Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38228889

ABSTRACT

Early and progressive dysfunctions of the dopaminergic system from the Ventral Tegmental Area (VTA) have been described in Alzheimer's Disease (AD). During the long pre-symptomatic phase, alterations in the function of Parvalbumin interneurons (PV-INs) are also observed, resulting in cortical hyperexcitability represented by subclinical epilepsy and aberrant gamma-oscillations. However, it is unknown whether the dopaminergic deficits contribute to brain hyperexcitability in AD. Here, using the Tg2576 mouse model of AD, we prove that reduced hippocampal dopaminergic innervation, due to VTA dopamine neuron degeneration, impairs PV-IN firing and gamma-waves, weakens the inhibition of pyramidal neurons and induces hippocampal hyperexcitability via lower D2-receptor-mediated activation of the CREB-pathway. These alterations coincide with reduced PV-IN numbers and Perineuronal Net density. Importantly, L-DOPA and the selective D2-receptor agonist quinpirole rescue p-CREB levels and improve the PV-IN-mediated inhibition, thus reducing hyperexcitability. Moreover, similarly to quinpirole, sumanirole - another D2-receptor agonist and a known anticonvulsant - not only increases p-CREB levels in PV-INs but also restores gamma-oscillations in Tg2576 mice. Conversely, blocking the dopaminergic transmission with sulpiride (a D2-like receptor antagonist) in WT mice reduces p-CREB levels in PV-INs, mimicking what occurs in Tg2576. Overall, these findings support the hypothesis that the VTA dopaminergic system integrity plays a key role in hippocampal PV-IN function and survival, disclosing a relevant contribution of the reduced dopaminergic tone to aberrant gamma-waves, hippocampal hyperexcitability and epileptiform activity in early AD.

2.
Neurobiol Dis ; 191: 106403, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182074

ABSTRACT

Loss-of-function mutations in the GNAL gene are responsible for DYT-GNAL dystonia. However, how GNAL mutations contribute to synaptic dysfunction is still unclear. The GNAL gene encodes the Gαolf protein, an isoform of stimulatory Gαs enriched in the striatum, with a key role in the regulation of cAMP signaling. Here, we used a combined biochemical and electrophysiological approach to study GPCR-mediated AC-cAMP cascade in the striatum of the heterozygous GNAL (GNAL+/-) rat model. We first analyzed adenosine type 2 (A2AR), and dopamine type 1 (D1R) receptors, which are directly coupled to Gαolf, and observed that the total levels of A2AR were increased, whereas D1R level was unaltered in GNAL+/- rats. In addition, the striatal isoform of adenylyl cyclase (AC5) was reduced, despite unaltered basal cAMP levels. Notably, the protein expression level of dopamine type 2 receptor (D2R), that inhibits the AC5-cAMP signaling pathway, was also reduced, similar to what observed in different DYT-TOR1A dystonia models. Accordingly, in the GNAL+/- rat striatum we found altered levels of the D2R regulatory proteins, RGS9-2, spinophilin, Gß5 and ß-arrestin2, suggesting a downregulation of D2R signaling cascade. Additionally, by analyzing the responses of striatal cholinergic interneurons to D2R activation, we found that the receptor-mediated inhibitory effect is significantly attenuated in GNAL+/- interneurons. Altogether, our findings demonstrate a profound alteration in the A2AR/D2R-AC-cAMP cascade in the striatum of the rat DYT-GNAL dystonia model, and provide a plausible explanation for our previous findings on the loss of dopamine D2R-dependent corticostriatal long-term depression.


Subject(s)
Dystonia , Dystonic Disorders , Rats , Animals , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Dopamine/metabolism , Cyclic AMP/metabolism , Dystonia/genetics , Signal Transduction/physiology , Corpus Striatum/metabolism , Receptors, Dopamine/metabolism , Protein Isoforms/metabolism
3.
Cell Rep ; 42(9): 113066, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37656620

ABSTRACT

Fear-related disorders arise from inefficient fear extinction and have immeasurable social and economic costs. Here, we characterize mouse phenotypes that spontaneously show fear-independent behavioral traits predicting adaptive or maladaptive fear extinction. We find that, already before fear conditioning, specific morphological, electrophysiological, and transcriptomic patterns of cortical and amygdala pyramidal neurons predispose to fear-related disorders. Finally, by using an optogenetic approach, we show the possibility to rescue inefficient fear extinction by activating infralimbic pyramidal neurons and to impair fear extinction by activating prelimbic pyramidal neurons.


Subject(s)
Fear , Prefrontal Cortex , Mice , Animals , Prefrontal Cortex/physiology , Fear/physiology , Transcriptome/genetics , Extinction, Psychological/physiology , Amygdala/physiology , Pyramidal Cells/physiology
4.
Mov Disord ; 37(5): 949-961, 2022 05.
Article in English | MEDLINE | ID: mdl-35420219

ABSTRACT

BACKGROUND: The neuronal protein alpha-synuclein (α-Syn) is crucially involved in Parkinson's disease pathophysiology. Intriguingly, torsinA (TA), the protein causative of DYT1 dystonia, has been found to accumulate in Lewy bodies and to interact with α-Syn. Both proteins act as molecular chaperones and control synaptic machinery. Despite such evidence, the role of α-Syn in dystonia has never been investigated. OBJECTIVE: We explored whether α-Syn and N-ethylmaleimide sensitive fusion attachment protein receptor proteins (SNAREs), that are known to be modulated by α-Syn, may be involved in DYT1 dystonia synaptic dysfunction. METHODS: We used electrophysiological and biochemical techniques to study synaptic alterations in the dorsal striatum of the Tor1a+ /Δgag mouse model of DYT1 dystonia. RESULTS: In the Tor1a+/Δgag DYT1 mutant mice, we found a significant reduction of α-Syn levels in whole striata, mainly involving glutamatergic corticostriatal terminals. Strikingly, the striatal levels of the vesicular SNARE VAMP-2, a direct α-Syn interactor, and of the transmembrane SNARE synaptosome-associated protein 23 (SNAP-23), that promotes glutamate synaptic vesicles release, were markedly decreased in mutant mice. Moreover, we detected an impairment of miniature glutamatergic postsynaptic currents (mEPSCs) recorded from striatal spiny neurons, in parallel with a decreased asynchronous release obtained by measuring quantal EPSCs (qEPSCs), which highlight a robust alteration in release probability. Finally, we also observed a significant reduction of TA striatal expression in α-Syn null mice. CONCLUSIONS: Our data demonstrate an unprecedented relationship between TA and α-Syn, and reveal that α-Syn and SNAREs alterations characterize the synaptic dysfunction underlying DYT1 dystonia. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.


Subject(s)
Dystonia Musculorum Deformans , Dystonia , Dystonic Disorders , alpha-Synuclein/metabolism , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dystonia Musculorum Deformans/metabolism , Humans , Mice , Mice, Transgenic , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , alpha-Synuclein/genetics
5.
Handb Clin Neurol ; 184: 199-206, 2022.
Article in English | MEDLINE | ID: mdl-35034734

ABSTRACT

Dystonia represents a group of movement disorders characterized by involuntary muscle contractions that result in abnormal posture and twisting movements. In the last 20 years several animal models have been generated, greatly improving our knowledge of the neural and molecular mechanism underlying this pathological condition, but the pathophysiology remains still poorly understood. In this review we will discuss recent genetic factors related to dystonia and the current understanding of synaptic plasticity alterations reported by both clinical and experimental research. We will also present recent evidence involving epigenetics mechanisms in dystonia.


Subject(s)
Dystonia , Dystonic Disorders , Movement Disorders , Animals , Dystonia/genetics , Dystonic Disorders/genetics , Epigenesis, Genetic/genetics , Humans , Neuronal Plasticity/genetics
6.
FEBS J ; 289(19): 5834-5849, 2022 10.
Article in English | MEDLINE | ID: mdl-34217152

ABSTRACT

Alcohol consumption affects motor behavior and motor control. Both acute and chronic alcohol abuse have been extensively investigated; however, the therapeutic efficacy of alcohol on some movement disorders, such as myoclonus-dystonia or essential tremor, still does not have a plausible mechanistic explanation. Yet, there are surprisingly few systematic trials with known GABAergic drugs mimicking the effect of alcohol on neurotransmission. In this brief survey, we aim to summarize the effects of EtOH on striatal function, providing an overview of its cellular and synaptic actions in a 'circuit-centered' view. In addition, we will review both experimental and clinical evidence, in the attempt to provide a plausible mechanistic explanation for alcohol-responsive movement disorders, with particular emphasis on dystonia. Different hypotheses emerge, which may provide a rationale for the utilization of drugs that mimic alcohol effects, predicting potential drug repositioning.


Subject(s)
Dystonia , Dystonic Disorders , Movement Disorders , Dystonia/drug therapy , Dystonic Disorders/drug therapy , Ethanol , Humans , Synaptic Transmission
7.
Mov Disord ; 36(12): 2768-2779, 2021 12.
Article in English | MEDLINE | ID: mdl-34173686

ABSTRACT

BACKGROUND: Acetylcholine-mediated transmission plays a central role in the impairment of corticostriatal synaptic activity and plasticity in multiple DYT1 mouse models. However, the nature of such alteration remains unclear. OBJECTIVE: The aim of the present work was to characterize the mechanistic basis of cholinergic dysfunction in DYT1 dystonia to identify potential targets for pharmacological intervention. METHODS: We utilized electrophysiology recordings, immunohistochemistry, enzymatic activity assays, and Western blotting techniques to analyze in detail the cholinergic machinery in the dorsal striatum of the Tor1a+/- mouse model of DYT1 dystonia. RESULTS: We found a significant increase in the vesicular acetylcholine transporter (VAChT) protein level, the protein responsible for loading acetylcholine (ACh) from the cytosol into synaptic vesicles, which indicates an altered cholinergic tone. Accordingly, in Tor1a+/- mice we measured a robust elevation in basal ACh content coupled to a compensatory enhancement of acetylcholinesterase (AChE) enzymatic activity. Moreover, pharmacological activation of dopamine D2 receptors, which is expected to reduce ACh levels, caused an abnormal elevation in its content, as compared to controls. Patch-clamp recordings revealed a reduced effect of AChE inhibitors on cholinergic interneuron excitability, whereas muscarinic autoreceptor function was preserved. Finally, we tested the hypothesis that blockade of VAChT could restore corticostriatal long-term synaptic plasticity deficits. Vesamicol, a selective VAChT inhibitor, rescued a normal expression of synaptic plasticity. CONCLUSIONS: Overall, our findings indicate that VAChT is a key player in the alterations of striatal plasticity and a novel target to normalize cholinergic dysfunction observed in DYT1 dystonia. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Dystonia , Acetylcholinesterase/metabolism , Animals , Cholinergic Agents/metabolism , Corpus Striatum/metabolism , Dystonia Musculorum Deformans , Mice , Molecular Chaperones/metabolism , Neuronal Plasticity , Vesicular Acetylcholine Transport Proteins/metabolism
8.
Neurosci Biobehav Rev ; 127: 334-352, 2021 08.
Article in English | MEDLINE | ID: mdl-33964307

ABSTRACT

The acquisition of fear associative memory requires brain processes of coordinated neural activity within the amygdala, prefrontal cortex (PFC), hippocampus, thalamus and brainstem. After fear consolidation, a suppression of fear memory in the absence of danger is crucial to permit adaptive coping behavior. Acquisition and maintenance of fear extinction critically depend on amygdala-PFC projections. The robust correspondence between the brain networks encompassed cortical and subcortical hubs involved into fear processing in humans and in other species underscores the potential utility of comparing the modulation of brain circuitry in humans and animals, as a crucial step to inform the comprehension of fear mechanisms and the development of treatments for fear-related disorders. The present review is aimed at providing a comprehensive description of the literature on recent clinical and experimental researches regarding the noninvasive brain stimulation and optogenetics. These innovative manipulations applied over specific hubs of fear matrix during fear acquisition, consolidation, reconsolidation and extinction allow an accurate characterization of specific brain circuits and their peculiar interaction within the specific fear processing.


Subject(s)
Extinction, Psychological , Fear , Animals , Brain , Humans , Optogenetics , Writing
9.
Neurobiol Dis ; 153: 105319, 2021 06.
Article in English | MEDLINE | ID: mdl-33647447

ABSTRACT

Visual recognition of facial expression modulates our social interactions. Compelling experimental evidence indicates that face conveys plenty of information that are fundamental for humans to interact. These are encoded at neural level in specific cortical and subcortical brain regions through activity- and experience-dependent synaptic plasticity processes. The current pandemic, due to the spread of SARS-CoV-2 infection, is causing relevant social and psychological detrimental effects. The institutional recommendations on physical distancing, namely social distancing and wearing of facemasks are effective in reducing the rate of viral spread. However, by impacting social interaction, facemasks might impair the neural responses to recognition of facial cues that are overall critical to our behaviors. In this survey, we briefly review the current knowledge on the neurobiological substrate of facial recognition and discuss how the lack of salient stimuli might impact the ability to retain and consolidate learning and memory phenomena underlying face recognition. Such an "abnormal" visual experience raises the intriguing possibility of a "reset" mechanism, a renewed ability of adult brain to undergo synaptic plasticity adaptations.


Subject(s)
Brain/physiology , COVID-19/prevention & control , Facial Recognition/physiology , Masks , Neuronal Plasticity/physiology , Communicable Disease Control , Humans , Occipital Lobe/physiology , Prefrontal Cortex/physiology , SARS-CoV-2 , Social Perception , Temporal Lobe/physiology , Visual Pathways/physiology
10.
Int J Mol Sci ; 22(2)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467450

ABSTRACT

Fear extinction requires coordinated neural activity within the amygdala and medial prefrontal cortex (mPFC). Any behavior has a transcriptomic signature that is modified by environmental experiences, and specific genes are involved in functional plasticity and synaptic wiring during fear extinction. Here, we investigated the effects of optogenetic manipulations of prelimbic (PrL) pyramidal neurons and amygdala gene expression to analyze the specific transcriptional pathways associated to adaptive and maladaptive fear extinction. To this aim, transgenic mice were (or not) fear-conditioned and during the extinction phase they received optogenetic (or sham) stimulations over photo-activable PrL pyramidal neurons. At the end of behavioral testing, electrophysiological (neural cellular excitability and Excitatory Post-Synaptic Currents) and morphological (spinogenesis) correlates were evaluated in the PrL pyramidal neurons. Furthermore, transcriptomic cell-specific RNA-analyses (differential gene expression profiling and functional enrichment analyses) were performed in amygdala pyramidal neurons. Our results show that the optogenetic activation of PrL pyramidal neurons in fear-conditioned mice induces fear extinction deficits, reflected in an increase of cellular excitability, excitatory neurotransmission, and spinogenesis of PrL pyramidal neurons, and associated to strong modifications of the transcriptome of amygdala pyramidal neurons. Understanding the electrophysiological, morphological, and transcriptomic architecture of fear extinction may facilitate the comprehension of fear-related disorders.


Subject(s)
Amygdala/physiology , Conditioning, Classical/physiology , Extinction, Psychological/physiology , Fear/physiology , Pyramidal Cells/physiology , Transcriptome/genetics , Amygdala/cytology , Amygdala/metabolism , Animals , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/physiology , Fear/psychology , Male , Memory/physiology , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/metabolism , Neural Pathways/physiology , Optogenetics/methods , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Pyramidal Cells/metabolism , Synaptic Transmission/physiology
11.
Cell Rep ; 31(7): 107644, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433955

ABSTRACT

Firing activity of external globus pallidus (GPe) is crucial for motor control and is severely perturbed in dystonia, a movement disorder characterized by involuntary, repetitive muscle contractions. Here, we show that GPe projection neurons exhibit a reduction of firing frequency and an irregular pattern in a DYT1 dystonia model. Optogenetic activation of the striatopallidal pathway fails to reset pacemaking activity of GPe neurons in mutant mice. Abnormal firing is paralleled by alterations in motor learning. We find that loss of dopamine D2 receptor-dependent inhibition causes increased GABA input at striatopallidal synapses, with subsequent downregulation of hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels. Accordingly, enhancing in vivo HCN channel activity or blocking GABA release restores both the ability of striatopallidal inputs to pause ongoing GPe activity and motor coordination deficits. Our findings demonstrate an impaired striatopallidal connectivity, supporting the central role of GPe in motor control and, more importantly, identifying potential pharmacological targets for dystonia.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Molecular Chaperones/metabolism , Neurons/metabolism , Optogenetics/methods , Animals , Mice
12.
Neurobiol Dis ; 134: 104634, 2020 02.
Article in English | MEDLINE | ID: mdl-31678405

ABSTRACT

Dystonia is a neurological movement disorder characterized by sustained or intermittent involuntary muscle contractions. Loss-of-function mutations in the GNAL gene have been identified to be the cause of "isolated" dystonia DYT25. The GNAL gene encodes for the guanine nucleotide-binding protein G(olf) subunit alpha (Gαolf), which is mainly expressed in the olfactory bulb and the striatum and functions as a modulator during neurotransmission coupling with D1R and A2AR. Previously, heterozygous Gαolf -deficient mice (Gnal+/-) have been generated and showed a mild phenotype at basal condition. In contrast, homozygous deletion of Gnal in mice (Gnal-/-) resulted in a significantly reduced survival rate. In this study, using the CRISPR-Cas9 system we generated and characterized heterozygous Gnal knockout rats (Gnal+/-) with a 13 base pair deletion in the first exon of the rat Gnal splicing variant 2, a major isoform in both human and rat striatum. Gnal+/- rats showed early-onset phenotypes associated with impaired dopamine transmission, including reduction in locomotor activity, deficits in rotarod performance and an abnormal motor skill learning ability. At cellular and molecular level, we found down-regulated Arc expression, increased cell surface distribution of AMPA receptors, and the loss of D2R-dependent corticostriatal long-term depression (LTD) in Gnal+/- rats. Based on the evidence that D2R activity is normally inhibited by adenosine A2ARs, co-localized on the same population of striatal neurons, we show that blockade of A2ARs restores physiological LTD. This animal model may be a valuable tool for investigating Gαolf function and finding a suitable treatment for dystonia associated with deficient dopamine transmission.


Subject(s)
Adenosine/metabolism , Disease Models, Animal , Dopamine/metabolism , Dystonia , Long-Term Synaptic Depression/physiology , Animals , Dystonia/metabolism , Dystonia/physiopathology , GTP-Binding Protein alpha Subunits/genetics , Gene Knockout Techniques , Male , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/metabolism , Signal Transduction/physiology
13.
EMBO Mol Med ; 11(1)2019 01.
Article in English | MEDLINE | ID: mdl-30552094

ABSTRACT

Dopamine D2 receptor signaling is central for striatal function and movement, while abnormal activity is associated with neurological disorders including the severe early-onset DYT1 dystonia. Nevertheless, the mechanisms that regulate D2 receptor signaling in health and disease remain poorly understood. Here, we identify a reduced D2 receptor binding, paralleled by an abrupt reduction in receptor protein level, in the striatum of juvenile Dyt1 mice. This occurs through increased lysosomal degradation, controlled by competition between ß-arrestin 2 and D2 receptor binding proteins. Accordingly, we found lower levels of striatal RGS9-2 and spinophilin. Further, we show that genetic depletion of RGS9-2 mimics the D2 receptor loss of DYT1 dystonia striatum, whereas RGS9-2 overexpression rescues both receptor levels and electrophysiological responses in Dyt1 striatal neurons. This work uncovers the molecular mechanism underlying D2 receptor downregulation in Dyt1 mice and in turn explains why dopaminergic drugs lack efficacy in DYT1 patients despite significant evidence for striatal D2 receptor dysfunction. Our data also open up novel avenues for disease-modifying therapeutics to this incurable neurological disorder.


Subject(s)
Corpus Striatum/pathology , Dystonia Musculorum Deformans/pathology , Dystonia Musculorum Deformans/physiopathology , Molecular Chaperones/genetics , RGS Proteins/analysis , Receptors, Dopamine D2/analysis , Signal Transduction , Animals , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Mice, Inbred C57BL , Microfilament Proteins/analysis , Nerve Tissue Proteins/analysis , RGS Proteins/genetics
14.
Curr Opin Neurol ; 31(4): 498-503, 2018 08.
Article in English | MEDLINE | ID: mdl-29746398

ABSTRACT

PURPOSE OF REVIEW: This survey takes into consideration the most recent advances in both human degenerative ataxias, disorders with a well established cerebellar origin, and discoveries from dystonia rodent models aimed at discussing the pathogenesis of dystonia. RECENT FINDINGS: One common recurrent term that emerges when describing dystonia is heterogeneity. Indeed, dystonia encompasses a wide group of 'hyperkinetic' movement disorders, with heterogeneous causes, classification, anatomical and physiological substrates. In addition, the clinical heterogeneity of age at onset, symptom distribution and appearance of non-motor symptoms has supported the concept of dystonia as 'network' disorder. Pathophysiological alterations are thought to arise from dysfunction at cortico-thalamic-basal ganglia level, whereas, more recently, a role for cerebellar pathways emerged. Results from human and animal studies thus fuel the evolving concept of the network disorder. SUMMARY: Current evidence suggests the involvement of multiple brain regions and cellular mechanisms, as part of the neural dysfunction observed at system level in dystonia.


Subject(s)
Biological Evolution , Dystonia/physiopathology , Dystonic Disorders/physiopathology , Nerve Net/physiopathology , Ataxia/physiopathology , Humans
15.
Elife ; 72018 03 05.
Article in English | MEDLINE | ID: mdl-29504938

ABSTRACT

The onset of abnormal movements in DYT1 dystonia is between childhood and adolescence, although it is unclear why clinical manifestations appear during this developmental period. Plasticity at corticostriatal synapses is critically involved in motor memory. In the Tor1a+/Δgag DYT1 dystonia mouse model, long-term potentiation (LTP) appeared prematurely in a critical developmental window in striatal spiny neurons (SPNs), while long-term depression (LTD) was never recorded. Analysis of dendritic spines showed an increase of both spine width and mature mushroom spines in Tor1a+/Δgag neurons, paralleled by an enhanced AMPA receptor (AMPAR) accumulation. BDNF regulates AMPAR expression during development. Accordingly, both proBDNF and BDNF levels were significantly higher in Tor1a+/Δgag mice. Consistently, antagonism of BDNF rescued synaptic plasticity deficits and AMPA currents. Our findings demonstrate that early loss of functional and structural synaptic homeostasis represents a unique endophenotypic trait during striatal maturation, promoting the appearance of clinical manifestations in mutation carriers.


Subject(s)
Corpus Striatum/growth & development , Corpus Striatum/physiopathology , Dystonia/genetics , Dystonia/pathology , Molecular Chaperones/genetics , Neuronal Plasticity , Animals , Disease Models, Animal , Long-Term Potentiation , Mice
16.
Parkinsonism Relat Disord ; 46 Suppl 1: S10-S14, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28760592

ABSTRACT

BACKGROUND: In the past decade, the study of the pathogenic mechanisms underlying neurodegeneration in Parkinson's disease (PD) has revealed a genetic component, often associated with a number of environmental risk factors. Animal models have improved our understanding of disease pathogenesis, providing significant insights into the understanding of novel molecular pathways. Each model has its own specific features and limitations, and the choice of the most appropriate one depends on the specific question that has to be answered. AIM: To provide an overview of some of the models supporting the hypothesis that early synaptic dysfunction represents a central event in the course of the disease. DEVELOPMENT: Along with "classical" models, based on the administration of neurotoxins and capable of replicating the neuropathological hallmarks of the disease, a number of genetic models, reproducing the disease-causing mutations of monogenic forms of familial PD, have been generated. More recently, novel models have been developed, based on the combination of a toxic insult together with PD mutations, allowing for the identification of dysfunction at a prodromal disease stage. CONCLUSIONS: The development and characterization of new models is crucial for a better understanding of PD related-synaptopathy, and hold promise for the identification of novel therapeutics.


Subject(s)
Brain/pathology , Disease Models, Animal , Parkinson Disease , Synapses/pathology , Animals , Animals, Genetically Modified , Brain/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation/genetics , Neurotoxins/toxicity , Optogenetics , Parkinson Disease/etiology , Parkinson Disease/pathology , Parkinson Disease/therapy , Protein Kinases/genetics , Rodentia
17.
Mov Disord ; 33(2): 310-320, 2018 02.
Article in English | MEDLINE | ID: mdl-29150865

ABSTRACT

BACKGROUND: Mu opioid receptor activation modulates acetylcholine release in the dorsal striatum, an area deeply involved in motor function, habit formation, and reinforcement learning as well as in the pathophysiology of different movement disorders, such as dystonia. Although the role of opioids in drug reward and addiction is well established, their involvement in motor dysfunction remains largely unexplored. METHODS: We used a multidisciplinary approach to investigate the responses to mu activation in 2 mouse models of DYT1 dystonia (Tor1a+/Δgag mice, Tor1a+/- torsinA null mice, and their respective wild-types). We performed electrophysiological recordings to characterize the pharmacological effects of receptor activation in cholinergic interneurons as well as the underlying ionic currents. In addition, an analysis of the receptor expression was performed both at the protein and mRNA level. RESULTS: In mutant mice, selective mu receptor activation caused a stronger G-protein-dependent, dose-dependent inhibition of firing activity in cholinergic interneurons when compared with controls. In Tor1a+/- mice, our electrophysiological analysis showed an abnormal involvement of calcium-activated potassium channels. Moreover, in both models we found increased levels of mu receptor protein. In addition, both total mRNA and the mu opioid receptor splice variant 1S (MOR-1S) splice variant of the mu receptor gene transcript, specifically enriched in striatum, were selectively upregulated. CONCLUSION: Mice with the DYT1 dystonia mutation exhibit an enhanced response to mu receptor activation, dependent on selective receptor gene upregulation. Our data suggest a novel role for striatal opioid signaling in motor control, and more important, identify mu opioid receptors as potential targets for pharmacological intervention in dystonia. © 2017 International Parkinson and Movement Disorder Society.


Subject(s)
Acetylcholine/metabolism , Corpus Striatum/metabolism , Dystonia/genetics , Gene Expression Regulation/genetics , Molecular Chaperones/genetics , Receptors, Opioid, mu/metabolism , Action Potentials/physiology , Adenosine Triphosphate/pharmacology , Analgesics, Opioid/pharmacology , Animals , Calcium/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/drug effects , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Dystonia/pathology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Gene Expression Regulation/drug effects , Male , Mice , Mice, Transgenic , Patch-Clamp Techniques , Receptors, Opioid, mu/genetics , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
18.
Sci Rep ; 5: 16742, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26572101

ABSTRACT

Parvalbumin-containing fast-spiking interneurons (FSIs) exert a powerful feed-forward GABAergic inhibition on striatal medium spiny neurons (MSNs), playing a critical role in timing striatal output. However, how glutamatergic inputs modulate their firing activity is still unexplored. Here, by means of a combined optogenetic and electrophysiological approach, we provide evidence for a differential modulation of cortico- vs thalamo-striatal synaptic inputs to FSIs in transgenic mice carrying light-gated ion channels channelrhodopsin-2 (ChR2) in glutamatergic fibers. Corticostriatal synapses show a postsynaptic facilitation, whereas thalamostriatal synapses present a postsynaptic depression. Moreover, thalamostriatal synapses exhibit more prominent AMPA-mediated currents than corticostriatal synapses, and an increased release probability. Furthermore, during current-evoked firing activity, simultaneous corticostriatal stimulation increases bursting activity. Conversely, thalamostriatal fiber activation shifts the canonical burst-pause activity to a more prolonged, regular firing pattern. However, this change in firing pattern was accompanied by a significant rise in the frequency of membrane potential oscillations. Notably, the responses to thalamic stimulation were fully abolished by blocking metabotropic glutamate 1 (mGlu1) receptor subtype, whereas both acetylcholine and dopamine receptor antagonists were ineffective. Our findings demonstrate that cortical and thalamic glutamatergic input differently modulate FSIs firing activity through specific intrinsic and synaptic properties, exerting a powerful influence on striatal outputs.


Subject(s)
Corpus Striatum/physiology , Interneurons/physiology , Thalamus/physiology , Animals , Calcium/metabolism , Channelrhodopsins , Electric Stimulation , Excitatory Postsynaptic Potentials , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , N-Methylaspartate/metabolism , Patch-Clamp Techniques , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Synapses/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
19.
Sci Rep ; 5: 10933, 2015 Jul 20.
Article in English | MEDLINE | ID: mdl-26190541

ABSTRACT

Mechanisms of gender-specific synaptic plasticity in the striatum, a brain region that controls motor, cognitive and psychiatric functions, remain unclear. Here we report that Rhes, a GTPase enriched in medium spiny neurons (MSNs) of striatum, alters the striatal cAMP/PKA signaling cascade in a gender-specific manner. While Rhes knockout (KO) male mice, compared to wild-type (WT) mice, had a significant basal increase of cAMP/PKA signaling pathway, the Rhes KO females exhibited a much stronger response of this pathway, selectively under the conditions of dopamine/adenosine-related drug challenge. Corticostriatal LTP defects are exclusively found in A2AR/D2R-expressing MSNs of KO females, compared to KO males, an effect that is abolished by PKA inhibitors but not by the removal of circulating estrogens. This suggests that the synaptic alterations found in KO females could be triggered by an aberrant A2AR/cAMP/PKA activity, but not due to estrogen-mediated effect. Consistent with increased cAMP signaling, D1R-mediated motor stimulation, haloperidol-induced catalepsy and caffeine-evoked hyper-activity are robustly enhanced in Rhes KO females compared to mutant males. Thus Rhes, a thyroid hormone-target gene, plays a relevant role in gender-specific synaptic and behavioral responses.


Subject(s)
Corpus Striatum/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , GTP-Binding Proteins/genetics , Neuronal Plasticity , Signal Transduction , Animals , Corpus Striatum/drug effects , Cortical Spreading Depression/genetics , Dopamine/metabolism , Dopamine/pharmacology , Female , GABAergic Neurons/metabolism , Gene Expression , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Long-Term Potentiation/genetics , Male , Mice , Mice, Knockout , Motor Activity , Mutation , Neuronal Plasticity/genetics , RNA, Messenger , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Sex Factors , Signal Transduction/drug effects
20.
Neurobiol Dis ; 78: 146-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818655

ABSTRACT

Ras homolog enriched in striatum (Rhes) is highly expressed in striatal medium spiny neurons (MSNs) of rodents. In the present study, we characterized the expression of Rhes mRNA across species, as well as its functional role in other striatal neuron subtypes. Double in situ hybridization analysis showed that Rhes transcript is selectively localized in striatal cholinergic interneurons (ChIs), but not in GABAergic parvalbumin- or in neuropeptide Y-positive cell populations. Rhes is closely linked to dopamine-dependent signaling. Therefore, we recorded ChIs activity in basal condition and following dopamine receptor activation. Surprisingly, instead of an expected dopamine D2 receptor (D2R)-mediated inhibition, we observed an aberrant excitatory response in ChIs from Rhes knockout mice. Conversely, the effect of D1R agonist on ChIs was less robust in Rhes mutants than in controls. Although Rhes deletion in mutants occurs throughout the striatum, we demonstrate that the D2R response is altered specifically in ChIs, since it was recorded in pharmacological isolation, and prevented either by intrapipette BAPTA or by GDP-ß-S. Moreover, we show that blockade of Cav2.2 calcium channels prevented the abnormal D2R response. Finally, we found that the abnormal D2R activation in ChIs was rescued by selective PI3K inhibition thus suggesting that Rhes functionally modulates PI3K/Akt signaling pathway in these neurons. Our findings reveal that, besides its expression in MSNs, Rhes is localized also in striatal ChIs and, most importantly, lack of this G-protein, significantly alters D2R modulation of striatal cholinergic excitability.


Subject(s)
Corpus Striatum/physiology , GTP-Binding Proteins/physiology , Receptors, Dopamine D2/physiology , Synaptic Transmission , Action Potentials , Adolescent , Adult , Animals , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Corpus Striatum/metabolism , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...