44
1 Identification of antiparkinsonian drugs in the 6-hydroxydopamine zebrafish model 1 Rita L. Vaz 1,2,† , Sara Sousa 1,†,* , Diana Chapela 1,3 , Herma C. van der Linde 4 , Rob 2 Willemsen 4 , Ana D. Correia 3, & , Tiago F. Outeiro 5,6,7,8 and Nuno D. Afonso 1* 3 4 1 TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal 5 2 Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal 6 3 Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. 7 Prof. Egas Moniz, 1649-028 Lisboa, Portugal 8 4 Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands. 9 5 Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and 10 Molecular Physiology of the Brain, Center for Biostructural Imaging of 11 Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany 12 6 CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de 13 Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 14 1169-056 Lisboa, Portugal. 15 7 Max Planck Institute for Experimental Medicine, Göttingen, Germany 16 8 Institute of Neuroscience, Medical School, Newcastle University, Framlington Place, 17 Newcastle Upon Tyne, NE2 4HH, UK 18 19 Equal contribution 20 & Current address: Battelle UK Limited, Chelmsford Business Park, Springfield, 21 Chelmsford CM2 5LB, United Kingdom 22 23 *Co-corresponding authors: 24 Dr. Nuno D. Afonso 25 TechnoPhage, SA, 26 Av. Prof. Egas Moniz 27 1649-028 Lisboa 28 Portugal 29 Telephone: +(351) 217999545 30 Email: [email protected] 31 32 Dr. Sara Sousa 33

3 4 - Newcastle University ePrints

Embed Size (px)

Citation preview

1

Identification of antiparkinsonian drugs in the 6-hydroxydopamine zebrafish model 1

Rita L. Vaz 1,2,†, Sara Sousa 1,†,*, Diana Chapela 1,3, Herma C. van der Linde 4, Rob 2

Willemsen 4, Ana D. Correia 3, &, Tiago F. Outeiro 5,6,7,8 and Nuno D. Afonso1* 3

4

1 TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal 5

2 Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal 6

3 Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. 7

Prof. Egas Moniz, 1649-028 Lisboa, Portugal 8

4 Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands. 9

5 Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and 10

Molecular Physiology of the Brain, Center for Biostructural Imaging of 11

Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany 12

6 CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de 13

Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 14

1169-056 Lisboa, Portugal. 15

7 Max Planck Institute for Experimental Medicine, Göttingen, Germany 16

8 Institute of Neuroscience, Medical School, Newcastle University, Framlington Place, 17

Newcastle Upon Tyne, NE2 4HH, UK 18

19

† Equal contribution 20

& Current address: Battelle UK Limited, Chelmsford Business Park, Springfield, 21

Chelmsford CM2 5LB, United Kingdom 22

23

*Co-corresponding authors: 24

Dr. Nuno D. Afonso 25

TechnoPhage, SA, 26

Av. Prof. Egas Moniz 27

1649-028 Lisboa 28

Portugal 29

Telephone: +(351) 217999545 30

Email: [email protected] 31

32

Dr. Sara Sousa 33

2

TechnoPhage, SA, 34

Av. Prof. Egas Moniz 35

1649-028 Lisboa 36

Portugal 37

Telephone: +(351) 217999545 38

Email: [email protected] 39

40

3

Abstract 41

Parkinson’s disease (PD) is known as a movement disorder due to characteristic motor 42

features. Existing therapies for PD are only symptomatic, and their efficacy decreases as 43

disease progresses. Zebrafish, a vertebrate in which parkinsonism has been modelled, 44

offers unique features for the identification of molecules with antiparkinsonian properties. 45

Here, we developed a screening assay for the selection of neuroactive agents with 46

antiparkinsonian potential. First, we performed a pharmacological validation of the 47

phenotypes exhibited by the 6-hydroxydopamine zebrafish model, by testing the effects 48

of known antiparkinsonian agents. These drugs were also tested for disease-modifying 49

properties by whole mount immunohistochemistry to TH+ neurons and confocal 50

microscopy in the dopaminergic diencephalic cluster of zebrafish. Next, we optimized a 51

phenotypic screening using the 6-hydroxydopamine zebrafish model and tested 1600 52

FDA-approved bioactive drugs. We found that 6-hydroxydopamine-lesioned zebrafish 53

larvae exhibit bradykinetic and dyskinetic-like behaviours that are rescued by the 54

administration of levodopa, rasagiline, isradipine or amantadine. The rescue of 55

dopaminergic cell loss by isradipine was also verified, through the observation of a higher 56

number of TH+ neurons in 6-OHDA-lesioned zebrafish larvae treated with this compound 57

as compared to untreated lesioned larvae. The phenotypic screening enabled us to identify 58

several compounds previously positioned for PD, as well as, new molecules with potential 59

antiparkinsonian properties. Among these, we selected stavudine, tapentadol and 60

nabumetone as the most promising candidates. Our results demonstrate the functional 61

similarities of the motor impairments exhibited by 6-hydroxydopamine-lesioned 62

zebrafish with mammalian models of PD and with PD patients, and highlights novel 63

molecules with antiparkinsonian potential. 64

65

Keywords: Drug screening, Parkinson’s disease, repositioning, zebrafish. 66

67

List of Abbreviations 68

6-OHDA, 6-hydroxydopamine AIMs, abnormal involuntary movements 69

BBB, blood brain barrier BDNF, brain derived neurotrophic factor 70

dpf, days post fertilization EM, embryo medium 71

4

L-dopa, levodopa PBS, phosphate buffer saline 72

PD, Parkinson’s disease PFA, paraformaldehyde 73

TH, tyrosine hydroxylase TU, Tubingen 74

75

5

Introduction 76

Parkinson’s disease (PD) is the most common movement disorder, affecting around 5 77

million people worldwide (Dorsey et al., 2007). The characteristic motor features of this 78

neurodegenerative disorder include bradykinesia, resting tremor, postural instability and 79

muscular rigidity. While the causes of PD are still not completely understood, the 80

degeneration of dopaminergic neurons in the substantia nigra is a major pathological 81

feature of the disease (Hirsch et al., 2013). Current pharmacological therapies, such as 82

levodopa (L-dopa), dopamine agonists (apomorphine, bromocriptine, among others), 83

monoamine oxidase (MAO)-B inhibitors (rasagiline, selegiline and safinamide) and 84

catechol-o-methyl transferase inhibitors (entacapone, tolcapone, nitecapone and 85

opicapone), address the loss of dopamine by either replacing it or controlling its 86

metabolism (Oertel and Schulz, 2016). Although these drugs substantially improve 87

quality of life, they lose efficacy as the disease progresses, and give rise to L-dopa induced 88

motor fluctuations and dyskinesia. Amantadine, an NMDA antagonist, is the most 89

effective anti-dyskinetic drug, but the associated side effects limit its use (Vijayakumar 90

and Jankovic, 2016). Non-dopaminergic agents, including A2A-receptor antagonists and 91

modulators of glutamate receptors, as well as nicotine, caffeine and isradipine are 92

currently under clinical trials (Oertel and Schulz, 2016). Despite the number of new 93

candidate agents that have successfully displayed antiparkinsonian effects in preclinical 94

studies, there is a growing demand for the development of alternative drugs. 95

Zebrafish is particularly suitable for large-scale drugs screening due to its small size, 96

transparency and high permeability to compounds diluted in the surrounding media. In 97

addition, studies have explored the organization and function of the zebrafish 98

dopaminergic system and suggest its overall conserved when compared to mammalian 99

vertebrates (Godoy et al., 2015; Rico et al., 2011; Rink and Wullimann, 2002, 2001), 100

rendering zebrafish as a practical and economic alternative vertebrate model for testing 101

the effect of neuroactive compounds (Parng et al., 2006; Sun et al., 2012). The sensitivity 102

of zebrafish to specific neurotoxins known to induce dopaminergic cell loss in rodent 103

models of PD has also been well validated (Anichtchik et al., 2004; Babu et al., 2016; 104

Wang et al., 2017). Specifically, the exposure of zebrafish larvae to 6-hydroxydopamine 105

(6-OHDA) induces the typical phenotypic features of PD, namely death of dopaminergic 106

neurons and bradykinesia (Feng et al., 2014). Furthermore, 6-OHDA-lesioned zebrafish 107

respond to antiparkinsonian compounds, such as levodopa or rasagiline (Cronin and 108

6

Grealy, 2017; Feng et al., 2014). Therefore, this model constitutes an excellent tool for 109

evaluating novel therapeutic options, not only as an alternative but also to complement 110

studies in mammalian models (Chong et al., 2013; Flinn et al., 2008; Vaz et al., 2018; 111

Wang et al., 2011; Xi et al., 2011; Zhang et al., 2012, 2011). 112

Using the 6-OHDA zebrafish model, we developed a platform for the selection of novel 113

potential antiparkinsonian molecules. First, we evaluated the effects of established anti-114

parkinsonian agents, such as L-dopa, rasagiline and isradipine on two specific measures 115

of motor performance, total distance moved and burst swimming. These three compounds 116

were also tested for their disease-modifying properties as determined by dopaminergic 117

neuronal loss. We then evaluated a new parameter, immobile events, as a surrogate 118

marker for dyskinetic-like behaviour (Babu et al., 2016), using the previously described 119

drugs and amantadine. Finally, we performed a phenotypic screen of 1600 FDA approved 120

drugs and selected the most promising candidates based on their ability to rescue motor 121

impairments in 6-OHDA-lesioned zebrafish larvae and evaluated their potential for 122

repositioning. 123

124

7

Material and methods 125

Chemicals and reagents 126

6-hydroxydopamine hydrobromide (6-OHDA) stock solution was prepared in 0.2% (w/v) 127

ascorbic acid and tested at 250, 500, 600, 750 and 800 µM, (Supplementary fig. S1A and 128

B). Tapentadol hydrochloride was purchased from Thonson Technology Ltd. (Shanghai, 129

China). The library of bioactive, FDA approved drugs was purchased from Microsource 130

(Gaylordsville, CT, USA). Nabumetone (Cat# N6142), stavudine (Cat# Y0000408), 6-131

OHDA (Cat# 162957), levodopa methyl ester (L-dopa, Cat# D9628), rasagiline (Cat# 132

SML0124), isradipine (Cat# I6658), paraformaldehyde (Cat# P6148), bovine serum 133

albumin (Cat# A2153) and triton X-100 (Cat# X100) were purchased from Sigma-134

Aldrich (St Louis, MO, USA). Amantadine was used directly from the library of drugs. 135

DABCO (Cat# 803456) was obtained from Millipore (Darmstadt, Germany). Mouse anti-136

tyrosine hydroxylase (TH, Cat# 22941) antibody was purchased from ImmunoStar 137

(Hudson, WI, USA) and AlexaFluor 568 (Cat# A11004) secondary antibody was 138

obtained from ThermoFisher Scientific (Waltham, MA, USA). All other reagents were 139

purchased from AppliChem (Darmstadt, Germany). 140

141

Zebrafish maintenance 142

Animal procedures were performed in accordance with the European Community 143

guidelines (Directive 2010/63/EU), Portuguese law on animal care (DL 113/2013), and 144

approved by the Instituto de Medicina Molecular Internal Committee and the Portuguese 145

Animal Ethics Committee (Direcção Geral de Alimentação e Veterinária). Tubingen (TU) 146

wild-type zebrafish were obtained from ZIRC (University of Oregon, Eugene, USA), 147

maintained and bred in constant conditions, by following standard guidelines for fish care 148

and maintenance protocols (Westerfield, 2000). 149

150

Treatment protocol 151

The protocol optimized for exposure of zebrafish larvae to the compounds was the same 152

for testing therapeutic controls, screening the library and determining dose-response 153

curves (Fig. 1). All experimental procedures were conducted between 8AM and 8PM. 154

8

Investigators were blind to the experimental groups during the screening protocol. 155

Briefly, zebrafish larvae were allowed to develop in embryo medium (EM [5 mM NaCl, 156

0.17 mM KCl, 0.33 mM CaCl2, 0.33 mM MgSO4, pH 7.4]) with 1µM methylene blue 157

until 4 days post fertilization (dpf). At this developmental stage, the zebrafish 158

catecholaminergic system is fully developed and the blood brain barrier (BBB) is already 159

functional (Du et al., 2016; Jeong et al., 2008; Kastenhuber et al., 2010). On the other 160

hand, 6-OHDA induces dopaminergic lesion in zebrafish larvae with up to 5 dpf, 161

suggesting the BBB permeability to this neurotoxin (Feng et al., 2014). 4 dpf zebrafish 162

larvae were arbitrarily distributed into 24-well plate (8 larvae/well) containing EM + 10 163

mM HEPES and treated with 750 µM of 6-OHDA, for 24h. The next day, compounds 164

were added to the medium at the established concentration and larvae (n= 8-16 larvae) 165

were incubated for 24h. To counteract zebrafish larvae neuroregenerative capability, 166

further 6-OHDA was added to each well simultaneously. For each experiment, healthy 167

larvae treated with 0.2% ascorbic acid (vehicle, n= 8-16 larvae) and untreated 6-OHDA-168

lesioned larvae (6-OHDA, n= 8-16 larvae) were used as controls. The sample size was 169

predetermined by non-statistical methods. Particularly, the exposure to 6-OHDA resulted 170

in a variable rate of survival dependent on zebrafish larvae fitness. For suitable statistical 171

analysis and in compliance with the 3Rs, this study was designed to ensure a minimum 172

of 5 zebrafish larvae per group and, given only experiments with more than 65% of 173

surviving animals were considered, minimum 8 zebrafish larvae were assigned to each 174

group. Behavioural assessment was performed at the end of the incubation with the 175

compounds. 176

9

177

Figure 1 – Schematic representation of the platform developed for the selection of 178

novel compounds with antiparkinsonian potential. The protocol of the phenotypic 179

screening starts with dopaminergic lesion by exposure of the zebrafish larvae to 6-OHDA. 180

The next day, the compounds are added to the medium and zebrafish larvae are incubated 181

for 24h. Finally, the behavioural analysis is conducted with a video tracking system and 182

drugs are selected for their capability to improve the motor performance of 6-OHDA-183

lesioned zebrafish larvae. 184

185

Behavioural analysis 186

Spontaneous locomotion was recorded using the DanioVision™ (Noldus Information 187

Technology, the Netherlands) automated tracking system for zebrafish larvae. Larvae 188

were allowed to swim freely in a 96-well plate (1 larva/well) with 1200 µl of EM + 10 189

mM HEPES and their swimming activity was tracked for 90 minutes, under 10 min light-190

dark cycles. 30 min acclimatization period was done followed by 60 min test. The 191

acquired track data was analysed using the Ethovision X.T. 10 software (Noldus, 192

10

Wageningen, Netherlands). Only swimming activity obtained in the dark periods (i.e. 193

under infrared light) were analysed (Esch et al., 2012). The parameters automatically 194

measured were total distance moved (mm), burst swimming (number of times larvae 195

reached velocities higher than 25 mm/s) and immobile events (mean time spent moving 196

less than 2 mm/s divided by number of events; protocol adapted from noldus technical 197

specifications (http://www.noldus.com/EthoVision-XT/Gathering-data; 198

http://www.noldus.com/animal-behavior-research/solutions/research-small-lab-199

animals/open-field-set)). To filter system noise, 0.2 mm was defined as the minimum 200

distance of movement. All values were normalized as a percentage of the mean of the 201

healthy control. 202

203

Dose-response curves 204

Dose-response curves were determined by adding to 6-OHDA-lesioned larvae different 205

doses of the compound under test (n= 8 larvae per condition). Dopaminergic lesion 206

induction, exposure to the compounds and behavioural analysis were conducted as 207

described above. Exceptionally, to depict L-dopa curve, larvae were treated 30 min prior 208

to the behavioural evaluation. The parameters were normalized as a percentage of the 209

mean of healthy control. Anti-TH immunostaining was performed after behavioural 210

evaluation to test the viability of dopaminergic neurons in zebrafish larvae treated with 211

an effective dose of the compound. 212

213

Screening of the library of FDA-approved compounds 214

To screen for compounds that can rescue 6-OHDA-induced motor impairments in 215

zebrafish, the treatment protocol and behavioural analysis were performed as described 216

above. 6-OHDA-lesioned larvae treated with 1250 µM of L-dopa were concomitantly 217

tested as positive control. L-dopa treated larvae were exposed to freshly prepared L-dopa, 218

30 min prior to behavioural evaluation. All experimental conditions (n= 8 larvae per 219

condition) were performed at 1% DMSO and with compounds at 25 µM, labelled with a 220

predefined code by an investigator not involved in the analysis, to ensure the blinding 221

procedure. Healthy larvae treated with vehicle and untreated 6-OHDA-lesioned larvae 222

were also exposed to 1% DMSO, a non-toxic concentration to zebrafish (Hallare et al., 223

11

2006). Compounds were considered as a positive hit, when there was a statistically 224

significant recovery of motor performance (total distance moved and burst swimming) in 225

the 3 independent experiments performed. 226

227

Evaluation of repositioning potential 228

After the first round of screenings, the feasibility of repositioning of the compounds that 229

rescued motor performance of 6-OHDA-lesioned zebrafish larvae was evaluated by an 230

investigator not involved in the screening experiments. Relevant data about the selected 231

molecules was searched in publicly available scientific databases and revised. All 232

compounds with active intellectual property, prior art for PD, prone to off-label use (e.g. 233

vitamins) and with safety concerns were excluded from further screenings. The 234

compounds that rescued motor performance in the three independent rounds of screenings 235

and feasible for repositioning were further investigated. Previous medical indications, 236

targets and BBBpermeability were evaluated. 237

238

Whole mount immunostaining and confocal microscopy 239

Whole mount immunostaining in zebrafish was performed as previously described (Wang 240

et al., 2011) with modifications. Zebrafish larvae were fixed overnight at 4 °C in 4% 241

paraformaldehyde (PFA). Larvae were then gradually dehydrated to methanol (Cat# 242

A3493) 100% and stored at −20 °C. For whole mount immunostaining, larvae were 243

gradually rehydrated to phosphate buffer saline (PBS). The tissue was then permeated in 244

100% acetone (Cat# 211007) for 15 min at -20°C, washed with 0.5% PBS-Triton X-100 245

and blocked in blocking solution (1% bovine serum albumin in PBS with 1% DMSO and 246

0.05% Triton X-100) for 2 hours at room temperature. Whole mount tissues were 247

incubated overnight at 4 ºC with anti-TH primary antibody (1:200 in blocking solution; 248

ImmunoStar Cat# 22941), washed in 0.1% PBS-Triton X-100, PBS and re-incubated 249

overnight at 4 ºC with AlexaFluor 568 secondary antibody (1:1000 in blocking solution; 250

ThermoFisher Scientific Cat# A-11004). After staining, larvae were washed in PBS and 251

flat-mounted on a fluorescent mounting medium with DABCO, under a stereoscope. Z-252

stack compositions of the dopaminergic diencephalic cluster were acquired in a confocal 253

microscope (Zeiss LSM 510 META, Carl Zeiss MicroImaging, Göttingen, Germany) 254

with 40x magnification. Dopaminergic cell content was assessed by counting the number 255

of TH+ cells from average intensity projections with Image J software (Schneider et al., 256

12

2012), as described by (Wang et al., 2011). The zebrafish dopaminergic diencephalic 257

cluster was outlined according to (Kastenhuber et al., 2010; Tay et al., 2011) (Figure 3D). 258

The number of TH+ neurons ranged between X and X in the 6-OHDA-lesioned zebrafish 259

larvae as compared to Y and Y in the healthy larvae. Results are expressed as a percentage 260

of the mean of TH+ cells in healthy controls. 261

262

Statistical analysis 263

Data analysis and graphical representation were performed using Prism 5 software 264

(GraphPad Software, Inc., San Diego, CA, USA). All values were normalized as 265

percentage of the mean of healthy control, because substantial variability was evidenced 266

in the behaviour of zebrafish larvae (previously described at this developmental stage 267

(Farrell et al., 2011). This variability was also observed in the number of TH+ neurons in 268

the dopaminergic diencephalic cluster, as well as, in the extent of the lesion induced by 269

different lots of 6-OHDA. This variability probably resulted from the instability of 6-270

OHDA, which is highly sensitive to light and easily oxidized. Values presented are mean 271

± s.e.m. of n animals. All statistical tests used were two-tailed and chosen according to 272

the distribution of the data. Mean comparisons between the different groups and 6-273

OHDA-lesioned untreated larvae were performed using one-way ANOVA with 274

Dunnett’s post-hoc test for experiments independently replicated or Kruskal-Wallis 275

ANOVA with Dunn’s post-hoc test for experiments performed once. Difference was 276

considered significant when P value < 0.05. 277

278

13

Results 279

L-dopa, rasagiline and isradipine rescue bradykinesia in zebrafish larvae 280

6-OHDA-lesioned zebrafish present bradykinetic-like behaviour that can be depicted 281

from quantification of the total distance moved (Feng et al., 2014). To further characterize 282

the motor impairments in this model, we calculated the number of times that larvae 283

reached velocities corresponding to escaping behaviour (>25 mm/s, burst swimming). 284

This parameter allows the evaluation of motor fitness. After 2 days of exposure to 6-285

OHDA, lesioned larvae exhibit a decrease of the burst swimming when compared to 286

healthy larvae, as well as, a reduction of the total distance moved (Fig. 2). We then 287

assessed whether, in this model, the motor impairments could be rescued by L-dopa (the 288

most effective treatment for PD to date, (Oertel and Schulz, 2016)), rasagiline (showed 289

disease-modifying properties in preclinical models of PD and induces motor 290

improvement in patients with PD (Oertel and Schulz, 2016)) and isradipine (in phase III 291

clinical trials as a disease-modifying agent for PD (Oertel and Schulz, 2016)). To 292

determine the effective doses, 6-OHDA-lesioned larvae were incubated with different 293

concentrations of each compound and the dose-response curves were outlined. First, we 294

tested a larger range of concentrations and determined the LD50 of each drug. The LD50 295

of L-dopa, rasagiline and isradipine on 6-OHDA-lesioned zebrafish larvae was above 296

5000, 150 and 40 µM, respectively. Then, we tested concentrations around the optimal 297

dose. As shown by the dose-response curve, L-dopa rescued the total distance moved at 298

125 and 1250 µM (Supplementary fig. S2A), but only at 125 µM there was rescue of the 299

burst swimming (Supplementary fig. S2B). Rasagiline was effective at lower 300

concentrations, 0.8 and 0.9 µM rescued the total distance moved and burst swimming 301

(Supplementary fig. S2C and D). Isradipine showed a wider range of effective doses, 0.04 302

µM of this drug rescued the total distance moved, but did not affect the burst swimming 303

(Supplementary fig. S2E). Additionally, several concentrations between 0.08 and 0.8 µM 304

rescued both parameters, with a peak effective dose at 0.5 µM (Supplementary fig. S2E 305

and F). To confirm the effective doses obtained from this data, three independent 306

experiments were performed with each of the compounds at two different concentrations, 307

125 and 1250 µM for L-dopa, 0.8 and 0.9 µM for rasagiline and 0.04 and 0.5 µM of 308

isradipine. 0.8 µM and 0.5 µM were confirmed to be the effective doses of rasagiline (Fig. 309

2C and D) and isradipine (Fig. 2E and F), respectively, as highlighted by the dose-310

response curves of each compound. In turn, while the dose of 125 µM was highlighted 311

14

by the dose-response curves of L-dopa, the dose of 1250 µM revealed more consistent 312

rescue of motor performance of 6-OHDA-lesioned zebrafish larvae in the three 313

independent experiments performed. The specificity of the motor effects induced by L-314

dopa, rasagiline and isradipine was then confirmed, as none of the compounds induced 315

behavioural changes in healthy zebrafish larvae (Supplementary fig. S3). This data shows 316

that the bradykinetic-like behaviour in 6-OHDA-lesioned zebrafish larvae can be 317

recovered by antiparkinsonian compounds, namely L-dopa, rasagiline and isradipine, and 318

that the effects observed are specific. 319

320

15

321

Figure 2 – Rescue of bradykinetic-like behaviour by L-dopa, rasagiline and 322

isradipine in 6-OHDA-lesioned zebrafish larvae. Motor performance depicted from 323

(A, C and E) total distance moved and (B, D and F) burst swimming in 6-OHDA-lesioned 324

zebrafish larvae treated with 1250 µM of L-dopa (n= 24 larvae), 0.8 µM of rasagiline (n= 325

36 larvae) and 0.5 µM of isradipine (n= 24 larvae), as compared to untreated larvae (n= 326

35-44 larvae). Percentage relative to the mean of vehicle treated larvae (healthy control, 327

n= 24-32 larvae). Mean ± s.e.m. of three independent experiments is presented. 328

***p<0.001, one-way ANOVA with Dunnett’s post-hoc test. 329

16

330

331

Isradipine rescues dopaminergic cell loss in zebrafish larvae 332

Previous studies demonstrated neuronal loss in the dopaminergic diencephalic cluster of 333

larvae lesioned with 6-OHDA (Feng et al., 2014). Therefore, we explored disease-334

modifying properties of L-dopa, rasagiline and isradipine in this model. Larvae were 335

treated with each compound at the effective dose and, after behavioural analysis, the 336

cellular content in the dopaminergic diencephalic cluster was determined by 337

immunohistochemistry against TH. 6-OHDA-lesioned larvae presented a reduction in the 338

number of dopaminergic cells, that was partially recovered by 0.5 µM of isradipine (Fig. 339

3C and D). On the contrary, neither L-dopa (Fig. 3A), nor rasagiline showed disease-340

modifying properties in this zebrafish model, at the optimal dose. Considering that, in 341

rats, rasagiline shows disease-modifying properties (Blandini et al., 2004), in addition to 342

the optimal dose, we also tested this compound at the concentration used in the phenotypic 343

screening, 25 µM, but no statistically significant cell recovery was observed (Fig. 3B). 344

This result indicates that the 6-OHDA-lesioned zebrafish larvae has a limited predictive 345

value to test disease-modifying agents for PD. 346

347

17

348

Figure 3 – Rescue of dopaminergic cell loss by isradipine, but not rasagiline and L-349

dopa in 6-OHDA-lesioned zebrafish larvae. Neuronal death depicted from percentage 350

of TH+ neurons in 6-OHDA-lesioned zebrafish larvae treated with (A) 1250 µM of L-351

dopa (n= 18 larvae), (B) 25 µM of rasagiline (n= 16 larvae) and (C) 0.5 µM of isradipine 352

(n= 18 larvae), as compared to untreated larvae (n= 19-32 larvae), determined by whole 353

mount immunohistochemistry for TH. Percentage relative to the mean of vehicle treated 354

larvae (healthy control, n= 18-21 larvae). Mean ± s.e.m. of three independent experiments 355

is presented. Ns – not significant, *p<0.05 and ***p<0.001, one-way ANOVA with 356

Dunnett’s post-hoc test. Representative Z-projections (D) of confocal stacks of 357

wholemount anti-TH immunohistochemistry in the dopaminergic diencephalic cluster of 358

6-OHDA-lesioned zebrafish larvae treated with isradipine (isradipine) as compared to 6-359

OHDA-lesioned untreated larvae (6-OHDA) and healthy larvae (vehicle). Ventral views, 360

anterior to the top and posterior to the bottom. Scale bar is 20 µM. 361

362

Zebrafish larvae exhibit dyskinetic-like behaviour that is reduced by isradipine and 363

amantadine 364

18

A dyskinetic-like behaviour has been previously described in MPTP-lesioned adult 365

zebrafish as diminished voluntary movements (Babu et al., 2016). Here, we investigated 366

whether 6-OHDA-lesioned larvae present a similar phenotype, observed as immobile 367

events, and tested the effects of L-dopa, rasagiline and isradipine. In addition, we tested 368

amantadine, the only anti-dyskinetic agent clinically available for PD (Vijayakumar and 369

Jankovic, 2016). 6-OHDA-lesioned larvae were exposed to each compound and their 370

behaviour was analysed. This zebrafish model showed an increase of the duration of the 371

immobile events, that was rescued by isradipine, at 0.06 µM (Fig. 4C), and amantadine, 372

at 25 µM (Fig. 4D). L-dopa was tested at 125, 250 (Fig. 4A) and 1250 µM, and rasagiline 373

at 0.95 (Fig. 4B) and 25 µM, but both compounds failed to alter the parameter, at the 374

doses tested. This result reveals that 6-OHDA-lesioned zebrafish larvae exhibit a 375

behaviour that resembles dyskinesia, sensitive to isradipine and amantadine. 376

377

378

19

Figure 4 – Rescue of dyskinetic-like behaviour by isradipine and amantadine, but 379

not L-dopa and rasagiline in 6-OHDA-lesioned zebrafish larvae. Dyskinetic-like 380

behaviour depicted from immobile events in 6-OHDA-lesioned zebrafish larvae treated 381

with (A) 250 µM of L-dopa (n= 30 larvae), (B) 0.95 µM of rasagiline (n= 31 larvae), (C) 382

0.06 µM of isradipine (n= 26 larvae) and (D) 25 µM of amantadine (n= 23 larvae), as 383

compared to untreated larvae (n= 44-49 larvae). Percentage relative to the mean of vehicle 384

treated larvae (healthy control, n= 23-31 larvae). Mean ± s.e.m. of three independent 385

experiments is presented. Ns – not significant, **p<0.01 and ***p<0.001, one-way 386

ANOVA with Dunnett’s post-hoc test. 387

388

A phenotypic-based screen identifies compounds with antiparkinsonian potential 389

To select candidate compounds capable of rescuing motor impairments in zebrafish 390

larvae lesioned with 6-OHDA, we developed a phenotypic assay that enabled the 391

screening of a library of 1600 FDA approved drugs (Fig. 1). The assay was three days 392

long and encompassed dopaminergic lesion with 6-OHDA (0h-24h), incubation with the 393

screening compounds (24h-48h) and behavioural evaluation (48h-72h). The locomotor 394

behaviour was tested by automatic measurement of total distance moved and burst 395

swimming, as previously described. From the 1600 compounds screened (Fig. 5), 258 396

(16%) rescued motor impairments in 6-OHDA-lesioned zebrafish larvae during the first 397

round of experiments performed (Table 1). These compounds were then evaluated for 398

repositioning and 83 (32%) were excluded from further analysis, based on pre-determined 399

exclusion criteria. From these 83 compounds, 26 (31%) had prior art for PD, 25 (30%) 400

had an active patent for other indications or were prone to off-label use, and 7 (8%) raised 401

safety concerns (Table 2). Nevertheless, these compounds were useful for validation of 402

the screen. This was the case, for example, of caffeine and carbinoxamine maleate, which 403

were blindly selected during the screen for rescuing motor impairments in 6-OHDA-404

lesioned zebrafish larvae. In contrast, bromocriptine mesylate and apomorphine 405

hydrochloride, two dopamine agonists used in the clinic, were also screened, but showed 406

no effect in the 6-OHDA-lesioned zebrafish larvae at the concentration tested. In the 407

second and third rounds of screenings, 78 (30%) and 23 (29%) compounds, respectively, 408

rescued motor impairments in 6-OHDA-lesioned zebrafish larvae (Table 1). After further 409

analysis, which included the evaluation of previous medical indications and targets and 410

20

BBB permeability, 3 drugs (13%), stavudine, tapentadol and nabumetone, showed to be 411

particularly promising for further experiments (Table 3). These three compounds had 412

minimally explored targets for the treatment of PD, high permeability to the BBB and no 413

further concerns from previous indications. The other 20 compounds had either no known 414

targets, low or non-described BBB permeability or concerns regarding safety, and were 415

discarded. Overall, the screening protocol developed was suitable for a quick selection of 416

neuroactive drugs with antiparkinsonian potential, but also presented limitations 417

concerning the activity of dopamine agonists. 418

419

21

420

421

Round I Round II Round III

-3 Intelectual property issues

-2 Toxic to larvae

-1 No rescue of motor performance

0 Rescue of burst swimming (BS)

1 Rescue of total distance moved (TDM)

2 Rescue of TDM + BS

Not Applicable

22

Figure 5 – Hit map representation of the screening of 1600 bioactive drugs. Each 422

square corresponds to a compound, identified as rescuing none of the parameters 423

measured (burst swimming, BS, and total distance moved, TDM), rescuing one of the two 424

parameters measured or rescuing both parameters measured in 6-OHDA-lesioned 425

zebrafish larvae. Only compounds that rescued both, BS and TDM, are represented in the 426

subsequent round of screenings. Compounds that presented toxic effects on zebrafish 427

larvae and intellectual property concerns, and therefore excluded from further screenings 428

are also represented. 429

430

431

Table 1 – Overview results of the screening of 1600 bioactive drugs. Number and 432

percentage of compounds with no effects, and with effects on one or both parameters 433

(burst swimming, BS, and total distance moved, TDM) measured during each round of 434

the screening assay, to depict motor performance in 6-OHDA-lesioned zebrafish larvae. 435

Number and percentage of compounds with toxic effects on zebrafish larvae and 436

intellectual property concerns also indicated. Percentage relative to the total of 437

compounds screened (1600) for round I and to the number of positive hits in the previous 438

round, for rounds II and III. 439

440

441

Table 2 – Intellectual property issues considered for exclusion of positive hits from 442

round I of phenotypic screenings. Number and percentage of compounds with prior art 443

23

for PD, active patent, prone to off-label use and safety concerns. Percentage relative to 444

the total of compounds selected for rescuing motor performance in 6-OHDA-lesioned 445

zebrafish larvae during round I of screenings. 446

447

24

448

Hits Targets BBB permeability

1 Human immunodeficiency virus type 1 protease inhibitor

Nuclear receptor subfamily 1 group I member 2 activator

Low

2 Tubulin beta-1 chain

Apoptosis regulator Bcl-2

Microtubule-associated protein 2 and 4

Microtubule-associated protein tau

Nuclear receptor subfamily 1 group I member 2

High

3 Not found High

4 Sodium channel protein type 5 subunit alpha antagonist High

5 Not found Not found

6 cAMP and cGMP-specific 3',5'-cyclic phosphodiesterases inhibitor

Adenosine deaminase inhibitor

Calcipressin-1

Alpha-1-acid glycoprotein 1

Low

7 Not found Not found

8 5-hydroxytryptamine receptors agonist

Alpha-2A adrenergic receptor agonist

High

9 Cross-linking/alkylation of DNA

Nuclear receptor subfamily 1 group I member 2

High

10 Arachidonate 5-lipoxygenase inhibitor

Prostaglandin G/H synthase 1 inhibitor

High

11 Prostaglandin G/H synthase 1 and 2 inhibitor High

12 Not found Not found

13 Reverse transcriptase/RNaseH inhibitor High

14 Mu-type opioid receptor agonist

Sodium-dependent noradrenaline transporter inhibitor

Kappa-type opioid receptor

Delta-type opioid receptor

5-hydroxytryptamine receptor 3A

Sodium-dependent serotonin transporter inhibitor

High

15 Alpha-1A, 1D and 1B adrenergic receptor antagonist Low

16 3 beta-hydroxysteroid dehydrogenase/Delta 5-->4-isomerase type 1 and 2 inhibitor

Estrogen receptor alpha and beta allosteric modulator

Low

17 Not found Not found

18 Not found Not found

19 Beta-1 and 2 adrenergic receptor antagonist

Alpha-1A, 1B, 1D, 2A-2C adrenergic receptor antagonist

NADH dehydrogenase [ubiquinone] 1 subunit C2 inhibitor

Vascular endothelial growth factor A

Natriuretic peptides B

Gap junction alpha-1 protein

Potassium voltage-gated channel subfamily H member 2 inhibitor

Vascular cell adhesion protein 1 inhibitor

E-selectin inhibitor

Hypoxia-inducible factor 1-alpha modulator

Inward rectifier potassium channel 4

Low

20 Penicillin binding protein 2a, 1A and 1B inhibitor

Peptidoglycan synthase FtsI inhibitor

D-alanyl-D-alanine carboxypeptidase DacA and DacC inhibitor

High

21 Muscarinic acetylcholine receptor M1 antagonist High

22 Not found Not found

23 Potassium voltage-gated channel subfamily H member 2 Not found

25

Table 3 – Overview of the 23 lead compounds of the screening assay. Targets and 449

BBB permeability of the 23 compounds selected for rescuing motor performance in 6-450

OHDA-lesioned zebrafish larvae during the screening assay. Data obtained from an initial 451

search at https://www.drugbank.ca/. 452

453

454

Three new compounds display antiparkinsonian activity in 6-OHDA-lesioned 455

zebrafish larvae 456

Stavudine is a nucleoside reverse transcriptase inhibitor, that stimulates the production of 457

brain derived neurotrophic factor (BDNF) (Renn et al., 2011), tapentadol is a µ-opioid 458

receptor agonist and norepinephrine reuptake inhibitor (Tzschentke et al., 2007) and 459

nabumetone is a cyclooxygenase enzyme inhibitor (Davies, 1997). These three 460

compounds were selected from the screening assay, for their antiparkinsonian properties 461

in 6-OHDA-lesioned zebrafish larvae (Supplementary fig. S4). Next, we determined the 462

effective dose of each of the purified compounds and evaluated their effect on dyskinetic-463

like behaviour. 6-OHDA-lesioned larvae were treated with different concentrations of 464

each compound and behavioural analysis was conducted. For both parameters, total 465

distance moved and burst swimming, we observed a significant recovery of larvae treated 466

with 50 µM of stavudine (Fig. 6A and B), 49 µM of tapentadol (Fig. 6D and E) and 0.9 467

µM of nabumetone (Fig. 6G and H). Stavudine and tapentadol also rescued dyskinetic-468

like behaviour at these doses, as opposed to nabumetone, in 6-OHDA-lesioned zebrafish 469

larvae (Fig. 6C, F and I). This data confirms that stavudine, tapentadol and nabumetone 470

are potential antiparkinsonian agents and supports further studies in mammalian models 471

of PD. 472

473

26

474

Figure 6 – Rescue of motor impairments by stavudine, tapentadol and nabumetone 475

in 6-OHDA-lesioned zebrafish larvae. Motor performance and dyskinetic-like 476

behaviour depicted from (A, D and G) total distance moved, (B, E and H) burst swimming 477

and (C, F and I) immobile events in 6-OHDA-lesioned zebrafish larvae treated with 50 478

µM of stavudine (n= 38-40 larvae), 49 µM of tapentadol (n= 32-46 larvae) and 0.9 µM 479

of nabumetone (n= 38-49 larvae), as compared to untreated larvae (n= 30-66 larvae). 480

Percentage relative to the mean of vehicle treated larvae (healthy control, n= 23-31 481

larvae). Mean ± s.e.m. of three independent experiments is presented. Ns – not significant, 482

and ***p<0.001, one-way ANOVA with Dunnett’s post-hoc test. 483

484

27

Discussion 485

Here, we developed a phenotypic screening assay for the selection of new drugs with 486

antiparkinsonian potential, using 6-OHDA-lesioned zebrafish larvae. This model exhibits 487

death of dopaminergic neurons, accompanied by a decrease of dopamine levels and by 488

motor impairments (Anichtchik et al., 2004; Feng et al., 2014; Vijayanathan et al., 2017). 489

These phenotypes can be rescued by antiparkinsonian compounds (L-dopa and rasagiline) 490

(Cronin and Grealy, 2017; Feng et al., 2014). After deeper characterization of this model, 491

we used it for screening of 1600 FDA approved drugs and identified 23 drugs with 492

antiparkinsonian potential. We selected stavudine, tapentadol and nabumetone due to 493

their effects on relevant targets for the treatment of PD. 494

In agreement with previous studies (Anichtchik et al., 2004; Cronin and Grealy, 2017; 495

Feng et al., 2014), we observed that zebrafish larvae exposed to 6-OHDA display 496

bradykinetic-like behaviour and loss of dopaminergic neurons. We also introduce the 497

burst swimming as a suitable parameter to assess motor performance. This parameter 498

measures the number of times that larvae reach velocities correspondent to escaping 499

behaviour (>25 mm/s). Burst swimming has been catalogued as a swimming behaviour 500

of zebrafish larvae, which exhibit locomotion at slow speed, sometimes followed by a 501

faster and more vigorous swimming, that is typically above 20 mm/s (Budick and 502

O’Malley, 2000; Kalueff et al., 2013). Importantly, we showed that burst swimming can 503

be rescued by L-dopa and rasagiline, as previously demonstrated for total distance moved 504

(Cronin and Grealy, 2017; Feng et al., 2014). 505

Since isradipine has been described as a disease-modifying agent for PD (Chan et al., 506

2007; Ilijic et al., 2011), we also explored the antiparkinsonian potential of this drug. We 507

report, for the first time, that isradipine reduces the loss of dopaminergic neurons and 508

rescues bradykinetic-like behaviour in 6-OHDA-lesioned zebrafish larvae. It has been 509

suggested that isradipine (calcium channel blocker) prevents the death of dopaminergic 510

cells through the reduction of calcium influx in neurons and consequently the decrease of 511

mitochondrial activity (Chan et al., 2007). Therefore, isradipine could have induced 512

changes in the mitochondrial activity of zebrafish larvae which decreased the oxidative 513

stress and cytotoxicity induced by 6-OHDA over dopaminergic neurons. In contrast, we 514

did not detect disease-modifying effect of rasagiline. This finding differs from previous 515

reports in zebrafish and mice (Blandini et al., 2004; Cronin and Grealy, 2017), likely due 516

28

to differences in the treatment protocols adopted. The time between dopaminergic lesion 517

and the exposure to rasagiline is a key factor that may dramatically influence the outcome 518

in the zebrafish model, where the regeneration capacity is greater in comparison to 519

mammals (Zupanc, 2008). In fact, rasagiline failed to demonstrate disease-modifying 520

properties in humans and isradipine is still under clinical evaluation (Oertel and Schulz, 521

2016; Olanow et al., 2009). 522

Previously, dyskinetic-like behaviour in MPTP-lesioned adult zebrafish was reported 523

(Babu et al., 2016). Now, we observed that 6-OHDA-lesioned zebrafish larvae exhibit a 524

similar behaviour, as measured by the average duration of immobile events (velocity <2 525

mm/s). While the evaluation of an hyperkinetic disease through complete lack of 526

movement might appear counter-intuitive, there is already evidence supporting this 527

approach in a zebrafish model of dystonia (Friedrich et al., 2012). On the other hand, 528

dyskinesia only occurs in PD patients under L-dopa or dopamine-agonist treatment. 529

Therefore, although the parameter we measured cannot be directly related with L-dopa-530

induced dyskinesia, it reports on non-induced dyskinesias, indicative of abnormal 531

plasticity of the motor circuitry. This could result from the dysregulation of different 532

neurotransmitters, sensitive to the impairment of the dopaminergic system, that are 533

essential for generating a normal swimming behaviour. Consistently, L-dopa and 534

rasagiline, two drugs that rely on the dopaminergic system, did not alter the average 535

duration of immobile events in 6-OHDA-lesioned zebrafish larvae. Accordingly, these 536

drugs do not have an anti-dyskinetic effect in humans (Pistacchi et al., 2014). In turn, one 537

would expect this parameter to worsen in larvae treated with L-dopa. However, this may 538

only be observed with chronic treatment, so it could not be assessed. Additionally, we 539

found that the drugs with non-dopaminergic targets, isradipine and amantadine, reduced 540

the average duration of immobile events. Amantadine is widely used in the clinic as an 541

anti-dyskinetic agent for PD (Vijayakumar and Jankovic, 2016). In the case of isradipine, 542

the pre-clinical evidence for anti-dyskinetic properties is limited (Rylander et al., 2009; 543

Schuster et al., 2009) and reports of this therapeutic indication in humans do not exist to 544

date. Whereas, amantadine is an NMDA antagonist and could have balanced the 545

dysregulation induced by the impaired dopaminergic system in zebrafish, isradipine does 546

not act in a specific neuronal circuitry. Therefore, the changes observed in the duration 547

of immobile events of zebrafish larvae treated with isradipine could have resulted from 548

the rescue of dopaminergic neurons and, consequently, from a lower dysregulation of this 549

29

and other neuronal systems. Additional studies will be necessary to further investigate the 550

predictive value of this parameter. As such, the phenotypic screening assay we developed 551

relied on the total distance moved and burst swimming parameters for the selection of 552

compounds with antiparkinsonian potential. 553

The phenotypic screening of 1600 FDA approved drugs, resulted in the identification of 554

26 drugs (31% of the drugs selected during round I) with prior art for PD, revealing the 555

sensitivity of the assay. In contrast, the screening set up failed to identify some dopamine 556

agonists used as monotherapy or in combination with L-dopa (Oertel and Schulz, 2016). 557

In zebrafish, eight proteins homologous to the human dopamine receptors have been 558

identified (Panula et al., 2010), but their affinity for dopamine agonists has not been 559

explored. Therefore, one possibility is that the dose tested during the screening was not 560

effective. Previously, it was reported that the non-selective dopamine agonist, 561

apomorphine, has a biphasic effect on the locomotor activity of zebrafish, that depends 562

on the dose applied (Irons et al., 2013). This dose-dependent effect has also been 563

evidenced in mice treated with dopamine receptor agonists (Lundblad et al., 2005). All 564

drugs were screened at a concentration of 25 µM, the highest concentration more 565

commonly used for drug screenings in zebrafish (Rennekamp and Peterson, 2015). 566

Importantly, only 16% of the screened drugs showed toxic effects in these conditions, 567

which did not compromise the screening. 568

Overall, the phenotypic screening identified 23 drugs (1.4% of the total drugs screened) 569

with antiparkinsonian potential in 6-OHDA-lesioned zebrafish larvae. Previous studies 570

allowed the analysis of targets, BBB permeability and clinical indications, and selection 571

of three drugs that were considered particularly interesting for further studies. Stavudine 572

is a reverse transcriptase inhibitor and previous reports have described the stimulation of 573

BDNF expression by this drug (Renn et al., 2011). Neurotrophic factors have proven their 574

efficacy in animal models of PD, but their inability to cross the BBB has limited their 575

application (Aron and Klein, 2011). In turn, tapentadol is a µ-opioid receptor agonist and 576

norepinephrine reuptake inhibitor (Tzschentke et al., 2007), two pathways long 577

implicated in PD (Espay et al., 2014; Samadi et al., 2006). Lastly, nabumetone is a 578

prostaglandin synthase inhibitor with anti-inflammatory properties (Davies, 1997). 579

Neuroinflammation is a key pathogenic mechanism of PD and anti-inflammatory agents 580

have also shown promising results in preclinical studies, although convincing clinical data 581

is still missing (Athauda and Foltynie, 2015). Interestingly, the three drugs rescued 582

30

bradykinetic-like behaviour, but nabumetone failed to alter the duration of immobile 583

events in 6-OHDA-lesioned zebrafish larvae. As discussed above, the target described for 584

nabumetone is not specific of a neuronal circuitry, while stavudine induces the expression 585

of BDNF and tapentadol acts on the opioid and noradrenergic systems. Further studies 586

need to be conducted to determine the exact mechanism of action behind the therapeutic 587

potential of these three compounds and to validate their antiparkinsonian efficacy in other 588

vertebrate models. Since the effects of stavudine, tapentadol and nabumetone in the motor 589

performance of healthy zebrafish larvae were not assessed, further studies should 590

determine the specificity of the therapeutic effects to dopaminergic disfunction. In 591

contrast to other screenings previously reported (Buckley et al., 2010; Parng et al., 2006; 592

Robertson et al., 2016; Sun et al., 2012), our selection of drugs was completely agnostic 593

to target or mechanism of action, which is a key distinction of the phenotypic screening, 594

as it can result in the identification of drugs with new mechanisms of action. 595

596

31

Conclusions 597

In general, our study provides further demonstration that the 6-OHDA-lesioned zebrafish 598

larvae exhibit bradykinetic-like behaviours that are sensitive to the motor improvement 599

of antiparkinsonian compounds. A dyskinetic-like behaviour was also observed, but 600

further investigations will be necessary in order to fully demonstrate the predictive value 601

of this parameter in zebrafish. Although we did not detect disease-modifying properties 602

for rasagiline or anti-bradykinetic properties for dopamine agonists, this might be due to 603

the screening protocol, and will require further investigation. Nevertheless, the 604

phenotypic screening we describe is a valid strategy for rapid selection of potential 605

antiparkinsonian agents. Importantly, it is simple, is based in objective and automated 606

parameters, and does not require invasive or stressful manipulation of the animals. The 607

screening led to the identification of three neuroactive drugs, stavudine, tapentadol and 608

nabumetone, and further studies regarding their mechanism of action could lead to the 609

discovery of targets or pathways relevant for PD mechanisms. 610

32

Acknowledgements 611

Funding: This study was sponsored by TechnoPhage S.A. and Eurostars program 612

(ES#5553) from EUREKA (a program run by the European Commission). Rita L. Vaz 613

was supported by a grant (SFRH/BD/78077/2011) from Fundação para a Ciência e 614

Tecnologia. Tiago F. Outeiro was supported by the DFG Center for Nanoscale 615

Microscopy and Molecular Physiology of the Brain (CNMPB). 616

617

Conflict of interest: S.S., D.C. and N.D.A. were employees of Technophage SA, at the 618

time of the study. The other authors declare no conflicts of interest. 619

620

Author contributions: S.S., R.W., T.F.O. and N.D.A. did study conception and design. 621

R.L.V., D.C. and S.S. performed experiments, did data acquisition and analysed the data. 622

All authors interpreted the data. R.L.V. drafted the paper. S.S. and N.D.A. did critical 623

revision. 624

625

33

References 626

Anichtchik, O. V., Kaslin, J., Peitsaro, N., Scheinin, M., Panula, P., 2004. 627

Neurochemical and behavioural changes in zebrafish Danio rerio after systemic 628

administration of 6-hydroxydopamine and 1-methyl-4-phenyl-1,2,3,6-629

tetrahydropyridine. J. Neurochem. 88, 443–453. doi:10.1046/j.1471-630

4159.2003.02190.x 631

Aron, L., Klein, R., 2011. Repairing the parkinsonian brain with neurotrophic factors. 632

Trends Neurosci. 34, 88–100. doi:10.1016/j.tins.2010.11.001 633

Athauda, D., Foltynie, T., 2015. The ongoing pursuit of neuroprotective therapies in 634

Parkinson disease. Nat. Rev. Neurol. 11, 25–40. doi:10.1038/nrneurol.2014.226 635

Babu, N.S., Murthy, C.L.N., Kakara, S., Sharma, R., Swamy, C.V.B., Idris, M.M., 636

2016. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine induced Parkinson’s disease 637

in zebrafish. Proteomics 16, 1407–1420. doi:10.1002/pmic.201500291 638

Blandini, F., Armentero, M.T., Fancellu, R., Blaugrund, E., Nappi, G., 2004. 639

Neuroprotective effect of rasagiline in a rodent model of Parkinson’s disease. Exp. 640

Neurol. 187, 455–459. doi:10.1016/j.expneurol.2004.03.005 641

Buckley, C.E., Marguerie, A., Roach, A.G., Goldsmith, P., Fleming, A., Alderton, 642

W.K., Franklin, R.J.M., 2010. Drug reprofiling using zebrafish identifies novel 643

compounds with potential pro-myelination effects. Neuropharmacology 59, 149–644

159. doi:10.1016/j.neuropharm.2010.04.014 645

Budick, S.A., O’Malley, D.M., 2000. Locomotor repertoire of the larval zebrafish: 646

swimming, turning and prey capture. J. Exp. Biol. 203, 2565–2579. 647

doi:10.1242/jeb.01529 648

Chan, C.S., Guzman, J.N., Ilijic, E., Mercer, J.N., Rick, C., Tkatch, T., Meredith, G.E., 649

Surmeier, D.J., 2007. “Rejuvenation” protects neurons in mouse models of 650

Parkinson’s disease. Nature 447, 1081–1086. doi:10.1038/nature05865 651

Chong, C., Zhou, Z., Razmovski-Naumovski, V., Cui, G., Zhang, L., Sa, F., Hoi, P., 652

Chan, K., Lee, S.M., 2013. Danshensu protects against 6-hydroxydopamine-653

induced damage of PC12 cells in vitro and dopaminergic neurons in zebrafish. 654

34

Neurosci. Lett. 543, 121–125. doi:10.1016/j.neulet.2013.02.069 655

Cronin, A., Grealy, M., 2017. Neuroprotective and Neuro-restorative Effects of 656

Minocycline and Rasagiline in a Zebrafish 6-Hydroxydopamine Model of 657

Parkinson’s Disease. Neuroscience 367, 34–46. 658

doi:10.1016/j.neuroscience.2017.10.018 659

Davies, N.M., 1997. Clinical pharmacokinetics of nabumetone. The dawn of selective 660

cyclo-oxygenase-2 inhibition? Clin. Pharmacokinet. 33, 404–416. 661

Dorsey, E.R., Constantinescu, R., Thompson, J.P., Biglan, K.M., Holloway, R.G., 662

Kieburtz, K., Marshall, F.J., Ravina, B.M., Schifitto, G., Siderowf, A., Tanner, 663

C.M., 2007. Projected number of people with Parkinson disease in the most 664

populous nations, 2005 through 2030. Neurology 68, 384–386. 665

doi:10.1212/01.wnl.0000247740.47667.03 666

Du, Y., Guo, Q., Shan, M., Wu, Y., Huang, S., Zhao, H., 2016. Spatial and Temporal 667

Distribution of Dopaminergic Neurons during Development in Zebrafish 10, 1–7. 668

doi:10.3389/fnana.2016.00115 669

Esch, C. De, Linde, H. Van Der, Slieker, R., Willemsen, R., Wolterbeek, A., 670

Woutersen, R., Groot, D. De, 2012. Locomotor activity assay in zebrafish larvae: 671

Influence of age, strain and ethanol. Neurotoxicol. Teratol. 34, 425–433. 672

doi:10.1016/j.ntt.2012.03.002 673

Espay, A.J., Lewitt, P.A., Kaufmann, H., 2014. Norepinephrine deficiency in 674

Parkinson’s disease: The case for noradrenergic enhancement. Mov. Disord. 29, 675

1710–1719. doi:10.1002/mds.26048 676

Farrell, T.C., Cario, C.L., Milanese, C., Vogt, A., Jeong, J.-H., Burton, E.A., 2011. 677

Evaluation of spontaneous propulsive movement as a screening tool to detect 678

rescue of Parkinsonism phenotypes in zebrafish models. Neurobiol. Dis. 44, 9–18. 679

doi:10.1016/j.nbd.2011.05.016 680

Feng, C., Wen, Z., Huang, S., Hung, H., Chen, C., Yang, S., Chen, N., Wang, H., Hsiao, 681

C., Chen, W., 2014. Effects of 6-hydroxydopamine exposure on motor activity and 682

biochemical expression in zebrafish (Danio rerio) larvae. Zebrafish 11, 227–239. 683

doi:10.1089/zeb.2013.0950 684

35

Flinn, L., Bretaud, S., Lo, C., Ingham, P.W., Bandmann, O., 2008. Zebrafish as a new 685

animal model for movement disorders. J. Neurochem. 106, 1991–1997. 686

doi:10.1111/j.1471-4159.2008.05463.x 687

Friedrich, T., Lambert, A.M., Masino, M.A., Downes, G.B., 2012. Mutation of 688

zebrafish dihydrolipoamide branched-chain transacylase E2 results in motor 689

dysfunction and models maple syrup urine disease. Dis. Model. Mech. 5, 248–258. 690

doi:10.1242/dmm.008383 691

Godoy, R., Noble, S., Yoon, K., Anisman, H., Ekker, M., 2015. Chemogenetic ablation 692

of dopaminergic neurons leads to transient locomotor impairments in zebrafish 693

larvae. J. Neurochem. 135, 249–260. doi:10.1111/jnc.13214 694

Hallare, A., Nagel, K., Köhler, H.R., Triebskorn, R., 2006. Comparative embryotoxicity 695

and proteotoxicity of three carrier solvents to zebrafish (Danio rerio) embryos. 696

Ecotoxicol. Environ. Saf. 63, 378–388. doi:10.1016/j.ecoenv.2005.07.006 697

Hirsch, E.C., Jenner, P., Przedborski, S., 2013. Pathogenesis of Parkinson’s disease. 698

Mov. Disord. 28, 24–30. doi:10.1002/mds.25032 699

Ilijic, E., Guzman, J.N., Surmeier, D.J., 2011. The L-type channel antagonist isradipine 700

is neuroprotective in a mouse model of Parkinson’s disease. Neurobiol. Dis. 43, 701

364–371. doi:10.1097/MPG.0b013e3181a15ae8.Screening 702

Irons, T.D., Kelly, P.E., Hunter, D.L., MacPhail, R.C., Padilla, S., 2013. Acute 703

administration of dopaminergic drugs has differential effects on locomotion in 704

larval zebrafish. Pharmacol. Biochem. Behav. 103, 792–813. 705

doi:10.1016/j.pbb.2012.12.010 706

Jeong, J., Kwon, H., Ahn, J., Kang, D., Kwon, S., Park, J.A., Kim, K., 2008. Functional 707

and developmental analysis of the blood-brain barrier in zebrafish. Brain Res. Bull. 708

75, 619–628. doi:10.1016/j.brainresbull.2007.10.043 709

Kalueff, A. V, Gebhardt, M., Stewart, A.M., Cachat, J.M., Brimmer, M., Chawla, J.S., 710

Craddock, C., Kyzar, E.J., Roth, A., Landsman, S., Gaikwad, S., Robinson, K., 711

Baatrup, E., Tierney, K., Shamchuk, A., Norton, W., Miller, N., Nicolson, T., 712

Braubach, O., Gilman, C.P., Pittman, J., Rosemberg, D.B., Gerlai, R., Echevarria, 713

D., Lamb, E., Neuhauss, S.C.F., Weng, W., Bally-Cuif, L., Schneider, H., 2013. 714

36

Towards a comprehensive catalog of zebrafish behavior 1.0 and beyond. Zebrafish 715

10, 70–86. doi:10.1089/zeb.2012.0861 716

Kastenhuber, E., Kratochwil, C.F., Ryu, S., Schweitzer, J., Driever, W., 2010. Genetic 717

dissection of dopaminergic and noradrenergic contributions to catecholaminergic 718

tracts in early larval zebrafish. J. Comp. Neurol. 518, 439–458. 719

doi:10.1002/cne.22214 720

Lundblad, M., Usiello, A., Carta, M., Håkansson, K., Fisone, G., Cenci, M.A., 2005. 721

Pharmacological validation of a mouse model of L-DOPA-induced dyskinesia. 722

Exp. Neurol. 194, 66–75. doi:10.1016/j.expneurol.2005.02.002 723

Oertel, W., Schulz, J.B., 2016. Current and experimental treatments of Parkinson 724

disease: A guide for neuroscientists. J. Neurochem. 1–13. doi:10.1111/jnc.13750 725

Olanow, C.W., Rascol, O., Hauser, R., Feigin, P.D., Jankovic, J., Lang, A., Langston, 726

W., Melamed, E., Poewe, W., Stocchi, F., Tolosa, E., ADAGIO Study 727

Investigators, 2009. A Double-Blind, Delayed-Start Trial of Rasagiline in 728

Parkinson’s Disease. N. Engl. J. Med. 361, 1268–1278. 729

doi:10.1056/NEJMoa0809335 730

Panula, P., Chen, Y.C., Priyadarshini, M., Kudo, H., Semenova, S., Sundvik, M., 731

Sallinen, V., 2010. The comparative neuroanatomy and neurochemistry of 732

zebrafish CNS systems of relevance to human neuropsychiatric diseases. 733

Neurobiol. Dis. 40, 46–57. doi:10.1016/j.nbd.2010.05.010 734

Parng, C., Ton, C., Lin, Y.-X., Roy, N.M., McGrath, P., 2006. A zebrafish assay for 735

identifying neuroprotectants in vivo. Neurotoxicol. Teratol. 28, 509–516. 736

doi:10.1016/j.ntt.2006.04.003 737

Pistacchi, M., Martinello, F., Gioulis, M., Marsala, S.Z., 2014. Rasagiline and Rapid 738

Symptomatic Motor Effect in Parkinson’s Disease: Review of Literature. Neurol. 739

Ther. 3, 41–66. doi:10.1007/s40120-013-0014-1 740

Renn, C.L., Leitch, C.C., Lessans, S., Rhee, P., Mcguire, W.C., Smith, B.A., Traub, 741

R.J., Dorsey, S.G., 2011. Brain-derived neurotrophic factor modulates 742

antiretroviral-induced mechanical allodynia in the mouse. J. Neurosci. Res. 89, 743

1551–1565. doi:10.1002/jnr.22685 744

37

Rennekamp, A.J., Peterson, R.T., 2015. 15 Years of Zebrafish Chemical Screening. 745

Curr. Opin. Chem. Biol. 24, 58–70. doi:10.1016/j.cbpa.2014.10.025 746

Rico, E.P., Rosemberg, D.B., Seibt, K.J., Capiotti, K.M., Da Silva, R.S., Bonan, C.D., 747

2011. Zebrafish neurotransmitter systems as potential pharmacological and 748

toxicological targets. Neurotoxicol. Teratol. 33, 608–617. 749

doi:10.1016/j.ntt.2011.07.007 750

Rink, E., Wullimann, M.F., 2002. Connections of the ventral telencephalon and tyrosine 751

hydroxylase distribution in the zebrafish brain (Danio rerio) lead to identification 752

of an ascending dopaminergic system in a teleost. Brain Res. Bull. 57, 385–387. 753

doi:10.1016/S0361-9230(01)00696-7 754

Rink, E., Wullimann, M.F., 2001. The teleostean (zebrafish) dopaminergic system 755

ascending to the subpallium (striatum) is located in the basal diencephalon 756

(posterior tuberculum). Brain Res. 889, 316–330. doi:10.1016/S0006-757

8993(00)03174-7 758

Robertson, A.L., Ogryzko, N. V., Henry, K.M., Loynes, C.A., Foulkes, M.J., Meloni, 759

M.M., Wang, X., Ford, C., Jackson, M., Ingham, P.W., Wilson, H.L., Farrow, 760

S.N., Solari, R., Flower, R.J., Jones, S., Whyte, M.K.B., Renshaw, S.A., 2016. 761

Identification of benzopyrone as a common structural feature in compounds with 762

anti-inflammatory activity in a zebrafish phenotypic screen. Dis. Model. Mech. 9, 763

621–632. doi:10.1242/dmm.024935 764

Rylander, D., Recchia, A., Mela, F., Dekundy, A., Danysz, W., Cenci, M.A., 2009. 765

Pharmacological modulation of glutamate transmission in a rat model of L-DOPA-766

induced dyskinesia: effects on motor behavior and striatal nuclear signaling. J. 767

Pharmacol. Exp. Ther. 330, 227–235. doi:10.1124/jpet.108.150425 768

Samadi, P., Bédard, P.J., Rouillard, C., 2006. Opioids and motor complications in 769

Parkinson’s disease. Trends Pharmacol. Sci. 27, 512–517. 770

doi:10.1016/j.tips.2006.08.002 771

Schneider, C.A., Rasband, W.S., Eliceiri, K.W., 2012. NIH Image to ImageJ: 25 years 772

of image analysis. Nat. Methods 671–675. doi:10.1038/nmeth.2089 773

Schuster, S., Doudnikoff, E., Rylander, D., Berthet, A., Aubert, I., Ittrich, C., Bloch, B., 774

38

Cenci, M.A., Surmeier, D.J., Hengerer, B., Bezard, E., 2009. Antagonizing L-type 775

Ca2+ Channel Reduces Development of Abnormal Involuntary Movement in the 776

Rat Model of L-3,4-Dihydroxyphenylalanine-Induced Dyskinesia. Biol. Psychiatry 777

65, 518–526. doi:10.1016/j.biopsych.2008.09.008 778

Sun, Y., Dong, Z., Khodabakhsh, H., Chatterjee, S., Guo, S., 2012. Zebrafish chemical 779

screening reveals the impairment of dopaminergic neuronal survival by cardiac 780

glycosides. PLoS One 7, 1–11. doi:10.1371/journal.pone.0035645 781

Tay, T.L., Ronneberger, O., Ryu, S., Nitschke, R., Driever, W., 2011. Comprehensive 782

catecholaminergic projectome analysis reveals single-neuron integration of 783

zebrafish ascending and descending dopaminergic systems. Nat. Commun. 2, 171. 784

doi:10.1038/ncomms1171 785

Tzschentke, T.M., Christoph, T., Kögel, B., Schiene, K., Hennies, H., Englberger, W., 786

Haurand, M., Jahnel, U., Cremers, T.I.F.H., Friderichs, E., Vry, J. De, 2007. (-)-787

(1R,2R)-3-(3-Dimethylamino-1-ethyl-2-methyl-propyl)-phenol hydrochloride 788

(Tapentadol HCl): a Novel u-Opioid Receptor Agonist / Norepinephrine Reuptake 789

Inhibitor with Broad-Spectrum Analgesic Properties. J Pharmacol Exp Ther. 323, 790

265–276. doi:10.1124/jpet.107.126052.2002 791

Vaz, R.L., Outeiro, T.F., Ferreira, J.J., 2018. Zebrafish as an animal model for drug 792

discovery in Parkinson’s disease and other movement disorders: a systematic 793

review. Front. Neurol. 9, 1–23. doi:10.3389/FNEUR.2018.00347 794

Vijayakumar, D., Jankovic, J., 2016. Drug-Induced Dyskinesia, Part 1: Treatment of 795

Levodopa-Induced Dyskinesia. Drugs 76, 759–777. doi:10.1007/s40265-016-0566-796

3 797

Vijayanathan, Y., Lim, F.T., Lim, S.M., Long, C.M., Tan, M.P., Majeed, A.B.A., 798

Ramasamy, K., 2017. 6-OHDA-Lesioned Adult Zebrafish as a Useful Parkinson’s 799

Disease Model for Dopaminergic Neuroregeneration. Neurotox. Res. Epub ahead, 800

1–13. doi:10.1007/s12640-017-9778-x 801

Wang, M., Zhang, Z., Cheang, L.C., Lin, Z., Lee, S.M., 2011. Eriocaulon buergerianum 802

extract protects PC12 cells and neurons in zebrafish against 6-hydroxydopamine-803

induced damage. Chin. Med. 6, 1–10. doi:10.1186/1749-8546-6-16 804

39

Wang, Y., Liu, W., Yang, J., Wang, F., Sima, Y., Zhong, Z., Wang, H., Hu, L.-F., Liu, 805

C.-F., 2017. Parkinson’s disease-like motor and non-motor symptoms in rotenone-806

treated zebrafish. Neurotoxicology 58, 103–109. doi:10.1016/j.neuro.2016.11.006 807

Westerfield, M., 2000. The zebrafish book. A guide for the laboratory use of zebrafish 808

(Danio rerio)., 4th ed. Univ. of Oregon Press, Eugene. 809

Xi, Y., Noble, S., Ekker, M., 2011. Modeling neurodegeneration in zebrafish. Curr. 810

Neurol. Neurosci. Rep. 11, 274–282. doi:10.1007/s11910-011-0182-2 811

Zhang, Z., Cheang, L.C.V., Wang, M., Li, G., Chu, I.K., Lin, Z., Lee, S.M.Y., 2012. 812

Ethanolic extract of fructus alpinia oxyphylla protects against 6-hydroxydopamine-813

induced damage of PC12 cells in vitro and dopaminergic neurons in zebrafish. 814

Cell. Mol. Neurobiol. 32, 27–40. doi:10.1007/s10571-011-9731-0 815

Zhang, Z.J., Cheang, L.C.V., Wang, M.W., Lee, S.M., 2011. Quercetin exerts a 816

neuroprotective effect through inhibition of the iNOS/NO system and pro-817

inflammation gene expression in PC12 cells and in zebrafish. Int. J. Mol. Med. 27, 818

195–203. doi:10.3892/ijmm.2010.571 819

Zupanc, G.K.H., 2008. Adult neurogenesis and neuronal regeneration in the brain of 820

teleost fish. J. Physiol. Paris 102, 357–373. doi:10.1016/j.jphysparis.2008.10.007 821

822

823

40

Supplementary material 824

825

Supplementary figure S1 – Sensitivity of zebrafish larvae to different concentrations 826

of 6-OHDA. Motor performance depicted from (A) total distance moved and (B) burst 827

swimming in zebrafish larvae lesioned with different concentrations of 6-OHDA (n= 8 828

larvae per concentration) as compared to vehicle treated larvae (n= 8 larvae). Percentage 829

relative to the mean of vehicle treated larvae. Mean ± s.e.m. of one experiment is 830

presented. *p<0.05, **p<0.01, ***p<0.001, δp<0.05 500 µM vs 750 µM, Kruskal-Wallis 831

ANOVA with Dunn’s post-hoc test. 832

833

41

834

Supplementary figure S2 – Dose-response curves of motor performance of 6-OHDA-835

lesioned zebrafish larvae treated with L-dopa, rasagiline and isradipine. Motor 836

performance depicted from (A, C and E) total distance moved and (B, D and F) burst 837

swimming in 6-OHDA-lesioned zebrafish larvae treated with different doses of L-dopa 838

(n= 7-16 larvae), rasagiline (n= 7-8 larvae) and isradipine (n= 6-8 larvae) as compared to 839

untreated larvae (n= 13-14 larvae). Percentage relative to the mean of vehicle treated 840

42

larvae (healthy control, n= 8 larvae). Dashed lines signalize healthy and disease state, on 841

the top and at the bottom, respectively. Peak effective doses are highlighted by arrows. 842

843

844

Supplementary figure S3 – Motor performance of healthy zebrafish larvae treated 845

with L-dopa, rasagiline and isradipine. Motor performance depicted from (A) total 846

distance moved and (B) burst swimming in healthy zebrafish larvae treated with 1250 847

µM of L-dopa (n= 16 larvae), 0.8 µM of rasagiline (n= 16 larvae) and 0.5 µM of isradipine 848

(n= 15 larvae), as compared to untreated healthy zebrafish larvae (n= 16 larvae). 849

Percentage relative to the mean of healthy larvae. Mean ± s.e.m. of one experiment is 850

presented. Ns – not significant, Kruskal-Wallis ANOVA with Dunn’s post-hoc test. 851

43

852

853

Supplementary figure S4 - Rescue of motor impairments by stavudine, tapentadol 854

and nabumetone in 6-OHDA-lesioned zebrafish larvae, during the phenotypic 855

screening. Motor performance depicted from (A, C and E) total distance moved and (B, 856

D and F) burst swimming in 6-OHDA-lesioned zebrafish larvae treated with 25 µM of 857

44

stavudine (n= 19 larvae), tapentadol (n= 24 larvae) and nabumetone (n= 20 larvae), 858

picked from a library of FDA approved drugs, as compared to untreated larvae (n= 38-45 859

larvae). Percentage relative to the mean of vehicle treated larvae (healthy control, n= 23-860

24 larvae). Mean ± s.e.m. of three independent experiments is presented. ***p<0.001, 861

one-way ANOVA with Dunnett’s post-hoc test. 862

863