J Neuropathol Exp Neurol Vol. 64, No. 11 Copyright Ó 2005 by the American Association of Neuropathologists, Inc. November 2005 pp. 936–947

ORIGINAL ARTICLE

Coordinated and Spatial Upregulation of Arc in Striatonigral Correlates With L-Dopa-Induced Behavioral Sensitization in Dyskinetic Rats

Ve´ronique Sgambato-Faure, PhD, Virginie Buggia, MSc, Francxois Gilbert, MSc, Daniel Le´vesque, PhD, Alim-Louis Benabid, MD, PhD, and Francxois Berger, MD, PhD Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021

remains the best symptomatic treatment (6). However, its long- Abstract term use is commonly associated with side effects such as Although oral administration of L-Dopa remains the best therapy motor fluctuations and abnormal involuntary movements, for Parkinson disease, its long-term administration causes the appear- namely L-Dopa-induced dyskinesia (LID) (7–10; for review, ance of abnormal involuntary movements such as dyskinesia. Although [11]). The genesis of LID is not fully understood, but major persistent striatal induction of some has already been asso- factors include degree of presynaptic nigral denervation (12– ciated with such pathologic profiles in hemiparkinsonian rats, molec- 15); onset, dose, and manner of administration of L-Dopa (16); ular and cellular mechanisms underlying such long-term adaptations sensitization of dopaminergic receptors and their pulsatile remain to be elucidated. In this study, using a rat model of L-Dopa- stimulation (17, 18); imbalance between the output structures induced dyskinesia, we report that activity regulated cytoskeletal (Arc)- of basal ganglia (3, 4); as well as alteration of activity and associated protein is strongly upregulated in the lesioned striatum and discharge pattern of neurons of basal ganglia (19, 20). that the extent of its induction further varies according to the However, the precise mechanisms by which chronic L-Dopa occurrence or absence of locomotor sensitization. Moreover, Arc is leads to such long-term motor alterations require elucidation. preferentially induced, along with FosB, nur77, and homer-1a, in From experimental studies using animal models of LID striatonigral neurons, which express mRNA encoding the precursor (i.e. dopaminergic-lesioned animals chronically treated with of dynorphin. Given the likely importance of Arc in the regulation of L-Dopa given intermittently to simulate dosing schedules in cytoskeleton during synaptic plasticity, its upregulation supports the parkinsonian patients), it has been shown that abnormal in- hypothesis that a relationship exists between cytoskeletal modifica- formation storage in the corticostriatal pathways correlates with tions and the longlasting action of chronically administrated L-Dopa. LID (21). Moreover, it has been shown that in the striatum, Key Words: Arc, Dynorphin, Dyskinesia, homer-1a, L-Dopa, persistent molecular alterations noticeably related to glutamate Locomotor sensitization, nur77. signaling are involved in LID (22, 23). Many of these modulations are observed in striatonigral-projecting cells of the striatum, suggesting that dynorphin-containing cells may INTRODUCTION be associated with the development of behavioral sensitization Parkinson disease (PD) is a neurologic disorder that occurs in animal models of PD after L-Dopa treatment, characterized by an extensive loss of dopaminergic neurons although the involvement of opioid receptors is still a matter in the substantia nigra pars compacta (1, 2). The loss of of debate (24–29). Often compared with an aberrant form of dopamine within the striatum disturbs the functional organi- motor (30, 31), LID might also involve synaptic re- zation of the basal ganglia networks by causing an imbalance modeling. Among the cytoskeleton-related genes, the activity- between the 2 efferent striatal output pathways (direct: regulated cytoskeletal (Arc)-associated gene is of particular striatonigral; indirect: striatopallido–subthalamonigral) (3). interest. Initially identified in differential screens for seizure- Dopamine deficiency causes the motor symptoms of PD such stimulated hippocampal mRNA (32, 33), Arc mRNA rapidly as akinesia, rigidity, and tremor (3, 4). Despite the intense localizes to discrete dendritic regions that have received direct development of new therapeutic approaches (5), administra- synaptic stimulation (34, 35). Moreover, Arc is required for tion of the natural dopamine precursor levodopa (L-Dopa) still long-term potentiation as well as in the hippocampus (36–38). In the present study, we asked whether chronic L-Dopa From Inserm U318 (VS-F, VB, A-LB, FB), Laboratoire de Neurosciences Pre´cliniques, CHU Pavillon B, Grenoble, France; and Centre de recherche increased the expression of Arc and whether its L-Dopa-driven en Neuroscience (FG, DL), CHUL, Sainte Foy, Que´bec, Canada. induction could be correlated with dyskinesia or behavioral Send correspondence and reprint requests to: Ve´ronique Sgambato-Faure, sensitization. Using hemiparkinsonian rats that were chron- PhD, INSERM U704, Dynamique des Re´seaux Neuronaux, 2280 Rue de ically treated with L-Dopa and developed dyskinetic profiles, la Piscine, BP53, 38041 Grenoble cedex 9, France; E-mail: veronique. [email protected] we observed that a strong upregulation of Arc at both gene and This work was supported by grants from Institut National de la Sante´ et de la protein levels, in response to L-Dopa administration in the Recherche Me´dicale. dopaminergic-depleted striatum, was spatially coincident with

936 J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Arc Correlates With L-Dopa-Induced Motor Behavior that of the FosB-like proteins, homer-1a, nur77 (also known as practice. Rats were observed individually after each injection. nerve growth factor-inducible B [NGFI-B]), and preprodynor- Another small group of hemiparkinsonian rats (n = 3) were phin (PPDyn) mRNA levels, in the striatal subregion according treated for a 3-week period with lower doses of L-Dopa to the subtype of motor behavior displayed. Moreover, we (10 mg/kg) and benserazide (2 mg/kg). For pharmacologic observed that Arc is colocalized with PPDyn mRNA in neurons studies, 4 groups of hemiparkinsonian rats (n = 12) were treated of the direct striatonigral pathway. Finally, we found that with L-Dopa (at 50 mg/kg plus benserazide at 10 mg/kg) L-Dopa-induced motor behavioral response and Arc upregulation twice daily for 10 days as follows: administration of L-Dopa are not affected by a modulation of opioid receptor activity. alone, coadministration of L-Dopa plus naloxone dihydro- Altogether, these findings highlight a possible involvement of chloride (at 10 mg/kg), coadministration of L-Dopa plus Arc in the development or maintenance of LID and locomotor U-50,488H (trans-3,4-di-chloro-N-methyl-N[2]-[(1-pyrrolidinyl) sensitization and further support a role for cytoskeletal remod- cyclohexyl] benzeneacetamide methane-sulfonate salt) (at

eling in the regulation of such long-term motor alterations. 1 mg/kg), and administration of L-Dopa plus an acute Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021 injection of nor-binaltorphimine di-hydrochloride (20 mg/kg) 1 hour before L-Dopa on the last day of treatment. MATERIALS AND METHODS The animal experimental protocols performed in this study strictly conformed to the guidelines of the French Agriculture Motor Behavior Analysis and Forestry Ministry and were approved by INSERM. Briefly, individual rats were observed daily in their home cage 1 hour after the injection, when the response to L-Dopa Lesion Surgery was maximal. Movements were considered as dyskinetic when Male adult Wistar rats, weighing 260 to 300 g, were both of the following criteria were fulfilled: induction by maintained on a 12-hour light/dark cycle with food and tap L-Dopa of repetitive and abnormal movements of the body water available ad libitum. All animals in this study were contralateral to the lesion. Abnormal involuntary movements anesthetized with chloral hydrate (400 mg/kg) and subjected to were subdivided into 3 subtypes: forelimb dyskinesia, orolin- unilateral 6-OHDA lesion in the left substantia nigra pars gual dyskinesia, and axial dyskinesia (40). The severity of each compacta (SNc) using a conventional stereotaxic instrument dyskinesia subtype was evaluated for each animal by assigning (David Kopf, Phymep, Paris, France). The lesions were per- a rating scale (0–4) (14, 41). Locomotor sensitization was also formed according to a standard procedure. Briefly, 12 mgof evaluated by counting the number of contralateral rotations per 6-OHDA-HCl (Sigma, Saint Quentin Fallavier, France) dis- minute. Finally, animals were videotaped during the treatment solved in 0.1% ascorbate saline were injected in the left SNc at and before death. the following coordinates (according to the atlas of Paxinos and Watson (39)) in millimeters and relative to Bregma and Tissue Preparation cortical surface: A = 5.3; L = 2.1; H = 7.8. To preserve norad- Two or 24 hours after the last L-Dopa injection, animals renergic fibers, all animals were intraperitoneally (IP) injected received a lethal dose of chloral hydrate and were killed by with desipramine-HCl (25 mg/kg; Sigma) at least 30 minutes intracardiac perfusion of RingerÕs solution followed by 4% before 6-OHDA injection. paraformaldehyde in 0.1 M Na2HPO4/NaH2PO4 buffer, pH 7.5 Amphetamine-Induced Rotational Behavior (phosphate buffer). were removed and postfixed in the same fixative solution for 2 hours, washed overnight in 0.1 M Ten to 15 days after lesion, animals were tested for phosphate buffer containing 15% sucrose, and then frozen. amphetamine-induced rotation. Rats received amphetamine Rostrocaudal series of coronal sections (20 mm) were cut on sulfate salt (2.5 mg/kg, IP; Sigma) and were placed in a vertical a cryostat and then kept in a solution containing 30% ethylene cylinder. After a period of 30 to 45 minutes, circling behavior glycol, 30% glycerol, 0.1 M phosphate buffer, and 0.1% diethyl was observed for 15 minutes. Only animals showing individ- pyrocarbonate (DEPC) at 20°C until they were processed for $ ual means 5 full turns/minute in the direction ipsilateral to either in situ hybridization or immunohistochemistry. the lesion were selected for the study. This score has been shown to correspond to at least 90% of DA depletion in the striatum (14). The effectiveness of the lesion was further In Situ Hybridization 35 confirmed postmortem by means of tyrosine hydroxylase (TH) The antisense probes used in this study were a[ -S]UTP immunohistochemistry and preproenkephalin (PPE) or pre- radiolabeled riboprobes. Preproenkephalin and homer-1a prodynorphin (PPDyn) in situ hybridization. cDNAs were linearized after Apa I digestion and transcribed with SP6 RNA polymerase. The preprodynorphin and Arc Drug Treatments antisense probes were linearized after EcoR I digestion and All the drugs were purchased from Sigma. Four to transcribed with T7 RNA polymerase. The nur77 (NGFI-B) 5 weeks after lesion, rats were treated with chronic (twice daily antisense probe was linearized with BamH I and transcribed for 10 days) IP injection of physiological saline (lesioned-only with T3 (42). Transcription reaction contained 6 mL a[35S]- animals, n = 3) or L-Dopa (L-3,4-Dihydroxyphenylalanine UTP (20 mCi/mL; Amersham Biosciences, Orsay, France), methyl ester hydrochloride) (50 mg/kg) combined with 250 mM adenosine triphosphate (ATP), cytidine triphosphate benserazide hydrochloride (10 mg/kg) (L-Dopa-treated ani- (CTP), and guanosine triphosphate (GTP), and unlabeled mals, n = 12). Benserazide was given to prevent decarbox- uridine triphosphate (UTP) (10.5 mM); the reaction was ylation of L-Dopa in the periphery, as is common in clinical incubated at 37°C for 2 hours. After DNase I digestion for q 2005 American Association of Neuropathologists, Inc. 937 Sgambato-Faure et al J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005

15 minutes, the labeled RNA was purified on microspin G50 tories; ABCYS, Paris, France). The sections were then rinsed columns (Amersham Biosciences). Free-floating sections were 2 times in TBS and 2 times in TB (0.25M Tris, pH 7.5) for mounted on SuperFrost Plus slides in RNase-free conditions. 10 minutes each, placed in a solution of TB containing 0.1% Once the sections were dried, in situ hybridization was per- 3,3#-diaminobenzidine (DAB; 50 mg/100 mL), and developed formed at 60°C in humid chambers for 16 hours according to by H2O2 addition (0.02%). the previous protocol (43). After coverslips were removed in 4 3 SSC and 10 mM DTT, the slides were washed in the same Double Labeling Combining solution for 1 hour at room temperature and then in 50% Immunohistochemistry and formamide, 10 mM Tris-HCl, pH 8, 75 mM NaCl, and 2.5 mM In Situ Hybridization EDTA. Sections were treated with RNase A (20 mg/mL; Free-floating sections were processed for FosB or Arc Sigma) in 400 mM NaCl, 10 mM Tris-HCl, pH 7.5, and immunohistochemistry as described previously, except that all

50 mM EDTA for 30 minutes at 37°C and then were rinsed for buffers used were autoclaved. Sections were mounted on Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021 15 minutes at 60°Cin23 SSC followed by 0.1 3 SSC. After SuperFrost Plus slides in RNase-free conditions and air-dried dehydration, the sections were air-dried and exposed with for at least 60 minutes followed by the indicated in situ Biomax-MR films (Kodak). hybridization. The slides were then coated with LM-1 emulsion (Amersham Biosciences) and exposed. After development, Immunohistochemistry slides were mounted with Eukitt. Free-floating sections were rinsed in Tris-buffered saline (TBS; 0.25M Tris and 0.25M NaCl, pH 7.5), incubated Double In Situ Hybridization 5 minutes in TBS containing 3% H2O2 and 10% methanol, and The double in situ hybridization procedure was thenrinsed3timesfor10minuteseachinTBS.After performed as previously described (42). Briefly, the proportion 15-minute incubation in 0.2% triton X-100 in TBS, the sections of nur77 colocalization with PPE or PPDyn mRNA was were rinsed 3 times in TBS. These were incubated with the evaluated using double in situ hybridization with a [35S]UTP- primary antibody (anti-FosB 1/400 rabbit polyclonal H-75 labeled nur77 probe and a nonradioactive digoxigenin (Dig)- [SC-7203] from Tebu-Bio, Le Parray en Yvelines, France), labeled PPE or PPDyn riboprobe. PPE and PPDyn riboprobes anti-TH 1/2000 rabbit polyclonal from Jacques Boy (Reims, were labeled with using the Riboprobe System of Promega France), or anti-Arc 1/300 mouse monoclonal from BD (Madison, WI) with the Dig-RNA labeling mix (Roche Biosciences (Pont de Claix, France) for 72 hours at 4°C. After Diagnostics, Laval, Canada). Double in situ hybridization was 3 rinses in TBS, the sections were then incubated 2 hours at performed in the same conditions as for single in situ pro- 4°C with the corresponding secondary biotinylated antibody cedure. The Dig-cRNA probe (approximately 50 ng) was in TBS. After being washed, the sections were incubated simply added in the same hybridization solution with the 90 minutes at room temperature in avidin-biotin-peroxidase radioactive (2 3 106 cpm) cRNA probe for nur77. An complex (ABC) solution (final dilution, 1/50; Vector Labora- additional step using a 50% formamide solution in 2 3 SSC

FIGURE 1. Characterization of the unilateral 6-hydroxydopamine le- sion. (A) Representative photomi- crographs of sections showing TH immunoreactivity after unilateral 6-OHDA injection (n = 30) (the lesioned side is indicated with an asterisk) in the substantia nigra and ventral tegmental areas (left panels) and striatum (right panels). (B) Quantification of the in situ hybrid- ization signals of preproenkephalin (PPE) and preprodynorphin (PPDyn) mRNA probes in the ipsilateral (ipsi) and contralateral (contra) striatal sides after unilateral 6-OHDA injec- tion. ***, p , 0.001 or **, p , 0.01 when compared with the ipsilateral striatum of unlesioned rats. 6-OHDA, 6-hydroxydopamine; acb, accumbens nucleus; cc, corpus callosum; CPu, caudate-putamen; lv, lateral ventri- cle; PPDyn, preprodynorphin; PPE, preproenkephalin; SNc, substantia nigra pars compacta; SNr, substantia nigra pars reticulata; TH, tyrosine hydrox- ylase; VTA, ventral tegmental area.

938 q 2005 American Association of Neuropathologists, Inc. J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Arc Correlates With L-Dopa-Induced Motor Behavior Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021

FIGURE 2. Effect of chronic L-Dopa on locomotor sensitization and dys- kinesia. 6-OHDA rats were chroni- cally treated with L-Dopa (50 mg/kg; n = 12) for 10 days, during which locomotor sensitization ([A]; aver- age standard error of the mean comprises between 0, 9 and 3, 1) as well as the different subtypes of dyskinesia (B) were analyzed. Note that all rats (groups 1 [n = 7] and 2 [n = 5]) develop forelimb, orolin- gual, and axial dyskinesia but only rats from group 1 develop locomo- tor sensitization. dysk, dyskinesia. buffer after hybridization was performed to reduce nonspecific Data Analysis Dig labeling. Detection of Dig-labeled RNA was performed Tissue sections were examined under light microscope. with an anti-Dig antibody conjugated to alkaline phosphatase TH, FosB, and Arc-positive neurons were digitalized at (Boehringer Mannheim, Laval, Canada) visualized using various magnifications with a computerized image analyzer a nitroblue tetrazolium chloride, 5-bromo-4-chloro-3-indolyl (Analysis; Soft Imaging System, Munster, Germany). Cell phosphate, and levamisole chromogen solution. Slides were counts were performed in the lateral part of the striatum then dipped in LM-1 photographic emulsion (Amersham, ipsilateral to the SNc lesion (3 fields per section). Cells labeled Oakville, Ontario, Canada) melted at 42°C, air-dried, and with PPE, PPDyn, and Arc probes were visualized by the stored in the dark for 12 days at 4°C. The emulsion was accumulation of silver grains over the soma. Densitometric developed in D-19 developer and fixed (Kodak, New Haven, measurements of mRNA signals were obtained from auto- CT). Slides were coverslipped using a water-soluble mounting radiograms using the NIH Image J 1.29 software (Wayne medium (Permafluor; Lipshaw Immunon, Pittsburgh, PA). Rasband, NIMH) after subtraction of background value. They Single- or double-labeled cells were visualized and manually were performed in the lateral part of the striatum on 4 different counted under bright-field illumination with a Zeiss photo- sections per rat. Histogram shows mean 6 standard error of microscope at a magnification of 4003. Cell counts were mean. Statistical analyses were assessed by analysis of performed in the lateral part of the striatum on 4 different variance with multiple comparisons (Newman-Keuls multiple sections (3 fields per section). comparison test). q 2005 American Association of Neuropathologists, Inc. 939 Sgambato-Faure et al J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005

animals. An analysis of the densitometric measurements of TH immunostaining in both SNc and ventral tegmental area (VTA) did not produce a statistical difference between the 2 groups (data not shown). At the striatal level, mRNA levels encoding PPE (p , 0.001) and PPDyn (p , 0.01) were respectively induced or repressed by the lesion (Fig. 1B). Evaluation of L-Dopa-Induced Motor Behavior The motor behavior of hemiparkinsonian rats was observed and evaluated as described in the ‘‘Materials and Methods’’ section. Although rats treated chronically with the

vehicle did not show any abnormal motor activity (data not Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021 shown), rats treated with high doses of L-Dopa could be subdivided into 2 groups according to their rotational behavior (Fig. 2A). The first group (group 1, n = 7) developed severe locomotor sensitization over time (linear regression coefficient R2 = 0.9195, p , 0.001), whereas the second group (group 2, n = 5) displayed almost no locomotor sensitization (R2 = 0.5363, p = 0.057) despite the fact that they received the same L-dopa regimen and displayed the same levels of denervation. The 3 subtypes of dyskinesia (i.e. forelimb, orolingual, and axial) were also evaluated (Fig. 2B). For each, a gradual increase of the scores was observed over time (Fig. 2B), but no further difference was noted between the first and second groups (compare left and right columns for each dyskinesia subtype). The dispersion of the results reflects the variability in the severity of the dyskinesia displayed by each animal. No dyskinesia was observed for vehicle-treated hemiparkinsonian rats (data not shown). Effect of Chronic L-Dopa Treatment on FosB Expression in the Depleted Striatum Figure 3 shows the effect of chronic L-Dopa treatment on the expression of FosB-like proteins after use of an antibody directed against the N-terminal part of FosB and therefore able to recognize both FosB and its chronic isoforms such as DFosB (44). As expected, FosB-positive nuclei were detected in the striatum ipsilateral to the lesion in response to FIGURE 3. FosB-like proteins are induced in the denervated chronic L-Dopa administration but not in response to chronic striatum by chronic L-Dopa. (A–C) Representative photo- vehicle (compare Fig. 3B and C to A). Although rats from micrographs of brain sections showing FosB/DFosB immuno- group 1 displayed a widespread expression of FosB-like proteins reactivity after unilateral 6-OHDA injection and chronic in both the lateral and the medial parts of the striatum (Fig. treatment with vehicle (n = 3) (A-A$) or L-Dopa (50 mg/kg) 3B#-B$), rats from group 2 showed an expression restricted to (n = 12) (B-B$ and C-C$). Note that FosB-like proteins are the lateral part of the striatum (Fig. 3C#-C$). These results induced by L-Dopa (B-B$ and C-C$) and not by the vehicle concur with previous data showing that the extent of FosB $ (A-A ). Note also the lack of staining in the medial part of expression varies according to the subtype of motor behavior the striatum for group 2. (D) Quantification of the number of developed (40). Of note, the pattern of induction was not FosB-positive cells in the lateral part of the denervated stria- related to the pattern of TH depletion because all animals tum 2 or 24 hours after the last injection. ***, p , 0.001 when compared with the ipsilateral striatum from unlesioned rats. displayed a complete loss of TH in both lateral and medial parts of the striatum. FosB-like proteins induction in the RESULTS lesioned striatum is similar at 2 and 24 hours after L-dopa administration (Fig. 3D) for all dyskinetic animals, suggesting Characterization of the Unilateral that persistent forms of FosB-like proteins have been induced. 6-hydroxydopamine Lesion The dopaminergic lesion was evaluated postmortem by Effect of Chronic L-Dopa Treatment on PPDyn, histochemical analysis (Fig. 1). A complete loss of TH im- Arc, and homer-1a mRNA Expression in the munostaining was observed ipsilaterally in both the SNc and Depleted Striatum the striatum of all animals used in the present study (Fig. 1A). Figure 4 shows the effect of chronic L-Dopa treatment The levels of denervation were the same in all groups of on the striatal levels of mRNA encoding for PPDyn, as well as

940 q 2005 American Association of Neuropathologists, Inc. J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Arc Correlates With L-Dopa-Induced Motor Behavior Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021

FIGURE 4. Induction of PPDyn, Arc, and homer-1a mRNA in the lesioned striatum by chronic L-Dopa. (A–K) Representative autoradiograms from adjacent brain sections of 6-OHDA-lesioned rats treated chronically with vehicle (n = 3) (A, E, I) or L-Dopa (n = 12) (50 mg/kg) (B, F, J, C, G, K) showing in situ hybridization of PPDyn (A–C), Arc (E, F, G), and homer-1a (I–K) mRNA. Note that inductions of Arc and homer-1a correlate spatially with PPDyn. Groups 1 and 2 were defined according to their locomotor behavior. Group 1 displayed a strong sensitization of their turning behaviors during L-Dopa treatment, whereas group 2 developed almost no sensitization (Fig. 1). Patterns of PPDyn, Arc, and Homer-1a gene induction also differ between group 1 and 2. Quantification of PPDyn (D), Arc (H), and homer-1a (L) mRNA levels in the lesioned striatum. ***, p , 0.001 when compared with their respective intact side.

2 other markers of synaptic plasticity, namely Arc (34, 35) and expressed in PPDyn mRNA-containing neurons (Fig. 6B2). homer-1a (45–47) on adjacent brain sections. Two hours after The quantification of this result was addressed by combining the last L-Dopa injection (Fig. 4), PPDyn mRNAwere strongly the immunohistochemistry of FosB with the in situ hybrid- upregulated (p , 0.001) in response to L-Dopa compared with ization of PPE, PPDyn, or Arc mRNA (Fig. 5). Indeed, after vehicle (Fig. 4A–D), the extent of its mRNA induction cor- unilateral medial forebrain bundle lesion and chronic L-Dopa relating with the occurrence or absence of locomotor sensitization treatment, FosB is expressed by PPDyn-positive neurons (40). as for FosB-like proteins. Strikingly, both Arc (Fig. 4E–H; p , We addressed this point in our model by using striatal sections 0.001) and homer-1a (Fig. 4I–L; p , 0.001) mRNA were from both groups of rats (1 and 2) and found that more than induced in response to L-Dopa in the denervated striatum of 80% of FosB-positive cells colocalized with PPDyn-positive dyskinetic rats. Their expressions were also spatially correlated neurons (Fig. 5A, D) compared with 20% colocalizing with with that of PPDyn and FosB. Whereas PPDyn and Arc mRNA PPE-positive neurons (Fig. 5B, D). We also observed that 65% remained upregulated 24 hours after the last L-Dopa injection, of FosB-positive cells expressing Arc mRNA (Fig. 5C, D) homer-1a mRNA returned to basal levels (data not shown). showed a preferential Arc expression in PPDyn-positive neurons. The occurrence of locomotor sensitization did not alter Identification of the Striatal Cell Type in colocalization, indicating that dyskinesia and locomotor sen- Which Arc Is Upregulated sitization are associated with distinct spatial expression of The combination of Arc immunohistochemistry with those markers in the striatum in a majority of cells expressing PPDyn in situ hybridization revealed that Arc protein was PPDyn in the areas examined. q 2005 American Association of Neuropathologists, Inc. 941 Sgambato-Faure et al J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021

FIGURE 5. FosB-positive cells express PPDyn and Arc mRNA. (A–C) Repre- sentative photomicrographs of stria- tal sections (from L-Dopa-treated [n = 12] [50 mg/kg] rats) that were processed for FosB immunohisto- chemistry and in situ hybridization of PPDyn (A), PPE (B), or Arc (C) mRNA. Note that FosB-positive cells express PPDyn and Arc, but not PPE mRNA. Arrowheads point to double- labeled cells: FosB- and PPDyn- positive cells in panel (A); FosB- and Arc-positive cells in panel (C). Arrows point to single-labeled cells (PPE-positive cells in panel [B]). (D) Quantification of FosB-positive cells expressing PPDyn, PPE, and Arc in the lesioned striatum in response to the chronic L-Dopa treatment. Effect of Lower Doses of Chronic L-Dopa on we administered k opioid antagonists (naloxone [49] and Striatal Gene Regulation nor-binaltorphimine [50]) or agonist (U-50,488H [51]) in Having shown that the Arc gene (and the protein, data not denervated-rats treated with L-Dopa. At the behavioral level, shown) is upregulated in the denervated striatum in response to opioid antagonist or agonist administration did not alter the high doses of L-Dopa, we further analyzed if its induction could development of dyskinesia (Fig. 7A) as well as locomotor still be evoked in responses to more therapeutic doses of L-Dopa sensitization (data not shown). Similarly, at the histochemical treatment (10 mg/kg) (Fig. 6). This point was addressed by level, PPDyn (data not shown) and Arc (Fig. 7B) mRNA immunohistochemistry with an antibody against the Arc protein. upregulation in the lesioned striatum remained the same in the As shown in Figure 6A1, the Arc protein was strongly induced presence of opioid antagonists. Gene regulation was still in the lateral part of the striatum, and the extent of its expression spatially correlated to the L-Dopa-driven motor behavior, that was again spatially coincident with that of PPDyn mRNA (Fig. is, either restricted to the lateral part of the striatum or in 6B1) detected on the adjacent brain section from a dyskinetic rat the whole striatum according to the absence or occurrence (2 of 3 animals treated with low doses of L-Dopa displayed (Fig. 7B) of locomotor sensitization, respectively. Similarly, dyskinetic movements without locomotor sensitization, whereas FosB and Arc proteins were still induced (data not shown). the third rat displayed no motor behavior). Interestingly, nur77 These data suggest that in our model, modulation of k opioid mRNA (48) was also strongly upregulated (Fig. 6C1, C2; p , receptors does not alter the mechanisms underlying gene in- 0.001) in the same striatal territory (compare Figure 6C1 duction or those involved in the development of LID and with A1 and B1). Of note, although 95% of PPDyn-positive locomotor sensitization. cells also express nur77 (Fig. 6C3), only 35% of PPE-containing neurons express nur77 (Fig. 6C4). Similar results were ob- tained regardless of when the animals were killed. DISCUSSION The main goal of the present study using a rat model of Effect of Coadministration of Naloxone, U-50, LID was to investigate the effect of chronic L-Dopa treatment 488H, or Nor-Binaltorphimine With L-Dopa on the regulation of Arc mRNA and protein in the dopamine- on Motor Behavior and denervated striatum and on its coordinate expression with To test the involvement of endogenous opioid dynorphin striatal neuropeptides belonging to the striatonigral and in the development of dyskinesia and behavioral sensitization, striatopallidal output pathways. The main finding of our study

942 q 2005 American Association of Neuropathologists, Inc. J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Arc Correlates With L-Dopa-Induced Motor Behavior Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021

FIGURE 6. Induction of Arc protein and nur77 mRNA in the lesioned striatum in response to a chronic treatment with low doses of L-Dopa. Representative photomicrographs of brain sections (from 6-OHDA rats treated with low doses of L-Dopa [n = 3] [10 mg/kg]) were processed for Arc immunohistochemistry (A1–A3, B2) and in situ hybridization of PPDyn (B1, B2) or double in situ hybridization combining nur77 (C1, C3, C4) with PPDyn (C3) or PPE (C4) mRNA. Note that Arc and nur77 upregulations are coincident to that of PPDyn (A1, B1, C1). Note also that Arc and PPDyn mRNA are expressed in the same striatal cells (B2). (C2) Quantification of nur77 mRNA induction in the lateral part of the lesioned striatum. ***, p , 0.001 when compared with the ipsilateral striatum of unlesioned rats. Note that nur77 is colocalized in dynorphin-expressing cells and not enkephalin-expressing neurons. was that intermittent and chronic L-Dopa led to strong CaM kinase II-dependent neurite extension (58). The short upregulation of Arc mRNA, which is spatially coincident with half-life of Arc mRNA contrasts with its persistent striatal that of PPDyn, homer-1a, and nur77 mRNA as well as FosB- induction and indicates that Arc upregulation is unlikely to like proteins, their striatal territories of expression being result from a direct action of L-Dopa but rather a consequence correlated with the motor behavior developed (i.e. dyskinesia of a L-Dopa-driven long-term adaptation of the structural and accompanied or not with sensitization of their turning molecular network of the striatum. This hypothesis is further behaviors). Moreover, these biochemical alterations of Arc supported by the induction of other structural- and synaptic- levels, along with homer-1a and nur77, were preferentially related genes (Sgambato-Faure et al, unpublished data; and observed in striatonigral neurons of the direct striatal output [54]). Finally, it has been shown that Arc mRNA induction pathway that express PPDyn mRNA, the precursor of the depends on NMDA glutamatergic receptors (59, 60) and is re- dynorphin peptide. quired for long-term potentiation as well as memory process Arc mRNA is induced in the striatum by acute cocaine in the hippocampus (36–38). It is therefore of particular in- (52, 53) and this effect is inhibited by a D1 antagonist, terest to confront our data with other studies in the striatum reserpine treatment, or 6-hydroxydopamine lesion (52). In the showing persistent increases of extracellular glutamate levels current study, we found that Arc mRNA was upregulated in the (61) and lack of depotentiation after induction of long-term striatum after chronic L-Dopa administration, validating potentiation (21) in dyskinetic rats. previous data obtained by a microarray study (54). The Homer proteins have gained particular attention because induction of Arc after a chronic L-Dopa treatment contrasts they are part of postsynaptic densities and are involved in with the fact that most IEG show reduced induction after both the localization and function of type I metabotropic chronic exposure to a drug. However, Arc mRNA is also glutamatergic receptors (45–47) as well as in the coupling of upregulated after chronic cocaine in the frontal cortex (55) and NMDA-binding ionotropic glutamate receptors to metabo- chronic morphine in the striatum (56). The exact function of tropic glutamatergic receptors (62). The dimerization of long Arc is unknown, but its induction and subcellular localization homer proteins can serve to link mGluR receptors to inositol (35) led to the hypothesis of its involvement in cytoskeletal triphosphate receptors and thus link extracellular signals to the rearrangements during the process of synaptic plasticity. release of calcium from intracellular stores. The shorter Highly enriched in neuronal dendrites, where it localizes in homer-1a protein can be induced in the brain in response to a distribution that parallels that of F-actin (33), Arc interacts various stimuli, including long-term potentiation, electrocon- with cytoskeletal proteins such as MAP-2 (57) and promotes vulsive shocks, and drugs of abuse (63–65). This truncated q 2005 American Association of Neuropathologists, Inc. 943 Sgambato-Faure et al J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021

FIGURE 7. Modulation of opioid transmission does not impair chronic L-Dopa-driven motor behavior or Arc mRNA induction. (A) 6-OHDA- lesioned rats were treated chronically for 10 days with L-Dopa (50 mg/kg) combined with vehicle, naloxone (10 mg/kg), U-50,488H (1 mg/kg), or with an acute injection of nor- binaltorphimine (20 mL/kg) on the last day of the treatment. Each day, forelimb, orolingual, and axial dyski- nesia was evaluated using the rating scale. Note that there is an increase of the scores over time for the 4 exper- imental groups. (B) Quantification of Arc mRNA induction, detected by in situ hybridization, in the lesioned striatum from chronically L-Dopa- treated rats (50 mg/kg) combined with vehicle, naloxone (10 mg/kg), U-50,488H (1 mg/kg) or nor-binaltor- phimine (20 mg/kg). Note that Arc remains upregulated in the lesioned striatum whatever the treatment used. Note also that the extension of gene expression correlates with L-Dopa- driven motor behavior. isoform may serve to competitively interfere with the normal of dopaminergic neurotransmission does not desensitize after linkage between homer-binding proteins (66–69). Given the chronic haloperidol (42) or L-Dopa (48) treatment. These connection between dopaminergic receptors and calcium (70), findings strongly suggest that nur77 along with Arc and our data showing upregulation of homer-1a in response to homer-1a may contribute to the dopamine-driven behavioral chronic L-Dopa further supports a link between intracellular sensitization. Interestingly, nur77 is also modulated in trans- calcium signaling and dyskinesia. However, the more transient genic mice overexpressing presenilin-1 that display memory character of its induction compared with Arc suggests that consolidation deficits (71). Considering that LID may be distinct mechanisms are involved to underlie the transient assimilated to a pathologic form of learning (21), it is tempting expression of dyskinesia or its long-term maintenance through to speculate that this set of signaling-related genes may par- a possible synaptic remodeling process. ticipate in the cell that generate an abnormal Similar to Arc, we found that nur77 was upregulated activity of the striatonigral pathway leading to abnormal preferentially in selective dynorphin overexpressing striatal involuntary movements and behavioral sensitization. areas. This gene encodes an orphan nuclear receptor closely Intermittent treatment with L-Dopa markedly elevated related to members of the steroid/thyroid hormone receptor PPDyn mRNA levels in the dopamine-denervated striatum. family but also acts as an IEG. It is induced in the striatum in Dynorphin, the endogenous agonist for the k opioid receptor response to dopamine denervation (48, 64) and chronic (72), is enriched in striatonigral neurons (73, 74). An increased haloperidol (42). Like Arc, nur77 upregulation after alteration stimulation of k receptors distributed on glutamatergic terminals

944 q 2005 American Association of Neuropathologists, Inc. J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Arc Correlates With L-Dopa-Induced Motor Behavior

(75) and striatonigral neurons (76) may therefore contribute to REFERENCES the sensitization of dopamine agonist-induced behavioral 1. Ehringer H, Hornykiewicz O. Distribution of noradrenaline and dopamine response. It has been shown that the blockade of opioid (3-hydroxytyramine) in the human brain and their behavior in diseases of transmission allows attenuation of LID in rats (25, 26). the extrapyramidal system. Klin Wochenschr 1960;38:1236–39 2. Agid Y. Parkinson’s disease: Some physiopathological aspects and clinical However, a more recent study indicated that opioid antagonist practice. Rev Prat 1989;39:641–46 treatment increased LID in a nonhuman primate model of PD 3. Obeso JA, Rodriguez-Oroz MC, Rodriguez M, et al. The basal ganglia (77). We found that blockade of opioid receptors did not alter and disorders of movement: Pathophysiological mechanisms. News the development of dyskinesia or the gene modifications that Physiol Sci 2002;17:51–55 are associated with it in our rat LID model. This might be the 4. Obeso JA, Rodriguez-Oroz M, Marin C, et al. The origin of motor fluctuations in Parkinson’s disease: Importance of dopaminergic inner- result of the downregulation of kappa opioid-binding sites in vation and basal ganglia circuits. Neurology 2004;62:S17–30 the striatum and substantia nigra after chronic L-Dopa (78). 5. Johnston TH, Brotchie JM. Drugs in development for Parkinson’s disease. Curr Opin Investig Drugs 2004;5:720–26 Moreover, upregulation of endogenous dynorphin may trigger Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021 desensitization and downregulation of opioid receptors, be- 6. Birkmayer W, Hornykiewicz O. Der L-Dioxyphenylalanin (4DOPA)- Effekt bei der Parkinson-Akinese. Wien Klin Wschr 1961;73:787–88 cause it is known that opioid receptors desensitize rapidly after 7. Barbeau A. L-Dopa therapy in Parkinsonvs disease: A critical review of opioid administration (79). nine years experience. Can Med Assoc J 1969;101:791–800 Another striking observation made in this study (besides 8. Krack P,Pollak P,Limousin P,et al. From off-period dystonia to peak-dose the perfect topographic superimposition between striatal chorea: The clinical spectrum of varying subthalamic nucleus activity. regions of gene upregulations) is the spatial correlation of Brain 1999;122:1133–46 9. Nutt JG. Motor fluctuations and dyskinesia in Parkinson’s disease. gene expression with dyskinesia accompanied (whole stria- Parkinsonism Relat Disord 2001;8:101–8 tum) or not (lateral part of the striatum) with locomotor 10. Rascol O, Fabre N. Dyskinesia: L-Dopa-induced and tardive dyskinesia. sensitization. This particular pattern of expression have been Clin Neuropharmacol 2001;24:313–23 observed in other studies (40) and is not the result of an 11. Defebvre L. Motor complications in dopa treatment of Parkinson disease: incomplete denervation of the striatum because we did not Clinical description and evaluation. The´rapie 2004;59:93–6 12. Di Monte DA, McCormack A, Petzinger G, et al. Relationship among report any significant difference in TH immunoreactivity nigrostriatal denervation, parkinsonism, and dyskinesias in the MPTP levels between these experimental groups. Furthermore, the primate model. Mov Disord 2000;15:459–66 absence of locomotor sensitization was not the result of the 13. Baas H, Fuchs G, Gemende I, et al. Which factors influence therapeutic fact that rats were tested in their home cages, because placing decisions in Parkinson’s disease? J Neurol 2002;249:49–52 14. Winkler C, Kirik D, Bjorklund A, et al. L-DOPA-induced dyskinesia in them in a cylinder did not increase the number of rotations, the intrastriatal 6-hydroxydopamine model of ParkinsonÕs disease: Rela- whatever the group involved. Besides dopaminergic input (80, tion to motor and cellular parameters of nigrostriatal function. Neurobiol 81), the striatum receives major glutamatergic afferents from Dis 2002;10:165–86 the cerebral cortex and the thalamus (82–85). The cortical and 15. Paille V, Brachet P, Damier P. Role of nigral lesion in the genesis of dys- thalamic afferents are topographically organized along its kinesias in a rat model of Parkinson’s disease. Neuroreport 2004;15:561–64 16. Jenner P. Avoidance of dyskinesia: Preclinical evidence for continuous rostrocaudal axis and mainly target the lateral portion of the dopaminergic stimulation. Neurology 2004;62:S47–55 striatum (86–88). Therefore, the lateral and medial parts of the 17. Calon F, Tahar AH, Blanchet PJ, et al. Dopamine-receptor stimulation: striatum differ in terms of afferent inputs. The similarity Biobehavioral and biochemical consequences. Trends Neurosci 2000;23: between the striatal territory targeted by cortical and thalamic S92–S100 afferents and the selective spatial induction of gene expression 18. Graybiel AM, Canales JJ, Capper-Loup C. Levodopa-induced dyskinesias and dopamine-dependent stereotypies: A new hypothesis. Trends in the group of dyskinetic animals that did not show behavioral Neurosci 2000;23:S71–77 sensitization suggests that the development of sensitization 19. Bevan MD, Magill PJ, Terman D, et al. Move to the rhythm: Oscillations may be associated with corticothalamostriatal individual in the subthalamic nucleus–external globus pallidus network. Trends vulnerability. Neurosci 2002;25:525–31 Together, these findings demonstrate that Arc along 20. Hutchison WD, Dostrovsky JO, Walters JR, et al. Neuronal oscillations in the basal ganglia and movement disorders: Evidence from whole animal with homer-1a and nur77 is upregulated in the dynorphin- and human recordings. J Neurosci 2004;24:9240–43 containing striatonigral neurons from dyskinetic rats support 21. Picconi B, Centonze D, Hakansson K, et al. Loss of bidirectional striatal a role for cytoskeletal remodeling in the regulation of L-Dopa- synaptic plasticity in L-DOPA-induced dyskinesia. Nat Neurosci 2003;6: induced long-term motor alterations. Further studies are required 501–6 to elucidate if these molecular changes are responsible for or 22. Chase TN. Levodopa therapy: Consequences of the nonphysiologic replacement of dopamine. Neurology 1998;50:S17–25 protective against LID. 23. Chase TN. Striatal plasticity and extrapyramidal motor dysfunction. Parkinsonism Relat Disord 2004;10:305–13 24. Matsumoto RR, Brinsfield KH, Patrick RL, et al. Rotational behavior mediated by dopaminergic and nondopaminergic mechanisms after intranigral microinjection of specific mu, delta and kappa opioid agonists. J Pharmacol Exp Ther 1988;246:196–203 25. Carey RJ. Naloxone reverses L-Dopa induced overstimulation effects in ACKNOWLEDGMENTS a Parkinson’s disease animal model analogue. Life Sci 1991;48:1303–8 The authors thank Drs. L. Kerkerian-Le Goff and P. 26. Henry B, Brotchie JM. Potential of opioid antagonists in the treatment of Salin for critical reading of the manuscript; Drs. R. Paletzki levodopa-induced dyskinesias in Parkinson’sdisease. Drugs Aging 1996;9: 149–58 for generously providing Arc, PPE, and homer-1a cDNAs; and 27. Marin C, Engber TM, Chaudhuri P, et al. Effects of kappa receptor Drs. S. Watson and H. Akil for the gift of PPDyn cDNA. The agonists on D1 and D2 dopamine agonist and antagonist-induced be- authors also thank M. El Atifi for her help with Excel graphs. haviors. Psychopharmacology 1996;123:215–21 q 2005 American Association of Neuropathologists, Inc. 945 Sgambato-Faure et al J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005

28. Newman DD, Rajakumar N, Flumerfelt BA, et al. A kappa opioid 53. Tan A, Moratalla R, Lyford GL, et al. The activity-regulated cytoskeletal- antagonist blocks sensitization in a rodent model of Parkinson’s disease. associated protein Arc is expressed in different striosome-matrix patterns Neuroreport 1997;8:669–72 following exposure to amphetamine and cocaine. J Neurochem 2000;74: 29. Henry B, Fox SH, Crossman AR, et al. Mu- and delta-opioid receptor 2074–78 antagonists reduce levodopa-induced dyskinesia in the MPTP-lesioned 54. Konradi C, Westin JE, Carta M, et al. Transcriptome analysis in a rat primate model of Parkinson’s disease. Exp Neurol 2001;171:139–46 model of L-DOPA-induced dyskinesia. Neurobiol Dis 2004;17:219–36 30. Calabresi P, Giacomini P, Centonze D, et al. Levodopa-induced dyskinesia: 55. Freeman WM, Brebner K, Lynch WJ, et al. Changes in rat frontal cortex A pathological form of striatal synaptic plasticity? Ann Neurol 2000;47:60–68 gene expression following chronic cocaine. Brain Res Mol Brain Res 31. Chase TN, Oh JD. Striatal dopamine- and glutamate-mediated dysregu- 2002;104:11–20 lation in experimental parkinsonism. Trends Neurosci 2000;23:86–91 56. Marie-Claire C, Courtin C, Roques BP, et al. Cytoskeletal genes regu- 32. Link W, Konietzko U, Kauselmann G et al. Somatodendritic expression of lation by chronic morphine treatment in rat striatum. Neuropsychophar- an immediate early gene is regulated by synaptic activity. Proc Natl Acad macology 2004;29:2208–15 Sci U S A 1995;92:5734–38 57. Fujimoto T, Tanaka H, Kumamaru E, et al. Arc interacts with 33. Lyford GL, Yamagata K, Kaufmann WE, et al. Arc, a growth factor and microtubules/microtubule-associated protein 2 and attenuates microtu- activity-regulated gene, encodes a novel cytoskeleton-associated protein bule-associated protein 2 immunoreactivity in the dendrites. J Neurosci Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021 that is enriched in neuronal dendrites. 1995;14:433–45 Res 2004;76:51–63 34. Steward O, Wallace CS, Lyford GL, et al. Synaptic activation causes the 58. Donai H, Sugiura H, Ara D, et al. Interaction of Arc with CaM kinase II mRNA for the IEG Arc to localize selectively near activated postsynaptic and stimulation of neurite extension by Arc in neuroblastoma cells sites on dendrites. Neuron 1998;21:741–51 expressing CaM kinase II. Neurosci Res 2003;47:399–408 35. Steward O, Worley P. Local synthesis of proteins at synaptic sites on 59. Steward O, Worley PF. Selective targeting of newly synthesized Arc dendrites: Role in synaptic plasticity and ? mRNA to active synapses requires NMDA receptor activation. Neuron Neurobiol Learn Mem 2002;78:508–27 2001;30:227–40 36. Guzowski JF, Lyford GL, Stevenson GD, et al. Inhibition of activity- 60. Martin KC. Arc mRNA dynamics: Return to sender—the NMDA receptor dependent Arc protein expression in the rat hippocampus impairs the provides the targeting address for Arc mRNA. Trends Neurosci 2001;24: maintenance of long-term potentiation and the consolidation of long-term 621–23 memory. J Neurosci 2000;20:3993–4001 61. Robelet S, Melon C, Guillet B, et al. Chronic L-DOPA treatment increases 37. Plath N, Ohana O, Dammermann B, et al. Aberrant LTP in arg3.1/ARC extracellular glutamate levels and GLT1 expression in the basal ganglia in knockout animals. Soc Neurosci Abstr 2001;27:1598 a rat model of Parkinson’s disease. Eur J Neurosci 2004;20:1255–66 38. Ying SW,Futter M, Rosenblum K, et al. Brain-derived neurotrophic factor 62. Sheng M, Kim E. The Shank family of scaffold proteins. J Cell Sci induces long-term potentiation in intact adult hippocampus: Requirement 2000;113:1851–56 for ERK activation coupled to CREB and up-regulation of Arc synthesis. 63. Brakeman PR, Lanahan AA, O’Brian R, et al. Homer: A protein that J Neurosci 2002;22:1532–40 selectively binds to metabotropic glutamate receptors. Nature 1997;386: 39. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates.New 284–88 York: Academic Press, 1986 64. Berke JD, Paletzki RF, Aronson GJ, et al. A complex program of stria- 40. Andersson M, Hilbertson A, Cenci MA. Striatal fosB expression is tal gene expression induced by dopaminergic stimulation. J Neurosci causally linked with l-DOPA-induced abnormal involuntary movements 1998;18:5301–10 and the associated upregulation of striatal prodynorphin mRNA in a rat 65. Bottai D, Guzowski JF, Schwarz MK, et al. Synaptic activity-induced model of Parkinson’s disease. Neurobiol Dis 1999;6:461–74 conversation of intronic to exonic sequence in Homer 1 immediate early 41. Cenci MA, Lee CS, Bjorklund A. L-DOPA-induced dyskinesia in the rat gene expression. J Neurosci 2002;22:167–75 is associated with striatal overexpression of prodynorphin- and glutamic 66. Xiao B, Tu JC, Petralia RS, et al. Homer regulates the association of group acid decarboxylase mRNA. Eur J Neurosci 1998;10:2694–2706 1 metabotropic glutamate receptors with multivalent complexes of homer- 42. Beaudry G, Langlois MC, Weppe I, et al. Contrasting patterns and cellular related, synaptic proteins. Neuron 1998;21:707–16 specificity of transcriptional regulation of the nuclear receptor nerve 67. Tu JC, Xiao B, Yuan JP, et al. Homer binds a novel proline-rich motif and growth factor-inducible B by haloperidol and clozapine in the rat links group 1 metabotropic glutamate receptors with IP3 receptors. forebrain. J Neurochem 2000;75:1694–1702 Neuron 1998;21:717–26 43. Sgambato V, Pages C, Rogard M, et al. Extracellular signal-regulated 68. Ango F, Pre´zeau L, Muller T, et al. Agonist-independent activation of kinase (ERK) controls immediate early gene induction on corticostriatal metabotropic glutamate receptors by the intracellular protein Homer. stimulation. J Neurosci 1998;18:8814–25 Nature 2001;411:962–65 44. Nestler EJ, Kelz MB, Chen J. DFosB: A molecular mediator of long-term 69. Ango F, Robbe D, Tu JC, et al. Homer-dependent cell surface expression neural and behavioral plasticity. Brain Res 1999;835:10–17 of metabotropic glutamate receptor type 5 in neurons. Mol Cell Neurosci 45. Thomas U. Modulation of synaptic signalling complexes by homer 2002;20:323–29 proteins. J Neurochem 2002;81:407–13 70. Bergson C, Levenson R, Goldman-Rakic PS, et al. Dopamine receptor- 46. Fagni L, Worley PF, Ango F. Homer as both a scaffold and transduction interacting proteins: The Ca(21) connection in dopamine signaling. molecule. Sci STKE 2002;137:RE8 Trends Pharmacol Sci 2003;24:486–92 47. Fagni L, Ango F, Perroy J, et al. Identification and functional roles of 71. Dickey CA, Loring JF, Montgomery J, et al. Selectively reduced metabotropic glutamate receptor-interacting proteins. Semin Cell Dev expression of synaptic plasticity-related genes in amyloid precursor Biol 2004;15:289–98 protein 1 presenilin-1 transgenic mice. J Neurosci 2003;23:5219–26 48. St-Hilaire M, Landry E, Levesque D, et al. Denervation and repeated L- 72. Chavkin C, James IF, Goldstein A. Dynorphin is a specific endogenous DOPA induce a coordinate expression of the NGFI-B ligand of the kappa opioid receptor. Science 1982;215:413–15 in striatal projection pathways in hemi-parkinsonian rats. Neurobiol Dis 73. Vincent S, Hokfelt T, Christensson I, et al. Immunohistochemical 2003;14:98–109 evidence for a dynorphin immunoreactive striato-nigral pathway. Eur J 49. Hill RG. The status of naloxone in the identification of pain control mech- Pharmacol 1982;85:251–52 anisms operated by endogenous opioids. Neurosci Lett 1981;21:217–22 74. Gerfen CR, YoungWS. 3rd Distribution of striatonigral and striatopallidal 50. Portoghese PS, Lipkowski AW, Takemori AE. Binaltorphimine and nor- peptidergic neurons in both patch and matrix compartments: An in situ binaltorphimine, potent and selective kappa-opioid receptor antagonists. hybridization histochemistry and fluorescent retrograde tracing study. Life Sci 1987;40:1287–92 Brain Res 1988;460:161–67 51. Piercey MF, Lahti RA, Schroeder LA, et al. U-50,488H, a pure kappa 75. Meshul CK, McGinty JF. Kappa opioid receptor immunoreactivity in the receptor agonist with spinal analgesic loci in the mouse. Life Sci 1982;31: nucleus accumbens and caudate–putamen is primarily associated with 1197–1200 synaptic vesicles in axons. Neuroscience 2000;96:91–99 52. Fosnaugh JS, Bhat RV, Yamagata K, et al. Activation of Arc, a putative 76. Abou-Khalil B, Young AB, Penney JB. Evidence for the presynaptic ÔeffectorÕ immediate early gene, by cocaine in rat brain. J Neurochem localization of opiate binding sites on striatal efferent fibers. Brain Res 1995;64:2377–80 1984;323:21–29

946 q 2005 American Association of Neuropathologists, Inc. J Neuropathol Exp Neurol  Volume 64, Number 11, November 2005 Arc Correlates With L-Dopa-Induced Motor Behavior

77. Samadi P,Gregoire L, Bedard PJ. Opioid antagonists increase the dyskinetic 83. Parent A. Extrinsic connections of the basal ganglia. Trends Neurosci response to dopaminergic agents in parkinsonian monkeys: Interaction be- 1990;13:254–58 tween dopamine and opioid systems. Neuropharmacology 2003;45:954–63 84. Smith AD, Bolam JP. The neural network of the basal ganglia as revealed 78. Johansson PA, Andersson M, Andersson KE, et al. Alterations in cortical by the study of synaptic connections of identified neurones. Trends and basal ganglia levels of opioid receptor binding in a rat model of Neurosci 1990;13:259–65 l-DOPA-induced dyskinesia. Neurobiol Dis 2001;8:220–39 85. Smith Y, Raju DV, Pare JF, et al. The thalamostriatal system: A highly 79. Morris BJ, Herz A. Control of opiate receptor number in vivo: specific network of the basal ganglia circuitry. Trends Neurosci 2004;27: Simultaneous kappa-receptor down-regulation and mu-receptor up- 520–27 regulation following chronic agonist/antagonist treatment. Neuroscience 86. McGeorge AJ, Faull RL. The organization of the projection from the 1989;29:433–42 cerebral cortex to the striatum in the rat. Neuroscience 1989;29:503–37 80. Moore RY, Bhatnagar RK, Heller A. Anatomical and chemical studies of 87. Berendse HW, Galis-de Graaf Y, Groenewegen HJ. Topographical a nigro-neostriatal projection in the cat. Brain Res 1971;30:119–35 organization and relationship with ventral striatal compartments of 81. Doucet G, Descarries L, Garcia S. Quantification of the dopamine prefrontal corticostriatal projections in the rat. J Comp Neurol 1992;316: innervation in adult rat neostriatum. Neuroscience 1986;19:427–45 314–47

82. Parent A, Mackey A, De Bellefeuille L. The subcortical afferents to 88. Van der Kooy D. The organization of the thalamic, nigral and raphe cells Downloaded from https://academic.oup.com/jnen/article/64/11/936/2916615 by guest on 30 September 2021 caudate nucleus and putamen in primate: A fluorescence retrograde projecting to the medial vs lateral caudate-putamen in rat. A fluorescent double labeling study. Neuroscience 1983;10:1137–50 retrograde double labeling study. Brain Res 1979;169:381–87

q 2005 American Association of Neuropathologists, Inc. 947