Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.464
Filter
Add more filters

Publication year range
1.
J Neurosci ; 41(12): 2668-2683, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33563724

ABSTRACT

l-3,4-dihydroxyphenylalanine (l-DOPA) is an effective treatment for Parkinson's disease (PD); however, long-term treatment induces l-DOPA-induced dyskinesia (LID). To elucidate its pathophysiology, we developed a mouse model of LID by daily administration of l-DOPA to PD male ICR mice treated with 6-hydroxydopamine (6-OHDA), and recorded the spontaneous and cortically evoked neuronal activity in the external segment of the globus pallidus (GPe) and substantia nigra pars reticulata (SNr), the connecting and output nuclei of the basal ganglia, respectively, in awake conditions. Spontaneous firing rates of GPe neurons were decreased in the dyskinesia-off state (≥24 h after l-DOPA injection) and increased in the dyskinesia-on state (20-100 min after l-DOPA injection while showing dyskinesia), while those of SNr neurons showed no significant changes. GPe and SNr neurons showed bursting activity and low-frequency oscillation in the PD, dyskinesia-off, and dyskinesia-on states. In the GPe, cortically evoked late excitation was increased in the PD and dyskinesia-off states but decreased in the dyskinesia-on state. In the SNr, cortically evoked inhibition was largely suppressed, and monophasic excitation became dominant in the PD state. Chronic l-DOPA treatment partially recovered inhibition and suppressed late excitation in the dyskinesia-off state. In the dyskinesia-on state, inhibition was further enhanced, and late excitation was largely suppressed. Cortically evoked inhibition and late excitation in the SNr are mediated by the cortico-striato-SNr direct and cortico-striato-GPe-subthalamo-SNr indirect pathways, respectively. Thus, in the dyskinesia-on state, signals through the direct pathway that release movements are enhanced, while signals through the indirect pathway that stop movements are suppressed, underlying LID.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is caused by progressive loss of midbrain dopaminergic neurons, characterized by tremor, rigidity, and akinesia, and estimated to affect around six million people world-wide. Dopamine replacement therapy is the gold standard for PD treatment; however, control of symptoms using l-3,4-dihydroxyphenylalanine (l-DOPA) becomes difficult over time because of abnormal involuntary movements (AIMs) known as l-DOPA-induced dyskinesia (LID), one of the major issues for advanced PD. Our electrophysiological data suggest that dynamic changes in the basal ganglia circuitry underlie LID; signals through the direct pathway that release movements are enhanced, while signals through the indirect pathway that stop movements are suppressed. These results will provide the rationale for the development of more effective treatments for LID.


Subject(s)
Basal Ganglia/physiopathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Dyskinesia, Drug-Induced/physiopathology , Levodopa/toxicity , Synaptic Transmission/physiology , Animals , Basal Ganglia/drug effects , Cerebral Cortex/drug effects , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Motor Activity/physiology , Synaptic Transmission/drug effects
2.
Neurobiol Dis ; 159: 105491, 2021 11.
Article in English | MEDLINE | ID: mdl-34461264

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative movement disorder that is routinely treated with levodopa. Unfortunately, long-term dopamine replacement therapy using levodopa leads to levodopa-induced dyskinesias (LID), a significant and disabling side-effect. Clinical findings indicate that LID typically only occurs following the progression of PD motor symptoms from the unilateral (Hoehn and Yahr (HY) Stage I) to the bilateral stage (HY Stage II). This suggests the presence of some compensatory interhemispheric mechanisms that delay the occurrence of LID. We therefore investigated the role of interhemispheric connections of the nigrostriatal pathway on LID expression in a rat model of PD. The striatum of one hemisphere of rats was first injected with a retrograde tracer to label the ipsi- and cross-hemispheric nigrostriatal pathways. Rats were then split into groups and unilaterally lesioned in the striatum or medial forebrain bundle of the tracer-injected hemisphere to induce varying levels of hemiparkinsonism. Finally, rats were treated with levodopa and tested for the expression of LID. Distinct subsets emerged from rats that underwent the same lesioning paradigm based on LID. Strikingly, non-dyskinetic rats had significant sparing of their cross-hemispheric nigrostriatal pathway projecting from the unlesioned hemisphere. In contrast, dyskinetic rats only had a small proportion of this cross-hemispheric nigrostriatal pathway survive lesioning. Crucially, both non-dyskinetic and dyskinetic rats had nearly identical levels of ipsi-hemispheric nigrostriatal pathway survival and parkinsonian motor deficits. Our data suggest that the survival of the cross-hemispheric nigrostriatal pathway plays a crucial role in preventing the expression of LID and represents a potentially novel target to halt the progression of this devastating side-effect of a common anti-PD therapeutic.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Neostriatum/physiology , Parkinsonian Disorders/physiopathology , Substantia Nigra/physiology , Animals , Disease Progression , Dyskinesia, Drug-Induced/etiology , Medial Forebrain Bundle/physiopathology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Rats , Sympatholytics/toxicity
3.
Neurobiol Dis ; 157: 105429, 2021 09.
Article in English | MEDLINE | ID: mdl-34153463

ABSTRACT

L-DOPA-induced dyskinesia (LID) is a significant complication of dopamine replacement therapy in Parkinson's disease (PD), and the specific role of different dopamine receptors in this disorder is poorly understood. We set out to compare patterns of dyskinetic behaviours induced by the systemic administration of L-DOPA and D1 or D2 receptor (D1R, D2R) agonists in mice with unilateral 6-hydroxydopamine lesions. Mice were divided in four groups to receive increasing doses of L-DOPA, a D1R agonist (SKF38393), a D2/3 agonist (quinpirole), or a selective D2R agonist (sumanirole). Axial, limb and orofacial abnormal involuntary movements (AIMs) were rated using a well-established method, while dystonic features were quantified in different body segments using a new rating scale. Measures of abnormal limb and trunk posturing were extracted from high-speed videos using a software for markerless pose estimation (DeepLabCut). While L-DOPA induced the full spectrum of dyskinesias already described in this mouse model, SKF38393 induced mostly orofacial and limb AIMs. By contrast, both of the D2-class agonists (quinpirole, sumanirole) induced predominantly axial AIMs. Dystonia ratings revealed that these agonists elicited marked dystonic features in trunk/neck, forelimbs, and hindlimbs, which were overall more severe in sumanirole-treated mice. Accordingly, sumanirole induced pronounced axial bending and hindlimb divergence in the automated video analysis. In animals treated with SKF38393, the only appreciable dystonic-like reaction consisted in sustained tail dorsiflexion and stiffness. We next compared the effects of D1R or D2R selective antagonists in L-DOPA-treated mice, where only the D2R antagonist had a significant effect on dystonic features. Taken together these results indicate that the dystonic components of LID are predominantly mediated by the D2R.


Subject(s)
Dyskinesia, Drug-Induced/physiopathology , Dystonia/physiopathology , Movement/drug effects , Parkinsonian Disorders/physiopathology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Antiparkinson Agents/adverse effects , Benzimidazoles/pharmacology , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/metabolism , Dystonia/chemically induced , Dystonia/metabolism , Mice , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Quinpirole/pharmacology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists
4.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R541-R546, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33533311

ABSTRACT

Physical exercise attenuates the development of l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia (LID) in 6-hydroxydopamine-induced hemiparkinsonian mice through unknown mechanisms. We now tested if exercise normalizes the aberrant corticostriatal neuroplasticity associated with experimental murine models of LID. C57BL/6 mice received two unilateral intrastriatal injections of 6-hydroxydopamine (12 µg) and were treated after 3 wk with l-DOPA/benserazide (25/12.5 mg/kg) for 4 wk, with individualized moderate-intensity running (60%-70% V̇o2peak) or not (untrained). l-DOPA converted the pattern of plasticity in corticostriatal synapses from a long-term depression (LTD) into a long-term potentiation (LTP). Exercise reduced LID severity and decreased aberrant LTP. These results suggest that exercise attenuates abnormal corticostriatal plasticity to decrease LID.


Subject(s)
Antiparkinson Agents/toxicity , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/prevention & control , Exercise Therapy , Levodopa/toxicity , Neuronal Plasticity/drug effects , Parkinsonian Disorders/drug therapy , Animals , Benserazide/toxicity , Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Dihydroxyphenylalanine/analogs & derivatives , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Running , Time Factors
5.
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
6.
Expert Opin Emerg Drugs ; 25(2): 131-144, 2020 06.
Article in English | MEDLINE | ID: mdl-32366130

ABSTRACT

INTRODUCTION: Prolonged treatment with L-3,4-dihydroxyphenylalanine (L-DOPA) leads to the development of uncontrolled movements (L-DOPA-induced dyskinesias (LID)) in Parkinson's disease (PD). There is currently only a single approved drug for the treatment of LID, a long-acting preparation of the NMDA antagonist, amantadine, that has variable benefits and side-effects. Therefore, new treatments for LID remain an unmet in PD. AREAS COVERED: We review the current strategies for the management of LID; the pathogenic mechanisms underlying the development of LID, which provides the rationale for clinical trials of novel targets for LID and provide a review of phase II/III trials for emerging drugs for LID, with either positive results, or ongoing studies, reported between January 2014 and December 2019. EXPERT OPINION: There are several ongoing studies for agents that showed possible benefit at phase Ib/IIa for reducing LID. However, there are no new positive phase III double-blind randomized controlled clinical trials (DBRCT) for emerging treatments for LID. Generating better preclinical models, more precise recruitment tools and better outcome measures remain a priority. The pharmacology of drugs investigated for LID may be too selective; therefore, evaluating combinations of drugs is worthy of consideration as is the repurposing of existing drugs with multiple pharmacological targets.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Amantadine/administration & dosage , Amantadine/pharmacology , Animals , Antiparkinson Agents/administration & dosage , Drug Development , Drug Repositioning , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Humans , Levodopa/administration & dosage , Parkinson Disease/drug therapy , Randomized Controlled Trials as Topic
7.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33374986

ABSTRACT

In previous work we evaluated an opioid glycopeptide with mixed µ/δ-opioid receptor agonism that was a congener of leu-enkephalin, MMP-2200. The glycopeptide analogue showed penetration of the blood-brain barrier (BBB) after systemic administration to rats, as well as profound central effects in models of Parkinson's disease (PD) and levodopa (L-DOPA)-induced dyskinesia (LID). In the present study, we tested the glycopeptide BBI-11008 with selective δ-opioid receptor agonism, an analogue of deltorphin, a peptide secreted from the skin of frogs (genus Phyllomedusa). We tested BBI-11008 for BBB-penetration after intraperitoneal (i.p.) injection and evaluated effects in LID rats. BBI-11008 (10 mg/kg) demonstrated good CNS-penetrance as shown by microdialysis and mass spectrometric analysis, with peak concentration levels of 150 pM in the striatum. While BBI-11008 at both 10 and 20 mg/kg produced no effect on levodopa-induced limb, axial and oral (LAO) abnormal involuntary movements (AIMs), it reduced the levodopa-induced locomotor AIMs by 50% after systemic injection. The N-methyl-D-aspartate receptor antagonist MK-801 reduced levodopa-induced LAO AIMs, but worsened PD symptoms in this model. Co-administration of MMP-2200 had been shown prior to block the MK-801-induced pro-Parkinsonian activity. Interestingly, BBI-11008 was not able to block the pro-Parkinsonian effect of MK-801 in the LID model, further indicating that a balance of mu- and delta-opioid agonism is required for this modulation. In summary, this study illustrates another example of meaningful BBB-penetration of a glycopeptide analogue of a peptide to achieve a central behavioral effect, providing additional evidence for the glycosylation technique as a method to harness therapeutic potential of peptides.


Subject(s)
Disease Models, Animal , Dyskinesia, Drug-Induced/physiopathology , Glycopeptides/pharmacology , Motor Activity/drug effects , Parkinson Disease, Secondary/physiopathology , Receptors, Opioid, delta/agonists , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/pharmacology , Animals , Corpus Striatum/metabolism , Dizocilpine Maleate/pharmacology , Dyskinesia, Drug-Induced/metabolism , Glycopeptides/administration & dosage , Glycopeptides/pharmacokinetics , Levodopa , Male , Motor Activity/physiology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism
8.
Neurobiol Dis ; 132: 104579, 2019 12.
Article in English | MEDLINE | ID: mdl-31445160

ABSTRACT

Dystonia and levodopa-induced dyskinesia (LID) are both hyperkinetic movement disorders. Dystonia arises most often spontaneously, although it may be seen after stroke, injury, or as a result of genetic causes. LID is associated with Parkinson's disease (PD), emerging as a consequence of chronic therapy with levodopa, and may be either dystonic or choreiform. LID and dystonia share important phenomenological properties and mechanisms. Both LID and dystonia are generated by an integrated circuit involving the cortex, basal ganglia, thalamus and cerebellum. They also share dysregulation of striatal cholinergic signaling and abnormalities of striatal synaptic plasticity. The long duration nature of both LID and dystonia suggests that there may be underlying epigenetic dysregulation as a proximate cause. While both may improve after interventions such as deep brain stimulation (DBS), neither currently has a satisfactory medical therapy, and many people are disabled by the symptoms of dystonia and LID. Further study of the fundamental mechanisms connecting these two disorders may lead to novel approaches to treatment or prevention.


Subject(s)
Brain/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Dystonic Disorders/physiopathology , Animals , Antiparkinson Agents/adverse effects , Humans , Levodopa/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology
9.
BMC Neurosci ; 20(1): 5, 2019 Feb 13.
Article in English | MEDLINE | ID: mdl-30760214

ABSTRACT

BACKGROUND: Accurately assessing promising therapeutic interventions for human diseases depends, in part, on the reproducibility of preclinical disease models. With the development of transgenic mice, the rapid adaptation of a 6-OHDA mouse model of Parkinson's disease that was originally described for the use in rats has come with a lack of a comprehensive characterization of lesion progression. In this study we therefore first characterised the time course of neurodegeneration in the substantia nigra pars compacta and striatum over a 4 week period following 6-OHDA injection into the medial forebrain bundle of mice. We then utilised the model to assess the anti-dyskinetic efficacy of recombinant activin A, a putative neuroprotectant and anti-inflammatory that is endogenously upregulated during the course of Parkinson's disease. RESULTS: We found that degeneration of fibers in the striatum was fully established within 1 week following 6-OHDA administration, but that the loss of neurons continued to progress over time, becoming fully established 3 weeks after the 6-OHDA injection. In assessing the anti-dyskinetic efficacy of activin A using this model we found that treatment with activin A did not significantly reduce the severity, or delay the time-of-onset, of dyskinesia. CONCLUSION: First, the current study concludes that a 3 week duration is required to establish a complete lesion of the nigrostriatal tract following 6-OHDA injection into the medial forebrain bundle of mice. Second, we found that activin A was not anti-dyskinetic in this model.


Subject(s)
Activins/pharmacology , Dyskinesia, Drug-Induced/drug therapy , Medial Forebrain Bundle/physiopathology , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Disease Progression , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Levodopa/pharmacology , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/pathology , Mice, Inbred C57BL , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Oxidopamine , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Random Allocation , Treatment Failure
10.
Mov Disord ; 34(6): 832-844, 2019 06.
Article in English | MEDLINE | ID: mdl-30759320

ABSTRACT

OBJECTIVE: Spreading depolarization (SD) is a transient self-propagating wave of neuronal and glial depolarization coupled with large membrane ionic changes and a subsequent depression of neuronal activity. Spreading depolarization in the cortex is implicated in migraine, stroke, and epilepsy. Conversely, spreading depolarization in the striatum, a brain structure deeply involved in motor control and in Parkinson's disease (PD) pathophysiology, has been poorly investigated. METHODS: We characterized the participation of glutamatergic and dopaminergic transmission in the induction of striatal spreading depolarization by using a novel approach combining optical imaging, measurements of endogenous DA levels, and pharmacological and molecular analyses. RESULTS: We found that striatal spreading depolarization requires the concomitant activation of D1-like DA and N-methyl-d-aspartate receptors, and it is reduced in experimental PD. Chronic l-dopa treatment, inducing dyskinesia in the parkinsonian condition, increases the occurrence and speed of propagation of striatal spreading depolarization, which has a direct impact on one of the signaling pathways downstream from the activation of D1 receptors. CONCLUSION: Striatal spreading depolarization might contribute to abnormal basal ganglia activity in the dyskinetic condition and represents a possible therapeutic target. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Corpus Striatum/physiopathology , Dopaminergic Neurons/physiology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/pharmacology , Neurons/physiology , Parkinsonian Disorders/physiopathology , Synaptic Transmission/physiology , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antiparkinson Agents/pharmacology , Corpus Striatum/drug effects , Nitrogen Mustard Compounds/metabolism , Prednisolone/metabolism , Procarbazine/metabolism , Rats , Rats, Wistar , Vincristine/metabolism
11.
J Neural Transm (Vienna) ; 126(6): 789-793, 2019 06.
Article in English | MEDLINE | ID: mdl-31098725

ABSTRACT

We tested in a translational approach the usefulness of plasma creatine kinase (CK) as an objective biomarker for levodopa-induced dyskinesia (LID). Plasma CK levels were measured in five dyskinetic parkinsonian non-human primates (NHP) and in ten PD patients with LID who participated in a treatment trial with simvastatin. Plasma CK levels were increased in dyskinetic NHP and correlated with LID severity while they were not affected by LID severity in PD patients.


Subject(s)
Antiparkinson Agents/adverse effects , Creatine Kinase/blood , Dyskinesia, Drug-Induced/blood , Levodopa/adverse effects , Parkinson Disease/drug therapy , Aged , Animals , Biomarkers/blood , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Humans , Macaca , Middle Aged , Randomized Controlled Trials as Topic , Severity of Illness Index
12.
J Neural Transm (Vienna) ; 126(4): 411-422, 2019 04.
Article in English | MEDLINE | ID: mdl-30937538

ABSTRACT

The striatum is richly innervated by mesencephalic dopaminergic neurons that modulate a diverse array of cellular and synaptic functions that control goal-directed actions and habits. The loss of this innervation has long been thought to be the principal cause of the cardinal motor symptoms of Parkinson's disease (PD). Moreover, chronic, pharmacological overstimulation of striatal dopamine (DA) receptors is generally viewed as the trigger for levodopa-induced dyskinesia (LID) in late-stage PD patients. Here, we discuss recent advances in our understanding of the relationship between the striatum and DA, particularly as it relates to PD and LID. First, it has become clear that chronic perturbations of DA levels in PD and LID bring about cell type-specific, homeostatic changes in spiny projection neurons (SPNs) that tend to normalize striatal activity. Second, perturbations in DA signaling also bring about non-homeostatic aberrations in synaptic plasticity that contribute to disease symptoms. Third, it has become evident that striatal interneurons are major determinants of network activity and behavior in PD and LID. Finally, recent work examining the activity of SPNs in freely moving animals has revealed that the pathophysiology induced by altered DA signaling is not limited to imbalance in the average spiking in direct and indirect pathways, but involves more nuanced disruptions of neuronal ensemble activity.


Subject(s)
Corpus Striatum/physiopathology , Dopamine/metabolism , Dyskinesia, Drug-Induced/physiopathology , Parkinson Disease/physiopathology , Animals , Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Dyskinesia, Drug-Induced/metabolism , Humans , Levodopa/adverse effects , Parkinson Disease/metabolism
13.
Neuroimage ; 183: 132-141, 2018 12.
Article in English | MEDLINE | ID: mdl-30102999

ABSTRACT

BACKGROUND: Dopaminergic and serotonergic degenerations alter pharmacological neurotransmission and structural markers in Parkinson's disease (PD). Alteration of diffusion measures in key brain regions depict MPTP/MDMA lesions in the monkey model of PD. Whether dopatherapy impacts such diffusion measures remains an open question. OBJECTIVES: The aim of this study was to investigate the consequences of l-DOPA treatment on diffusion alterations, PET imaging and immunohistochemical markers in MPTP/MDMA-intoxicated monkeys. METHODS: We acquired PET imaging and measures of mean diffusivity and fractional anisotropy longitudinally and correlated them with behavior and post-mortem fiber quantification. RESULTS: Severity of l-DOPA-induced dyskinesia was correlated to serotonin transporter radioligand binding increases in the ventral striatum and the anterior cingulate cortex and decreases of mean diffusivity in the ventral striatum. After lesion of serotonergic fibers by MDMA and the second l-DOPA period, diffusion measures were no more altered while the serotonergic binding still increased in all regions of interest, despite abolition of dyskinesia. Interestingly, in the anterior cingulate cortex, the SERT radioligand binding was negatively correlated to the number of SERT fibers. CONCLUSION: These results show that the increase of SERT radioligand binding is not systematically paralleled by an increase of SERT fibers and does not always reflect the presence of LID. More specifically, our study suggest that SERT increase may be underpinned by an increased density of serotonergic fibers after MPTP and the first l-DOPA period, and by an elevation of SERT itself after MDMA and the second l-DOPA period. This highlights that DTI is complementary to PET imaging to decipher pathophysiological mechanisms underlying l-DOPA-induced dyskinesia in a non-human primate model of PD.


Subject(s)
Brain , Dopamine Agents/pharmacology , Dyskinesia, Drug-Induced , Levodopa/pharmacology , Nerve Fibers , Parkinson Disease, Secondary , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Diffusion Tensor Imaging , Disease Models, Animal , Dyskinesia, Drug-Induced/diagnostic imaging , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Immunohistochemistry , MPTP Poisoning/diagnostic imaging , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Macaca fascicularis , Multimodal Imaging , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Nerve Fibers/drug effects , Nerve Fibers/pathology , Parkinson Disease, Secondary/diagnostic imaging , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Parkinson Disease, Secondary/physiopathology , Positron-Emission Tomography
14.
Neurobiol Dis ; 109(Pt A): 148-162, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29037828

ABSTRACT

The intricate balance between dopaminergic and cholinergic neurotransmission in the striatum has been thoroughly difficult to characterize. It was initially described as a seesaw with a competing function of dopamine versus acetylcholine. Recent technical advances however, have brought this view into question suggesting that the two systems work rather in concert with the cholinergic interneurons (ChIs) driving dopamine release. In this study, we have utilized two transgenic Cre-driver rat lines, a choline acetyl transferase ChAT-Cre transgenic rat and a novel double-transgenic tyrosine hydroxylase TH-Cre/ChAT-Cre rat to further elucidate the role of striatal ChIs in normal motor function and in Parkinson's disease. Here we show that selective and reversible activation of ChIs using chemogenetic (DREADD) receptors increases locomotor function in intact rats and potentiate the therapeutic effect of L-DOPA in the rats with lesions of the nigral dopamine system. However, the potentiation of the L-DOPA effect is accompanied by an aggravation of L-DOPA induced dyskinesias (LIDs). These LIDs appear to be driven primarily through the indirect striato-pallidal pathway since the same effect can be induced by the D2 agonist Quinpirole. Taken together, the results highlight the intricate regulation of balance between the two output pathways from the striatum orchestrated by the ChIs.


Subject(s)
Cholinergic Neurons/physiology , Corpus Striatum/physiology , Dopaminergic Neurons/physiology , Interneurons/physiology , Parkinson Disease/physiopathology , Animals , Choline O-Acetyltransferase/genetics , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/physiopathology , Female , Interneurons/cytology , Interneurons/metabolism , Levodopa/administration & dosage , Locomotion , Male , Parkinson Disease/metabolism , Rats, Long-Evans , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/physiology , Tyrosine 3-Monooxygenase/genetics
15.
J Neural Transm (Vienna) ; 125(8): 1157-1169, 2018 08.
Article in English | MEDLINE | ID: mdl-29704061

ABSTRACT

With the advent of rodent models of L-DOPA-induced dyskinesia (LID), a growing literature has linked molecular changes in the striatum to the development and expression of abnormal involuntary movements. Changes in information processing at the striatal level are assumed to impact on the activity of downstream basal ganglia nuclei, which in turn influence brain-wide networks, but very little is actually known about systems-level mechanisms of dyskinesia. As an aid to approach this topic, we here review the anatomical and physiological organisation of cortico-basal ganglia-thalamocortical circuits, and the changes affecting these circuits in animal models of parkinsonism and LID. We then review recent findings indicating that an abnormal cerebellar compensation plays a causal role in LID, and that structures outside of the classical motor circuits are implicated too. In summarizing the available data, we also propose hypotheses and identify important knowledge gaps worthy of further investigation. In addition to informing novel therapeutic approaches, the study of LID can provide new clues about the interplay between different brain circuits in the control of movement.


Subject(s)
Antiparkinson Agents/toxicity , Brain/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/toxicity , Neural Pathways/physiopathology , Animals
16.
J Neural Transm (Vienna) ; 125(8): 1263-1271, 2018 08.
Article in English | MEDLINE | ID: mdl-29492662

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive degeneration of dopaminergic neurons located in the midbrain. The gold-standard therapy for PD is the restoration of dopamine (DA) levels through the chronic administration of the DA precursor levodopa (L-DOPA). Although levodopa therapy is the main therapeutic approach for PD, its use is limited by the development of very disabling dyskinetic movements, mainly due to the fluctuation of DA cerebral content. Experimental animal models of PD identified in DA D1/ERK-signaling pathway aberrant activation, occurring in striatal projection neurons, coupled with structural spines abnormalities, the molecular and neuronal basis of L-DOPA-induced dyskinesia (LIDs) occurrence. Different electrophysiological approaches allowed the identification of  the alteration of homeostatic structural and synaptic changes, the neuronal bases of LIDs either in vivo in parkinsonian patients or in vitro in experimental animals. Here, we report the most recent studies showing electrophysiological and morphological evidence of aberrant synaptic plasticity in parkinsonian patients during LIDs in different basal ganglia nuclei and also in cortical transmission, accounting for the complexity of the synaptic changes during dyskinesias. All together, these studies suggest that LIDs are associated with a loss of homeostatic synaptic mechanisms.


Subject(s)
Corpus Striatum/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Neuronal Plasticity/physiology , Animals , Antiparkinson Agents/adverse effects , Humans , Levodopa/adverse effects
17.
J Neural Transm (Vienna) ; 125(8): 1145-1156, 2018 08.
Article in English | MEDLINE | ID: mdl-29502255

ABSTRACT

The MPTP monkey model of Parkinson's disease (PD) has allowed huge advances regarding the understanding of the pathological mechanisms of PD and L-DOPA-induced adverse effects. Among the main findings were the imbalance between the efferent striatal pathways in opposite directions between the hypokinetic and hyperkinetic states of PD. In both normal and parkinsonian monkeys, the combination of behavioral and anatomical studies has allowed the deciphering of the cortico-basal ganglia circuits involved in both movement and behavioral disorders. A major breakthrough has then been made regarding the hypothesis of the involvement of serotonergic fibers in the conversion of L-DOPA to dopamine when dopaminergic neurons are dying and to release it, in an uncontrolled manner, as serotonergic neurons are deprived from the machinery required for buffering dopamine from the synaptic cleft. The crucial involvement of serotonergic fibers underlying L-DOPA-induced dyskinesia (LID) has been demonstrated in both rodent and monkey models of PD, in which dyskinesia induced by L-DOPA is abolished following lesion of the serotonergic system. Moreover, the role of serotonergic fibers goes well beyond dyskinesia, as lesioning of such serotonergic fibers by MDMA in the monkey also decreased other L-DOPA-induced adverse effects such as impulsive compulsive behaviors and visual hallucinations. The same pathological mechanism, i.e., an imbalance between serotonin and dopamine terminals may, therefore, favor L-DOPA-induced adverse effects according to the basal ganglia territory it inhabits. Further non-human primate studies will be needed to demonstrate the role of such a pathological mechanism in both movement and behavioral disorders driven by L-DOPA therapy but also to determine the causal link between serotonin lesions and the expression of non-motor symptoms like apathy, depression and anxiety, frequently observed in PD patients.


Subject(s)
Dyskinesia, Drug-Induced/physiopathology , Mental Disorders/physiopathology , Parkinsonian Disorders/physiopathology , Serotonergic Neurons/physiology , Animals , Disease Models, Animal , Haplorhini
18.
J Neural Transm (Vienna) ; 125(8): 1273-1286, 2018 08.
Article in English | MEDLINE | ID: mdl-29396609

ABSTRACT

Adenosine is an endogenous purine nucleoside that regulates several physiological functions, at the central and peripheral levels. Besides, adenosine has emerged as a major player in the regulation of motor behavior. In fact, adenosine receptors of the A2A subtype are highly enriched in the caudate-putamen, which is richly innervated by dopamine. Moreover, several studies in experimental animals have consistently demonstrated that the pharmacological antagonism of A2A receptors has a facilitatory influence on motor behavior. Taken together, these findings have envisaged A2A receptors as a promising target for symptomatic therapies aimed at ameliorating motor deficits. Accordingly, A2A receptor antagonists have been extensively studied as new agents for the treatment of Parkinson's disease (PD), the epitome of motor disorders. In this review, we provide an overview of the effects that adenosine A2A receptor antagonists elicit in rodent and primate experimental models of PD, with regard to the counteraction of motor deficits as well as to manifestation of dyskinesia and motor fluctuations. Moreover, we briefly present the results of clinical trials of A2A receptor antagonists in PD patients experiencing motor fluctuations, with particular regard to dyskinesia. Finally, we discuss the interaction between A2A receptor antagonists and serotonin receptor agonists, since combined administration of these drugs has recently emerged as a new potential therapeutic strategy in the treatment of dyskinesia.


Subject(s)
Dyskinesia, Drug-Induced/metabolism , Parkinson Disease/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/physiopathology , Humans , Levodopa/adverse effects , Parkinson Disease/physiopathology
19.
J Neural Transm (Vienna) ; 125(8): 1195-1202, 2018 08.
Article in English | MEDLINE | ID: mdl-29480391

ABSTRACT

During the last decade, the serotonergic system has emerged as a key player in the appearance of L-DOPA-induced dyskinesia in animal models of Parkinson's disease. Clinical investigations, based on imaging and postmortem analyses, suggest that the serotonin neurons are also involved in the etiology of this complication of long-term L-DOPA treatment in parkinsonian patients. These findings have stimulated efforts to develop new therapies using drugs targeting the malfunctioning serotonin neurons. In this review, we summarize the experimental and clinical data obtained so far and discuss the prospects for further development of this therapeutic strategy.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Serotonergic Neurons/metabolism , Serotonergic Neurons/pathology , Animals , Dyskinesia, Drug-Induced/metabolism , Humans
20.
J Neural Transm (Vienna) ; 125(8): 1251-1262, 2018 08.
Article in English | MEDLINE | ID: mdl-29492663

ABSTRACT

Cholinergic signaling plays a key role in regulating striatal function. The principal source of acetylcholine in the striatum is the cholinergic interneurons which, although low in number, densely arborize to modulate striatal neurotransmission. This modulation occurs via strategically positioned nicotinic and muscarinic acetylcholine receptors that influence striatal dopamine, GABA and other neurotransmitter release. Cholinergic interneurons integrate multiple striatal synaptic inputs and outputs to regulate motor activity under normal physiological conditions. Consequently, an imbalance between these systems is associated with basal ganglia disorders. Here, we provide an overview of how striatal cholinergic interneurons modulate striatal activity under normal and pathological conditions. Numerous studies show that nigrostriatal damage such as that occurs with Parkinson's disease affects cholinergic receptor-mediated striatal activity. This altered cholinergic signaling is an important contributor to Parkinson's disease as well as to the dyskinesias that develop with L-dopa therapy, the gold standard for treatment. Indeed, multiple preclinical studies show that cholinergic receptor drugs may be beneficial for the treatment of L-dopa-induced dyskinesias. In this review, we discuss the evidence indicating that therapeutic modulation of the cholinergic system, particularly targeting of nicotinic cholinergic receptors, may offer a novel approach to manage this debilitating side effect of dopamine replacement therapy for Parkinson's disease.


Subject(s)
Corpus Striatum/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Interneurons/metabolism , Parkinson Disease/physiopathology , Animals , Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Humans , Levodopa/adverse effects , Parkinson Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL