Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Neuroscience ; 536: 21-35, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-37952579

ABSTRACT

The primary motor cortex (M1) receives dopaminergic (DAergic) projections from the midbrain which play a key role in modulating motor and cognitive processes, such as motor skill learning. However, little is known at the level of individual neurons about how dopamine (DA) and its receptors modulate the intrinsic properties of the different neuronal subpopulations in M1 and if this modulation depends on age. Using immunohistochemistry, we first mapped the cells expressing the DA D1 receptor across the different layers in M1, and quantified the number of pyramidal neurons (PNs) expressing the D1 receptor in the different layers, in young and adult mice. This work reveals that the spatial distribution and the molecular profile of D1 receptor-expressing neurons (D1+) across M1 layers do not change with age. Then, combining whole-cell patch-clamp recordings and pharmacology, we explored ex vivo in young and adult mice the impact of activation or blockade of D1 receptors on D1+ PN intrinsic properties. While the bath application of the D1 receptor agonist induced an increase in the excitability of layer V PNs both in young and adult, we identified a distinct modulation of intrinsic electrical properties of layer V D1+ PNs by D1 receptor antagonist depending on the age of the animal.


Subject(s)
Dopamine Agonists , Motor Cortex , Rats , Mice , Animals , Dopamine Agonists/pharmacology , Rats, Sprague-Dawley , Dopamine/pharmacology , Pyramidal Cells/physiology , Receptors, Dopamine D1/metabolism , Prefrontal Cortex/metabolism
2.
Cell Rep ; 42(11): 113328, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37925641

ABSTRACT

The subthalamic nucleus (STN) is critical for behavioral control; its dysregulation consequently correlated with neurological and neuropsychiatric disorders, including Parkinson's disease. Deep brain stimulation (DBS) targeting the STN successfully alleviates parkinsonian motor symptoms. However, low mood and depression are affective side effects. STN is adjoined with para-STN, associated with appetitive and aversive behavior. DBS aimed at STN might unintentionally modulate para-STN, causing aversion. Alternatively, the STN mediates aversion. To investigate causality between STN and aversion, affective behavior is addressed using optogenetics in mice. Selective promoters allow dissociation of STN (e.g., Pitx2) vs. para-STN (Tac1). Acute photostimulation results in aversion via both STN and para-STN. However, only STN stimulation-paired cues cause conditioned avoidance and only STN stimulation interrupts on-going sugar self-administration. Electrophysiological recordings identify post-synaptic responses in pallidal neurons, and selective photostimulation of STN terminals in the ventral pallidum replicates STN-induced aversion. Identifying STN as a source of aversive learning contributes neurobiological underpinnings to emotional affect.


Subject(s)
Deep Brain Stimulation , Parkinson Disease , Parkinsonian Disorders , Subthalamic Nucleus , Animals , Mice , Avoidance Learning , Deep Brain Stimulation/methods , Parkinson Disease/therapy
3.
Curr Biol ; 33(22): 5011-5022.e6, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37879332

ABSTRACT

Repeated exposure to psychostimulants, such as amphetamine, causes a long-lasting enhancement in the behavioral responses to the drug, called behavioral sensitization.1 This phenomenon involves several neuronal systems and brain areas, among which the dorsal striatum plays a key role.2 The endocannabinoid system (ECS) has been proposed to participate in this effect, but the neuronal basis of this interaction has not been investigated.3 In the CNS, the ECS exerts its functions mainly acting through the cannabinoid type-1 (CB1) receptor, which is highly expressed at terminals of striatal medium spiny neurons (MSNs) belonging to both the direct and indirect pathways.4 In this study, we show that, although striatal CB1 receptors are not involved in the acute response to amphetamine, the behavioral sensitization and related synaptic changes require the activation of CB1 receptors specifically located at striatopallidal MSNs (indirect pathway). These results highlight a new mechanism of psychostimulant sensitization, a phenomenon that plays a key role in the health-threatening effects of these drugs.


Subject(s)
Cannabinoids , Central Nervous System Stimulants , Amphetamine/pharmacology , Amphetamine/metabolism , Receptors, Cannabinoid/metabolism , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/metabolism , Neurons/metabolism , Corpus Striatum/physiology , Endocannabinoids/pharmacology , Cannabinoids/pharmacology
4.
Cell Rep ; 42(10): 113287, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37843977

ABSTRACT

The activity of substantia nigra pars reticulata (SNr) neurons, the main output structure of basal ganglia, is altered in Parkinson's disease (PD). However, neither the underlying mechanisms nor the type of neurons responsible for PD-related motor dysfunctions have been elucidated yet. Here, we show that parvalbumin-expressing SNr neurons (SNr-PV+) occupy dorsolateral parts and possess specific electrophysiological properties compared with other SNr cells. We also report that only SNr-PV+ neurons' intrinsic excitability is reduced by downregulation of sodium leak channels in a PD mouse model. Interestingly, in anesthetized parkinsonian mice in vivo, SNr-PV+ neurons display a bursty pattern of activity dependent on glutamatergic tone. Finally, we demonstrate that chemogenetic inhibition of SNr-PV+ neurons is sufficient to alleviate motor impairments in parkinsonian mice. Overall, our findings establish cell-type-specific dysfunction in experimental parkinsonism in the SNr and provide a potential cellular therapeutic target to alleviate motor symptoms in PD.


Subject(s)
Parkinson Disease , Pars Reticulata , Mice , Animals , Substantia Nigra , Parvalbumins , Neurons/physiology
5.
Curr Neuropharmacol ; 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37475558

ABSTRACT

The external globus pallidus (GPe) is part of the basal ganglia circuit and plays a key role in controlling the actions. Although, many evidence indicate that dopamine through its activation of D2 receptors (D2Rs) modulates the GPe neuronal activity, the precise spatiomolecular characterization of cell populations expressing D2Rs in the mouse GPe is still lacking. By combining single molecule in situ hybridization, cell type-specific imaging analyses, and electrophysiology slice recordings, we found that GPe D2R cells are neurons preferentially localized in the caudal portion of GPe. These neu- rons comprising pallido-striatal, pallido-nigral, and pallido-cortical neurons segregate into two distinct populations displaying molecular and electrophysiological features of GPe GABAergic PV/NKX2.1 and cholinergic neurons respectively. By clarifying the spatial molecular identity of GPe D2R neurons in the mouse, this work provides the basis for future studies aiming at disentangling the action of do- pamine within the GPe.

6.
Curr Neuropharmacol ; 21(1): 22-30, 2023.
Article in English | MEDLINE | ID: mdl-35850655

ABSTRACT

The subthalamic nucleus (STN) is classically subdivided into sensori-motor, associative and limbic regions, which is consistent with the involvement of this structure in not only motor control, but also in cognitive and emotional tasks. However, the function of the sensory inputs to the STN's sensori-motor territory is comparatively less well explored, although sensory responses have been reported in this structure. There is still a paucity of information regarding the characteristics of that subdivision and its potential functional role in basal ganglia processing and more widely in associated networks. In this perspective paper, we summarize the type of sensory stimuli that have been reported to activate the STN, and describe the complex sensory properties of the STN and its anatomical link to a sensory network involving the brainstem, characterized in our recent work. Analyzing the sensory input to the STN led us to suggest the existence of previously unreported threelateral subcortical loops between the basal ganglia and the brainstem which do not involve the cortex. Anatomically, these loops closely link the STN, the substantia nigra pars reticulata and various structures from the brainstem such as the superior colliculus and the parabrachial nucleus. We also discuss the potential role of the STN in the control of sensory activity in the brainstem and its possible contribution to favoring sensory habituation or sensitization over brainstem structures to optimize the best selection of action at a given time.


Subject(s)
Subthalamic Nucleus , Humans , Subthalamic Nucleus/physiology , Basal Ganglia , Brain Stem
7.
Neurobiol Dis ; 167: 105674, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35245676

ABSTRACT

The primary motor cortex (M1) is crucial for movement execution, especially dexterous ones, but also for cognitive functions like motor learning. The acquisition of motor skills to execute dexterous movements requires dopamine-dependent and -independent plasticity mechanisms within M1. In addition to the basal ganglia, M1 is disturbed in Parkinson's disease (PD). However, little is known about how the lack of dopamine (DA), characteristic of PD, directly or indirectly impacts M1 circuitry. Here we review data from studies of PD patients and the substantial research in non-human primate and rodent models of DA depletion. These models enable us to understand the importance of DA in M1 physiology at the behavioral, network, cellular, and synaptic levels. We first summarize M1 functions and neuronal populations in mammals. We then look at the origin of M1 DA and the cellular location of its receptors and explore the impact of DA loss on M1 physiology, motor, and executive functions. Finally, we discuss how PD treatments impact M1 functions.


Subject(s)
Motor Cortex , Parkinson Disease , Animals , Basal Ganglia , Cognition , Dopamine , Humans , Mammals
8.
Neuron ; 109(9): 1513-1526.e11, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33770505

ABSTRACT

Recent advances in neuroscience have positioned brain circuits as key units in controlling behavior, implying that their positive or negative modulation necessarily leads to specific behavioral outcomes. However, emerging evidence suggests that the activation or inhibition of specific brain circuits can actually produce multimodal behavioral outcomes. This study shows that activation of a receptor at different subcellular locations in the same neuronal circuit can determine distinct behaviors. Pharmacological activation of type 1 cannabinoid (CB1) receptors in the striatonigral circuit elicits both antinociception and catalepsy in mice. The decrease in nociception depends on the activation of plasma membrane-residing CB1 receptors (pmCB1), leading to the inhibition of cytosolic PKA activity and substance P release. By contrast, mitochondrial-associated CB1 receptors (mtCB1) located at the same terminals mediate cannabinoid-induced catalepsy through the decrease in intra-mitochondrial PKA-dependent cellular respiration and synaptic transmission. Thus, subcellular-specific CB1 receptor signaling within striatonigral circuits determines multimodal control of behavior.


Subject(s)
Brain/metabolism , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/physiology , Synaptic Transmission/physiology , Animals , Brain/drug effects , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Catalepsy/chemically induced , Cell Membrane/metabolism , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nociception/drug effects , Nociception/physiology , Signal Transduction/drug effects , Synaptic Transmission/drug effects
9.
Curr Biol ; 31(4): 707-721.e7, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33306949

ABSTRACT

The basal ganglia (BG) inhibit movements through two independent circuits: the striatal neuron-indirect and the subthalamic nucleus-hyperdirect pathways. These pathways exert opposite effects onto external globus pallidus (GPe) neurons, whose functional importance as a relay has changed drastically with the discovery of two distinct cell types, namely the prototypic and the arkypallidal neurons. However, little is known about the synaptic connectivity scheme of different GPe neurons toward both motor-suppressing pathways, as well as how opposite changes in GPe neuronal activity relate to locomotion inhibition. Here, we optogenetically dissect the input organizations of prototypic and arkypallidal neurons and further define the circuit mechanism and behavioral outcome associated with activation of the indirect or hyperdirect pathways. This work reveals that arkypallidal neurons are part of a novel disynaptic feedback loop differentially recruited by the indirect or hyperdirect pathways and that broadcasts inhibitory control onto locomotion only when arkypallidal neurons increase their activity.


Subject(s)
Globus Pallidus/cytology , Locomotion/physiology , Neural Pathways , Synapses , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neurons , Optogenetics , Subthalamic Nucleus/cytology
10.
Cells ; 11(1)2021 12 23.
Article in English | MEDLINE | ID: mdl-35011592

ABSTRACT

Long-term exposition to morphine elicits structural and synaptic plasticity in reward-related regions of the brain, playing a critical role in addiction. However, morphine-induced neuroadaptations in the dorsal striatum have been poorly studied despite its key function in drug-related habit learning. Here, we show that prolonged treatment with morphine triggered the retraction of the dendritic arbor and the loss of dendritic spines in the dorsal striatal projection neurons (MSNs). In an attempt to extend previous findings, we also explored whether the dopamine D4 receptor (D4R) could modulate striatal morphine-induced plasticity. The combined treatment of morphine with the D4R agonist PD168,077 produced an expansion of the MSNs dendritic arbors and restored dendritic spine density. At the electrophysiological level, PD168,077 in combination with morphine altered the electrical properties of the MSNs and decreased their excitability. Finally, results from the sustantia nigra showed that PD168,077 counteracted morphine-induced upregulation of µ opioid receptors (MOR) in striatonigral projections and downregulation of G protein-gated inward rectifier K+ channels (GIRK1 and GIRK2) in dopaminergic cells. The present results highlight the key function of D4R modulating morphine-induced plasticity in the dorsal striatum. Thus, D4R could represent a valuable pharmacological target for the safety use of morphine in pain management.


Subject(s)
Corpus Striatum/physiology , Morphine/pharmacology , Neuronal Plasticity/physiology , Receptors, Dopamine D4/metabolism , Animals , Benzamides/pharmacology , Corpus Striatum/drug effects , Dendritic Spines/drug effects , Dendritic Spines/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Male , Morphine/administration & dosage , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Piperazines/pharmacology , Rats, Sprague-Dawley , Receptors, Dopamine D4/agonists , Receptors, Opioid, mu/metabolism
11.
Neurobiol Dis ; 145: 105076, 2020 11.
Article in English | MEDLINE | ID: mdl-32898646

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder that usually starts during midlife with progressive alterations of motor and cognitive functions. The disease is caused by a CAG repeat expansion within the huntingtin gene leading to severe striatal neurodegeneration. Recent studies conducted on pre-HD children highlight early striatal developmental alterations starting as soon as 6 years old, the earliest age assessed. These findings, in line with data from mouse models of HD, raise the questions of when during development do the first disease-related striatal alterations emerge and whether they contribute to the later appearance of the neurodegenerative features of the disease. In this review we will describe the different stages of striatal network development and then discuss recent evidence for its alterations in rodent models of the disease. We argue that a better understanding of the striatum's development should help in assessing aberrant neurodevelopmental processes linked to the HD mutation.


Subject(s)
Corpus Striatum/growth & development , Corpus Striatum/pathology , Huntington Disease/pathology , Neurogenesis/physiology , Animals , Humans , Mice
12.
Autophagy ; 16(12): 2289-2291, 2020 12.
Article in English | MEDLINE | ID: mdl-32981464

ABSTRACT

The recreational and medical use of cannabis is largely increasing worldwide. Cannabis use, however, can cause adverse side effects, so conducting innovative studies aimed to understand and potentially reduce cannabis-evoked harms is important. Previous research conducted on cultured neural cells had supported that CNR1/CB1R (cannabinoid receptor 1), the main molecular target of cannabis, affects macroautophagy/autophagy. However, it was not known whether CNR1 controls autophagy in the brain in vivo, and, eventually, what the functional consequences of a potential CNR1-autophagy connection could be. We have now found that Δ9-tetrahydrocannabinol (THC), the major intoxicating constituent of cannabis, impairs autophagy in the mouse striatum. Administration of autophagy activators (specifically, the rapalog temsirolimus and the disaccharide trehalose) rescues THC-induced autophagy inhibition and motor dyscoordination. The combination of various genetic strategies in vivo supports the idea that CNR1 molecules located on neurons belonging to the direct (striatonigral) pathway are required for the autophagy- and motor-impairing activity of THC. By identifying autophagy as a mechanistic link between THC and motor performance, our findings may open a new conceptual view on how cannabis acts in the brain.


Subject(s)
Cannabinoids , Animals , Autophagy , Brain , Dronabinol/pharmacology , Mice
13.
Elife ; 92020 08 10.
Article in English | MEDLINE | ID: mdl-32773031

ABSTRACT

The use of cannabis is rapidly expanding worldwide. Thus, innovative studies aimed to identify, understand and potentially reduce cannabis-evoked harms are warranted. Here, we found that Δ9-tetrahydrocannabinol, the psychoactive ingredient of cannabis, disrupts autophagy selectively in the striatum, a brain area that controls motor behavior, both in vitro and in vivo. Boosting autophagy, either pharmacologically (with temsirolimus) or by dietary intervention (with trehalose), rescued the Δ9-tetrahydrocannabinol-induced impairment of motor coordination in mice. The combination of conditional knockout mouse models and viral vector-mediated autophagy-modulating strategies in vivo showed that cannabinoid CB1 receptors located on neurons belonging to the direct (striatonigral) pathway are required for the motor-impairing activity of Δ9-tetrahydrocannabinol by inhibiting local autophagy. Taken together, these findings identify inhibition of autophagy as an unprecedented mechanistic link between cannabinoids and motor performance, and suggest that activators of autophagy might be considered as potential therapeutic tools to treat specific cannabinoid-evoked behavioral alterations.


Subject(s)
Autophagy/drug effects , Cannabinoids/pharmacology , Psychomotor Performance/drug effects , Putamen/physiology , Substantia Nigra/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Putamen/drug effects , Substantia Nigra/drug effects
14.
eNeuro ; 7(3)2020.
Article in English | MEDLINE | ID: mdl-32321772

ABSTRACT

Dopamine (DA) plays a crucial role in the control of motor and higher cognitive functions such as learning, working memory, and decision making. The primary motor cortex (M1), which is essential for motor control and the acquisition of motor skills, receives dopaminergic inputs in its superficial and deep layers from the midbrain. However, the precise action of DA and DA receptor subtypes on the cortical microcircuits of M1 remains poorly understood. The aim of this work was to investigate in mice how DA, through the activation of D2-like receptors (D2Rs), modulates the cellular and synaptic activity of M1 parvalbumin-expressing interneurons (PVINs) which are crucial to regulate the spike output of pyramidal neurons (PNs). By combining immunofluorescence, ex vivo electrophysiology, pharmacology and optogenetics approaches, we show that D2R activation increases neuronal excitability of PVINs and GABAergic synaptic transmission between PVINs and PNs in Layer V of M1. Our data reveal how cortical DA modulates M1 microcircuitry, which could be important in the acquisition of motor skills.


Subject(s)
Dopamine , Motor Cortex , Animals , Interneurons , Mice , Parvalbumins , Synaptic Transmission
15.
J Physiol ; 598(10): 1897-1927, 2020 05.
Article in English | MEDLINE | ID: mdl-32112413

ABSTRACT

KEY POINTS: Reciprocally connected GABAergic external globus pallidus (GPe) and glutamatergic subthalamic nucleus (STN) neurons form a key network within the basal ganglia. In Parkinson's disease and its models, abnormal rates and patterns of GPe-STN network activity are linked to motor dysfunction. Using cell class-specific optogenetic identification and inhibition during cortical slow-wave activity and activation, we report that, in dopamine-depleted mice, (1) D2 dopamine receptor expressing striatal projection neurons (D2-SPNs) discharge at higher rates, especially during cortical activation, (2) prototypic parvalbumin-expressing GPe neurons are excessively patterned by D2-SPNs even though their autonomous activity is upregulated, (3) despite being disinhibited, STN neurons are not hyperactive, and (4) STN activity opposes striatopallidal patterning. These data argue that in parkinsonian mice abnormal, temporally offset prototypic GPe and STN neuron firing results in part from increased striatopallidal transmission and that compensatory plasticity limits STN hyperactivity and cortical entrainment. ABSTRACT: Reciprocally connected GABAergic external globus pallidus (GPe) and glutamatergic subthalamic nucleus (STN) neurons form a key, centrally positioned network within the basal ganglia. In Parkinson's disease and its models, abnormal rates and patterns of GPe-STN network activity are linked to motor dysfunction. Following the loss of dopamine, the activities of GPe and STN neurons become more temporally offset and strongly correlated with cortical oscillations below 40 Hz. Previous studies utilized cortical slow-wave activity and/or cortical activation (ACT) under anaesthesia to probe the mechanisms underlying the normal and pathological patterning of basal ganglia activity. Here, we combined this approach with in vivo optogenetic inhibition to identify and interrupt the activity of D2 dopamine receptor-expressing striatal projection neurons (D2-SPNs), parvalbumin-expressing prototypic GPe (PV GPe) neurons, and STN neurons. We found that, in dopamine-depleted mice, (1) the firing rate of D2-SPNs was elevated, especially during cortical ACT, (2) abnormal phasic suppression of PV GPe neuron activity was ameliorated by optogenetic inhibition of coincident D2-SPN activity, (3) autonomous PV GPe neuron firing ex vivo was upregulated, presumably through homeostatic mechanisms, (4) STN neurons were not hyperactive, despite being disinhibited, (5) optogenetic inhibition of the STN exacerbated abnormal GPe activity, and (6) exaggerated beta band activity was not present in the cortex or GPe-STN network. Together with recent studies, these data suggest that in dopamine-depleted mice abnormally correlated and temporally offset PV GPe and STN neuron activity is generated in part by elevated striatopallidal transmission, while compensatory plasticity prevents STN hyperactivity and limits cortical entrainment.


Subject(s)
Globus Pallidus , Subthalamic Nucleus , Animals , Basal Ganglia , Dopamine , Mice , Neural Pathways , Neurons
16.
Cells ; 8(9)2019 08 29.
Article in English | MEDLINE | ID: mdl-31470672

ABSTRACT

The basal ganglia (BG) are a collection of interconnected subcortical nuclei that participate in a great variety of functions, ranging from motor programming and execution to procedural learning, cognition, and emotions. This network is also the region primarily affected by the degeneration of midbrain dopaminergic neurons localized in the substantia nigra pars compacta (SNc). This degeneration causes cellular and synaptic dysfunctions in the BG network, which are responsible for the appearance of the motor symptoms of Parkinson's disease. Dopamine (DA) modulation and the consequences of its loss on the striatal microcircuit have been extensively studied, and because of the discrete nature of DA innervation of other BG nuclei, its action outside the striatum has been considered negligible. However, there is a growing body of evidence supporting functional extrastriatal DA modulation of both cellular excitability and synaptic transmission. In this review, the functional relevance of DA modulation outside the striatum in both normal and pathological conditions will be discussed.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Globus Pallidus/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Synaptic Transmission , Animals , Dopaminergic Neurons/pathology , Globus Pallidus/pathology , Humans , Parkinson Disease/pathology , Substantia Nigra/pathology
17.
Sci Rep ; 8(1): 8858, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891970

ABSTRACT

Corticofugal fibers target the subthalamic nucleus (STN), a component nucleus of the basal ganglia, in addition to the striatum, their main input. The cortico-subthalamic, or hyperdirect, pathway, is thought to supplement the cortico-striatal pathways in order to interrupt/change planned actions. To explore the previously unknown properties of the neurons that project to the STN, retrograde and anterograde tools were used to specifically identify them in the motor cortex and selectively stimulate their synapses in the STN. The cortico-subthalamic neurons exhibited very little sag and fired an initial doublet followed by non-adapting action potentials. In the STN, AMPA/kainate synaptic currents had a voltage-dependent conductance, indicative of GluA2-lacking receptors and were partly inhibited by Naspm. AMPA transmission displayed short-term depression, with the exception of a limited bandpass in the 5 to 15 Hz range. AMPA synaptic currents were negatively controlled by dopamine D5 receptors. The reduction in synaptic strength was due to postsynaptic D5 receptors, mediated by a PKA-dependent pathway, but did not involve a modified rectification index. Our data indicated that dopamine, through post-synaptic D5 receptors, limited the cortical drive onto STN neurons in the normal brain.


Subject(s)
Dopamine/metabolism , Motor Cortex/metabolism , Neurons/metabolism , Receptors, Dopamine D5/physiology , Subthalamic Nucleus/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , Animals , Corpus Striatum/metabolism , Kainic Acid/metabolism , Mice, Inbred C57BL , Neural Pathways , Neurons/cytology , Synapses/metabolism , Synaptic Transmission
18.
Mov Disord ; 33(10): 1632-1642, 2018 10.
Article in English | MEDLINE | ID: mdl-29756234

ABSTRACT

Parkinson's disease motor symptoms are treated with levodopa, but long-term treatment leads to disabling dyskinesia. Altered synaptic transmission and maladaptive plasticity of corticostriatal glutamatergic projections play a critical role in the pathophysiology of dyskinesia. Because the noble gas xenon inhibits excitatory glutamatergic signaling, primarily through allosteric antagonism of the N-methyl-d-aspartate receptors, we aimed to test its putative antidyskinetic capabilities. We first studied the direct effect of xenon gas exposure on corticostriatal plasticity in a murine model of levodopa-induced dyskinesia We then studied the impact of xenon inhalation on behavioral dyskinetic manifestations in the gold-standard rat and primate models of PD and levodopa-induced dyskinesia. Last, we studied the effect of xenon inhalation on axial gait and posture deficits in a primate model of PD with levodopa-induced dyskinesia. This study shows that xenon gas exposure (1) normalized synaptic transmission and reversed maladaptive plasticity of corticostriatal glutamatergic projections associated with levodopa-induced dyskinesia, (2) ameliorated dyskinesia in rat and nonhuman primate models of PD and dyskinesia, and (3) improved gait performance in a nonhuman primate model of PD. These results pave the way for clinical testing of this unconventional but safe approach. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Parkinsonian Disorders/drug therapy , Xenon/therapeutic use , Administration, Inhalation , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/etiology , Gait Disorders, Neurologic/drug therapy , Gait Disorders, Neurologic/etiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , MPTP Poisoning/drug therapy , Mice , Mice, Transgenic , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/complications , Rats , Sensation Disorders/drug therapy , Sensation Disorders/etiology , Sympatholytics/toxicity , Time Factors
19.
Cell Rep ; 23(6): 1678-1690, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29742425

ABSTRACT

The external globus pallidus (GP) is a key GABAergic hub in the basal ganglia (BG) circuitry, a neuronal network involved in motor control. In Parkinson's disease (PD), the rate and pattern of activity of GP neurons are profoundly altered and contribute to the motor symptoms of the disease. In rodent models of PD, the striato-pallidal pathway is hyperactive, and extracellular GABA concentrations are abnormally elevated in the GP, supporting the hypothesis of an alteration of neuronal and/or glial clearance of GABA. Here, we discovered the existence of persistent GABAergic tonic inhibition in GP neurons of dopamine-depleted (DD) rodent models. We showed that glial GAT-3 transporters are downregulated while neuronal GAT-1 function remains normal in DD rodents. Finally, we showed that blocking GAT-3 activity in vivo alters the motor coordination of control rodents, suggesting that GABAergic tonic inhibition in the GP contributes to the pathophysiology of PD.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Globus Pallidus/pathology , Globus Pallidus/physiopathology , Neural Inhibition , Neurons/pathology , Parkinson Disease/physiopathology , Animals , Dopamine/deficiency , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Globus Pallidus/drug effects , Mice, Inbred C57BL , Motor Activity/drug effects , Neural Inhibition/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Receptors, GABA/metabolism , Synapses/drug effects , Synapses/metabolism , gamma-Aminobutyric Acid/pharmacology
20.
CNS Neurosci Ther ; 24(4): 292-300, 2018 04.
Article in English | MEDLINE | ID: mdl-29464851

ABSTRACT

Hereditary Huntington's disease (HD) is characterized by cell dysfunction and death in the brain, leading to progressive cognitive, psychiatric, and motor impairments. Despite molecular and cellular descriptions of the effects of the HD mutation, no effective pharmacological treatment is yet available. In addition to well-established alterations of glutamatergic and dopaminergic neurotransmitter systems, it is becoming clear that the GABAergic systems are also impaired in HD. GABA is the major inhibitory neurotransmitter in the brain, and GABAergic neurotransmission has been postulated to be modified in many neurological and psychiatric diseases. In addition, GABAergic neurotransmission is the target of many drugs that are in wide clinical use. Here, we summarize data demonstrating the occurrence of alterations of GABAergic markers in the brain of HD carriers as well as in rodent models of the disease. In particular, we pinpoint HD-related changes in the expression of GABAA receptors (GABAA Rs). On the basis that a novel GABA pharmacology of GABAA Rs established with more selective drugs is emerging, we argue that clinical treatments acting specifically on GABAergic neurotransmission may be an appropriate strategy for improving symptoms linked to the HD mutation.


Subject(s)
Huntington Disease/metabolism , Receptors, GABA-A/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Humans , Huntington Disease/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...