Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 35(1): 96-111, 2015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25568106

RESUMO

In Parkinson's disease, long-term dopamine replacement therapy is complicated by the appearance of L-DOPA-induced dyskinesia (LID). One major hypothesis is that LID results from an aberrant transcriptional program in striatal neurons induced by L-DOPA and triggered by the activation of ERK. To identify these genes, we performed transcriptome analyses in the striatum in 6-hydroxydopamine-lesioned mice. A time course analysis (0-6 h after treatment with L-DOPA) identified an acute signature of 709 genes, among which genes involved in protein phosphatase activity were overrepresented, suggesting a negative feedback on ERK activation by l-DOPA. l-DOPA-dependent deregulation of 28 genes was blocked by pretreatment with SL327, an inhibitor of ERK activation, and 26 genes were found differentially expressed between highly and weakly dyskinetic animals after treatment with L-DOPA. The intersection list identified five genes: FosB, Th, Nptx2, Nedd4l, and Ccrn4l. Nptx2 encodes neuronal pentraxin II (or neuronal activity-regulated pentraxin, Narp), which is involved in the clustering of glutamate receptors. We confirmed increased Nptx2 expression after L-DOPA and its blockade by SL327 using quantitative RT-PCR in independent experiments. Using an escalating L-DOPA dose protocol, LID severity was decreased in Narp knock-out mice compared with their wild-type littermates or after overexpression of a dominant-negative form of Narp in the striatum. In conclusion, we have identified a molecular signature induced by L-DOPA in the dopamine-denervated striatum that is dependent on ERK and associated with LID. Here, we demonstrate the implication of one of these genes, Nptx2, in the development of LID.


Assuntos
Antiparkinsonianos/toxicidade , Proteína C-Reativa/biossíntese , Proteína C-Reativa/genética , Discinesia Induzida por Medicamentos/genética , Discinesia Induzida por Medicamentos/metabolismo , Levodopa/toxicidade , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Animais , Discinesia Induzida por Medicamentos/patologia , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
J Neurosci ; 34(13): 4728-40, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24672017

RESUMO

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.


Assuntos
Corpo Estriado/metabolismo , Doença de Parkinson/patologia , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Dopamina D1/metabolismo , Adrenérgicos/toxicidade , Animais , Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Agonistas de Dopamina/farmacologia , Antagonistas de Aminoácidos Excitatórios/toxicidade , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxidopamina/toxicidade , Doença de Parkinson/etiologia , Piridinas/toxicidade , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/genética , Receptores de Dopamina D1/genética , Tiazóis/toxicidade , Tirosina 3-Mono-Oxigenase/metabolismo
3.
bioRxiv ; 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39314449

RESUMO

L-DOPA-induced dyskinesia (LID) is a debilitating complication of dopamine replacement therapy in Parkinsons disease and the most common hyperkinetic disorder of basal ganglia origin. Abnormal activity of striatal D1 and D2 spiny projection neurons (SPNs) is critical for LID, yet the link between SPN activity patterns and specific dyskinetic movements remains unknown. To explore this, we developed a novel method for clustering movements based on high-resolution motion sensors and video recordings. In a mouse model of LID, this method identified two main dyskinesia types and pathological rotations, all absent during normal behavior. Using single-cell resolution imaging, we found that specific sets of both D1 and D2-SPNs were abnormally active during these pathological movements. Under baseline conditions, the same SPN sets were active during behaviors sharing physical features with LID movements. These findings indicate that ensembles of behavior-encoding D1- and D2-SPNs form new combinations of hyperactive neurons mediating specific dyskinetic movements.

4.
J Neurosci ; 32(17): 5900-10, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539851

RESUMO

Although L-3,4-dihydroxyphenylalanine (L-DOPA) remains the reference treatment of Parkinson's disease, its long-term beneficial effects are hindered by L-DOPA-induced dyskinesia (LID). In the dopamine (DA)-denervated striatum, L-DOPA activates DA D1 receptor(D1R) signaling, including cAMP-dependent protein kinase A (PKA) and extracellular signal-regulated kinase (ERK), two responses associated with LID. However, the cause of PKA and ERK activation, their respective contribution to LID, and their relationship are not known. In striatal neurons, D1R activates adenylyl-cyclase through Gα(olf), a protein upregulated after lesion of DA neurons in rats and inpatients. We report here that increased Gα(olf) levels in hemiparkinsonian mice are correlated with LID after chronic L-DOPA treatment. To determine the role of this upregulation, we performed unilateral lesion in mice lacking one allele of the Gnal gene coding for Gα(olf) (Gnal⁺/⁻). Despite an increase in the lesioned striatum,Gα(olf) levels remained below those of unlesioned wild-type mice. In Gnal⁺/⁻ mice, the lesion-induced L-DOPA stimulation of cAMP/PKA-mediated phosphorylation of GluA1 Ser845 and DARPP-32 (32 kDa DA- and cAMP-regulated phosphoprotein) Thr34 was dramatically reduced, whereas ERK activation was preserved. LID occurrence was similar in Gnal⁺/⁺ and Gnal⁺/⁻ mice after a 10-d L-DOPA (20 mg/kg) treatment. Thus, in lesioned animals, Gα(olf) upregulation is critical for the activation by L-DOPA of D1R-stimulated cAMP/PKA but not ERK signaling. Although the cAMP/PKA pathway appears to be required for LID development, our results indicate that its activation is unlikely to be the main source of LID. In contrast, the persistence of L-DOPA-induced ERK activation in Gnal⁺/⁻ mice supports its causal role in LID development.


Assuntos
Discinesia Induzida por Medicamentos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Mutação/fisiologia , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Antiparkinsonianos/efeitos adversos , Benserazida/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Corpo Estriado/efeitos dos fármacos , Interações Medicamentosas , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/genética , Inibidores Enzimáticos/farmacologia , Lateralidade Funcional/efeitos dos fármacos , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Histonas/metabolismo , Levodopa/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Movimento/efeitos dos fármacos , Mutação/genética , Oxidopamina/farmacologia , Desempenho Psicomotor/efeitos dos fármacos , Receptores de AMPA/metabolismo , Transdução de Sinais/genética , Simpatolíticos/farmacologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
5.
J Neurochem ; 108(3): 621-33, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19187092

RESUMO

In the dopamine-depleted striatum, extracellular signal-regulated kinase (ERK) signaling is implicated in the development of L-DOPA-induced dyskinesia. To gain insights on its role in this disorder, we examined the effects of L-DOPA on the state of phosphorylation of ERK and downstream target proteins in striatopallidal and striatonigral medium spiny neurons (MSNs). For this purpose, we employed mice expressing enhanced green fluorescent protein (EGFP) under the control of the promoters for the dopamine D(2) receptor (Drd2-EGFP mice) or the dopamine D(1) receptor (Drd1a-EGFP mice), which are expressed in striatopallidal and striatonigral MSNs, respectively. In 6-hydroxydopamine-lesioned Drd2-EGFP mice, L-DOPA increased the phosphorylation of ERK, mitogen- and stress-activated kinase 1 and histone H3, selectively in EGFP-negative MSNs. Conversely, a complete co-localization between EGFP and these phosphoproteins was observed in Drd1a-EGFP mice. The effect of L-DOPA was prevented by blockade of dopamine D(1) receptors. The same pattern of activation of ERK signaling was observed in dyskinetic mice, after repeated administration of L-DOPA. Our results demonstrate that in the dopamine-depleted striatum, L-DOPA activates ERK signaling specifically in striatonigral MSNs. This regulation may result in ERK-dependent changes in striatal plasticity leading to dyskinesia.


Assuntos
Antiparkinsonianos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Histonas/metabolismo , Levodopa/farmacologia , Neostriado/metabolismo , Neurônios/metabolismo , Doença de Parkinson Secundária/metabolismo , Transdução de Sinais/efeitos dos fármacos , Substância Negra/metabolismo , Animais , Western Blotting , Discinesias/fisiopatologia , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/citologia , Neostriado/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Oxidopamina , Doença de Parkinson Secundária/induzido quimicamente , Fosforilação , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Substância Negra/citologia , Substância Negra/efeitos dos fármacos , Simpatolíticos
6.
Eur J Neurosci ; 30(10): 1923-30, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19895565

RESUMO

The ability to form long-term memories exists very early during ontogeny; however, the properties of early memory processes, brain structures involved and underlying cellular mechanisms are poorly defined. Here, we examine the role of extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase/ERK signaling cascade, which is crucial for adult memory, in the consolidation and reconsolidation of an early memory using a conditioned taste aversion paradigm in 3-day-old rat pups. We show that intraperitoneal injection of SL327, the upstream mitogen-activated protein kinase kinase inhibitor, impairs both consolidation and reconsolidation of early memory, leaving short-term memory after acquisition and after reactivation intact. The amnesic effect of SL327 diminishes with increasing delays after acquisition and reactivation. Biochemical analyses revealed ERK hyperphosphorylation in the amygdala but not the hippocampus following acquisition, suggesting functional activation of the amygdala as early as post-natal day 3, although there was no clear evidence for amygdalar ERK activation after reactivation. These results indicate that, despite an immature brain, the basic properties of memory and at least some of the molecular mechanisms and brain structures implicated in aversion memory share a number of similarities with the adult and emerge very early during ontogeny.


Assuntos
Envelhecimento , Aprendizagem da Esquiva/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Memória/fisiologia , Paladar/fisiologia , Aminoacetonitrila/análogos & derivados , Aminoacetonitrila/farmacologia , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Masculino , Memória/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Paladar/efeitos dos fármacos , Fatores de Tempo
7.
J Clin Invest ; 127(2): 720-734, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28112685

RESUMO

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.


Assuntos
Levodopa/efeitos adversos , Vias Neurais/metabolismo , Doença de Parkinson Secundária/tratamento farmacológico , Receptor Muscarínico M3/agonistas , Receptores de Dopamina D2/agonistas , Córtex Visual/metabolismo , Animais , Humanos , Levodopa/farmacologia , Camundongos , Camundongos Transgênicos , Vias Neurais/patologia , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Doença de Parkinson Secundária/patologia , Ratos , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Córtex Visual/patologia
8.
Prog Neurobiol ; 132: 96-168, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26209473

RESUMO

Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.


Assuntos
Antiparkinsonianos/efeitos adversos , Sistema Nervoso Central/fisiopatologia , Discinesia Induzida por Medicamentos/fisiopatologia , Levodopa/efeitos adversos , Animais , Sistema Nervoso Central/efeitos dos fármacos , Humanos , Doença de Parkinson/tratamento farmacológico
9.
Neurosci Lett ; 583: 76-80, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25233866

RESUMO

In advanced Parkinson's disease, l-DOPA treatment causes the appearance of abnormal involuntary movements or l-DOPA-induced dyskinesia (LID). LID results in part from l-DOPA-induced activation of extracellular signal-regulated kinase (ERK) in the dopamine-denervated striatum. Activated ERK triggers nuclear responses, including phosphorylation of mitogen- and stress-activated protein kinase 1 (MSK1) and histone H3, and transcription of genes such as FosB. To determine the role of MSK1, wild type and MSK1 knockout mice with unilateral 6-hydroxydopamine lesion in the dorsolateral striatum were chronically treated with l-DOPA. The absence of MSK1 had no effect on the lesion or l-DOPA-induced ERK activation, but reduced l-DOPA-induced phosphorylation of histone H3 and FosB accumulation in the dopamine-denervated striatum. MSK1 deficiency also prevented the increase in Gαolf, the stimulatory α subunit of G protein coupling striatal dopamine D1 receptor to adenylyl cyclase. However, the intensity of LID was similar in MSK1-deficient and wild type mice. In conclusion, l-DOPA-induced activation of MSK1 contributes to histone H3 phosphorylation, induction of FosB, and Gαolf up-regulation but appears not to be necessary for the development of LID.


Assuntos
Antiparkinsonianos/farmacologia , Corpo Estriado/metabolismo , Di-Hidroxifenilalanina/efeitos adversos , Neurônios Dopaminérgicos/patologia , Discinesia Induzida por Medicamentos/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Corpo Estriado/patologia , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/patologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Oxidopamina , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais
10.
Nat Commun ; 5: 5316, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25360704

RESUMO

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.


Assuntos
Corpo Estriado/fisiopatologia , Neurônios Dopaminérgicos/fisiologia , Discinesia Induzida por Medicamentos/fisiopatologia , Plasticidade Neuronal , Transtornos Parkinsonianos/fisiopatologia , Animais , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Discinesia Induzida por Medicamentos/metabolismo , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Transtornos Parkinsonianos/patologia , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
11.
PLoS One ; 6(5): e19415, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21559295

RESUMO

Activation of dopamine D1 receptors (D1Rs) has been shown to induce epileptiform activity. We studied the molecular changes occurring in the hippocampus in response to the administration of the D1-type receptor agonist, SKF 81297. SKF 81297 at 2.5 and 5.0 mg/kg induced behavioural seizures. Electrophysiological recordings in the dentate gyrus revealed the presence of epileptiform discharges peaking at 30-45 min post-injection and declining by 60 min. Seizures were prevented by the D1-type receptor antagonist, SCH 23390, or the cannabinoid CB1 receptor agonist, CP 55,940. The effect of SKF 81297 was accompanied by increased phosphorylation of the extracellular signal-regulated protein kinases 1 and 2 (ERK), in the granule cells of the dentate gyrus. This effect was also observed in response to administration of other D1-type receptor agonists, such as SKF83822 and SKF83959. In addition, SKF 81297 increased the phosphorylation of the ribosomal protein S6 and histone H3, two downstream targets of ERK. These effects were prevented by genetic inactivation of D1Rs, or by pharmacological inhibition of ERK. SKF 81297 was also able to enhance the levels of Zif268 and Arc/Arg3.1, two immediate early genes involved in transcriptional regulation and synaptic plasticity. These changes may be involved in forms of activity-dependent plasticity linked to the manifestation of seizures and to the ability of dopamine to affect learning and memory.


Assuntos
Convulsivantes/farmacologia , Proteínas do Citoesqueleto/biossíntese , Giro Denteado/enzimologia , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/biossíntese , Receptores de Dopamina D1/agonistas , Animais , Benzazepinas/farmacologia , Cicloexanóis/farmacologia , Giro Denteado/efeitos dos fármacos , Eletrofisiologia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA