16
Uncorrected Author Proof Journal of Alzheimer’s Disease xx (20xx) x–xx DOI 10.3233/JAD-141014 IOS Press 1 Cannabis-Based Medicine Reduces Multiple Pathological Processes in APP/PS1 Mice 1 2 Ester Aso a,b,, Alexandre S´ anchez-Pla c,d , Esteban Vegas-Lozano c , Rafael Maldonado e and Isidro Ferrer a,b 3 4 a Institut de Neuropatologia, Servei d’Anatomia Patol` ogica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, L’Hospitalet de Llobregat, Spain 5 6 b CIBERNED, Centro de Investigaci´ on Biom´ edica en Red de Enfermedades Neurodegenerativas, Instituto Carlos III, Spain 7 8 c Departament d’Estad´ ıstica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain 9 d Statistics and Bioinformatics Unit, Institut de Recerca de l’Hospital Universitari de Vall d’Hebron, Barcelona, Spain 10 11 e Laboratori de Neurofarmacologia, Departament de Ci ` encies Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain 12 13 Handling Associate Editor: Tommaso Cassano 14 Accepted 8 July 2014 Abstract. Several recent findings suggest that targeting the endogenous cannabinoid system can be considered as a potential therapeutic approach to treat Alzheimer’s disease (AD). The present study supports this hypothesis demonstrating that delta-9- tetrahydrocannabinol (THC) or cannabidiol (CBD) botanical extracts, as well as the combination of both natural cannabinoids, which are the components of an already approved cannabis-based medicine, preserved memory in APP/PS1 transgenic mice when chronically administered during the early symptomatic stage. Moreover, THC + CBD reduced learning impairment in APP/PS1 mice. A significant decrease in soluble A 42 peptide levels and a change in plaques composition were also observed in THC + CBD-treated APP/PS1 mice, suggesting a cannabinoid-induced reduction in the harmful effect of the most toxic form of the A peptide. Among the mechanisms related with these positive cognitive effects, the anti-inflammatory properties of cannabinoids may also play a relevant role. Here we observed reduced astrogliosis, microgliosis, and inflammatory-related molecules in treated APP/PS1 mice, which were more marked after treatment with THC + CBD than with either THC or CBD. Moreover, other cannabinoid-induced effects were uncovered by a genome-wide gene expression study. Thus, we have identified the redox protein thioredoxin 2 and the signaling protein Wnt16 as significant substrates for the THC + CBD-induced effects in our AD model. In summary, the present findings show that the combination of THC and CBD exhibits a better therapeutic profile than each cannabis component alone and support the consideration of a cannabis-based medicine as potential therapy against AD. 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 Keywords: Alzheimer’s disease, animal model, cannabidiol, tetrahydrocannabinol, therapy 30 Correspondence to: Ester Aso, Institut de Neuropatologia, Servei d’Anatomia Patol` ogica, IDIBELL-Hospital Universitari de Bellvitge, C/Feixa Llarga s/n, 08907 L’Hospitalet de Llobregat, Spain. Tel.: +34 93 2607452; Fax: +34 93 2607503; E-mail: [email protected]. INTRODUCTION 31 Alzheimer’s disease (AD) is the most common neu- 32 rodegenerative disease associated with dementia in the 33 elderly. While a small proportion of AD cases have a 34 genetic basis, the majority of cases are sporadic with 35 unknown etiology. A consistent feature of the AD 36 ISSN 1387-2877/14/$27.50 © 2014 – IOS Press and the authors. All rights reserved

Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

Journal of Alzheimer’s Disease xx (20xx) x–xxDOI 10.3233/JAD-141014IOS Press

1

Cannabis-Based Medicine Reduces MultiplePathological Processes in A�PP/PS1 Mice

1

2

Ester Asoa,b,∗, Alexandre Sanchez-Plac,d, Esteban Vegas-Lozanoc, Rafael Maldonadoe andIsidro Ferrera,b

3

4

aInstitut de Neuropatologia, Servei d’Anatomia Patologica, IDIBELL-Hospital Universitari de Bellvitge, Universitatde Barcelona, L’Hospitalet de Llobregat, Spain

5

6

bCIBERNED, Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas, Instituto CarlosIII, Spain

7

8

cDepartament d’Estadıstica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain9

dStatistics and Bioinformatics Unit, Institut de Recerca de l’Hospital Universitari de Vall d’Hebron, Barcelona,Spain

10

11

eLaboratori de Neurofarmacologia, Departament de Ciencies Experimentals i de la Salut, Universitat PompeuFabra, Barcelona, Spain

12

13

Handling Associate Editor: Tommaso Cassano14

Accepted 8 July 2014

Abstract. Several recent findings suggest that targeting the endogenous cannabinoid system can be considered as a potentialtherapeutic approach to treat Alzheimer’s disease (AD). The present study supports this hypothesis demonstrating that delta-9-tetrahydrocannabinol (THC) or cannabidiol (CBD) botanical extracts, as well as the combination of both natural cannabinoids,which are the components of an already approved cannabis-based medicine, preserved memory in A�PP/PS1 transgenic micewhen chronically administered during the early symptomatic stage. Moreover, THC + CBD reduced learning impairment inA�PP/PS1 mice. A significant decrease in soluble A�42 peptide levels and a change in plaques composition were also observedin THC + CBD-treated A�PP/PS1 mice, suggesting a cannabinoid-induced reduction in the harmful effect of the most toxicform of the A� peptide. Among the mechanisms related with these positive cognitive effects, the anti-inflammatory propertiesof cannabinoids may also play a relevant role. Here we observed reduced astrogliosis, microgliosis, and inflammatory-relatedmolecules in treated A�PP/PS1 mice, which were more marked after treatment with THC + CBD than with either THC or CBD.Moreover, other cannabinoid-induced effects were uncovered by a genome-wide gene expression study. Thus, we have identifiedthe redox protein thioredoxin 2 and the signaling protein Wnt16 as significant substrates for the THC + CBD-induced effectsin our AD model. In summary, the present findings show that the combination of THC and CBD exhibits a better therapeuticprofile than each cannabis component alone and support the consideration of a cannabis-based medicine as potential therapyagainst AD.

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

Keywords: Alzheimer’s disease, animal model, cannabidiol, tetrahydrocannabinol, therapy30

∗Correspondence to: Ester Aso, Institut de Neuropatologia,Servei d’Anatomia Patologica, IDIBELL-Hospital Universitari deBellvitge, C/Feixa Llarga s/n, 08907 L’Hospitalet de Llobregat,Spain. Tel.: +34 93 2607452; Fax: +34 93 2607503; E-mail:[email protected].

INTRODUCTION 31

Alzheimer’s disease (AD) is the most common neu- 32

rodegenerative disease associated with dementia in the 33

elderly. While a small proportion of AD cases have a 34

genetic basis, the majority of cases are sporadic with 35

unknown etiology. A consistent feature of the AD 36

ISSN 1387-2877/14/$27.50 © 2014 – IOS Press and the authors. All rights reserved

Page 2: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

2 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

brain is the presence of senile plaques composed of37

pathogenic extracellular deposits of amyloid-� (A�),38

a peptide derived from the aberrant processing of the39

trans-membrane amyloid-� protein precursor (A�PP).40

A� fragments are believed to play a central role41

in the genesis of the disease resulting in memory42

loss and behavioral changes. A second pathologi-43

cal hallmark of the disease is hyperphosphorylation44

of the microtubule-associated protein tau that forms45

intracellular neurofibrillary tangles. AD is also associ-46

ated with neuroinflammation and oxidative stress thus47

exacerbating neurodegenerative damage [1, 2]. The48

feeble effectiveness of current therapies against AD49

highlights the need for urgent development of new50

agents geared to preventing the disease or curbing its51

progression.52

Targeting the endocannabinoid system offers a53

multi-faceted approach to the treatment of AD as54

cannabinoid compounds provide neuroprotection by55

reducing neuronal damage, neuroinflammation, and56

oxidative stress, as well as by promoting intrinsic repair57

mechanisms [3–5]. Recent studies have demonstrated58

that chronic stimulation with selective synthetic ago-59

nists of CB1 and CB2 receptors, the most well-known60

cannabinoid receptors, reduce cognitive impairment61

and brain alterations associated with A� produc-62

tion, in at least three different animal models of AD63

[6–9]. Promising results have also been obtained in a64

murine model of tauopathy using treatment with nat-65

ural cannabinoids [10]. Moreover, several in vitro and66

in vivo observations support the beneficial effects of67

CB1 and CB2 stimulation in AD models. Thus, the68

activation of CB1 receptor in vitro preserves neuron69

viability by reducing A�-induced lysosomal mem-70

brane permeability [11] and suppressing pro-apoptotic71

signaling pathways [12]. CB2 receptor agonists induce72

A� removal by human macrophages [13] and reduce73

microglial response to A� [7, 14]. In addition, cer-74

tain cannabinoids are also capable of decreasing tau75

phosphorylation via CB1 or CB2 receptor activation [7,76

15, 16].77

The aim of the present study was to test78

the therapeutic properties of the combination of79

delta-9-tetrahydrocannabinol (THC) and cannabidiol80

(CBD), two phytocannabinoids produced by the plant81

Cannabis sativa that are known to modulate the82

endogenous cannabinoid system, in an animal model83

of AD. The compounds are the two main components84

of Sativex®, which is a cannabinoid-based medicine85

already launched in eleven countries (including the86

UK, Canada, Spain, Italy, and Germany), and approved87

in a further thirteen countries. Sativex® is a well-88

tolerated medicine prescribed for the treatment of 89

spasticity associated with multiple sclerosis and it 90

is also undergoing development for other therapeu- 91

tic applications including pain of various origins (i.e., 92

cancer) and Huntington’s disease [17, 18], a fact that 93

can facilitate the translation from basic research in AD 94

models to human cases. We have used A�PP/PS1 mice 95

as an animal model because they replicate the most 96

relevant features of AD, including cognitive impair- 97

ment and several pathological alterations such as A� 98

deposition, dystrophic neurites, synaptic failure, mito- 99

chondrial dysfunction, and oxidative stress damage 100

[19, 20]. 101

MATERIALS AND METHODS 102

Animals 103

The experiments were carried out in male A�PP/PS1 104

mice and wild-type littermates aged 6 months (early 105

symptomatic phase) at the outset of the study. The gen- 106

eration of mice expressing the human mutated forms 107

A�PPswe and PS1dE9 has already been described 108

[19]. Animals were maintained under standard ani- 109

mal housing conditions in a 12-h dark-light cycle with 110

free access to food and water. Mice were randomly 111

assigned to treatment groups and the experiments 112

were conducted under blind experimental conditions. 113

All animal procedures were carried out following 114

the guidelines of the European Communities Coun- 115

cil Directive 2010/63/EU and with the approval of the 116

local ethical committees of the University of Barcelona 117

and University Pompeu Fabra. 118

Pharmacological treatment 119

THC enriched botanical extract (containing 120

67.1% THC, 0.3% CBD, 0.9% cannabigerol, 0.9% 121

cannabichromene, and 1.9% other phytocannabinoids) 122

and CBD enriched botanical extract (containing 123

64.8% CBD, 2.3% THC, 1.1% cannabigerol, 3.0% 124

cannabichromene, and 1.5% other phytocannabinoids) 125

were supplied by GW Pharmaceuticals Ltd (Cam- 126

bridge, UK). The extracts (THC, 0.75 mg/kg; CBD, 127

0.75 mg/kg; THC + CBD, 0.75 mg/kg each) were 128

dissolved in 5% ethanol, 5% Tween, and 90% saline, 129

and these mixtures were injected intra-peritoneally 130

(i.p.) in a volume of 10 mL/kg body weight. The 131

human equivalent dose (HED) calculated with the 132

formula for dose translation based on body surface 133

area [20] corresponds to 0.04 mg/kg for each cannabi- 134

noid, what is equivalent to the administration of 135

Page 3: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 3

a single Sativex® oromucosal spray (2.8 mg THC136

+2.8 mg CBD) in a human being weighting 70 kg,137

and is lacking of psychoactivity. Animals were138

treated once a day for 5 weeks with the extracts139

or the corresponding vehicle (wild-type, n = 7–11;140

A�PP/PS1, n = 7–8 per group). After 10 days of141

washing period, animals were subjected to behavioral142

evaluation.143

Behavioral evaluation of cognitive performance144

and sample collection145

Two-object recognition test: This paradigm was146

performed in a V-maze (Panlab, Barcelona, Spain)147

because it improves the exploration time of the ani-148

mals with respect to a classical open field. On day149

1, mice were habituated for 9 min, allowing them to150

freely explore the apparatus. On the second day, mice151

were placed for 9 min in the maze, where two identi-152

cal objects were situated at the end of the arms, and153

the time that the mice spent exploring each object was154

recorded. Then, 24 h after the training session, animals155

were placed again in the V-maze where one of the156

two familiar objects was replaced by a novel object.157

The time that the animals spent exploring the two158

objects was recorded and an object recognition index159

(RI) was calculated as the difference between the time160

spent exploring the novel (TN) and the familiar object161

(TF), divided by the total time spent exploring the two162

objects [RI = (TN-TF)/(TN + TF)]. Animals exhibiting163

memory impairments revealed a lower object recogni-164

tion index.165

Active avoidance test: After the two-object recogni-166

tion test, the animals were allowed to rest for 4 days167

before starting the active avoidance test. Then, the mice168

were trained to avoid an aversive stimulus associated169

with the presentation of a conditioned stimulus (CS) in170

a two-way shuttle box apparatus (Panlab, Barcelona,171

Spain). The CS was a light (10 W) switched on in172

the compartment in which the mouse was placed. The173

CS was received 5 s before the onset of the uncondi-174

tioned stimulus (US) and overlapped it for 25 s. At the175

end of the 30-s period, both CS and US were auto-176

matically turned off. The US was an electric shock177

(0.2 mA) continuously applied to the grid of the floor.178

A conditioned response was recorded when the animal179

avoided the US by changing from the compartment180

where it received the CS to the opposite compartment181

within the 5-s period after the onset of the CS. If ani-182

mals failed to avoid the shock, they could escape it by183

crossing during the US (25 s), and this was recorded184

as unconditioned response. Between each trial session,185

there was an inter-trial interval of 30 s. Animals were 186

subjected to five daily 100-trial active avoidance ses- 187

sions. Each day, the mice were placed in the shuttle 188

box for 10 min before the start of each session to allow 189

them to explore the box. Data are expressed as the 190

total number of conditioned changes, converted to the 191

area under the curve (AUC) using a standard trapezoid 192

method. 193

At the end of the behavioral testing, the animals 194

were sacrificed by cervical dislocation and their brains 195

rapidly removed from the skull and processed for study. 196

One hemisphere was dissected on ice, immediately 197

frozen, and stored at −80◦C until used for the pro- 198

tein quantification and the gene expression study. The 199

other hemisphere was fixed in 4% paraformaldehyde 200

and processed for immunohistochemistry. 201

Aβ immunohistochemistry 202

Fixed tissue samples were embedded in paraffin, and 203

coronal sections, 4 �m thick, were cut with a micro- 204

tome. Consecutive de-waxed sections were incubated 205

with 98% formic acid (3 min) and then treated with 206

citrate buffer (20 min) to enhance antigenicity. Then 207

endogenous peroxidases were blocked by incubation 208

in 10% methanol-1% H2O2 solution (15 min). Sections 209

were blocked with 3% normal horse serum solution 210

and then incubated at 4◦C overnight with the primary 211

antibody against A�40 (1 : 100, Merck Millipore, Bil- 212

lerica, MA, USA) or A�42 (1 : 50, Merck Millipore). 213

Sections were subsequently rinsed and incubated with 214

biotinylated secondary antibody (Dako), followed by 215

EnVision + system peroxidase (Dako), and finally with 216

chromogen diaminobenzidine and H2O2. Sections 217

were lightly counterstained with hematoxylin. After 218

staining, the sections were dehydrated and cover- 219

slipped for observation under a Nikon Eclipse E800 220

microscope (Nikon Imaging Inc., Tokyo, Japan; Objec- 221

tive: 10x). The cortical total A�42 and A�40 burden 222

was calculated as the percentage of the area of amy- 223

loid deposition in plaques with respect to the total 224

area in 9 representative pictures taken from the cere- 225

bral cortex of each animal, corresponding to the main 226

regions where A�42 and A�40 deposition is observed 227

in A�PP/PS1 mice. The ratio between A�42 and A�40 228

deposition in each plaque was calculated by com- 229

paring the specific staining with each antibody in at 230

least 10 plaques per animal in consecutive sections. 231

A� quantification was calculated using the Adobe®232

Photoshop® CS4 software (Adobe Systems Inc., San 233

Jose, CA, USA), as previously described [20]. All the 234

A�PP/PS1 treated animals were analyzed.

Page 4: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

4 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

Aβ soluble quantification: Enzyme-linked235

immunosorbent assay (ELISA)236

Fresh-frozen mouse brain cortex was homoge-237

nized in 4 volumes (wt:vol) of TBS extraction238

buffer (140 mM NaCl, 3 mM KCl, 25 mM Tris (pH239

7.4), 5 mM EDTA, and protease inhibitor cocktail240

(Roche Molecular Systems, Pleasanton, CA, USA).241

Homogenate was spun 100,000 g × 1 h, and the super-242

natant was saved as the soluble fraction for A�243

quantification. A�40 and A�42 Human ELISA kits244

(InvitrogenTM Corporation, Camarillo, CA, USA)245

were used to quantify the levels of A�40 and A�42 pep-246

tides in the brain soluble fractions. Quantitative deter-247

mination was carried out according to the manufac-248

turer’s instructions, as previously described [21]. A�40249

and A�42 levels were normalized to the total amount250

of protein from each individual sample (BCA method,251

Thermo Fisher Scientific, Wilmington, DE, USA). Six252

A�PP/PS1 mice per treatment were analyzed.253

Double-labeling immunofluorescence254

De-waxed sections were incubated with 98% formic255

acid (3 min) for A� immunofluorescence and then256

treated with citrate buffer (20 min) to enhance anti-257

genicity. Sections were stained with a saturated258

solution of Sudan black B for 30 min (Merck Milli-259

pore) to block lipofuscin autofluorescence, then rinsed260

in 70% ethanol and washed in distilled water. After a261

blockade with 10% fetal bovine serum (90 min), the262

sections were incubated at 4◦C overnight with combi-263

nations of primary antibodies against A� (clone 6F/3D264

1 : 50, Dako), glial fibrillary acidic protein (GFAP;265

1 : 250, Dako) or IBA1 (1 : 250, Wako, Richmond, VA,266

USA). After washing, the sections were incubated with267

Alexa488 or Alexa546 fluorescence secondary anti-268

bodies against the corresponding host species (1 : 400,269

Molecular Probes, Eugene, OR, USA). Then they270

were washed and mounted in Immuno-Fluore Mount-271

ing medium (ICN Biomedicals, Solon, OH, USA),272

sealed, dried overnight, and examined with a Nikon273

Eclipse E800 microscope. The specific GFAP and274

IBA1 immunostaining density was calculated in refer-275

ence to the A� plaque area in 5 representative pictures276

taken from the cortex of each animal using the Adobe®277

Photoshop® CS4 software. Six animals per each group278

were used for quantifications.279

RNA microarray studies280

RNA from frozen cortex samples of treated mice281

was extracted following the instructions of the sup-282

plier (Rneasy Mini Kit, Qiagen® GmbH, Hilden, 283

Germany). RNA quality control was tested with the 284

Agilent Bioanalyzer (Agilent Technologies Inc, Santa 285

Clara, CA, USA), and the RNA concentration was 286

evaluated using a NanoDrop™ Spectrophotometer 287

(Thermo Fisher Scientific). A total of 24 samples (6 288

A�PP/PS1 samples per treatment) were analyzed by 289

microarray hybridization with the GeneChip® Mouse 290

Gene 1.0 ST Array from Affimetrix (Santa Clara, 291

CA, USA). Bioinformatic analysis was performed with 292

a three (+1) step on the probe values to turn them 293

into comparable gene-level expression values: back- 294

ground correction (RMA), normalization (Quantiles), 295

summarization (Median Polish), and transcript-level 296

summarization (Average). Non-specific filtering was 297

applied to rule out controls, low signal genes, and low 298

variability genes. This pre-processing left 5,606 genes 299

for further study. Functional annotation and biological 300

term enrichment analysis were carried out using the 301

DAVID database (http://david.abcc.ncifcrf.gov/). We 302

used p < 0.05 as the cut-off point to determine whether 303

Kyoto Encyclopedia of Genes and Genomes (KEGG) 304

pathways were significantly enriched. Each group was 305

composed by 6 samples. 306

Quantitative PCR 307

1 �g total RNA was reverse-transcribed with cDNA 308

synthesized with the High-Capacity cDNA Reverse 309

Transcription kit (Applied Biosystems). Quantifica- 310

tion of the mRNA levels was performed in duplicate 311

reactions with gene-specific TaqMan® probes and 312

the TaqMan® Universal PCR Master Mix (Applied 313

Biosystems). House-keeping genes used were Aars, 314

Hprt, and Xpnpep1 [22]. QPCR was performed using 315

the Applied Biosystems 7900HT Fast Real-Time PCR 316

System. Samples were analyzed with the double delta 317

CT (��CT) method using vehicle-treated A�PP/PS1 318

samples as control. Six animals per group were 319

analyzed. 320

Gel electrophoresis and western blotting 321

Samples of the cerebral cortex were homogenized 322

in RIPA lysis buffer (50 mM Tris/HCl buffer, pH 7.4 323

containing 2 mM EDTA, 0.2% Nonidet P-40, 1 mM 324

PMSF, protease, and phosphatase inhibitor cocktails, 325

Roche Molecular Systems, USA). The homogenates 326

were centrifuged for 15 min at 13,000 rpm. Pro- 327

tein concentration was determined with the BCA 328

method (Thermo Scientific). Equal amounts of protein 329

(20 �g) for each sample were loaded and separated 330

Page 5: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 5

by electrophoresis on sodium dodecyl sulfate poly-331

acrylamide gel electrophoresis (SDS-PAGE) (10%)332

gels and transferred onto nitrocellulose membranes333

(Amersham, Freiburg, Germany). Non-specific bind-334

ings were blocked by incubation in 3% albumin in PBS335

containing 0.2% Tween for 1 h at room temperature.336

After washing, membranes were incubated overnight337

at 4◦C with the antibodies against extracellular signal-338

regulated kinase (ERK)1/2 phospho Thr202/Tyr204339

Thr185/Tyr187 (1 : 1,000, Millipore), ERK1/2 (1 : 200,340

Santa Cruz Biotechnology, Dallas, TX, USA), thiore-341

doxin 2 (Txn2, 1 : 1,000, Proteintech, Chicago, IL,342

USA) and wingless-related integration site 16 (Wnt16,343

1 : 5,000, GeneTex, Irvine, CA, USA). Protein load-344

ing was monitored using an antibody against �-tubulin345

(1 : 10,000, Abcam). Membranes were then incubated346

for 1 h in the appropriate HRP- conjugated secondary347

antibodies (1 : 2,000, Dako), and immunocomplexes348

were revealed by chemiluminescence reagent (ECL,349

Amersham). Densitometric quantification was carried350

out with TotalLab v2.01 software (Pharmacia, Swe-351

den). Bands were normalized to �-tubulin. Six animals352

per group were analyzed.353

Statistical analysis354

The sample size for experimentation was com-355

puted using the Power and Precision software (Biostat,356

Englewood, NJ, USA), assuming a power of 95% and357

no missing data. Statistical analysis was performed358

with the SPSS® Statistics v21.0 software (IBM, New359

York, NY, USA). The normality of the data was360

assessed with the Shapiro-Wilk test and as a conse-361

quence parametric statistical tests were used for the362

analysis of all the data in the study. Data were analyzed363

with two-way ANOVA with genotype and treatment364

as between factors (memory, AUC, western blotting365

quantifications) or one-way ANOVA with treatment as366

between factor (A�, glia, and gene expression quantifi-367

cations), followed by Tukey’s post hoc when required.368

Learning data (conditioned changes) were analyzed by369

two-way ANOVA with day of training as within factor370

and genotype as between factor. In all the experiments,371

the significance level was set at p < 0.05.372

RESULTS373

Natural cannabinoids reduce cognitive deficits in374

AβPP/PS1 mice375

Daily administration of THC (0.75 mg/kg, i.p.),376

CBD (0.75 mg/kg, i.p.) botanical extracts, or the com-377

bination of THC and CBD (0.75 mg/kg each botanical 378

extract, i.p.) during 5 weeks at the early stages of 379

the symptomatic phase (6 months) blunted the mem- 380

ory impairment observed in vehicle-treated A�PP/PS1 381

mice when compared to wild-type animals on the 382

two-object recognition test (Fig. 1A). Thus, two- 383

way ANOVA revealed a significant treatment effect 384

(F(3,55) = 3.57, p < 0.05) and interaction between gen- 385

type and treatment (F(3,55) = 12.92, p < 0.001), but 386

not genotype effect. Subsequent Tukey’s post hoc 387

tests revealed that THC (p < 0.001), CBD (p < 0.01), 388

and THC + CBD (p < 0.05) significantly increased the 389

recognition index of A�PP/PS1 mice when com- 390

pared to vehicle-treated littermates. Chronic exposure 391

to THC botanical extract resulted in reduced mem- 392

ory performance in wild-type mice when compared 393

to vehicle-treated littermates (p < 0.05). However, 394

this deleterious effect was not seen in CBD- and 395

THC + CBD-treated wild mice as no impaired memory 396

performance was observed in these animals. No signif- 397

icant difference in the total exploration time during the 398

memory acquisition session or the memory test was 399

observed between groups (Supplementary Table 1), 400

discarding any possible impact of the treatments on the 401

anxiety levels or the activity of mice. Animals exhib- 402

ited no preference for any object during the acquisition 403

session. 404

The learning performance of mice was evalu- 405

ated in the active avoidance test by recording the 406

number of conditioned changes during 5 consecu- 407

tive training days. The AUC revealed a significant 408

reduction in the learning performance of vehicle- 409

(p < 0.01) and CBD-treated (p < 0.05) but not in THC- 410

or THC + CBD-treated A�PP/PS1 mice when com- 411

pared to wild littermates (Fig. 1B). When compared 412

day by day, the number of conditioned changes 413

achieved by mice was reduced in vehicle-treated 414

A�PP/PS1 mice on day 3 (p < 0.05), day 4 (p < 0.01), 415

and day 5 (p < 0.001; Fig. 1 C), in THC-treated on 416

day 5 (Fig. 1D) and in CBD-treated mice on day 417

3 (p < 0.05), day 4 (p < 0.01) and day 5 (p < 0.01; 418

Fig. 1E) when compared with wild-type animals. In 419

contrast, A�PP/PS1 mice chronically treated with the 420

combination of THC + CBD did not evidence such 421

learning impairment at any day (Fig. 1F). No signif- 422

icant treatment effect was observed respect vehicle 423

group neither in wild-type nor A�PP/PS1 mice. These 424

results demonstrate that the THC + CBD combination 425

rescued A�PP/PS1 learning impairment in the active 426

avoidance paradigm when administered at the begin- 427

ning of the symptomatic stage. See Supplementary 428

Table 2 for statistical details. 429

Page 6: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

6 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

A B

C D

E F

Fig. 1. A) Memory performance of animals treated during the early symptomatic stage (6 months). A�PP/PS1 mice chronically treated withvehicle exhibit a significant reduction in the recognition index when compared to corresponding wild-type littermates. However, chronic THC(0.75 mg/kg, i.p.), CBD (0.75 mg/kg, i.p.) botanical extracts, and THC + CBD (0.75 mg/kg each, i.p.) administration induce memory improvementin A�PP/PS1 when compared to wild-type animals. Interestingly, chronic THC induces a significant reduction in the memory performance ofwild-type animals. B-F) The number of conditioned changes in the active avoidance test was recorded during 5 consecutive days in order toevaluate the learning performance of mice. B) Statistical analysis from the Area Under the Curve (AUC) reveals a global reduction in thelearning performance of vehicle- and CBD-treated but not in THC- or THC + CBD-treated A�PP/PS1 mice when compared to wild littermates.The comparison of the conditioned changes achieved by mice every training day reveals a significant reduction in A�PP/PS1 mice treated withvehicle from day 3 to day 5 (C), in THC-treated on day 5 (D), and in CBD-treated mice from day 3 to day 5 (E) when compared with wild-typeanimals. In contrast, A�PP/PS1 mice chronically treated with the combination of THC + CBD do not evidence such learning impairment at anyday, thus demonstrating a positive effect (F). No significant treatment effect is observed respect vehicle group either in wild-type (light graydashed line) or A�PP/PS1 mice (dark gray dashed line). Data are expressed as the mean values ± SEM. �p < 0.05, ��p < 0.01, ���p < 0.001genotype effect; p < 0.05, p < 0.01, p < 0.001 compared to vehicle. §p < 0.05 compared to THC group.

Page 7: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 7

The combination of THC and CBD alters Aβ430

processing in AβPP/PS1 mice431

Chronic treatment with THC, CBD, or the combi-432

nation of both did not significantly modify the total433

A� burden (F(3,28) = 0.73, N.S.; Fig. 2B) or the A�42434

(F(3,22) = 0.62, N.S.) and A�40 burden (F(3,22) = 0.30,435

N.S.; Fig. 2C) in the cortex of A�PP/PS1 mice,436

although there was a tendency to reduced A� depo-437

sition in THC + CBD-treated animals. Similarly, no438

significant treatment effect was observed in the total439

A� burden in the hippocampus of A�PP/PS1 mice440

(F(3,17) = 0.83, N.S.; Fig. 2B), which is much lower441

than the A� burden observed in the A�PP/PS1 mice442

cortex, as expected. However, a significant reduc-443

tion in A�42 (F(3,22) = 7.88, p < 0.001), but not A�40444

(F(3,22) = 1.62, N.S), protein levels was observed in445

the cortical soluble fraction of THC + CBD-treated446

A�PP/PS1 mice when compared to vehicle- (p < 0.01),447

THC- (p < 0.01), and CBD-treated mice (p < 0.05), thus448

demonstrating a protective effect of the combination449

of both cannabinoids in A�PP/PS1 animals by reduc-450

ing the most toxic form of the A� peptide (Fig. 2D).451

The THC + CBD treatment also induced a change452

(F(3,23) = 3.169, p < 0.05) in the composition of A�453

plaques since the ratio A�42/A�40 in each plaque was454

increased in treated A�PP/PS1 mice when compared455

to control group (p < 0.05) (Fig. 2E, F), suggesting a456

facilitation of A�42 deposition that could be related457

to the reduction of the most toxic A�42 soluble con-458

tents. None of the A� forms studied was detectable in459

wild-type animals, as expected (data not shown).460

Natural cannabinoids reduce Aβ461

deposition-related astrogliosis and cytokine462

expression in AβPP/PS1 mice463

One-way ANOVA revealed a treatment effect464

in the astrogliosis (F(3,20) = 10.86, p < 0.001) and465

microgliosis (F(3,20) = 2.53, p < 0.05) associated to A�466

deposition in A�PP/PS1 mice. A significant reduc-467

tion in the number of astrocytes around A� plaques468

was observed in mice treated with THC (p < 0.01),469

CBD (p < 0.001), or the combination of the two470

compounds (p < 0.05) when compared with vehicle-471

treated A�PP/PS1 mice, as revealed with quantitative472

double-labeling immunofluorescence (Fig. 3A, B).473

However, the number of microglial cells associated474

with A� plaques was only significantly reduced by the475

THC + CBD combination (p < 0.05) when compared476

to vehicle-treated A�PP/PS1 animals (Fig. 3A, C).477

No significant effect on the number of astrocytes and478

microglial cells was observed in the cortex of treated 479

wild-type mice (data not shown). To assess possible 480

inflammatory changes associated with cannabinoid 481

compounds, we evaluated the expression levels of a 482

panel of cytokine-related genes, which have been pre- 483

viously demonstrated to underlie the inflammatory 484

response in A�PP/PS1 mice and AD brains (Lopez- 485

Gonzalez et al., in preparation), by quantitative PCR. 486

As shown in Table 1, the combination of THC + CBD 487

resulted in a marked modification of the neuroin- 488

flammatory responses, which was greater than that 489

resulting from treatment with THC or CBD alone. 490

Reduced inflammatory responses involved a colony 491

stimulating factor receptor (Csf3r), a complement sys- 492

tem component (C1qtnf7), a cell surface adhesion 493

protein (Itgb2), Fc receptors (Fcgr1, Fcgr2b), a pro- 494

inflammatory cytokine (Il6st), a regulator of myeloid 495

cell cycle (Inpp5d), and toll-like receptors (Tlr4, 496

Tlr7). The THC + CBD combination also reduced the 497

expression of two genes related to anti-inflammatory 498

cytokines (Il10rb, Tgfb1). 499

Natural cannabinoids modify brain gene 500

expression in AβPP/PS1 mice 501

Additional transcription modifications associated 502

with cannabinoid effects in A�PP/PS1 mice were 503

assessed with RNA microarrays. Natural cannabi- 504

noids induced a differential gene expression profile 505

in A�PP/PS1 mice as revealed the heatmap obtained 506

from microarrays studies (Fig. 4A). The number of 507

genes significantly modulated in relation to vehicle- 508

treated A�PP/PS1 mice was 142 upregulated and 142 509

down-regulated in THC-treated mice; 125 upregu- 510

lated and 166 down-regulated in CBD-treated mice; 511

and 187 upregulated and 136 down-regulated in the 512

THC + CBD group (p < 0.05). The Venn’s diagram 513

shows that only 23 genes were commonly regu- 514

lated by the three treatments (Fig. 4B). The KEGG 515

enrichment analysis of the results allowed to dis- 516

cover functional-related gene groups significantly 517

modulated by treatments and pointed to degradation 518

processes, immunomodulation, mitochondrial func- 519

tion, and mitogen-activated protein kinase 3 (Mapk3) 520

and wingless-type MMTV integration site family, 521

member 16 (Wnt16) signaling pathways, among oth- 522

ers, as relevant molecular mechanisms underlying the 523

effects of natural cannabinoids in A�PP/PS1 trans- 524

genic mice (Supplementary Table 3). Eight candidate 525

genes were chosen for validation on the basis of 526

their potential functional relevance and their high- 527

fold change in treated A�PP/PS1 mice. The statistical 528

Page 8: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

8 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

A B

C D

E F

Fig. 2. A) Scheme showing the cortical brain areas (dashed squares) analyzed for A� burden quantification in each animal. Neither total A�burden (B) nor A�42 or A�40 burden (C) are significantly modified in A�PP/PS1 mice cortex by chronic treatment with THC, CBD, or thecombination of the two, in spite of the tendency toward decrease in THC + CBD-treated animals. D) Soluble A�40 and A�42 levels in corticalhomogenates from A�PP/PS1 mice chronically treated with THC, CBD, and THC + CBD during the early symptomatic phase. The THC + CBDcombination significantly reduces protein levels of soluble A�42 when compared to vehicle-treated controls, revealing the protective effect ofthe combination of the natural cannabinoids. E) Reduction in the A�42 soluble contents can be related, in part, to a change in the composition ofplaques since THC + CBD-treated A�PP/PS1 mice present increased A�42 respect A�40 deposition in each plaque when compared to vehicle-treated animals. F) Representative images of the A�42 (right) and A�40 (left) specific immunoreactivity in consecutive cortical sections ofA�PP/PS1 mice treated during the early symptomatic phase. Scale bar represents 100 �m. Counts are expressed as the mean values ± SEM.�p < 0.05, ��p < 0.01 compared to vehicle. §p < 0.05 compared to THC group. &p < 0.05 compared to CBD group.

Page 9: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 9

Fig. 3. A) Representative images of double GFAP (red, upper panels) or IBA1 (red, lower panels) and A� (green) immunoreactivity in corticalsections of A�PP/PS1 mice chronically treated during the early symptomatic phase with natural cannabinoids. Scale bar represents 25 �m.B) Quantification of the GFAP staining around the A� plaques reveals a significant reduction of the astroglial response in A�PP/PS1 micechronically treated with THC, CBD, or the combination of the two. C) Quantification of the IBA1 staining around the A� plaques reveals asignificant reduction in microglial response only in A�PP/PS1 mice chronically treated with the combination of THC + CBD. Data are expressedas the mean values ± SEM. �p < 0.05, ��p < 0.01 ���p < 0.001 compared to vehicle.

analysis of the quantitative PCR resulted in: adeny-529

late cyclase 3 (Adcy3; F(3,20) = 1.54, N.S.), cytochrome530

c oxidase subunit VIIc (Cox7c; F(3,20) = 2.30, N.S.),531

Mapk3 (F(3,20) = 5.76, p < 0.01), nitric oxide synthase532

1 (Nos1; F(3,20) = 3.76, p < 0.05), proteasome subunit,533

beta type, 2 (Psmb2; F(3,20) = 3.37, p < 0.05), thiore-534

doxin 2 (Txn2; F(3,20) = 5.08, p < 0.01), ubiquitin (Ubb;535

F(3,20) = 3.182, p < 0.05), and Wnt16 (F(3,20) = 2.22,536

p < 0.05). Thus, a Mapk3, Psmb2, Txn2, and Wnt16537

decrease was validated in THC + CBD-treated mice538

(Fig. 4C). Decrease expression of Nos1 and Ubb539

was observed by quantitative PCR in THC + CBD,540

which was in contrast with the increase found in RNA541

microarray. Finally, Adcy3 and Cox7c modifications542

seen in microarrays were not validated with PCR.

Natural cannabinoids modulate MAPK3, Txn2, 543

and Wnt16 protein levels in AβPP/PS1 mice 544

We assessed the correlation between the 545

cannabinoid-induced alteration of Mapk3, Txn2, 546

and Wnt16 gene expression and the levels of the 547

proteins coded by those genes using western blotting. 548

In spite of decreased Mapk3 mRNA, no modifica- 549

tions in the expression of ERK1 (Genotype effect: 550

F(1,31) = 3.13, N.S.; Treatment effect: F(3,31) = 2.15, 551

N.S.; Interaction: F(3,31) = 1.26, N.S.) were seen in 552

treated A�PP/PS1 mice (Fig. 5A). However, natural 553

cannabinoids induced a significant modulation of 554

ERK1, but not ERK2, phosphorylation (Geno- 555

type effect: F(1,31) = 0.93, N.S.; Treatment effect: 556

Page 10: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

10 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

Table 1mRNA expression levels of several cytokine-related genes involved in the inflammatory response in A�PP/PS1 mice

Cytokine-related genes A�PP/PS1

Vehicle THC CBD THC + CBD

Anti-inflammatory cytokines Il10ra 1.02 ± 0.09 0.90 ± 0.05 0.92 ± 0.07 0.88 ± 0.06Il10rb 1.01 ± 0.07 1.08 ± 0.04 0.96 ± 0.08 0.78 ± 0.03∗.§Tgfb1 1.03 ± 0.12 0.86 ± 0.05 0.84 ± 0.08 0.71 ± 0.07∗

Cell Surface Adhesion Itgb2 1.01 ± 0.07 0.95 ± 0.05 1.04 ± 0.15 0.75 ± 0.08∗Chemokines Ccl3 1.04 ± 0.12 1.21 ± 0.14 1.19 ± 0.10 0.82 ± 0.16

Ccl4 1.03 ± 0.10 1.16 ± 0.10 1.39 ± 0.10∗ 0.97 ± 0.17Ccl6 1.04 ± 0.12 1.24 ± 0.09 1.19 ± 0.12 0.99 ± 0.09

CxCl10 1.22 ± 0.35 1.21 ± 0.21 1.04 ± 0.19 0.95 ± 0.18Complement system C1ql1 1.01 ± 0.05 1.15 ± 0.05 1.04 ± 0.03 1.11 ± 0.13

C1qtnf7 1.08 ± 0.19 0.95 ± 0.08 0.99 ± 0.03 0.75 ± 0.08&

C3ar1 1.00 ± 0.04 0.99 ± 0.04 1.03 ± 0.07 0.91 ± 0.06C4b 1.02 ± 0.09 0.89 ± 0.03 1.07 ± 0.12 0.87 ± 0.12

Colony stimulating factor receptors Csf1r 1.01 ± 0.05 1.01 ± 0.03 0.96 ± 0.05 0.90 ± 0.05Csf3r 1.02 ± 0.08 1.02 ± 0.10 0.86 ± 0.05 0.71 ± 0.07∗,§

Fc receptors Fcgr1 1.02 ± 0.09 1.08 ± 0.06 1.00 ± 0.06 0.85 ± 0.08§Fcgr2b 1.01 ± 0.07 1.11 ± 0.07 1.08 ± 0.09 0.87 ± 0.05§

Pro-inflammatory cytokines Il6st 1.01 ± 0.06 0.95 ± 0.07 0.98 ± 0.07 0.77 ± 0.07∗Tnfrsf1a 1.02 ± 0.08 1.15 ± 0.08 1.15 ± 0.05 1.12 ± 0.09

Regulator of myeloid cells Inpp5d 1.01 ± 0.05 0.92 ± 0.08 0.82 ± 0.10 0.60 ± 0.09∗,§Toll-like receptors Tlr4 1.02 ± 0.09 0.90 ± 0.11 0.80 ± 0.08 0.68 ± 0.05∗

Tlr7 1.06 ± 0.15 1.01 ± 0.11 0.96 ± 0.16 0.63 ± 0.06∗,§

Values are calculated with the ��Ct method, using the mean of three housekeeping genes (Aars, Hprt, Xpnpep1) and vehicle-treated A�PP/PS1as references. ∗p < 0.05 versus Vehicle; §p < 0.05 versus THC, &p < 0.05 versus CBD.

F(3,31) = 5.18, p < 0.01; Interaction: F(3,31) = 3.73,557

p < 0.05). Thus, CBD increased the levels of phospho-558

ERK1 in wild-type animals when compared to the559

vehicle (p < 0.05) or THC + CBD (p < 0.01) groups.560

In contrast, THC and THC + CBD induced a ten-561

dency to reduce the phosphorylation of ERK1 in562

A�PP/PS1 mice, which was apparently enhanced in563

vehicle-treated transgenic animals (Fig. 5A). Those564

results indicate that cannabinoid compounds could565

differentially regulate ERK1 signaling.566

Natural cannabinoids modulated the levels of Txn2567

in treated mice (Genotype effect: F(1,31) = 0.71, N.S.;568

Treatment effect: F(3,31) = 5.56, p < 0.01; Interaction:569

F(3,31) = 9.22, p < 0.001). A�PP/PS1 mice exhibited570

decreased Txn2 protein levels after treatment with571

vehicle (p < 0.05) and THC (p < 0.05), which was572

also apparent but not significant after CBD exposure,573

when compared to wild-type littermates (Fig. 5B).574

This deficiency in Txn2 levels could account to575

impaired capability to cope with oxidative components576

in A�PP/PS1 mice. Interestingly, the combination of577

THC + CBD induced a strong increase in the Txn2 pro-578

tein levels (p < 0.01 with respect to vehicle or CBD;579

p < 0.001 with respect to THC), which completely580

reversed this Txn2 deficiency observed in A�PP/PS1581

mice (Fig. 5B).582

Regarding the signaling protein Wnt16, a significant 583

effect of treatment was also observed (Genotype effect: 584

F(1,31) = 2.59, N.S.; Treatment effect: F(3,31) = 5.64, 585

p < 0.01; Interaction: F(3,31) = 1.67, N.S.). Both THC 586

and the combination of THC + CBD increased the 587

levels in A�PP/PS1 mice when compared to vehicle- 588

treated animals (p < 0.05). THC-treated A�PP/PS1 589

mice exhibited significantly higher Wnt16 protein lev- 590

els than corresponding wild-type controls (p < 0.01) 591

(Fig. 5C). 592

DISCUSSION 593

According to the protective hypothesis of cannabi- 594

noid compounds in neurodegenerative diseases, the 595

present findings show that treatment with natural 596

cannabinoids at non-psychoactive doses reduces cog- 597

nitive impairment and several pathological processes 598

occurring in A�PP/PS1, a model of AD, when chron- 599

ically administered at the early symptomatic phase. 600

Thus, THC and CBD, as well as the combina- 601

tion of both natural cannabinoids, reduces memory 602

impairment exhibited by A�PP/PS1 mice in the two- 603

object recognition test, but only the combination of 604

THC + CBD was able to prevent learning deficiency 605

Page 11: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 11

Fig. 4. A) Heat map generated from RNA microarray data reflecting the differential gene expression profile induced by cannabinoid compounds.Blue: decreased expression. Red: increased expression. Headings: Vehicle (yellow), THC (blue), CBD (green), THC + CBD (red). B) Venn’sdiagram showing the number of genes significantly regulated by natural cannabinoids. C) Real-time PCR validated the results obtained withmicroarray techniques in at least 4 out of 8 candidate genes, confirming decreased expression of Mapk3, Psmb2, Txn2, and Wnt16 genes inTHC + CBD-treated A�PP/PS1 mice. Data are expressed as the mean values ± SEM. �p < 0.05 compared to vehicle. &p < 0.05, &&p < 0.01compared to CBD.

of transgenic mice in the active avoidance test, con-606

sidered a complex cognitive task. As THC and CBD607

are supposed to produce their effects by acting on608

different signaling pathways [23], the present results609

with combined THC and CBD can be interpreted610

as a summative effect or as an interaction of the611

two compounds resulting in the potentiation of each612

cannabinoid, as previously suggested [24, 25]. The613

present findings are in agreement with a recent report614

conducted in parallel demonstrating positive behav-615

ioral effects of THC + CBD in a murine model of616

tauopathy [10]. Importantly, the cannabinoid doses 617

employed in this study are devoid of psychoactivity 618

[26] and their HED corresponds to a single Sativex®619

administration, what means that the potential transla- 620

tion of our results to human beings might result in a 621

safe and well-tolerated approach taking into consider- 622

ation that multiple sclerosis patients receiving up to 12 623

Sativex® administrations per day reported a relatively 624

low side-effect profile [27]. 625

A collateral observation deserves attention. In con- 626

trast to A�PP/PS1 mice, memory impairment occurs 627

Page 12: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

12 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

Fig. 5. Western blot quantification of proteins codified by genes differentially expressed in treated mice: ERK1 (Mapk3), thioredoxin 2 (Txn2),and wingless-related integration site (Wnt16). A) No significant change in the total amount of ERK1 is observed in any treatment group, in spiteof the tendency toward increased total ERK1 in THC + CBD-treated A�PP/PS1 mice. CBD significantly increases the levels of phosphorylatedERK1 in wild-type animals. In contrast, THC and THC + CBD slightly decrease ERK1 phosphorylation without statistical significance. B)THC + CBD completely reverses the Txn2 deficiency exhibited by vehicle- and THC-treated A�PP/PS1 mice. C) THC and THC + CBD increasethe levels of Wnt16 protein in cortical homogenates of A�PP/PS1 treated mice. In the upper part of each panel are representative immunoblots forERK1/2, Txn2, and Wnt16, and corresponding tubulin loading control. Densitometric quantifications are expressed as the mean values ± SEM.�p < 0.05, ��p < 0.01 genotype effect. p < 0.05, p < 0.01, compared to vehicle. §§§p < 0.001 compared to THC. &&p < 0.01 compared toCBD.

in wild-type mice chronically exposed to the THC-628

enriched extract at doses that are known not to produce629

acute amnesia-like effects in mice [26]. This obser-630

vation warns about the chronic effects of THC in631

healthy individuals and is in accordance with sev-632

eral human studies revealing that long-term use of633

cannabis can be associated with disruption of short- 634

term memory, working memory, and attention skills 635

[28, 29]. It is known that certain cannabinoids, such 636

as THC, affect cognitive function modulating sig- 637

naling pathways critically implicated in learning and 638

memory [30]. The molecular reorganization of endoge- 639

Page 13: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 13

nous cannabinoid system in AD [31] and the altered640

neuronal signaling occurring during the neurodegen-641

erative processes may account for the discrepancy642

between the effects of THC in wild-type and AD-like643

transgenic mice. However, wild-type mice chroni-644

cally receiving THC + CBD do not exhibit memory645

impairment. This observation supports previous work646

showing that CBD is able to antagonize THC-induced647

deficits in memory tasks [32], and highlights the rele-648

vance of combining the two natural cannabinoids, THC649

and CBD, to mitigate the negative consequences of650

THC administration.651

A remarkable finding of this study is the altered652

A� processing induced by the THC + CBD combi-653

nation in A�PP/PS1 mice. Even though THC, CBD,654

and the combination of both did not significantly mod-655

ify cortical or hippocampal A� burden in A�PP/PS1656

mice in spite of a tendency to decrease in the ani-657

mals treated with THC + CBD, the combination of658

both compounds reduced soluble A�42, but not A�40659

protein levels, thus showing a protective effect by660

reducing the quantity of the most toxic soluble A� form661

in A�PP/PS1 animals [33]. We have also observed662

a change in amyloid plaques composition since an663

increase in the A�42/A�40 ratio in each plaque was664

observed in THC + CBD-treated A�PP/PS1 mice, sug-665

gesting a cannabinoid-induced facilitation of the A�42666

deposition that could account at least in part for the667

specific reduction of soluble A�42 observed and likely668

to decrease its toxicity. The recently described A�42669

clearance facilitation across the blood-brain barrier by670

cannabinoids [8, 34], might also contribute to the THC-671

CBD-induced reduction of the A� toxicity in our AD672

model.673

AD progression involves aberrant glial activation674

and neuroinflammation that contribute to neuronal dys-675

function, which in turn drives a vicious cycle of further676

glial activation and neuronal damage [35]. Several677

studies have shown anti-inflammatory effects of nat-678

ural and synthetic CB1 or CB2 agonists, as well as679

CBD, in multiple in vitro and in vivo AD models [6–8,680

14, 36–38]. The present observations confirm previous681

findings by demonstrating a reduction of the astroglio-682

sis associated with A� deposition in A�PP/PS1 mice683

treated with THC, CBD, or the combination of both. In684

addition, THC + CBD significantly reduced microglio-685

sis and the expression of several cytokines and related686

molecules in A�PP/PS1 mice. Most importantly, the687

combination of THC + CBD resulted more effective688

than either THC or CBD alone.689

The ubiquitous distribution of endocannabinoid sys-690

tem and its polyvalent functionality suggest that the691

positive cognitive effects observed in A�PP/PS1 after 692

chronic treatment with natural cannabinoids might be 693

due to multiple mechanisms run in parallel, beyond 694

to the already known anti-inflammatory properties or 695

the role in reducing A� toxicity. A useful tool to iden- 696

tify novel mechanisms that may contribute to a certain 697

effect is the microarrays technology. This technique 698

involves large-scale monitoring of relative differ- 699

ences in RNA abundance between samples. Thus, we 700

identified additional mechanisms contributing to the 701

natural cannabinoid effects in A�PP/PS1 mice by RNA 702

microarrays. The functional analysis of the results 703

pointed to molecular degradation, immunomodulation, 704

mitochondrial function, and Mapk3 and Wnt16 sig- 705

naling pathways, among others, as relevant pathways 706

targeted by cannabinoids. First, we focused on validat- 707

ing the cannabinoid effects on the Mapk3 signaling. 708

Previous in vitro studies have shown that the stimu- 709

lation of endogenous cannabinoid system decreases 710

ERK1/2 pro-inflammatory signaling in response to 711

A�, resulting in reduced toxicity [12, 39]. Although 712

the total amount of ERK1, the protein coded by 713

Mapk3, is not significantly modulated by cannabi- 714

noids in the present model, THC and THC + CBD 715

decrease ERK1 phosphorylation. We also observed an 716

increase in ERK1/2 phosphorylation in wild-type ani- 717

mals receiving CBD, which is contrast to a previous 718

study showing reduced phospho-ERK1/2 in the cor- 719

tex of rats chronically exposed to CBD [40]. These 720

discrepancies could be due to different experimen- 721

tal conditions. Together, these observations point to 722

the need for further studies geared to elucidating the 723

ERK response in wild and A�PP/PS1 mice treated with 724

cannabinoids. 725

Another important contribution of the present study 726

is the induction of Txn2 protein levels by the 727

THC + CBD combination, in contrast to the reduced 728

Txn2 mRNA expression observed in the microarray 729

study as well as by quantitative PCR. The diver- 730

gence between the mRNA and protein levels could 731

account for compensatory mechanisms directed to 732

regulate Txn2 functionality. THC + CBD completely 733

reversed Txn2 deficiency in A�PP/PS1 mice, which 734

also occurs in AD patients [41]. This nuclear gene 735

encodes a mitochondrial member of the thioredoxin 736

family, a group of small multifunctional redox-active 737

proteins [42]. The encoded protein is a key compo- 738

nent of the mitochondrial antioxidant system which 739

is responsible for the clearance of reactive intermedi- 740

ates and repairs proteins with oxidative damage and 741

may play important roles in the regulation of the mito- 742

chondrial membrane potential and in protection against 743

Page 14: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

14 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

oxidant-induced apoptosis [43, 44]. Therefore, it can744

be assumed that increased Txn2 levels provide protec-745

tion against oxidative damage in our model.746

Finally, little is known about the role of Wnt16747

signaling in cells and to our knowledge there is no748

specific information about Wnt16 function in brain.749

The Wnt gene family consists of structurally related750

genes which encode secreted signaling proteins. These751

proteins have been implicated in oncogenesis and in752

several developmental processes, including regulation753

of cell fate and patterning during embryogenesis, as754

well as in axon guidance during development and in755

response to traumatic injury in adult central nervous756

system [45]. Moreover, activation of the Wnt signaling757

pathway prevents A�-induced neurotoxicity in vitro,758

probably through the modulation of the GSK3�/�-759

catenin pathway [46]. Wnt16 gene is a member of the760

Wnt gene family. It contains two transcript variants761

diverging at the 5’ termini. These two variants are pro-762

posed to be the products of separate promoters and not763

to be splice variants from a single promoter. They are764

differentially expressed in normal tissues, one of which765

(variant 2) is expressed at significant levels only in the766

pancreas, whereas another one (variant 1) is expressed767

more ubiquitously with highest levels in adult kidney,768

placenta, brain, heart, and spleen [47]. Thus, it is tempt-769

ing to speculate that increased cannabinoid-induced770

Wnt16 expression may reduce A� neurotoxicity and771

contribute to maintain axon integrity in vivo. Never-772

theless, additional experiments are required to validate773

this hypothesis.774

In summary, here we provide evidence of the ther-775

apeutic effects of the THC + CBD combination, over776

THC or CBD alone, by acting at different levels mod-777

ifying A� metabolism, reducing soluble A�42 levels,778

astrogliosis, microglia, and several molecules of neu-779

roinflammation. Speculatively, it is conceivable that780

the effects of THC + CBD combination are also due781

to the increase protein expression of thioredoxin 2782

and Wnt16. Nevertheless, additional experiments are783

required to validate this hypothesis. This is accompa-784

nied by a reduction of memory deficits and increased785

learning capacity in A�PP/PS1 transgenic mice used786

as a model of AD. The present findings give insights787

for a further clinical trial to test the effectiveness of788

THC + CBD in AD patients.789

ACKNOWLEDGMENTS790

We thank T. Yohannan for editorial help and GW791

Pharmaceuticals Ltd for the supply of the botanical792

extracts. This study was supported by grants from 793

the Agrupacio Mutua Foundation (XVII Award in the 794

Elderly Field, to IF), Mutua Madrilena Foundation 795

(IF), and BESAD-P project, CIBERNED, Instituto 796

Carlos III (IF). 797

Authors’ disclosures available online (http://www.j- 798

alz.com/disclosures/view.php?id=2441). 799

SUPPLEMENTARY MATERIAL 800

The supplementary material is available in the elect- 801

ronic version of this article: http://dx.doi.org/10.3233/ 802

JAD-141014. 803

REFERENCES 804

[1] Ferrer I (2012) Defining Alzheimer as a common age-related 805

neurodegenerative process not inevitably leading to dementia. 806

Prog Neurobiol 97, 38-51. 807

[2] Selkoe DJ (2012) Preventing Alzheimer’s disease. Science 808

337, 1488-1492. 809

[3] Campbell VA, Gowran A (2007) Alzheimer’s disease; taking 810

the edge off with cannabinoids? Br J Pharmacol 152, 655- 811

662. 812

[4] Koppel J, Davies P (2008) Targeting the endocannabinoid 813

system in Alzheimer’s disease. J Alzheimers Dis 15, 495-504. 814

[5] Aso E, Ferrer I (2014) Cannabinoids for treatment of 815

Alzheimer’s disease: Moving toward the clinic. Front Phar- 816

macol 5, 37. 817

[6] Aso E, Palomer E, Juves S, Maldonado R, Munoz FJ, Ferrer I 818

(2012) CB1 agonist ACEA protects neurons and reduces the 819

cognitive impairment of A�PP/PS1 mice. J Alzheimers Dis 820

30, 439-459. 821

[7] Aso E, Juves S, Maldonado R, Ferrer I (2013) CB2 cannabi- 822

noid receptor agonist ameliorates Alzheimer-like phenotype 823

in A�PP/PS1 mice. J Alzheimers Dis 35, 847-858. 824

[8] Martın-Moreno AM, Brera B, Spuch C, Carro E, Garcıa- 825

Garcıa L, Delgado M, Pozo MA, Innamorato NG, Cuadrado 826

A, de Ceballos ML (2012) Prolonged oral cannabinoid admin- 827

istration prevents neuroinflammation, lowers �-amyloid 828

levels and improves cognitive performance in Tg APP 2576 829

mice. J Neuroinflammation 9, 8. 830

[9] Wu J, Bie B, Yang H, Xu JJ, Brown DL, Naguib M 831

(2013) Activation of the CB(2) receptor system reverses 832

amyloid-induced memory deficiency. Neurobiol Aging 34, 833

791-804. 834

[10] Casarejos MJ, Perucho J, Gomez A, Munoz MP, Fernandez- 835

Estevez M, Sagredo O, Fernandez Ruiz J, Guzman M, de 836

Yebenes JG, Mena MA (2013) Natural cannabinoids improve 837

dopamine neurotransmission and tau and amyloid pathology 838

in a mouse model of tauopathy. J Alzheimers Dis 35, 525-539. 839

[11] Noonan J, Tanveer R, Klompas A, Gowran A, McK- 840

iernan J, Campbell VA (2010) Endocannabinoids prevent 841

beta-amyloid-mediated lysosomal destabilization in cultured 842

neurons. J Biol Chem 285, 38543-38554. 843

[12] Chen X, Zhang J, Chen C (2011) Endocannabinoid 2- 844

arachidonoylglycerol protects neurons against �-amyloid 845

insults. Neuroscience 178, 159-168. 846

[13] Tolon RM, Nunez E, Pazos MR, Benito C, Castillo AI, 847

Martınez-Orgado JA, Romero J (2009) The activation of 848

cannabinoid CB2 receptors stimulates in situ and in vitro beta- 849

Page 15: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice 15

amyloid removal by human macrophages. Brain Res 1283,850

148-154.851

[14] Martın-Moreno AM, Reigada D, Ramırez BG, Mechoulam852

R, Innamorato N, Cuadrado A, de Ceballos ML (2011)853

Cannabidiol and other cannabinoids reduce microglial acti-854

vation in vitro and in vivo: Relevance to Alzheimer’s disease.855

Mol Pharmacol 79, 964-973.856

[15] Esposito G, De Filippis D, Carnuccio R, Izzo AA, Iuvone T857

(2006a) The marijuana component cannabidiol inhibits beta-858

amyloid-induced tau protein hyperphosphorylation through859

Wnt/beta-catenin pathway rescue in PC12 cells. J Mol Med860

84, 253-258.861

[16] Esposito G, De Filippis D, Steardo L, Scuderi C, Savani C,862

Cuomo V, Iuvone T (2006b) CB1 receptor selective activa-863

tion inhibits beta-amyloid-induced iNOS protein expression864

in C6 cells and subsequently blunts tau protein hyper-865

phosphorylation in co-cultured neurons. Neurosci Lett 404,866

342-346.867

[17] Barnes MP (2006) Sativex: Clinical efficacy and tolerabil-868

ity in the treatment of symptoms of multiple sclerosis and869

neuropathic pain. Expert Opin Pharmacother 7, 607-615.870

[18] Pazos MR, Sagredo O, Fernandez-Ruiz J (2008) The endo-871

cannabinoid system in Huntington’s disease. Curr Pharm Des872

14, 2317-2325.873

[19] Borchelt DR, Ratovitski T, van Lare J, Lee MK, Gonzales874

V, Jenkins NA, Copeland NG, Price DL, Sisodia SS (1997)875

Accelerated amyloid deposition in the brains of transgenic876

mice coexpressing mutant presenilin 1 and amyloid precursor877

proteins. Neuron 19, 939-945.878

[20] Reagan-Shaw S, Nihal M, Ahmad N (2008) Dose translation879

from animal to human studies revisited. FASEB J 22, 659-661.880

[21] Aso E, Lomoio S, Lopez-Gonzalez I, Joda L, Carmona881

M, Fernandez-Yague N, Moreno J, Juves S, Pujol A, Pam-882

plona R, Portero-Otın M, Martın V, Dıaz M, Ferrer I (2012)883

Amyloid generation and dysfunctional immunoproteasome884

activation with disease progression in animal model of famil-885

ial Alzheimer’s disease. Brain Pathol 22, 636-653.886

[22] Durrenberger PF, Fernando FS, Magliozzi R, Kashefi SN,887

Bonnert TP, Ferrer I, Seilhean D, Nait-Oumesmar B, Schmitt888

A, Gebicke-Haerter PJ, Falkai P, Grunblatt E, Palkovits M,889

Parchi P, Capellari S, Arzberger T, Kretzschmar H, Ron-890

caroli F, Dexter DT, Reynolds R (2012) Selection of novel891

reference genes for use in the human central nervous sys-892

tem: A BrainNet Europe Study. Acta Neuropathol 124,893

893-903.894

[23] Huestis MA (2005) Pharmacokinetics and metabolism of the895

plant cannabinoids, delta9-tetrahydrocannabinol, cannabidiol896

and cannabinol. Handb Exp Pharmacol 168, 657-690.897

[24] Varvel SA, Wiley JL, Yang R, Bridgen DT, Long K, Licht-898

man AH, Martin BR (2006) Interactions between THC and899

cannabidiol in mouse models of cannabinoid activity. Psy-900

chopharmacology (Berl) 186, 226-234.901

[25] Klein C, Karanges E, Spiro A, Wong A, Spencer J, Huynh902

T, Gunasekaran N, Karl T, Long LE, Huang XF, Liu903

K, Arnold JC, McGregor IS (2011) Cannabidiol potenti-904

ates �9-tetrahydrocannabinol (THC) behavioural effects and905

alters THC pharmacokinetics during acute and chronic treat-906

ment in adolescent rats. Psychopharmacology (Berl) 218,907

443-457.908

[26] Puighermanal E, Marsicano G, Busquets-Garcia A, Lutz B,909

Maldonado R, Ozaita A (2009) Cannabinoid modulation of910

hippocampal long-term memory is mediated by mTOR sig-911

naling. Nat Neurosci 12, 1152-1158.912

[27] Wade DT, Collin C, Stott C, Duncombe P (2010) Meta-913

analysis of the efficacy and safety of Sativex (nabiximols),914

on spasticity in people with multiple sclerosis. Mult Scler 16, 915

707-714. 916

[28] Fletcher JM, Page JB, Francis DJ, Copeland K, Naus MJ, 917

Davis CM, Morris R, Krauskopf D, Satz P (1996) Cognitive 918

correlates of long-term cannabis use in Costa Rican men. Arch 919

Gen Psychiatry 53, 1051-1057. 920

[29] Iversen L (2003) Cannabis and the brain. Brain 126, 1252- 921

1270. 922

[30] Puighermanal E, Busquets-Garcia A, Maldonado R, Ozaita 923

A (2012) Cellular and intracellular mechanisms involved in 924

the cognitive impairment of cannabinoids. Philos Trans R Soc 925

Lond B Biol Sci 367, 3254-3263. 926

[31] Mulder J, Zilberter M, Pasquare SJ, Alpar A, Schulte G, 927

Ferreira SG, Kofalvi A, Martın-Moreno AM, Keimpema E, 928

Tanila H, Watanabe M, Mackie K, Hortobagyi T, de Ceballos 929

ML, Harkany T (2011) Molecular reorganization of endo- 930

cannabinoide signalling in Alzheimer’s disease. Brain 134, 931

1041-1060. 932

[32] Fadda P, Robinson L, Fratta W, Pertwee RG, Riedel G (2004) 933

Differential effects of THC- or CBD-rich cannabis extracts on 934

working memory in rats. Neuropharmacology 47, 1170-1179. 935

[33] Mucke L, Selkoe DJ (2012) Neurotoxicity of amyloid �- 936

protein: Synaptic and network dysfunction. Cold Spring Harb 937

Perspect Med 2, a006338. 938

[34] Bachmeier C, Beaulieu-Abdelahad D, Mullan M, Paris D 939

(2013) Role of the cannabinoid system in the transit of beta- 940

amyloid across the blood-brain barrier. Mol Cell Neurosci 56, 941

255-262. 942

[35] Mrak RE, Griffin WS (2005) Glia and their cytokines in pro- 943

gression of neurodegeneration. Neurobiol Aging 26, 349-354. 944

[36] van der Stelt M, Mazzola C, Esposito G, Matias I, Petrosino 945

S, De Filippis D, Micale V, Steardo L, Drago F, Iuvone T, 946

Di Marzo V (2006) Endocannabinoids and beta-amyloid- 947

induced neurotoxicity in vivo: Effect of pharmacological 948

elevation of endocannabinoid levels. Cell Mol Life Sci 63, 949

1410-1424. 950

[37] Esposito G, Iuvone T, Savani C, Scuderi C, De Filippis D, 951

Papa M, Di Marzo V, Steardo L (2007) Opposing control 952

of cannabinoid receptor stimulation on amyloid-beta-induced 953

reactive gliosis: In vitro and in vivo evidence. J Pharmacol 954

Exp Ther 322, 1144-1152. 955

[38] Ramırez BG, Blazquez C, Gomez del Pulgar T, Guzman 956

M, de Ceballos ML (2005) Prevention of Alzheimer’s dis- 957

ease pathology by cannabinoids: Neuroprotection mediated 958

by blockade of microglial activation. J Neurosci 25, 1904- 959

1913. 960

[39] Benito C, Tolon RM, Castillo AI, Ruiz-Valdepenas L, 961

Martınez-Orgado JA, Fernandez-Sanchez FJ, Vazquez C, 962

Cravatt BF, Romero J (2012) �-Amyloid exacerbates inflam- 963

mation in astrocytes lacking fatty acid amide hydrolase 964

through a mechanism involving PPAR-�, PPAR-� and 965

TRPV1, but not CB1 or CB2 receptors. Br J Pharmacol 166, 966

1474-1489. 967

[40] ElBatsh MM, Assareh N, Marsden CA, Kendall DA (2012) 968

Anxiogenic-like effects of chronic cannabidiol administration 969

in rats. Psychopharmacology (Berl) 221, 239-247. 970

[41] Lovell MA, Xie C, Gabbita SP, Markesbery WR (2000) 971

Decreased thioredoxin and increased thioredoxin reductase 972

levels in Alzheimer’s disease brain. Free Radic Biol Med 28, 973

418-427. 974

[42] Arner ES (2009) Focus on mammalian thioredoxin 975

reductases-important selenoproteins with versatile functions. 976

Biochim Biophys Acta 1790, 495-526. 977

[43] Chen Y, Yu M, Jones DP, Greenamyre JT, Cai J (2006) Pro- 978

tection against oxidant-induced apoptosis by mitochondrial 979

Page 16: Journal of Alzheimer’s Disease xx (20xx) x–xx IOS Press Cannabis …diposit.ub.edu/dspace/bitstream/2445/140650/1/647690.pdf · Among the mechanisms related with these positive

Unc

orre

cted

Aut

hor P

roof

16 E. Aso et al. / Cannabinoids Reduce AD-Like Phenotype in Mice

thioredoxin in SH-SY5Y neuroblastoma cells. Toxicol Appl980

Pharmacol 216, 256-262.981

[44] Damdimopoulos AE, Miranda-Vizuete A, Pelto-Huikko M,982

Gustafsson JA, Spyrou G (2002) Human mitochondrial983

thioredoxin. Involvement in mitochondrial membrane poten-984

tial and cell death. J Biol Chem 277, 33249-33257.985

[45] Onishi K, Hollis E, Zou Y (2014) Axon guidance and injury-986

lessons from Wnts and Wnt signaling. Curr Opin Neurobiol987

27C, 232-240.

[46] Inestrosa NC, Montecinos-Oliva C, Fuenzalida M (2012) Wnt 988

signaling: Role in Alzheimer disease and schizophrenia. J 989

Neuroimmune Pharmacol 7, 788-807. 990

[47] Fear MW, Kelsell DP, Spurr NK, Barnes MR (2000) Wnt-16a, 991

a novel Wnt-16 isoform, which shows differential expression 992

in adult human tissues. Biochem Biophys Res Commun 278, 993

814-820. 994