Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 586
Filtrar
1.
Biomed Pharmacother ; 139: 111525, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33882412

RESUMO

3-Hydroxytyrosol (HXT) is a natural polyphenol present in extra virgin olive oil. It is a key component of Mediterranean diet and is known for its strong antioxidant activity. The present study evaluated the potential of HXT as an anti-parkinsonian molecule in terms of its ability to inhibit MAO-B and thereby maintaining dopamine (DA) levels in Parkinson's disease (PD). In-silico molecular docking study followed by MMGBSA binding free energy calculation revealed that HXT has a strong binding affinity for MAO-B in comparison to MAO-A. Moreover, rasagiline and HXT interacted with the similar binding sites and modes of interactions. Additionally, molecular dynamics simulation studies revealed stable nature of HXT-MAO-B interaction and also provided information about the amino acid residues involved in binding. Moreover, in vitro studies revealed that HXT inhibited MAO-B in human platelets with IC50 value of 7.78 µM. In vivo studies using MPTP-induced mouse model of PD revealed increase in DA levels with concomitant decrease in DA metabolites (DOPAC and HVA) on HXT treatment. Furthermore, MAO-B activity was also inhibited on HXT administration to PD mice. In addition, HXT treatment prevented MPTP-induced loss of DA neurons in substantia nigra and their nerve terminals in the striatum. HXT also attenuated motor impairments in PD mice assessed by catalepsy bar, narrow beam walk and open field tests. Thus, the present findings reveal HXT as a potential inhibitor of MAO-B, which may be used as a lead molecule for the development of therapeutics for PD.


Assuntos
Antiparkinsonianos/farmacologia , Intoxicação por MPTP/tratamento farmacológico , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/tratamento farmacológico , Álcool Feniletílico/análogos & derivados , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/enzimologia , Simulação por Computador , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Humanos , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Monoaminoxidase/metabolismo , Neostriado/efeitos dos fármacos , Neostriado/patologia , Azeite de Oliva/química , Doença de Parkinson Secundária/patologia , Álcool Feniletílico/farmacologia , Substância Negra/efeitos dos fármacos , Substância Negra/patologia
2.
Purinergic Signal ; 17(2): 247-254, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33548045

RESUMO

6-Hydroxydopamine (6-OHDA) is the most used toxin in experimental Parkinson's disease (PD) models. 6-OHDA shows high affinity for the dopamine transporter and once inside the neuron, it accumulates and undergoes non-enzymatic auto-oxidation, promoting reactive oxygen species (ROS) formation and selective damage of catecholaminergic neurons. In this way, our group has established a 6-OHDA in vitro protocol with rat striatal slices as a rapid and effective model for screening of new drugs with protective effects against PD. We have shown that co-incubation with guanosine (GUO, 100 µM) prevented the 6-OHDA-induced damage in striatal slices. As the exact GUO mechanism of action remains unknown, the aim of this study was to investigate if adenosine A1 (A1R) and/or A2A receptors (A2AR) are involved on GUO protective effects on striatal slices. Pre-incubation with DPCPX, an A1R antagonist prevented guanosine effects on 6-OHDA-induced ROS formation and mitochondrial membrane potential depolarization, while CCPA, an A1R agonist, did not alter GUO effects. Regarding A2AR, the antagonist SCH58261 had similar protective effect as GUO in ROS formation and mitochondrial membrane potential. Additionally, SCH58261 did not affect GUO protective effects. The A2AR agonist CGS21680, although, completely blocked GUO effects. Finally, the A1R antagonist DPCPX, and the A2AR agonist CGS21680 also abolished the preventive guanosine effect on 6-OHDA-induced ATP levels decrease. These results reinforce previous evidence for a putative interaction of GUO with A1R-A2AR heteromer as its molecular target and clearly indicate a dependence on adenosine receptors modulation to GUO protective effect.


Assuntos
Guanosina/farmacologia , Doenças Mitocondriais/prevenção & controle , Neostriado/metabolismo , Fármacos Neuroprotetores/farmacologia , Oxidopamina/toxicidade , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/efeitos dos fármacos , Explosão Respiratória/efeitos dos fármacos , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Avaliação Pré-Clínica de Medicamentos , Técnicas In Vitro , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Xantinas/uso terapêutico
3.
Am J Physiol Endocrinol Metab ; 319(4): E734-E743, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32865008

RESUMO

Autophagy dysfunctions are involved in the pathogenesis of Parkinson's disease (PD). In the present study, we aimed to evaluate the involvement of G protein-coupled estrogen receptor (GPER) in the inhibitory effect of insulin-like growth factor-1 (IGF-1) against excessive autophagy in PD animal and cellular models. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment significantly induced mouse movement disorder and decreased the protein level of tyrosine hydroxylase (TH) in the substantia nigra (SN) and dopamine (DA) content in striatum. Along with the dopamine neuron injury, we observed significant upregulations of microtubule-associated light chain-3 II (LC3-II) and α-synuclein as well as a downregulation of P62 in MPTP-treated mice. These changes could be restored by IGF-1 pretreatment. Cotreatment with IGF-1R antagonist JB-1 or GPER antagonist G15 could block the neuroprotective effects of IGF-1. 1-Methy-4-phenylpyridinium (MPP+) treatment could also excessively activate autophagy along with the reduction of cell viability in SH-SY5Y cells. IGF-1 could inhibit the neurotoxicity through promoting the phosphorylation of Akt and mammalian target of rapamycin (mTOR), which could also be antagonized by JB-1 or G15. These data suggest that IGF-1 inhibits MPTP/MPP+-induced autophagy on dopaminergic neurons through the IGF-1R/PI3K-Akt-mTOR pathway and GPER.


Assuntos
Autofagia/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Intoxicação por MPTP/prevenção & controle , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Humanos , Intoxicação por MPTP/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Equilíbrio Postural/efeitos dos fármacos , Receptor IGF Tipo 1 , Serina-Treonina Quinases TOR/metabolismo
4.
Addict Biol ; 25(3): e12757, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-30969011

RESUMO

Nicotine is recognized as one of the most addictive drugs, which in part could be attributed to progressive neuroadaptations and rewiring of dorsal striatal circuits. Since motor-skill learning produces neuroplasticity in the same circuits, we postulate that rotarod training could be sufficient to block nicotine-induced rewiring and thereby prevent long-lasting impairments of neuronal functioning. To test this hypothesis, Wistar rats were subjected to 15 days of treatment with either nicotine (0.36 mg/kg) or vehicle. After treatment, a subset of animals was trained on the rotarod. Ex vivo electrophysiology was performed 1 week after the nicotine treatment period and after up to 3 months of withdrawal to define neurophysiological transformations in circuits of the striatum and amygdala. Our data demonstrate that nicotine alters striatal neurotransmission in a distinct temporal and spatial sequence, where acute transformations are initiated in dorsomedial striatum (DMS) and nucleus accumbens (nAc) core. Following 3 months of withdrawal, synaptic plasticity in the form of endocannabinoid-mediated long-term depression (eCB-LTD) is impaired in the dorsolateral striatum (DLS), and neurotransmission is altered in DLS, nAc shell, and the central nucleus of the amygdala (CeA). Training on the rotarod, performed after nicotine treatment, blocks neurophysiological transformations in striatal subregions, and prevents nicotine-induced impairment of eCB-LTD. These datasets suggest that nicotine-induced rewiring of striatal circuits can be extinguished by other behaviors that induce neuroplasticity. It remains to be determined if motor-skill training could be used to prevent escalating patterns of drug use in experienced users or facilitate the recovery from addiction.


Assuntos
Aprendizagem/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Destreza Motora , Neostriado/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Animais , Corpo Estriado/efeitos dos fármacos , Endocanabinoides , Masculino , Neostriado/metabolismo , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Teste de Desempenho do Rota-Rod , Transmissão Sináptica/efeitos dos fármacos
5.
Physiol Behav ; 215: 112787, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31866232

RESUMO

Phytoestrogens are plant-derived compounds that can modulate estrogen activity in the brain and periphery. Laboratory rodent diets are typically high in soy-based phytoestrogens and therefore may influence neurophysiological and behavioural measures that are sensitive to estrogen signaling. Here we assessed such measures in rats (males and females) fed Australian made diets that varied in their soy levels. We found that a low-soy diet promoted greater weight, and lower levels of plasma estradiol, particularly in male rats. It also produced sex-specific effects on estrogen receptor gene expression in the brain, increasing ESR2 expression in the hippocampus and prefrontal cortex in female rats, and decreasing dopamine D1 receptor gene expression in the striatum of both male and female rats. We also found a dietary effect on short-term place recognition memory, but this was independent of soy levels in the diet. These results demonstrate that the choice of rodent laboratory diet can influence physiology, neurobiology and behavior, particularly on measures related to estrogen signaling.


Assuntos
Dieta , Estrogênios/fisiologia , Transdução de Sinais/fisiologia , Memória Espacial/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Ciclo Estral , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Fitoestrógenos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/biossíntese , Receptores de Dopamina D1/genética , Caracteres Sexuais , Alimentos de Soja
6.
Neurochem Int ; 131: 104583, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31654678

RESUMO

Several studies have suggested the association between neurodegenerative diseases and diabetes mellitus (DM), DM causes cognitive impairment with age, but its effect is not well known in Parkinson's disease (PD). As a member of the incretin family, Glucagon-like peptide-1 (GLP-1) has glycemic regulation functions. It also exerts many additional effects on different tissues through its receptor's widespread expression. OBJECTIVE: our aim is to investigate the effect of pre-existing diabetes on the severity of PD in male albino rats, and to find out whether GLP-1 could improve PD symptoms in diabetic animals in addition to its hypoglycemic effect, and how it could do that. METHODS: 75 adult male albino rats were equally divided into: Control, Parkinson's, Diabetic Parkinson's, Diabetic Parkinson's + low dose exenatide (GLP-1 receptor agonist), Diabetic Parkinson's + high dose exenatide group. Blood glucose and insulin, striatal dopamine, some striatal oxidative stress and inflammatory markers, and the catalepsy score were measured. RESULTS: Pre-existing of diabetes before initiation of PD raises the severity of PD shown by the more significant increase in catalepsy score, and the more significant decrease in striatal dopamine level. GLP-1 effects extend beyond their hypoglycemic effects only since it has a direct anti-oxidant, and anti-inflammatory neuronal effect with increasing the striatal dopamine and improving the catalepsy score in a dose dependent manner. CONCLUSIONS: Diabetes increases the severity of impairment in PD, and GLP-1 improve it through its direct neuronal effect in addition to its indirect effect through producing hypoglycemia.


Assuntos
Complicações do Diabetes/tratamento farmacológico , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Animais , Comportamento Animal , Glicemia/metabolismo , Catalepsia/induzido quimicamente , Catalepsia/psicologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/psicologia , Dopamina/metabolismo , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/agonistas , Peptídeo 1 Semelhante ao Glucagon/antagonistas & inibidores , Insulina/sangue , Interleucina-1beta/metabolismo , Masculino , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Estresse Oxidativo , Doença de Parkinson/psicologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa
7.
Neurochem Int ; 131: 104543, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31491493

RESUMO

Prolonged administration of Levodopa (L-dopa) therapy can generate L-dopa-induced dyskinesia (LID). Accumulating evidence indicates that hyper-activation of the dopamine D1 receptor (D1R) and the cAMP signaling cascade in the medium spiny neurons (MSNs) of the striatum are involved in LID. Previous studies have shown that striatal ß-arrestin2 overexpression significantly reduces LID severity and have indicated that ß-arrestin2 may play a causal role in the dyskinesia sensitization process. L-dopa-induced changes in the expression of signaling molecules and other proteins in the striatum were examined immunohistochemically and by western blot. A rAAV (recombinant adeno-associated virus) vector was used to overexpress and ablate ß-arrestin2. We found that striatal overexpression of AAV-mediated ß-arrestin2 produced less severe AIMs (abnormal involuntary movements) in response to L-dopa, whereas selective deletion of ß-arrestin2 in the striatal neurons dramatically enhanced the severity of dyskinesia induced by L-dopa. Furthermore, no significant improvements in motor behavior (FFT: forelimb functional test) were seen with the inhibition or overexpression of ß-arrestin2. Finally, overexpression of ß-arrestin2 diminished L-dopa-induced D1R and phosphor-DARPP32/ERK levels. Viral deletion of ß-arrestin2 markedly enhanced the key biochemical markers in the direct pathway. We found that increased availability of ß-arrestin2 ameliorated dyskinesia severity with no influence on the anti-Parkinsonian action of L-dopa, suggesting a promising approach for controlling LID in Parkinson's disease. In addition, overexpression of ß-Arrestin2 prevented the development of LID by inhibiting G protein-dependent D1R and phosphor-DARPP32/ERK signaling in dyskinetic rats.


Assuntos
Antiparkinsonianos , Discinesia Induzida por Medicamentos/terapia , Levodopa , Neostriado/metabolismo , Doença de Parkinson Secundária/terapia , beta-Arrestina 2/biossíntese , beta-Arrestina 2/genética , Adenoviridae/genética , Animais , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Discinesia Induzida por Medicamentos/psicologia , Deleção de Genes , Terapia Genética , Vetores Genéticos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Neostriado/efeitos dos fármacos , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/psicologia , Fosfoproteínas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fatores de Transcrição/efeitos dos fármacos
8.
Neuron ; 103(6): 1056-1072.e6, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31324539

RESUMO

Motor and cognitive functions depend on the coordinated interactions between dopamine (DA) and acetylcholine (ACh) at striatal synapses. Increased ACh availability was assumed to accompany DA deficiency based on the outcome of pharmacological treatments and measurements in animals that were critically depleted of DA. Using Slc6a3DTR/+ diphtheria-toxin-sensitive mice, we demonstrate that a progressive and L-dopa-responsive DA deficiency reduces ACh availability and the transcription of hyperpolarization-activated cation (HCN) channels that encode the spike timing of ACh-releasing tonically active striatal interneurons (ChIs). Although the production and release of ACh and DA are reduced, the preponderance of ACh over DA contributes to the motor deficit. The increase in striatal ACh relative to DA is heightened via D1-type DA receptors that activate ChIs in response to DA release from residual axons. These results suggest that stabilizing the expression of HCN channels may improve ACh-DA reciprocity and motor function in Parkinson's disease (PD). VIDEO ABSTRACT.


Assuntos
Acetilcolina/metabolismo , Neurônios Colinérgicos/metabolismo , Dopamina/deficiência , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Interneurônios/metabolismo , Neostriado/metabolismo , Doença de Parkinson/metabolismo , Anfetamina/farmacologia , Animais , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Dopaminérgicos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Camundongos , Neostriado/citologia , Neostriado/efeitos dos fármacos , Neostriado/fisiopatologia , Doença de Parkinson/fisiopatologia , Técnicas de Patch-Clamp , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Transcrição Gênica
9.
Int J Mol Sci ; 20(7)2019 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-30934738

RESUMO

Parkinson's disease (PD), a multifactorial movement disorder that involves progressive degeneration of the nigrostriatal system affecting the movement ability of the patient. Oxidative stress and neuroinflammation both are shown to be involved in the etiopathogenesis of PD. The aim of this study was to evaluate the therapeutic potential of thymol, a dietary monoterpene phenol in rotenone (ROT)-induced neurodegeneration in rats that precisely mimics PD in humans. Male Wistar rats were injected ROT at a dose of 2.5 mg/kg body weight for 4 weeks, to induce PD. Thymol was co-administered for 4 weeks at a dose of 50 mg/kg body weight, 30 min prior to ROT injection. The markers of dopaminergic neurodegeneration, oxidative stress and inflammation were estimated using biochemical assays, enzyme-linked immunosorbent assay, western blotting and immunocytochemistry. ROT challenge increased the oxidative stress markers, inflammatory enzymes and cytokines as well as caused significant damage to nigrostriatal dopaminergic system of the brain. Thymol treatment in ROT challenged rats appears to significantly attenuate dopaminergic neuronal loss, oxidative stress and inflammation. The present study showed protective effects of thymol in ROT-induced neurotoxicity and neurodegeneration mediated by preservation of endogenous antioxidant defense networks and attenuation of inflammatory mediators including cytokines and enzymes.


Assuntos
Dieta , Neurônios Dopaminérgicos/patologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Timol/uso terapêutico , Animais , Catalase/metabolismo , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Glutationa/metabolismo , Mediadores da Inflamação/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Neostriado/efeitos dos fármacos , Neostriado/patologia , Degeneração Neural/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos Wistar , Rotenona , Substância Negra/efeitos dos fármacos , Substância Negra/patologia , Superóxido Dismutase/metabolismo , Timol/química , Timol/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Exp Neurol ; 316: 52-62, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30981804

RESUMO

OCT4 is a key regulator in maintaining the pluripotency and self-renewal of embryonic stem cells (ESCs). Human OCT4 gene has three mRNA isoforms, termed OCT4A, OCT4B and OCT4B1. The 190-amino-acid protein isoform (OCT4B-190) is one of the major products of OCT4B mRNA, the biological function of which is still not well defined. Recent evidence suggests that OCT4B-190 may function in the cellular stress response. The glycogen synthase kinase-3ß (GSK-3ß) and histone deacetylase 6 (HDAC6) are also key stress modulators that play critical roles in the ischemic cascades of stroke. Hence, we here further investigated the effects of OCT4B-190 in the experimental stroke, and explored the underlying roles of GSK-3ß and HDAC6. We found that OCT4B-190 overexpression enhanced neuronal viability at 24 h after oxygen-glucose deprivation (OGD) treatment. Moreover, in male C57BL/6 mice subjected to transit middle cerebral artery occlusion (MCAO), OCT4B-190 overexpression reduced infarct volume and improved neurological function after stroke. Notably, we found spatio-temporal alterations of GSK-3ß and HDAC6 in the ischemic cortex and striatum, which were affected by adenovirus-mediated OCT4B-190 overexpression. OCT4B-190 demonstrated similar impacts on neuronal cultures in vitro, downregulating OGD-induced GSK-3ß activity and HDAC6 expression. In addition, we found that GSK-3ß and HDAC6 were co-expressed in the cytoplasm of neurons, and OCT4B-190 had an effect on interactions between GSK-3ß and HDAC6 in neuronal cultures subjected to OGD treatment. These findings suggest that OCT4B-190 exerts neuroprotection in the experimental stroke potentially by regulating actions of GSK-3ß and HDAC6 simultaneously, which may be an attractive therapeutic strategy for ischemic stroke.


Assuntos
Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Desacetilase 6 de Histona/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Animais , Isquemia Encefálica/patologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Citoplasma/metabolismo , Glicogênio Sintase Quinase 3 beta/biossíntese , Glicogênio Sintase Quinase 3 beta/genética , Desacetilase 6 de Histona/biossíntese , Desacetilase 6 de Histona/genética , Hipóxia Encefálica/tratamento farmacológico , Hipóxia Encefálica/patologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/psicologia , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/biossíntese , Acidente Vascular Cerebral/patologia
11.
J Pharmacol Exp Ther ; 369(3): 364-374, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30918068

RESUMO

Disease modification in Parkinson's disease (PD) is an unmet medical need. In the current study, we evaluated trehalose, a safe and well-tolerated disaccharide that has previously demonstrated efficacy in rodent models of neurodegenerative diseases, including PD. In a rat model of PD, based on delivery of adeno-associated virus serotype 1/2 containing the mutated human A53T α-synuclein gene (AAV1/2-hourA53T-aSyn) to the substantia nigra (SN), we showed that rats administered trehalose (2.67 g/kg per day, by mouth) for 6 weeks had less forelimb asymmetry (93% reduction) and higher striatal dopamine (54% increase) compared with rats receiving vehicle. In a pharmacokinetic study, we determined that efficacy was associated with plasma C max of 8900 ng/ml and area under the curve from time 0 to infinity (AUC0-inf) of 11,136 hour⋅ng/ml. We then showed, in macaques, that oral administration of trehalose (2.67 g/kg per day) produced plasma exposures of similar magnitude, with plasma C max of 10,918 ng/ml and AUC0-inf of 27,445 hour⋅ng/ml. In a macaque model of PD, also based on delivery of AAV1/2-hourA53T-aSyn to the SN, trehalose (2.67 g/kg per day, by mouth), administered for 142 days, produced higher striatal dopamine (by 39%) and dopamine transporter levels (by 50%), compared with macaques receiving vehicle. In neither model did trehalose treatment prevent loss of tyrosine hydroxylase (TH) positive (TH+ve) cells in the SN or alter α-synuclein levels in the striatum. These studies demonstrated that trehalose reduces striatal dopaminergic deficits in a rodent and macaque model of synucleinopathy in PD. Furthermore, we have determined the pharmacokinetic parameters associated with efficacy, and thus defined exposures to target in future clinical trials.


Assuntos
Dopamina/metabolismo , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Doença de Parkinson/tratamento farmacológico , Trealose/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Macaca fascicularis , Doença de Parkinson/sangue , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Ratos , Distribuição Tecidual , Trealose/sangue , Trealose/farmacocinética , Trealose/uso terapêutico
12.
Neuroscience ; 401: 106-116, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668973

RESUMO

The striatum of the basal ganglia is pivotal for voluntary movements and is implicated in debilitating movement disorders such as Parkinsonism and dystonia. Striatum projects to downstream nuclei through direct (dSPN) and indirect (iSPN) pathway projection neurons thought to exert opposite effects on movement. In rodent models of striatal function, unilateral dopamine deprivation induces ipsiversive rotational behavior. The dSPNs of the dorsal striatum are believed to engage distinct motor programs but underlying mechanisms remain unclear. Here, we show by employing chemogenetics [Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)] that unilateral inhibition of dorsomedial dSPNs is sufficient to selectively impair contraversive movement and elicit ipsiversive rotational behavior in mice. Adeno-associated virus (AAV) encoding Cre-dependent Gi-coupled DREADD was injected unilaterally into the dorsomedial striatum of Drd1-Cre mice, resulting in expression of the modified human M4 muscarinic receptor (hM4Di) in ∼20% of dorsostriatal dSPNs. Upon hM4Di activation, a striking positive linear correlation was found between turn ratio and viral expression, which corroborates a relationship between unilateral inhibition of dorsomedial dSPNs and rotational behavior. Bursts of ipsiversive rotations were interspersed with normal ambulation. However, partial unilateral inhibition of ∼20% of dorsostriatal dSPNs did not affect horizontal and vertical locomotion or forelimb use preference. Overall, our results substantiate a unique role of dSPNs in promoting response bias in rotational behavior and show this to be a highly sensitive measure of dSPN performance.


Assuntos
Drogas Desenhadas/farmacologia , Neostriado/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Animais , Gânglios da Base/metabolismo , Comportamento Animal , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Neostriado/citologia , Neostriado/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Transtornos Parkinsonianos/metabolismo , Receptor Muscarínico M4/metabolismo , Receptores de Dopamina D1/metabolismo , Rotação
13.
Toxicology ; 411: 110-121, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30391265

RESUMO

Exposure to herbicides can induce long-term chronic adverse effects such as respiratory diseases, malignancies and neurodegenerative diseases. Oxadiazon, a pre-emergence or early post-emergence herbicide, despite its low acute toxicity, may induce liver cancer and may exert adverse effects on reproductive and on endocrine functions. Unlike other herbicides, there are no indications on neurotoxicity associated with long-term exposure to oxadiazon. Therefore, we have analyzed in primary neuronal precursor cells isolated from human striatal primordium the effects of non-cytotoxic doses of oxadiazon on neuronal cell differentiation and migration, and on the expression and activity of the mitochondrial aldehyde dehydrogenase 2 (ALDH2) and of the acylphosphatase (ACYP). ALDH2 activity protects neurons against neurotoxicity induced by toxic aldehydes during oxidative stress and plays a role in neurodegenerative conditions such as Alzheimer's disease and Parkinson's disease. ACYP is involved in ion transport, cell differentiation, programmed cell death and cancer, and increased levels of ACYP have been revealed in fibroblasts from patients affected by Alzheimer's disease. In this study we demonstrated that non-cytotoxic doses of oxadiazon were able to inhibit neuronal striatal cell migration and FGF2- and BDNF-dependent differentiation towards neuronal phenotype, and to inhibit the expression and activity of ALDH2 and to increase the expression and activity of ACYP2. In addition, we have provided evidence that in human primary neuronal precursor striatal cells the inhibitory effects of oxadiazon on cell migration and differentiation towards neuronal phenotype were achieved through modulation of ACYP2. Taken together, our findings reveal for the first time that oxadiazon could exert neurotoxic effects by impairing differentiative capabilities of primary neuronal cells and indicate that ALDH2 and ACYP2 are relevant molecular targets for the neurotoxic effects of oxadiazon, suggesting a potential role of this herbicide in the onset of neurodegenerative diseases.


Assuntos
Hidrolases Anidrido Ácido/biossíntese , Aldeído-Desidrogenase Mitocondrial/biossíntese , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Herbicidas/toxicidade , Neostriado/enzimologia , Células-Tronco Neurais/enzimologia , Síndromes Neurotóxicas/enzimologia , Oxidiazóis/toxicidade , Hidrolases Anidrido Ácido/antagonistas & inibidores , Aldeído-Desidrogenase Mitocondrial/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Ensaio Cometa , Humanos , Neostriado/citologia , Neostriado/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Síndromes Neurotóxicas/patologia , Estresse Oxidativo/efeitos dos fármacos
14.
Neuropharmacology ; 146: 184-197, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30496754

RESUMO

Previous findings demonstrate a homeostatic role for oleoylethanolamide (OEA) signaling in the ethanol-related neuroinflammation and behavior. However, extensive research is still required in order to unveil the effects of OEA on a number of neurobiological functions such as adult neurogenesis, cell survival and resident neuroimmunity that become notably altered by alcohol. Daily consumption of ethanol (10%) for 2 weeks (6.3 ± 1.1 g/kg/day during last 5 days) caused hypolocomotor activity in rats. This effect appears to rely on central signaling mechanisms given that alcohol increased the OEA levels, the gene expression of OEA-synthesizing enzyme Nape-pld and the number of PPARα-immunoreactive neurons in the striatum. Ethanol-related neurobiological alterations such as a reduction in the number of microglial cells expressing iNOS (a cytokine-inducible immune defense) and in adult neural stem/progenitor cell (NSPC) proliferation (phospho-H3 and BrdU) and maturation (BrdU/ß3-tubulin), as well as an increase in damage cell activity (FosB) and apoptosis (cleaved caspase 3) were also observed in the rat striatum. Pharmacological administration of OEA (10 mg/kg) for 5 days during ethanol exposure exacerbated ethanol-induced hypolocomotion and cell apoptosis in the striatum. Interestingly, OEA abrogated the impaired effects of ethanol on PPARα-positive cell population and NSPC proliferation and maturation. OEA also decreased astrocyte-related vimentin immunoreactivity and increased microglial cell population (Iba-1, iNOS) in the striatum. These results suggest that OEA-PPARα signaling modulates glial activation, cell apoptosis and NSPC proliferation and maturation in response to striatal-specific neurobiological alterations induced by prolonged ethanol intake in rats.


Assuntos
Proliferação de Células/efeitos dos fármacos , Endocanabinoides/farmacologia , Etanol/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Ácidos Oleicos/farmacologia , Alanina Transaminase/sangue , Consumo de Bebidas Alcoólicas/tratamento farmacológico , Amidoidrolases/sangue , Animais , Apoptose/efeitos dos fármacos , Ácidos Araquidônicos/farmacologia , Aspartato Aminotransferases/sangue , Proteínas de Ligação ao Cálcio/metabolismo , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Etanolaminas/análise , Etanolaminas/sangue , Proteína Glial Fibrilar Ácida/metabolismo , Eliminação Hepatobiliar , Locomoção/efeitos dos fármacos , Masculino , Proteínas dos Microfilamentos/metabolismo , Neurônios/efeitos dos fármacos , PPAR alfa/metabolismo , Fosfolipase D/sangue , Alcamidas Poli-Insaturadas/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , gama-Glutamiltransferase/sangue
15.
Addict Biol ; 24(6): 1227-1234, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30536923

RESUMO

Fyn is a member of the Src family of protein tyrosine kinases (PTKs) that plays an important role not only in normal synaptic functions but also in brain pathologies including alcohol use disorder. We previously reported that repeated cycles of binge drinking and withdrawal activate Fyn in the dorsomedial striatum (DMS) of rodents, and that Fyn signaling in the DMS contributes to rat alcohol intake and relapse. Here, we used AZD0530, a CNS penetrable inhibitor of Src PTKs developed for the treatment of Alzheimer disease and cancer and tested its efficacy to suppress alcohol-dependent molecular and behavioral effects. We show that systemic administration of AZD0530 prevents alcohol-induced Fyn activation and GluN2B phosphorylation in the DMS of mice. We further report that a single dose of AZD0530 reduces alcohol operant self-administration and promotes extinction of alcohol self-administration without altering basal and dopamine D1 receptor-dependent locomotion. Together, our findings suggest that AZD0530, through its inhibitory actions on Fyn kinase, dampens alcohol seeking and drinking.


Assuntos
Comportamento Animal/efeitos dos fármacos , Benzodioxóis/farmacologia , Depressores do Sistema Nervoso Central/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Etanol/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fyn/antagonistas & inibidores , Quinazolinas/farmacologia , Animais , Condicionamento Operante/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Camundongos , Neostriado/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Autoadministração
16.
Oxid Med Cell Longev ; 2018: 4235695, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30228854

RESUMO

Excessive manganese (Mn) can accumulate in the striatum of the brain following overexposure. Oxidative stress is a well-recognized mechanism in Mn-induced neurotoxicity. It has been proven that glutathione (GSH) depletion is a key factor in oxidative damage during Mn exposure. However, no study has focused on the dysfunction of GSH synthesis-induced oxidative stress in the brain during Mn exposure. The objective of the present study was to explore the mechanism of Mn disruption of GSH synthesis via EAAC1 and xCT in vitro and in vivo. Primary neurons and astrocytes were cultured and treated with different doses of Mn to observe the state of cells and levels of GSH and reactive oxygen species (ROS) and measure mRNA and protein expression of EAAC1 and xCT. Mice were randomly divided into seven groups, which received saline, 12.5, 25, and 50 mg/kg MnCl2, 500 mg/kg AAH (EAAC1 inhibitor) + 50 mg/kg MnCl2, 75 mg/kg SSZ (xCT inhibitor) + 50 mg/kg MnCl2, and 100 mg/kg NAC (GSH rescuer) + 50 mg/kg MnCl2 once daily for two weeks. Then, levels of EAAC1, xCT, ROS, GSH, malondialdehyde (MDA), protein sulfhydryl, carbonyl, 8-hydroxy-2-deoxyguanosine (8-OHdG), and morphological and ultrastructural features in the striatum of mice were measured. Mn reduced protein levels, mRNA expression, and immunofluorescence intensity of EAAC1 and xCT. Mn also decreased the level of GSH, sulfhydryl, and increased ROS, MDA, 8-OHdG, and carbonyl in a dose-dependent manner. Injury-related pathological and ultrastructure changes in the striatum of mice were significantly present. In conclusion, excessive exposure to Mn disrupts GSH synthesis through inhibition of EAAC1 and xCT to trigger oxidative damage in the striatum.


Assuntos
Sistema y+ de Transporte de Aminoácidos/genética , Astrócitos/metabolismo , Cloretos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Transportador 3 de Aminoácido Excitatório/genética , Glutationa/biossíntese , Compostos de Manganês/farmacologia , Neostriado/patologia , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , 8-Hidroxi-2'-Desoxiguanosina , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Asparagina/análogos & derivados , Asparagina/farmacologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Transportador 3 de Aminoácido Excitatório/metabolismo , Feminino , Glutationa/metabolismo , Masculino , Camundongos , Modelos Biológicos , Neostriado/efeitos dos fármacos , Neostriado/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfassalazina/farmacologia
17.
Eur J Pharmacol ; 828: 31-41, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29577894

RESUMO

Huntington's disease (HD) is an inherited complex progressive neurodegenerative disorder with an established etiopathology linked to neuronal oxidative stress and corticostriatal excitotoxicity. Present study explores the effects of glucose-dependent insulinotropic polypeptide (GIP) receptor agonist on the neurobehavioral sequelae of quinolinic acid-induced phenotype of Huntington's disease in rats. Bilateral administration of quinolinic acid (300 nmol/4 µl) to the rat striatum led to characteristic deficits in, locomotor activity, motor coordination, neuromuscular coordination and short-term episodic memory. Therapeutic treatment for 14 days with a stable and brain penetrating GIP receptor agonist, D-Ala2GIP (100 nmol/kg, i.p.), attenuated the neurobehavioral deficits due to quinolinic acid (QA) administration. Protective actions of D-Ala2GIP were sensitive to blockade with a GIP receptor antagonist, (Pro3)GIP (50 nmol/kg, i.p.), indicating specific involvement of GIP receptor signaling pathway. Stimulation of GIP receptor with D-Ala2GIP attenuated lipid peroxidation, evidenced by reduced levels of brain malondialdehyde (MDA), and restoration of reduced glutathione (GSH) levels in brain. Quinolinic acid administration led to significant loss of striatal monoamines, e.g., norepinephrine, epinephrine, serotonin, dopamine, and metabolites, 3,4-Dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA) and 5-Hydroxyindoleacetic acid (5-HIAA). D-Ala2GIP attenuated the QA-induced depletion of striatal monoamines, without affecting the monoamine degradation pathways. Thus, observed effects with D-Ala2GIP in the QA-induced Huntington's disease model could be attributable to reduction in lipid peroxidation, restoration of endogenous antioxidants and decreased striatal monoamine levels. These findings together suggest that stimulation of GIP receptor signaling pathway in brain could be a potential therapeutic strategy in the symptomatic management of Huntington's disease.


Assuntos
Comportamento Animal/efeitos dos fármacos , Monoaminas Biogênicas/metabolismo , Polipeptídeo Inibidor Gástrico/farmacologia , Doença de Huntington/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Neostriado/metabolismo , Receptores dos Hormônios Gastrointestinais/agonistas , Animais , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Interações Medicamentosas , Marcha/efeitos dos fármacos , Polipeptídeo Inibidor Gástrico/química , Glutationa/metabolismo , Força da Mão , Doença de Huntington/induzido quimicamente , Doença de Huntington/fisiopatologia , Locomoção/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Memória Episódica , Neostriado/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ácido Quinolínico/efeitos adversos , Ratos , Ratos Wistar
18.
Mol Neurobiol ; 55(9): 7579-7587, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29429051

RESUMO

Nuclear receptor related 1 (Nurr1) orphan receptor has emerged as a promising contender in ameliorating Parkinson's disease; thus, finding a suitable activator of Nurr1 receptor is an attracting target for treating PD. Cilostazol, a phosphodiesterase-3 inhibitor, recently showed a favorable neuroprotective activity in multiple devastating central disorders, yet the possible antiparkinsonian activity of the drug has not been fully elucidated. Thus, the aim of this study is to explore the neuroprotective effect of cilostazol in rotenone-induced PD model in rats. Cilostazol successfully upregulated Nurr1 expression in PD rats, which resulted in successful preservation of the dopaminergic neuron functionality and integrity as verified by the marked improvement of motor performance in rotarod and open field tests, as well as the increased striatal tyrosine hydroxylase content. Moreover, cilostazol revealed an anti-inflammatory activity as manifested by hampering the global controller of inflammatory signaling pathway, nuclear factor-kappa B, together with its downstream pro-inflammatory cytokines, namely tumor necrosis factor-alpha and interleukin-1 beta, via Nurr-1 upregulation and glycogen synthase kinase 3 beta GSK-3ß inhibition. In turn, the increase in GSK-3ß inhibited form suppressed the measured downstream apoptotic biomarkers, viz. cytochrome C and caspase-3. Remarkably, cilostazol enhanced autophagy as depicted by hampering both LC3-II and P62 levels possibly through the prominent rise in sirtuin 1 level. In conclusion, cilostazol could be a promising candidate for PD treatment through modulating Nurr1 expression, as well as SIRT-1/autophagy, and GSK-3ß/apoptosis cross-regulation. Graphical Abstract In the rat rotenone model of Parkinson's disease (PD), Nurr1 expression was downregulated, GSK-3ß was activated, and autophagic flux was inhibited. Those deleterious effects were associated with deteriorated motor functions, striatal TH content, enhanced inflammatory state, and apoptotic cascade. Cilostazol, a phosphodiesterase-3 inhibitor, exerted a potential protective effect against PD through Nurr1 enhancement, GSK-3ß/apoptosis modulation, and SIRT-1/autophagy enhancement. Nurr1 nuclear receptor related 1, TH tyrosine hydroxylase, NF-κB nuclear factor κB, TNFα tumor necrosis factor alpha, ILs interleukins, GSK-3ß glycogen synthase kinase 3 beta, SIRT-1 sirtuin 1.


Assuntos
Autofagia/efeitos dos fármacos , Cilostazol/farmacologia , Fármacos Neuroprotetores/farmacologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Doença de Parkinson/patologia , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Biomarcadores/metabolismo , Modelos Animais de Doenças , Glicogênio Sintase Quinase 3 beta/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Atividade Motora/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Neostriado/patologia , Doença de Parkinson/enzimologia , Doença de Parkinson/fisiopatologia , Ratos Wistar , Teste de Desempenho do Rota-Rod , Rotenona , Sirtuína 1/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
19.
Mol Neurobiol ; 55(8): 6337-6346, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29294248

RESUMO

Huntington's disease (HD) is an autosomal dominantly inherited neurodegenerative disorder caused by expansion of CAG repeats in the coding area of huntingtin gene. In the HD brain, mutant huntingtin protein goes through proteolysis, and its amino-terminal portion consisting of polyglutamine repeats accumulate as inclusions that result in progressive impairment of cellular protein quality control system. Here, we demonstrate that partial rescue of the defective protein quality control in HD model mouse by azadiradione (a bioactive limonoids found in the seed of Azadirachta indica) could potentially improve the disease pathology. Prolonged treatment of azadiradione to HD mice significantly improved the progressive deterioration in body weight, motor functioning along with extension of lifespan. Azadiradione-treated HD mice brain also exhibited considerable decrease in mutant huntingtin aggregates load and improvement of striatal pathology in comparison with age-matched saline-treated HD controls. Biochemical analysis further revealed upregulation and activation of not only HSF1 (master regulator of protein folding) but also Ube3a (an ubiquitin ligase involved in the clearance of mutant huntingtin) in azadiradione-treated mice. Our results indicate that azadiradione-mediated enhanced folding and clearance of mutant huntingtin might underlie improved disease pathology in HD mice and suggests that it could be a potential therapeutic molecule to delay the progression of HD.


Assuntos
Progressão da Doença , Proteína Huntingtina/metabolismo , Doença de Huntington/tratamento farmacológico , Doença de Huntington/patologia , Limoninas/uso terapêutico , Animais , Atrofia , Modelos Animais de Doenças , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Doença de Huntington/fisiopatologia , Limoninas/administração & dosagem , Limoninas/farmacologia , Longevidade , Camundongos Transgênicos , Modelos Biológicos , Atividade Motora/efeitos dos fármacos , Proteínas Mutantes/metabolismo , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Neostriado/patologia , Neostriado/fisiopatologia , Agregados Proteicos/efeitos dos fármacos , Controle de Qualidade , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima
20.
Exp Neurol ; 299(Pt A): 137-147, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29056363

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by abnormal expansion of the polyglutamine tract in the huntingtin protein (HTT). The toxicity of mutant HTT (mHTT) is associated with intermediate mHTT soluble oligomers that subsequently form intranuclear inclusions. Thus, interventions promoting the clearance of soluble mHTT are regarded as neuroprotective. Striatal neurons are particularly vulnerable in HD. Their degeneration underlies motor symptoms and striatal atrophy, the anatomical hallmark of HD. Recent studies indicate that autophagy may be activated by dopamine D2 and D3 receptor (D2R/D3R) agonists. Since autophagy plays a central role in the degradation of misfolded proteins, and striatal neurons express D2R and D3R, D2R/D3R agonists may promote the clearance of mHTT in striatal neurons. Here, this hypothesis was tested by treating 8-week old R6/1 mice with the D2R/D3R agonist pramipexole for 4weeks. Pramipexole reduced striatal levels of soluble mHTT and increased the size of intranuclear inclusions in R6/1 mice. Furthermore, striatal DARPP-32 levels and motor functions were recovered. These effects were accompanied by an increase in LC3-II and a decrease in p62 in the striatum. Tollip, a selective adaptor of ubiquitinated polyQ proteins to LC3, was also reduced in the striata of R6/1mice but not in their wild-type littermates. No changes were detected in the cerebral cortex where D3R expression is very low, and behavioral and biochemical effects in the striatum were prevented by a D3R antagonist. The findings indicate that PPX protects striatal neurons by promoting the clearance of soluble mHTT through a D3R-mediated mechanism. The evidence of autophagy markers suggests that autophagy is activated, although it is not efficient at removing all mHTT recruited by the autophagic machinery as indicated by the increase in the size of intranuclear inclusions.


Assuntos
Benzotiazóis/uso terapêutico , Agonistas de Dopamina/uso terapêutico , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Neostriado/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Receptores de Dopamina D3/efeitos dos fármacos , Animais , Autofagia , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Movimento , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Pramipexol , Complexo de Endopeptidases do Proteassoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA