Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease

Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease

NSC 16249 No. of Pages 19 6 May 2015 Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but no...

4MB Sizes 1 Downloads 56 Views

NSC 16249

No. of Pages 19

6 May 2015 Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069 1

Neuroscience xxx (2015) xxx–xxx

4

INTERVENTION WITH EXERCISE RESTORES MOTOR DEFICITS BUT NOT NIGROSTRIATAL LOSS IN A PROGRESSIVE MPTP MOUSE MODEL OF PARKINSON’S DISEASE I

5 6

M. D. SCONCE, a M. J. CHURCHILL, a R. E. GREENE a AND C. K. MESHUL a,b,c*

7 8 9

a Research Services, VA Medical Center/Portland, Mail Code: RD-29, Research Services, 3710 SW Veterans Hospital Road, Portland, OR 97239, United States

2 3

10 11 12

b Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, United States

13 14 15

c Department of Pathology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, Oregon 97239, United States

16

Abstract—Many studies have investigated exercise therapy in Parkinson’s disease (PD) and have shown benefits in improving motor deficits. However, exercise does not slow down the progression of the disease or induce the revival of lost nigrostriatal neurons. To examine the dichotomy of behavioral improvement without the slowing or recovery of dopaminergic cell or terminal loss, we tested exercise therapy in an intervention paradigm where voluntary running wheels were installed half-way through our progressive PD mouse model. In our model, 1-methyl-4-phenyl-1,2,3,6-tetra hydropyridine (MPTP) is administered over 4 weeks with increased doses each week (8, 16, 24, 32-kg/mg). We found that after 4 weeks of MPTP treatment, mice that volunteered to exercise had behavioral recovery in several measures despite the loss of 73% and 53% tyrosine hydroxylase (TH) within the dorsolateral (DL) striatum and the substantia nigra (SN), respectively which was equivalent to the loss seen in the mice that did not exercise but were also administered MPTP for 4 weeks. Mice treated with 4 weeks of MPTP showed a 41% loss of vesicular monoamine

transporter II (VMAT2), a 71% increase in the ratio of glycosylated/non-glycosylated dopamine transporter (DAT), and significant increases in glutamate transporters including VGLUT1, GLT-1, and excitatory amino acid carrier 1. MPTP mice that exercised showed recovery of all these biomarkers back to the levels seen in the vehicle group and showed less inflammation compared to the mice treated with MPTP for 4 weeks. Even though we did not measure tissue dopamine (DA) concentration, our data suggest that exercise does not alleviate motor deficits by sparing nigrostriatal neurons, but perhaps by stabilizing the extraneuronal neurotransmitters, as evident by a recovery of DA and glutamate transporters. However, suppressing inflammation could be another mechanism of this locomotor recovery. Although exercise will not be a successful treatment alone, it could supplement other pharmaceutical approaches to PD therapy. Ó 2015 Published by Elsevier Ltd. on behalf of IBRO.

Key words: exercise, motor behavior, MPTP, Parkinson’s disease, glutamate transporters, tyrosine hydroxylase. 17

I

This work was supported by Merit Review #1BX 001643 to CKM from the United States (U.S.) Department of Veterans Affairs Biomedical Laboratory Research and Development. The contents do not represent the views of the U.S. Department of Veterans Affairs or the United States Government. *Correspondence to: C. K. Meshul, VA Medical Center/Portland, Mail Code: RD-29, Research Services, 3710 SW Veterans Hospital Road, Portland, OR 97239, United States. Tel: +1-503-220-8262x56788. E-mail address: [email protected] (C. K. Meshul). Abbreviations: BDNF, brain-derived neurotrophic factor; DA, dopamine; DAT, dopamine transporter; DL, dorsolateral; EAAC1, excitatory amino acid carrier 1; GFAP, glial fibrillary acidic protein; GLAST, glutamate aspartate transporter; GLT-1, glutamate transporter 1; IHC, immunohistochemistry; ir, immunoreactive; NFATc3, nuclear factor of activated T-cells cytoplasmic 3; PD, Parkinson’s disease; pTrkB, phosphorylated tyrosine kinase receptor B; MPTP, 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine; SN, substantia nigra; SNpc, substantia nigra pars compacta; TBST, tris-buffered saline tween 20; TH, tyrosine hydroxylase; TrkB, tyrosine kinase receptor B; VGLUT1, vesicular glutamate transporter 1; VGLUT2, vesicular glutamate transporter 2; VMAT2, vesicular mono amine transporter 2. http://dx.doi.org/10.1016/j.neuroscience.2015.04.069 0306-4522/Ó 2015 Published by Elsevier Ltd. on behalf of IBRO. 1

INTRODUCTION

18

In the clinic and in animal models, exercise has been consistently shown to alleviate some of the motor deficits associated with Parkinson’s disease (PD). PD is a common neurodegenerative disorder caused by the loss of dopaminergic neurons in the nigrostriatal pathway. This loss of dopamine (DA) also results in changes in striatal glutamate (Klockgether et al., 1991; Meshul et al., 1999; Robinson et al., 2003; Walker et al 2009) which creates an imbalance between DA and glutamate neurotransmitters. Whether this imbalance of DA and glutamate is a cause or a result of PD is unknown, and unfortunately, as the degeneration progresses over time so does the impairment of basal ganglion-stimulated motor behaviors (Meshul et al., 1999; Touchon et al., 2004; Holmer et al., 2005; Smith et al., 2011). As seen clinically and in animal models, exercise has shown to attenuate motor deficits in PD, but it does not seem to provoke recovery of the lost neurons nor prevent the progressive nature of the disease (Fisher et al., 2004; Fisher et al., 2008; Al-Jarrah et al., 2007; Petzinger et al., 2007; Pothakos et al., 2009; Petzinger et al., 2010; Vucˇkovic´ et al., 2010; Petzinger et al., 2013). This suggests that the behavioral recovery observed must be

19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

NSC 16249

No. of Pages 19

6 May 2015

2 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

due to exercise inducing other compensatory mechanisms that are separate from the mechanisms of dopaminergic survival and recovery. In this study, we looked at how intervening with voluntary exercise using running wheels would affect behavioral deficits as well as dopaminergic, glutamatergic and inflammatory biomarkers in our progressive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Of interest is a previous study reporting exercise-induced increases in both extracellular glutamate and DA levels within the striatum (Meeusen et al., 1997). However, following exercise training for several weeks, there was an overall decrease in the basal levels of both striatal DA and glutamate compared to the control group. MPTP has been extensively used in neurotoxin animal models of PD because it selectively lesions and causes neuronal death within the dopaminergic neurons in the basal ganglia (Meredith et al., 2008; Tieu, 2011; Blandini and Armentero, 2012). In contrast to other more acute/subacute MPTP/toxin models that lesion a large percent of the dopaminergic neurons in a short period of time, our progressive model better mimics the progressive temperament of PD by long-term gradual neurodegeneration of the nigrostriatal pathway where mice are injected with MPTP for 4 weeks (5 days/week) with increasing doses for the given week (8, 16, 24, and 32-mg/kg). In order to prevent inflammation due to possible stress of forced treadmill running (Howells et al., 2005), running wheels were installed into each singly housed mouse cage after the 2nd week of MPTP treatment (i.e. intervention) so that animals had the voluntary option to exercise. Although exercise has been shown to be protective against nigrostriatal degeneration in animal models (Lau et al., 2011; Gerecke et al., 2010), most patients upon diagnosis show a substantial amount of nigrostriatal terminal and cell loss (Bernheimer et al., 1973; Riederer and Wuketich, 1976; Kordower et al., 2013) which cedes rodent protection studies to be clinically irrelevant. Testing a therapy via intervention in our progressive mouse model is a more clinically appropriate approach because like the patients, the mice will already have dopaminergic cell and terminal loss; therefore, we can gauge if exercise therapy in our model can prevent or slow the progression of the nigrostriatal loss. Furthermore, due to the progressive nature of our animal model compared to the more acute/subacute models that have been used in previous exercise studies, we hypothesized that intervention with voluntary exercise would attenuate motor deficits and be disease modifying. To our knowledge, this is the first time that intervention with voluntary exercise therapy has been tested in a progressive mouse model of PD.

EXPERIMENTAL PROCEDURES

94 95

Animals

96

31 male C57BL/6J mice (Jackson Labs, Bar Harbor, ME, USA; 8-weeks old at arrival) were housed 3–4/cage and maintained on a 12-h light/dark cycle throughout (lights on 0600). They had ad libitum access to food and water. Mice were randomized into six groups: four mice

97 98 99 100

in the vehicle group (VEH), eight mice in the MPTP group (4WK_MPTP), six mice in the vehicle/exercise group (VEH + Ex), five mice in the MPTP/exercise (4WK_MPTP + Ex), four mice in the 2-week vehicle group (2WK_VEH), and four mice in the 2-week MPTP group (2WK_MPTP). For the exercise groups, mice were housed 1/cage and running wheels (Mini-Mitter Co., Inc, Bend, OR, USA) were installed. Mice were not forced to exercise but were able to voluntarily and at anytime. Each wheel had a digital attachment that counted the number of revolutions regardless of direction. The number of revolutions was recorded each week day and averaged over the number of hours between each recording. Handling and care of mice was consistent with federal guidelines of the Public Health Service Policy on the Humane Care and Use of Laboratory Animals and protocols were approved by the Portland VA IACUC.

101

MPTP administration

119

MPTP (Santa Cruz Biotechnology, Dallas, TX, USA)treated animals received intraperitoneal injections 5 days/week for 4 weeks in total. The dosing of MPTP (calculated as the base) increased each week with 8 mg/kg for week 1, 16 mg/kg for week 2, 24 mg/kg for week 3, and 32 mg/kg for week 4 (4WK_MPTP). Normal saline (SA) (0.1 ml/0.1 kg) was used as the vehicle (VEH) for MPTP. After the first two weeks of MPTP treatment, a group of mice treated with MPTP and another group of mice treated with vehicle were separated and housed individually with cages that had running wheels (4WK_MPTP + Ex or VEH + Ex). Two additional groups of animals were separated and euthanized after the first two weeks of MPTP or saline treatment for striatal and midbrain/nigral analysis (2WK_MPTP or 2WK_VEH).

120

Grip test analysis

136

Mice were tested for grip strength at the conclusion of either the 2 weeks MPTP/vehicle treatment or at the end of the MPTP/exercise treatment with the Grip Strength Meter (Columbus Instruments, Columbus, Ohio, USA). Using the Mesh Pull Bar attachment, mice were handled by the tail so that only the forepaws could touch the apparatus. When the forepaws were clearly latched to the Mesh Pull Bar, the mouse was slowly and steadily pulled away from the apparatus exactly parallel from the counter top until the mouse released its forepaws from the Mesh Pull Bar. Each animal’s grip strength was an average of five grip strength tests and animals were given a 5-min rest in between each trial. The protocol was repeated additionally, as explained above, for all the paws latching on the Mesh Pull Bar together. Grip strength was measured in Newtons (N). Mice in the 2WK_MPTP and 2WK_VEH groups were tested 10 days after the last 16 mg/kg dose of MPTP. All of the other groups of mice (VEH, 4WK_MPTP, VEH + Ex, and 4WK_MPTP + Ex) were tested 25 days after the last 32 mg/kg dose of MPTP.

137

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118

121 122 123 124 125 126 127 128 129 130 131 132 133 134 135

138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 158

Motor behavior analysis

159

171

Gait was assessed using a DigiGait apparatus (Mouse Specifics, Quincy, MA, USA). Ventral plane videography captured the gait of each mouse through a transparent, motor-driven treadmill belt (Kale et al., 2004; Amende et al., 2005). Digital images of the paws of each mouse were taken at 150 frames/s as mice ran at a speed of 24 cm/s. The area of each paw relative to the treadmill belt at each frame was used for spatial and temporal measurements. Mice in the 2WK_MPTP and 2WK_VEH groups were tested 9 days after the last 16 mg/kg dose of MPTP. All the other groups of mice (VEH, 4WK_MPTP, VEH + Ex, and 4WK_MPTP + Ex) were tested 24 days after the last 32 mg/kg dose of MPTP.

172

Harvesting & tissue fixation

173

194

All the animals were euthanized by cervical dislocation. The fresh brain was cut coronally in half at the level of the hypothalamus, where the rostral half containing the striatum was fixed with 2.5% glutaraldehdye/0.5% paraformaldehyde/0.1% picric acid in 0.1 M phosphate buffer (pH 7.3) using a microwave tissue processor (Pelco BioWave Ted Pella, Inc, Redding, CA, USA) as follows: the tissue was placed in a temperature controlled bath with fixative using a thermoelectric recirculating chiller (Pelco SteadyTemp Pro, Ted Pella, Inc) in the microwave for 90 min total [45 min., 150 watts(W) at 30 °C/30 min., 150 W at 25 °C/15 min., 650 W at 25 °C]. The tissue was then left in 2% paraformaldehyde/0.1 M phosphate buffer (pH 7.3) at 4 °C for 48-72 hrs, then rinsed and left in 0.1 M phosphate buffer at 4 °C until cut for future tyrosine hydroxylase (TH) immunohistochemistry (IHC) (see below). For the caudal portion of the brain, the left and right substantia nigra (SN) were micro-dissected, using a stereomicroscope and a 3-mm microdissection knife, and frozen at 80 °C for future protein/western blot analysis (see below).

195

Immunohistochemistry

196

A vibratome (Ted Pella Inc., Redding, CA, USA) was used to cut consecutive 60-lm thick sections of the striatum (starting at Bregma + 1.2 mm and ending at the level of the anterior commissure). Six sections were chosen that extended throughout the rostrocaudal portion of the striatum and these sections were matched anatomically in each animal to verify that the coronal sections used were similar in all groups of mice. The following incubations were carried out in the PELCO BioWaveÒ Pro microwave (Ted Pella Inc., Redding, CA, USA) with the temperature limited to 35 °C. Rinsing solutions were under normal pressure unless otherwise stated. Sections were incubated in 10 mM sodium citrate (pH 6) for 5 min at 550 W for antigen retrieval in a vacuum chamber that cycles the pressure down to 20 Hg and back to atmosphere repeatedly during this step (cycling vacuum), rinsed in 0.01 M phosphate-buffered saline (PBS) at 150 W for 1 min, rinsed with 0.3% hydrogen peroxide (150 W, 1 min), two PBS rinses for 1 min, and

160 161 162 163 164 165 166 167 168 169 170

174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193

197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214

3

then incubated in 0.5% Triton X-100 in PBS (550 W, 5 min, cycling vacuum). Sections were incubated with tyrosine hydroxylase (TH) antibody (mouse monoclonal, 1:2000 for the striatum; Immunostar, Hudson, WI, USA) or with ionizing calcium-binding adaptor molecule 1 (Iba-1) antibody (rabbit polyclonal, 1:200 for the striatum; Proteintech, Chicago, IL, USA) at 200 W for 36 min and 20 s under continuous vacuum (20 Hg, cycling the magnetron for 2 min on/3 min off/2 min on/5 min off repeating). Sections were incubated in blocking solution [10% goat serum/0.5% Triton-X 100/0.1 M phosphate buffer; pH 7.4] for two, 1 min washes at 150 W, and then exposed to biotinylated goat anti-mouse secondary antibody (1:400; Vector, Burlingame, CA, USA) for 16 min and 20 sec under continuous vacuum (10 s at 150 W, 4 min at 200 W, 3 min at 0 W, 4 min at 200 W, 5 min at 0 W, and 10 s at 150 W), rinsed with PBS, then washed with imidazole working buffer [5% Imidazole buffer(0.2 M), pH 9.0/16% sodium acetate (0.1 M), pH7.2] (1 min at 150 W), and finally incubated with avidin-biotin complex solution (ABC) (diluted according to manufacturer instruction; Vector) for 16 min and 10 sec under continuous vacuum (4 min at 200 W, 3 min at 0 W, 4 min at 200 W, 5 min at 0 W, and 10 sec at 200 W). Tissue was then rinsed with imidazole working buffer (1 min at 150 W), incubated with diaminobenzidine (DAB) (0.1% (Sigma Chemical Co., St Louis, MO; Cat #: D5637) + 1.5% hydrogen peroxide in 0.1 M phosphate buffer) for 10 min 20 sec under continuous vacuum (10 s at 200 W, 10 min at 200 W, 10 s at 0 W), rinsed with imidazole working buffer, and finally in PBS. Tissue was mounted on gel-coated slides, dehydrated at room temperature overnight and cover-slipped using Pro-TexxÒ medium (Lerner, Pittsburgh, PA, USA). Tissue from all treatment groups was processed on the same day, and all reacted with DAB for the same length of time. Optical density of the dorsolateral (DL) region of the striatum for each section for every animal was analyzed using light microscopy (1.25x magnification, images analyzed using ImagePro Plus 6.3, Media Cybernetics, Rockville, MD, USA). Treatment groups were analyzed in a blinded manner.

215

Western blots

258

Dissected SN tissue was thawed on ice. Protein was extracted from the tissue by sonication in lysis buffer [5% 1 M Tris, 2% 0.5 M EDTA, 1% Triton-X 100, 0.5% Protease Inhibitor Cocktail III (EMD Millipore’s Calbiochem, Darmstadt, Germany)]. Protein concentrations of the tissue from each individual animal were measured using the BCA Protein Assay Kit (Thermo Scientific, Rockford, IL, USA). 10 lg of protein from each sample was mixed with XT Sample Buffer and XT Reducing Agent (1:10; Bio-Rad, Hercules, CA, USA) and underwent electrophoresis on a 4-12% Bis-Tris XT Precast Gel (Bio-Rad, Hercules, CA, USA). Separated proteins were transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, MA, USA). The membranes were blocked in either a 5% non-fat dry milk in Tris–buffered saline with Tween-20 (TBST) for

259

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257

260 261 262 263 264 265 266 267 268 269 270 271 272 273 274

NSC 16249

No. of Pages 19

6 May 2015

4

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

319

60 min. Membranes were then washed three times for 5 min each in TBST, and probed with the following primary antibodies: tyrosine hydroxylase (TH at 62 kDa; Immunostar, Inc., Hudson, WI, 1:40,000, mouse monoclonal), phospho-tyrosine repeat kinase B (pTrkB at 145 kDa; Abcam (Cambridge, MA) 1:1000; rabbit polyclonal), vesicular monoamine transporter 2 (VMAT2 at 57 kDa; Synaptic Systems (Goettingen Germany); 1:200 rabbit polyclonal), dopamine transporter (DAT at 80 and 50 kDa; Proteintech (Chicago, IL); 1:1000; rabbit polyclonal), vesicular glutamate transporter 1 and 2 (VGLUT1 at 62 kDa, 1:20,000, and VGLUT2 at 52 kDa, Synaptic Systems; 1:2000 rabbit polyclonal), glutamate transporter 1 (GLT-1 or known as EAAT2 at 70 kDa, Santa Cruz Biotech Inc (Dallas, TX); 1:1000, rabbit polyclonal), excitatory amino acid carrier 1 (EAAC1 or known as EAAT3 at 57 kDa, Santa Cruz Biotech Inc; 1:1000; rabbit polyclonal), glutamate aspartate transporter (GLAST or known as EAAT1 at 64 kDa, Santa Cruz Biotech Inc; 1:1000; rabbit polyclonal) glial fibrillary acidic protein (GFAP at 55 kDa, Sigma Aldrich; 1:6000; mouse monoclonal), nuclear factor of activated T-cells cytoplasmic 3 (NFATc3 at 190 and 130 kDa, Santa Cruz Biotech Inc; 1:1000; rabbit polyclonal), and b-actin (at 47 kDa, Sigma Aldrich; 1:65,000; mouse monoclonal) . After three 5-min washes in TBST, membranes were probed with secondary antibodies for 1 h (ap-bovine anti-goat IgG H + L, Jackson ImmunoResearch Laboratories Inc (West Grove, PA); ap-goat anti-mouse IgG H + L, Bio-Rad, ap-goat anti-rabbit IgG H + L, Bio-Rad) then washed again in TBST for 3  5 min. Enhanced chemifluoresence (ECF) substrate (GE Healthcare, Piscataway, NJ, USA) was added to the membrane prior to visualization. Visualization and quantification of the antigen-antibody binding density were performed using the Typhoon HLA7000 imaging system (GE Healthcare, Piscataway, NJ, USA), and ImagePro Plus 6.3 software (Media Cybernetics, Rockville, MD, USA), respectively. Protein densities were analyzed relative to individual b-actin densities and the normalized optical density was determined for each animal. Each sample was at least duplicated on separate membranes and averaged for each animal. All the animals were averaged according to their group.

320

Statistical analysis

321

To analyze the amount of exercise at the end of each week, repeated measures analysis of variance (ANOVA) was used to compare the amount of exercise pursued between the VEH + Ex and 4WK_MPTP + Ex groups. A two-way ANOVA was then used for each group to compare the averages at the end of each week. Significant interactions were subject to post hoc Tukey– Kramer HSD tests for multiple comparisons. For behavioral, IHC, and western blot analysis, two-way ANOVA’s were used to compare the MPTP groups to each other as well as their respective vehicle groups. All significant interactions were subjected to post hoc Tukey–Kramer HSD tests for multiple comparisons. If there were no differences between the vehicle groups,

275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318

322 323 324 325 326 327 328 329 330 331 332 333 334

then those groups were combined before calculating the percent of vehicle against the MPTP-treated groups. In the analyses for which significant interactions were not observed, the a priori hypothesis that the population (exercise versus no exercise) had a greater effect in the MPTP-compared to the vehicle-treated animals was addressed in planned comparisons between the exercise (4WK_MPTP + Ex/VEH + Ex) and no exercise pairs (4WK_MPTP/vehicle) by the Student’s paired t-tests. Pearson correlations with significant r-values were used to evaluate the correlations between motor behaviors and biochemical markers (Fig. 7). All statistical analyses were considered significant at p < 0.05.

335

RESULTS

349

336 337 338 339 340 341 342 343 344 345 346 347 348

Behavioral Analysis

350

Despite 4 weeks of MPTP administration, mice that exercised showed behavioral recovery. Voluntary wheel running exercise was available to the mice starting during the 3rd week of MPTP administration (i.e. intervention). There was a main effect of MPTP and the distance the mice voluntarily exercised (Fig.1; F(4,5) = 6.3634; p = 0.0337). Compared to the vehicle group, mice administered MPTP ran 90% less during the 3rd week of the treatment (p = 0.0062) and 74% less during the 4th week of treatment (p = 0.0256). The 4WK_MPTP + Ex group then recovered to similar distances of voluntary exercise of the vehicle group after the last dosing week of MPTP. Furthermore, comparing among the MPTP group to itself after each week pre/post toxin administration, there was a

351

Fig. 1. Voluntary exercise running rate during and following the final two weeks of the MPTP dosing paradigm. While mice were administered MPTP (n = 5), their rate of wheel running decreased 90% and 74% compared to the vehicle mice (n = 6) (3rd week and 4th week, respectively). After MPTP administration (off_1WK-3WK), voluntary wheel running recovered to the rates of the vehicle mice. Values are mean ± S.E.M. ⁄p = 0.0062 vs VEH + Ex (3rd week), ⁄⁄p = 0.0256 vs VEH + Ex (4th week), dp = 0.0169 vs 4WK_MPTP + Ex (3rd week), and ddp = 0.0190 vs 4WK_MPTP + Ex (4th week).

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

352 353 354 355 356 357 358 359 360 361 362 363 364 365

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426

significant increase in the amount of voluntary exercise observed between the first week off treatment and the 3rd week of MPTP treatment (p = 0.0169) as well as with the 4th week of MPTP treatment (p = 0.0190). Because there was recovery in the amount of exercise for mice treated with MPTP, other physical, behavioral, and biochemical measures were analyzed including the weight gained from the beginning to end of the experiment. All mice were weighed before and after any treatment. Analysis showed that there was a main effect of MPTP on the difference in weight gain/loss from the beginning to the end of the study (Fig. 2A; F(5,20) = 3.213; p = 0.0273). Mice treated with MPTP for 2 weeks had an averaged loss of 0.15 g of weight, which was statistically significant compared to the vehicle group’s averaged weight gain of 1.11 g (p = 0.0054). The mice treated with MPTP for 4 weeks gained 0.43 g on average, which was trending from the weight difference in the vehicle group (p = 0.054), though not statistically different. Mice that were treated with MPTP and given the option to exercise on free-running wheels gained 1.5 g of weight after the study which was similar to the vehicle group as well as statically different from the 2WK_MPTP (p = 0.0015) and the 4WK_MPTP (p = 0.0119) groups. Despite 4 weeks of MPTP treatment, animals allowed to exercise had similar weight gain to the vehicle group. Further analyses of grip test behavior were then elucidated. The grip test investigates the force of grip strength. The grip strength in the forepaws and all paws together of all mice was tested before harvesting the animals. Statistically there were no differences in the grip strength of all four paws together within the groups. However, there was a main effect of MPTP on the percent of forepaw grip strength (Fig. 2B; F(5,19) = 7.1535; p = 0.0006) with respect to the strength in all the paws together. The animals treated with MPTP for 4 weeks showed an increase of 31% in the grip strength of the forepaws compared to the vehicle group (p < 0.0001). The 4WK_MPTP group increased 27% and 19% compared to the 2WK_MPTP group (p = 0.0031) and the 4WK_MPTP + Ex group (p = 0.0361), respectively. The 2WK_MPTP and 4WK_MPTP + Ex groups were not statistically different compared to the vehicle group. Animals that were administered MPTP for 4 weeks with the option to exercise showed a grip strength similar to that of the vehicle group. In additional to analyzing motor deficits by physical weight changes and grip strength, different parameters of gait dynamics were also measured. Looking at the paws while the mice were running revealed that MPTP had a main effect of the paw area at peak stance (Fig. 2C; F(5,16) = 8.4496; p = 0.0005) as well as the maximal rate of change of paw area in contact with the treadmill belt during the breaking phase (MAX dA/dT) (Fig. 2D; F(5,16) = 7.9748; p = 0.0006). Mice treated with MPTP for 4 weeks had a significant 45% increase in the overall paw area at peak stance compared to the vehicle group (p = 0.0045), which was also significantly

5

increased compared to the 2WK_MPTP (p = 0.0125) and 4WK_MPTP + Ex (p = 0.0014) groups. The MAX dA/dT in the 4WK_MPTP mice also significantly increased by 72% compared to the vehicle group (p < 0.0001), which was similarly increased compared to the 2WK_MPTP (p = 0.0019) and the 4WK_MPTP + Ex (p = 0.0002) groups. An increase in the paw area at peak stance and in the rate that the animal decelerates may suggest that the behavior deficits in the animals administered MPTP include a decrease in the typical agile running dynamics. Furthermore, MPTP had a main effect on the stance width (Fig. 2E; F(4,15) = 8.6484; p = 0.0056) of the mice as well as the percent of shared stance (Fig. 2F; F(5,15) = 9.0554; p = 0.0004). In animals treated with MPTP for 4 weeks, stance width increased 27% compared to the vehicle (p = 0.003), which was also similarly increased compared to the 2WK_MPTP (p = 0.0084) and 4WK_MPTP + Ex (p = 0.0291) groups. The percent of stance that was shared or on the treadmill belt at the same time while the mice were running had significantly decreased by 50% in the 4WK_MPTP animals compared to the vehicle group (p = 0.0220). The 4WK_MPTP + Ex mice had a recovery of this deficit to vehicle levels and was significantly increased compared to the 4WK_MPTP mice (p = 0.0062). An increase in stance width and a decrease in the percent of shared stance while running suggest that the motor deficits in our progressive model include postural changes and instability which would correlate with the PD patients who experience changes in their stance and posture. Taken together, among all the measures of behavior, animals who were administered MPTP and volunteered to exercise showed a recovery in all the motor deficits to vehicle group levels.

427

TH-immunoreactivity (TH-ir) Analysis

463

Exercise did not recover or slow down the progressive loss of TH in the DL striatum, nor the SN. The levels of the dopaminergic biomarker TH were assessed and compared within the DL striatum by IHC and within the SN by western blot. There was a main effect of MPTP on the levels of TH-ir within the DL striatum (Fig. 3A, C; F(2,12) = 8.924; p = 0.0042) as well as within the SN (Fig. 3B, D; F(5,19) = 41.7; p < 0.0001). Within the DL striatum, mice administered MPTP for 2 weeks showed a significant decrease of 42% compared to the respective vehicle group (p = 0.0019). Progressively, the animals administered MPTP for 4 weeks had a significant decrease of 70% compared to the respective vehicle group (p < 0.0001) with the DL striatum. Similarly, the mice that were administered MPTP for 4 weeks but also exercised showed a significant 73% loss of TH compared to the respective vehicle group (p < 0.0001) within the DL striatum. However, despite the continued loss of TH expression within the DL striatum similar to that observed in the MPTP-treated animals, the animals that were administered MPTP and

464

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462

465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484

NSC 16249

No. of Pages 19

6 May 2015

6

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

Fig. 2. Weight changes and behavioral analysis by grip test and gait dynamics. (A) The 2WK_MPTP group (n = 4) lost 0.15 g of body weight while the 4WK_MPTP group (n = 8) gained 0.43 g of body weight, which is a 61% decrease compared to the 1.11 g gained in the vehicle group (n = 14). The 4WK_MPTP + Ex group (n = 5) showed a weight gain of 1.5 g which was significantly different compared to the other MPTP groups but similar to the vehicle group. (B) Grip strength in the forepaw increased by 31% in the 4WK_MPTP group compared to the vehicle group. The 2WK_MPTP, 4WK_MPTP + Ex, and vehicle groups had similar averaged forepaw grip strength. (C) 4WK_MPTP increased their paw area at peak stance by 45%, while the 2WK_MPTP and 4WK_MPTP + Ex groups had similar paw areas compared to the vehicle group. (D) Assessing how quickly the mice decelerate, 4WK_MPTP mice showed a 72% increase in MAX dA/dT compared to the vehicle group, while the 2WK_MPTP and 4WK_MPTP + Ex mice did not show a change with respect to the vehicle group. (E) The stance width of the mice increased 27% in 4WK_MPTP animals compared to the vehicle and there were no changes in the 2WK_MPTP and 4WK_MPTP + Ex groups. (F) The percent of the stride duration that the paw was in contact with the belt decreased 50% in the 4WK_MPTP mice compared to the vehicle group. The 2WK_MPTP and 4WK_MPTP + Ex animals maintained a similar % stride in stance compared to the vehicle group. Values are mean ± S.E.M.

485 486 487 488

exercised showed behavioral recovery. Additional sections of the DL striatum were analyzed for Iba-1, to determine the relative expression of the microglia marker by IHC. There was a main effect of MPTP on

the relative optical density of Iba-1 levels within the DL striatum (F(3,13) = 5.1509; p = 0.0145). The animals treated with MPTP had a statistically significant 65 ± 14.8% (values are means of %/respective

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

489 490 491 492

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

A

DL striatum

7

B TH (62kDa) β-actin (43 kDa)

C

D

Fig. 3. TH-ir IHC of the DL striatum and western blot analysis of the SN. Changes in the relative optical density within the (A) DL striatum by IHC and (B) SN by western blot. (C) Compared to the respective vehicle group (n = 3-4), the 2WK_MPTP group (n = 4) showed a 42% loss in TH of the DL striatum and the 4WK_MPTP group (n = 8) showed a 70% loss in TH. Similarly, the 4WK_MPTP + Ex group (n = 5) had a 73% loss. (D) By western blot analysis of the SN, only a 9% loss of TH was observed in the 2WK_MPTP group compared to the respective vehicle group. The 4WK_MPTP and 4WK_MPTP + Ex displayed similar losses of 67% and 53%, respectively, compared to their respective vehicle group. Values are mean ± S.E.M.

493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520

vehicle ± S.E.M) increase in Iba-1 expression with respect to its vehicle group (p = 0.0287). The animals treated with MPTP and exercised had a slight but nonsignificant 21% ± 3.7 increase in Iba-1 levels compared to its respective vehicle group and was trending toward a 44% decrease compared to the MPTP animals (p = 0.0925). Within the SN, mice that were administered MPTP for 4 weeks, with or without exercise, showed a significant decrease of 52% and 63%, respectively, in TH compared to the vehicle group (p < 0.0001). The 2WK_MPTP group had similar levels of TH within the SN compared to the vehicle group. Additionally, the 2WK_MPTP group was significantly different verses the 4WK_MPTP and 4WK_MPTP + Ex groups (p < 0.0001). Because the levels of TH expression in the DL striatum and SN did not recovery via voluntary exercise, this suggests that exercise did not induce restoration or prevent loss of the nigro-striatal neurons. In contrast, the effects of exercise on levels of brain-derived neurotrophic factor (BDNF) have been extensively studied within the hippocampus (Cotman and Berchtold, 2002). BDNF binds to tyrosine repeat kinase (TrkB) causing the dimerization and autophosphorylation (pTrkB) of the receptor which leads to the signaling cascades that promotes cell survival and growth (Guillin et al., 2001; Carvalho et al., 2008; Cobb, 1999; Grewal et al., 1999; Chao, 2003; Minichiello., 2009; Skaper., 2012). To investigate if exercise had an effect

on the levels of TrkB activation, pTrkB expression was analyzed by western blot. There was a main effect of MPTP administration on the expression of pTrkB (F(5,18) = 3.4243; p = 0.0239). The 2WK_MPTP mice maintained similar levels of pTrkB compared to the respective vehicle group and the 4WK_MPTP mice had a 27% ± 4.5 (values are means of %/respective vehicle ± S.E.M) decrease of pTrkB compared to the respective vehicle group (p = 0.0296). Additionally, the 4WK_MPTP + Ex group had a similar decrease of 32% ± 11.5 compared to the respective vehicle group (p = 0.0112). Our data suggest that at least within the SN, exercise does not rescue pTrkB levels.

521

Western blot analysis

534

Despite dopaminergic cell/terminal loss, exercise maintained other regulatory biomarkers of dopamine in the SN. VMAT2 is a vesicular transmembrane protein responsible for uptake of DA into the synaptic vesicles. Within the SNpc cell bodies, there is also dendritic release of DA, meaning that VMAT2 can be detected within the SN (Geffen et al., 1976; Cheramy et al., 1981; Greenfield, 1985). MPTP had a main effect on the expression levels of VMAT2 within the SN (Fig. 4A, C; F(5,16) = 3.8144; p = 0.0182). There was a 41% loss of VMAT2 in the 4WK_MPTP mice compared to the

535

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

522 523 524 525 526 527 528 529 530 531 532 533

536 537 538 539 540 541 542 543 544 545

NSC 16249

No. of Pages 19

6 May 2015

8

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

A

VMAT2 (57kDa)

B

β-actin (43 kDa)

C

DAT<

(80kDa) (50kDa) β-actin (43 kDa)

D

Fig. 4. SN analysis of VMAT2 and DAT by western blot. (A) Resulting western blot of the SN for VMAT2 and (B) DAT. (C) The 2WK_MPTP mice (n = 3) maintained VMAT2 protein levels similar to the vehicle group (n = 11) while the 4WK_MPTP mice (n = 8) had a 41% loss compared to the vehicle group. Similar to the vehicle as well as the 2WK_MPTP group, the 4WK_MPTP + Ex mice (n = 4) maintained VMAT2 levels which were increased by 44% compared to animals administered MPTP for 4 weeks. (D) Taking the ratio of glycosylated DAT to non-glycolsylated DAT showed that the 2WK_MPTP mice had a significant 43% decrease compared to the respective vehicle group (n = 3–4) while the 4WK_MPTP mice had a significant 71% increase in the ratio compared to the respective vehicle group. The 4WK_MPTP + Ex group had a similar ratio of DAT compared to the respective vehicle group. Values are mean ± S.E.M.

546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573

respective vehicle group (p = 0.0203). The 2WK_MPTP and 4WK_MPTP + Ex groups had similar levels of VMAT2 compared to the respective vehicle groups and were significantly increased compared to the 4WK_MPTP group by 50% (p = 0.0014) and 44% (p = 0.0008), respectively. Mice administered MPTP for 4 weeks and were allowed to exercise maintained VMAT2 expression levels where the mice administered MPTP for 4 weeks without exercise had decreased expression levels. Further analysis of potential mechanisms of DA homeostasis was determined by investigating levels of DAT expression. The amount of post-translational protein modification of DAT by glycosylation can play a role in the activity of the transporter’s function of clearing extraneuronal DA as well as the degradation of the protein (Li et al., 2004). The relative levels of glycosylated and non-glycosylated DAT were analyzed and expressed as a ratio. There was a main effect of MPTP on the ratio of glycosylated to non-glycosylated DAT within the SN (Fig. 4B, D; F(5,20 = 3.0354; p = 0.0337). The 2WK_MPTP mice showed a significant 43% decrease in the ratio compared to the respective vehicle group (p = 0.0076) while the 4WK_MPTP showed a trending 71% increase in the ratio compared to the respective vehicle (p = 0.0730). There was a significant 114% difference between the 2- and 4-week-treated MPTP groups (p = 0.0026). The ratio of glycosylated to

non-glycosylated DAT in the 4WK_MPTP + Ex mice was similar to the respective vehicle group, and 84% less compared to the 4WK_MPTP group (p = 0.0257). These data suggest that despite 4 weeks of MPTP administration, animals who exercised maintained comparable levels of VMAT2 and the ratio of glycosylated to un-glycosylated DAT as the vehicle group in the remaining dopaminergic neurons within the SN. This could help explain behavioral recovery without dopaminergic cell/terminal recovery.

574

Exercise may help achieve glutamate homeostasis through glutamate transporters. Aside from VMAT2 and DAT, exercise could also affect the glutamatergic system. A debated hypothesis in PD is that over stimulation of the STN results in excess glutamate levels at the SNpc dendrites and soma. Excess glutamate could then induce toxicity and perpetuate additional DA cell loss. Because the previous results showed behavioral recovery despite continued dopaminergic cell/terminal loss, it is possible that exercise may be evoking regulation of glutamatergic mechanisms instead of contributing to the survival of dopaminergic cells/terminals. VGLUT1 is a vesicular transmembrane protein responsible for uptake of glutamate into the synaptic vesicles for transmission. The motor cortex projects terminals onto the SNpc and SNpr (pars reticulata) and utilizes VGLUT1 as the

584

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

575 576 577 578 579 580 581 582 583

585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 601 602 603 604 605

protein associated with vesicular glutamate uptake. The STN also has a glutamatergic projection to the SNpc/SNpr but uses the vesicular glutamate transporter-2 (VGLUT2) instead of the analogous VGLUT1 (Kaneko et al., 2002). Despite lack of any

A

VGLUT1 (61.6 kDa)

differences between the vehicle and MPTP groups in terms of VGLUT2 expression levels within the SN (data not shown), there was a main effect of VGLUT1 expression following MPTP administration (Fig. 5A, C; F(5,18) = 4.4476; p = 0.0082). The 2WK_MPTP mice

B

β-actin (43 kDa)

C

E

GLT-1 (70 kDa) β-actin (43 kDa)

D

EAAC1 (57 kDa)

F

GLAST (64 kDa) β-actin (43 kDa)

β-actin (43 kDa)

G

9

H

Fig. 5. SN analysis of glutamate biomarkers by western blot. (A) Resulting western blot of the SN for VGLUT1 and (B) GLT-1. (C) Changes in VGLUT1 levels were seen in the 4WK_MPTP (n = 5) group with a significant 120% increase compared to the vehicle group (n = 12). The 2WK_MPTP (n = 3) group was similar to the vehicle group with only a 22% non-significant increase. The 4WK_MPTP + Ex (n = 4) was statistically between the other two MPTP groups with a 54% non-significant increase compared to the vehicle group. (D) GLT-1 protein levels were both significantly increased in the 2WK_MPTP (n = 4) and 4WK_MPTP (n = 7) groups by 60% and 89%, respectively, compared to the vehicle group (n = 10). The 4WK_MPTP + Ex (n = 4) group maintained similar levels of GLT-1 compared to the respective vehicle group. (E) Resulting western blot of the SN pc for EAAC1 and (F) GLAST. (G) Expression of EAAC1 maintained similar levels compared to the respective vehicle group for the 2WK_MPTP (n = 3) and 4WK_MPTP + Ex (n = 3) groups. The 4WK_MPTP (n = 6) mice showed a 101% increase in EAAC1 levels, which were significantly different compared to the respective vehicle groups. (H) Levels of GLAST in the 2WK_MPTP (n = 3) and 4WK_MPTP (n = 7) groups were not different compared to the vehicle group (n = 11). The 4WK_MPTP + Ex mice had a 31% decrease compared to the vehicle group. Values are mean ± S.E.M. Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

606 607 608 609 610

NSC 16249

No. of Pages 19

6 May 2015

10 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

showed a 22% increase in VGLUT1 expression which was not statistically different compared to the vehicle group. The 4WK_MPTP mice had a 120% increase in VGLUT1 levels compared to the vehicle group (p = 0.0010), which was also increased compared to the 2WK_MPTP (p = 0.0133) and 4WK_MPTP + Ex (p = 0.0469) groups. The 4WK_MPTP + Ex group also had elevated VGLUT1 expression with a 49% increase compared to the vehicle group, which was not statistically significant (p = 0.0993). A decrease in VGLUT1 expression in the 4WK_MPTP + Ex mice relative to the 4WK_MPTP mice suggests that exercise helps alleviate increased glutamate input from the cortex. Other plasma membrane transporters of glutamate, such as GLT-1, EAAC1, and GLAST, could be regulated if there was increased activity of the cortico-nigral pathway. These different subtypes of glutamate transporters all function to clear extracellular glutamate. The GLT-1 transporter is located on microglia and astrocytes (Rothstein et al., 1994; Shashidharan et al., 1997; Maragakis and Rothstein, 2004). There was a main effect of MPTP on the expression of GLT-1 (Fig. 5B, D; F(5,19) = 3.3005; p = 0.0259). The 2WK_MPTP mice had a 60% increase in GLT-1 levels compared to the vehicle group (p = 0.0150), while the 4WK_MPTP mice had an 89% increase compared to the vehicle group (p = 0.0155). The 4WK_MPTP + Ex mice had similar

A

+phos~ (190 kDa) NFATc3<

-phos~ (130 kDa)

levels of GLT-1 compared to the vehicle group and had decreased levels compared to the 4WK_MPTP (p = 0.0419). Additionally, EAAC1 regulates neurotransmitter homeostasis by clearing extraneuronal glutamate and is found on the SNpc cell bodies and dendrites (Shashidharan et al., 1997; Plaitakis., 2000). After administration of MPTP, there was a main effect on levels of EAAC1 (Fig. 5E, G; F(5,18) = 4.9386; p = 0.0051). The 2WK_MPTP and 4WK_MPTP + Ex group preserved similar levels of EAAC1 compared to their respective vehicle groups. The 4WK_MPTP group had increased EAAC1 levels to 101% compared to the respective vehicle group (p = 0.0021) and the 2WK_MPTP (p = 0.0063) and 4WK_MPTP + Ex (p = 0.0049) groups. Increased expression of GLT-1 and EAAC1, along with VGLUT1, would be consistent with the suggestion that there is an increase in extracellular glutamate levels within the SN of the 4WK_MPTP mice, as we have previously reported in another MPTP model (Meredith et al., 2009). Additionally, the expression of GLAST was determined because it is also involved in regulating glutamate transport and can be found in both astroglia and neurons (Rothstein et al., 1994; Shashidharan et al., 1997; Plaitakis, 2000). There was a main effect of exercise on the expression of GLAST (Fig. 5F, H; F(5,9) = 6.1421; p = 0.0208). The 2WK_MPTP and 4WK_MPTP groups had similar and non-significant changes in the expression

B

β-actin (43 kDa)

β-actin (43 kDa)

C

GFAP (55 kDa)

D

Fig. 6. SN analysis of GFAP and NFATc3 by western blot. (A) Resulting western blot of the SN for NFATc3 and (B) GFAP. (C) Taking the ratio of phosphorylated to non-phosphorylated NFATc3 showed that the 4WK_MPTP + Ex mice had a significant increase of 25% compared to the vehicle group while the 2WK_MPTP and 4WK_MPTP were similar to the vehicle group. The 4WK_MPTP + Ex was also significantly increased by 42% compared to the 4WK_MPTP mice. (D) There were no differences between the 2WK_MPTP (n = 4) and the respective vehicle group (n = 3-4) in GFAP expression. The 4WK_MPTP mice (n = 8) showed a significant 116% increase compared to the respective vehicle group, while the 4WK_MPTP + Ex group (n = 4) had a 41% increase in GFAP protein expression compared to the respective vehicle group. Additionally, the 4WK_MPTP group was significantly increased by 108% and 75% compared to the 2WK_MPTP and 4WK_MPTP + Ex groups, respectively. Values are mean ± S.E.M. Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 665 666 667 668 669 670 671 672 673

674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724

levels of GLAST comparable to the vehicle group. The 4WK_MPTP + Ex group had decreased levels of GLAST by 31% compared to the vehicle group (p = 0.0265), which was also significantly less than the 2WK_MPTP (p = 0.0156) and 4WK_MPTP (p = 0.0030) groups. A decrease in GLAST expression in the 4WK_MPTP + Ex mice could imply that glutamate concentrations within the SN decreased compared to the 4WK_MPTP mice due to exercise. Animals administered MPTP and allowed to exercise had lower levels of inflammatory markers within the SN. Mechanistically, excess glutamate levels would increase inflammation due to the influx of Ca2+ upon binding to the glutamate receptors, which would lead to an increase in reactive oxygen species (Choi, 1988; Coyle and Puttfarcken, 1993; Blandini et al., 1996; Plaitakis and Shashidharan, 2000; Shashidharan et al., 2000). NFATc3 (component 3 of the nuclear factor of activated T-cells) is regulated in part by phosphorylation where it can only transverse the nucleus to turn on genes that signal apoptosis when it is non-phosphorylated (Crabtree and Olson, 2002; Macian, 2005). There was a main effect of exercise on the ratio of phosphorylated to non-phosphorylated NFATc3 (Fig. 6B, D; F(5,17) = 3.1389; p = 0.0346). Both the 2WK_MPTP and 4WK_MPTP mice maintained similar ratios of phosphorylated to non-phosphorylated NFATc3 compared to each other and their respective vehicle group. The 4WK_MPTP + Ex group had a significantly elevated ratio of 25% compared to the respective vehicle group (p = 0.0101) and a significant increase of 42% compared to the 4WK_MPTP group (p = 0.0130). An increased ratio of phosphorylated NFATc3 would mean that there is an increase in the percent of the NFATc3 protein population that cannot enter into the nucleus to regulate genes that would signal apoptosis. This piece of evidence further demonstrates that the behavioral recovery without dopaminergic cell/terminal recovery could be due in part by exercise normalizing DA release (as evidenced by DAT levels), regulating appropriate glutamate levels (as evidenced by VGLUT1, GLT-1, GLAST and EAAC1 levels) and thereby suppressing inflammation. The expression of GFAP levels was additionally assessed to indirectly gauge the relative amounts of inflammation. There was a main effect of MPTP on the levels of GFAP (Fig. 6A, C; F(5,19) = 15.4820; p < 0.0001). The 2WK_MPTP mice showed little astrocytic reaction and had similar GFAP levels compared to the vehicle group. The 4WK_MPTP mice had increased GFAP levels of 116%, which were significantly different compared to the vehicle group (p = 0.0043) as well as the 2WK_MPTP group (p = 0.0082). The 4WK_MPTP + Ex mice had GFAP expression levels that ranged between the other two MPTP groups, with an increase of 41% compared to the vehicle group. Levels of GFAP in the 4WK_MPTP + Ex mice were decreased by 75% compared to the 4WK_MPTP mice (p = 0.0373). These results with both NFATc3 and GFAP suggest that exercise may have helped alleviate the degree of inflammation within the SN; therefore, perhaps with less inflammation,

11

the remaining TH-ir cells within the SNpc can compensate by normalizing DA transmission to restore behavioral deficits without the need for either neurogenesis or axonal sprouting.

725

Correlation analysis

729

Dopaminergic, glutamatergic, and inflammatory biomarkers correlated with behavioral measures. By Pearson correlations, additional analysis showed a moderate negative Pearson correlation between VMAT2 and the paw area at peak stance (Fig. 7A; r = 0.5476; p = 0.0056) as well as the MAX dA/dT (Fig. 7B; r = 0.5787; p = 0.0031). There was a moderate positive correlation between VLGUT1 and the forepaw grip strength (Fig. 7C; r = 0.6433; p = 0.0007) as well as the MAX dA/dT (Fig. 7D; r = 0.5313; p = 0.0075) and a moderate positive correlation was observed between GLAST and the paw area at peak stance (Fig. 7E; r = 0.5245; p = 0.0085) as well as the MAX dA/dT (Fig. 7F; r = 0.5369; p = 0.0068). Furthermore, there was a moderate negative Pearson correlation between the ratio of phosphorylated to nonphosphorylated NFATc3 and the paw area at peak stance (Fig. 7G; r = 0.5351; p = 0.0071). Additional analysis showed a moderate negative Pearson correlation between levels of GFAP and the percent of shared stance (Fig. 7H; r = 0.5125; p = 0.0105).

730

DISCUSSION

751

Generally, exercise has many known benefits for overall health and has been shown to be protective against PD locomotor deficits in animal models (Lau et al., 2011) like many other pharmacological treatments (Ross and Petrovitch, 2001; Checkoway et al., 2002; Ravina et al., 2003). However, it is generally accepted that when patients are diagnosed with PD, dopaminergic loss of the nigrostriatal terminals is greater than 70%, while about 60% of the cell bodies of the SNpc are lost (Bernheimer et al., 1973; Riederer and Wuketich, 1976; Kordower et al., 2013), thus, rendering the protection study design in animal models much less translational to clinical PD. To more closely mimic the progressive dopaminergic cell loss and behavioral deficits seen clinically, we administered MPTP chronically and with increasing doses (Goldberg et al., 2011a,b; Sconce et al., 2015). Furthermore, PD patients are encouraged to exercise to help lighten motor symptoms and with the optimism that it might slow the progression of the disease. We are reporting in the current study that in our progressive mouse model of PD, intervening half-way through the progression with exercise alleviated motor deficits but did not prevent continued loss of the dopaminergic biomarker TH within either the striatum or SN. After the final administration of MPTP, the amount of exercise pursued by the mice increased back to the vehicle levels. Additionally, despite 4 weeks of MPTP administration, mice that pursued exercise gained similar weight and had similar grip strength, paw area at peak stance, MAX dA/dT, stance width, and percent shared stance, compared to the vehicle-

752

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

726 727 728

731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750

753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781

NSC 16249

No. of Pages 19

6 May 2015

12

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

Fig. 7. Pearson correlations of motor behaviors and biochemical markers. The expression of VMAT2 negatively correlated with (A) the paw area at peak stance and (B) the MAX dA/dT. The expression of VGLUT1 shows a significant positive correlation with (C) the % forepaw grip strength and (D) the MAX dA/dT. The relative expression levels of GLAST shows a significant positive correlation with (E) the paw area at peak stance and (F) the MAX dA/dT. (G) A moderate negative correlation was seen between the ratio of phosphorylated to non-phosphorylated NFATc3 and the paw area at peak stance, while (H) GFAP showed a negative correlation with the percent of shared stance (n = 24). Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822

823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841

treated mice. In comparison, mice that were administered MPTP for 4 weeks without exercise had a deficit in weight gain, which was associated with increased grip strength, stance width, paw area at peak stance, percent shared stance, and a decreased MAX dA/dT. By IHC and western blot, there were no differences in TH-ir within the DL striatum or the SN between the 4WK_MPTP and 4WK_MPTP + Ex groups despite the behavioral recovery observed in the mice that exercised. This suggests that exercise had an effect on other factors besides TH recovery that may be contributing to behavioral restoration. While 4 weeks of MPTP administration caused a decrease in VMAT2 and an increase in the ratio of glycosylated/non-glycosylated DAT within the SN, mice that were also administered 4 weeks of MPTP and pursued exercise showed the opposite trends which were similar to the vehicle group. Analysis of glutamate transporters within the SN revealed an analogous theme where the 4WK_MPTP + Ex mice maintained similar levels of VGLUT1, GLT-1, and EAAC1 compared to the vehicle and the 4WK_MPTP mice had significantly increased levels of these glutamate biomarkers, suggesting that exercised mice were capable of maintaining normal glutamate function. Additionally, animals that were administered 4 weeks of MPTP and pursued exercise showed a decrease in inflammatory markers that were increased in the 4WK_MPTP mice. Taken together, the return to vehicle levels of the glutamate markers and the association with the decrease in astrocytic/inflammatory markers suggests that following MPTP, there is an increase in extracellular glutamate within the SN via activation of the cortico-nigral/VGLUT1 pathway, which is reversed by exercise. These findings demonstrate that exercise did not modify the progressive loss of DA (as measured by TH) but perhaps alleviated motor deficits by compensating for DA release (as revealed by the changes in VMAT and DAT), maintaining glutamate homeostasis, and decreasing inflammation. Future studies will focus on measuring the extracellular levels of glutamate and DA within the SN and DL striatum using in vivo microdialysis (Meshul et al., 1999; Meredith et al., 2009). Exercise restored motor behaviors despite continued loss of TH in the DL striatum and SNpc Exercise has been known to alleviate some motor symptoms in animal models of PD (Tillerson et al., 2003; Al-Jarrah et al., 2007; Petzinger et al., 2007; Pothakos et al., 2009; Gorton et al., 2010; Vucˇkovic´ et al., 2010; Lau et al., 2011; Smith et al., 2011) as well as in the clinic (Bilowit, 1956; Szekely et al., 1982 ; Baatile et al., 2000; Toole et al., 2000; Petzinger et al., 2010). It is debated which kind of exercise is most effective and many mouse models have looked at forced exercise by treadmill running (Tillerson et al., 2003; Petzinger et al., 2007; Yoon et al., 2007; Pothakos et al., 2009; Vucˇkovic´ et al., 2010; Lau et al., 2011; Smith et al., 2011). To bypass the potential stress due to forced exercise that could interfere with exercise therapy (Howells et al., 2005), we installed running wheels into the mouse cages so each animal could volunteer to run at anytime. We found that following 2 weeks of MPTP treatment,

13

when running wheels were then installed, mice volunteered to exercise 74–90% less than the vehicle-treated animals during the 3rd and 4th week of toxin administration. During the following weeks off any MPTP treatment, the amount of volunteered exercise increased back to the vehicle-treated mice. Gorton et al. (2010) compared both forced exercise via treadmill running and voluntary exercise by cage wheels in the same study. In contrast to our findings, they found that an MPTP lesion did not reduce voluntary exercise. This could be attributed to the differences in their acute MPTP model of four high doses of toxin injected in the same day, versus our progressive toxin administration over 4 weeks with increasing doses. Another difference that could explain why the MPTP mice in our study had a decrease in voluntary exercise is because we administered MPTP during the same time they were allowed to exercise versus after the MPTP lesion. It is possible that the decrease in exercise is simply due to the ill-effects of MPTP toxicity, suppressing any typical mouse movement and hence, the increased exercise after MPTP treatment. However, if the 4WK_MPTP + Ex mice were not exercising as much during the last two weeks of MPTP administration because of toxicity, the intervention experimental design still holds clinical value where patients with progressed PD have trouble getting out of bed, let alone doing cardiovascular exercise. Furthermore, other behavioral measures followed the same trend of recovery in the 4WK_MPTP + Ex mice and these tests were administered 25 days after the last injection of MPTP. As mice age, they typically gain weight. In our progressive MPTP studies, we consistently see that mice administered MPTP either gain little weight or lose it (Goldberg et al., 2012). Animals were weighed before the experiment and again before they were euthanized. The animals administered MPTP for 2 and 4 weeks did not gain the typical weight as seen with the vehicle-treated mice. Interestingly, the 4WK_MPTP + Ex mice gained the most weight compared to all the other groups. In addition to weight changes, the grip strength test was also implemented before harvesting. Nowak and Hermsdo¨rfer (2006) reported that patients increased their grip force earlier in anticipation of the opposite hand dropping a weight in a container compared to healthy controls. In addition the PD patients produced more variable force levels at the impact of the weight which, according to the authors, could indicate deficits of sensorimotor integration associated with motor dysfunction in basal ganglia disorders. For a similar correlate in our study, mice administered MPTP for 4 weeks had increased forepaw grip strength. Fellows and Noth (2004) reported that grip force was abnormally high in the hold and lifting phases for PD patients regardless of DA medication. Potentially comparable with basal ganglia dysfunction in mice, perhaps the 4WK_MPTP mice have increased forepaw grip strength because they have trouble letting go of the apparatus. Nonetheless, the mice that exercised had restored grip strength comparable to the vehicle group despite 4 weeks of MPTP treatment. Our findings also indicate that other motor behaviors associated with gait dynamics were restored in animals

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902

NSC 16249

No. of Pages 19

6 May 2015

14 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

that exercise and were administered MPTP. By analyzing the area of the paws, we found that there was a significant increase in the paw area at peak stance as well as the MAX dA/dT or rate that the animal decelerates via paw area in contact with the ground, in the animals treated with MPTP for 4 weeks. This could indicate that the 4WK_MPTP mice were not picking up their paws as quickly or had a decrease in agility. Additionally, we found a significant increase in the stance width and a decrease in the percent that the stance was shared during the running form with mice administered MPTP for 4 weeks. Patients with PD are reported to have motor symptoms of postural changes and instability/falling (Politis et al., 2010; Jankovic, 2008). An increase in stance width could suggest that the animals are compensating for postural instability by forming a wider stance which coincides with increased paw to ground surface area at peak stance. The deficit in the percent of shared stance further supports a decrease in the typical running agility and postural changes in the MPTP-treated mice. Taken together, animals that exercised had behavioral and motor recovery in all measures despite 4 weeks of MPTP treatment. Even though many studies have shown that exercise is of benefit in the recovery of motor deficits, it has not been shown to be disease modifying, such that it neither attenuates the progression of the disease nor induces recovery in the population of neurons within the SNpc that are degenerating (Al-Jarrah et al., 2007; Petzinger et al., 2007; Pothakos et al., 2009; Petzinger et al., 2010; Vucˇkovic´ et al., 2010). Although we used a different model of PD, our data are in agreement with the observation from other studies that exercise does not spare or result in any recovery of dopaminergic neurons when initiated following toxin administration (Al-Jarrah et al., 2007; Moroz et al., 2004; O’Dell et al., 2007). The 4WK_MPTP + Ex group had complete recovery of motor deficits but no such improvement or prevention of dopaminergic loss as gauged by TH-ir within the DL striatum and SN. Nonetheless, the 4WK_MPTP and 4WK_MPTP + Ex groups had equivalent and significant loss of TH within the DL striatum and SN compared to the vehicle group. This indicates that in our progressive model, behavioral recovery does not imply nigrostriatal sparing or recovery. To elucidate the lack of neuronal recovery further, the levels of pTrkB were analyzed within the SN. It has been repeatedly reported that BDNF levels increase in hippocampal neurons after exercise (Oliff et al., 1998; Russo-Neustadt et al., 1999; Cotman and Berchtold, 2002; Adlard et al., 2005). BDNF is needed for dopaminergic neurotransmission (Guillin et al., 2001; Carvalho et al., 2008) and binds to the TrkB receptor, causing the receptor to dimerize and autophosphorylate, which then results in the promotion of a series of signaling cascades that regulate cell growth/differentiation, neuronal survival, and synaptic plasticity (Guillin et al., 2001; Carvalho et al., 2008; Cobb, 1999; Grewal et al., 1999; Chao, 2003; Minichiello, 2009; Skaper, 2012). Instead of analyzing the expression of BDNF directly within the SN, measuring the levels of the phosphorylated form of TrkB (pTrkB) was

felt to be a more straight forward and sensitive assessment of neuronal restoration. We are reporting that even though exercise evoked behavioral recovery, it did not result in any stimulation of the TrkB receptor in animals treated with MPTP within the SN. This suggests that other compensatory mechanisms that participate in DA regulation may have played a role in the behavioral recovery that was observed.

964

VMAT2 and DAT-glycosylation within the SN recovered to normal levels in animals that exercised

972

Even though mice that were administered MPTP and voluntarily exercised showed just as much TH-ir loss as the mice that were administered MPTP, we looked at the relative levels of VMAT2 and DAT-glycosylation within the SN to determine if there was differential regulation between DA biomarkers. Our results show that VMAT2 levels significantly decreased in the 4WK_MPTP mice but had fully recovered in the 4WK_MPTP + Ex mice. This suggests that exercise could have evoked the remaining SN cells to increase their overall dopaminergic function as previously reported (O’Dell et al., 2007). It has previously been reported in the 6-hydroxydopamine (6-OHDA) lesion models, that after initial nigrostriatal loss, the remaining cells normalize striatal DA signaling in part by increasing DA release (Zigmond and Stricker, 1984; Zigmond et al., 1984, 1989, 1992; Abercrombie et al., 1990; Robinson et al., 1990; Snyder et al., 1990). However, with progressive MPTP lesions over 4 weeks, we previously reported that striatal DA is significantly decreased and DOPAC/DA turnover is increased (Goldberg et al., 2011a, 2012). In addition we have reported a direct correlation between levels of TH in the SN by western blot analysis and the average number of TH neurons within the SNpc (Goldberg et al., 2011a), suggesting that the loss of TH within the SN in the current study is most likely due to the loss of SNpc DA neurons. Therefore, the results of the current study suggest that within the remaining nigrostriatal neurons, exercise either induced an increase of dopaminergic function or prolonged initial normalization of striatal DA signaling rather than sparing the dopaminergic neurons. Another way to gauge dopaminergic function is by determining DAT levels. Aside from the DL striatum, DAT can also be localized to the dendrites of the SN cell bodies and is thought to be crucial for terminating DA action by clearing extraneuronal DA (Iversen, 1971). DAT is a heavily glycosylated protein (Li et al., 2004). Generally, including carbohydrates in post-translational modifications of proteins can contribute to protein folding and its stability at that conformation, regulating protein trafficking, and protecting against proteolysis (Lis and Sharon, 1998). Li et al. (2004) carefully characterized DAT stability and activity when all or individual glycosylation sites were mutated and found that prevention of N-glycosylation decreased DA transport efficiency but did not terminate function. We analyzed both the fully glycosylated DAT at 80 kDa as well as the non-glycosylated DAT at 50 kDa. Calculating a ratio between the two forms

974

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

965 966 967 968 969 970 971

973

975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058

1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082

indicated that the 4WK_MPTP mice had elevated levels of the 80-kDa glycosylated DAT with respect to nonglycosylated DAT, whereas the 4WK_MPTP + Ex mice recovered to a DAT ratio similar to the vehicle group. These findings are consistent with the hypothesis that exercise could be provoking normal DA signaling through DAT regulation, but further studies are still needed to support this idea. Li et al. (2004) also observed that prevention of N-glycosylation reduced surface DAT, suggesting that non-glycosylated DAT could be more susceptible to degradation and that changes in DAT glycosylation are likely to impact DA clearance. Taken together with Li et al. (2004) reporting that non-glycosylated DAT still maintained transporter function, glycosylation of DAT could be a regulated mechanism contributing to normalizing DA extra/intra neuronal levels. Essentially an increase in glycosylated DAT as seen in the 4WK_MPTP mice implies that within the DAT protein population, increased levels of the transporter were more stable, efficient, and likely to reach the membrane surface. This suggests that the 4WK_MPTP mice may be compensating for abnormal DA levels by increasing the levels of a more stable DAT. Of note is a recent study in which a full unilateral nigrostriatal lesion with 6-OHDA resulted in a greater than 70% loss of striatal DAT, but with a surprising decrease in DA uptake of only 25% (Chotibut et al., 2012). As the loss of DAT increased, the degree of DA uptake continued to increase. Our findings of a more stable DAT following nigrostriatal DA loss would also be consistent with the observations of Chotibut et al. (2012). Even though DA levels were not determined in the current study, the data regarding VMAT2 and DAT recovery suggest the possibility that exercise results in increased DA levels (Hattori et al., 1994; Wilson and Marsden, 1995; Meeusen et al., 1995), resulting in behavioral improvement despite continued TH loss. Glutamate transporters were up-regulated in mice treated with MPTP for 4 weeks A common hypothesis within the PD field is that when the dopaminergic neurons degenerate, there is over activation of the glutamatergic STN neurons that project to the remaining SNpc neurons, resulting in further neuronal degeneration (Albin et al., 1989; Maesawa et al., 2004; however see Paul et al., 2004). Whether initial over activity of the STN is a cause or an effect of the DA neuronal loss is unknown, but the loss of dopaminergic function that contributes to the regulation of glutamatergic corticostriatal neurotransmission perpetuates a state of hyperexcitability (Day et al., 2006). Our results indicate that animals administered progressive doses of MPTP had significant increases in glutamate transporter expression of GLT-1, and EAAC1 within the SN. We suggest that this is due to an increase in extracellular glutamate levels. This hypothesis is consistent with our previous report that using another more chronic toxin animal model of PD, we find that following 5 weeks of MPTP/probenecid treatment, there was an increase in the basal extracellular level of glutamate within the SN (Meredith et al., 2009). However, the origin of that increased SN extracellular glutamate is not known.

15

VGLUT2 is a vesicular glutamate transporter utilized in the subthalamo-nigral projection and VGLUT1 is specific for the cortico-nigral terminal projections. VGLUT2 expression levels (data not shown) revealed a nonsignificant 20% increase in the 4WK_MPTP mice. Lack of a significant increase in VGLUT2 levels within the SN following MPTP does not necessarily imply that the STN glutamate neurons are not hyperexcitable. It could simply indicate that the number of glutamatergic vesicles remained the same with an increase in glutamate concentration or there were more vesicles within the terminal but with less VGLUT2 protein per vesicle. Interestingly, VGLUT1 expression significantly increased in the 4WK_MPTP mice, suggesting the possibility of increased glutamate originating from the motor cortex. This observation is contrary to the notion that depleting DA signaling in the SN would affect the direct and indirect pathways with the same outcome of quenching the thalamus and thereby suppressing glutamate signaling from the motor cortex to at least the SN. However, it is possible that the origin of the motor cortex input to the SN is different compared to that of other parts of the basal ganglia, including the striatum and STN. Nonetheless, mice that were administered MPTP initially, followed by intervention with the wheel running, showed recovery of VGLUT1, GLT-1, and EAAC1 protein levels within the SN that were associated with behavioral recovery. Also, GLAST levels in the mice that exercised showed significantly decreased levels. Exercise may be compensating for dopaminergic loss by increasing the activity of the thalamo-corticostriatal circuit (Meeusen et al., 1997; Bezard et al., 2003; O’Dell et al., 2007; Massie et al., 2010; Petzinger et al., 2010).

1083

Mice treated with MPTP for 4 weeks and exercised had less inflammation in the SN

1116

Perhaps by better regulation of glutamate excitotoxicity, mice that exercised also had decreased inflammatory markers. It is not known whether inflammation is a cause or outcome of PD, but it does seem to play a role in the progression of the disease. Postmortem PD brain tissue and other animal models indicate that production of pro-inflammatory factors by microglia mediates continued neurodegeneration (Liu, 2006; Chen et al., 2008). Other studies have shown that suppressing inflammatory factors associated with either astrocytes or microglial are protective against the loss of dopaminergic neurons (Chung et al., 2010). NFAT proteins are hyperphosphorylated and located within the cytoplasm during the resting state of the microglia, where activation contributes to the loss of neurons (Luo et al., 2014). Calcineurin (CN) targets the NFAT family of transcription factors (Crabtree and Olson, 2002) and after activation, CN rapidly dephosphorylates the NFAT proteins, which are then able to translocate into the nucleus to regulate gene transcription (Macian, 2005). Luo et al. (2014) showed that alpha-synuclein contributed to NFATc3 nuclear import, or increased non-phosphorylated NFATc3, by instigating CN phosphotase activity which leads to dopaminergic cell loss in their alpha-synuclein A53T conditional transgenic mouse model of PD. Our

1118

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115

1117

1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142

NSC 16249

No. of Pages 19

6 May 2015

16

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

1172

results show that animals that were administered increased doses of MPTP over 4 weeks and volunteered to exercise showed a significant increase in the ratio of resting-state phosphorylated NFATc3 compared to the 4WK_MPTP and vehicle groups. This suggests that exercise decreased inflammation by regulating the ratio of phosphorylated to non-phosphorylated NFATc3. Furthermore, most neurological disorders show microglia activation (Chen and Trapp, 2015) and the invasion of blood-borne macrophages which leads to astrocytosis (Mirza et al., 1999), that is, the increase in number or phenotypic change in astrocytes. In MPTP mouse models or 6-OHDA lesion models, the astrocytosis response begins after initial dopaminergic cell loss and is elevated after most dopaminergic neurons have died as evidenced by an increase in GFAP immunoreactivity (Teismann and Schulz, 2004; Maragakis and Rothstein, 2006). As an intermediate filament protein, GFAP is localized to astrocytes and has been pathologically observed to be up regulated for a long time post injury (Maragakis and Rothstein, 2006). Our results show that within the SN, animals treated with increased doses of MPTP over 4 weeks had greatly increased GFAP levels, which suggests astrocytosis processes were activated. Interestingly, the 4WK_MPTP + Ex group also had elevated GFAP levels compared to the vehicle group, however, the levels had significantly decreased compared to the 4WK_MPTP group. This suggests that exercise suppressed the inflammatory response sufficiently, resulting in a decrease in astrocytosis.

1173

Mechanistic links between exercise and inflammation

1174

Generally, there are many known benefits of exercise on overall well-being, including brain health. While exercise has shown many benefits in the hippocampus including enhancements in learning and memory, and improving executive function (Cotman et al., 2007), it has also been shown to increase angiogenesis or the proliferation of new blood vessels and improvement of blood flow in some brain regions (Black et al., 1990). Ehninger and Kempermann (2003) have previously reported that voluntary running wheel exercise results in an increase of microglia in several brain regions and this increase may assist in perpetuating better brain health and function. Furthermore, microglia can either suppress neuroinflammation and restore homeostasis by producing antiinflammatory cytokines (Colton, 2009) or microglia can promote pro-inflammatory markers when activated which has been shown to mediate continued neurodegeneration (Liu, 2006; Chen et al., 2008). Since the distinction between pro- and anti-inflammatory microglia cells was not determined in the current study, we can only speculate that the exercise-induced decrease in both the ratio of phosphorylated NFATc3 and in GFAP expression could be due to increased levels of anti-inflammatory microglia cells. Aside from the astrocytes and microglia, one of the interesting findings of this study was the observation that animals which exercised while being administered MPTP behaviorally recovered despite lack of TH

1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171

1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201

resurgence within the DL striatum and SN. It has been previously shown using in vivo microdialysis that exercise increases the DA concentration in the rat striatum (Hattori et al., 1994; Wilson and Marsden, 1995; Meeusen et al., 1995), which could be due in part to an increase in TH enzyme activity (Hattori et al., 1994). It is possible that within the remaining nigrostriatal cells of the mice administered MPTP, exercise increased DA concentration through increased TH enzyme activity, which would help explain the behavioral recovery. Additionally, mice administered MPTP alone showed significant increases in glutamate transporters indicating increased levels of extracellular glutamate, which can result in neuroinflammation in and or itself through excitotoxicity. Also by in vivo microdialysis, it has been shown that MPTP administration increases extracellular glutamate levels within the SN (Meredith et al., 2009), a finding consistent with the increase in SN glutamate transporters in the current study. Perhaps by normalizing DA output by exercise, the mice administered MPTP and exercised could suppress glutamate excitotoxicity by regulating the activity of the DA–D2 receptors located on the SN cell bodies (i.e., DA release resulting in DA–D2 inhibition of the DA neurons) or by activating pre-synaptic DA–D2 receptors on glutamate terminals within the SN, resulting in a decrease in glutamate release (Xu et al., 1999; Hatzipetros and Yamamoto, 2006). This would explain why the mice that exercised showed glutamate transporter and GFAP levels within the SN that were similar to the vehicle group. Taken together, our data indicate that intervention with exercise can alleviate behavioral deficits despite 4 weeks of MPTP and do so without restoring or even preventing continued loss of nigrostriatal TH levels. We are reporting that this dichotomy could be explained by exercise increasing DA release within the remaining cells (Hattori et al., 1994; Wilson and Marsden, 1995; Meeusen et al., 1997) as evident by the restoration of VMAT2 levels (i.e., increased uptake of DA into either more synaptic vesicles or increase in VMAT protein expression/vesicle) and glycosylated DAT [i.e., as intracellular DA levels increase due to exercise, the more stable form of DAT (i.e., glycosylated) decreases to vehicle levels]. Furthermore, our results suggest that exercise alleviated excess glutamate within the SN as indicated by restored levels of VGLUT1, EAAC1, and GLT-1. These data are consistent with the decrease in basal levels of glutamate following exercise (Meeusen et al., 1997). We also observed a suppression of the inflammatory response in the MPTP group that exercised. Future studies should not solely rely on different kinds of exercise as a means of treatment, but rather in addition to pharmaceutical approaches that could provoke recovery or sprouting of the remaining dopaminergic neurons/terminals (Sconce et al., 2015).

1202

UNCITED REFERENCES

1257

Cepeda et al. (1993), Levine et al. (1996), Smith and Villalba (2008), Umemiya and Raymond (1997).

1258

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256

1259

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 1260

REFERENCES

1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329

Abercrombie ED, Bonatz AE, Zigmond MJ (1990) Effects of L-dopa on extracellular dopamine in striatum of normal and 6hydroxydopamine-treated rats. Brain Res 525(1):36–44. Adlard PA, Perreau VM, Cotman CW (2005) The exercise-induced expression of BDNF within the hippocampus varies across lifespan. Neurobiol Aging 26(4):511–520. Albin RL, Young AB, Penney JB (1989) The functional anatomy of basal ganglia disorders. Trends Neurosci 12(10):366–375. Al-Jarrah M, Pothakos K, Novikova L, Smirnova IV, Kurz MJ, StehnoBittel L, Lau YS (2007) Endurance exercise promotes cardiorespiratory rehabilitation without neurorestoration in the chronic mouse model of parkinsonism with severe neurodegeneration. Neuroscience 149(1):28–37. Amende I, Kale A, McCue S, Glazier S, Morgan JP, Hampton TG (2005) Gait dynamics in mouse models of Parkinson’s disease and Huntington’s disease. J Neuroeng Rehabil 2:20. Baatile J, Langbein WE, Weaver F, Maloney C, Jost MB (2000) Effect of exercise on perceived quality of life of individuals with Parkinson’s disease. J Rehabil Res Dev 37(5):529–534. Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F1 (1973) Brain dopamine and the syndromes of Parkinson and Huntington Clinical, morphological and neurochemical correlations. J Neurol Sci 20(4):415–455. Bezard E, Gross CE, Brotchie JM (2003) Presymptomatic compensation in Parkinson’s disease is not dopamine-mediated. Trends Neurosci 26(4):215–221. Bilowit DS (1956) Establishing physical objectives in the rehabilitation of patients with Parkinson’s disease; gymnasium activities. Phys Ther Rev 36(3):176. Black JE, Isaacs KR, Anderson BJ, Alcantara AA, Greenough WT (1990) Learning causes synaptogenesis, whereas motor activity causes angiogenesis, in cerebellar cortex of adult rats. Proc Natl Acad Sci 87(14):5568–5572. Blandini F, Porter RH, Greenamyre JT (1996) Glutamate and Parkinson’s disease. Mol Neurobiol 12(1):73–94. Blandini F, Armentero MT (2012) Animal models of Parkinson’s disease. FEBS J 279(7):1156–1166. Carvalho AL, Caldeira MV, Santos SD, Duarte CB (2008) Role of the brain-derived neurotrophic factor at glutamatergic synapses. Br J Pharmacol 153(Suppl. 1):S310–S324. Cepeda C, Buchwald NA, Levine MS (1993) Neuromodulatory actions of dopamine in the neostriatum are dependent upon the excitatory amino acid receptor subtypes activated. Proc Natl Acad Sci USA 90:9576–9580. Chao MV (2003) Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat Rev Neurosci 4(4):299–309. Checkoway H, Powers K, Smith-Weller T, Franklin GM, Longstreth Jr WT, Swanson PD (2002) Parkinson’s disease risks associated with cigarette smoking, alcohol consumption, and caffeine intake. Am J Epidemiol 155(8):732–738. Chen H, O’Reilly EJ, Schwarzschild MA, Ascherio A (2008) Peripheral inflammatory biomarkers and risk of Parkinson’s disease. Am J Epidemiol 167(1):90–95. Chen Z, Trapp BD (2015) Microglia and neuroprotection. J Neurochem. http://dx.doi.org/10.1111/jnc.13062. Cheramy A, Leviel V, Glowinski J (1981) Dendritic release of dopamine in the substantia nigra. Nature 289(5798):537–543. Choi DW (1988) Glutamate neurotoxicity and diseases of the nervous system. Neuron 1(8):623–634. Chotibut T, Apple DM, Jefferis R, Salvatore MF (2012) Dopamine transporter loss in 6-OHDA Parkinson’s model is unmet by parallel reduction in dopamine uptake. PLoS One 7(12):e52322. Colton CA (2009) Heterogeneity of microglial activation in the innate immune response in the brain. J Neuroimmune Pharmacol 4(4):399–418. Cotman CW, Berchtold NC, Christie LA (2007) Exercise builds brain health: key roles of growth factor cascades and inflammation. Trends Neurosci 30(9):464–472.

17

Coyle JT, Puttfarcken P (1993) Oxidative stress, glutamate, and neurodegenerative disorders. Science 262(5134):689–695. Chung YC, Kim SR, Jin BK (2010) Paroxetine prevents loss of nigrostriatal dopaminergic neurons by inhibiting brain inflammation and oxidative stress in an experimental model of Parkinson’s disease. J Immunol 185(2):1230–1237. Cobb MH (1999) MAP kinase pathways. Prog Biophys Mol Biol 71(3– 4):479–500. Cotman CW, Berchtold NC (2002) Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 25(6):295–301. Crabtree GR, Olson EN (2002) NFAT signaling: choreographing the social lives of cells. Cell 109:S67–S79. Day M, Wang Z, Ding J, An X, Ingham CA, Shering AF, Surmeier DJ (2006) Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nat Neurosci 9(2):251–259. Ehninger D, Kempermann G (2003) Regional effects of wheel running and environmental enrichment on cell genesis and microglia proliferation in the adult murine neocortex. Cerebral Cortex 13(8):845–851. Fellows SJ, Noth J (2004) Grip force abnormalities in de novo Parkinson’s disease. Mov Disord 19(5):560–565. Fisher BE, Petzinger GM, Nixon K, Hogg E, Bremmer S, Meshul CK, Jakowec MW (2004) Exercise-induced behavioral recovery and neuroplasticity in the 1-methyl-4-phenyl-1, 2, 3,6tetrahydropyridine-lesioned mouse basal ganglia. J Neurosci Res 77(3):378–390. Fisher BE, Wu AD, Salem GJ, Song J, Lin CHJ, Yip J, Petzinger G (2008) The effect of exercise training in improving motor performance and corticomotor excitability in people with early Parkinson’s disease. Arch Phys Med Rehabil 89(7):1221–1229. Geffen LB, Jessell TM, Cuello AC, Iversen LL (1976) Release of dopamine from dendrites in rat substantia nigra. Nature:258–260. Gerecke KM, Jiao Y, Pani A, Pagala V, Smeyne RJ (2010) Exercise protects against MPTP-induced neurotoxicity in mice. Brain Res 1341:72–83. Goldberg NR, Haack AK, Lim NS, Janson OK, Meshul CK (2011a) Dopaminergic and behavioral correlates of progressive lesioning of the nigrostriatal pathway with 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine. Neuroscience 180:256–271. Goldberg NR, Hampton T, McCue S, Kale A, Meshul CK (2011b) Profiling changes in gait dynamics resulting from progressive 1methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced nigrostriatal lesioning. J Neurosci Res 89(10):1698–1706. Goldberg NR, Fields V, Pflibsen L, Salvatore MF, Meshul CK (2012) Social enrichment attenuates nigrostriatal lesioning and reverses motor impairment in a progressive 1-methyl-2-phenyl-1, 2, 3, 6tetrahydropyridine (MPTP) mouse model of Parkinson’s disease. Neurobiol Dis 45(3):1051–1067. Gorton LM, Vuckovic MG, Vertelkina N, Petzinger GM, Jakowec MW, Wood RI (2010) Exercise effects on motor and affective behavior and catecholamine neurochemistry in the MPTP-lesioned mouse. Behav Brain Res 213(2):253–262. Greenfield SA (1985) The significance of dendritic release of transmitter and protein in the substantia nigra. Neurochem Int 7(6):887–901. Grewal SS, York RD, Stork PJ (1999) Extracellular-signal-regulated kinase signalling in neurons. Curr Opin Neurobiol 9(5):544–553. Guillin O, Diaz J, Carroll P, Griffon N, Schwartz JC, Sokoloff P (2001) BDNF controls dopamine D3 receptor expression and triggers behavioural sensitization. Nature 411(6833):86–89. Hattori S, Naoi M, Nishino H (1994) Striatal dopamine turnover during treadmill running in the rat: relation to the speed of running. Brain Res Bull 35(1):41–49. Hatzipetros T, Yamamoto BK (2006) Dopaminergic and GABAergic modulation of glutamate release from rat subthalamic nucleus efferents to the substantia nigra. Brain Res 1076(1):60–67. Holmer HK, Keyghobadi M, Moore C, Meshul CK (2005) L-dopainduced reversal in striatal glutamate following partial depletion of

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400

NSC 16249

No. of Pages 19

6 May 2015

18 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx

nigrostriatal dopamine with 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine. Neuroscience 136(1):333–341. Howells FM, Russell VA, Mabandla MV, Kellaway LA (2005) Stress reduces the neuroprotective effect of exercise in a rat model for Parkinson’s disease. Behav Brain Res 165(2):210–220. Iversen LL (1971) Role of transmitter uptake mechanisms in synaptic neurotransmission. Brit J Pharmacol 41(4):571–591. Jankovic J (2008) Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry 79(4):368–376. Kale A, Amende I, Meyer GP, Crabbe JC, Hampton TG (2004) Ethanol’s effects on gait dynamics in mice investigated by ventral plane videography. Alcohol Clin Exp Res 28(12):1839–1848. Kaneko T, Fujiyama F, Hioki H (2002) Immunohistochemical localization of candidates for vesicular glutamate transporters in the rat brain. J Comp Neurol 444(1):39–62. Klockgether T, Turski L, Honore´ T, Zhang Z, Gash DM, Kurlan R, Greenamyre JT (1991) The AMPA receptor antagonist NBQX has antiparkinsonian effects in monoamine-depleted rats and MPTPtreated monkeys. Ann Neurol 30(5):717–723. Kordower JH, Olanow CW, Dodiya HB, Chu Y, Beach TG, Adler CH, Bartus RT (2013) Disease duration and the integrity of the nigrostriatal system in Parkinson’s disease. Brain 136(8):2419–2431. Lau YS, Patki G, Das-Panja K, Le WD, Ahmad SO (2011) Neuroprotective effects and mechanisms of exercise in a chronic mouse model of Parkinson’s disease with moderate neurodegeneration. Eur J Neurosci 33(7):1264–1274. Levine MS, Altemus KL, Cepeda C, et al. (1996) Modulatory actions of dopamine on NMDA receptor-mediated responses are reduced in D1A-deficient mutant mice. J Neurosci 16:5870–5882. Li Li-Bin, Chen Nianhang, Ramamoorthy Sammanda, Chi Limen, Cui Xiao-Nan, Wang Lijuan C, Reith Maarten EA (2004) The role of Nglycosylation in function and surface trafficking of the human dopamine transporter. J Biol Chem 279(20):21012–21020. Lis H, Sharon N (1998) Lectins: carbohydrate-specific proteins that mediate cellular recognition. Chem Rev 98(2):637–674. Liu B (2006) Modulation of microglial pro-inflammatory and neurotoxic activity for the treatment of Parkinson’s disease. AAPS J 8(3):E606–E621. Luo J, Sun L, Lin X, Liu G, Yu J, Parisiadou L, Cai H (2014) A calcineurin and NFAT-dependent pathway is involved in asynuclein-induced degeneration of midbrain dopaminergic neurons. Hum Mol Genet:ddu377. Macian F (2005) NFAT proteins: key regulators of T-cell development and function. Nat Rev Immunol 5:472–484. Maesawa S, Kaneoke Y, Kajita Y, Usui N, Misawa N, Nakayama A, Yoshida J (2004) Long-term stimulation of the subthalamic nucleus in hemiparkinsonian rats: neuroprotection of dopaminergic neurons. J Neurosurg 100(4):679–687. Maragakis NJ, Rothstein JD (2004) Glutamate transporters: animal models to neurologic disease. Neurobiol Dis 15(3):461–473. Maragakis NJ, Rothstein JD (2006) Mechanisms of disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neurol 2(12):679–689. Massie A, Goursaud S, Schallier A, Vermoesen K, Meshul CK, Hermans E, Michotte Y (2010) Time-dependent changes in GLT-1 functioning in striatum of hemi-Parkinson rats. Neurochem Int 57(5):572–578. Meeusen R, Smolders I, Sarre S, De Meirleir K, Keizer H, Serneels M, Michotte Y (1997) Endurance training effects on neurotransmitter release in rat striatum: an in vivo microdialysis study. Acta Physiol Scand 159(4):335–341. Meredith GE, Totterdell S, Potashkin JA, Surmeier DJ (2008) Modeling PD pathogenesis in mice: advantages of a chronic MPTP protocol. Parkinsonism Relat Disord 14(Suppl. 2):S112–S115. Meredith GE, Totterdell S, Beales M, Meshul CK (2009) Impaired glutamate homeostasis and programmed cell death in a chronic MPTP mouse model of Parkinson’s disease. Exp Neurol 219(1):334–340.

Meshul CK, Emre N, Nakamura CM, Allen C, Donohue MK, Buckman JF (1999) Time-dependent changes in striatal glutamate synapses following a 6-hydroxydopamine lesion. Neuroscience 88(1):1–16. Minichiello L (2009) TrkB signalling pathways in LTP and learning. Nat Rev Neurosci 10(12):850–860. Mirza B, Hadberg H, Thomsen P, Moos T (1999) The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson’s disease. Neuroscience 95(2):425–432. Nowak DA, Hermsdo¨rfer J (2006) Predictive and reactive control of grasping forces: on the role of the basal ganglia and sensory feedback. Exp Brain Res 173(4):650–660. O’dell SJ, Gross NB, Fricks AN, Casiano BD, Nguyen TB, Marshall JF (2007) Running wheel exercise enhances recovery from nigrostriatal dopamine injury without inducing neuroprotection. Neuroscience 144(3):1141–1151. Oliff HS, Berchtold NC, Isackson P, Cotman CW (1998) Exerciseinduced regulation of brain-derived neurotrophic factor (BDNF) transcripts in the rat hippocampus. Mol Brain Res 61(1):147–153. Paul G, Meissner W, Rein S, Harnack D, Winter C, Hosmann K, Kupsch A (2004) Ablation of the subthalamic nucleus protects dopaminergic phenotype but not cell survival in a rat model of Parkinson’s disease. Exp Neurol 185(2):272–280. Politis M, Wu K, Molloy S, Bain PG, Chaudhuri K, Piccini P (2010) Parkinson’s disease symptoms: the patient’s perspective. Mov Disord 25(11):1646–1651. Pothakos K, Kurz MJ, Lau YS (2009) Restorative effect of endurance exercise on behavioral deficits in the chronic mouse model of Parkinson’s disease with severe neurodegeneration. BMC Neurosci 10(1):6. Petzinger GM, Walsh JP, Akopian G, Hogg E, Abernathy A, Arevalo P, Jakowec MW (2007) Effects of treadmill exercise on dopaminergic transmission in the 1-methyl-4-phenyl-1, 2, 3, 6tetrahydropyridine-lesioned mouse model of basal ganglia injury. J Neurosci 27(20):5291–5300. Petzinger GM, Fisher BE, Van Leeuwen JE, Vukovic M, Akopian G, Meshul CK, Jakowec MW (2010) Enhancing neuroplasticity in the basal ganglia: the role of exercise in Parkinson’s disease. Mov Disord 25(S1):S141–S145. Petzinger GM, Fisher BE, McEwen S, Beeler JA, Walsh JP, Jakowec MW (2013) Exercise-enhanced neuroplasticity targeting motor and cognitive circuitry in Parkinson’s disease. Lancet Neurol 12(7):716–726. Plaitakis A, Shashidharan P (2000) Glutamate transport and metabolism in dopaminergic neurons of substantia nigra: implications for the pathogenesis of Parkinson’s disease. J Neurol 247(2):II25–II35. Ravina BM, Fagan SC, Hart RG, Hovinga CA, Murphy DD, Dawson TM, Marler JR (2003) Neuroprotective agents for clinical trials in Parkinson’s disease: a systematic assessment. Neurology 60(8):1234–1240. Riederer P, Wuketich S (1976) Time course of nigrostriatal degeneration in Parkinson’s disease. J Neural Transm 38(3– 4):277–301. Robinson TE, Castan eda E, Whishaw IQ (1990) Compensatory changes in striatal dopamine neurons following recovery from injury induced by 6-OHDA or methamphetamine: a review of evidence from microdialysis studies. Can J Psychol 44(2):253–275. Robinson S, Freeman P, Moore C, Touchon JC, Krentz L, Meshul CK (2003) Acute and subchronic MPTP administration differentially affects striatal glutamate synaptic function. Exp Neurol 180(1):74–87. Ross GW, Petrovitch H (2001) Current evidence for neuroprotective effects of nicotine and caffeine against Parkinson’s disease. Drugs Aging 18(11):797–806. Rothstein JD, Martin L, Levey AI, Dykes-Hoberg M, Jin L, Wu D, Kuncl RW (1994) Localization of neuronal and glial glutamate transporters. Neuron 13(3):713–725. Russo-Neustadt A, Beard RC, Cotman CW (1999) Exercise, antidepressant medications, and enhanced brain derived

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540

NSC 16249

No. of Pages 19

6 May 2015

M. D. Sconce et al. / Neuroscience xxx (2015) xxx–xxx 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1620 1621 1622

neurotrophic factor expression. Neuropsychopharmacology 21(5):679–682. Sconce MD, Churchill MJ, Moore C, Meshul CK (2015) Intervention with 7,8-dihydroxyflavone blocks further striatal terminal loss and restores motor deficits in a progressive mouse model of Parkinson’s disease. Neuroscience 290:454–471. Shashidharan P, Huntley GW, Murray JM, Buku A, Moran T, Walsh MJ, Plaitakis A (1997) Immunohistochemical localization of the neuron-specific glutamate transporter EAAC1 (EAAT3) in rat brain and spinal cord revealed by a novel monoclonal antibody. Brain Res 773(1):139–148. Skaper SD (2012) The neurotrophin family of neurotrophic factors: an overview. Methods Mol Biol 846:1–12. Smith BA, Goldberg NR, Meshul CK (2011) Effects of treadmill exercise on behavioral recovery and neural changes in the substantia nigra and striatum of the 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine-lesioned mouse. Brain Res 1386:70–80. Smith Y,, Villalba R (2008) Striatal and extrastriatal dopamine in the basal ganglia: An overview of its anatomical organization in normal and Parkinsonian brains. Mov Disord 23(Suppl. 3):S534–S547. Snyder GL, Keller Jr RW, Zigmond MJ (1990) Dopamine efflux from striatal slices after intracerebral 6-hydroxydopamine: evidence for compensatory hyperactivity of residual terminals. J Pharmacol Exp Ther 253(2):867–876. Szekely BC, Kosanovich NN, Sheppard W (1982) Adjunctive treatment in Parkinson’s disease: physical therapy and comprehensive group therapy. Rehabil Lit 43:72–76. Teismann P, Schulz JB (2004) Cellular pathology of Parkinson’s disease: astrocytes, microglia and inflammation. Cell Tissue Res 318:149–161. Tieu K (2011) A guide to neurotoxic animal models of Parkinson’s disease. Cold Spring Harb Perspect Med 1(1):a009316. Tillerson JL, Caudle WM, Reveron ME, Miller GW (2003) Exercise induces behavioral recovery and attenuates neurochemical deficits in rodent models of Parkinson’s disease. Neuroscience 119(3):899–911. Toole T, Hirsch MA, Forkink A, Lehman DA, Maitland CG (2000) The effects of a balance and strength training program on equilibrium in Parkinsonism: a preliminary study. NeuroRehabilitation 14(3):165–174.

19

Touchon JC, Moore C, Frederickson J, Meshul CK (2004) Lesion subthalamic or motor thalamic nucleus in 6-hydroxydopaminetreated rats: effects on striatal glutamate and apomorphineinduced contralateral rotations. Synapse 51(4):287–298. Umemiya M, Raymond LA (1997) Dopaminergic modulation of excitatory postsynaptic currents in rat neostriatal neurons. J Neurophysiol 78:1248–1255. Vucˇkovic´ MG, Li Q, Fisher B, Nacca A, Leahy RM, et al. (2010) Exercise elevates dopamine D2 receptor in a mouse model of Parkinson’s disease: in vivo imaging with [18F]fallypride. Mov Disord. 25(16):2777–2784. Walker RH, Koch RJ, Sweeney JE, Moore C, Meshul CK (2009) Effects of subthalamic nucleus lesions and stimulation upon glutamate levels in the dopamine-depleted rat striatum. Neuroreport 20(8):770–775. Wilson WM, Marsden CA (1995) Extracellular dopamine in the nucleus accumbens of the rat during treadmill running. Acta Physiol Scand 155(4):465–466. Xu SG, Prasad C, Smith DE (1999) Neurons exhibiting dopamine d 2 receptor immunoreactivity in the substantia nigra of the mutant weaver mouse. Neuroscience 89(1):191–207. Yoon MC, Shin MS, Kim TS, Kim BK, Ko IG, Sung YH, Kim CJ (2007) Treadmill exercise suppresses nigrostriatal dopaminergic neuronal loss in 6-hydroxydopamine-induced Parkinson’s rats. Neurosci Lett 423(1):12–17. Zigmond MJ, Acheson AL, Stachowiak MK, Stricker EM (1984) Neurochemical compensation after nigrostriatal bundle injury in an animal model of preclinical parkinsonism. Arch Neurol 41(8):856–861. Zigmond MJ, Stricker EM (1984) Parkinson’s disease: studies with an animal model. Life Sci 35(1):5–18. Zigmond MJ, Berger TW, Grace AA, Stricker EM (1989) Compensatory responses to nigrostriatal bundle injury. Studies with 6-hydroxydopamine in an animal model of parkinsonism. Mol Chem Neuropathol 10(3):185–200. Zigmond MJ, Hastings TG, Abercrombie ED (1992) Neurochemical responses to 6-hydroxydopamine and L-dopa therapy: implications for Parkinson’s disease. Ann N Y Acad Sci 648:71–86.

(Accepted 28 April 2015) (Available online xxxx)

Please cite this article in press as: Sconce MD et al. Intervention with exercise restores motor deficits but not nigrostriatal loss in a progressive MPTP mouse model of Parkinson’s disease. Neuroscience (2015), http://dx.doi.org/10.1016/j.neuroscience.2015.04.069

1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618 1619