Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(37): 18664-18672, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31455727

RESUMO

Long-term dopamine (DA) replacement therapy in Parkinson's disease (PD) leads to the development of abnormal involuntary movements known as l-Dopa-induced dyskinesia (LID). The transcription factor ΔFosB that is highly up-regulated in the striatum following chronic l-Dopa exposure may participate in the mechanisms of altered neuronal responses to DA generating LID. To identify intrinsic effects of elevated ΔFosB on l-Dopa responses, we induced transgenic ΔFosB overexpression in the striatum of parkinsonian nonhuman primates kept naïve of l-Dopa treatment. Elevated ΔFosB levels led to consistent appearance of LID since the initial acute l-Dopa tests. In line with this motor response, striatal projection neurons (SPNs) responded to DA with changes in firing frequency that reversed at the peak of the motor response, and these unstable SPN activity changes in response to DA are typically associated with the emergence of LID. Transgenic ΔFosB overexpression also induced up-regulation of other molecular markers of LID. These results support an autonomous role of striatal ΔFosB in the adaptive mechanisms altering motor responses to chronic DA replacement in PD.


Assuntos
Discinesia Induzida por Medicamentos/patologia , Levodopa/efeitos adversos , Neostriado/patologia , Doença de Parkinson/tratamento farmacológico , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Feminino , Humanos , Macaca fascicularis , Masculino , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Regulação para Cima/efeitos dos fármacos
2.
Neurotox Res ; 35(3): 575-583, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30729450

RESUMO

Organophosphates (OPs) inhibit cholinesterase and hyperactivate the acetylcholinergic nervous system in the brain, causing motor disorders (e.g., tremor and seizures). Here, we performed behavioral and immunohistochemical studies in mice and rats to investigate the tremorgenic mechanism of paraoxon, an active metabolite of parathion. Treating animals with paraoxon (0.15-0.6 mg/kg, i.p.) elicited kinetic tremor in a dose-dependent manner. Expressional analysis of Fos protein, a biomarker of neural excitation, revealed that a tremorgenic dose of paraoxon (0.6 mg/kg) significantly and region-specifically elevated Fos expression in the cerebral cortex (e.g., sensory cortex), hippocampal CA1, globus pallidus, medial habenula, and inferior olive (IO) among 48 brain regions examined. A moderate increase in Fos expression was also observed in the dorsolateral striatum while the change was not statistically significant. Paraoxon-induced tremor was inhibited by the nicotinic acetylcholine (nACh) receptor antagonist mecamylamine (MEC), but not affected by the muscarinic acetylcholine receptor antagonist trihexyphenidyl (THP). In addition, paraoxon-induced Fos expression in the IO was also antagonized by MEC, but not by THP, and lesioning of the IO markedly suppressed tremorgenic action of paraoxon. The present results suggest that OPs elicit kinetic tremor at least partly by activating IO neurons via nACh receptors.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Discinesia Induzida por Medicamentos/metabolismo , Paraoxon/efeitos adversos , Tremor/induzido quimicamente , Tremor/metabolismo , Animais , Encéfalo/patologia , Relação Dose-Resposta a Droga , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/patologia , Expressão Gênica/efeitos dos fármacos , Masculino , Mecamilamina/farmacologia , Camundongos , Antagonistas Muscarínicos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Antagonistas Nicotínicos/farmacologia , Proteínas Oncogênicas v-fos/metabolismo , Ratos , Receptores Nicotínicos/metabolismo , Tremor/tratamento farmacológico , Tremor/patologia , Triexifenidil/farmacologia
3.
Mol Neurobiol ; 56(6): 4037-4050, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30259400

RESUMO

L-DOPA is the main pharmacological therapy for Parkinson's disease. However, long-term exposure to L-DOPA induces involuntary movements termed dyskinesia. Clinical trials show that dyskinesia is attenuated by metabotropic glutamate receptor type 5 (mGluR5) antagonists. Further, the onset of dyskinesia is delayed by nicotine and mGluR5 expression is lower in smokers than in non-smokers. However, the mechanisms by which mGluR5 modulates dyskinesia and how mGluR5 and nicotine interact have not been established. To address these issues, we studied the role of mGluR5 in D1R-containing neurons in dyskinesia and examined whether nicotine reduces dyskinesia via mGluR5. In the aphakia mouse model of Parkinson's disease, we selectively knocked down mGluR5 in D1R-containing neurons (aphakia-mGluR5KD-D1). We found that genetic downregulation of mGluR5 decreased dyskinesia in aphakia mice. Although chronic nicotine increased the therapeutic effect of L-DOPA in both aphakia and aphakia-mGluR5KD-D1 mice, it caused a robust reduction in dyskinesia only in aphakia, and not in aphakia-mGluR5KD-D1 mice. Downregulating mGluR5 or nicotine treatment after L-DOPA decreased ERK and histone 3 activation, and FosB expression. Combining nicotine and mGluR5 knockdown did not have an added antidyskinetic effect, indicating that the effect of nicotine might be mediated by downregulation of mGluR5 expression. Treatment of aphakia-mGluR5KD-D1 mice with a negative allosteric modulator did not further modify dyskinesia, suggesting that mGluR5 in non-D1R-containing neurons does not play a role in its development. In conclusion, this work suggests that mGluR5 antagonists reduce dyskinesia by mainly affecting D1R-containing neurons and that the effect of nicotine on dyskinetic signs in aphakia mice is likely via mGluR5.


Assuntos
Afacia/complicações , Corpo Estriado/patologia , Discinesia Induzida por Medicamentos/genética , Técnicas de Silenciamento de Genes , Levodopa/efeitos adversos , Neurônios/metabolismo , Receptor de Glutamato Metabotrópico 5/genética , Receptores de Dopamina D1/metabolismo , Regulação Alostérica/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Regulação para Baixo/efeitos dos fármacos , Discinesia Induzida por Medicamentos/complicações , Discinesia Induzida por Medicamentos/patologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neurônios/patologia , Nicotina/farmacologia , Receptor de Glutamato Metabotrópico 5/metabolismo
4.
BMC Neurosci ; 19(1): 77, 2018 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-30497382

RESUMO

BACKGROUND: The treatment of Parkinson's disease is often complicated by levodopa-induced dyskinesia (LID). Nicotinic acetylcholine receptor agonists can alleviate LID in animal models but may be less effective in conditions of severe dopaminergic denervation. While the mechanisms of LID remain incompletely understood, elevated corticostriatal levels of the brain-derived neurotrophic factor (BDNF) have been suggested to play a role. Here, female mice with near-total unilateral 6-hydroxydopamine-induced nigrostriatal lesions were chronically treated with levodopa, and the effects of the α7 nicotinic receptor partial agonist AZD0328 and nicotine on LID were assessed. At the end of the experiment, BDNF protein levels in the prefrontal cortex and striatum were measured. RESULTS: Five-day treatments with three escalating doses of AZD0328 and a 10-week treatment with nicotine failed to alleviate LID. BDNF levels in the lesioned striatum correlated positively with LID severity, but no evidence was found for a levodopa-induced elevation of corticostriatal BDNF in the lesioned hemisphere. The nicotine treatment decreased BDNF levels in the prefrontal cortex but had no effect on striatal BDNF. CONCLUSIONS: The findings suggest that treatment of LID with nicotinic agonists may lose its effectiveness as the disease progresses, represent further evidence for a role for BDNF in LID, and expand previous knowledge on the effects of long-term nicotine treatment on BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dopaminérgicos/farmacologia , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/metabolismo , Agonistas Nicotínicos/farmacologia , Transtornos Parkinsonianos/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Dopamina/deficiência , Dopaminérgicos/efeitos adversos , Discinesia Induzida por Medicamentos/patologia , Feminino , Furanos/farmacologia , Levodopa/efeitos adversos , Levodopa/farmacologia , Camundongos Endogâmicos C57BL , Nicotina/farmacologia , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Quinuclidinas/farmacologia , Distribuição Aleatória , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
5.
Sci Rep ; 8(1): 15381, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30337665

RESUMO

Increasing evidence supports a close relationship between Ras-ERK1/2 activation in the striatum and L-DOPA-induced dyskinesia (LID). ERK1/2 activation by L-DOPA takes place through the crosstalk between D1R/AC/PKA/DARPP-32 pathway and NMDA/Ras pathway. Compelling genetic and pharmacological evidence indicates that Ras-ERK1/2 inhibition prevents LID onset and may even revert already established dyskinetic symptoms. However, it is currently unclear whether exacerbation of Ras-ERK1/2 activity in the striatum may further aggravate dyskinesia in experimental animal models. Here we took advantage of two genetic models in which Ras-ERK1/2 signaling is hyperactivated, the Nf1+/- mice, in which the Ras inhibitor neurofibromin is reduced, and the Ras-GRF1 overexpressing (Ras-GRF1 OE) transgenic mice in which a specific neuronal activator of Ras is enhanced. Nf1+/- and Ras-GRF1 OE mice were unilaterally lesioned with 6-OHDA and treated with an escalating L-DOPA dosing regimen. In addition, a subset of Nf1+/- hemi-parkinsonian animals was also co-treated with the Ras inhibitor lovastatin. Our results revealed that Nf1+/- and Ras-GRF1 OE mice displayed similar dyskinetic symptoms to their wild-type counterparts. This observation was confirmed by the lack of differences between mutant and wild-type mice in striatal molecular changes associated to LID (i.e., FosB, and pERK1/2 expression). Interestingly, attenuation of Ras activity with lovastatin does not weaken dyskinetic symptoms in Nf1+/- mice. Altogether, these data suggest that ERK1/2-signaling activation in dyskinetic animals is maximal and does not require further genetic enhancement in the upstream Ras pathway. However, our data also demonstrate that such a genetic enhancement may reduce the efficacy of anti-dyskinetic drugs like lovastatin.


Assuntos
Discinesia Induzida por Medicamentos/tratamento farmacológico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Levodopa/toxicidade , Lovastatina/farmacologia , Neurofibromina 1/fisiologia , Proteínas ras/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Dopaminérgicos/toxicidade , Discinesia Induzida por Medicamentos/metabolismo , Discinesia Induzida por Medicamentos/patologia , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais , Proteínas ras/genética
6.
J Neural Transm (Vienna) ; 125(8): 1287-1297, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29541852

RESUMO

Neuroinflammation is a main component of Parkinson's disease (PD) neuropathology, where unremitting reactive microglia and microglia-secreted soluble molecules such as cytokines, contribute to the neurodegenerative process as part of an aberrant immune reaction. Besides, pro-inflammatory cytokines, predominantly TNF-α, play an important neuromodulatory role in the healthy and diseased brain, being involved in neurotransmitter metabolism, synaptic scaling and brain plasticity. Recent preclinical studies have evidenced an exacerbated neuroinflammatory reaction in the striatum of parkinsonian rats that developed dyskinetic responses following L-DOPA administration. These findings prompted investigation of non-neuronal mechanisms of L-DOPA-induced dyskinesia (LID) involving glial cells and glial-secreted soluble molecules. Hence, besides the classical mechanisms of LID that include abnormal corticostriatal neurotransmission and maladaptive changes in striatal medium spiny neurons (MSNs), here we review studies supporting a role of striatal neuroinflammation in the development of LID, with a focus on microglia and the pro-inflammatory cytokine TNF-α. Moreover, we discuss several mechanisms that have been involved in the development of LID, which are directly or indirectly under the control of TNF-α, and might be abnormally affected by its chronic overproduction and release by microglia in PD. It is proposed that TNF-α may contribute to the altered neuronal responses occurring in LID by targeting receptor trafficking and function in MSNs, but also dopamine synthesis in preserved dopaminergic terminals and serotonin metabolism in serotonergic neurons. Therapeutic approaches specifically targeting glial-secreted cytokines may represent a novel target for preventing or treating LID.


Assuntos
Discinesia Induzida por Medicamentos/imunologia , Inflamação/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Antiparkinsonianos/efeitos adversos , Discinesia Induzida por Medicamentos/patologia , Humanos , Inflamação/patologia , Levodopa/efeitos adversos
7.
Mol Neurobiol ; 55(6): 5125-5136, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28840468

RESUMO

Dopamine replacement therapy with L-DOPA is the treatment of choice for Parkinson's disease; however, its long-term use is frequently associated with L-DOPA-induced dyskinesia (LID). Many molecules have been implicated in the development of LID, and several of these have been proposed as potential therapeutic targets. However, to date, none of these molecules have demonstrated full clinical efficacy, either because they lie downstream of dopaminergic signaling, or due to adverse side effects. Therefore, discovering new strategies to reduce LID in Parkinson's disease remains a major challenge. Here, we have explored the tyrosine kinase Fyn, as a novel intermediate molecule in the development of LID. Fyn, a member of the Src kinase family, is located in the postsynaptic density, where it regulates phosphorylation of the NR2B subunit of the N-methyl-D-aspartate (NMDA) receptor in response to dopamine D1 receptor stimulation. We have used Fyn knockout and wild-type mice, lesioned with 6-hydroxydopamine and chronically treated with L-DOPA, to investigate the role of Fyn in the induction of LID. We found that mice lacking Fyn displayed reduced LID, ΔFosB accumulation and NR2B phosphorylation compared to wild-type control mice. Pre-administration of saracatinib (AZD0530), an inhibitor of Fyn activity, also significantly reduced LID in dyskinetic wild-type mice. These results support that Fyn has a critical role in the molecular pathways affected during the development of LID and identify Fyn as a novel potential therapeutic target for the management of dyskinesia in Parkinson's disease.


Assuntos
Discinesia Induzida por Medicamentos/complicações , Discinesia Induzida por Medicamentos/enzimologia , Doença de Parkinson/complicações , Doença de Parkinson/enzimologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Animais , Benzodioxóis/farmacologia , Discinesia Induzida por Medicamentos/patologia , Discinesia Induzida por Medicamentos/fisiopatologia , Feminino , Levodopa , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Movimento , Neostriado/metabolismo , Neostriado/patologia , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Fosforilação , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-fyn/antagonistas & inibidores , Quinazolinas/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
8.
J Neural Transm (Vienna) ; 125(8): 1217-1223, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29264660

RESUMO

The purpose of review is to review the current status of positron emission tomography (PET) molecular imaging of serotonergic system in Parkinson's patients who experience levodopa-induced (LIDs) and graft-induced dyskinesias (GIDs). PET imaging studies have shown that Parkinson's disease is characterized by progressive loss of dopaminergic and serotonergic neurons. Parkinson's patients who experienced LIDs and GIDs have an aberrant spreading of serotonergic terminals, which lead to an increased serotonergic/dopaminergic terminals ratio within the putamen. Serotonergic terminals convert exogenous levodopa into dopamine in a non-physiological manner and release an abnormal amount of dopamine without an auto-regulatory feedback. This results in higher swings in synaptic levels of dopamine, which leads to the development of LIDs and GIDs. The modulation of serotonergic terminals with 5-HT1A and 5-HT1B receptors agonists partially reduced these motor complications. In vivo PET studies confirmed that abnormal spreading of serotonergic terminals within the putamen has a pivotal role in the development of LIDs and GIDs. However, glutamatergic, adenosinergic, opioid systems, and phosphodiesterases 10A may also play a role in the development of these motor complications. An integrative multimodal imaging approach combining PET and MRI imaging techniques is needed to fully understand the mechanisms underlying the development of LIDs and GIDs.


Assuntos
Discinesia Induzida por Medicamentos/diagnóstico por imagem , Doença de Parkinson/diagnóstico por imagem , Neurônios Serotoninérgicos/patologia , Discinesia Induzida por Medicamentos/metabolismo , Discinesia Induzida por Medicamentos/patologia , Humanos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Tomografia por Emissão de Pósitrons , Neurônios Serotoninérgicos/metabolismo
9.
Artigo em Russo | MEDLINE | ID: mdl-29076470

RESUMO

OBJECTIVE: to determine the efficacy of unilateral posteroventral pallidotomy (PVP) in the treatment of drug-induced dyskinesia (DID) in Parkinson's disease (PD). MATERIAL AND METHODS: We analyzed surgical treatment of 14 patients with PD complicated by DID who underwent unilateral PVP at the Research Center of Neurology in the period between 2012 and 2015. The clinical type of DID was mainly represented by peak-dose choreoathetoid dyskinesia, more pronounced in the distal limbs, and predominantly unilateral. The severity of drug-induced dyskinesia was assessed on the UPDRS scale (part IV-A) before surgery and at 1 week and 6 months after surgery. RESULTS: One week after pallidotomy, all of the 14 patients had a regression of contralateral dyskinesia by 68.3±9.7%; 50% of patients had a regression of ipsilateral dyskinesias by 43%, on average. In 50% of cases, the dose of levodopa was reduced by 15%, on average. On examination at 6 months after surgery, regression of contralateral dyskinesia was 55.7±8.8%, and the severity of ipsilateral DID returned to the preoperative level. The use of pallidotomy significantly improved the indicators of daily activity and quality of life of patients. There were no significant postoperative complications. Three patients had mild speech disorders in the form of dysarthria, which regressed 2-3 weeks after surgery.


Assuntos
Discinesia Induzida por Medicamentos/cirurgia , Palidotomia/métodos , Doença de Parkinson/cirurgia , Idoso , Discinesia Induzida por Medicamentos/patologia , Discinesia Induzida por Medicamentos/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia
10.
Exp Neurol ; 297: 73-81, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28757258

RESUMO

In addition to its role in neuronal survival, the brain neurotrophic factor (BDNF) has been shown to influence serotonin transmission and synaptic plasticity, events strongly implicated in the appearance of l-DOPA-induced dyskinesia (LID), a motor complication occurring in parkinsonian patients after long-term treatment with the dopamine precursor. In order to evaluate a possible influence of BDNF in the appearance of LID, 6-OHDA-lesioned rats received a striatal injection of different concentrations of an adeno-associated viral (AAV) vector over-expressing either BDNF or GFP, as control vector. Eight weeks later, animals started to receive a daily treatment with l-DOPA (4-6mg/kg plus benserazide 4-6mg/kg, s.c.) or saline, and dyskinesias, as well as l-DOPA-induced rotations, were evaluated at several time-points. Moreover, molecular changes in striatal D1 receptor-dependent cAMP/PKA and ERK/mTORC signaling pathways, as well as, sprouting of striatal serotonin axons, were measured. Results showed that the AAV-BDNF vector injection induced striatal over-expression of BDNF, as well as striatal and pallidal serotonin axon hyperinnervation. Moreover, rats that over-expressed BDNF were more prone to develop LID and l-DOPA-induced rotations, compared to the GFP-treated control group. Finally, rats that over-expressed BDNF showed increased levels of striatal D1R-dependent signaling phospho-proteins in response to l-DOPA administration. This study suggests that BDNF over-expression, by inducing changes in pre-synaptic serotonin axonal trophism, is able to exacerbate maladaptive responses to l-DOPA administration.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Corpo Estriado/metabolismo , Discinesia Induzida por Medicamentos/metabolismo , Levodopa/toxicidade , Oxidopamina/toxicidade , Neurônios Serotoninérgicos/metabolismo , Animais , Antiparkinsonianos/toxicidade , Fator Neurotrófico Derivado do Encéfalo/genética , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Discinesia Induzida por Medicamentos/patologia , Expressão Gênica , Células HEK293 , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/patologia
11.
Neurochem Res ; 41(12): 3386-3398, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27752803

RESUMO

The symptoms of Parkinsonism and oral dyskinesia have been showing to be induced by neuroleptics that significantly affect its clinical use. In this study, we investigate whether Nigella sativa-oil (NS) (black cumin seeds)-a traditional medicine used for the seizure treatment in eastern country-may reduce the haloperidol (HAL)-induced extrapyramidal symptoms (EPS)-like behavior in rats. After combine treatment with HAL (1 mg/kg) on NS (0.2 ml/rat), rats displayed a significant decreased EPS-like behavior including movement disorders and oral dyskinesia as compared to controls. Immunohistochemical analysis indicates that NS reduced astrogliosis in caudate and accumbens nuclei. These results suggest that NS may consider as an adjunct to antipsychotics to reduce the EPS-like side effect.


Assuntos
Antipsicóticos/efeitos adversos , Doenças dos Gânglios da Base/tratamento farmacológico , Discinesia Induzida por Medicamentos/tratamento farmacológico , Haloperidol/efeitos adversos , Nigella sativa/química , Óleos de Plantas/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Doenças dos Gânglios da Base/induzido quimicamente , Doenças dos Gânglios da Base/patologia , Núcleo Caudado/efeitos dos fármacos , Núcleo Caudado/patologia , Discinesia Induzida por Medicamentos/patologia , Gliose/induzido quimicamente , Gliose/tratamento farmacológico , Gliose/patologia , Masculino , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/patologia , Ratos Wistar , Discinesia Tardia/tratamento farmacológico , Discinesia Tardia/patologia
12.
Neuroscience ; 332: 149-59, 2016 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-27365175

RESUMO

Short-term exposure to nicotine induces positive effects in mice, monkeys and humans, including mild euphoria, hyperactivity, and enhanced cognition. However, the underlying neural basis and molecular mechanisms for these effects remain poorly understood. Here, using a video recording system, we find that acute nicotine administration induces locomotor hyperactivity in Drosophila, similar to observations made in higher model organisms. Suppressing dopaminergic neurons or down-regulating dopamine 1-like receptor (DopR) abolishes this acute nicotine response, but surprisingly, does so only in male flies. Using a GFP reconstitution across synaptic partners (GRASP) approach, we show that dopaminergic neurons possess potential synaptic connections with acetylcholinergic neurons in wide regions of the brain. Furthermore, dopaminergic neurons are widely activated upon nicotine perfusion in both sexes, while the response curve differs significantly between the sexes. Moreover, knockdown of the ß1 nicotine acetylcholine receptor (nAChR) in dopaminergic neurons abolishes the acute nicotine response only in male flies, while panneural knock-down occurs in both sexes. Taken together, our results reveal that in fruit flies, dopaminergic neurons mediate nicotine-induced acute locomotor hyperactivity in a sexually dimorphic manner, and Drosophila ß1 nAChR subunit plays a crucial role in this nicotine response. These findings provide important insights into the molecular and neural basis of acute nicotine effects, and the underlying mechanisms may play conserved roles across species.


Assuntos
Discinesia Induzida por Medicamentos/fisiopatologia , Atividade Motora/efeitos dos fármacos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Caracteres Sexuais , Animais , Animais Geneticamente Modificados , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/patologia , Neurônios Colinérgicos/fisiologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Neurônios Dopaminérgicos/fisiologia , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Discinesia Induzida por Medicamentos/patologia , Feminino , Técnicas de Silenciamento de Genes , Masculino , Atividade Motora/fisiologia , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo
13.
Neurobiol Dis ; 89: 169-79, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26875664

RESUMO

The dopamine precursor 3,4-dihydroxyphenyl-l-alanine (L-DOPA) is currently the most efficacious pharmacotherapy for Parkinson's disease (PD). However, long-term L-DOPA treatment leads to the development of abnormal involuntary movements (AIMs) in patients and animal models of PD. Recently, involvement of growth arrest and DNA damage-inducible 45ß (Gadd45ß) was reported in neurological and neurobehavioral dysfunctions. However, little is known about the role of Gadd45ß in the dopaminergic nigrostriatal pathway or L-DOPA-induced dyskinesia (LID). To address this issue, we prepared an animal model of PD using unilateral 6-hydroxydopamine (6-OHDA) lesions in the substantia nigra of Gadd45ß(+/+) and Gadd45ß(-/-) mice. Dyskinetic symptoms were triggered by repetitive administration of L-DOPA in these 6-OHDA-lesioned mice. Whereas dopamine denervation in the dorsal striatum decreased Gadd45ß mRNA, chronic L-DOPA treatment significantly increased Gadd45ß mRNA expression in the 6-OHDA-lesioned striatum of wild-type mice. Using unilaterally 6-OHDA-lesioned Gadd45ß(+/+) and Gadd45ß(-/-) mice, we found that mice lacking Gadd45ß exhibited long-lasting increases in AIMs following repeated administration of L-DOPA. By contrast, adeno-associated virus-mediated expression of Gadd45ß in the striatum reduced AIMs in Gadd45ß knockout mice. The deficiency of Gadd45ß in LID increased expression of ΔFosB and c-Fos in the lesioned striatum 90 min after the last administration of L-DOPA following 11days of daily L-DOPA treatments. These data suggest that the increased expression of Gadd45ß induced by repeated administration of L-DOPA may be beneficial in patients with PD.


Assuntos
Antígenos de Diferenciação/metabolismo , Discinesia Induzida por Medicamentos/metabolismo , Levodopa/toxicidade , Transtornos Parkinsonianos/metabolismo , Animais , Antígenos de Diferenciação/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Discinesia Induzida por Medicamentos/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Oxidopamina , Transtornos Parkinsonianos/patologia , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/metabolismo , Substância Negra/metabolismo , Substância Negra/patologia
14.
Neuroscience ; 310: 12-26, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26363150

RESUMO

Treatment of Parkinson's disease (PD) with dopamine replacement relieves symptoms of poverty of movement, but often causes drug-induced dyskinesias. Accumulating clinical and pre-clinical evidence suggests that the primary motor cortex (M1) is involved in the pathophysiology of PD and that modulating cortical activity may be a therapeutic target in PD and dyskinesia. However, surprisingly little is known about how M1 neurotransmitter tone or gene expression is altered in PD, dyskinesia or associated animal models. The present study utilized the rat unilateral 6-hydroxydopamine (6-OHDA) model of PD/dyskinesia to characterize structural and functional changes taking place in M1 monoamine innervation and gene expression. 6-OHDA caused dopamine pathology in M1, although the lesion was less severe than in the striatum. Rats with 6-OHDA lesions showed a PD motor impairment and developed dyskinesia when given L-DOPA or the D1 receptor agonist, SKF81297. M1 expression of two immediate-early genes (c-Fos and ARC) was strongly enhanced by either L-DOPA or SKF81297. At the same time, expression of genes specifically involved in glutamate and GABA signaling were either modestly affected or unchanged by lesion and/or treatment. We conclude that M1 neurotransmission and signal transduction in the rat 6-OHDA model of PD/dyskinesia mirror features of human PD, supporting the utility of the model to study M1 dysfunction in PD and the elucidation of novel pathophysiological mechanisms and therapeutic targets.


Assuntos
Complexo Relacionado com a AIDS/metabolismo , Discinesia Induzida por Medicamentos/patologia , Expressão Gênica/fisiologia , Córtex Motor/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transmissão Sináptica/fisiologia , Complexo Relacionado com a AIDS/genética , Animais , Benzazepinas/efeitos adversos , Modelos Animais de Doenças , Dopaminérgicos/efeitos adversos , Discinesia Induzida por Medicamentos/etiologia , Lateralidade Funcional/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Levodopa/uso terapêutico , Masculino , Feixe Prosencefálico Mediano/lesões , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Transmissão Sináptica/efeitos dos fármacos
15.
Neuroscience ; 298: 302-17, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25892702

RESUMO

L-3,4-Dihydroxyphenylalanine (L-DOPA) is the therapeutic gold standard in Parkinson's disease. However, long-term treatment is complicated by the induction of debilitating abnormal involuntary movements termed L-DOPA-induced dyskinesias (LIDs). Until today the underlying mechanisms of LID pathogenesis are not fully understood. The aim of this study was to reveal new factors, which may be involved in the induction of LID. We have focused on the expression of striatal tyrosine hydroxylase-positive (TH+) neurons, which are capable of producing either L-DOPA or dopamine (DA) in target areas of ventral midbrain DAergic neurons. To address this issue, a daily L-DOPA dose was administered over the course of 15 days to mice with unilateral 6-hydroxydopamine-induced lesions of the medial forebrain bundle and LIDs were evaluated. Remarkably, the number of striatal TH+ neurons strongly correlated with both induction and severity of LID as well as ΔFosB expression as an established molecular marker for LID. Furthermore, dyskinetic mice showed a marked augmentation of serotonergic fiber innervation in the striatum, enabling the decarboxylation of L-DOPA to DA. Axial, limb and orolingual dyskinesias were predominantly associated with TH+ neurons in the lateral striatum, whereas medially located TH+ neurons triggered locomotive rotations. In contrast, identified accumbal and cortical TH+ cells did not contribute to the generation of LID. Thus, striatal TH+ cells and serotonergic terminals may cooperatively synthesize DA and subsequently contribute to supraphysiological synaptic DA concentrations, an accepted cause in LID pathogenesis.


Assuntos
Corpo Estriado/patologia , Discinesia Induzida por Medicamentos/patologia , Lateralidade Funcional/fisiologia , Neurônios/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Anfetamina/farmacologia , Animais , Antiparkinsonianos/efeitos adversos , Modelos Animais de Doenças , Discinesia Induzida por Medicamentos/etiologia , Levodopa/efeitos adversos , Masculino , Feixe Prosencefálico Mediano/efeitos dos fármacos , Feixe Prosencefálico Mediano/lesões , Camundongos , Camundongos Endogâmicos C57BL , Oxidopamina/toxicidade , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/etiologia , Fosfopiruvato Hidratase/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Estatísticas não Paramétricas
16.
Cell Mol Life Sci ; 72(11): 2107-17, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25681866

RESUMO

Levodopa-induced dyskinesias (LIDs) occur in the majority of patients with Parkinson's disease (PD) following years of levodopa treatment. The pathophysiology underlying LIDs in PD is poorly understood, and current treatments generate only minor benefits for the patients. Studies with positron emission tomography (PET) molecular imaging have demonstrated that in advanced PD patients, levodopa administration induces sharp increases in striatal dopamine levels, which correlate with LIDs severity. Fluctuations in striatal dopamine levels could be the result of the attenuated buffering ability in the dopaminergically denervated striatum. Lines of evidence from PET studies indicate that serotonergic terminals could also be responsible for the development of LIDs in PD by aberrantly processing exogenous levodopa and by releasing dopamine in a dysregulated manner from the serotonergic terminals. Additionally, other downstream mechanisms involving glutamatergic, cannabinoid, opioid, cholinergic, adenosinergic, and noradrenergic systems may contribute in the development of LIDs. In this article, we review the findings from preclinical, clinical, and molecular imaging studies, which have contributed to our understanding the pathophysiology of LIDs in PD.


Assuntos
Antiparkinsonianos/efeitos adversos , Discinesia Induzida por Medicamentos/patologia , Levodopa/efeitos adversos , Antiparkinsonianos/uso terapêutico , Dopamina/metabolismo , Dopaminérgicos/efeitos adversos , Dopaminérgicos/uso terapêutico , Humanos , Levodopa/uso terapêutico , Imagem Molecular , Doença de Parkinson/tratamento farmacológico , Receptores Dopaminérgicos/metabolismo
17.
Int Rev Neurobiol ; 119: 151-67, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25175965

RESUMO

Altered dopaminergic neurotransmission in the basal ganglia is observed in Parkinson's disease (PD) and L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesias (LID). An attractive alternative for treating LID is to use adjunct drugs to modulate nondopaminergic neurotransmitter systems in the basal ganglia. For example, adenosine receptors have received attention over the past years for the treatment of PD and LID. Adenosine interacts closely with dopamine and plays an important role in the function of striatal GABAergic efferent neurons. Excitatory glutamatergic neurotransmission is also modulated by adenosine in the striatum. Hence, based on the unique cellular and regional distribution of this system, adenosine neurotransmission could have an important implication for the development of new therapeutic strategies targeting the basal ganglia disorders. Indeed, A2A adenosine receptor antagonists were shown to improve motor deficits in PD and to reduce the severity of LID. A2A receptor subtypes are selectively found on striatopallidal neurons and can couple with receptors of interest in PD, such as D2 dopamine and metabotropic glutamate receptor type 5 (mGlu5) receptors, and form functional heteromeric complexes. This chapter will review relevant studies investigating the role and contribution of adenosine receptor subtypes in pathophysiology of PD and LID. The interactions of adenosine receptors, especially A1 and A2A receptor subtypes, with other receptors implicated in the pathophysiology of PD and LID such as dopaminergic and glutamatergic receptors will be reviewed. The implication of these interactions in the development and expression of PD symptoms and LID needs further investigation to find novel drug targets.


Assuntos
Antiparkinsonianos/uso terapêutico , Neurônios GABAérgicos/metabolismo , Doença de Parkinson/terapia , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Gânglios da Base/patologia , Discinesia Induzida por Medicamentos/patologia , Neurônios GABAérgicos/efeitos dos fármacos , Humanos , Levodopa/efeitos adversos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Receptores Dopaminérgicos/metabolismo
18.
Neurobiol Dis ; 71: 334-44, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25171793

RESUMO

Parkinsonian symptoms arise due to over-activity of the indirect striatal output pathway, and under-activity of the direct striatal output pathway. l-DOPA-induced dyskinesia (LID) is caused when the opposite circuitry problems are established, with the indirect pathway becoming underactive, and the direct pathway becoming over-active. Here, we define synaptic plasticity abnormalities in these pathways associated with parkinsonism, symptomatic benefits of l-DOPA, and LID. We applied spike-timing dependent plasticity protocols to cortico-striatal synapses in slices from 6-OHDA-lesioned mouse models of parkinsonism and LID, generated in BAC transgenic mice with eGFP targeting the direct or indirect output pathways, with and without l-DOPA present. In naïve mice, bidirectional synaptic plasticity, i.e. LTP and LTD, was induced, resulting in an EPSP amplitude change of approximately 50% in each direction in both striatal output pathways, as shown previously. In parkinsonism and dyskinesia, both pathways exhibited unidirectional plasticity, irrespective of stimulation paradigm. In parkinsonian animals, the indirect pathway only exhibited LTP (LTP protocol: 143.5±14.6%; LTD protocol 177.7±22.3% of baseline), whereas the direct pathway only showed LTD (LTP protocol: 74.3±4.0% and LTD protocol: 63.3±8.7%). A symptomatic dose of l-DOPA restored bidirectional plasticity on both pathways to levels comparable to naïve animals (Indirect pathway: LTP protocol: 124.4±22.0% and LTD protocol: 52.1±18.5% of baseline. Direct pathway: LTP protocol: 140.7±7.3% and LTD protocol: 58.4±6.0% of baseline). In dyskinesia, in the presence of l-DOPA, the indirect pathway exhibited only LTD (LTP protocol: 68.9±21.3% and LTD protocol 52.0±14.2% of baseline), whereas in the direct pathway, only LTP could be induced (LTP protocol: 156.6±13.2% and LTD protocol 166.7±15.8% of baseline). We conclude that normal motor control requires bidirectional plasticity of both striatal outputs, which underlies the symptomatic benefits of l-DOPA. Switching from bidirectional to unidirectional plasticity drives global changes in striatal pathway excitability, and underpins parkinsonism and dyskinesia.


Assuntos
Antiparkinsonianos/efeitos adversos , Corpo Estriado/patologia , Discinesia Induzida por Medicamentos/patologia , Levodopa/efeitos adversos , Vias Neurais/patologia , Plasticidade Neuronal/fisiologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , Discinesia Induzida por Medicamentos/etiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Lateralidade Funcional , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal/efeitos dos fármacos , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Purinérgicos/farmacologia , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
19.
PLoS One ; 9(3): e90759, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24614598

RESUMO

Dyskinesia is a major side effect of an otherwise effective L-DOPA treatment in Parkinson's patients. The prevailing view for the underlying presynaptic mechanism of L-DOPA-induced dyskinesia (LID) suggests that surges in dopamine (DA) via uncontrolled release from serotonergic terminals results in abnormally high level of extracellular striatal dopamine. Here we used high-sensitivity online microdialysis and PET imaging techniques to directly investigate DA release properties from serotonergic terminals both in the parkinsonian striatum and after neuronal transplantation in 6-OHDA lesioned rats. Although L-DOPA administration resulted in a drift in extracellular DA levels, we found no evidence for abnormally high striatal DA release from serotonin neurons. The extracellular concentration of DA remained at or below levels detected in the intact striatum. Instead, our results showed that an inefficient release pool of DA associated with low D2 receptor binding remained unchanged. Taken together, these findings suggest that differential DA receptor activation rather than excessive release could be the underlying mechanism explaining LID seen in this model. Our data have important implications for development of drugs targeting the serotonergic system to reduce DA release to manage dyskinesia in patients with Parkinson's disease.


Assuntos
Discinesia Induzida por Medicamentos/metabolismo , Levodopa/efeitos adversos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Receptores Dopaminérgicos/metabolismo , Animais , Benzamidas , Modelos Animais de Doenças , Dopamina/metabolismo , Discinesia Induzida por Medicamentos/patologia , Espaço Extracelular/metabolismo , Feminino , Levodopa/administração & dosagem , Levodopa/farmacologia , Levodopa/uso terapêutico , Microdiálise , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Nomifensina/uso terapêutico , Doença de Parkinson/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Proteínas Proto-Oncogênicas c-fos/metabolismo , Pirrolidinas , Ratos Sprague-Dawley , Serotonina/metabolismo
20.
PLoS One ; 9(3): e92605, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24663062

RESUMO

BACKGROUND: RGS9-deficient mice show drug-induced dyskinesia but normal locomotor activity under unchallenged conditions. RESULTS: Genes related to Ca2+ signaling and their functions were regulated in RGS9-deficient mice. CONCLUSION: Changes in Ca2+ signaling that compensate for RGS9 loss-of-function can explain the normal locomotor activity in RGS9-deficient mice under unchallenged conditions. SIGNIFICANCE: Identified signaling components may represent novel targets in antidyskinetic therapy. The long splice variant of the regulator of G-protein signaling 9 (RGS9-2) is enriched in striatal medium spiny neurons and dampens dopamine D2 receptor signaling. Lack of RGS9-2 can promote while its overexpression prevents drug-induced dyskinesia. Other animal models of drug-induced dyskinesia rather pointed towards overactivity of dopamine receptor-mediated signaling. To evaluate changes in signaling pathways mRNA expression levels were determined and compared in wild-type and RGS9-deficient mice. Unexpectedly, expression levels of dopamine receptors were unchanged in RGS9-deficient mice, while several genes related to Ca2+ signaling and long-term depression were differentially expressed when compared to wild type animals. Detailed investigations at the protein level revealed hyperphosphorylation of DARPP32 at Thr34 and of ERK1/2 in striata of RGS9-deficient mice. Whole cell patch clamp recordings showed that spontaneous synaptic events are increased (frequency and size) in RGS9-deficient mice while long-term depression is reduced in acute brain slices. These changes are compatible with a Ca2+-induced potentiation of dopamine receptor signaling which may contribute to the drug-induced dyskinesia in RGS9-deficient mice.


Assuntos
Regulação da Expressão Gênica , Neostriado/metabolismo , Proteínas RGS/deficiência , Receptores de Dopamina D2/metabolismo , Animais , Sinalização do Cálcio , AMP Cíclico/metabolismo , Discinesia Induzida por Medicamentos/genética , Discinesia Induzida por Medicamentos/metabolismo , Discinesia Induzida por Medicamentos/patologia , Discinesia Induzida por Medicamentos/fisiopatologia , Depressão Sináptica de Longo Prazo , Masculino , Camundongos , Plasticidade Neuronal , Fosforilação , Sinapses/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA