+ All Categories
Home > Documents > Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al,...

Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al,...

Date post: 07-Nov-2020
Category:
Upload: others
View: 3 times
Download: 0 times
Share this document with a friend
17
RESEARCH ARTICLE Differential involvement of Ras-GRF1 and Ras-GRF2 in L-DOPA-induced dyskinesia Simone Bido 1 , Nicola Solari 2,3 , Marzia Indrigo 2,3 , Angela D’Antoni 2 , Riccardo Brambilla 2,4 , Michele Morari 1,5 & Stefania Fasano 2,4 1 Section of Pharmacology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy 2 Division of Neuroscience, Institute of Experimental Neurology, IRCSS San Raffaele Scientific Institute, Milan, Italy 3 Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy 4 Division of Neuroscience, School of Biosciences, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom 5 Neuroscience Centre and National Institute of Neuroscience, Ferrara, Italy Correspondence Stefania Fasano, Division of Neuroscience, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy. Tel: +390226434877; Fax: +390226434767; E-mail: [email protected] Funding Information The work was supported by the Michael J. Fox Foundation for Parkinson’s Research (to R. B.), by Parkinson’s UK (to R. B.), the Italian Ministry of Health (to R. B., and S. F.), the Compagnia di San Paolo (to R. B. and M. M.). Received: 16 September 2014; Revised: 13 March 2015; Accepted: 13 March 2015 Annals of Clinical and Translational Neurology 2015; 2(6): 662–678 doi: 10.1002/acn3.202 S. B. and N. S. contributed equally to this work. Abstract Objective: Recent findings have shown that pharmacogenetic manipulations of the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy- phenylalanine (L-DOPA)-induced dyskinesia (LID). First, we investigated whether a prolonged L-DOPA treatment differentially affected ERK signaling in medium spiny neurons of the direct pathway (dMSNs) and in cholinergic asp- iny interneurons (ChIs) and assessed the role of Ras-GRF1 in both subpopula- tions. Second, using viral-assisted technology, we probed Ras-GRF1 and Ras-GRF2 as potential targets in this pathway. We investigated how selective blockade of striatal Ras-GRF1 or Ras-GRF2 expression impacted on LID (induction, maintenance, and reversion) and its neurochemical correlates. Methods: We used both Ras-GRF1 knockout mice and lentiviral vectors (LVs) delivering short-hairpin RNA sequences (shRNAs) to obtain striatum-specific gene knockdown of Ras-GRF1 and Ras-GRF2. The consequences of these genetic manipulations were evaluated in the 6-hydroxydopamine mouse model of Parkinson’s disease. Escalating doses of L-DOPA were administered and then behavioral analysis with immunohistochemical assays and in vivo microdialysis were performed. Results: Ras-GRF1 was found essential in controlling ERK sig- naling in dMSNs, but its ablation did not prevent ERK activation in ChIs. Moreover, striatal injection of LV-shRNA/Ras-GRF1 attenuated dyskinesia development and ERK-dependent signaling, whereas LV-shRNA/Ras-GRF2 was without effect, ruling out the involvement of Ras-GRF2 in LID expression. Accordingly, Ras-GRF1 but not Ras-GRF2 striatal gene-knockdown reduced L-DOPA-induced GABA and glutamate release in the substantia nigra pars reticulata, a neurochemical correlate of dyskinesia. Finally, inactivation of Ras-GRF1 provided a prolonged anti-dyskinetic effect for up to 7 weeks and significantly attenuated symptoms in animals with established LID. Interpreta- tion: Our results suggest that Ras-GRF1 is a promising target for LID therapy based on Ras-ERK signaling inhibition in the striatum. Introduction L-DOPA-induced dyskinesia (LID) is one of the unwanted and debilitating motor side effects ensuing from pro- longed treatment with L-DOPA, the gold standard for the symptomatic treatment of Parkinson’s disease (PD). 1 Indeed, the majority of PD patients develop abnormal involuntary movements (AIMs) within 510 years of L- DOPA therapy. 2 LID is still a significant clinical problem since no truly effective treatment has been developed so 662 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
Transcript
Page 1: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

RESEARCH ARTICLE

Differential involvement of Ras-GRF1 and Ras-GRF2 inL-DOPA-induced dyskinesiaSimone Bido1, Nicola Solari2,3, Marzia Indrigo2,3, Angela D’Antoni2, Riccardo Brambilla2,4,Michele Morari1,5 & Stefania Fasano2,4

1Section of Pharmacology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy2Division of Neuroscience, Institute of Experimental Neurology, IRCSS San Raffaele Scientific Institute, Milan, Italy3Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy4Division of Neuroscience, School of Biosciences, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom5Neuroscience Centre and National Institute of Neuroscience, Ferrara, Italy

Correspondence

Stefania Fasano, Division of Neuroscience,

Institute of Experimental Neurology, IRCCS

San Raffaele Scientific Institute, Via Olgettina

58, 20132 Milan, Italy.

Tel: +390226434877;

Fax: +390226434767;

E-mail: [email protected]

Funding Information

The work was supported by the Michael J.

Fox Foundation for Parkinson’s Research (to

R. B.), by Parkinson’s UK (to R. B.), the Italian

Ministry of Health (to R. B., and S. F.), the

Compagnia di San Paolo (to R. B. and M.

M.).

Received: 16 September 2014; Revised: 13

March 2015; Accepted: 13 March 2015

Annals of Clinical and Translational

Neurology 2015; 2(6): 662–678

doi: 10.1002/acn3.202

S. B. and N. S. contributed equally to this

work.

Abstract

Objective: Recent findings have shown that pharmacogenetic manipulations ofthe Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine (L-DOPA)-induced dyskinesia (LID). First, we investigatedwhether a prolonged L-DOPA treatment differentially affected ERK signaling inmedium spiny neurons of the direct pathway (dMSNs) and in cholinergic asp-iny interneurons (ChIs) and assessed the role of Ras-GRF1 in both subpopula-tions. Second, using viral-assisted technology, we probed Ras-GRF1 andRas-GRF2 as potential targets in this pathway. We investigated how selectiveblockade of striatal Ras-GRF1 or Ras-GRF2 expression impacted on LID(induction, maintenance, and reversion) and its neurochemical correlates.Methods: We used both Ras-GRF1 knockout mice and lentiviral vectors (LVs)delivering short-hairpin RNA sequences (shRNAs) to obtain striatum-specificgene knockdown of Ras-GRF1 and Ras-GRF2. The consequences of thesegenetic manipulations were evaluated in the 6-hydroxydopamine mouse modelof Parkinson’s disease. Escalating doses of L-DOPA were administered and thenbehavioral analysis with immunohistochemical assays and in vivo microdialysiswere performed. Results: Ras-GRF1 was found essential in controlling ERK sig-naling in dMSNs, but its ablation did not prevent ERK activation in ChIs.Moreover, striatal injection of LV-shRNA/Ras-GRF1 attenuated dyskinesiadevelopment and ERK-dependent signaling, whereas LV-shRNA/Ras-GRF2 waswithout effect, ruling out the involvement of Ras-GRF2 in LID expression.Accordingly, Ras-GRF1 but not Ras-GRF2 striatal gene-knockdown reducedL-DOPA-induced GABA and glutamate release in the substantia nigra parsreticulata, a neurochemical correlate of dyskinesia. Finally, inactivation ofRas-GRF1 provided a prolonged anti-dyskinetic effect for up to 7 weeks andsignificantly attenuated symptoms in animals with established LID. Interpreta-tion: Our results suggest that Ras-GRF1 is a promising target for LID therapybased on Ras-ERK signaling inhibition in the striatum.

Introduction

L-DOPA-induced dyskinesia (LID) is one of the unwantedand debilitating motor side effects ensuing from pro-longed treatment with L-DOPA, the gold standard for the

symptomatic treatment of Parkinson’s disease (PD).1

Indeed, the majority of PD patients develop abnormalinvoluntary movements (AIMs) within 5–10 years of L-DOPA therapy.2 LID is still a significant clinical problemsince no truly effective treatment has been developed so

662 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use anddistribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.

Page 2: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

Figure 1. LID profile and ERK activation in dMSNs and ChIs during chronic L-DOPA administration. (A) Experimental design 1: 2 weeks after 6-

OHDA lesion, different groups of C57BL/6 mice were treated with L-DOPA: one group (n = 8) received an acute dose (3 mg/kg, 1 day) of L-DOPA

(hereinafter 1 day) and was perfused 20 min thereafter without performing AIMs; one group (n = 11) was treated with a 9 days protocol

(hereinafter 9 days) (days 1–3, 3 mg; days 4–6, 6 mg; days 6–9, 12 mg/kg) and lastly, two groups after the 9 days protocol underwent a

dyskinesia maintenance protocol consisting of twice a week injections (12 mg/kg L-DOPA plus 12 mg/kg of benserazide) for either 2 weeks

(n = 11) or 4 weeks (n = 11). For each time point, a small group of mice (n = 3) was treated with saline as internal control for subsequent

postmortem analysis. (B) Temporal profiles of all groups throughout the 9 days, 2 weeks, and 4 weeks protocol expressed as a sum of Axial,

Limb, and Orolingual AIMs (ALO AIMs) are shown. No differences in dyskinesia expression levels were found amongst groups in either the initial

9 days period (P = 0.259) or throughout the maintenance period (2 weeks vs. first 2 weeks of 4 weeks P = 0.577; vs. last 2 weeks of 4 weeks

P = 0.238). (C) Pearson’s correlation index revealed a positive, linear correlation between pERK levels in dMSNs and AIMs in the 9 days, but not

in the 4 weeks group (9 days: r = 0.802, P < 0.05; 4 weeks: r = 0.084, P = 0.878). (D) Representative micrographs of ERK1/2 immunoreactivity

(pERK) in the dorsolateral part of the striatum after completion of different L-DOPA treatments (1 day, 9 days and maintenance protocol). Scale

bar 30 lm. (E) Quantification of pERK levels observed in all L-DOPA-treated groups with statistical difference between 1 day and 9 days protocol

(one way ANOVA, treatment effect F(4, 38) = 20.884, P < 0.0001, Tukey’s post hoc 1 day vs. 9 days P < 0.05). (F) Immunofluorescent

micrographs of ERK activation in the dMSNs: pERK (green), DR1 (red) and NeuN (blue) cells in dorsal striatal. Scale bar 20 lm. Small insets of split

channels of pERK and DR1-positive cells are also reported. (G) Quantification of pERK-positive cells in D1R-positive and -negative cells, with

statistical difference after 1 day and 9 days protocol (one way ANOVA, treatment effect F(3, 34) = 8.2302, P < 0.001 Tukey’s post hoc 1 day vs.

9 days P < 0.001). (H) Immunofluorescent micrographs of ERK activation in cholinergic interneurons: pERK (green), ChAT (red) and NeuN (blue)

cells. Scale bar 30 lm. Small insets of split channels of ChAT and pERK-positive cells are also reported. (I) A raise of pERK levels was observed in

ChIs after 9 days (Kruskal–Wallis Test P < 0.0001; Mann–Whitney Test 1 day vs. 9 days, 2 weeks and 4 weeks P < 0.01) and persisted through

the prolonged administration of L-DOPA (Mann–Whitney Test 9 days vs. 2 weeks P = 0.124, 9 days vs. 4 weeks P = 0.787, 2 weeks vs. 4 weeks

P = 0.916). *P < 0.05, **P < 0.0.01, ***P < 0.001. LID, L-DOPA-induced dyskinesia; L-DOPA, L-3,4-dihydroxyphenylalanine; 6-OHDA,

6-hydroxydopamine; AIMs, abnormal involuntary movements.

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 663

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 3: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

far. Besides the classical pharmacological approach target-ing neurotransmitter receptors, accumulating evidencefrom animal models supports a causative role for dysreg-ulated D1 receptor intracellular signaling in striatal med-ium-sized spiny neurons of the direct pathway (dMSNs).These observations have opened new perspectives forinnovative therapeutic approaches against LID, based onthe inhibition of either the canonical PKA/DARPP-32 cas-cade or the non-canonical Ras-ERK and mTOR path-ways.3–11

The Ras-ERK cascade is an evolutionarily conservedneuronal pathway involved in several survival processesand an important regulator of behavioral plasticity.12–19

Its sustained activation leads to synaptic rearrangementsrequiring de novo gene expression and protein synthe-sis. In striatal cells, glutamate (GLU) and dopaminereceptors interact and provide a route to ERK activa-tion.20–24 Importantly, in animal models of PD, includ-ing the unilaterally 6-hydroxydopamine (6-OHDA)lesioned rodent and the 1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine (MPTP)-treated non-human primate(NHP), the supersensitivity of dopamine D1 receptorsleads to aberrant ERK activation in response to L-DOPA, which correlates with LID severity.3–5,25,26 Inparticular, our recent study indicated that Ras-GRF1, aRas activator (Ras guanine-nucleotide exchange factor,Ras-GEF) expressed only in mature neurons of the cen-tral nervous system, is necessary for the integration ofGLU and dopamine signaling that leads to ERK activa-tion.23 Importantly, Ras-GRF1 specifically controlsdownstream ERK signaling in a neurotrophin-indepen-dent manner, suggesting that its inhibition would onlyaffect plasticity-related ERK signaling without alteringcell survival mechanisms. Consistently, Ras-GRF1 abla-tion by conventional gene targeting27 does not affectthe ability of 6-OHDA to deplete dopamine-producingcells but strongly attenuates ERK activation and AIMsappearance in the rodent lesion model of LID.28 Nota-bly, whilst ERK activity is required in all striatal cells toinduce long-term potentiation (LTP), Ras-GRF1 is nec-essary only in striatal dMSNs, that is, those cells moredirectly implicated in LID.11 Moreover, attenuation ofRas-GRF1 and ERK signaling in the NHP model of PDresults in a strong reduction in dyskinetic symptomswithout compromising the antiparkinsonian effect of L-DOPA, providing a more clinically valuable approachvia targeting Ras-ERK, which may ameliorate this path-ological condition.28

A recent study showed that in Pitx3-deficient mice, agenetic model of PD, the abnormal activation of ERKsurprisingly diminishes in MSNs but increases in the

large aspiny cholinergic interneurons (ChIs), upon con-tinuous administration of L-DOPA.29 In addition, in asubsequent study Won and colleagues demonstratedthat selective depletion of striatal ChIs via Cre-depen-dent viral expression of the diphtheria toxin A signifi-cantly attenuated LID without affecting the therapeuticefficacy of L-DOPA.30 This evidence prompted us toinvestigate in the first part of our work whether inter-mittent but prolonged administration of high doses ofL-DOPA could lead to different ERK activation profilesin dMSNs and ChIs and whether this activation in cho-linergic interneurons is somehow regulated by Ras-GRF1. Our previous observation that Ras-GRF1 inhibi-tion in the brain only leads to ~50% reduction inAIMs could indeed suggest that other factors may regu-late striatal ERK activity in response to L-DOPA,including a potentially Ras-GRF1-independent ERK acti-vation in ChIs. Moreover, Ras-GRF2, a close homologof Ras-GRF1 in the striatum, may be an additional fac-tor controlling ERK activity in dyskinesia. To addressthis point in the second part of the work, we tookadvantage of short hairpin RNA sequences (shRNA)technology to specifically target the dorsolateral stria-tum, a key area involved in the development of dyski-netic movements to obtain striatal-specific geneknockdown of Ras-GRF1 and Ras-GRF2. We then ana-lyzed the behavioral consequences of these geneticmanipulations in the development of dyskinesia andtheir molecular and neurochemical correlates. Finally,we analyzed the possibility of reversing an alreadyestablished dyskinetic state.Our findings showed that a sustained activation of

ERK is present in both dMSNs and ChIs after 4 weeks ofintermittent high doses of L-DOPA but in ChIs this eventis Ras-GRF1 independent. In addition, only Ras-GRF1,but not Ras-GRF2 was critically involved in the develop-ment of LID and in the correlated molecular changes.Importantly, Ras-GRF1 silencing had a long-lasting thera-peutic effect, up to 7 weeks and was also effective in alle-viating established dyskinesia, demonstrating the potentialas a therapeutic intervention.

Material and Methods

For detailed description of material and methods see DataS1.

Animals

In this study, we used the following: (1) C57BL/6 malemice (Charles River Laboratories, Calco, Italy); (2) Ras-

664 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 4: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

GRF1 KO males and their littermates.27 All experimentalanimal procedures were conducted according to the EUDirective 2010/63/EU and to experimental animal licenses

of the Fondazione San Raffaele del Monte Tabor andUniversity of Ferrara approved by the Italian Ministry ofHealth and the local authorities.

Figure 2. Reduced ERK signaling in dMSNs but not ChIs population in Ras-GRF1 KO mice. (A) Immunofluorescent photomicrographs of ERK

activation in the dMSNs: pERK (green), DR1 (red) and NeuN (blue) cells in dorsal striatal area of WT and Ras-GRF1 KO mice after the 4 weeks

regimen of L-DOPA. Scale bar 20 lm. (B) pERK-positive cells colocalizing with D1R-positive cells were counted, with statistical difference between

WT and Ras-GRF1 KO mice (Independent-samples t-test, P < 0.001). (C) Immunofluorescent micrographs of acetyl-histone H3 activation (pAcH3)

in the dMSNs: pAcH3 (green), DR1 (red) and NeuN (blue) cells in dorsal striatal area of WT and Ras-GRF1 KO mice after the 4 weeks protocol of

L-DOPA. Scale bar 20 lm. (D) pAcH3-positive cells colocalizing with D1R-positive cells were counted, with statistical difference between WT and

Ras-GRF1 KO mice (Independent-samples t-test, P < 0.05). (E) Immunofluorescent micrographs of ERK activation in ChIs: pERK (green), ChAT (red)

and NeuN (blue) cells in dorsal striatal area of WT and Ras-GRF1 KO mice after 4 weeks regimen of L-DOPA. Scale bar 20 lm. (F) pERK-positive

cells colocalizing with ChAT-positive cells were observed, without statistical difference between WT and Ras-GRF1 KO mice (Independent-samples

t-test, P = 0.415). (G) Immunofluorescent micrographs of AcH3 activation in the ChIs: pAcH3 (green), ChAT (red) and NeuN (blue) cells in dorsal

striatal area of WT and Ras-GRF1 KO mice after the 4 weeks regimen of L-DOPA. Scale bar 20 lm. (H) pAcH3-positive cells colocalizing with

ChAT-positive cells were observed, without statistical difference between WT and Ras-GRF1 KO mice (Independent-samples t-test, P = 0.295).

Data are mean ! SEM of 8–10 animals for each group. *P < 0.05, ***P < 0.001.

Figure 3. Ras-GRFs-specific gene silencing in vitro and in vivo. Ras-GRF1 and Ras-GRF2 residual protein expression levels in neurons were

determined by Western blotting 48 h after LVs infection with three different shRNA cassettes (Seq 3, 2, 1 for (A) Ras-GRF1 and Seq 1,2,3 for (C)

Ras-GRF2) or the relative control sequence (Seq Ctr). Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as a loading control. (B and

D) Quantification of three independent experiments showed that all sequences were able to reduce the expression of Ras-GRF proteins. (E) For

in vivo delivery, Seq 1 of sh-Ras-GRF1 and Seq 3 of sh-Ras-GRF2 were injected in striatum of wild type mice. Representative immunoblots of p140

Ras-GRF1 and p135 Ras-GRF2 are shown. (F) Densitometry analysis demonstrated that p140 Ras-GRF1 was reduced by sh-Ras-GRF1 (one way

ANOVA, main effect F(2, 9) = 10.517, P < 0.01, Bonferroni’s post hoc sh-Ctr vs. sh-Ras-GRF1 P < 0.01) without alterations in Ras-GRF2 protein

levels (Bonferroni’s post hoc sh-Ctr vs. sh-Ras-GRF2 P = 1.000). Similarly, p135 Ras-GRF2 was reduced by sh-Ras-GRF2 expression (main effect F

(2, 9) = 5.45, P < 0.05, Bonferroni’s post hoc sh-Ctr vs. sh-Ras-GRF2 P < 0.05) with no variation in Ras-GRF1 protein levels (Bonferroni’s post hoc

sh-Ctr vs. sh-Ras-GRF1 P = 0.282). (G) Immunoreactivity to both anti-phospho-ERK and anti-ERK antibodies in striata of sh-Ras-GRF1, sh-Ras-GRF2

and sh-Ctr injected mice. (H—I) Quantification of normalized p44 and p42 bands intensities revealed no changes in either protein levels or basal

phosphorylation state of ERK1/2. *Non-specific band. *P < 0.05, **P < 0.0.01.

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 665

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 5: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

shRNA constructs and LV production

Three shRNA constructs were probed against Ras-GRF1and Ras-GRF2 genes in primary cultures. The sequencetargeting the Ras-GEF region of Ras-GRF1 (sequence 1)

and PH domain of Ras-GRF2 (sequence 3) were usedin vivo. The Vesicular stomatitis virus (VSV) pseudotypedthird-generation lentiviral vectors (LVs) were produced aspreviously described.31,32

666 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 6: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

Stereotaxic Surgery and L-DOPA treatments

6-OHDA lesion

Unilateral 6-OHDA MFB lesions were performed asdescribed in Fasano et al.28

LV-shRNA injections

Two weeks post-6-OHDA lesions, mice with confirmedipsilateral rotations received unilateral striatal injections(2 9 1 lL) of LVs as described in Fasano et al.28

L-DOPA administration

L-DOPA (Sigma Aldrich, Milan, Italy) was injectedaccordingly to the following protocols: (1) daily injectionof 3 (day 1–3), 6 (day 4–6) or 12 (day 7–9) mg/kg incombination with benserazide (Sigma-Aldrich) (12 mg/kg) in the 9 days protocol; (2) twice a week 12 mg/kgwith 12 mg/kg benserazide in the maintenance protocolof 4 and 7 weeks and in the reversal protocol of 4 weeks(experimental design 1, 4, and 5, respectively). Mice nottreated with L-DOPA received an equivalent volume of

Figure 4. Striatal-specific gene knockdown of Ras-GRF1 or Ras-GRF2 does not impact on basal motor behavior of 6-OHDA lesioned mice. (A)

Experimental design 2: 2 weeks after 6-OHDA lesion mice performed spontaneous rotations test and were split in four groups, injected with sh-

Ras-GRF1, sh-Ras-GRF2 and sh-Ctr, respectively. Three weeks post-shRNA injection, mice performed spontaneous rotations, rotarod and drag

test. (B) Spontaneous full rotations were found equivalent among the experimental groups (one way ANOVA, F(3, 35) = 0.282, P = 0.838). (C)

Motor learning during 5 days on the rotarod was measured. A significant effect of training but not of treatment was observed in sh-Ras-GRF1

and sh-Ctr groups (Repeated measures ANOVA, F(1, 17) = 56.336, P < 0.0001). Similarly, sh-Ras-GRF2 and sh-Ctr mice displayed only a

significant effect of training (Repeated measures ANOVA, F(1, 17) = 30.073, P < 0.0001). (D) Akinesia was evaluated using drag test. On day 1,

the number of steps of the contralateral forepaw was significantly less than the one performed with the ipsilateral forepaw in all experimental

groups (Paired-samples t-test, sh-Ras-GRF1: t(8) = "11.300, P < 0.0001; sh-Ctr: t(9) = "8.899, P < 0.0001; sh-Ras-GRF2: t(9) = "5.349,

P < 0.001; sh-Ctr: t(9) = "5.365, P < 0.0001). On the subsequent test on day 5, the number of contralateral steps continued to be significantly

lower than the one of ipsilateral steps in all groups (Paired-samples t-test, sh-Ras-GRF1: t(8) = "5.635, P = 0.001; sh-Ctr: t(9) = "12.825,

P < 0.0001; sh-GRF2: t(9) = 5.312, P < 0.001; sh-Ctr: t(9) = 4.589, P < 0.001). Data are mean ! SEM of 9–11 animals for each group.

***P < 0.001, ****P < 0.0001.

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 667

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 7: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

Saline. Both drugs were injected i.p. in a total volume of10 mL/kg body weight.

Behavioral analysis, in vivo microdialysis and postmor-tem examination were performed as described in refs[28,33–36].

Figure 5. Striatal downregulation of Ras-GRF1 but not Ras-GRF2 is able to attenuate LID expression. Four weeks after sh-injection, 9 days

L-DOPA protocol was administered and AIMs were daily scored. (A) A gradual development of dyskinesia was observed (repeated measure

ANOVA, time effect, F(2.182, 47.999) = 29.703, e = 0.273 P < 0.0001). However, sh-Ras-GRF1 mice developed dyskinesia to a lesser extent

in comparison to their controls (repeated measure ANOVA, shRNA effect, F(1, 22) = 6.944, P < 0.05), with the difference becoming evident

on day 4 of L-DOPA exposure and persisting till day 9 (P < 0.05). (B) The analysis of the area under curve (AUC) of AIMs score confirmed

this reducing effect of sh-Ras-GRF1 (mean ! SEM) (Independent-samples t-test: t(22) = 2.618, P < 0.05). (C) Sh-Ras-GRF1 expression lessened

the Limb and Axial components of AIMs in comparison to sh-Ctr mice (ANOVA, shRNA effect, P < 0.05) while the Orolingual subtype was

found only almost significantly different from sh-Ctr animals (ANOVA, shRNA effect, P = 0.06). (D) Sh-Ras-GRF2 injected mice showed rising

dyskinesia levels over time but the intensity displayed was equal to sh-Ctr mice (repeated measure ANOVA, shRNA effect, F(1, 20) = 0.497,

P = 0.489). (E) Analysis of AUC (mean ! SEM) (Independent-samples t-test: t(20) = "0.773, P = 0.448) and (F) individual components of ALO

AIMs in Ras-GRF2 silenced mice were similar to those observed in sh-Ctr animals (ANOVA, shRNA effect, P > 0.05). Data are mean ! SEM

of 9–11 animals for each group. *P < 0.05.

668 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 8: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

Results

Prolonged and intermittent L-DOPAtreatment in dopamine-depleted miceinduces persistent and long-lasting ERKactivation in both dMSNs and ChIs

In order to define ERK activation profile in dMSNs and ChIsin LID, we first applied different protocols of L-DOPAadministration in 6-OHDA lesioned mice: (1) 1 day acuteprotocol; (2) 9 days dose-escalating protocol and (3) 9 daysprotocol followed by LID maintenance protocol of either 2or 4 weeks (Fig. 1A). In the acute protocol, mice wereinjected with 3 mg/kg of L-DOPA and 20 min after theywere perfused to detect molecular changes without scoringAIMs. In the 9 days protocol, mice developed a clear dyski-netic state that remained stable over 4 weeks (Fig. 1B). Acorrelation was found between ERK1/2 immunoreactivity(pERK levels) in dMSNs and axial, limb and orolingual(ALO) AIMs at 9 days but not 4 weeks (Fig. 1C). In fact,ERK activity in the dMSNs became maximal already afteracute L-DOPA challenge and remained high also after 9 daysof treatment in concomitance to an exacerbated dyskineticbehavior. However, we detected only a small but significantdecrease after 9 days of treatment in comparison to theacute challenge that was not observed after the intermittentadministration of L-DOPA in the maintenance protocol(Fig. 1D–G). In contrast, in ChIs, ERK activation was evi-dent only after 9 days of L-DOPA administration (Fig. 1Hand I). The results thus far indicate that ERK activityremains high in dMSNs in response to a prolonged butintermittent L-DOPA administration and appears also inChIs only after a repeated challenge with L-DOPA.

Ras-GRF1 controls ERK signaling in dMSNsbut not in ChIs

As described earlier, Ras-GRF1 KO animals are signifi-cantly less dyskinetic than controls and exhibit lower lev-els of ERK activation and FosB/DFosB expression. Thisevidence was not due to a different expression of Ras-GRF1 in striatonigral MSNs but to the specific engage-ment of Ras-GRF1 by D1 receptors.11,28

Thus, we next examined whether Ras-GRF1 may alsoregulate ERK signaling in ChIs using the Ras-GRF1 KOmodel. We found that pERK was significantly reduced indMSNs (Fig. 2A and B) but not in ChIs (Fig. 2E and F)of Ras-GRF1 KO animals upon a 4 weeks dyskinesiamaintenance protocol. Similarly, downstream phosphory-lation of acetyl-histone H3 (AcH3), also known to beassociated with LID but never measured in ChIs4,25 wasonly attenuated in dMSNs (Fig. 2C and D) but not inChIs (Fig. 2G and H). These data strongly indicate thatRas-GRF1 does not control ERK activity in ChIs of thestriatum, suggesting that other factors may regulate ERKsignaling in these cells.

Validation of LV-assisted gene knockdownof Ras-GRF1 and Ras-GRF2

Ras-GRF2 is a close homolog of Ras-GRF1, and isexpressed, although to a lesser extent, in the striatum.23

To address the possibility that Ras-GRF2 could play arole in LID, we used a sophisticated viral-mediatedapproach to knockdown both Ras-GRF1 and Ras-GRF2gene in striatal cells, in vitro and in vivo. Expression cas-settes containing 3 distinct Ras-GRF1 and Ras-GRF2-spe-

Figure 6. Striatal-specific gene knockdown of Ras-GRF1 but not Ras-GRF2 reduces ERK phosphorylation and FosB/DFosB accumulation associated

with LID. (A) Photomicrographs of ERK1/2 immunoreactivity in the dorsal striatum of sh-Ctr and sh-Ras-GRF1 groups after 9 days L-DOPA or

Saline treatment. (B) Quantification of pERK-positive cells revealed a significant effect of L-DOPA in both shRNA injected groups in comparison to

saline (two way ANOVA, treatment effect, F(1, 29) = 41.890, P < 0.0001). However, a significant reduction of pERK was observed in sh-Ras-GRF1

mice (two way ANOVA, interaction genotype 9 treatment effect, F(1, 29) = 6.319, P < 0.05). (C) Pearson’s correlation index revealed a strong,

linear correlation between ERK levels and AIMS in both experimental groups (sh-Ctr: r = 0.935, P < 0.001; sh-Ras-GRF1: r = 0.914, P < 0.0001).

(D) Photomicrographs of pERK in sh-Ctr and sh-Ras-GRF2 groups after 9 days L-DOPA protocol. (E) Quantification of pERK-positive cells revealed a

significant effect of L-DOPA (two way ANOVA, treatment effect, F(1, 26) = 84.954, P < 0.0001) without difference between sh-Ctr and sh-Ras-

GRF2 mice (two way ANOVA, shRNA effect, F(1, 26) = 0.026, P = 0.874). (F) Pearson’s correlation index revealed a positive, linear correlation

between ERK levels and AIMS in both experimental groups (sh-Ctr: r = 0.850, P < 0.001; sh-Ras-GRF2: r = 0.895, P < 0.0001). (G)

Photomicrographs of FosB/DFos Bimmunoreactive cells in the dorsal striatum of sh-Ctr and sh-Ras-GRF1 groups after 9 days L-DOPA or Saline

treatment. (H) Ras-GRF1 knockdown significantly reduced FosB/DFosB accumulation (two way ANOVA, interaction genotype 9 treatment effect, F

(1, 29) = 4.402, P < 0.05). (I) A positive, linear correlation was found between FosB levels and AIMs in both experimental groups (sh-Ctr:

r = 0.931, P < 0.001; sh-Ras-GRF1: r = 0.921, P < 0.0001). (J) Photomicrographs of FosB/DFosB-immunoreactive cells in the dorsal striatum of

sh-Ctr and sh-Ras-GRF2 groups after 9 days L-DOPA or Saline treatment. (K) Equivalent levels of FosB/DFosB accumulation were induced by

L-DOPA treatment (two way ANOVA, treatment effect, F(1, 26) = 83.857, P < 0.0001) without difference between sh-Ras-GRF2 mice and their

controls (two way ANOVA, shRNA effect, F(1, 26) = 0.353, P = 0.557). (L) A positive, linear correlation was found between FosB levels and AIMs

in both experimental groups (sh-Ctr: r = 0.850, P < 0.001; sh-Ras-GRF2: r = 0.895, P < 0.0001). Scale bar 30 lm. Data are mean ! SEM of

9–11 animals for each group. *P < 0.05.

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 669

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 9: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

cific short hairpin RNAs (shRNA) were transferred intogreen fluorescent protein (GFP)-tagged LV in order totest their inhibitory potential in neurons on p140Ras-GRF1

and p135Ras-GRF2, respectively. We prepared in vitro neu-ronal cultures from newborn (P1) wild type mice, whichwere subsequently infected with equal amounts of LV.

Then, cells were processed by Western blot analysis usingspecific antibodies against Ras-GRF1 and Ras-GRF2 pro-teins (Fig. 3A and C). All selected sequences were able toreduce the expression of Ras-GRF proteins in comparisonwith control sequence (hereinafter sh-Ctr). We selectedthe most effective sequences, that is, sequence 1 of LV-

670 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 10: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

shRNA/Ras-GRF1 (hereinafter sh-Ras-GRF1) andsequence 3 of LV-shRNA/Ras-GRF2 (hereinafter sh-Ras-GRF2), for further in vivo experiments (Fig. 3B and Drespectively). To confirm in vivo efficacy of the selectedshRNA sequences, we performed unilateral stereotaxicinjections of sh-Ras-GRF1, sh-Ras-GRF2 or sh-Ctr in themotor striatum of wild type mice, and 3 weeks later,protein content in dorsal striata was determined by Wes-tern blot (Fig. 3E). Expression level of p140Ras-GRF1 wasreduced by sh-Ras-GRF1 with no alterations in Ras-GRF2 levels. Similarly, sh-Ras-GRF2 significantlydecreased the expression level of p135Ras-GRF2 withoutaffecting Ras-GRF1 levels (Fig. 3F). The observed in vivoreduction in p140 and p135 was likely to be an underes-timation due to the significant presence of non-infectedcells in the tissue samples (see also Fig. 7). Importantly,no changes in either ERK1/2 protein levels or basal phos-phorylation were observed (Fig. 3G and H).

Ras-GRF1 and Ras-GRF2 gene knockdowndoes not interfere with basal motorbehavior in 6-OHDA lesioned mice

Next, we asked whether Ras-GRF1 or Ras-GRF2 knock-down could affect the course of 6-OHDA lesioning.Mice were hemilesioned with 6-OHDA then sorted bycomparable levels of rotational behavior 2 weeks later,and finally unilaterally injected with LVs in the dorsalstriatum. Three weeks later, lesion-induced motor deficitswere examined using spontaneous rotations, rotarod,and drag test28,33 (Fig. 4A). All four shRNA groupsshowed a similar number of spontaneous ipsilateral rota-tions during a 10 min session, suggesting an equivalentextent of 6-OHDA damage, regardless of the injectedshort hairpins (Fig. 4B). Motor abilities were also evalu-ated on the rotarod during a 5-days training protocol.Latency to fall was similar in sh-Ras-GRF1, sh-Ras-GRF2and sh-Ctr animals at day 1, and significantly increasedto the same extent in all groups at day 5, indicating asignificant effect of training (Fig. 4C). A marked reduc-tion in stepping activity at the contralateral forepaw inthe drag test was observed in sh-Ras-GRF1, sh-Ras-GRF2and sh-Ctr mice at both day 1 and day 5 of training,demonstrating a uniform effect of 6-OHDA acrossgroups (Fig. 4D).

Ras-GRF1 but not Ras-GRF2 striatal silencingattenuates LID

To evaluate whether specific knockdown of striatal Ras-GRF1 and Ras-GRF2 would affect behavioral responses torepeated L-DOPA treatment, we applied the 9 days

L-DOPA protocol.28 A gradual development of dyskinesiain both sh-Ras-GRF1 and sh-Ctr injected animals wasobserved. However, LID scores were significantly lower insh-Ras-GRF1 mice than in controls (Fig. 5A and B). Fur-ther analysis indicated that in sh-Ras-GRF1 mice all AIMssubtypes were weakened, confirming the prominent roleof Ras-GRF1 in LID development (Fig. 5C). On the con-trary, time course and intensity of the L-DOPA responsein sh-Ras-GRF2 mice were similar to those observed incontrol animals, ruling out the involvement of Ras-GRF2in LID formation (Fig. 5D–F).On day 10, mice were challenged with a final dose of

12 mg/kg of L-DOPA and transcardially perfused 20 minlater in order to analyze the involvement of Ras-GRF1and Ras-GRF2 in the downstream signaling associatedwith the severity of AIMs. Only sh-Ras-GRF1 miceshowed a significant reduction in ERK activation andFosB/DFosB immunoreactivity (Fig. 6A, B, G and H)whilst knockdown of Ras-GRF2 did not affect thesemolecular changes (Fig. 6D, E, J and K). Furthermore,positive correlations between the severity of AIMs andERK phosphorylation or FosB/DFosB accumulation wereobserved (Fig. 6C, F, I, and L).To confirm that reduction in ERK signaling was spe-

cific to silenced cells we examined the involvement ofRas-GRF1 and Ras-GRF2 in the phosphorylation ofAcH3. Triple-labeling experiments showed that reductionin AcH3 phosphorylation was only restricted to sh-Ras-GRF1 silenced cells (Fig. 7A and B). Indeed, nodifferences were found in non-infected cells of sh-Ctr andsh-Ras-GRF1 mice though the percentage of infection wassimilar in both groups and accounted for ~35% of cells(Fig. 7C and D). Not surprisingly, sh-Ras-GRF2 silencingdid not alter AcH3 activation in L-DOPA-treated mice(Fig. 7E–H). Altogether, these data not only confirm aprominent role of Ras-GRF1 in LID formation and ERKregulation in response to chronic L-DOPA but also ruleout an involvement of Ras-GRF2 in this process.

Ras-GRF1 but not Ras-GRF2 striatalknockdown reduces L-DOPA-induced GABArelease in the substantia nigra

The alterations caused by Ras-GRF1 gene knockdown onbehavioral and cellular correlates of LID prompted us todetermine its possible impact on substantia nigra parsreticulata (SNr) and globus pallidus (GP) neurochemicalchanges after L-DOPA administration. Indeed, previouswork showed that an increase in c-Aminobutyric acid(GABA) and glutamate (GLU) levels in the SNr but notin the GP correlated with the intensity of dyskinesia.34,37

After the 9 days protocol, mice underwent dual probe

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 671

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 11: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

Figure 7. Silencing of Ras-GRF1 in the striatum dampens the L-DOPA-induced phospho-acetylated histone H3. (A) Photomicrographs of pAcH3

(red), shRNA constructs (green) and NeuN (blue) in the dorsal striata of sh-Ras-GRF1 mice after the 9 days L-DOPA protocol. (B) A significant

reduction in pAcH3 was found in successfully silenced cells of mice treated with sh-Ras-GRF1 in comparison to sh-Ctr injected mice (two way

ANOVA, shRNA effect, F(1, 20) = 7.765, **P < 0.001). (C) PAcH3 quantification among non-infected neurons (sh-cells- no GFP) was instead

found equivalent in L-DOPA-treated mice (two way ANOVA, shRNA effect, F(1, 20) = 0.01, P = 0.980). (D) Percentage of sh-infected cells

counted on total NeuN-positive neurons was found comparable in both experimental groups (two way ANOVA, shRNA infection effect, F(1,

20) = 0.666, P = 0.424). (E) Equivalent photomicrographs of pAcH3 in sh-Ras-GRF2 mice after the 9 days L-DOPA protocol. (F) Identical activation

of AcH3 was found in successfully silenced cells of mice treated with sh-Ras-GRF2 in comparison to sh-Ctr injected mice (two way ANOVA,

shRNA effect, F(1, 20) = 0.024, P = 0.878). (G) PAcH3 quantification among non infected neurons (sh-cells- no GFP) was found equivalent in L-

DOPA-treated mice (two way ANOVA, shRNA effect, F(1, 20) = 0.342, P = 0.565). (H) Percentage of sh-infected cells counted on total NeuN-

positive neurons was found comparable in both experimental groups (two way ANOVA, shRNA infection effect, F(1, 20) = 0.010, P = 0.922).

Scale bar, 50 lm. Data are mean ! SEM of 10–11 animals for each group ** P < 0.001.

672 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 12: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

microdialysis implantation and received a challenge of L-DOPA 24 h later: GABA and GLU release was monitoredsimultaneously with AIMs scoring (Fig. 8A). Sh-Ras-GRF1 mice showed less severe dyskinesia in response toL-DOPA than controls (Fig. 8B), this effect being paral-

leled by a milder increase in GABA levels in SNr(Fig. 8D). Conversely, sh-Ras-GRF2 animals were as dys-kinetic as sh-Ctr mice (Fig. 8C) and, consistently, no dif-ference in the extent of the GABA levels rise was observedbetween the two groups (Fig. 8E). Moreover, GLU over-

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 673

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 13: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

flow in the SNr of sh-Ras-GRF1 mice did not show anyincrease over time, contrary to what can be observed insh-Ctr mice (Fig. 8F). On the other hand, both sh-Ras-GRF2 mice and sh-Ctr animals showed a significantincrease in the GLU overflow over time (Fig. 8G). Finally,no changes in either GABA or GLU levels were detectedin the GP of any group of mice in response to L-DOPA(Fig. 8H–K).

These data clearly indicate that Ras-GRF1 inhibitionand the associated attenuation of LID are linked to signif-icant neurochemical changes in the substantia nigra.

Gene knockdown of Ras-GRF1 provides long-term beneficial effects on LID

Finally, we investigated whether Ras-GRF1 gene knock-down could either attenuate LID in a prolonged L-DOPAprotocol or could affect already established dyskineticstate. First, we applied the 9 days protocol followed bytwice a week L-DOPA treatment for 7 weeks (Fig. 9A): asexpected, sh-Ras-GRF1 silenced animals showed a lowerprogression of LID that was maintained throughout theexperiment (Fig. 9B and C). The beneficial effect wasconfirmed on the last day of treatment showing a reducedand shortened ALO profile (Fig. 9D). Second, we treated6-OHDA-lesioned mice with L-DOPA accordingly to our9 days protocol. Then mice were equally balanced in twogroups following an unbiased design and submitted tointrastriatal LVs injection (Fig. 9E). After a short recoveryperiod mice were then challenged with 12 mg/kg ofL-DOPA twice a week for 4 weeks. The dyskinetic profilebefore LVs injection was the same in both groups. How-ever, 3 weeks post-LVs injection, AIMs were significantlyattenuated in sh-Ras-GRF1 mice and were comparable tothe extent observed in the initial phase of the treatment(Fig. 9F–H).

These data demonstrated that Ras-GRF1 inhibition notonly provides a long-term beneficial therapeutic effect inattenuating LID but also can significantly ameliorate asevere dyskinetic state.

Discussion

The Ras-ERK cascade appears to be particularly promis-ing for the treatment of LID since its involvement hasbeen confirmed by various independent studies. However,ERK signaling is not only involved in plasticity processes,like MSNs dysregulation in LID, but also in cell survivalmechanisms. Therefore, identification of key componentsof this signaling pathway selectively involved in LIDwould be highly advantageous since the repeated use ofavailable MEK inhibitors in PD patients may result inintolerable side effects, including an exacerbation of dopa-mine cell loss in the SN.38

Here we found that an acute dose of L-DOPA was ableto trigger maximal ERK activation in dMSNs and dyski-nesia appearance, while a prolonged and intermittentdrug administration resulted in a sustained ERK activityin dMSNs and a stable dyskinetic behavior. Differentlyfrom Ding and colleagues, we observed an engagement ofthe ChIs upon chronic L-DOPA treatment not paralleledby a concomitant ERK downregulation in dMSNs.29

These discrepancies are likely due to a different dose andregimen of L-DOPA administration. Moreover, we didnot observe a reduction in ERK activation after 4 weeksmaintenance protocol as reported by Santini et al., in dys-kinetic NHPs after 3 months of L-DOPA administrationlikely for the same reasons.7 Nevertheless, we consideredthe contribution of ERK signaling in these two cell popu-lations. Thus, we explored the involvement of Ras-GRF1in MSNs and ChIs activity using the Ras-GRF1-deficientmice. Surprisingly, we found that Ras-GRF1 controls ERKactivity exclusively in dMSNs. Indeed in dopamine-depleted striatum, untreated or acutely exposed to L-DOPA, ERK signaling in ChIs is not altered, but increasesonly upon repeated L-DOPA administration, in a Ras-GRF1-independent manner. This finding may explain thepresence of residual dyskinetic symptoms in Ras-GRF1-deficient mice, possibly due to the intact ERK activity inChIs. Thus, unlike dMSNs, where ERK activation uponD1 and GLU receptor interplay requires Ras-GRF1,23 in

Figure 8. Ras-GRF1 reduces L-DOPA-induced GABA release in the substantia nigra. (A) Experimental design 3: after the 9 days L-DOPA protocol,

sh-Ctr, sh-Ras-GRF1 and sh-Ras-GRF2 dyskinetic mice underwent surgery for microdialysis probe implantation, and were challenged with L-DOPA

(12 mg/kg, i.p.) or saline 24 h later. Three baseline samples were collected before the time point 0, indicated with an arrow, corresponding to

L-DOPA i.p. injection. ALO AIMs were scored every 20 min over 120 min after L-DOPA administration. (B) A significant reduction in LID was

observed in L-DOPA-treated sh-Ras-GRF1 in comparison to sh-Ctr mice (two way ANOVA, shRNA effect F(1, 60) = 41.14, P < 0.05). (C)

Comparable levels of dyskinesia were observed in sh-Ras-GRF2 and sh-Ctr mice (two-way ANOVA, shRNA effect F(1, 114) = 3.445, P = 0.066).

(D) A significant reduction in GABA levels in SNr of L-DOPA-treated sh-Ras-GRF1 mice compared to sh-Ctr animals was observed (two-way

ANOVA, shRNA effect F(1, 81) = 29.82, P < 0.0001). (E) Equivalent levels of GABA in SNr were displayed by L-DOPA-treated sh-Ras-GRF2 and sh-

Ctr mice (two-way ANOVA, shRNA effect F(1, 126) = 1.927, P = 0.1675). (F) A reduction in GLU levels in SNr of L-DOPA injected sh-Ras-GRF1

mice compared to sh-Ctr group was found (two-way ANOVA, shRNA effect F(1, 81) = 4.074, P < 0.05). (G) In SNr of sh-Ras-GRF2 groups, GLU

levels were equally altered by L-DOPA treatment (two-way ANOVA, shRNA effect F(1, 108) = 0.7117, P = 0.4008). No relevant differences were

observed among groups for GABA (H and I) and GLU (J and K) levels in GP. Data are mean ! SEM of 6–12 animals. *P < 0.05.

674 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 14: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

cholinergic cells the route of activation may be distinctand the underlying mechanisms are yet to be elucidated.

Regardless of the exact mechanism controlling ERKactivity in ChIs, the reduction in ERK signaling indMSNs of Ras-GRF1 knockout and knockdown mice notonly correlates well with LID attenuation, but also con-firms that blockade of Ras-GRF1 mediated signaling inthe dorsostriatal dMSNs provides a significant antidyski-

netic effect. However, the reduction in ERK activityobserved in Ras-GRF1-deficient dMSNs is not complete,strongly suggesting the involvement of other factors. Wepreviously showed that a suboptimal dose of a MEKinhibitor, SL327, could further reduce AIMs (up to 80%)in Ras-GRF1 KO mice indicating that LID symptoms arelargely ERK dependent.28 For this reason, we hypothe-sized the involvement of a close Ras-GRF1 homolog, Ras-

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 675

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 15: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

GRF2. Unfortunately, our results provide compellingbehavioral, histochemical and neurochemical evidenceexcluding its involvement in this process. This finding isin line with the idea that despite their homology, Ras-GRF1 and Ras-GRF2 display non-overlapping functionalroles.39

In addition to the molecular control of ERK at theinput level, that is, through Ras-GRF1 or other Ras-GEFs,other downstream mechanisms have been involved inLID. The first is the D1R mediated cAMP-PKA-DARPP32cascade whose activation leads to ERK enhancement.4,22

The second, attractive pathway relies on the protein tyro-sine phosphatase Shp-2 that also acts downstream to D1Rto control ERK activity, both in normal and dyskineticstates.9,40

Future work will assess the potential contribution ofthese two additional pathways in the Ras-GRF1-indepen-dent regulation of ERK activity in LID. Finally, it isimportant to note that additional levels of ERK, histoneH3 and/or DFosB regulation could be achieved throughother mechanisms, either involving mGluR5/PLC/PKCmodulation, cAMP/PKA/DREAM-dependent transcrip-tional repression or NO/cGMP signaling as recentlydescribed.10,41,42

One final important point refers to the poorly exploredrelationship between changes in striatal cell signaling andneurotransmitter release. Previous in vivo microdialysisstudies in dyskinetic rats and mice have shown that AIMsappearance following a dyskinetogenic dose of L-DOPA isassociated with an increase in GABA and GLU levels inSNr.34,37,43 These changes are consistent with dMSNshyperactivity since they are prevented by intrastriatal per-fusion with a D1 receptor antagonist.43 In line with thesefindings, silencing Ras-GRF1 in dMSNs caused LID atten-

uation and no surge in nigral amino acid levels uponL-DOPA challenge. Interestingly, however, we found thatalso perfusion with a D1 receptor antagonist in the SNrof dyskinetic rats provides attenuation of both LID andthe accompanying increase in nigral GABA.43 Consis-tently, D1 receptor agonist induced [3H]-GABA releasewas found to be upregulated in ex vivo nigral slicesobtained from dyskinetic rats.44 This suggests that also ni-gral D1 receptors contribute to LID, and that the L-DOPAinduced increase of nigral GABA release might beachieved through activation of both striatal postsynapticD1 receptors located on dMSNs and nigral D1 receptorslocated on striato-nigral GABA afferents. Silencing Ras-GRF1 in dMSNs would therefore prevent up-regulationof both striatal and nigral D1 receptors, making L-DOPAunable to elevate nigral GABA release.LID is a chronic disorder and, once established, it will

be affecting PD patients throughout their life. Thus, inorder to translate our initial findings in clinically relevantobservations, we investigated two important aspects asso-ciated with the Ras-GRF1 role in this disorder. First, weconfirmed that Ras-GRF1 attenuation ameliorates LIDsymptoms also in the maintenance phase, up to 7 weeksof chronic L-DOPA treatment. Second, we found thatalready established dyskinesia could be significantly atten-uated by Ras-GRF1 knockdown. These data strongly sup-port the therapeutic potential of Ras-GRF1 modulationand are in line with our previous work on the NHPmodel.28

Altogether, our work provides important insights for abetter interpretation of the molecular mechanisms under-lying LID and also relevant information for devising suc-cessful therapeutic approaches based on intracellularsignaling inhibition in the striatum.

Figure 9. Ras-GRF1 inhibition reduces appearance of dyskinesia over prolonged L-DOPA treatment and also alleviates its expression. (A)

Experimental design 4: 3 week post-sh-injection, mice underwent first the 9 days L-DOPA protocol followed by 7 weeks maintenance protocol

consisting of twice a week injections (12 mg/kg L-DOPA plus benserazide). (B) Daily scoring of AIMs during the 9 days protocol showed a

significant reduction in dyskinetic behavior in sh-Ras-GRF1 silenced mice over time (repeated measure ANOVA, shRNA effect, F(1, 22) = 15.163,

P < 0.001, time effect, F(8, 176) = 34.263, P < 0.0001; Independent sample T-test, from day 4 to day 9 P < 0.01). This effect persisted over

7 weeks of treatment (repeated measure ANOVA, shRNA effect, F(1, 22) = 41.585, P < 0.001, independent sample T-test, all days of

maintenance P < 0.001). (C) The analysis of the area under curve (AUC) of AIMs score during the maintenance protocol confirmed the protective

effect of sh-Ras-GRF1 (mean ! SEM) (Independent-samples t-test: t(22) = 5.360, P < 0.001). (D) Time profile of the sum of ALO AIMs on the last

day (day 56) of treatment. A significant effect of shRNA, F(1, 22) = 19.412, P < 0.001 and time, F(6, 132) = 141.383, P < 0.001 was found. (E)

Experimental design 5: after the 9 days L-DOPA protocol, dyskinetic mice were divided in two equivalent groups and subsequently underwent

surgery for LVs-injection in the dorsal striatum. Few days later, mice were subjected to a dyskinesia maintenance protocol of 4 weeks (12 mg/kg

of L-DOPA, twice a week). (F) the expression of LID upon 9 days protocol was equivalent in both groups before LVs injection. However, during

the maintenance protocol, sh-Ras-GRF1 showed a significantly reduction in dyskinetic behavior (repeated measure ANOVA, shRNA effect, F(1,

22) = 6.7733, P < 0.05, independent sample T-test, from day 27 to day 41 P < 0.05. (G) The analysis of the area under curve (AUC) of AIMs

score confirmed the antidyskinetic effect of sh-Ras-GRF1 (mean ! SEM) (independent-samples t-test: t(22) = 2.564, P < 0.05). (H) Time profile of

the sum of ALO AIMs on the last day (day 41) of treatment. A significant effect of shRNA, (F(1, 22) = 5.991, P < 0.05) and time, (F(7,

154) = 42.073, P < 0.05) was found. Data are mean ! SEM of 10–12 animals for each group. *P < 0.05, **P < 0.01, ***P < 0.001.

676 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.

Page 16: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

Acknowledgments

This work is dedicated to the memory of the PD patientUmberto d’Isa (1932–2014). The work was supported bythe Michael J. Fox Foundation for Parkinson’s Research(to R. B.), by Parkinson’s UK (to R. B.), the Italian Min-istry of Health (to R. B., and S. F.), the Compagnia diSan Paolo (to R. B. and M. M.). The authors thank Raffa-ele d’Isa for useful suggestions on statistical analysis, Eve-line Doudnikoff and Martina Berganton for technicalassistance. Part of this work was carried out in ALEMBICmicroscopy laboratory at the San Raffaele Scientific Insti-tute. The authors report no biomedical financial interestsor potential conflicts of interest.

Conflict of Interest

None declared.

References

1. Olanow CW, Schapira AH. Therapeutic prospects for

Parkinson disease. Ann Neurol 2013;74:337–347.2. Brichta L, Greengard P, Flajolet M. Advances in the

pharmacological treatment of Parkinson’s disease: targeting

neurotransmitter systems. Trends Neurosci 2013;36:543–554.3. Pavon N, Martin AB, Mendialdua A, Moratalla R. ERK

phosphorylation and FosB expression are associated with

L-DOPA-induced dyskinesia in hemiparkinsonian mice.

Biol Psychiatry 2006;59:64–74.4. Santini E, Valjent E, Usiello A, et al. Critical involvement

of cAMP/DARPP-32 and extracellular signal-regulated

protein kinase signaling in L-DOPA-induced dyskinesia.

J Neurosci 2007;27:6995–7005.5. Westin JE, Vercammen L, Strome EM, et al.

Spatiotemporal pattern of striatal ERK1/2 phosphorylation

in a rat model of L-DOPA-induced dyskinesia and the role

of dopamine D1 receptors. Biol Psychiatry 2007;62:

800–810.6. Santini E, Heiman M, Greengard P, et al. Inhibition of

mTOR signaling in Parkinson’s disease prevents L-DOPA-

induced dyskinesia. Sci Signal 2009;2:ra36.

7. Santini E, Sgambato-Faure V, Li Q, et al. Distinct changes

in cAMP and extracellular signal-regulated protein kinase

signaling in L-DOPA-induced dyskinesia. PLoS One 2010;5:

e12322.

8. Decressac M, Bjorklund A. mTOR inhibition alleviates

L-DOPA-induced dyskinesia in parkinsonian rats.

J Parkinsons Dis 2013;3:13–17.9. Fiorentini C, Savoia P, Savoldi D, et al. Persistent

activation of the D1R/Shp-2/Erk1/2 pathway in L-DOPA-

induced dyskinesia in the 6-hydroxy-dopamine rat model

of Parkinson’s disease. Neurobiol Dis 2013;54:339–348.

10. Fieblinger T, Sebastianutto I, Alcacer C, et al. Mechanisms

of dopamine D1 receptor-mediated ERK1/2 activation in

the parkinsonian striatum and their modulation by

metabotropic glutamate receptor type 5. J Neurosci

2014;34:4728–4740.11. Cerovic M, Bagetta V, Pendolino V, et al. Derangement of

Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1) and

extracellular signal-regulated kinase (ERK) dependent

striatal plasticity in L-DOPA-induced dyskinesia. Biol

Psychiatry 2015;77:106–115.12. Mazzucchelli C, Brambilla R. Ras-related and MAPK

signalling in neuronal plasticity and memory formation.

Cell Mol Life Sci 2000;57:604–611.13. Fasano S, Brambilla R. Cellular mechanisms of striatum-

dependent behavioral plasticity and drug addiction. Curr

Mol Med 2002;2:649–665.14. Girault JA, Valjent E, Caboche J, Herve D. ERK2: a logical

AND gate critical for drug-induced plasticity? Curr Opin

Pharmacol 2007;7:77–85.15. Santini E, Valjent E, Fisone G. Parkinson’s disease:

levodopa-induced dyskinesia and signal transduction. FEBS

J 2008;275:1392–1399.16. Murer MG, Moratalla R. Striatal signaling in L-DOPA-

induced dyskinesia: common mechanisms with drug abuse

and long term memory involving D1 dopamine receptor

stimulation. Front Neuroanat 2011;5:51.

17. Feyder M, Bonito-Oliva A, Fisone G. L-DOPA-induced

dyskinesia and abnormal signaling in striatal medium

spiny neurons: focus on dopamine D1 receptor-mediated

transmission. Front Behav Neurosci 2011;5:71.

18. Fasano S, Brambilla R. Ras-ERK signaling in behavior: old

questions and new perspectives. Front Behav Neurosci

2011;5:1–6.19. Cerovic M, d’Isa R, Tonini R, Brambilla R. Molecular and

cellular mechanisms of dopamine-mediated behavioral

plasticity in the striatum. Neurobiol Learn Mem

2013;105:63–80.20. Valjent E, Corvol JC, Pages C, et al. Involvement of

the extracellular signal-regulated kinase cascade for

cocaine-rewarding properties. J Neurosci 2000;20:8701–8709.

21. Mazzucchelli C, Vantaggiato C, Ciamei A, et al. Knockout

of ERK1 MAP kinase enhances synaptic plasticity in the

striatum and facilitates striatal-mediated learning and

memory. Neuron 2002;34:807–820.22. Valjent E, Pascoli V, Svenningsson P, et al. Regulation of a

protein phosphatase cascade allows convergent dopamine

and glutamate signals to activate ERK in the striatum.

Proc Natl Acad Sci USA 2005;102:491–496.23. Fasano S, D’Antoni A, Orban PC, et al. Ras-guanine

nucleotide-releasing factor 1 (Ras-GRF1) controls

activation of extracellular signal-regulated kinase (ERK)

signaling in the striatum and long-term behavioral

ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association. 677

S. Bido et al. Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia

Page 17: Differential involvement of Ras‐GRF1 and Ras‐GRF2 in L ...orca.cf.ac.uk/81147/1/Bido et al, 2015.pdf · the Ras-ERK pathway provide a therapeutic means to tackle L-3,4-dihydroxy-phenylalanine

responses to cocaine. Biol Psychiatry 2009;66:

758–768.24. Pascoli V, Besnard A, Herve D, et al. Cyclic adenosine

monophosphate-independent tyrosine phosphorylation of

NR2B mediates cocaine-induced extracellular

signal-regulated kinase activation. Biol Psychiatry

2010;69:218–227.25. Darmopil S, Martin AB, De Diego IR, et al. Genetic

inactivation of dopamine D1 but not D2 receptors inhibits

L-DOPA-induced dyskinesia and histone activation. Biol

Psychiatry 2009;66:603–613.26. Alcacer C, Santini E, Valjent E, et al. Galpha(olf) mutation

allows parsing the role of cAMP-dependent and

extracellular signal-regulated kinase-dependent signaling in

L-3,4-dihydroxyphenylalanine-induced dyskinesia.

J Neurosci 2012;32:5900–5910.27. Brambilla R, Gnesutta N, Minichiello L, et al. A role for

the Ras signalling pathway in synaptic transmission and

long-term memory. Nature 1997;390:281–286.28. Fasano S, Bezard E, D’Antoni A, et al. Inhibition of Ras-

guanine nucleotide-releasing factor 1 (Ras-GRF1) signaling

in the striatum reverts motor symptoms associated with

L-DOPA-induced dyskinesia. Proc Natl Acad Sci USA

2010;107:21824–21829.29. Ding Y, Won L, Britt JP, et al. Enhanced striatal

cholinergic neuronal activity mediates L-DOPA-induced

dyskinesia in parkinsonian mice. Proc Natl Acad Sci USA

2011;108:840–845.30. Won L, Ding Y, Singh P, Kang UJ. Striatal cholinergic

cell ablation attenuates L-DOPA induced dyskinesia

in Parkinsonian mice. J Neurosci 2014;34:3090–3094.

31. Indrigo M, Papale A, Orellana D, Brambilla R. Lentiviral

vectors to study the differential function of ERK1

and ERK2 MAP kinases. Methods Mol Biol

2010;661:205–220.32. Papale A, Brambilla R. Lentiviral vectors as research tools

in neurobiology: design and production. In: Brambilla R,

ed. Viral vector approaches in neurobiology and brain

diseases. New York: Springer Science+Business Media,

2014. p. 3–10.33. Viaro R, Sanchez-Pernaute R, Marti M, et al. Nociceptin/

orphanin FQ receptor blockade attenuates MPTP-induced

parkinsonism. Neurobiol Dis 2008;30:430–438.34. Bido S, Marti M, Morari M. Amantadine attenuates

levodopa-induced dyskinesia in mice and rats preventing

the accompanying rise in nigral GABA levels. J Neurochem

2011;118:1043–1055.35. Marti M, Trapella C, Viaro R, Morari M. The nociceptin/

orphanin FQ receptor antagonist J-113397 and L-DOPA

additively attenuate experimental parkinsonism through

overinhibition of the nigrothalamic pathway. J Neurosci

2007;27:1297–1307.36. Mabrouk OS, Marti M, Morari M. Endogenous

nociceptin/orphanin FQ (N/OFQ) contributes to

haloperidol-induced changes of nigral amino acid

transmission and parkinsonism: a combined microdialysis

and behavioral study in naive and nociceptin/orphanin FQ

receptor knockout mice. Neuroscience 2010;166:40–48.37. Mela F, Marti M, Cenci MA, Morari M. Differential role

of striatal and nigral D1 receptors in the expression of

L-DOPA induced dyskinesia and its neurochemical

correlates. Soc Neurosci Meet 2007; Abst. 590.26.

38. McCubrey JA, Steelman LS, Chappell WH, et al.

Mutations and deregulation of Ras/Raf/MEK/ERK and

PI3K/PTEN/Akt/mTOR cascades which alter therapy

response. Oncotarget 2012;3:954–987.39. Fernandez-Medarde A, Santos E. The RasGrf family of

mammalian guanine nucleotide exchange factors. Biochim

Biophys Acta 2011;1815:170–188.40. Fiorentini C, Mattanza C, Collo G, et al. The

tyrosine phosphatase Shp-2 interacts with the dopamine

D(1) receptor and triggers D(1) -mediated Erk

signaling in striatal neurons. J Neurochem 2011;117:

253–263.41. Ruiz-DeDiego I, Mellstrom B, Vallejo M, et al. Activation

of DREAM (downstream regulatory element antagonistic

modulator), a calcium-binding protein, reduces L-DOPA-

induced dyskinesias in mice. Biol Psychiatry 2015;

77:95–105.42. Solis O, Espadas I, Del-Bel EA, Moratalla R. Nitric oxide

synthase inhibition decreases L-DOPA-induced dyskinesia

and the expression of striatal molecular markers in Pitx3

(-/-) aphakia mice. Neurobiol Dis 2015;73:49–59.43. Mela F, Marti M, Bido S, et al. In vivo evidence for a

differential contribution of striatal and nigral D1 and D2

receptors to L-DOPA-induced dyskinesia and the

accompanying surge of nigral amino acid levels. Neurobiol

Dis 2012;45:573–582.44. Rangel-Barajas C, Silva I, Lopez-Santiago LM, et al.

L-DOPA-induced dyskinesia in hemiparkinsonian rats is

associated with up-regulation of adenylyl cyclase

type V/VI and increased GABA release in the

substantia nigra reticulata. Neurobiol Dis 2011;41:

51–61.

Supporting Information

Additional Supporting Information may be found in theonline version of this article:

Data S1. Supplemental information.

678 ª 2015 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals, Inc on behalf of American Neurological Association.

Ras-GRF1 and ERK in Levodopa-Induced Dyskinesia S. Bido et al.


Recommended