Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neurosci Methods ; 339: 108728, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32289333

ABSTRACT

Although dystonia represents the third most common movement disorder, its pathophysiology remains still poorly understood. In the past two decades, multiple models have been generated, improving our knowledge on the molecular and cellular bases of this heterogeneous group of movement disorders. In this short survey, we will focus on recently generated novel models of DYT1 dystonia, the most common form of genetic, "isolated" dystonia. These models clearly indicate the existence of multiple signaling pathways affected by the protein mutation causative of DYT1 dystonia, torsinA, paving the way for potentially multiple, novel targets for pharmacological intervention.


Subject(s)
Dystonia , Dystonic Disorders , Movement Disorders , Dystonia/genetics , Dystonic Disorders/genetics , Humans , Mutation/genetics , Signal Transduction
2.
Rev Neurol (Paris) ; 174(9): 608-614, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30153948

ABSTRACT

Dystonia refers to a heterogeneous group of movement disorders characterized by involuntary, sustained muscle contractions leading to repetitive twisting movements and abnormal postures. A better understanding of the etiology, pathogenesis and molecular mechanisms underlying dystonia may be obtained from animal models. Indeed, while studies in vitro using cell and tissue models are helpful for investigating molecular pathways, animal models remain essential for studying the pathogenesis of these disorders and exploring new potential treatment strategies. To date, the mouse is the most common choice for mammalian models in most laboratories, particularly when manipulations of the genome are planned. Dystonia animal models can be classified into two categories, etiological and symptomatic, although neither is able to recapitulate all features of these disorders in humans. Nevertheless, etiological and symptomatic animal models have advantages and limitations that should be taken into consideration according to the specific proposed hypothesis and experimental goals. Etiological mouse models of inherited dystonia can reproduce the etiology of the disorder and help to reveal biochemical and cellular alterations, although a large majority of them lack motor symptoms. Conversely, symptomatic models can partially mimic the phenotype of human dystonia and test novel pharmacological agents, and also identify the anatomical and physiological processes involved, although the etiology remains unknown. Thus, our brief survey aims to review the state of the art as regards most of the commonly used animal models available for dystonia research.


Subject(s)
Dystonia/physiopathology , Dystonia/therapy , Animals , Disease Models, Animal , Dystonic Disorders , Humans , Species Specificity
3.
Neurobiol Dis ; 108: 128-139, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823931

ABSTRACT

Striatal dysfunction is implicated in many movement disorders. However, the precise nature of defects often remains uncharacterized, which hinders therapy development. Here we examined striatal function in a mouse model of the incurable movement disorder, myoclonus dystonia, caused by SGCE mutations. Using RNAseq we found surprisingly normal gene expression, including normal levels of neuronal subclass markers to strongly suggest that striatal microcircuitry is spared by the disease insult. We then functionally characterized Sgce mutant medium spiny projection neurons (MSNs) and cholinergic interneurons (ChIs). This revealed normal intrinsic electrophysiological properties and normal responses to basic excitatory and inhibitory neurotransmission. Nevertheless, high-frequency stimulation in Sgce mutants failed to induce normal long-term depression (LTD) at corticostriatal glutamatergic synapses. We also found that pharmacological manipulation of MSNs by inhibiting adenosine 2A receptors (A2AR) restores LTD, again pointing to structurally intact striatal circuitry. The fact that Sgce loss specifically inhibits LTD implicates this neurophysiological defect in myoclonus dystonia, and emphasizes that neurophysiological changes can occur in the absence of broad striatal dysfunction. Further, the positive effect of A2AR antagonists indicates that this drug class be tested in DYT11/SGCE dystonia.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Corpus Striatum/drug effects , Dystonic Disorders/drug therapy , Neuronal Plasticity/drug effects , Animals , Corpus Striatum/physiopathology , Disease Models, Animal , Dystonic Disorders/physiopathology , Female , Glutamic Acid/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , RNA, Messenger/metabolism , Receptor, Adenosine A2A/metabolism , Sarcoglycans/genetics , Sarcoglycans/metabolism , Tissue Culture Techniques
4.
Neuropharmacology ; 85: 440-50, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24951854

ABSTRACT

Early onset torsion dystonia (DYT1) is an autosomal dominantly inherited disorder caused by deletion in TOR1A gene. Evidence suggests that TOR1A mutation produces dystonia through an aberrant neuronal signalling within the striatum, where D2 dopamine receptors (D2R) produce an abnormal excitatory response in cholinergic interneurons (ChIs) in different models of DYT1 dystonia. The excitability of ChIs may be modulated by group I metabotropic glutamate receptor subtypes (mGlu1 and 5). We performed electrophysiological and calcium imaging recordings from ChIs of both knock-in mice heterozygous for Δ-torsinA (Tor1a(+/Δgag) mice) and transgenic mice overexpressing human torsinA (hMT1). We demonstrate that the novel negative allosteric modulator (NAM) of metabotropic glutamate 5 (mGlu) receptor, dipraglurant (ADX48621) counteracts the abnormal membrane responses and calcium rise induced either by the D2R agonist quinpirole or by caged dopamine (NPEC-Dopamine) in both models. These inhibitory effects were mimicked by two other well-characterized mGlu5 receptor antagonists, SIB1757 and MPEP, but not by mGlu1 antagonism. D2R and mGlu5 post-receptor signalling may converge on PI3K/Akt pathway. Interestingly, we found that the abnormal D2R response was prevented by the selective PI3K inhibitor, LY294002, whereas PLC and PKC inhibitors were both ineffective. Currently, no satisfactory pharmacological treatment is available for DYT1 dystonia patients. Our data show that negative modulation of mGlu5 receptors may counteract abnormal D2R responses, normalizing cholinergic cell excitability, by modulating the PI3K/Akt post-receptor pathway, thereby representing a novel potential treatment of DYT1 dystonia.


Subject(s)
Brain/drug effects , Corpus Striatum/drug effects , Dystonic Disorders/drug therapy , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Dopamine D2/metabolism , Animals , Brain/physiopathology , Calcium/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Corpus Striatum/physiology , Disease Models, Animal , Dystonic Disorders/physiopathology , Humans , Interneurons/drug effects , Interneurons/physiology , Mice, Transgenic , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Tissue Culture Techniques
5.
Neurobiol Dis ; 65: 124-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24503369

ABSTRACT

DYT1 dystonia is a movement disorder caused by a deletion in the C-terminal of the protein torsinA. It is unclear how torsinA mutation might disrupt cellular processes encoding motor activity, and whether this impairment occurs in specific brain regions. Here, we report a selective impairment of corticostriatal synaptic plasticity in knock-in mice heterozygous for Δ-torsinA (Tor1a(+/Δgag) mice) as compared to controls (Tor1a(+/+) mice). In striatal spiny neurons from Tor1a(+/Δgag) mice, high-frequency stimulation failed to induce long-term depression (LTD), whereas long-term potentiation (LTP) exhibited increased amplitude. Of interest, blockade of D2 dopamine receptors (D2Rs) increased LTP in Tor1a(+/+) mice to a level comparable to that measured in Tor1a(+/Δgag) mice and normalized the levels of potentiation across mouse groups. A low-frequency stimulation (LFS) protocol was unable to depotentiate corticostriatal synapses in Tor1a(+/Δgag) mice. Muscarinic M1 acetylcholine receptor (mAChR) blockade rescued plasticity deficits. Additionally, we found an abnormal responsiveness of cholinergic interneurons to D2R activation, consisting in an excitatory response rather than the expected inhibition, further confirming an imbalance between dopaminergic and cholinergic signaling in the striatum. Conversely, synaptic activity and plasticity in the CA1 hippocampal region were unaltered in Tor1a(+/Δgag) mice. Importantly, the M1 mAChR-dependent enhancement of hippocampal LTP was unaffected in both genotypes. Similarly, both basic properties of dopaminergic nigral neurons and their responses to D2R activation were normal. These results provide evidence for a regional specificity of the electrophysiological abnormalities observed and demonstrate the reproducibility of such alterations in distinct models of DYT1 dystonia.


Subject(s)
Brain/pathology , Dystonia/genetics , Dystonia/pathology , Molecular Chaperones/genetics , Neuronal Plasticity/genetics , Synapses/pathology , Animals , Disease Models, Animal , Dopamine/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/genetics , GABA Antagonists/pharmacology , Gene Expression Regulation/genetics , In Vitro Techniques , Mice , Mice, Transgenic , Muscarinic Antagonists/pharmacology , Mutation/genetics , Neurons/physiology , Picrotoxin/pharmacology , Pirenzepine/pharmacology , Synapses/genetics
6.
Neuropharmacology ; 75: 78-85, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23891638

ABSTRACT

Cholinergic interneurons (ChIs) of dorsal striatum play a key role in motor control and in behavioural learning. Neuropeptides regulate cholinergic transmission and mu opioid receptor (MOR) activation modulates striatal acetylcholine release. However, the mechanisms underlying this effect are yet uncharacterized. Here, we examined the electrophysiological responses of ChIs to the selective MOR agonist, DAMGO {[D-Ala2-MePhe4-Gly(ol)5] enkephalin}. We observed a robust, dose-dependent inhibition of spontaneous firing activity (0.06-3 µM) which was reversible upon drug washout and blocked by the selective antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) (1 µM). Voltage-clamp analysis of the reversal potential of the DAMGO effect did not provide univocal results, indicating the involvement of multiple membrane conductances. The MOR-dependent effect persisted in the presence of GABAA and ionotropic glutamate receptor antagonists, ruling out an indirect effect. Additionally, it depended upon G-protein activation, as it was prevented by intrapipette GDP-ß-S. Because D2 dopamine receptors (D2R) and MOR share a common post-receptor signalling pathway, occlusion experiments were performed with maximal doses of both D2R and MOR agonists. The D2R agonist quinpirole decreased spike discharge, which was further reduced by adding DAMGO. Then, D2R or MOR antagonists were used to challenge the response to the respective agonists, DAMGO or quinpirole. No cross-effect was observed, suggesting that the two receptors act independently. Our findings demonstrate a postsynaptic inhibitory modulation by MOR on ChIs excitability. Such opioidergic regulation of cholinergic transmission might contribute to shape information processing in basal ganglia circuits, and represent a potential target for pharmacological intervention.


Subject(s)
Action Potentials/physiology , Cholinergic Neurons/physiology , Corpus Striatum/cytology , Neural Inhibition/physiology , Receptors, Opioid, mu/metabolism , Action Potentials/drug effects , Analgesics, Opioid/pharmacology , Anesthetics, Local/pharmacology , Animals , Cadmium Chloride/pharmacology , Cholinergic Neurons/drug effects , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Excitatory Amino Acid Agents/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Neural Inhibition/drug effects , Quinpirole/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Tetrodotoxin/pharmacology
7.
Neuroscience ; 177: 240-51, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21195752

ABSTRACT

In the present work we analyzed the profile of high voltage-activated (HVA) calcium (Ca2+) currents in freshly isolated striatal medium spiny neurons (MSNs) from rodent models of both idiopathic and familial forms of Parkinson's disease (PD). MSNs were recorded from reserpine-treated and 6-hydroxydopamine (6-OHDA)-lesioned rats, and from DJ-1 and PINK1 (PTEN induced kinase 1) knockout (-/-) mice. Our analysis showed no significant changes in total HVA Ca2+ current. However, we recorded a net increase in the L-type fraction of HVA Ca2+ current in dopamine-depleted rats, and of both N- and P-type components in DJ-1-/- mice, whereas no significant change in Ca2+ current profile was observed in PINK1-/- mice. Dopamine modulates HVA Ca2+ channels in MSNs, thus we also analyzed the effect of D1 and D2 receptor activation. The effect of the D1 receptor agonist SKF 83822 on Ca2+ current was not significantly different among MSNs from control animals or PD models. However, in both dopamine-depleted rats and DJ-1-/- mice the D2 receptor agonist quinpirole inhibited a greater fraction of HVA Ca2+ current than in the respective controls. Conversely, in MSNs from PINK1-/- mice we did not observe alterations in the effect of D2 receptor activation. Additionally, in both reserpine-treated and 6-OHDA-lesioned rats, the effect of quinpirole was occluded by the selective L-type Ca2+ channel blocker nifedipine, while in DJ-1-/- mice it was mostly occluded by ω-conotoxin GVIA, blocker of N-type channels. These results demonstrate that both dopamine depletion and DJ-1 deletion induce a rearrangement in the HVA Ca2+ channel profile, specifically involving those channels that are selectively modulated by D2 receptors.


Subject(s)
Calcium Channels/metabolism , Dopamine/physiology , Neostriatum/metabolism , Neurons/metabolism , Oncogene Proteins/genetics , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Receptors, Dopamine D2/metabolism , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Disease Models, Animal , Dopamine/deficiency , Dopamine/genetics , Male , Mice , Mice, Knockout , Neostriatum/pathology , Neurons/drug effects , Neurons/pathology , Oncogene Proteins/deficiency , Organ Culture Techniques , Parkinsonian Disorders/genetics , Peroxiredoxins , Protein Deglycase DJ-1 , Rats , Rats, Wistar , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/deficiency
8.
Exp Neurol ; 215(2): 388-96, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19071114

ABSTRACT

An altered glutamatergic input at corticostriatal synapses has been shown in experimental models of Parkinson's disease (PD). In the present work, we analyzed the membrane and synaptic responses of striatal neurons to metabotropic glutamate (mGlu) receptor activation in two different mouse models of inherited PD, linked to mutations in PINK1 or Parkin genes. Both in PINK1 and Parkin knockout ((-/-)) mice, activation of group I mGlu receptors by 3,5-DHPG caused a membrane depolarization coupled to an increase in firing frequency in striatal cholinergic interneurons that was comparable to the response observed in the respective wild-type (WT) interneurons. The sensitivity to group II and III mGlu receptors was tested on cortically-evoked excitatory postsynaptic potentials (EPSPs) recorded from medium spiny neurons (MSNs). Both LY379268 and L-AP4, agonists for group II and III, respectively, had no effect on intrinsic membrane properties, but dose-dependently reduced the amplitude of corticostriatal EPSPs. However, both in PINK1(-/-) and Parkin(-/-) mice, LY379268, but not L-AP4, exhibited a greater potency as compared to WT in depressing EPSP amplitude. Accordingly, the dose-response curve for the response to LY379268 in both knockout mice was shifted leftward. Moreover, consistent with a presynaptic site of action, both LY379268 and L-AP4 increased the paired-pulse ratio either in PINK1(-/-) and Parkin(-/-) or in WT mice. Acute pretreatment with L-dopa did not rescue the enhanced sensitivity to LY379268. Together, these results suggest that the selective increase in sensitivity of striatal group II mGlu receptors represents an adaptive change in mice in which an altered dopamine metabolism has been documented.


Subject(s)
Cerebral Cortex/cytology , Corpus Striatum/cytology , Neurons/physiology , Protein Kinases/deficiency , Receptors, Metabotropic Glutamate/metabolism , Synapses/genetics , Ubiquitin-Protein Ligases/deficiency , Amino Acids/pharmacology , Animals , Biophysical Phenomena , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dopamine Agents/pharmacology , Electric Stimulation/methods , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , In Vitro Techniques , Levodopa/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques/methods , Propionates/pharmacology , Synapses/drug effects
9.
Neuropharmacology ; 55(4): 392-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18602651

ABSTRACT

In the recent past, evidence accumulated in favour of a central role of group I metabotropic glutamate (mGlu) receptors, mGlu1 and mGlu5, in the modulation of cell excitability both of striatal medium spiny projection neurons (MSNs) and interneuronal population. Electrophysiological and pharmacological studies have clearly shown that activation of mGlu1 and mGlu5 receptors exerts distinct actions, depending on the neuronal subtype involved. MGlu5 receptor activation mediates the potentiation of NMDA responses in MSNs, and underlies the retrograde inhibitory signaling by endocannabinoids at corticostriatal synapses. Conversely, both group I mGlu receptors are involved in long-term synaptic plasticity of MSNs. Likewise, either mGlu1 or mGlu5 receptors are engaged in shaping the excitability of large cholinergic interneurons, playing different roles in the membrane responses. Differently, although GABAergic parvalbumin-positive, fast-spiking interneurons have been shown to express both group I receptors, only mGlu1 receptor seems to mediate membrane and synaptic responses. This review provides a brief survey of the cellular and synaptic actions of group I mGlu receptors, and discusses the potential relevance of these findings in neostriatal function and motor control.


Subject(s)
Corpus Striatum/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Humans
10.
Neuroscience ; 152(2): 469-76, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18262727

ABSTRACT

By means of whole-cell patch-clamp recordings, we characterized the developmental profile of high-voltage-activated (HVA) calcium (Ca(2+)) channel subtypes in distinct neuronal populations of mouse striatum. Acutely dissociated medium spiny neurons (MSNs) and cholinergic interneurons (ChIs) were recorded from mice at five developmental stages: postnatal-days (PD) 14, 23, 40, 150 and 270. During ageing, total HVA Ca(2+) current recorded from both MSNs and ChIs was unchanged. However, the pharmacological analysis of the differential contribution of HVA Ca(2+) channel subtypes showed a significant rearrangement of each component. In both neuronal subtypes, a large fraction of the total HVA current recorded from PD14 mice was inhibited by the L-type HVA channel blocker nifedipine. This dihydropyridine-sensitive component accounted for nearly 50%, in MSNs, and 35%, in ChIs, of total current at PD14, but its contribution was down-regulated up to 20-25% at 9 months. Likewise, the N-type, omega-conotoxin GVIA-sensitive component decreased from 35% to 40% to about 25% in MSNs and 15% in ChIs. The P-type, omega-agatoxin-sensitive fraction did not show significant changes in both neuronal subtypes, whereas the Q-type, omega-conotoxin MVIIC-sensitive channels did show a significant up-regulation at 9 months. As compared with striatal neurons, we recorded pyramidal neurons dissociated from cortical layers IV-V and found no significant developmental change in the different components of HVA Ca(2+) currents. In conclusion, our data demonstrate a functional reconfiguration of HVA Ca(2+) channels in striatal but not cortical pyramidal neurons during mouse development. Such changes might have profound implications for physiological and pathophysiological processes of the striatum.


Subject(s)
Aging/physiology , Calcium Channels/physiology , Corpus Striatum/cytology , Neurons/classification , Neurons/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Calcium Channel Blockers/pharmacology , Calcium Channels/classification , Calcium Channels/drug effects , Calcium Channels/radiation effects , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...