Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Sci Adv ; 7(2)2021 01.
Article in English | MEDLINE | ID: mdl-33523980

ABSTRACT

l-DOPA treatment for Parkinson's disease frequently leads to dyskinesias, the pathophysiology of which is poorly understood. We used MALDI-MSI to map the distribution of l-DOPA and monoaminergic pathways in brains of dyskinetic and nondyskinetic primates. We report elevated levels of l-DOPA, and its metabolite 3-O-methyldopa, in all measured brain regions of dyskinetic animals and increases in dopamine and metabolites in all regions analyzed except the striatum. In dyskinesia, dopamine levels correlated well with l-DOPA levels in extrastriatal regions, such as hippocampus, amygdala, bed nucleus of the stria terminalis, and cortical areas, but not in the striatum. Our results demonstrate that l-DOPA-induced dyskinesia is linked to a dysregulation of l-DOPA metabolism throughout the brain. The inability of extrastriatal brain areas to regulate the formation of dopamine during l-DOPA treatment introduces the potential of dopamine or even l-DOPA itself to modulate neuronal signaling widely across the brain, resulting in unwanted side effects.


Subject(s)
Dyskinesia, Drug-Induced , Levodopa , Animals , Brain/metabolism , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Mass Spectrometry
2.
J Neurosci ; 40(35): 6812-6819, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32690616

ABSTRACT

Parkinson's disease (PD) is characterized by severe locomotor deficits and is commonly treated with the dopamine precursor L-DOPA, but its prolonged usage causes dyskinesias referred to as L-DOPA-induced dyskinesia (LID). Several studies in animal models of PD have suggested that dyskinesias are associated with a heightened opioid cotransmitter tone, observations that have led to the notion of a LID-related hyperactive opioid transmission that should be corrected by µ opioid receptor antagonists. Reports that both antagonists and agonists of the µ opioid receptor may alleviate LID severity in primate models of PD and LID, together with the failure of nonspecific antagonist to improve LID in pilot clinical trials in patients, raises doubt about the reliability of the available data on the opioid system in PD and LID. After in vitro characterization of the functional activity at the µ opioid receptor, we selected prototypical agonists, antagonists, and partial agonists at the µ opioid receptor. We then showed that both oral and discrete intracerebral administration of a µ receptor agonist, but not of an antagonist as long thought, ameliorated LIDs in the gold-standard bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned female macaque model of PD and LID. The results call for a reappraisal of opioid pharmacology in the basal ganglia as well as for the development of brain nucleus-targeted µ opioid receptor agonists.SIGNIFICANCE STATEMENT µ opioid receptors have long been considered as a viable target for alleviating the severity of L-DOPA-induced hyperkinetic side effects, induced by the chronic treatment of Parkinson's disease motor symptoms with L-DOPA. Conflicting results between experimental parkinsonism and Parkinson's disease patients, however, dampened the enthusiasm for the target. Here we reappraise the pharmacology and then demonstrate that both oral and discrete intracerebral administration of a µ receptor agonist, but not of an antagonist as long thought, ameliorates LIDs in the gold-standard bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson's disease, calling for a reappraisal of the opioid pharmacology as well as for the development of brain nucleus-targeted µ receptor agonists.


Subject(s)
Dyskinesias/drug therapy , MPTP Poisoning/physiopathology , Receptors, Opioid, mu/agonists , Animals , CHO Cells , Cricetinae , Cricetulus , Dyskinesias/etiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use , Female , Humans , Levodopa/adverse effects , Levodopa/therapeutic use , MPTP Poisoning/drug therapy , Macaca fascicularis , Neurotransmitter Agents/administration & dosage , Neurotransmitter Agents/pharmacology , Neurotransmitter Agents/therapeutic use , Receptors, Opioid, mu/antagonists & inhibitors
3.
Mov Disord ; 33(6): 889-899, 2018 07.
Article in English | MEDLINE | ID: mdl-29488257

ABSTRACT

Understanding the biological mechanisms of l-dopa-induced motor complications is dependent on our ability to investigate these phenomena in animal models of Parkinson's disease. The most common motor complications consist in wearing-off fluctuations and abnormal involuntary movements appearing when plasma levels of l-dopa are high, commonly referred to as peak-dose l-dopa-induced dyskinesia. Parkinsonian models exhibiting these features have been well-characterized in both rodent and nonhuman primate species. The first animal models of peak-dose l-dopa-induced dyskinesia were produced in monkeys lesioned with N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and treated chronically with l-dopa to elicit choreic movements and dystonic postures. Seminal studies were performed in these models using both metabolic mapping and electrophysiological techniques, providing fundamental pathophysiological insights that have stood the test of time. A decade later, it was shown possible to reproduce peak-dose l-dopa-induced dyskinesia in rats and mice rendered parkinsonian with nigrostriatal 6-hydroxydopamine lesions. When treated with l-dopa, these animals exhibit abnormal involuntary movements having both hyperkinetic and dystonic components. These models have enabled molecular- and cellular-level investigations into the mechanisms of l-dopa-induced dyskinesia. A flourishing literature using genetically engineered mice is now unraveling the role of specific genes and neural circuits in the development of l-dopa-induced motor complications. Both non-human primate and rodent models of peak-dose l-dopa-induced dyskinesia have excellent construct validity and provide valuable tools for discovering therapeutic targets and evaluating potential treatments. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/etiology , Levodopa/adverse effects , Parkinson Disease, Secondary/drug therapy , Animals , Disease Models, Animal , Dyskinesia, Drug-Induced/pathology , Parkinson Disease, Secondary/chemically induced
4.
Mov Disord ; 31(8): 1120-1, 2016 08.
Article in English | MEDLINE | ID: mdl-27506637
5.
Prog Neurobiol ; 132: 96-168, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26209473

ABSTRACT

Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.


Subject(s)
Antiparkinson Agents/adverse effects , Central Nervous System/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Animals , Central Nervous System/drug effects , Humans , Parkinson Disease/drug therapy
6.
Neuron ; 84(4): 697-707, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25453841

ABSTRACT

Current neuroimaging techniques have very limited abilities to directly identify and quantify neurotransmitters from brain sections. We have developed a molecular-specific approach for the simultaneous imaging and quantitation of multiple neurotransmitters, precursors, and metabolites, such as tyrosine, tryptamine, tyramine, phenethylamine, dopamine, 3-methoxytyramine, serotonin, GABA, glutamate, acetylcholine, and L-alpha-glycerylphosphorylcholine, in histological tissue sections at high spatial resolutions. The method is employed to directly measure changes in the absolute and relative levels of neurotransmitters in specific brain structures in animal disease models and in response to drug treatments, demonstrating the power of mass spectrometry imaging in neuroscience.


Subject(s)
Brain/metabolism , Molecular Imaging/methods , Neurotransmitter Agents/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Mice , Rats
7.
Neurobiol Dis ; 70: 138-48, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24969021

ABSTRACT

Regulators of G-protein signalling (RGS) proteins are implicated in striatal G-protein coupled receptor (GPCR) sensitisation in the pathophysiology of l-DOPA-induced abnormal involuntary movements (AIMs), also known as dyskinesia (LID), in Parkinson's disease (PD). In this study, we investigated RGS protein subtype 4 in the expression of AIMs in the unilateral 6-hydroxydopamine (6-OHDA)-lesioned rat model of LID. The effects of RGS4 antisense brain infusion on the behavioural and molecular correlates of l-DOPA priming in 6-OHDA-lesioned rats were assessed. In situ hybridisation revealed that repeated l-DOPA/benserazide treatment caused an elevation of RGS4 mRNA levels in the striatum, predominantly in the lateral regions. The increased expression of RGS4 mRNA in the rostral striatum was found to positively correlate with the behavioural (AIM scores) and molecular (pre-proenkephalin B, PPE-B expression) markers of LID. We found that suppressing the elevation of RGS4 mRNA in the striatum by continuous infusion of RGS4 antisense oligonucleotides, via implanted osmotic mini-pumps, during l-DOPA priming, reduced the induction of AIMs. Moreover, ex vivo analyses of the rostral dorsolateral striatum showed that RGS4 antisense infusion attenuated l-DOPA-induced elevations of PPE-B mRNA and dopamine-stimulated [(35)S]GTPγS binding, a marker used for measuring dopamine receptor super-sensitivity. Taken together, these data suggest that (i) RGS4 proteins play an important pathophysiological role in the development and expression of LID and (ii) suppressing the elevation of RGS4 mRNA levels in l-DOPA priming attenuates the associated pathological changes in LID, dampening its physiological expression. Thus, modulating RGS4 proteins could prove beneficial in the treatment of dyskinesia in PD.


Subject(s)
Antiparkinson Agents/adverse effects , Corpus Striatum/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Parkinsonian Disorders/physiopathology , RGS Proteins/metabolism , Animals , Antiparkinson Agents/pharmacology , Cells, Cultured , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/therapy , Enkephalins/metabolism , Functional Laterality , Gene Expression/drug effects , Genetic Therapy , Levodopa/pharmacology , Male , Oligonucleotides, Antisense/administration & dosage , Oxidopamine , Parkinsonian Disorders/drug therapy , Protein Precursors/metabolism , RGS Proteins/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Up-Regulation/drug effects
8.
Parkinsonism Relat Disord ; 20(4): 452-5, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24521874

ABSTRACT

BACKGROUND: The antiepileptic drug topiramate reduces levodopa-induced dyskinesia without exacerbating parkinsonism in animal models. We report a randomized, double-blind, placebo-controlled crossover trial in patients with Parkinson's disease and levodopa-induced dyskinesia. METHODS: Fifteen patients with Parkinson's disease and stable levodopa-induced dyskinesia were enrolled into the study, of whom 13 were randomized to topiramate or placebo. The study medication was titrated to 100 mg/day over four weeks, and assessments were carried out after a further two weeks. Dyskinesia severity assessed by a blinded rater from video recordings was the primary outcome measure. RESULTS: Seven patients (mean age 58.9 ± 12.8 years) completed the study. Patients taking topiramate vs. placebo showed a significant increase in dyskinesia severity compared to baseline (Wilcoxon signed rank test, P = 0.043). Five patients withdrew from the study whilst taking topiramate due to adverse effects. CONCLUSIONS: Topiramate tended to worsen dyskinesia in patients with Parkinson's disease, and was poorly tolerated.


Subject(s)
Anticonvulsants/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , Fructose/analogs & derivatives , Parkinson Disease/drug therapy , Adult , Aged , Antiparkinson Agents/adverse effects , Cross-Over Studies , Double-Blind Method , Female , Fructose/therapeutic use , Humans , Levodopa/adverse effects , Male , Middle Aged , Topiramate
9.
Neurobiol Dis ; 62: 307-12, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24148855

ABSTRACT

A role for enhanced peptidergic transmission, either opioidergic or not, has been proposed for the generation of l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia (LID) on the basis of in situ hybridization studies showing that striatal peptidergic precursor expression consistently correlates with LID severity. Few studies, however, have focused on the actual peptides derived from these precursors. We used mass-spectrometry to study peptide profiles in the putamen and globus pallidus (internalis and externalis) collected from 1-methyl-4-phenyl-1,2,4,6-tetrahydropyridine treated macaque monkeys, acutely or chronically treated with l-DOPA. We identified that parkinsonian and dyskinetic states are associated with an abnormal production of proenkephalin-, prodynorphin- and protachykinin-1-derived peptides in both segments of the globus pallidus. Moreover, we report that peptidergic processing is dopamine-state dependent and highly structure-specific, possibly explaining the failure of previous clinical trials attempting to rectify abnormal peptidergic transmission.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/metabolism , Globus Pallidus/metabolism , Levodopa/toxicity , Neuropeptides/metabolism , Parkinsonian Disorders/metabolism , Putamen/chemistry , Animals , Enkephalins/analysis , Enkephalins/metabolism , Female , Globus Pallidus/chemistry , Macaca mulatta , Neuropeptides/analysis , Protein Precursors/analysis , Protein Precursors/metabolism , Putamen/metabolism , Tachykinins/analysis , Tachykinins/metabolism
10.
Exp Neurol ; 247: 476-84, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23360800

ABSTRACT

Abnormal corticostriatal plasticity is a key mechanism of L-DOPA-induced dyskinesia (LID) in Parkinson's disease (PD). Antagonists at glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, such as IEM 1460, reduce induction and expression of dyskinesia in rat and non-human primate models of PD. AMPA receptor function is regulated by post-transcriptional splicing of subunit mRNA to produce flip and flop isoforms, which may therefore influence corticostriatal plasticity. The aim of this work was to evaluate alterations in alternative splicing of striatal AMPA receptor subunits in the unilateral 6-hydroxydopamine (6-OHDA)-lesioned rat model of LID and PD. Male Sprague-Dawley rats received 12.5 µg 6-OHDA injections into the right medial forebrain bundle. In experiment 1, to assess acute dyskinesia, rats received L-DOPA/benserazide (6/15 mg/kg, i.p.) or vehicle for 21 days. In experiment 2, to assess dyskinesia priming, rats received vehicle, L-DOPA+vehicle or L-DOPA+IEM 1460 (3 mg/kg, i.p.) for 21 days. Animals were humanely killed 1h following final treatment in experiment 1, and 48 h following final treatment in experiment 2. Coronal sections of rostral striatum were processed for in situ hybridisation histochemistry, using oligonucleotide probes specific for the GluR1 and GluR2 subunits and their flip and flop isoforms. L-DOPA treatment increased GluR2-flip mRNA expression in the lesioned striatum of both groups; this was blocked by the Ca(2+)-permeable AMPA receptor antagonist IEM 1460. GluR1-flip expression was increased after 48 h drug washout but not in acute LID. There were no changes in expression of flop isoforms. Alternative splicing of AMPAR subunits contributes to abnormal striatal plasticity in the induction and expression of LID. Increases in GluR2-flip expression depend on activation of Ca(2+)-permeable AMPA receptors, which are a potential target of anti-dyskinetic therapies.


Subject(s)
Alternative Splicing/drug effects , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Parkinson Disease , Receptors, AMPA/metabolism , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Cocaine/analogs & derivatives , Cocaine/pharmacokinetics , Corpus Striatum/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Iodine Isotopes/pharmacokinetics , Male , Motor Activity/drug effects , Oxidopamine/toxicity , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sympatholytics/toxicity
11.
Mov Disord ; 26(13): 2354-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21953539

ABSTRACT

L-Dopa-induced dyskinesia in patients with Parkinson's disease can be alleviated by amantadine, an antagonist at N-methyl-D-aspartate glutamate receptors. The antiepileptic drug topiramate, which blocks α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, has also been shown to reduce dyskinesia. The purpose of this study was to examine the behavioral pharmacology of topiramate alone and in combination with amantadine in animal models of PD and L-dopa-induced dyskinesia. The effects of topiramate (5-20 mg/kg) and amantadine (5-20 mg/kg) on abnormal involuntary movements (the rat homologue of dyskinesia) and Rotarod performance were assessed alone and in combination in the 6-hydroxydopamine-lesioned rat following chronic L-dopa treatment. Dyskinesia, parkinsonian disability, and "on-time" were assessed in the MPTP-lesioned nonhuman primate following administration of topiramate (5-20 mg/kg) and amantadine (0.1-1.0 mg/kg) alone and in combination. Topiramate and amantadine dose-dependently reduced dyskinesia in the 6-hydroxydopamine-lesioned rat, whereas topiramate reduced Rotarod performance; there was no effect on parkinsonian disability in the MPTP-lesioned nonhuman primate, in which both drugs reduced dyskinesia. Topiramate and amantadine exhibited differential antidyskinetic effects on dyskinesia elicited by the dopamine D1 receptor agonist SKF 38393 (2 mg/kg). Subthreshold doses of both drugs in combination had a synergistic effect on dyskinesia in the 6-hydroxydopamine-lesioned rat, with no worsening of motor performance; this effect was confirmed in the MPTP-lesioned nonhuman primate, with a selective reduction in "bad on-time." These data confirm the antidyskinetic potential of topiramate and suggest that combination with low-dose amantadine may allow better reduction of dyskinesia with no adverse motor effects.


Subject(s)
Amantadine/pharmacology , Antiparkinson Agents/therapeutic use , Behavior, Animal/drug effects , Dyskinesia, Drug-Induced/drug therapy , Fructose/analogs & derivatives , MPTP Poisoning/drug therapy , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Amantadine/administration & dosage , Animals , Antiparkinson Agents/toxicity , Callithrix , Disease Models, Animal , Dopamine Agonists/administration & dosage , Dopamine Agonists/toxicity , Drug Synergism , Dyskinesia, Drug-Induced/diagnosis , Fructose/administration & dosage , Fructose/pharmacology , Levodopa/toxicity , Male , Rats , Rats, Sprague-Dawley , Topiramate
12.
J Neurochem ; 114(2): 499-511, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20456008

ABSTRACT

Overactivity of striatal alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors is implicated in the pathophysiology of L-DOPA-induced dyskinesia (LID) in Parkinson's disease (PD). In this study, we evaluated the behavioural and molecular effects of acute and chronic blockade of Ca(2+)-permeable AMPA receptors in animal models of PD and LID. The acute effects of the Ca(2+)-permeable AMPA receptor antagonist 1-trimethylammonio-5-(1-adamantane-methylammoniopentane) dibromide hydrobromide (IEM 1460) on abnormal involuntary movements (AIMs) in the 6-hydroxydopamine (6-OHDA)-lesioned rat and LID in the MPTP-lesioned non-human primate were assessed. Subsequently, the effects of chronic treatment of 6-OHDA-lesioned rats with vehicle, L-DOPA/benserazide (6/15 mg/kg, i.p.) + vehicle or L-DOPA + IEM 1460 (3 mg/kg, i.p.) on behavioural and molecular correlates of priming for LID were evaluated. In the 6-OHDA-lesioned rat and MPTP-lesioned non-human primate, acute treatment with IEM 1460 (1-3 mg/kg) dose-dependently reduced LID without adverse effects on motor performance. Chronic co-treatment for 21 days with IEM 1460 reduced the induction of AIMs by L-DOPA in the 6-OHDA-lesioned rat without affecting peak rotarod performance, and attenuated AIMs score by 75% following l-DOPA challenge (p < 0.05). Chronic IEM 1460 treatment reversed L-DOPA-induced up-regulation of pre-proenkephalin-A, and normalised pre-proenkephalin-B mRNA expression in the lateral striatum, indicating an inhibition of both behavioural and molecular correlates of priming. These data suggest that Ca(2+)-permeable AMPA receptors are critically involved in both the induction and subsequent expression of LID, and represent a potential target for anti-dyskinetic therapies.


Subject(s)
Antiparkinson Agents/adverse effects , Behavior, Animal/drug effects , Calcium/metabolism , Dyskinesia, Drug-Induced/psychology , Levodopa/adverse effects , Parkinson Disease/drug therapy , Receptors, AMPA/physiology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Benserazide/adverse effects , Brain/drug effects , Brain/metabolism , Callithrix , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/metabolism , Enkephalins/biosynthesis , Male , Motor Activity/drug effects , Oxidopamine , Parkinson Disease/etiology , Parkinson Disease/metabolism , Protein Precursors/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors
13.
PLoS One ; 3(2): e1589, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18270577

ABSTRACT

L-3,4-dihydroxypheylalanine (L-dopa)-induced dyskinesia represent a debilitating complication of therapy for Parkinson's disease (PD) that result from a progressive sensitization through repeated L-dopa exposures. The MPTP macaque model was used to study the proteome in dopamine-depleted striatum with and without subsequent acute and chronic L-dopa treatment using two-dimensional difference in-gel electrophoresis (2D-DIGE) and mass spectrometry. The present data suggest that the dopamine-depleted striatum is so sensitive to de novo L-dopa treatment that the first ever administration alone would be able (i) to induce rapid post-translational modification-based proteomic changes that are specific to this first exposure and (ii), possibly, lead to irreversible protein level changes that would be not further modified by chronic L-dopa treatment. The apparent equivalence between first and chronic L-dopa administration suggests that priming would be the direct consequence of dopamine loss, the first L-dopa administrations only exacerbating the sensitization process but not inducing it.


Subject(s)
Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Parkinson Disease/complications , Proteomics/methods , Animals , Corpus Striatum/chemistry , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Levodopa/administration & dosage , Macaca , Mass Spectrometry , Parkinson Disease/drug therapy , Protein Processing, Post-Translational , Rats
14.
Biol Psychiatry ; 61(7): 836-44, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-16950226

ABSTRACT

BACKGROUND: A role for enhanced opioid peptide transmission has been suggested in the genesis of levodopa-induced dyskinesia. However, basal ganglia nuclei other than the striatum have not been regarded as potential sources, and the opioid precursors have never been quantified simultaneously with the levels of opioid receptors at the peak of dyskinesia severity. METHODS: The levels of messenger RNA (mRNA) encoding the opioid precursors preproenkephalin-A and preproenkephalin-B in the striatum and the subthalamic nucleus and the levels of mu, delta, and kappa opioid receptors were measured within the basal ganglia of four groups of nonhuman primates killed at the peak of effect: normal, parkinsonian, parkinsonian chronically-treated with levodopa without exhibiting dyskinesia, and parkinsonian chronically-treated with levodopa showing overt dyskinesia. RESULTS: Dyskinesia are associated with reduction in opioid receptor binding and specifically of kappa and mu receptor binding in the globus pallidus internalis (GPi), the main output structure of the basal ganglia. This decrease was correlated with enhancement of the expression of preproenkephalin-B mRNA but not that of preproenkephalin-A in the striatum and the subthalamic nucleus. CONCLUSIONS: Abnormal transmission of preproenkephalin-B-derived opioid coming from the striatum and the subthalamic nucleus converges upon GPi at the peak of dose to induce levodopa-induced dyskinesia.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced , Enkephalins/metabolism , Gene Expression Regulation/physiology , Protein Precursors/metabolism , Receptors, Opioid/metabolism , Subthalamic Nucleus/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Analysis of Variance , Animals , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Drug Interactions , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Enkephalins/genetics , Female , Gene Expression Regulation/drug effects , In Situ Hybridization/methods , Levodopa/administration & dosage , Levodopa/adverse effects , Macaca fascicularis , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Protein Precursors/genetics , RNA, Messenger/metabolism , Radioligand Assay/methods , Regression Analysis
15.
Exp Neurol ; 203(2): 415-22, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17045989

ABSTRACT

Clinical DA agonist monotherapy trials, which used in vivo imaging of the DA transporter (DAT) to assess the rate of progression of nigrostriatal degeneration, have failed to demonstrate consistent evidence for neuroprotection. The present study aims at reconciling these experimental and clinical data by testing the protective property of the continuously delivered D3/D2/D1 dopamine receptor agonist rotigotine. Using a progressive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned (MPTP) macaque model that mimics the progression of Parkinson's disease in vivo ([99mTc]-TRODAT-1 single photon emission computed tomography (SPECT)) and ex vivo ([125I]-nortropane DAT labelling) endpoints were evaluated. After 38 days of treatment followed by two weeks of washout, rotigotine-treated animals were significantly less parkinsonian than the vehicle-treated ones. Such behavioural difference is the consequence of a partial protection of the DA terminals as could be confirmed by ex vivo DAT labelling. However, the protection of nerve terminals was not detected using SPECT. The data suggest that rotigotine exerts partial protection but that conventional imaging would not be able to identify such protection.


Subject(s)
Dopamine Agonists/therapeutic use , MPTP Poisoning/prevention & control , Parkinson Disease, Secondary/prevention & control , Tetrahydronaphthalenes/therapeutic use , Thiophenes/therapeutic use , Animals , Autoradiography , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Immunohistochemistry , MPTP Poisoning/diagnostic imaging , Macaca fascicularis , Neostriatum/diagnostic imaging , Neostriatum/metabolism , Organotechnetium Compounds , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/diagnostic imaging , Radiopharmaceuticals , Tomography, Emission-Computed, Single-Photon , Tropanes , Tyrosine 3-Monooxygenase/metabolism
16.
BMC Bioinformatics ; 7: 475, 2006 Oct 26.
Article in English | MEDLINE | ID: mdl-17067368

ABSTRACT

BACKGROUND: Two-Dimensional Difference In Gel Electrophoresis (2D-DIGE) is a powerful tool for measuring differences in protein expression between samples or conditions. However, to remove systematic variability within and between gels the data has to be normalized. In this study we examined the ability of four existing and four novel normalization methods to remove systematic bias in data produced with 2D-DIGE. We also propose a modification of an existing method where the statistical framework determines whether a set of proteins shows an association with the predefined phenotypes of interest. This method was applied to our data generated from a monkey model (Macaca fascicularis) of Parkinson's disease. RESULTS: Using 2D-DIGE we analysed the protein content of the striatum from 6 control and 21 MPTP-treated monkeys, with or without de novo or long-term L-DOPA administration. There was an intensity and spatial bias in the data of all the gels examined in this study. Only two of the eight normalization methods evaluated ('2D loess+scale' and 'SC-2D+quantile') successfully removed both the intensity and spatial bias. In 'SC-2D+quantile' we extended the commonly used loess normalization method against dye bias in two-channel microarray systems to suit systems with three or more channels.Further, by using the proposed method, Differential Expression in Predefined Proteins Sets (DEPPS), several sets of proteins associated with the priming effects of L-DOPA in the striatum in parkinsonian animals were identified. Three of these sets are proteins involved in energy metabolism and one set involved proteins which are part of the microtubule cytoskeleton. CONCLUSION: Comparison of the different methods leads to a series of methodological recommendations for the normalization and the analysis of data, depending on the experimental design. Due to the nature of 2D-DIGE data we recommend that the p-values obtained in significance tests should be used as rankings only. Individual proteins may be interesting as such, but by studying sets of proteins the interpretation of the results are probably more accurate and biologically informative.


Subject(s)
Corpus Striatum/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Gene Expression Profiling/methods , Nerve Tissue Proteins/metabolism , Parkinson Disease, Secondary/metabolism , Proteome/metabolism , Animals , Corpus Striatum/drug effects , Disease Models, Animal , Levodopa , Macaca fascicularis , Movement Disorders/etiology , Movement Disorders/metabolism , Parkinson Disease, Secondary/chemically induced
17.
Mov Disord ; 21(12): 2090-5, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16991143

ABSTRACT

L-dopa remains the most common treatment for Parkinson's disease. However, there is considerable interest in D3/D2 receptor agonists such as the novel agent S32504, since they exert antiparkinsonian properties in the absence of dyskinesia. An important question concerns the roles of D2 vs. D3 receptors, an issue we addressed with the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned nonhuman primate model of Parkinson's disease. In L-dopa-primed animals, S32504 (0.16-2.5 mg/kg p.o.) dose-dependently enhanced locomotor activity. This action was abolished by the D2 antagonist, L741,626 (2.5 mg/kg), but potentiated by the D3 antagonist, S33084 (0.63 mg/kg). Both antagonists were inactive alone. In drug-naive animals, a maximally effective dose of S32504 (2.5 mg/kg p.o.) displayed pronounced antiparkinsonian properties from the third day of administration, and its actions were expressed rapidly and durably. Thus, on day 33, antiparkinsonian properties of S32504 were apparent within 5 minutes and present for > 4 hours. Moreover, they were associated with neither wearing off nor significant dyskinesia. In conclusion, the novel D3/D2 agonist S32504 may offer advantages over L-dopa in the treatment of newly diagnosed parkinsonian patients. Its actions are expressed primarily by activation of D2, not D3, receptors.


Subject(s)
Antiparkinson Agents/therapeutic use , MPTP Poisoning/drug therapy , Oxazines/therapeutic use , Receptors, Dopamine D2/physiology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Callithrix , Disease Models, Animal , Dopamine Antagonists/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Indoles/therapeutic use , Levodopa/therapeutic use , MPTP Poisoning/physiopathology , Motor Activity/drug effects , Piperidines/therapeutic use , Receptors, Dopamine D3/physiology , Time Factors
18.
J Neurosci ; 26(34): 8653-61, 2006 Aug 23.
Article in English | MEDLINE | ID: mdl-16928853

ABSTRACT

The classic view of anatomofunctional organization of the basal ganglia is that striatopallidal neurons of the "indirect" pathway express D2 dopamine receptors and corelease enkephalin with GABA, whereas striatopallidal neurons of the "direct" pathway bear D1 dopamine receptors and corelease dynorphin and substance P with GABA. Although many studies have investigated the pathophysiology of the basal ganglia after dopamine denervation and subsequent chronic levodopa (L-dopa) treatment, none has ever considered the possibility of plastic changes leading to profound reorganization and/or biochemical phenotype modifications of medium spiny neurons. Therefore, we studied the phenotype of striatal neurons in four groups of nonhuman primates, including the following: normal, parkinsonian, parkinsonian chronically treated with L-dopa without exhibiting dyskinesia, and parkinsonian chronically treated with L-dopa exhibiting overt dyskinesia. To identify striatal cells projecting to external (indirect) or internal (direct) segments of the globus pallidus, the retrograde tracer cholera toxin subunit B (CTb) was injected stereotaxically into the terminal areas. Using immunohistochemistry techniques, brain sections were double labeled for CTb and dopamine receptors, opioid peptides, or the substance P receptor (NK1). We also used HPLC-RIA to assess opioid levels throughout structures of the basal ganglia. Our results suggest that medium spiny neurons retain their phenotype because no variations were observed in any experimental condition. Therefore, it appears unlikely that dyskinesia is related to a phenotype modification of the striatal neurons. However, this study supports the concept of axonal collateralization of striatofugal cells that project to both globus pallidus pars externa and globus pallidus pars interna. Striatofugal pathways are not as segregated in the primate as previously considered.


Subject(s)
Basal Ganglia/physiopathology , Corpus Striatum/physiopathology , Dyskinesias/physiopathology , Neurons , Parkinsonian Disorders/physiopathology , Animals , Cholera Toxin/administration & dosage , Cholera Toxin/pharmacokinetics , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dyskinesias/pathology , Female , Globus Pallidus/metabolism , Immunohistochemistry , Injections , Macaca fascicularis , Macaca mulatta , Neurons/metabolism , Opioid Peptides/metabolism , Parkinsonian Disorders/pathology , Phenotype , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Neurokinin-1/metabolism , Synaptic Transmission , Tissue Distribution
19.
FASEB J ; 19(9): 1140-2, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15894565

ABSTRACT

Endocannabinoids and cannabinoid CB1 receptors play a role in the control of movement by modulating GABA, glutamate, and other neurotransmitters throughout the basal ganglia. Roles for abnormalities in endocannabinoid signaling in Parkinson's disease (PD) and the major side effect of current treatments, levodopa-induced dyskinesia (LID), have been suggested by rodent studies. Here we show that signaling by endocannabinoids contributes to the pathophysiology of parkinsonism and LID in MPTP-lesioned, non-human primate models of Parkinson's disease. In MPTP-lesioned marmosets previously treated with levodopa to establish LID, attenuation of CB1 signaling by systemic administration of rimonabant (1 and 3 mg/kg) had anti-parkinsonian actions, equivalent to a 71% increase in motor activity at 3 mg/kg. Rimonabant did not elicit dyskinesia. Co-administration of levodopa (8 mg/kg) and rimonabant (1 and 3 mg/kg) resulted in significantly less dyskinesia than levodopa alone, without significantly affecting the anti-parkinsonian action of levodopa. These data suggest that enhanced endocannabinoid signaling may be involved in the pathophysiology of both parkinsonism and LID. To define potential mechanisms by which such a role might be mediated, we determined the levels of the endocannabinoids anandamide and 2-arachidonyl glycerol (2-AG) throughout the basal ganglia in normal and three groups of MPTP-lesioned cynomolgus monkeys (untreated; acutely treated with L-DOPA, non-dyskinetic; long-term treated, with levodopa-induced dyskinesia). In the untreated, MPTP-lesioned primate, parkinsonism was associated with increases in both 2-AG (+88%) and anandamide (+49%) in the striatum, and of 2-AG (+97%) in the substantia nigra, changes that are consistent with the previously suggested role for endocannabinoids in mechanisms attempting to compensate for loss of dopamine in untreated parkinsonism. Increased levels of anandamide (+34%) in the external globus pallidus of MPTP-lesioned animals were normalized by levodopa treatment and may contribute to the generation of parkinsonian symptoms. However, no clear alteration in endocannabinoid levels could be correlated with the expression of LID. These data highlight the potential roles played by endocannabinoids and CB1 in PD and LID and suggest the need for further research to pursue the multiple therapeutic opportunities for manipulating this system in movement disorders.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Dyskinesia, Drug-Induced/etiology , Endocannabinoids , Levodopa/toxicity , MPTP Poisoning/etiology , Animals , Arachidonic Acids/analysis , Callithrix , Cannabinoid Receptor Modulators/analysis , Female , Glycerides/analysis , MPTP Poisoning/drug therapy , Male , Piperidines/therapeutic use , Polyunsaturated Alkamides , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/physiology , Rimonabant , gamma-Aminobutyric Acid/metabolism
20.
J Neurosci ; 25(8): 2102-7, 2005 Feb 23.
Article in English | MEDLINE | ID: mdl-15728850

ABSTRACT

Dyskinesia represents a debilitating complication of L-3,4-dihydroxyphenylalanine (L-dopa) therapy for Parkinson's disease. Such motor manifestations are attributed to pathological activity in the motor parts of basal ganglia. However, because consistent funneling of information takes place between the sensorimotor, limbic, and associative basal ganglia domains, we hypothesized that nonmotor domains play a role in these manifestations. Here we report the changes in 2-deoxyglucose (2-DG) accumulation in the sensorimotor, limbic, and associative domains of basal ganglia and thalamic nuclei of four groups of nonhuman primates: normal, parkinsonian, parkinsonian chronically treated with L-dopa without exhibiting dyskinesia, and parkinsonian chronically treated with L-dopa and exhibiting overt dyskinesia. Although nondyskinetic animals display a rather normalized metabolic activity, dyskinetic animals are distinguished by significant changes in 2-DG accumulation in limbic- and associative-related structures and not simply in sensorimotor-related ones, suggesting that dyskinesia is linked to a pathological processing of limbic and cognitive information. We propose that these metabolic changes reflect the underlying neural mechanisms of not simply motor dyskinesias but also affective, motivational, and cognitive disorders associated with long-term exposure to L-dopa.


Subject(s)
Basal Ganglia/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/toxicity , Limbic System/physiopathology , Animals , Basal Ganglia/chemistry , Deoxyglucose/pharmacokinetics , Dyskinesia, Drug-Induced/metabolism , Female , Globus Pallidus/chemistry , Globus Pallidus/physiopathology , Levodopa/therapeutic use , Limbic System/chemistry , Macaca fascicularis , Motor Cortex/chemistry , Motor Cortex/physiopathology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Septal Nuclei/chemistry , Septal Nuclei/physiopathology , Somatosensory Cortex/chemistry , Somatosensory Cortex/physiopathology , Substantia Nigra/chemistry , Substantia Nigra/physiopathology , Subthalamic Nucleus/chemistry , Subthalamic Nucleus/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...