Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180(7): 927-942, 2023 04.
Article in English | MEDLINE | ID: mdl-34767639

ABSTRACT

BACKGROUND AND PURPOSE: Regulator of G-protein signalling 4 (RGS4) is a signal transduction protein that accelerates intrinsic GTPase activity of Gαi/o and Gαq subunits, suppressing GPCR signalling. Here, we investigate whether RGS4 modulates nociceptin/orphanin FQ (N/OFQ) opioid (NOP) receptor signalling and if this modulation has relevance for l-Dopa-induced dyskinesia. EXPERIMENTAL APPROACH: HEK293T cells transfected with NOP, NOP/RGS4 or NOP/RGS19 were challenged with N/OFQ and the small-molecule NOP agonist AT-403, using D1-stimulated cAMP levels as a readout. Primary rat striatal neurons and adult mouse striatal slices were challenged with either N/OFQ or AT-403 in the presence of the experimental RGS4 chemical probe, CCG-203920, and D1-stimulated cAMP or phosphorylated extracellular signal regulated kinase 1/2 (pERK) responses were monitored. In vivo, CCG-203920 was co-administered with AT-403 and l-Dopa to 6-hydroxydopamine hemilesioned rats, and dyskinetic movements, striatal biochemical correlates of dyskinesia (pERK and pGluR1 levels) and striatal RGS4 levels were measured. KEY RESULTS: RGS4 expression reduced NOFQ and AT-403 potency and efficacy in HEK293T cells. CCG-203920 increased N/OFQ potency in primary rat striatal neurons and potentiated AT-403 response in mouse striatal slices. CCG-203920 enhanced AT-403-mediated inhibition of dyskinesia and its biochemical correlates, without compromising its motor-improving effects. Unilateral dopamine depletion caused bilateral reduction of RGS4 levels, which was reversed by l-Dopa. l-Dopa acutely up-regulated RGS4 in the lesioned striatum. CONCLUSIONS AND IMPLICATIONS: RGS4 physiologically inhibits NOP receptor signalling. CCG-203920 enhanced NOP responses and improved the antidyskinetic potential of NOP receptor agonists, mitigating the effects of striatal RGS4 up-regulation occurring during dyskinesia expression. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.


Subject(s)
Dyskinesia, Drug-Induced , Levodopa , Mice , Rats , Humans , Animals , Levodopa/pharmacology , Analgesics, Opioid , HEK293 Cells , Signal Transduction , Dyskinesia, Drug-Induced/drug therapy , Receptors, Opioid/metabolism , Nociceptin
2.
Neurobiol Dis ; 162: 105579, 2022 01.
Article in English | MEDLINE | ID: mdl-34871735

ABSTRACT

The G2019S mutation of LRRK2 represents a risk factor for idiopathic Parkinson's disease. Here, we investigate whether LRRK2 kinase activity regulates susceptibility to the environmental toxin 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP). G2019S knock-in mice (bearing enhanced kinase activity) showed greater nigro-striatal degeneration compared to LRRK2 knock-out, LRRK2 kinase-dead and wild-type mice following subacute MPTP treatment. LRRK2 kinase inhibitors PF-06447475 and MLi-2, tested under preventive or therapeutic treatments, protected against nigral dopamine cell loss in G2019S knock-in mice. MLi-2 also rescued striatal dopaminergic terminal degeneration in both G2019S knock-in and wild-type mice. Immunoblot analysis of LRRK2 Serine935 phosphorylation levels confirmed target engagement of LRRK2 inhibitors. However, MLi-2 abolished phosphoSerine935 levels in the striatum and midbrain of both wild-type and G2019S knock-in mice whereas PF-06447475 partly reduced phosphoSerine935 levels in the midbrain of both genotypes. In vivo and ex vivo uptake of the 18-kDa translocator protein (TSPO) ligand [18F]-VC701 revealed a similar TSPO binding in MPTP-treated wild-type and G2019S knock-in mice which was consistent with an increased GFAP striatal expression as revealed by Real Time PCR. We conclude that LRRK2 G2019S, likely through enhanced kinase activity, confers greater susceptibility to mitochondrial toxin-induced parkinsonism. LRRK2 kinase inhibitors are neuroprotective in this model.


Subject(s)
Parkinson Disease , Parkinsonian Disorders , Animals , Corpus Striatum/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mice , Mutation , Parkinson Disease/metabolism , Parkinsonian Disorders/metabolism , Phosphorylation
3.
Neurobiol Dis ; 144: 105044, 2020 10.
Article in English | MEDLINE | ID: mdl-32798726

ABSTRACT

Acetylcholine muscarinic receptors (mAChRs) contribute to both the facilitation and inhibition of levodopa-induced dyskinesia operated by striatal cholinergic interneurons, although the receptor subtypes involved remain elusive. Cholinergic afferents from the midbrain also innervate the substantia nigra reticulata, although the role of nigral mAChRs in levodopa-induced dyskinesia is unknown. Here, we investigate whether striatal and nigral M1 and/or M4 mAChRs modulate dyskinesia and the underlying striato-nigral GABAergic pathway activation in 6-hydroxydopamine hemilesioned rats. Reverse microdialysis allowed to deliver the mAChR antagonists telenzepine (M1 subtype preferring), PD-102807 and tropicamide (M4 subtype preferring), as well as the selective M4 mAChR positive allosteric modulator VU0152100 in striatum or substantia nigra, while levodopa was administered systemically. Dyskinetic movements were monitored along with nigral GABA (and glutamate) and striatal glutamate dialysate levels, taken as neurochemical correlates of striato-nigral pathway and cortico-basal ganglia-thalamo-cortical loop activation. We observed that intrastriatal telenzepine, PD-102807 and tropicamide alleviated dyskinesia and inhibited nigral GABA and striatal glutamate release. This was partially replicated by intrastriatal VU0152100. The M2 subtype preferring antagonist AFDX-116, used to elevate striatal acetylcholine levels, blocked the behavioral and neurochemical effects of PD-102807. Intranigral VU0152100 prevented levodopa-induced dyskinesia and its neurochemical correlates whereas PD-102807 was ineffective. These results suggest that striatal, likely postsynaptic, M1 mAChRs facilitate dyskinesia and striato-nigral pathway activation in vivo. Conversely, striatal M4 mAChRs can both facilitate and inhibit dyskinesia, possibly depending on their localization. Potentiation of striatal and nigral M4 mAChR transmission leads to powerful multilevel inhibition of striato-nigral pathway and attenuation of dyskinesia.


Subject(s)
Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Neostriatum/metabolism , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M4/metabolism , Substantia Nigra/metabolism , Allosteric Regulation , Animals , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Microdialysis , Muscarinic Antagonists/pharmacology , Neostriatum/drug effects , Neural Pathways , Oxidopamine/toxicity , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/etiology , Parkinsonian Disorders/metabolism , Rats , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M4/antagonists & inhibitors , Substantia Nigra/drug effects , Sympatholytics/toxicity , gamma-Aminobutyric Acid/drug effects , gamma-Aminobutyric Acid/metabolism
4.
Neuropharmacology ; 167: 108006, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32086070

ABSTRACT

To investigate whether the reversible MAO-B inhibitor and sodium channel blocker safinamide impairs glutamate release under parkinsonian conditions in vivo, and this effect is dependent on MAO-B inhibition, safinamide (and rasagiline as a comparator) were administered to 6-hydroxydopamine hemilesioned rats, a model of Parkinson's disease, and haloperidol-treated rats, a model of neuroleptic-induced parkinsonism. A microdialysis probe was implanted in the dopamine-depleted dorsolateral striatum, globus pallidus, subthalamic nucleus or substantia nigra reticulata of 6-hydroxydopamine hemilesioned rats. Glutamate and GABA release was stimulated by reverse dialysis of veratridine, and safinamide or rasagiline were acutely administered before veratridine at doses inhibiting MAO-B >50%. A microdialysis probe was implanted in the substantia nigra reticulata of naïve rats to monitor glutamate and GABA release following acute haloperidol and safinamide administration. Safinamide inhibited the veratridine-evoked glutamate release in the globus pallidus and subthalamic nucleus but not in the striatum and substantia nigra. Moreover, it reduced pallidal and nigral GABA release. Conversely, rasagiline failed to modify the veratridine-induced glutamate and GABA release in the basal ganglia. Safinamide also inhibited the haloperidol-induced nigral glutamate release. MAO-B inhibitors safinamide and rasagiline differ in their abilities to inhibit depolarization-evoked glutamate release in the basal ganglia of parkinsonian rats. The ineffectiveness of rasagiline suggests that MAO-B inhibition does not contribute to the antiglutamatergic activity of safinamide. The glutamate-inhibiting action of safinamide within the subthalamo-external pallidal loop, which shows abnormal activity in Parkinson's disease, might contribute to its therapeutic actions of improving motor performance without provoking troublesome dyskinesia.


Subject(s)
Alanine/analogs & derivatives , Benzylamines/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Glutamic Acid/metabolism , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Alanine/pharmacology , Alanine/therapeutic use , Animals , Benzylamines/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Male , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Rats , Rats, Sprague-Dawley
5.
J Med Chem ; 63(5): 2688-2704, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31951130

ABSTRACT

A novel series of C(3)-substituted piperdinylindoles were developed as nociceptin opioid receptor (NOP) partial agonists to explore a pharmacological hypothesis that NOP partial agonists would afford a dual pharmacological action of attenuating Parkinson's disease (PD) motor symptoms and development of levodopa-induced dyskinesias. SAR around the C-3 substituents investigated effects on NOP binding, intrinsic activity, and selectivity and showed that while the C(3)-substituted indoles are selective, high affinity NOP ligands, the steric, polar, and cationic nature of the C-3 substituents affected intrinsic activity to afford partial agonists with a range of efficacies. Compounds 4, 5, and 9 with agonist efficacies between 25% and 35% significantly attenuated motor deficits in the 6-OHDA-hemilesioned rat model of PD. Further, unlike NOP antagonists, which appear to worsen dyskinesia expression, these NOP partial agonists did not attenuate or worsen dyskinesia expression. The NOP partial agonists and their SAR reported here may be useful to develop nondopaminergic treatments for PD.


Subject(s)
Antiparkinson Agents/therapeutic use , Indoles/therapeutic use , Parkinson Disease/drug therapy , Receptors, Opioid/agonists , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/pharmacokinetics , Caco-2 Cells , Disease Models, Animal , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Male , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Opioid/metabolism , Structure-Activity Relationship , Nociceptin Receptor
6.
J Pharmacol Exp Ther ; 364(2): 198-206, 2018 02.
Article in English | MEDLINE | ID: mdl-29167350

ABSTRACT

Safinamide has been recently approved as an add-on to levodopa therapy for Parkinson disease. In addition to inhibiting monoamine oxidase type B, it blocks sodium channels and modulates glutamate (Glu) release in vitro. Since this property might contribute to the therapeutic action of the drug, we undertook the present study to investigate whether safinamide inhibits Glu release also in vivo and whether this effect is consistent across different brain areas and is selective for glutamatergic neurons. To this aim, in vivo microdialysis was used to monitor the spontaneous and veratridine-induced Glu and GABA release in the hippocampus and basal ganglia of naive, awake rats. Brain levels of safinamide were measured as well. To shed light on the mechanisms underlying the effect of safinamide, sodium currents were measured by patch-clamp recording in rat cortical neurons. Safinamide maximally inhibited the veratridine-induced Glu and GABA release in hippocampus at 15 mg/kg, which reached free brain concentrations of 1.89-1.37 µM. This dose attenuated veratridine-stimulated Glu (but not GABA) release in subthalamic nucleus, globus pallidus, and substantia nigra reticulata, but not in striatum. Safinamide was ineffective on spontaneous neurotransmitter release. In vitro, safinamide inhibited sodium channels, showing a greater affinity at depolarized (IC50 = 8 µM) than at resting (IC50 = 262 µM) potentials. We conclude that safinamide inhibits in vivo Glu release from stimulated nerve terminals, likely via blockade of sodium channels at subpopulations of neurons with specific firing patterns. These data are consistent with the anticonvulsant and antiparkinsonian actions of safinamide and provide support for the nondopaminergic mechanism of its action.


Subject(s)
Alanine/analogs & derivatives , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Benzylamines/pharmacology , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , gamma-Aminobutyric Acid/metabolism , Alanine/pharmacology , Animals , Basal Ganglia/cytology , Hippocampus/cytology , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley
7.
PLoS One ; 12(6): e0179082, 2017.
Article in English | MEDLINE | ID: mdl-28582422

ABSTRACT

Mutations in LRRK2 play a critical role in both familial and sporadic Parkinson's disease (PD). Up to date, the role of LRRK2 in PD onset and progression remains largely unknown. However, experimental evidence highlights a critical role of LRRK2 in the control of vesicle trafficking that in turn may regulate different aspects of neuronal physiology. We have analyzed the role of LRRK2 in regulating dopamine receptor D1 (DRD1) and D2 (DRD2) trafficking. DRD1 and DRD2 are the most abundant dopamine receptors in the brain. They differ in structural, pharmacological and biochemical properties, as well as in localization and internalization mechanisms. Our results indicate that disease-associated mutant G2019S LRRK2 impairs DRD1 internalization, leading to an alteration in signal transduction. Moreover, the mutant forms of LRRK2 affect receptor turnover by decreasing the rate of DRD2 trafficking from the Golgi complex to the cell membrane. Collectively, our findings are consistent with the conclusion that LRRK2 influences the motility of neuronal vesicles and the neuronal receptor trafficking. These findings have important implications for the complex role that LRRK2 plays in neuronal physiology and the possible pathological mechanisms that may lead to neuronal death in PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Neurons/metabolism , Parkinson Disease/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Knock-In Techniques , Golgi Apparatus/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Transport , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction
8.
Acta Neuropathol Commun ; 5(1): 22, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28292328

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic cause of Parkinson's disease. Here, we investigated whether the G2019S LRRK2 mutation causes morphological and/or functional changes at nigro-striatal dopamine neurons. Density of striatal dopaminergic terminals, nigral cell counts, tyrosine hydroxylase protein levels as well as exocytotic dopamine release measured in striatal synaptosomes, or striatal extracellular dopamine levels monitored by in vivo microdialysis were similar between ≥12-month-old G2019S knock-in mice and wild-type controls. In vivo striatal dopamine release was insensitive to the LRRK2 inhibitor Nov-LRRK2-11, and was elevated by the membrane dopamine transporter blocker GBR-12783. However, G2019S knock-in mice showed a blunted neurochemical and motor activation response to GBR-12783 compared to wild-type controls. Western blot and dopamine uptake analysis revealed an increase in dopamine transporter levels and activity in the striatum of 12-month-old G2019S KI mice. This phenotype correlated with a reduction in vesicular monoamine transporter 2 levels and an enhancement of vesicular dopamine uptake, which was consistent with greater resistance to reserpine-induced hypolocomotion. These changes were not observed in 3-month-old mice. Finally, Western blot analysis revealed no genotype difference in striatal levels of endogenous α-synuclein or α-synuclein bound to DOPAL (a toxic metabolite of dopamine). However, Serine129-phosphorylated α-synuclein levels were higher in 12-month-old G2019S knock-in mice. Immunohistochemistry confirmed this finding, also showing no genotype difference in 3-month-old mice. We conclude that the G2019S mutation causes progressive dysfunctions of dopamine transporters, along with Serine129-phosphorylated α-synuclein overload, at striatal dopaminergic terminals, which are not associated with dopamine homeostasis dysregulation or neuron loss but might contribute to intrinsic dopaminergic terminal vulnerability. We propose G2019S knock-in mice as a presymptomatic Parkinson's disease model, useful to investigate the pathogenic interaction among genetics, aging, and internal or environmental factors leading to the disease.


Subject(s)
Aging/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mutation , alpha-Synuclein/metabolism , Aging/pathology , Animals , Corpus Striatum/pathology , Dopamine Plasma Membrane Transport Proteins/metabolism , Gene Knock-In Techniques , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Phenotype , Phosphorylation , Prodromal Symptoms , Substantia Nigra/metabolism , Substantia Nigra/pathology , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Vesicular Monoamine Transport Proteins/metabolism , alpha-Synuclein/genetics
9.
Neuropsychopharmacology ; 42(7): 1491-1501, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28128334

ABSTRACT

Antipsychotic drugs are currently used in clinical practice for a variety of mental disorders. Among them, clozapine is the most effective medication for treatment-resistant schizophrenia and is most helpful in controlling aggression and the suicidal behavior in schizophrenia and schizoaffective disorder. Although clozapine is associated with a low likelihood of extrapyramidal symptoms and other neurological side effects, it is well known for the weight gain and metabolic side effects, which expose the patient to a greater risk of cardiovascular disorders and premature death, as well as psychosocial issues, leading to non-adherence to therapy. The mechanisms underlying these iatrogenic metabolic disorders are still controversial. We have therefore investigated the in vivo effects of the selective PKCß inhibitor, ruboxistaurin (LY-333531), in a preclinical model of long-term clozapine-induced weight gain. Cell biology, biochemistry, and behavioral tests have been performed in wild-type and PKCß knockout mice to investigate the contribution of endogenous PKCß and its pharmacological inhibition to the psychomotor effects of clozapine. Finally, we also shed light on a novel aspect of the mechanism underlying the clozapine-induced weight gain, demonstrating that the clozapine-dependent PKCß activation promotes the inhibition of the lipid droplet-selective autophagy process. This paves the way to new therapeutic approaches to this serious complication of clozapine therapy.


Subject(s)
Antipsychotic Agents/administration & dosage , Clozapine/administration & dosage , Protein Kinase C beta/antagonists & inhibitors , Protein Kinase C beta/deficiency , Weight Gain/drug effects , Animals , Antipsychotic Agents/toxicity , Cells, Cultured , Clozapine/toxicity , Drug Delivery Systems , Enzyme Inhibitors/administration & dosage , Indoles/administration & dosage , Male , Maleimides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors , Weight Gain/physiology
10.
Neurobiol Dis ; 85: 155-163, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26522958

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.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/metabolism , GTP-Binding Proteins/deficiency , Levodopa/toxicity , Multiprotein Complexes/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dyskinesia, Drug-Induced/pathology , Female , GTP-Binding Proteins/genetics , Glutamic Acid/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Multiprotein Complexes/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Sirolimus/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , TOR Serine-Threonine Kinases/metabolism
11.
Mov Disord ; 30(13): 1728-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26207892

ABSTRACT

BACKGROUND: Preclinical and clinical evidence that the serotonergic system plays a major role in levodopa-induced dyskinesias has been provided. Selective serotonin (5-hydroxytryptamine; 5-HT) 5-HT1A or 5-HT1B receptor agonists, and, very recently, the mixed 5-HT1A /5-HT1B receptor agonist, eltoprazine, proved effective in inhibiting L-dopa-induced dyskinesias in experimental animals and parkinsonian patients. Here, we investigate the mechanisms underlying this effect. METHODS: Microdialysis was employed in 6-hydroxydopamine-hemilesioned rats chronically treated with L-dopa alone or in combination with eltoprazine. Gamma-aminobutyric acid (GABA) and glutamate levels were monitored on L-dopa in the dopamine-depleted striatum and ipsilateral SNr. Motor activity on the rotarod was assessed, both off and on L-dopa. Western blot was used to quantify ex vivo striatal levels of phosphorylated extracellular signal-regulated kinase 1 and 2. Striatal and nigral amino acid levels, as well as striatal dopamine levels, were also monitored in L-dopa-primed dyskinetic rats acutely challenged with L-dopa and eltoprazine. RESULTS: Eltoprazine attenuated the development and expression of dyskinesias, preserving motor coordination on the rotarod. Eltoprazine prevented the rise of nigral amino acids and striatal glutamate levels, as well as the increase in striatal phosphorylated extracellular signal-regulated kinase 1 and 2, associated with dyskinesias. However, eltoprazine did not affect the L-dopa-induced increase in striatal dopamine. CONCLUSIONS: Eltoprazine inhibits the sensitization of striatonigral medium-sized GABA spiny neurons (the direct pathway) to L-dopa and their overactivation associated with dyskinesias appearance. Activation of 5-HT1A and 5-HT1B receptors regulating striatal glutamate transmission, but not striatal ectopic dopamine release, might underlie the symptomatic effect of eltoprazine.


Subject(s)
Antiparkinson Agents/adverse effects , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/prevention & control , Glutamic Acid/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Adrenergic Agents/pharmacology , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dyskinesia, Drug-Induced/etiology , Functional Laterality/drug effects , Gene Expression Regulation/drug effects , Levodopa/adverse effects , MAP Kinase Signaling System/drug effects , Male , Motor Activity/drug effects , Oxidopamine/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Rats , Rats, Sprague-Dawley , Serotonin Receptor Agonists/pharmacology , Serotonin Receptor Agonists/therapeutic use , Time Factors , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...