Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 191: 106398, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38182075

RESUMO

Parkinson's disease (PD) is characterized by the progressive and asymmetrical degeneration of the nigrostriatal dopamine neurons and the unilateral presentation of the motor symptoms at onset, contralateral to the most impaired hemisphere. We previously developed a rat PD model that mimics these typical features, based on unilateral injection of a substrate inhibitor of excitatory amino acid transporters, L-trans-pyrrolidine-2,4-dicarboxylate (PDC), in the substantia nigra (SN). Here, we used this progressive model in a multilevel study (behavioral testing, in vivo 1H-magnetic resonance spectroscopy, slice electrophysiology, immunocytochemistry and in situ hybridization) to characterize the functional changes occurring in the cortico-basal ganglia-cortical network in an evolving asymmetrical neurodegeneration context and their possible contribution to the cell death progression. We focused on the corticostriatal input and the subthalamic nucleus (STN), two glutamate components with major implications in PD pathophysiology. In the striatum, glutamate and glutamine levels increased from presymptomatic stages in the PDC-injected hemisphere only, which also showed enhanced glutamatergic transmission and loss of plasticity at corticostriatal synapses assessed at symptomatic stage. Surprisingly, the contralateral STN showed earlier and stronger reactivity than the ipsilateral side (increased intraneuronal cytochrome oxidase subunit I mRNA levels; enhanced glutamate and glutamine concentrations). Moreover, its lesion at early presymptomatic stage halted the ongoing neurodegeneration in the PDC-injected SN and prevented the expression of motor asymmetry. These findings reveal the existence of endogenous interhemispheric processes linking the primary injured SN and the contralateral STN that could sustain progressive dopamine neuron loss, opening new perspectives for disease-modifying treatment of PD.


Assuntos
Doença de Parkinson , Transtornos Parkinsonianos , Núcleo Subtalâmico , Ratos , Animais , Neurônios Dopaminérgicos/metabolismo , Dopamina/metabolismo , Glutamina/metabolismo , Transtornos Parkinsonianos/metabolismo , Doença de Parkinson/metabolismo , Substância Negra/metabolismo , Glutamatos/metabolismo , Oxidopamina/farmacologia
2.
Cell Rep ; 40(1): 111034, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35793632

RESUMO

Striatal cholinergic interneurons (CINs) respond to salient or reward prediction-related stimuli after conditioning with brief pauses in their activity, implicating them in learning and action selection. This pause is lost in animal models of Parkinson's disease. How this signal regulates the striatal network remains an open question. Here, we examine the impact of CIN firing inhibition on glutamatergic transmission between the cortex and the medium spiny neurons expressing dopamine D1 receptor (D1 MSNs). Brief interruption of CIN activity has no effect in control conditions, whereas it increases glutamatergic responses in D1 MSNs after dopamine denervation. This potentiation depends upon M4 muscarinic receptor and protein kinase A. Decreasing CIN firing by optogenetics/chemogenetics in vivo partially rescues long-term potentiation in MSNs and motor learning deficits in parkinsonian mice. Our findings demonstrate that the control exerted by CINs on corticostriatal transmission and striatal-dependent motor-skill learning depends on the integrity of dopaminergic inputs.


Assuntos
Interneurônios , Transtornos Parkinsonianos , Animais , Colinérgicos/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Interneurônios/metabolismo , Camundongos , Neurônios/metabolismo , Transtornos Parkinsonianos/metabolismo
3.
Cell Death Dis ; 12(5): 460, 2021 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-33966044

RESUMO

TP53INP1 is a stress-induced protein, which acts as a dual positive regulator of transcription and of autophagy and whose deficiency has been linked with cancer and metabolic syndrome. Here, we addressed the unexplored role of TP53INP1 and of its Drosophila homolog dDOR in the maintenance of neuronal homeostasis under chronic stress, focusing on dopamine (DA) neurons under normal ageing- and Parkinson's disease (PD)-related context. Trp53inp1-/- mice displayed additional loss of DA neurons in the substantia nigra compared to wild-type (WT) mice, both with ageing and in a PD model based on targeted overexpression of α-synuclein. Nigral Trp53inp1 expression of WT mice was not significantly modified with ageing but was markedly increased in the PD model. Trp53inp2 expression showed similar evolution and did not differ between WT and Trp53inp1-/- mice. In Drosophila, pan-neuronal dDOR overexpression improved survival under paraquat exposure and mitigated the progressive locomotor decline and the loss of DA neurons caused by the human α-synuclein A30P variant. dDOR overexpression in DA neurons also rescued the locomotor deficit in flies with RNAi-induced downregulation of dPINK1 or dParkin. Live imaging, confocal and electron microscopy in fat bodies, neurons, and indirect flight muscles showed that dDOR acts as a positive regulator of basal autophagy and mitophagy independently of the PINK1-mediated pathway. Analyses in a mammalian cell model confirmed that modulating TP53INP1 levels does not impact mitochondrial stress-induced PINK1/Parkin-dependent mitophagy. These data provide the first evidence for a neuroprotective role of TP53INP1/dDOR and highlight its involvement in the regulation of autophagy and mitophagy in neurons.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Choque Térmico/metabolismo , Neuroproteção/genética , Doença de Parkinson/genética , Estresse Fisiológico/genética , Fatores Etários , Animais , Humanos , Camundongos
4.
Stem Cells Transl Med ; 10(5): 725-742, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33528918

RESUMO

Enhancing the differentiation potential of human induced pluripotent stem cells (hiPSC) into disease-relevant cell types is instrumental for their widespread application in medicine. Here, we show that hiPSCs downregulated for the signaling modulator GLYPICAN-4 (GPC4) acquire a new biological state characterized by increased hiPSC differentiation capabilities toward ventral midbrain dopaminergic (VMDA) neuron progenitors. This biological trait emerges both in vitro, upon exposing cells to VMDA neuronal differentiation signals, and in vivo, even when transplanting hiPSCs at the extreme conditions of floor-plate stage in rat brains. Moreover, it is compatible with the overall neuronal maturation process toward acquisition of substantia nigra neuron identity. HiPSCs with downregulated GPC4 also retain self-renewal and pluripotency in stemness conditions, in vitro, while losing tumorigenesis in vivo as assessed by flank xenografts. In conclusion, our results highlight GPC4 downregulation as a powerful approach to enhance generation of VMDA neurons. Outcomes may contribute to establish hiPSC lines suitable for translational applications.


Assuntos
Diferenciação Celular , Neurônios Dopaminérgicos , Glipicanas , Células-Tronco Pluripotentes Induzidas , Animais , Células Cultivadas , Neurônios Dopaminérgicos/citologia , Regulação para Baixo , Glipicanas/genética , Xenoenxertos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Mesencéfalo , Células-Tronco Neurais/citologia , Ratos
5.
Brain Struct Funct ; 224(1): 363-372, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30341742

RESUMO

Recent studies have suggested deep brain stimulation (DBS) as a promising therapy in patients with Alzheimer's disease (AD). Particularly, the stimulation of the forniceal area was found to slow down the cognitive decline of some AD patients, but the biochemical and anatomical modifications underlying these effects remain poorly understood. We evaluated the effects of chronic forniceal stimulation on amyloid burden, inflammation, and neuronal loss in a transgenic Alzheimer rat model TgF344-AD, as well as in age-matched control rats. 18-month-old rats were surgically implanted with electrodes in stereotactic conditions and connected to a portable microstimulator for chronic DBS in freely moving rats. The stimulation was continuous during 5 weeks and animals were immediately sacrificed for immunohistochemical analysis of pathological markers. Implanted, but non-stimulated rats were used as controls. We found that chronic forniceal DBS in the Tg-AD rat significantly reduces amyloid deposition in the hippocampus and cortex, decreases astrogliosis and microglial activation and lowers neuronal loss, as determined by NeuN staining. In control animals, the stimulation neither affects neuroinflammation nor neuronal count. In the Tg-F344-AD rat model, 5 weeks of forniceal DBS decreased amyloidosis, inflammatory responses, and neuronal loss in both cortex and hippocampus. These findings strongly suggest a neuroprotective effect of DBS and support the beneficial effects of targeting the fornix in Alzheimer's disease patients.


Assuntos
Doença de Alzheimer/terapia , Encéfalo/patologia , Estimulação Encefálica Profunda/métodos , Fórnice , Inflamação/terapia , Degeneração Neural , Neurônios/patologia , Placa Amiloide , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Ratos Endogâmicos F344 , Ratos Transgênicos , Fatores de Tempo
6.
Front Cell Neurosci ; 11: 149, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28620281

RESUMO

The solute carrier family 25 (SLC25) drives the import of a large diversity of metabolites into mitochondria, a key cellular structure involved in many metabolic functions. Mutations of the mitochondrial glutamate carrier SLC25A22 (also named GC1) have been identified in early epileptic encephalopathy (EEE) and migrating partial seizures in infancy (MPSI) but the pathophysiological mechanism of GC1 deficiency is still unknown, hampered by the absence of an in vivo model. This carrier is mainly expressed in astrocytes and is the principal gate for glutamate entry into mitochondria. A sufficient supply of energy is essential for the proper function of the brain and mitochondria have a pivotal role in maintaining energy homeostasis. In this work, we wanted to study the consequences of GC1 absence in an in vitro model in order to understand if glutamate catabolism and/or mitochondrial function could be affected. First, short hairpin RNA (shRNA) designed to specifically silence GC1 were validated in rat C6 glioma cells. Silencing GC1 in C6 resulted in a reduction of the GC1 mRNA combined with a decrease of the mitochondrial glutamate carrier activity. Then, primary astrocyte cultures were prepared and transfected with shRNA-GC1 or mismatch-RNA (mmRNA) constructs using the Neon® Transfection System in order to target a high number of primary astrocytes, more than 64%. Silencing GC1 in primary astrocytes resulted in a reduced nicotinamide adenine dinucleotide (Phosphate) (NAD(P)H) formation upon glutamate stimulation. We also observed that the mitochondrial respiratory chain (MRC) was functional after glucose stimulation but not activated by glutamate, resulting in a lower level of cellular adenosine triphosphate (ATP) in silenced astrocytes compared to control cells. Moreover, GC1 inactivation resulted in an intracellular glutamate accumulation. Our results show that mitochondrial glutamate transport via GC1 is important in sustaining glutamate homeostasis in astrocytes. Main Points: The mitochondrial respiratory chain is functional in absence of GC1Lack of glutamate oxidation results in a lower global ATP levelLack of mitochondrial glutamate transport results in intracellular glutamate accumulation.

7.
J Neurochem ; 136(5): 1004-16, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26576509

RESUMO

The long-term effects and action mechanisms of subthalamic nucleus (STN) high-frequency stimulation (HFS) for Parkinson's disease still remain poorly characterized, mainly due to the lack of experimental models relevant to clinical application. To address this issue, we performed a multilevel study in freely moving hemiparkinsonian rats undergoing 5-week chronic STN HFS, using a portable constant-current microstimulator. In vivo metabolic neuroimaging by (1) H-magnetic resonance spectroscopy (11.7 T) showed that STN HFS normalized the tissue levels of the neurotransmission-related metabolites glutamate, glutamine and GABA in both the striatum and substantia nigra reticulata (SNr), which were significantly increased in hemiparkinsonian rats, but further decreased nigral GABA levels below control values; taurine levels, which were not affected in hemiparkinsonian rats, were significantly reduced. Slice electrophysiological recordings revealed that STN HFS was, uniquely among antiparkinsonian treatments, able to restore both forms of corticostriatal synaptic plasticity, i.e. long-term depression and potentiation, which were impaired in hemiparkinsonian rats. Behavior analysis (staircase test) showed a progressive recovery of motor skill during the stimulation period. Altogether, these data show that chronic STN HFS efficiently counteracts metabolic and synaptic defects due to dopaminergic lesion in both the striatum and SNr. Comparison of chronic STN HFS with acute and subchronic treatment further suggests that the long-term benefits of this treatment rely both on the maintenance of acute effects and on delayed actions on the basal ganglia network. We studied the effects of chronic (5 weeks) continuous subthalamic nucleus (STN) high-frequency stimulation (HFS) in hemiparkinsonian rats. The levels of glutamate and GABA in the striatum () and substantia nigra reticulata (SNr) (), measured by in vivo proton magnetic resonance spectroscopy ((1) H-MRS), were increased by 6-hydroxydopamine (6-OHDA) lesion, which also disrupted corticostriatal synaptic plasticity () and impaired forepaw skill () in the staircase test. Five-week STN HFS normalized glutamate and GABA levels and restored both synaptic plasticity and motor function. A partial behavioral recovery was observed at 2-week STN HFS.


Assuntos
Gânglios da Base/metabolismo , Comportamento Animal/efeitos dos fármacos , Estimulação Encefálica Profunda , Plasticidade Neuronal/efeitos dos fármacos , Substância Negra/metabolismo , Núcleo Subtalâmico/metabolismo , Animais , Gânglios da Base/fisiopatologia , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Estimulação Encefálica Profunda/métodos , Dopamina/metabolismo , Ácido Glutâmico/metabolismo , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo , Doença de Parkinson/fisiopatologia , Doença de Parkinson/terapia , Ratos , Substância Negra/fisiopatologia , Núcleo Subtalâmico/fisiopatologia , Tempo
8.
PLoS One ; 10(11): e0142838, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26571268

RESUMO

Recent evidence points to a neuroprotective action of bee venom on nigral dopamine neurons in animal models of Parkinson's disease (PD). Here we examined whether bee venom also displays a symptomatic action by acting on the pathological functioning of the basal ganglia in rat PD models. Bee venom effects were assessed by combining motor behavior analyses and in vivo electrophysiological recordings in the substantia nigra pars reticulata (SNr, basal ganglia output structure) in pharmacological (neuroleptic treatment) and lesional (unilateral intranigral 6-hydroxydopamine injection) PD models. In the hemi-parkinsonian 6-hydroxydopamine lesion model, subchronic bee venom treatment significantly alleviates contralateral forelimb akinesia and apomorphine-induced rotations. Moreover, a single injection of bee venom reverses haloperidol-induced catalepsy, a pharmacological model reminiscent of parkinsonian akinetic deficit. This effect is mimicked by apamin, a blocker of small conductance Ca2+-activated K+ (SK) channels, and blocked by CyPPA, a positive modulator of these channels, suggesting the involvement of SK channels in the bee venom antiparkinsonian action. In vivo electrophysiological recordings in the substantia nigra pars reticulata (basal ganglia output structure) showed no significant effect of BV on the mean neuronal discharge frequency or pathological bursting activity. In contrast, analyses of the neuronal responses evoked by motor cortex stimulation show that bee venom reverses the 6-OHDA- and neuroleptic-induced biases in the influence exerted by the direct inhibitory and indirect excitatory striatonigral circuits. These data provide the first evidence for a beneficial action of bee venom on the pathological functioning of the cortico-basal ganglia circuits underlying motor PD symptoms with potential relevance to the symptomatic treatment of this disease.


Assuntos
Gânglios da Base/fisiopatologia , Venenos de Abelha/farmacologia , Modelos Animais de Doenças , Atividade Motora/efeitos dos fármacos , Córtex Motor/fisiopatologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Animais , Gânglios da Base/efeitos dos fármacos , Venenos de Abelha/administração & dosagem , Venenos de Abelha/uso terapêutico , Catalepsia/complicações , Catalepsia/tratamento farmacológico , Catalepsia/fisiopatologia , Antagonistas de Dopamina/farmacologia , Estimulação Elétrica , Haloperidol , Masculino , Córtex Motor/efeitos dos fármacos , Oxidopamina , Doença de Parkinson/complicações , Ratos Wistar , Receptores Dopaminérgicos/metabolismo , Substância Negra/efeitos dos fármacos , Substância Negra/fisiopatologia
9.
J Neurochem ; 132(6): 703-12, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25533782

RESUMO

Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is an efficient neurosurgical treatment for advanced Parkinson's disease. Non-invasive metabolic neuroimaging during the course of DBS in animal models may contribute to our understanding of its action mechanisms. Here, DBS was adapted to in vivo proton magnetic resonance spectroscopy at 11.7 T in the rat to follow metabolic changes in main basal ganglia structures, the striatum, and the substantia nigra pars reticulata (SNr). Measurements were repeated OFF and ON acute and subchronic (7 days) STN-DBS in control and parkinsonian (6-hydroxydopamine lesion) conditions. Acute DBS reversed the increases in glutamate, glutamine, and GABA levels induced by the dopamine lesion in the striatum but not in the SNr. Subchronic DBS normalized GABA in both the striatum and SNr, and glutamate in the striatum. Taurine levels were markedly decreased under subchronic DBS in the striatum and SNr in both lesioned and unlesioned rats. Microdialysis in the striatum further showed that extracellular taurine was increased. These data reveal that STN-DBS has duration-dependent metabolic effects in the basal ganglia, consistent with development of adaptive mechanisms. In addition to counteracting defects induced by the dopamine lesion, prolonged DBS has proper effects independent of the pathological condition. Non-invasive metabolic neuroimaging might be useful to understand the physiological mechanisms of deep brain stimulation (DBS). Here, we demonstrate the feasibility of repeated high-field proton magnetic resonance spectroscopy of basal ganglia structures under subthalamic nucleus DBS in control and parkinsonian rats. Results show that DBS has both rapid and delayed effects either dependent or independent of disease state.


Assuntos
Gânglios da Base/metabolismo , Estimulação Encefálica Profunda/tendências , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/terapia , Núcleo Subtalâmico/metabolismo , Núcleo Subtalâmico/cirurgia , Animais , Encéfalo/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Masculino , Ratos , Ratos Wistar , Fatores de Tempo
10.
Neuropsychopharmacology ; 39(11): 2662-72, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24903652

RESUMO

The striatum is the input structure of the basal ganglia network that contains heterogeneous neuronal populations, including two populations of projecting neurons called the medium spiny neurons (MSNs), and different types of interneurons. We developed a transgenic mouse model enabling inducible ablation of the striatonigral MSNs constituting the direct pathway by expressing the human diphtheria toxin (DT) receptor under the control of the Slc35d3 gene promoter, a gene enriched in striatonigral MSNs. DT injection into the striatum triggered selective elimination of the majority of striatonigral MSNs. DT-mediated ablation of striatonigral MSNs caused selective loss of cholinergic interneurons in the dorsal striatum but not in the ventral striatum (nucleus accumbens), suggesting a region-specific critical role of the direct pathway in striatal cholinergic neuron homeostasis. Mice with DT injection into the dorsal striatum showed altered basal and cocaine-induced locomotion and dramatic reduction of L-DOPA-induced dyskinesia in the parkinsonian condition. In addition, these mice exhibited reduced anxiety, revealing a role of the dorsal striatum in the modulation of behaviors involving an emotional component, behaviors generally associated with limbic structures. Altogether, these results highlight the implication of the direct striatonigral pathway in the regulation of heterogeneous functions from cell survival to regulation of motor and emotion-associated behaviors.


Assuntos
Ansiedade/fisiopatologia , Neurônios Colinérgicos/fisiologia , Corpo Estriado/fisiologia , Interneurônios/fisiologia , Movimento/fisiologia , Substância Negra/fisiologia , Animais , Antiparkinsonianos/toxicidade , Neurônios Colinérgicos/efeitos dos fármacos , Cocaína/farmacologia , Corpo Estriado/efeitos dos fármacos , Toxina Diftérica/toxicidade , Inibidores da Captação de Dopamina/farmacologia , Discinesia Induzida por Medicamentos/fisiopatologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Humanos , Interneurônios/efeitos dos fármacos , Levodopa/toxicidade , Camundongos Transgênicos , Proteínas de Transporte de Monossacarídeos/genética , Movimento/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurotoxinas/toxicidade , Regiões Promotoras Genéticas , Substância Negra/efeitos dos fármacos
11.
J Neurosci ; 34(24): 8318-23, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920634

RESUMO

The heparan sulfate proteoglycan Glypican 4 (Gpc4) is strongly expressed in mouse embryonic stem (ES) cells where it controls the maintenance of self-renewal by modulating Wnt/ß-catenin signaling activities. Here we show that mouse ES cells carrying a hypomorphic Gpc4 allele, in a single-step neuronal differentiation protocol, show increased differentiation into dopaminergic neurons expressing tyrosine hydroxylase (TH) and nuclear receptor related-1 protein (Nurr1) 1. In contrast to wild-type cells, these differentiating Gpc4-mutant cells expressed high levels of DOPA decarboxylase and the dopamine transporter, two markers expressed by fully mature dopaminergic neurons. Intrastriatal transplantation of Gpc4 hypomorphic cells into a 6-OHDA rat model for Parkinson's disease improved motor behavior in the cylinder test and amphetamine-induced rotations at a higher level than transplanted wild-type cells. Importantly, Gpc4 hypomorphic cell grafts, in contrast to wild-type cells, did not generate teratomas in the host brains, leading to strongly enhanced animal survival. Therefore, control of Gpc4 activity level represents a new potential strategy to reduce ES cell tumorigenic features while at the same time increasing neuronal differentiation and integration.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Células-Tronco Embrionárias/transplante , Glipicanas/metabolismo , Doença de Parkinson/fisiopatologia , Doença de Parkinson/cirurgia , Teratoma/prevenção & controle , Animais , Calbindinas/metabolismo , Contagem de Células , Diferenciação Celular , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Glipicanas/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Camundongos , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Ratos , Receptores de Dopamina D2/metabolismo , Recuperação de Função Fisiológica/fisiologia , Teratoma/etiologia , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Neuropharmacology ; 85: 166-77, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24866785

RESUMO

Metabotropic glutamate 4 (mGlu4) receptor is a promising target for the treatment of motor deficits in Parkinson's disease (PD). This is due in part to its localization at key basal ganglia (BG) synapses that become hyperactive in this pathology, particularly striatopallidal synapses. In this context, mGlu4 receptor activation using either orthosteric agonists or positive allosteric modulators (PAMs) improves motor symptoms in rodent PD models in certain conditions. However, literature data show that mGlu4 receptor PAMs have no effect at striatopallidal GABAergic synapses (unless combined with an orthosteric agonist) and on the firing activity of pallidal neurons, and fail to provide significant motor improvement in relevant PD models. This questions the mechanistic hypothesis that mGlu4 receptor PAMs should act at striatopallidal synapses to alleviate PD motor symptoms. To shed light on this issue, we performed brain slice electrophysiology experiments. We show that Lu AF21934, an mGlu4 PAM small-molecule probe-compound, was ineffective at striatopallidal synapses at all concentrations tested, while it significantly inhibited corticostriatal synaptic transmission. Similarly, Lu AF21934 did not affect electrophysiology readouts at striatopallidal synapses in the presence of haloperidol or in 6-hydroxydopamine-lesioned rats. Interestingly, co-application of Lu AF21934 with a glutamate transporter inhibitor revealed a significant inhibitory action at striatopallidal synapses. Possibly, this effect could rely on increased level/permanence of glutamate in the synaptic cleft. Such differential efficacy of mGlu4 receptor PAMs at corticostriatal vs. striatopallidal synapses raises several issues regarding the synaptic target(s) of these drugs in the BG, and challenges the mechanisms by which they alleviate motor deficits in experimental PD models.


Assuntos
Anilidas/farmacologia , Antiparkinsonianos/farmacologia , Encéfalo/efeitos dos fármacos , Ácidos Cicloexanocarboxílicos/farmacologia , Receptores de Glutamato Metabotrópico/metabolismo , Sinapses/efeitos dos fármacos , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Encéfalo/fisiopatologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Antagonistas de Dopamina/farmacologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Globo Pálido/efeitos dos fármacos , Globo Pálido/fisiopatologia , Ácido Glutâmico/metabolismo , Haloperidol/farmacologia , Masculino , Oxidopamina , Transtornos Parkinsonianos , Ratos Wistar , Sinapses/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
13.
Neurobiol Dis ; 65: 69-81, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24480091

RESUMO

Parkinson's disease (PD) is characterized by the progressive degeneration of substantia nigra (SN) dopamine neurons, involving a multifactorial cascade of pathogenic events. Here we explored the hypothesis that dysfunction of excitatory amino acid transporters (EAATs) might be involved. Acutely-induced dysfunction of EAATs in the rat SN, by single unilateral injection of their substrate inhibitor l-trans-pyrrolidine-2,4-dicarboxylate (PDC), triggers a neurodegenerative process mimicking several PD features. Dopamine neurons are selectively affected, consistent with their sustained excitation by PDC measured by slice electrophysiology. The anti-oxidant N-acetylcysteine and the NMDA receptor antagonists ifenprodil and memantine provide neuroprotection. Besides oxidative stress and NMDA receptor-mediated excitotoxicity, glutathione depletion and neuroinflammation characterize the primary insult. Most interestingly, the degeneration progresses overtime with unilateral to bilateral and caudo-rostral evolution. Transient adaptive changes in dopamine function markers in SN and striatum accompany cell loss and axonal dystrophy, respectively. Motor deficits appear when neuron loss exceeds 50% in the most affected SN and striatal dopamine tone is dramatically reduced. These findings outline a functional link between EAAT dysfunction and several PD pathogenic mechanisms/pathological hallmarks, and provide a novel acutely-triggered model of progressive Parkinsonism.


Assuntos
Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Substância Negra/metabolismo , Acetilcisteína/uso terapêutico , Potenciais de Ação/efeitos dos fármacos , Animais , Ácidos Dicarboxílicos/toxicidade , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Membro Anterior/fisiopatologia , Sequestradores de Radicais Livres/uso terapêutico , Lateralidade Funcional , Glutamato Descarboxilase/metabolismo , Técnicas In Vitro , Masculino , Atividade Motora/efeitos dos fármacos , Neuroglia/patologia , Inibidores da Captação de Neurotransmissores/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Pirrolidinas/toxicidade , Ratos , Ratos Wistar , Substância Negra/efeitos dos fármacos , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
14.
Behav Brain Res ; 258: 80-9, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24144546

RESUMO

The increasing use of mobile phone technology over the last decade raises concerns about the impact of high frequency electromagnetic fields (EMF) on health. More recently, a link between EMF, iron overload in the brain and neurodegenerative disorders including Parkinson's and Alzheimer's diseases has been suggested. Co-exposure to EMF and brain iron overload may have a greater impact on brain tissues and cognitive processes than each treatment by itself. To examine this hypothesis, Long-Evans rats submitted to 900 MHz exposure or combined 900 MHz EMF and iron overload treatments were tested in various spatial learning tasks (navigation task in the Morris water maze, working memory task in the radial-arm maze, and object exploration task involving spatial and non spatial processing). Biogenic monoamines and metabolites (dopamine, serotonin) and oxidative stress were measured. Rats exposed to EMF were impaired in the object exploration task but not in the navigation and working memory tasks. They also showed alterations of monoamine content in several brain areas but mainly in the hippocampus. Rats that received combined treatment did not show greater behavioral and neurochemical deficits than EMF-exposed rats. None of the two treatments produced global oxidative stress. These results show that there is an impact of EMF on the brain and cognitive processes but this impact is revealed only in a task exploiting spontaneous exploratory activity. In contrast, there are no synergistic effects between EMF and a high content of iron in the brain.


Assuntos
Encéfalo/metabolismo , Dopamina/metabolismo , Campos Eletromagnéticos , Sobrecarga de Ferro/metabolismo , Aprendizagem em Labirinto/fisiologia , Estresse Oxidativo/fisiologia , Serotonina/metabolismo , Animais , Comportamento Animal/fisiologia , Comportamento Exploratório/fisiologia , Masculino , Memória de Curto Prazo/fisiologia , Ratos , Ratos Wistar
15.
Neuropharmacology ; 66: 158-69, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22491024

RESUMO

Group III metabotropic glutamate (mGlu) receptors are localized in presynaptic terminals within basal ganglia (BG) circuitry that become hyperactive due to dopamine depletion in Parkinson's disease (PD). For this reason, group III mGlu receptors, in particular mGlu4, have been considered as key strategic targets for non-dopaminergic pharmacological treatments aimed at modulating these synapses, without producing the well known side-effects of l-DOPA, in particular the highly disabling l-DOPA-induced dyskinesia (LID). Herein we add physiological and functional support to this hypothesis using Lu AF21934, a novel selective and brain-penetrant mGlu4 receptor positive allosteric modulator (PAM) tool compound. By in vitro electrophysiological recordings we demonstrate that Lu AF21934 inhibits corticostriatal synaptic transmission and enhances the effect of the orthosteric mGlu4 receptor-preferred agonist LSP1-2111. In naïve rats, Lu AF21934 dose-dependently (10 and 30 mg/kg) alleviated haloperidol-induced catalepsy. In hemiparkinsonian rats (unilateral 6-hydroxydopamine lesion of the substantia nigra pars compacta), Lu AF21934 alone did not affect akinesia at the doses tested (10 and 30 mg/kg). However, when Lu AF21934 was combined with sub-threshold doses of l-DOPA (1 and 5 mg/kg), it acted synergistically in alleviating akinesia in a dose-dependent manner and, notably, also reduced the incidence of LID but not its severity. Interestingly, these effects occurred at Lu AF21934 brain free concentrations that showed functional activity in in vitro screens (calcium flux and electrophysiology assays). These results support the potential for antiparkinsonian clinical use of a combined treatment consisting in l-DOPA and a mGlu4 receptor PAM to reduce efficacious l-DOPA doses (generally known as l-DOPA sparing), while maintaining the same benefit on PD motor troubles, and at the same time minimizing the development of LID. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Assuntos
Regulação Alostérica/fisiologia , Anilidas/farmacologia , Ácidos Cicloexanocarboxílicos/farmacologia , Discinesia Induzida por Medicamentos/tratamento farmacológico , Agonistas de Aminoácidos Excitatórios/farmacologia , Levodopa/farmacologia , Doença de Parkinson/tratamento farmacológico , Receptores de Glutamato Metabotrópico/agonistas , Regulação Alostérica/efeitos dos fármacos , Aminobutiratos/agonistas , Aminobutiratos/farmacologia , Aminobutiratos/uso terapêutico , Anilidas/farmacocinética , Anilidas/uso terapêutico , Animais , Catalepsia/induzido quimicamente , Catalepsia/tratamento farmacológico , Ácidos Cicloexanocarboxílicos/farmacocinética , Ácidos Cicloexanocarboxílicos/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Agonistas de Aminoácidos Excitatórios/farmacocinética , Agonistas de Aminoácidos Excitatórios/uso terapêutico , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Haloperidol/antagonistas & inibidores , Haloperidol/farmacologia , Levodopa/efeitos adversos , Levodopa/uso terapêutico , Masculino , Oxidopamina , Ácidos Fosfínicos/agonistas , Ácidos Fosfínicos/farmacologia , Ácidos Fosfínicos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
16.
J Neurosci Methods ; 209(1): 50-7, 2012 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-22659685

RESUMO

In the last decades, deep brain stimulation (DBS) has been widely used as a functional surgical strategy for the treatment of a variety of neurological and psychiatric disorders, including Parkinson's disease (PD), dystonia, epilepsy, depression or obsessive-compulsive disorder. While the therapeutic benefits of DBS are now recognized, experimental data on its mechanisms and impact at long term remain poor. This is mainly due to the lack of a microstimulation system adapted for chronic DBS in small laboratory animals. In this context, we have developed a microstimulator for DBS adapted to rat. This device, which has a size and weight compatible for use in freely moving rat, can be clipped to a support fixed on the animal's head. This easy "removal" property is crucial because it enables removing or even switching the microstimulator during the experiments without having to anaesthetize or to operate the animal, thus minimizing stress. The design of the microstimulator allows to set the DBS parameters easily (intensity, frequency and pulse width) and to replace the battery for long-term DBS. To validate our device, we performed continuous DBS of the subthalamic nucleus (known to improve motor deficits in clinic) in a classical rat model of PD during 5 weeks. We show that this long duration stimulation reduces significantly PD-induced akinesia without inducing animal discomfort and tissue damage. These first data demonstrated that long term DBS procedure in behaving rat is now workable.


Assuntos
Estimulação Encefálica Profunda/instrumentação , Modelos Animais de Doenças , Microeletrodos , Animais , Masculino , Movimento/fisiologia , Transtornos Parkinsonianos/terapia , Ratos , Ratos Wistar
17.
Neurobiol Dis ; 42(3): 284-91, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21296669

RESUMO

Chronic high frequency stimulation of the subthalamic nucleus (STN-HFS) efficiently alleviates motor symptoms of advanced Parkinson's disease (PD). Here, we looked for possible STN-HFS-induced changes on adult brain neurogenesis in the hippocampus and olfactory bulb that may be related to non-motor deficits associated to PD, such as mood disorders and olfaction deficits. Cell proliferation (Ki-67 immuno-positive-cells) and survival (bromodeoxyuridine (BrdU)-immuno-positive cells) were assessed in the subventricular zone-olfactory bulb continuum and the dentate gyrus of the hippocampus of hemiparkinsonian rats with or without continuous STN-HFS for 8 days. Dopamine lesion impaired cell proliferation and survival through different mechanisms, the effect on proliferation being correlated to the level of dopamine depletion whereas the effect on survival was not. Prolonged STN-HFS did not affect cell proliferation, but increased cell survival bilaterally. In these regions of constitutive neurogenesis, the percentage of new neuroblasts (BrdU-doublecortin-positive cells) was unchanged, suggesting that STN-HFS can lead to a net increase in newly formed neurons later on. STN-HFS also increased new cell survival in the striatum and promoted dopamine system recovery detected by tyrosine hydroxylase immunostaining. These data provide the first evidence that prolonged STN-HFS has a neurorestorative action and support the view that the action of this neurosurgical treatment can bypass the cortico-basal ganglia-thalamocortical loop circuits and largely impinge neuroplasticity and brain function.


Assuntos
Neurogênese/fisiologia , Doença de Parkinson/fisiopatologia , Núcleo Subtalâmico/fisiopatologia , Análise de Variância , Animais , Estimulação Encefálica Profunda , Modelos Animais de Doenças , Dopamina/metabolismo , Proteína Duplacortina , Eletrodos Implantados , Imuno-Histoquímica , Masculino , Doença de Parkinson/metabolismo , Ratos , Ratos Wistar , Núcleo Subtalâmico/metabolismo
18.
Eur J Neurosci ; 32(3): 423-34, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20673310

RESUMO

Dyskinesia is a major side-effect of chronic l-DOPA administration, the reference treatment for Parkinson's disease. High-frequency stimulation of the subthalamic nucleus (STN-HFS) alleviates parkinsonian motor symptoms and indirectly improves dyskinesia by decreasing the L-DOPA requirement. However, inappropriate stimulation can also trigger dyskinetic movements, in both human and rodents. We investigated whether STN-HFS-evoked forelimb dyskinesia involved changes in glutamatergic neurotransmission as previously reported for L-DOPA-induced dyskinesias, focusing on the role of NR2B-containing N-methyl-D-aspartate receptors (NR2B/NMDARs). We applied STN-HFS in normal rats at intensities above and below the threshold for triggering forelimb dyskinesia. Dyskinesiogenic STN-HFS induced the activation of NR2B (as assessed by immunodetection of the phosphorylated residue Tyr(1472)) in neurons of the subthalamic nucleus, entopeduncular nucleus, motor thalamus and forelimb motor cortex. The severity of STN-HFS-induced forelimb dyskinesia was decreased in a dose-dependent manner by systemic injections of CP-101,606, a selective blocker of NR2B/NMDARs, but was either unaffected or increased by the non-selective N-methyl-D-aspartate receptor antagonist, MK-801.


Assuntos
Discinesias/fisiopatologia , Membro Anterior/fisiopatologia , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Subtalâmico/fisiopatologia , Análise de Variância , Animais , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Discinesias/metabolismo , Estimulação Elétrica , Eletrodos Implantados , Membro Anterior/efeitos dos fármacos , Membro Anterior/metabolismo , Imuno-Histoquímica , Masculino , Córtex Motor/metabolismo , Neurônios/efeitos dos fármacos , Piperidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Estatísticas não Paramétricas , Núcleo Subtalâmico/efeitos dos fármacos , Núcleo Subtalâmico/metabolismo , Tálamo/metabolismo
19.
J Neurosci ; 30(29): 9919-28, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20660274

RESUMO

The thalamic centromedian-parafascicular (CM/Pf) complex, mainly represented by Pf in rodents, is proposed as an interesting target for the neurosurgical treatment of movement disorders, including Parkinson's disease. In this study, we examined the functional impact of subchronic high-frequency stimulation (HFS) of Pf in the 6-hydroxydopamine-lesioned hemiparkinsonian rat model. Pf-HFS had significant anti-akinetic action, evidenced by alleviation of limb use asymmetry (cylinder test). Whereas this anti-akinetic action was moderate, Pf-HFS totally reversed lateralized neglect (corridor task), suggesting potent action on sensorimotor integration. At the cellular level, Pf-HFS partially reversed the dopamine denervation-induced increase in striatal preproenkephalin A mRNA levels, a marker of the neurons of the indirect pathway, without interfering with the markers of the direct pathway (preprotachykinin and preprodynorphin). Pf-HFS totally reversed the lesion-induced changes in the gene expression of cytochrome oxidase subunit I in the subthalamic nucleus, the globus pallidus, and the substantia nigra pars reticulata, and partially in the entopeduncular nucleus. Unlike HFS of the subthalamic nucleus, Pf-HFS did not induce per se dyskinesias and directly, although partially, alleviated L-3,4-dihydroxyphenylalanine (L-DOPA)-induced forelimb dyskinesia. Conversely, L-DOPA treatment negatively interfered with the anti-parkinsonian effect of Pf-HFS. Altogether, these data show that Pf-DBS, by recruiting a large basal ganglia circuitry, provides moderate to strong anti-parkinsonian benefits that might, however, be affected by L-DOPA. The widespread behavioral and cellular outcomes of Pf-HFS evidenced here demonstrate that CM/Pf is an important node for modulating the pathophysiological functioning of basal ganglia and related disorders.


Assuntos
Gânglios da Base/patologia , Gânglios da Base/fisiopatologia , Estimulação Encefálica Profunda , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Tálamo/fisiopatologia , Animais , Denervação/métodos , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Retroalimentação Sensorial , Globo Pálido/metabolismo , Levodopa/administração & dosagem , Levodopa/metabolismo , Masculino , Oxidopamina , Doença de Parkinson/fisiopatologia , Doença de Parkinson Secundária/induzido quimicamente , Precursores de Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Substância Negra/metabolismo , Núcleo Subtalâmico/metabolismo , Taquicininas/genética
20.
Neurobiol Dis ; 36(1): 116-25, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19615446

RESUMO

Subthalamic nucleus high frequency stimulation (STN-HFS) efficiently alleviates L-DOPA-sensitive parkinsonian motor symptoms, but has no direct beneficial action on L-DOPA-induced dyskinesias (LID). Here, we provide evidence that anti-akinetic STN-HFS or dyskinesiogenic L-DOPA similarly reversed the dopamine lesion-induced increases in gene expression of cytochrome oxidase subunit I (CoI), a metabolic marker of neuronal activity, in the globus pallidus, STN and substantia nigra pars reticulata (SNr) in rats. In contrast, in entopeduncular nucleus (EP), STN-HFS did not modify the lesion-induced increase in CoI mRNA levels, whereas L-DOPA induced a marked decrease versus control. Combining the two treatments did not reveal significant interaction. Interestingly, CoI gene expression in EP but not in SNr was inversely correlated with striatal preprodynorphin mRNA level, a LID marker. This work suggests the existence of two functional basal ganglia subcircuits: the one, including STN and SNr, involved in antiparkinsonian action, and the other, including EP, preferentially involved in LID.


Assuntos
Antiparkinsonianos/efeitos adversos , Antiparkinsonianos/farmacologia , Gânglios da Base/patologia , Discinesia Induzida por Medicamentos/patologia , Levodopa/farmacologia , Rede Nervosa/patologia , Transtornos Parkinsonianos/patologia , Animais , Gânglios da Base/efeitos dos fármacos , Modelos Animais de Doenças , Dinorfinas/genética , Dinorfinas/metabolismo , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/metabolismo , Discinesia Induzida por Medicamentos/terapia , Estimulação Elétrica/métodos , Eletrodos Implantados , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Levodopa/efeitos adversos , Masculino , Rede Nervosa/efeitos dos fármacos , Oxidopamina/efeitos adversos , Oxidopamina/farmacologia , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/terapia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...