereb Blood Flow Metab 22, 935-950.752

[35] Li JH, Wang YH, Wolfe BB, Krueger KE, Corsi L, Stocca753

G, Vicini S (1998) Developmental changes in localization754

of NMDA receptor subunits in primary cultures of cortical755

neurons. Eur J Neurosci 10, 1704-1715.756

[36] Benussi L, Govoni S, Gasparini L, Binetti G, Trabucchi M,757

Bianchetti A, Racchi M (1998) Specific role for protein kinase758

C alpha in the constitutive and regulated secretion of amyloid759

precursor protein in human skin fibroblasts. Neurosci Lett 240,760

97-101.761

[37] Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy762

N, De Paepe A, Speleman F (2002) Accurate normalization763

of real-time quantitative RT-PCR data by geometric aver-764

aging of multiple internal control genes. Genome Biol 3,765

RESEARCH0034.766

[38] Zhao R, Zhang S, Xu Z, Ju L, Lu D, Yao G (2007) Studying767

gene expression profile of rat neuron exposed to 1800 MHz768

radiofrequency electromagnetic fields with cDNA microas-769

say. Toxicology 235, 167-175.770

[39] Chishti MA, Yang DS, Janus C, Phinney AL, Horne P, Pearson771

J, Strome R, Zuker N, Loukides J, French J, Turner S, Lozza772

G, Grilli M, Kunicki S, Morissette C, Paquette J, Gervais773

F, Bergeron C, Fraser PE, Carlson GA, George-Hyslop PS,774

Westaway D (2001) Early-onset amyloid deposition and cog-775

nitive deficits in transgenic mice expressing a double mutant776

form of amyloid precursor protein 695. J Biol Chem 276,777

21562-21570.778

[40] Repici M, Centeno C, Tomasi S, Forloni G, Bonny C, Vercelli779

A, Borsello T (2007) Time-course of c-Jun N-terminal kinase780

activation after cerebral ischemia and effect of D-JNKI1 on781

c-Jun and caspase-3 activation. Neuroscience 150, 40-49.782

[41] Bellucci A, Luccarini I, Scali C, Prosperi C, Giovannini MG,783

Pepeu G, Casamenti F (2006) Cholinergic dysfunction, neu-784

ronal damage and axonal loss in TgCRND8 mice. Neurobiol785

Dis 23, 260-272.786

[42] Cardozo AK, Buchillier V, Mathieu M, Chen J, Ortis F,787

Ladriere L, Allaman-Pillet N, Poirot O, Kellenberger S,788

Beckmann JS, Eizirik DL, Bonny C, Maurer F (2007)789

Cell-permeable peptides induce dose- and length-dependent790

cytotoxic effects. Biochim Biophys Acta 1768, 2222-2234.791

[43] Rossi G, Dalpra L, Crosti F, Lissoni S, Sciacca FL, Catania 792

M, Di Fede G, Mangieri M, Giaccone G, Croci D, Tagliavini 793

F (2008) A new function of microtubule-associated protein 794

tau: involvement in chromosome stability. Cell Cycle 7, 1788- 795

1794. 796

[44] Goedert M, Jakes R, Vanmechelen E (1995) Monoclonal anti- 797

body AT8 recognises tau protein phosphorylated at both serine 798

202 and threonine 205. Neurosci Lett 189, 167-169. 799

[45] Kimura T, Ono T, Takamatsu J, Yamamoto H, Ikegami K, 800

Kondo A, Hasegawa M, Ihara Y, Miyamoto E, Miyakawa T 801

(1996) Sequential changes of tau-site-specific phosphoryla- 802

tion during development of paired helical filaments. Dementia 803

7, 177-181. 804

[46] Lauckner J, Frey P, Geula C (2003) Comparative distribution 805

of tau phosphorylated at Ser262 in pre-tangles and tangles. 806

Neurobiol Aging 24, 767-776. 807

[47] Maurage CA, Sergeant N, Ruchoux MM, Hauw JJ, Delacourte 808

A (2003) Phosphorylated serine 199 of microtubule- 809

associated protein tau is a neuronal epitope abundantly 810

expressed in youth and an early marker of tau pathology. Acta 811

Neuropathol 105, 89-97. 812

[48] Bussiere T, Hof PR, Mailliot C, Brown CD, Caillet-Boudin 813

ML, Perl DP, Buee L, Delacourte A (1999) Phosphorylated 814

serine422 on tau proteins is a pathological epitope found in 815

several diseases with neurofibrillary degeneration. Acta Neu- 816

ropathol 97, 221-230. 817

[49] Johnson GV, Stoothoff WH (2004) Tau phosphorylation in 818

neuronal cell function and dysfunction. J Cell Sci 117, 5721- 819

5729. 820

[50] Ashe KH (2007) A tale about tau. N Engl J Med 357, 933-935. 821

[51] Lee VM, Trojanowski JQ (2001) Transgenic mouse models 822

of tauopathies: prospects for animal models of Pick’s disease. 823

Neurology 56, S26-S30. 824

[52] Buee-Scherrer V, Goedert M (2002) Phosphorylation of 825

microtubule-associated protein tau by stress-activated protein 826

kinases in intact cells. FEBS Lett 515, 151-154. 827

[53] Gallo JM, Hanger DP, Twist EC, Kosik KS, Anderton BH 828

(1992) Expression and phosphorylation of a three-repeat iso- 829

form of tau in transfected non-neuronal cells. Biochem J 286 830

(Pt 2), 399-404. 831

[54] Mandelkow EM, Drewes G, Biernat J, Gustke N, Van 832

Lint J, Vandenheede JR, Mandelkow E (1992) Glyco- 833

gen synthase kinase-3 and the Alzheimer-like state of 834

microtubule-associated protein tau. FEBS Lett 314, 315-321. 835

[55] Kobayashi S, Ishiguro K, Omori A, Takamatsu M, Arioka 836

M, Imahori K, Uchida T (1993) A cdc2-related kinase 837

PSSALRE/cdk5 is homologous with the 30 kDa subunit of tau 838

protein kinase II, a proline-directed protein kinase associated 839

with microtubule. FEBS Lett 335, 171-175. 840

[56] Paudel HK, Lew J, Ali Z, Wang JH (1993) Brain proline-direc- 841

ted protein kinase phosphorylates tau on sites that are abnor- 842

mally phosphorylated in tau associated with Alzheimer’s 843

paired helical filaments. J Biol Chem 268, 23512-23518. 844

[57] Reynolds CH, Nebreda AR, Gibb GM, Utton MA, Anderton 845

BH (1997) Reactivating kinase/p38 phosphorylates tau 846

protein in vitro. J Neurochem 69, 191-198. 847

[58] Aplin AE, Gibb GM, Jacobsen JS, Gallo JM, Anderton BH 848

(1996) In vitro phosphorylation of the cytoplasmic domain of 849

the amyloid precursor protein by glycogen synthase kinase- 850

3 beta. J Neurochem 67, 699-707. 851

[59] Iijima K, Ando K, Takeda S, Satoh Y, Seki T, Itohara S, 852

Greengard P, Kirino Y, Nairn AC, Suzuki T (2000) Neuron- 853

specific phosphorylation of Alzheimer’s beta-amyloid precur- 854

sor protein by cyclin-dependent kinase 5. J Neurochem 75, 855

1085-1091. 856

Page 15: IOS Press JNK Plays a Key Role in Tau Hyperphosphorylation ... · JNK Plays a Key Role in Tau Hyperphosphorylation in Alzheimer’s ... preservefunctionality.TheinactivepeptideD-TATwas

Unc

orre

cted

Aut

hor P

roof

C. Ploia et al. / JNK and Tau Phosphorylation in AD 15

[60] Standen CL, Brownlees J, Grierson AJ, Kesavapany S, Lau857

KF, McLoughlin DM, Miller CC (2001) Phosphorylation of858

thr(668) in the cytoplasmic domain of the Alzheimer’s disease859

amyloid precursor protein by stress-activated protein kinase860

1 b (Jun N-terminal kinase-3). J Neurochem 76, 316-320.861

[61] Lee MS, Kao SC, Lemere CA, Xia W, Tseng HC, Zhou Y,862

Neve R, Ahlijanian MK, Tsai LH (2003) APP processing is863

regulated by cytoplasmic phosphorylation. J Cell Biol 163,864

83-95.865

[62] Mohit AA, Martin JH, Miller CA (1995) p493F12 kinase: a866

novel MAP kinase expressed in a subset of neurons in the867

human nervous system. Neuron 14, 67-78.868

[63] Zheng-Fischhofer Q, Biernat J, Mandelkow EM, Illen-869

berger S, Godemann R, Mandelkow E (1998) Sequential870

phosphorylation of Tau by glycogen synthase kinase-3 beta871

and protein kinase A at Thr212 and Ser214 generates the872

Alzheimer-specific epitope of antibody AT100 and requires

a paired-helical-filament-like conformation. Eur J Biochem 873

252, 542-552. 874

[64] Shea TB, Cressman CM (1999) The order of exposure of tau to 875

signal transduction kinases alters the generation of “AD-like” 876

phosphoepitopes. Cell Mol Neurobiol 19, 223-233. 877

[65] Cruz JC, Tseng HC, Goldman JA, Shih H, Tsai LH (2003) 878

Aberrant Cdk5 activation by p25 triggers pathological events 879

leading to neurodegeneration and neurofibrillary tangles. 880

Neuron 40, 471-483. 881

[66] Lucas JJ, Hernandez F, Gomez-Ramos P, Moran MA, Hen R, 882

Avila J (2001) Decreased nuclear beta-catenin, tau hyperphos- 883

phorylation and neurodegeneration in GSK-3 beta conditional 884

transgenic mice. Embo J 20, 27-39. 885

[67] Hanger DP, Anderton BH, Noble W (2009) Tau phosphoryla- 886

tion: the therapeutic challenge for neurodegenerative disease. 887

Trends Mol Med 15, 112-119. 888