Elsevier

Neurobiology of Disease

Volume 85, January 2016, Pages 155-163
Neurobiology of Disease

Genetic deletion of Rhes or pharmacological blockade of mTORC1 prevent striato-nigral neurons activation in levodopa-induced dyskinesia

https://doi.org/10.1016/j.nbd.2015.10.020Get rights and content

Highlights

  • Rhes deletion and mTORC1 blockade attenuate levodopa-induced dyskinesia in mice.

  • Rapamycin but not Rhes deletion fully preserves rotarod performance on levodopa.

  • Rhes deletion and rapamycin prevent the levodopa-induced rise of nigral amino acids.

  • Rapamycin but not Rhes deletion prevents the rise of striatal glutamate.

Abstract

Ras homolog enriched in striatum (Rhes) is a small GTP-binding protein that modulates signal transduction at dopamine receptors, and also activates mammalian target of rapamycin complex 1 (mTORC1). Rhes binding to mTORC1 is hypothesized to play a role in motor disorders such as levodopa-induced dyskinesia. Here, we investigate the behavioral and in vivo neurocircuitry changes associated with genetic deletion of Rhes or inhibition of mTORC1 signaling in the mouse model of levodopa-induced dyskinesia. 6-Hydroxydopamine-hemilesioned Rhes knockout mice and wild-type littermates were chronically treated with levodopa. In parallel, 6-hydroxydopamine-hemilesioned naïve mice were chronically treated with levodopa or levodopa plus rapamycin, to block mTORC1 pathway activation. Dyskinetic movements were monitored during levodopa treatment along with motor activity on the rotarod. Finally, dyskinetic mice underwent microdialysis probe implantation in the dopamine-depleted striatum and ipsilateral substantia nigra reticulata, and GABA and glutamate levels were monitored upon acute challenge with levodopa. Both Rhes knockouts and rapamycin-treated mice developed less dyskinesia than controls, although only rapamycin-treated mice fully preserved rotarod performance on levodopa. Levodopa elevated nigral GABA and glutamate in controls but not in Rhes knockouts or rapamycin-treated mice. Levodopa also stimulated striatal glutamate in controls and Rhes knockouts but not in rapamycin-treated mice. We conclude that both genetic deletion of Rhes and pharmacological blockade of mTORC1 significantly attenuate dyskinesia development by reducing the sensitization of striato-nigral medium-sized spiny neurons to levodopa. However, mTORC1 blockade seems to provide a more favorable behavioral outcome and a wider effect on neurochemical correlates of dyskinesia.

Introduction

Ras homolog enriched in striatum (Rhes) is a 266 amino acid protein isolated in the rat brain through a subtractive hybridization procedure (Falk et al., 1999, Usui et al., 1994). Rhes belongs to the superfamily of Ras proteins, a group of small GTP-binding proteins that exert pleiotropic effects on cell function (Harrison, 2012). Moreover, Rhes has been shown to influence dopamine (DA) and adenosine-related behaviors and signal transduction pathways (Errico et al., 2008, Ghiglieri et al., 2015, Sciamanna et al., 2015, Vitucci et al., 2015). Rhes is highly expressed in striatum, and to a lesser extent in other areas of the rodent and human brain, including hippocampus (Ghiglieri et al., 2015, Spano et al., 2004). In human brain, the ortholog gene, RASD2, has been localized also in the V layer of prefrontal cortex, with a peculiar pattern of expression (Vitucci et al., 2015). Rhes mRNA expression is essentially postnatal and positively regulated by thyroid hormone (Vargiu et al., 2001). Likewise, DA is a positive regulator of Rhes expression in adult striatum (Harrison and LaHoste, 2006, Harrison et al., 2008). Rhes found in striatal medium-sized spiny neurons (MSNs) and cholinergic interneurons is involved in sensorimotor gating functions and modulates striatal DA receptor-dependent signaling and behaviors (Errico et al., 2008, Ghiglieri et al., 2015, Sciamanna et al., 2015, Vitucci et al., 2015). However, Rhes has received much attention for its pathogenic role in Huntington's disease (Mealer et al., 2013, Subramaniam et al., 2009, Subramaniam and Snyder, 2011). Consistently, Rhes acts as a specific striatal E3 ligase (Subramaniam et al., 2010) able to bind and SUMOylate mutant huntingtin, thereby inhibiting its aggregation and increasing its cytotoxicity (Subramaniam et al., 2009). Additionally, it has been postulated that binding to mutant huntingtin prevents Rhes from activating mammalian target of rapamycin complex 1 (mTORC1), thus removing its trophic effects on striatal cells (Lee et al., 2015, Subramaniam and Snyder, 2011). On the other hand, striatal Rhes-induced mTORC1 pathway activation has been proven to be crucial for another motor disorder, i.e. levodopa (L-DOPA) induced dyskinesia (LID) (Subramaniam et al., 2012). LID represents a group of dystonic and choreic involuntary movements emerging in the majority of Parkinson's disease (PD) patients chronically administered with L-DOPA (Bastide et al., 2015, Fabbrini et al., 2007). LID can be very disabling, particularly in the advanced stages of PD, and the yet incomplete knowledge of the pathophysiological mechanisms underlying LID has hindered the identification of a drug capable of preventing the sensitization of striatal MSNs to L-DOPA, the cellular mechanism associated with LID development (Bastide et al., 2015, Santini et al., 2010).

Nonetheless, a pathogenic role in LID onset for aberrant striatal D1 receptor (D1R) signaling in MSNs has been well established (Bastide et al., 2015, Feyder et al., 2011). Indeed, D1R coupling to Gαolf proteins and adenylyl cyclase is enhanced in the dyskinetic brain, which results in an increased activity of cAMP-related protein kinase A (PKA) and phosphorylation (activation) of downstream effectors such as Dopamine- and cAMP-regulated phosphoprotein Mr. 32,000 (DARPP-32) (Nishi et al., 1997). DARRP-32 phosphorylation at Thr34 residue promotes extracellular-regulated kinase 1 and 2 (ERK) signaling (Pavon et al., 2006) which, in turn, activates mTORC1, ultimately responsible for LID onset in mice and rats (Decressac and Bjorklund, 2013, Santini et al., 2008). ERK activates mTORC1 by maintaining Ras homolog enriched in brain (Rheb) in the active state. Until the discovery that Rhes is capable to bind and activate mTORC1 (Subramaniam et al., 2012, Subramaniam and Snyder, 2011), Rheb was believed to be the only striatal mTORC1 activator. Interestingly, in line with Rhes ability to bind and activate mTORC1 signaling, which is known to be instrumental to LID development (Santini et al., 2009), Rhes knockout (Rhes−/−) mice displayed significantly less dyskinesia than wild-type littermates during L-DOPA treatment (Subramaniam et al., 2012).

In the present study, we used an in vivo microdialysis approach to investigate the role of Rhes and mTORC1 in modulating the neurocircuitry underlying dyskinesia associated with striatal dopamine denervation and chronic L-DOPA administration. Specifically, we investigated whether L-DOPA is able to elevate GABA release in substantia nigra reticulata (SNr), an in vivo neurochemical marker of striato-nigral MSNs activation (Bido et al., 2011, Mela et al., 2012, Mela et al., 2007, Paolone et al., 2015), and glutamate release in striatum, a neurochemical LID correlate (Dupre et al., 2011, Paolone et al., 2015), in 6-OHDA-hemilesioned, L-DOPA primed Rhes−/− mice and rapamycin-treated mice.

Section snippets

Subjects

In order to prevent that the presence of the PGK-neo cassette might interfere with normal transcriptional regulation within the Rhes locus, mice used in this work were crossed Rhes+/loxP-neo with offsprings (Spano et al., 2004) and subsequently with Cre deleter mice. The validation of Cre-mediated loxP-flanked neo cassette excision was assessed by PCR analysis using the following primers: Rhes-EGFP_forward: 5′-CATGGTCCTGCTGGAGTTCGTGA-3′, and Rhes-Ex2_reverse: 5′-ACCACCATGCGGTAGGAGTTCT-3′. The

Data presentation and statistical analysis

Motor performance was expressed as time on bar or rod (in seconds, bar and rotarod tests) and number of steps (drag test). AIMs severity was expressed as ALO AIMs score, or total ALO AIMs score (i.e. the sum of ALO scores in the different behavioral sessions). GABA and glutamate release was expressed as percentage ± SEM of basal values (calculated as mean of the two samples before treatment). Statistical analysis was performed by repeated measure one-way ANOVA followed by the Newman–Keuls test,

Genetic deletion of Rhes prevents LID development without causing primary hypolocomotion

To confirm the involvement of Rhes in LID (Subramaniam et al., 2012), Rhes−/− and Rhes+/+ mice were hemilesioned with 6-OHDA and chronically treated with L-DOPA. Mice were submitted to behavioral tests in order to evaluate differences in motor phenotype before and after 6-OHDA lesion. No differences between genotypes were observed in motor activity in the bar, drag and rotarod tests before 6-OHDA (Table 1). Unilateral intrastriatal injections of 6-OHDA caused a significant increase of

Discussion

Rhes is a striatal-enriched small GTP-binding protein, likely anchored to the plasma membrane by farnesylation (Falk et al., 1999). In rodent and human striatum, Rhes is virtually expressed in all D1R-expressing striato-nigral and D2 receptor (D2R) expressing striato-pallidal MSNs (Errico et al., 2008, Ghiglieri et al., 2015, Sciamanna et al., 2015) as well as in striatal cholinergic interneurons (Sciamanna et al., 2015). Rhes modulates receptor signaling via GPCR (Vargiu et al., 2004). In

Concluding remarks

Rhes deletion and mTORC1 blockade attenuated LID development in mice and prevented the LID-associated rise in nigral GABA and glutamate release in SNr, the basal ganglia output. This suggests that both manipulations prevent sensitization of striato-nigral MSNs to L-DOPA, confirming the pathogenic role of dysfunctional striatal mTORC1 activation along the aberrant D1 signaling cascade in MSNs (Santini et al., 2009, Santini et al., 2010). These data also confirm Rhes as an attractive target in

Acknowledgments

This work was supported by grants from the Italian Ministry of University [PRIN 2010–2011 #2010AHHP5H to M. Morari and A. Usiello].

References (55)

  • C.Y. Ostock

    Role of the primary motor cortex in L-Dopa-induced dyskinesia and its modulation by 5-HT1A receptor stimulation

    Neuropharmacology

    (2011)
  • N. Pavon

    ERK phosphorylation and FosB expression are associated with L-DOPA-induced dyskinesia in hemiparkinsonian mice

    Biol. Psychiatry

    (2006)
  • C. Rangel-Barajas

    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)
  • G. Rozas

    An automated rotarod method for quantitative drug-free evaluation of overall motor deficits in rat models of parkinsonism

    Brain Res. Brain Res. Protoc.

    (1997)
  • E. Santini

    Dopamine- and cAMP-regulated phosphoprotein of 32-kDa (DARPP-32)-dependent activation of extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin complex 1 (mTORC1) signaling in experimental parkinsonism

    J. Biol. Chem.

    (2012)
  • G. Sciamanna

    Rhes regulates dopamine D2 receptor transmission in striatal cholinergic interneurons

    Neurobiol. Dis.

    (2015)
  • V. Sgambato-Faure et al.

    Glutamatergic mechanisms in the dyskinesias induced by pharmacological dopamine replacement and deep brain stimulation for the treatment of Parkinson's disease

    Prog. Neurobiol.

    (2012)
  • S. Subramaniam

    Rhes, a physiologic regulator of sumoylation, enhances cross-sumoylation between the basic sumoylation enzymes E1 and Ubc9

    J. Biol. Chem.

    (2010)
  • S. Subramaniam et al.

    Huntington's disease is a disorder of the corpus striatum: focus on Rhes (Ras homologue enriched in the striatum)

    Neuropharmacology

    (2011)
  • P. Vargiu

    Thyroid hormone regulation of Rhes, a novel Ras homolog gene expressed in the striatum

    Brain Res. Mol. Brain Res.

    (2001)
  • R. Viaro

    Nociceptin/orphanin FQ receptor blockade attenuates MPTP-induced parkinsonism

    Neurobiol. Dis.

    (2008)
  • I. Ahmed

    Glutamate NMDA receptor dysregulation in Parkinson's disease with dyskinesias

    Brain

    (2011)
  • H.S. Bateup

    Distinct subclasses of medium spiny neurons differentially regulate striatal motor behaviors

    Proc. Natl. Acad. Sci. U. S. A.

    (2010)
  • S. Bido

    Amantadine attenuates levodopa-induced dyskinesia in mice and rats preventing the accompanying rise in nigral GABA levels

    J. Neurochem.

    (2011)
  • S. Bido

    Differential involvement of Ras-GRF1 and Ras-GRF2 in L-DOPA-induced dyskinesia

    Ann Clin Transl Neurol.

    (2015)
  • M.A. Cenci et al.

    Ratings of L-DOPA-induced dyskinesia in the unilateral 6-OHDA lesion model of Parkinson's disease in rats and mice

    Curr. Protoc. Neurosci.

    (2007)
  • M. Decressac et al.

    mTOR inhibition alleviates L-DOPA-induced dyskinesia in parkinsonian rats

    J Parkinsons Dis.

    (2013)
  • Cited by (20)

    • Striatal and nigral muscarinic type 1 and type 4 receptors modulate levodopa-induced dyskinesia and striato-nigral pathway activation in 6-hydroxydopamine hemilesioned rats

      2020, Neurobiology of Disease
      Citation Excerpt :

      This might be due to an interaction with extrastriatal M1 mAChRs and/or with striatal or extrastriatal non-M1 mAChRs. To further confirm an action along the striato-nigral direct pathway, intrastriatal telenzepine also reduced the LID-associated elevation of striatal Glu release (Brugnoli et al., 2016; Gardoni et al., 2018; Ostock et al., 2011; Paolone et al., 2015). In fact, elevation of striatal Glu release might result from stimulation of striato-nigral MSNs leading to activation of cortico-basal ganglia-thalamo-cortical loop (Mark et al., 2004; Marti et al., 2005) and cortico-striatal terminals (Ostock et al., 2011).

    • Striatal overexpression of β-arrestin2 counteracts L-dopa-induced dyskinesia in 6-hydroxydopamine lesioned Parkinson's disease rats

      2019, Neurochemistry International
      Citation Excerpt :

      ERK activation facilitates the expression of the FosB transcription factor and p-Ac-H3, which are also implicated in LID. In addition, dyskinesia is related to ERK-dependent activation of mTORC1, which is likely to accelerate local protein synthesis (Brugnoli et al., 2016). In summary, phosphor-DARPP-32 and ERK are both key markers in the D1R signaling pathway in LID that can reflect the activation of D1R sensitization.

    • The Thyroid Hormone-target Gene Rhes a Novel Crossroad for Neurological and Psychiatric Disorders: New Insights from Animal Models

      2018, Neuroscience
      Citation Excerpt :

      Interestingly, both in vitro and in vivo studies clearly showed that Rhes has the ability to bind and activate mTOR pathway (Subramaniam et al., 2011). Accordingly, lack of Rhes in 6-OHDA-lesioned KO mice, chronically treated with l-DOPA, was associated with a dramatic LID reduction and, importantly, prevented the LID-associated rise in nigral GABA and glutamate release in SNr (Subramaniam et al., 2011; Brugnoli et al., 2016). Overall, these data suggest that Rhes might represent an attractive target for LID therapy with fewer side effects than global mTOR inhibition, known to cause a significant immunosuppression (Dumont and Su, 1996; Picconi and Calabresi, 2012).

    • BDNF over-expression induces striatal serotonin fiber sprouting and increases the susceptibility to L-DOPA-induced dyskinesia in 6-OHDA-lesioned rats

      2017, Experimental Neurology
      Citation Excerpt :

      Here, sprouting of serotonin axons was measured in terms of SERT immunoreactivity, which is considered a reliable index of increased density of 5-HT-positive fibers or axonal varicosities (Bez et al., 2016; Descarries et al., 1995; Rylander et al., 2010). Moreover, based on the existence of a correlation between LID and D1 receptor-dependent cAMP/PKA and ERK/mTORC signaling pathways (Brugnoli et al., 2016; Decressac and Björklund, 2013; Santini et al., 2009b, 2007; Subramaniam et al., 2011), we performed biochemical analysis in order to clarify the molecular features underlying the behavioral effects observed in this study. Transfer plasmids carrying adeno-associated viral (AAV5) inverted terminal repeat coding for either rat BDNF or enhanced green fluorescent protein (GFP), downstream of a cytomegalovirus enhancer hybrid synthetic chicken β-actin (CBA) promoter, were generated.

    View all citing articles on Scopus
    View full text