Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 59(6): 1227-1241, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37876330

ABSTRACT

The dendritic arbour of striatal projection neurons (SPNs) is the primary anatomical site where dopamine and glutamate inputs to the basal ganglia functionally interact to control movement. These dendritic arbourisations undergo atrophic changes in Parkinson's disease. A reduction in the dendritic complexity of SPNs is found also in animal models with severe striatal dopamine denervation. Using 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle as a model, we set out to compare morphological and electrophysiological properties of SPNs at an early versus a chronic stage of dopaminergic degeneration. Ex vivo recordings were performed in transgenic mice where SPNs forming the direct pathway (dSPNs) express a fluorescent reporter protein. At both the time points studied (5 and 28 days following 6-OHDA lesion), there was a complete loss of dopaminergic fibres through the dorsolateral striatum. A reduction in dSPN dendritic complexity and spine density was manifest at 28, but not 5 days post-lesion. At the late time point, dSPN also exhibited a marked increase in intrinsic excitability (reduced rheobase current, increased input resistance, more evoked action potentials in response to depolarising currents), which was not present at 5 days. The increase in neuronal excitability was accompanied by a marked reduction in inward-rectifying potassium (Kir) currents (which dampen the SPN response to depolarising stimuli). Our results show that dSPNs undergo delayed coordinate changes in dendritic morphology, intrinsic excitability and Kir conductance following dopamine denervation. These changes are predicted to interfere with the dSPN capacity to produce a normal movement-related output.


Subject(s)
Dopamine , Neurons , Mice , Animals , Dopamine/metabolism , Oxidopamine/toxicity , Neurons/physiology , Corpus Striatum/metabolism , Mice, Transgenic , Denervation
2.
Nat Commun ; 13(1): 6376, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289226

ABSTRACT

Mice display signs of fear when neurons that express cFos during fear conditioning are artificially reactivated. This finding gave rise to the notion that cFos marks neurons that encode specific memories. Here we show that cFos expression patterns in the mouse dentate gyrus (DG) change dramatically from day to day in a water maze spatial learning paradigm, regardless of training level. Optogenetic inhibition of neurons that expressed cFos on the first training day affected performance days later, suggesting that these neurons continue to be important for spatial memory recall. The mechanism preventing repeated cFos expression in DG granule cells involves accumulation of ΔFosB, a long-lived splice variant of FosB. CA1 neurons, in contrast, repeatedly expressed cFos. Thus, cFos-expressing granule cells may encode new features being added to the internal representation during the last training session. This form of timestamping is thought to be required for the formation of episodic memories.


Subject(s)
Dentate Gyrus , Spatial Learning , Animals , Mice , Dentate Gyrus/physiology , Hippocampus , Neurons/metabolism , Spatial Memory
3.
EMBO Rep ; 23(8): e54361, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35735260

ABSTRACT

The striatum is a subcortical brain region responsible for the initiation and termination of voluntary movements. Striatal spiny projection neurons receive major excitatory synaptic input from neocortex and thalamus, and cyclic nucleotides have long been known to play important roles in striatal function. Yet, the precise mechanism of action is unclear. Here, we combine optogenetic stimulation, 2-photon imaging, and genetically encoded scavengers to dissect the regulation of striatal synapses in mice. Our data show that excitatory striatal inputs are tonically depressed by phosphodiesterases (PDEs), in particular PDE1. Blocking PDE activity boosts presynaptic calcium entry and glutamate release, leading to strongly increased synaptic transmission. Although PDE1 degrades both cAMP and cGMP, we uncover that the concentration of cGMP, not cAMP, controls the gain of striatal inputs. Disturbing this gain control mechanism in vivo impairs motor skill learning in mice. The tight dependence of striatal excitatory synapses on PDE1 and cGMP offers a new perspective on the molecular mechanisms regulating striatal activity.


Subject(s)
Corpus Striatum , Synapses , Animals , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Mice , Neurons/metabolism , Synapses/physiology , Synaptic Transmission , Thalamus/metabolism
4.
Int J Mol Sci ; 23(10)2022 May 13.
Article in English | MEDLINE | ID: mdl-35628278

ABSTRACT

Non-apoptotic caspase-3 activation is critically involved in dendritic spine loss and synaptic dysfunction in Alzheimer's disease. It is, however, not known whether caspase-3 plays similar roles in other pathologies. Using a mouse model of clinically manifest Parkinson's disease, we provide the first evidence that caspase-3 is transiently activated in the striatum shortly after the degeneration of nigrostriatal dopaminergic projections. This caspase-3 activation concurs with a rapid loss of dendritic spines and deficits in synaptic long-term depression (LTD) in striatal projection neurons forming the indirect pathway. Interestingly, systemic treatment with a caspase inhibitor prevents both the spine pruning and the deficit of indirect pathway LTD without interfering with the ongoing dopaminergic degeneration. Taken together, our data identify transient and non-apoptotic caspase activation as a critical event in the early plastic changes of indirect pathway neurons following dopamine denervation.


Subject(s)
Corpus Striatum , Neostriatum , Caspase 3/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Neostriatum/metabolism , Neurons/metabolism
5.
Front Hum Neurosci ; 15: 697284, 2021.
Article in English | MEDLINE | ID: mdl-34354577

ABSTRACT

The striatum is a very heterogenous brain area, composed of different domains and compartments, albeit lacking visible anatomical demarcations. Two populations of striatal spiny projection neurons (SPNs) build the so-called direct and indirect pathway of the basal ganglia, whose coordinated activity is essential to control locomotion. Dysfunction of striatal SPNs is part of many movement disorders, such as Parkinson's disease (PD) and L-DOPA-induced dyskinesia. In this mini review article, I will highlight recent studies utilizing single-cell RNA sequencing to investigate the transcriptional profiles of striatal neurons. These studies discover that SPNs carry a transcriptional signature, indicating both their anatomical location and compartmental identity. Furthermore, the transcriptional profiles reveal the existence of additional distinct neuronal populations and previously unknown SPN sub-populations. In a parallel development, studies in rodent models of PD and L-DOPA-induced dyskinesia (LID) report that direct pathway SPNs do not react uniformly to L-DOPA therapy, and that only a subset of these neurons is underlying the development of abnormal movements. Together, these studies demonstrate a new level of cellular complexity for striatal (dys-) function and locomotor control.

6.
F1000Res ; 7: 1088, 2018.
Article in English | MEDLINE | ID: mdl-30109028

ABSTRACT

Authors of a recent paper demonstrate that, like ERMES (ER-mitochondria encounter structure) in fungal cells, PDZD8 (PDZ domain containing 8) tethers mitochondria to the ER in mammalian cells. However, identifying PDZD8 as a "functional ortholog" of yeast Mmm1 (maintenance of mitochondrial morphology protein 1) is at odds with the phylogenetic data. PDZD8 and Mmm1 are paralogs, not orthologs, which affects the interpretation of the data with respect to the evolution of ER-mitochondria tethering. Our phylogenetic analyses show that PDZD8 co-occurs with ERMES components in lineages closely related to animals solidifying its identity as a paralog of Mmm1. Additionally, we identify two related paralogs, one specific to flagellated fungi, and one present only in unicellular relatives of animals. These results point to a complex evolutionary history of ER-mitochondria tethering involving multiple gene gains and losses in the lineage leading to animals and fungi.


Subject(s)
Mitochondrial Proteins/genetics , Phylogeny , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Animals , Humans
7.
Neurobiol Dis ; 105: 117-131, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28578004

ABSTRACT

Striatal neurons forming the indirect pathway (iSPNs) are particularly vulnerable in Huntington's disease (HD). In this study we set out to investigate morphological and physiological alterations of iSPNs in two mouse models of HD with relatively slow disease progression (long CAG repeat R6/2 and zQ175-KI). Both were crossed with a transgenic mouse line expressing eGFP in iSPNs. Using the open-field and rotarod tests, we first defined two time points in relation to the occurrence of motor deficits in each model. Then, we investigated electrophysiological and morphological properties of iSPNs at both ages. Both HD models exhibited increased iSPN excitability already before the onset of motor deficits, associated with a reduced number of primary dendrites and decreased function of Kir- and voltage-gated potassium channels. Alterations that specifically occurred at symptomatic ages included increased calcium release by back-propagating action potentials in proximal dendrites, due to enhanced engagement of intracellular calcium stores. Moreover, motorically impaired mice of both HD models showed a reduction in iSPN spine density and progressive formation of huntingtin (Htt) aggregates in the striatum. Our study therefore reports iSPN-specific alterations relative to the development of a motor phenotype in two different mouse models of HD. While some alterations occur early and are partly non-progressive, others potentially provide a pathophysiological marker of an overt disease state.


Subject(s)
Disease Models, Animal , Huntington Disease/complications , Huntington Disease/physiopathology , Movement Disorders , Action Potentials/drug effects , Action Potentials/genetics , Animals , Cadmium Chloride/pharmacology , Cesium/pharmacology , Chlorides/pharmacology , Corpus Striatum/pathology , Dendrites/metabolism , Dendrites/pathology , Exploratory Behavior/physiology , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Movement Disorders/etiology , Movement Disorders/genetics , Movement Disorders/pathology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Neurons/ultrastructure , Potassium/pharmacology , Psychomotor Performance/physiology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Trinucleotide Repeat Expansion/genetics
8.
J Clin Invest ; 127(2): 720-734, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28112685

ABSTRACT

Parkinson's disease (PD) patients experience loss of normal motor function (hypokinesia), but can develop uncontrollable movements known as dyskinesia upon treatment with L-DOPA. Poverty or excess of movement in PD has been attributed to overactivity of striatal projection neurons forming either the indirect (iSPNs) or the direct (dSPNs) pathway, respectively. Here, we investigated the two pathways' contribution to different motor features using SPN type-specific chemogenetic stimulation in rodent models of PD (PD mice) and L-DOPA-induced dyskinesia (LID mice). Using the activatory Gq-coupled human M3 muscarinic receptor (hM3Dq), we found that chemogenetic stimulation of dSPNs mimicked, while stimulation of iSPNs abolished the therapeutic action of L-DOPA in PD mice. In LID mice, hM3Dq stimulation of dSPNs exacerbated dyskinetic responses to L-DOPA, while stimulation of iSPNs inhibited these responses. In the absence of L-DOPA, only chemogenetic stimulation of dSPNs mediated through the Gs-coupled modified rat muscarinic M3 receptor (rM3Ds) induced appreciable dyskinesia in PD mice. Combining D2 receptor agonist treatment with rM3Ds-dSPN stimulation reproduced all symptoms of LID. These results demonstrate that dSPNs and iSPNs oppositely modulate both therapeutic and dyskinetic responses to dopamine replacement therapy in PD. We also show that chemogenetic stimulation of different signaling pathways in dSPNs leads to markedly different motor outcomes. Our findings have important implications for the design of effective antiparkinsonian and antidyskinetic drug therapies.


Subject(s)
Levodopa/adverse effects , Neural Pathways/metabolism , Parkinson Disease, Secondary/drug therapy , Receptor, Muscarinic M3/agonists , Receptors, Dopamine D2/agonists , Visual Cortex/metabolism , Animals , Humans , Levodopa/pharmacology , Mice , Mice, Transgenic , Neural Pathways/pathology , Neurons/metabolism , Neurons/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Rats , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Visual Cortex/pathology
10.
Neuron ; 88(4): 762-73, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26590347

ABSTRACT

A balanced interaction between dopaminergic and cholinergic signaling in the striatum is critical to goal-directed behavior. But how this interaction modulates corticostriatal synaptic plasticity underlying learned actions remains unclear--particularly in direct-pathway spiny projection neurons (dSPNs). Our studies show that in dSPNs, endogenous cholinergic signaling through M4 muscarinic receptors (M4Rs) promoted long-term depression of corticostriatal glutamatergic synapses, by suppressing regulator of G protein signaling type 4 (RGS4) activity, and blocked D1 dopamine receptor dependent long-term potentiation (LTP). Furthermore, in a mouse model of L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia (LID) in Parkinson's disease (PD), boosting M4R signaling with positive allosteric modulator (PAM) blocked aberrant LTP in dSPNs, enabled LTP reversal, and attenuated dyskinetic behaviors. An M4R PAM also was effective in a primate LID model. Taken together, these studies identify an important signaling pathway controlling striatal synaptic plasticity and point to a novel pharmacological strategy for alleviating LID in PD patients.


Subject(s)
Dopamine Agents/toxicity , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Neostriatum/drug effects , Neuronal Plasticity/drug effects , Parkinsonian Disorders/drug therapy , RGS Proteins/metabolism , Receptor, Muscarinic M4/metabolism , Allosteric Regulation , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Glutamic Acid , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Macaca mulatta , Mice , Mice, Transgenic , Neostriatum/metabolism , Neurons , Signal Transduction
11.
Mov Disord ; 30(4): 484-93, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25759263

ABSTRACT

The spiny dendrites of striatal projection neurons integrate synaptic inputs of different origins to regulate movement. It has long been known that these dendrites lose spines and display atrophic features in Parkinson's disease (PD), but the significance of these morphological changes has remained unknown. Some recent studies reveal a remarkable structural plasticity of striatal spines in parkinsonian rodents treated with L-3,4-dihydroxyphenylalanine (L-DOPA), and they demonstrate an association between this plasticity and the development of dyskinesia. These studies used different approaches and animal models, which possibly explains why they emphasize different plastic changes as being most closely linked to dyskinesia (such as a growth of new spines in neurons of the indirect pathway, or a loss of spines in neurons of the direct pathway, or the appearance of spines with aberrant synaptic features). Clearly, further investigations are required to reconcile these intriguing findings and integrate them in a coherent pathophysiological model. Nevertheless, these studies may mark the beginning of a new era for dyskinesia research. In addition to addressing neurochemical and molecular events that trigger involuntary movements, there is a need to better understand the long-lasting structural reorganization of cells and circuits that maintain the brain in a "dyskinesia-prone" state. This may lead to the identification of new efficacious approaches to prevent the complications of dopaminergic therapies in PD.


Subject(s)
Corpus Striatum/pathology , Dendritic Spines/pathology , Dyskinesia, Drug-Induced/pathology , Neuronal Plasticity/physiology , Neurons/ultrastructure , Animals , Antiparkinson Agents/adverse effects , Dendritic Spines/drug effects , Dyskinesia, Drug-Induced/etiology , Humans , Levodopa/adverse effects , Neuronal Plasticity/drug effects , Neurons/pathology
12.
Nat Commun ; 5: 5316, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25360704

ABSTRACT

The striatum is widely viewed as the fulcrum of pathophysiology in Parkinson's disease (PD) and L-DOPA-induced dyskinesia (LID). In these disease states, the balance in activity of striatal direct pathway spiny projection neurons (dSPNs) and indirect pathway spiny projection neurons (iSPNs) is disrupted, leading to aberrant action selection. However, it is unclear whether countervailing mechanisms are engaged in these states. Here we report that iSPN intrinsic excitability and excitatory corticostriatal synaptic connectivity were lower in PD models than normal; L-DOPA treatment restored these properties. Conversely, dSPN intrinsic excitability was elevated in tissue from PD models and suppressed in LID models. Although the synaptic connectivity of dSPNs did not change in PD models, it fell with L-DOPA treatment. In neither case, however, was the strength of corticostriatal connections globally scaled. Thus, SPNs manifested homeostatic adaptations in intrinsic excitability and in the number but not strength of excitatory corticostriatal synapses.


Subject(s)
Corpus Striatum/physiopathology , Dopaminergic Neurons/physiology , Dyskinesia, Drug-Induced/physiopathology , Neuronal Plasticity , Parkinsonian Disorders/physiopathology , Animals , Dendritic Spines/pathology , Disease Models, Animal , Dyskinesia, Drug-Induced/metabolism , In Vitro Techniques , Male , Mice, Inbred C57BL , Parkinsonian Disorders/pathology , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
13.
J Neurosci ; 34(13): 4728-40, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24672017

ABSTRACT

In animal models of Parkinson's disease, striatal overactivation of ERK1/2 via dopamine (DA) D1 receptors is the hallmark of a supersensitive molecular response associated with dyskinetic behaviors. Here we investigate the pathways involved in D1 receptor-dependent ERK1/2 activation using acute striatal slices from rodents with unilateral 6-hydroxydopamine (6-OHDA) lesions. Application of the dopamine D1-like receptor agonist SKF38393 induced ERK1/2 phosphorylation and downstream signaling in the DA-denervated but not the intact striatum. This response was mediated through a canonical D1R/PKA/MEK1/2 pathway and independent of ionotropic glutamate receptors but blocked by antagonists of L-type calcium channels. Coapplication of an antagonist of metabotropic glutamate receptor type 5 (mGluR5) or its downstream signaling molecules (PLC, PKC, IP3 receptors) markedly attenuated SKF38393-induced ERK1/2 activation. The role of striatal mGluR5 in D1-dependent ERK1/2 activation was confirmed in vivo in 6-OHDA-lesioned animals treated systemically with SKF38393. In one experiment, local infusion of the mGluR5 antagonist MTEP in the DA-denervated rat striatum attenuated the activation of ERK1/2 signaling by SKF38393. In another experiment, 6-OHDA lesions were applied to transgenic mice with a cell-specific knockdown of mGluR5 in D1 receptor-expressing neurons. These mice showed a blunted striatal ERK1/2 activation in response to SFK38393 treatment. Our results reveal that D1-dependent ERK1/2 activation in the DA-denervated striatum depends on a complex interaction between PKA- and Ca(2+)-dependent signaling pathways that is critically modulated by striatal mGluR5.


Subject(s)
Corpus Striatum/metabolism , Parkinson Disease/pathology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Dopamine D1/metabolism , Adrenergic Agents/toxicity , Animals , Corpus Striatum/drug effects , Disease Models, Animal , Dopamine Agonists/pharmacology , Excitatory Amino Acid Antagonists/toxicity , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidopamine/toxicity , Parkinson Disease/etiology , Pyridines/toxicity , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Dopamine D1/genetics , Thiazoles/toxicity , Tyrosine 3-Monooxygenase/metabolism
14.
Nature ; 457(7233): 1079, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19242453
SELECTION OF CITATIONS
SEARCH DETAIL
...