Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Curr Biol ; 34(5): 1034-1047.e4, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38377999

ABSTRACT

Dopaminergic neurons (DANs) in the substantia nigra pars compacta (SNc) have been related to movement speed, and loss of these neurons leads to bradykinesia in Parkinson's disease (PD). However, other aspects of movement vigor are also affected in PD; for example, movement sequences are typically shorter. However, the relationship between the activity of DANs and the length of movement sequences is unknown. We imaged activity of SNc DANs in mice trained in a freely moving operant task, which relies on individual forelimb sequences. We uncovered a similar proportion of SNc DANs increasing their activity before either ipsilateral or contralateral sequences. However, the magnitude of this activity was higher for contralateral actions and was related to contralateral but not ipsilateral sequence length. In contrast, the activity of reward-modulated DANs, largely distinct from those modulated by movement, was not lateralized. Finally, unilateral dopamine depletion impaired contralateral, but not ipsilateral, sequence length. These results indicate that movement-initiation DANs encode more than a general motivation signal and invigorate aspects of contralateral movements.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Mice , Animals , Dopaminergic Neurons/physiology , Substantia Nigra/physiology , Movement/physiology , Pars Compacta
2.
Neurobiol Dis ; 176: 105930, 2023 01.
Article in English | MEDLINE | ID: mdl-36414182

ABSTRACT

Levodopa (L-DOPA) administration remains the gold standard therapy for Parkinson's disease (PD). Despite several pharmacological advances in the use of L-DOPA, a high proportion of chronically treated patients continues to suffer disabling involuntary movements, namely, L-DOPA-induced dyskinesias (LIDs). As part of the effort to stop these unwanted side effects, the present study used a rodent model to identify and manipulate the striatal outflow circuitry responsible for LIDs. To do so, optogenetic technology was used to activate separately the striatal direct (D1R- expressing) and indirect (D2R- expressing) pathways in a mouse model of PD. Firstly, D1-cre or A2a-cre animals received unilateral injections of neurotoxin 6-hydroxydopamine (6-OHDA) to simulate the loss of dopamine observed in PD patients. The effects of independently stimulating each pathway were tested to see if experimental dyskinesias could be induced. Secondly, dopamine depleted A2a-cre animals received systemic L-DOPA to evoke dyskinetic movements. The ability of indirect pathway optogenetic stimulation to suppress pre-established LIDs was then tested. Selective manipulation of direct pathway evoked optodyskinesias both in dopamine depleted and intact animals, but optical inhibition of these neurons failed to suppress LIDs. On the other hand, selective activation of indirect striatal projection neurons produced an immediate and reliable suppression of LIDs. Thus, a functional dissociation has been found here whereby activation of D1R- and D2R-expressing projection neurons evokes and inhibits LIDs respectively, supporting the notion of tight interaction between the two striatal efferent systems in both normal and pathological conditions. This points to the importance of maintaining an equilibrium in the activity of both striatal pathways to produce normal movement. Finally, the ability of selective indirect pathway optogenetic activation to block the expression of LIDs in an animal model of PD sheds light on intrinsic mechanisms responsible for striatal-based dyskinesias and identifies a potential therapeutic target for suppressing LIDs in PD patients.


Subject(s)
Dyskinesias , Parkinson Disease , Mice , Animals , Levodopa/pharmacology , Dopamine/metabolism , Parkinson Disease/metabolism , Corpus Striatum/metabolism , Oxidopamine/toxicity , Antiparkinson Agents/pharmacology , Disease Models, Animal
3.
Neurobiol Dis ; 167: 105669, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35219857

ABSTRACT

Dopaminergic denervation in patients with Parkinson's disease is associated with changes in brain metabolism. Cerebral in-vivo mapping of glucose metabolism has been studied in severe stable parkinsonian monkeys, but data on brain metabolic changes in early stages of dopaminergic depletion of this model is lacking. Here, we report cerebral metabolic changes associated with progressive nigrostriatal lesion in the pre-symptomatic and symptomatic stages of the progressive 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkey model of Parkinson's Disease. Monkeys (Macaca fascicularis) received MPTP injections biweekly to induce progressive grades of dopamine depletion. Monkeys were sorted according to motor scale assessments in control, asymptomatic, recovered, mild, and severe parkinsonian groups. Dopaminergic depletion in the striatum and cerebral metabolic patterns across groups were studied in vivo by positron emission tomography (PET) using monoaminergic ([11C]-dihydrotetrabenazine; 11C-DTBZ) and metabolic (2-[18F]-fluoro-2-deoxy-d-glucose; 18F-FDG) radiotracers. 11C-DTBZ-PET analysis showed progressive decrease of binding potential values in the striatum of monkeys throughout MPTP administration and the development of parkinsonian signs. 18F-FDG analysis in asymptomatic and recovered animals showed significant hypometabolism in temporal and parietal areas of the cerebral cortex in association with moderate dopaminergic nigrostriatal depletion. Cortical hypometabolism extended to involve a larger area in mild parkinsonian monkeys, which also exhibited hypermetabolism in the globus pallidum pars interna and cerebellum. In severe parkinsonian monkeys, cortical hypometabolism extended further to lateral-frontal cortices and hypermetabolism also ensued in the thalamus and cerebellum. Unbiased histological quantification of neurons in Brodmann's area 7 in the parietal cortex did not reveal neuron loss in parkinsonian monkeys versus controls. Early dopaminergic nigrostriatal depletion is associated with cortical, mainly temporo-parietal hypometabolism unrelated to neuron loss. These findings, together with recent evidence from Parkinson's Disease patients, suggest that early cortical hypometabolism may be associated and driven by subcortical changes that need to be evaluated appropriately. Altogether, these findings could be relevant when potential disease modifying therapies become available.


Subject(s)
Parkinsonian Disorders , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Humans , Parkinsonian Disorders/metabolism , Positron-Emission Tomography/methods , Primates/metabolism
4.
Eur J Neurosci ; 53(7): 2398-2413, 2021 04.
Article in English | MEDLINE | ID: mdl-32394612

ABSTRACT

Dopamine replacement therapy with L-DOPA remains the most widely prescribed treatment for Parkinson disease. However, prolonged treatment due to disease progression frequently causes unwanted motor movements known as levodopa-induced dyskinesias. Previous studies have established that alterations to the efferent circuitry of the striatum, a principal component of the basal ganglia, are in part responsible for the pathological motor consequences of prolonged levodopa treatment. While the role of the striatal direct pathway is widely accepted, the engagement of the striatal indirect pathway in dyskinetic pathophysiology is still under consideration. However, recent investigations have finally shown that the activity of both striatal pathways changes as a consequence of dopamine depletion and dyskinetic behavioural conditions. In addition, it has been reported that drug-induced structural alterations to indirect pathway medium spiny neurons, together with associated changes in synaptic plasticity and firing patterns, could contribute importantly to the development of dyskinesia. These findings, together with recent opto- and chemogenetic studies, suggest that a critical imbalance in the activity between both striatal pathways is sufficient to cause dyskinesia in both rodent and primate models of Parkinson disease. In animal models, and in human patients, dyskinetic behaviours elicited by this efferent pathway imbalance can be achieved even in the absence of dopamine denervation. In this review, we summarize recent and past findings to better understand this complex pathology with the aim of pursuing specific cell-type therapies to re-balance efferent striatal activity.


Subject(s)
Dyskinesias , Levodopa , Animals , Antiparkinson Agents , Basal Ganglia , Corpus Striatum , Disease Models, Animal , Dopamine , Humans
5.
Trends Neurosci ; 42(6): 375-383, 2019 06.
Article in English | MEDLINE | ID: mdl-31053241

ABSTRACT

Humans can simultaneously combine automatic/habitual and voluntary/goal-directed aspects of behavioral control. Habitual routines permit us to perform well practiced task-components with minimal or no voluntary attention. Evidence from animal and human investigations indicates that dopaminergic neurons in lateral substantia nigra, which innervate the sensorimotor striatum, are engaged during the acquisition and performance of automatized skills and habits. Typically, in Parkinson disease (PD), there is a differential loss of dopamine, which occurs earliest and most severely in the caudal sensorimotor striatum, a subdivision of the striatum implicated in habitual control. We suggest that frequent reliance on habitual performance may be a critical functional stressor, which, when combined with other more general risk factors, could explain the selective neurodegeneration of the nigrostriatal motor projection in PD.


Subject(s)
Brain/physiopathology , Dopaminergic Neurons , Habits , Parkinson Disease/physiopathology , Animals , Humans
6.
Mov Disord ; 33(6): 867-876, 2018 07.
Article in English | MEDLINE | ID: mdl-29219207

ABSTRACT

Long-term levodopa (l-dopa) treatment in patients with Parkinson´s disease (PD) is associated with the development of motor complications (ie, motor fluctuations and dyskinesias). The principal etiopathogenic factors are the degree of nigro-striatal dopaminergic loss and the duration and dose of l-dopa treatment. In this review article we concentrate on analysis of the mechanisms underlying l-dopa-induced dyskinesias, a phenomenon that causes disability in a proportion of patients and that has not benefited from major therapeutic advances. Thus, we discuss the main neurotransmitters, receptors, and pathways that have been thought to play a role in l-dopa-induced dyskinesias from the perspective of basic neuroscience studies. Some important advances in deciphering the molecular pathways involved in these abnormal movements have occurred in recent years to reveal potential targets that could be used for therapeutic purposes. However, it has not been an easy road because there have been a plethora of components involved in the generation of these undesired movements, even bypassing the traditional and well-accepted dopamine receptor activation, as recently revealed by optogenetics. Here, we attempt to unify the available data with the hope of guiding and fostering future research in the field of striatal activation and abnormal movement generation. © 2017 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Corpus Striatum/physiopathology , Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Parkinson Disease/drug therapy , Animals , Humans
7.
Exp Neurol ; 298(Pt B): 148-161, 2017 12.
Article in English | MEDLINE | ID: mdl-28987461

ABSTRACT

The motor features of Parkinson's disease (PD) are well known to manifest only when striatal dopaminergic deficit reaches 60-70%. Thus, PD has a long pre-symptomatic and pre-motor evolution during which compensatory mechanisms take place to delay the clinical onset of disabling manifestations. Classic compensatory mechanisms have been attributed to changes and adjustments in the nigro-striatal system, such as increased neuronal activity in the substantia nigra pars compacta and enhanced dopamine synthesis and release in the striatum. However, it is not so clear currently that such changes occur early enough to account for the pre-symptomatic period. Other possible mechanisms relate to changes in basal ganglia and motor cortical circuits including the cerebellum. However, data from early PD patients are difficult to obtain as most studies have been carried out once the diagnosis and treatments have been established. Likewise, putative compensatory mechanisms taking place throughout disease evolution are nearly impossible to distinguish by themselves. Here, we review the evidence for the role of the best known and other possible compensatory mechanisms in PD. We also discuss the possibility that, although beneficial in practical terms, compensation could also play a deleterious role in disease progression.


Subject(s)
Adaptation, Physiological/physiology , Dopamine/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Animals , Cerebellum/metabolism , Disease Progression , Humans
10.
Front Neuroanat ; 9: 146, 2015.
Article in English | MEDLINE | ID: mdl-26635540

ABSTRACT

[This corrects the article on p. 99 in vol. 9, PMID: 26300740.].

12.
Front Cell Neurosci ; 9: 245, 2015.
Article in English | MEDLINE | ID: mdl-26217176

ABSTRACT

Levodopa-induced dyskinesias (LIDs) are major complications in the pharmacological management of Parkinson's disease (PD). Abnormal glutamatergic transmission in the striatum is considered a key factor in the development of LIDs. This work aims at: (i) characterizing N-methyl-D-aspartate (NMDA) receptor GluN2A/GluN2B subunit ratio as a common synaptic trait in rat and primate models of LIDs as well as in dyskinetic PD patients; and (ii) validating the potential therapeutic effect of a cell-permeable peptide (CPP) interfering with GluN2A synaptic localization on the dyskinetic behavior of these experimental models of LIDs. Here we demonstrate an altered ratio of synaptic GluN2A/GluN2B-containing NMDA receptors in the striatum of levodopa-treated dyskinetic rats and monkeys as well as in post-mortem tissue from dyskinetic PD patients. The modulation of synaptic NMDA receptor composition by a cell-permeable peptide interfering with GluN2A subunit interaction with the scaffolding protein postsynaptic density protein 95 (PSD-95) leads to a reduction in the dyskinetic motor behavior in the two animal models of LIDs. Our results indicate that targeting synaptic NMDA receptor subunit composition may represent an intriguing therapeutic approach aimed at ameliorating levodopa motor side effects.

13.
J Neurosci ; 33(11): 4782-95, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486949

ABSTRACT

Despite evidence that dopamine neurotransmission in the striatum is critical for learning as well as for movement control, little is yet known about how the learning-related dynamics of striatal activity are affected by dopamine depletion, a condition faced in Parkinson's disease. We made localized intrastriatal 6-hydroxydopamine lesions in rats and recorded within the dopamine-depleted sensorimotor striatal zone and its contralateral correspondent as the animals learned a conditional maze task. Rather than producing global, nonspecific elevations in firing rate across the task, the dopamine depletion altered striatal projection neuron activity and fast-spiking interneuron activity selectively, with sharply task-specific and cell type-specific effects, and often, with learning-stage selective effects as well. Striatal projection neurons with strong responses during the maze runs had especially elevated responsiveness during the maze runs. Projection neurons that, instead, fired most strongly before maze running showed elevated pre-start firing rates, but not during maze running, as learning progressed. The intrastriatal dopamine depletion severely affected the learning-related patterning of fast-spiking interneuron ensembles, especially during maze running and after extended training. Remarkably, L-DOPA treatment almost entirely reversed the depletion-induced elevations in pre-run firing of the projection neurons, and elevated their responses around start and end of maze runs. By contrast, L-DOPA failed to normalize fast-spiking interneuron activity. Thus the effects of striatal dopamine depletion and restoration on striatal activity are highly dependent not only on cell type, as previously shown, but also on the behavioral activity called for and the state of behavioral learning achieved.


Subject(s)
Action Potentials/drug effects , Corpus Striatum , Dopamine Agents/therapeutic use , Dopamine/deficiency , Learning Disabilities/drug therapy , Learning Disabilities/pathology , Levodopa/therapeutic use , Analysis of Variance , Animals , Animals, Newborn , Conditioning, Operant/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Electric Stimulation , Electrochemical Techniques , Learning Disabilities/chemically induced , Male , Maze Learning/drug effects , Neural Pathways/drug effects , Neural Pathways/physiology , Neurotoxins/toxicity , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Time Factors
14.
Mov Disord ; 28(2): 125, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23339064
15.
Proc Natl Acad Sci U S A ; 109(44): 18126-31, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23074253

ABSTRACT

A major physiologic sign in Parkinson disease is the occurrence of abnormal oscillations in cortico-basal ganglia circuits, which can be normalized by L-DOPA therapy. Under normal circumstances, oscillatory activity in these circuits is modulated as behaviors are learned and performed, but how dopamine depletion affects such modulation is not yet known. We here induced unilateral dopamine depletion in the sensorimotor striatum of rats and then recorded local field potential (LFP) activity in the dopamine-depleted region and its contralateral correspondent as we trained the rats on a conditional T-maze task. Unexpectedly, the dopamine depletion had little effect on oscillations recorded in the pretask baseline period. Instead, the depletion amplified oscillations across delta (~3 Hz), theta (~8 Hz), beta (~13 Hz), and low-gamma (~48 Hz) ranges selectively during task performance times when each frequency band was most strongly modulated, and only after extensive training had occurred. High-gamma activity (65-100 Hz), in contrast, was weakened independent of task time or learning stage. The depletion also increased spike-field coupling of fast-spiking interneurons to low-gamma oscillations. L-DOPA therapy normalized all of these effects except those at low gamma. Our findings suggest that the task-related and learning-related dynamics of LFP oscillations are the primary targets of dopamine depletion, resulting in overexpression of behaviorally relevant oscillations. L-DOPA normalizes these dynamics except at low-gamma, linked by spike-field coupling to fast-spiking interneurons, now known to undergo structural changes after dopamine depletion and to lack normalization of spike activity following l-DOPA therapy.


Subject(s)
Corpus Striatum/physiology , Dopamine/genetics , Levodopa/therapeutic use , Maze Learning , Action Potentials , Animals , Corpus Striatum/metabolism , Male , Rats , Rats, Sprague-Dawley , Task Performance and Analysis
16.
Mov Disord ; 27(14): 1739, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23401891
17.
PLoS One ; 5(11): e13861, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21085660

ABSTRACT

BACKGROUND: Dyskinesias associated with involuntary movements and painful muscle contractions are a common and severe complication of standard levodopa (L-DOPA, L-3,4-dihydroxyphenylalanine) therapy for Parkinson's disease. Pathologic neuroplasticity leading to hyper-responsive dopamine receptor signaling in the sensorimotor striatum is thought to underlie this currently untreatable condition. METHODOLOGY/PRINCIPAL FINDINGS: Quantitative real-time polymerase chain reaction (PCR) was employed to evaluate the molecular changes associated with L-DOPA-induced dyskinesias in Parkinson's disease. With this technique, we determined that thyrotropin releasing hormone (TRH) was greatly increased in the dopamine-depleted striatum of hemi-parkinsonian rats that developed abnormal movements in response to L-DOPA therapy, relative to the levels measured in the contralateral non-dopamine-depleted striatum, and in the striatum of non-dyskinetic control rats. ProTRH immunostaining suggested that TRH peptide levels were almost absent in the dopamine-depleted striatum of control rats that did not develop dyskinesias, but in the dyskinetic rats, proTRH immunostaining was dramatically up-regulated in the striatum, particularly in the sensorimotor striatum. This up-regulation of TRH peptide affected striatal medium spiny neurons of both the direct and indirect pathways, as well as neurons in striosomes. CONCLUSIONS/SIGNIFICANCE: TRH is not known to be a key striatal neuromodulator, but intrastriatal injection of TRH in experimental animals can induce abnormal movements, apparently through increasing dopamine release. Our finding of a dramatic and selective up-regulation of TRH expression in the sensorimotor striatum of dyskinetic rat models suggests a TRH-mediated regulatory mechanism that may underlie the pathologic neuroplasticity driving dopamine hyper-responsivity in Parkinson's disease.


Subject(s)
Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Parkinson Disease, Secondary/drug therapy , Thyrotropin-Releasing Hormone/metabolism , Analysis of Variance , Animals , Antiparkinson Agents/toxicity , Behavior, Animal/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/genetics , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin-Releasing Hormone/genetics
18.
Proc Natl Acad Sci U S A ; 106(8): 2892-6, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19171906

ABSTRACT

Voluntary movement difficulties in Parkinson's disease are initially relieved by l-DOPA therapy, but with disease progression, the repeated l-DOPA treatments can produce debilitating motor abnormalities known as l-DOPA-induced dyskinesias. We show here that 2 striatum-enriched regulators of the Ras/Rap/ERK MAP kinase signal transduction cascade, matrix-enriched CalDAG-GEFI and striosome-enriched CalDAG-GEFII (also known as RasGRP), are strongly and inversely dysregulated in proportion to the severity of abnormal movements induced by l-DOPA in a rat model of parkinsonism. In the dopamine-depleted striatum, the l-DOPA treatments produce down-regulation of CalDAG-GEFI and up-regulation of CalDAG-GEFII mRNAs and proteins, and quantification of the mRNA levels shows that these changes are closely correlated with the severity of the dyskinesias. As these CalDAG-GEFs control ERK cascades, which are implicated in l-DOPA-induced dyskinesias, and have differential compartmental expression patterns in the striatum, we suggest that they may be key molecules involved in the expression of the dyskinesias. They thus represent promising new therapeutic targets for limiting the motor complications induced by l-DOPA therapy.


Subject(s)
Antiparkinson Agents/adverse effects , DNA-Binding Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Motor Activity , Animals , DNA-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Immunohistochemistry , Male , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...