Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 101: 194-210, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35032575

RESUMO

BACKGROUND: Antigen-specific neuroinflammation and neurodegeneration are characteristic for neuroimmunological diseases. In Parkinson's disease (PD) pathogenesis, α-synuclein is a known culprit. Evidence for α-synuclein-specific T cell responses was recently obtained in PD. Still, a causative link between these α-synuclein responses and dopaminergic neurodegeneration had been lacking. We thus addressed the functional relevance of α-synuclein-specific immune responses in PD in a mouse model. METHODS: We utilized a mouse model of PD in which an Adeno-associated Vector 1/2 serotype (AAV1/2) expressing human mutated A53T-α-Synuclein was stereotactically injected into the substantia nigra (SN) of either wildtype C57BL/6 or Recombination-activating gene 1 (RAG1)-/- mice. Brain, spleen, and lymph node tissues from different time points following injection were then analyzed via FACS, cytokine bead assay, immunohistochemistry and RNA-sequencing to determine the role of T cells and inflammation in this model. Bone marrow transfer from either CD4+/CD8-, CD4-/CD8+, or CD4+/CD8+ (JHD-/-) mice into the RAG-1-/- mice was also employed. In addition to the in vivo studies, a newly developed A53T-α-synuclein-expressing neuronal cell culture/immune cell assay was utilized. RESULTS: AAV-based overexpression of pathogenic human A53T-α-synuclein in dopaminergic neurons of the SN stimulated T cell infiltration. RNA-sequencing of immune cells from PD mouse brains confirmed a pro-inflammatory gene profile. T cell responses were directed against A53T-α-synuclein-peptides in the vicinity of position 53 (68-78) and surrounding the pathogenically relevant S129 (120-134). T cells were required for α-synuclein-induced neurodegeneration in vivo and in vitro, while B cell deficiency did not protect from dopaminergic neurodegeneration. CONCLUSIONS: Using T cell and/or B cell deficient mice and a newly developed A53T-α-synuclein-expressing neuronal cell culture/immune cell assay, we confirmed in vivo and in vitro that pathogenic α-synuclein peptide-specific T cell responses can cause dopaminergic neurodegeneration and thereby contribute to PD-like pathology.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Modelos Animais de Doenças , Dopamina , Neurônios Dopaminérgicos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doença de Parkinson/patologia , RNA , Substância Negra/metabolismo , Linfócitos T/metabolismo , alfa-Sinucleína/metabolismo
2.
Neurotherapeutics ; 18(2): 962-978, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33723752

RESUMO

Glucagon-like peptide-1 (GLP-1) receptor stimulation ameliorates parkinsonian motor and non-motor deficits in both experimental animals and patients; however, the disease-modifying mechanisms of GLP-1 receptor activation have remained unknown. The present study investigated whether exendin-4 (a GLP-1 analogue) can rescue motor deficits and exert disease-modifying effects in a parkinsonian rat model of α-synucleinopathy. This model was established by unilaterally injecting AAV-9-A53T-α-synuclein into the right substantia nigra pars compacta, followed by 4 or 8 weeks of twice-daily intraperitoneal injections of exendin-4 (5 µg/kg/day) starting at 2 weeks after AAV-9-A53T-α-synuclein injections. Positron emission tomography/computed tomography (PET/CT) scanning and immunostaining established that treatment with exendin-4 attenuated tyrosine-hydroxylase-positive neuronal loss and terminal denervation and mitigated the decrease in expression of vesicular monoamine transporter 2 within the nigrostriatal dopaminergic systems of rats injected with AAV-9-A53T-α-synuclein. It also mitigated the parkinsonian motor deficits assessed in behavioral tests. Furthermore, through both in vivo and in vitro models of Parkinson's disease, we showed that exendin-4 promoted autophagy and mediated degradation of pathological α-synuclein, the effects of which were counteracted by 3-methyladenine or chloroquine, the autophagic inhibitors. Additionally, exendin-4 attenuated dysregulation of the PI3K/Akt/mTOR pathway in rats injected with AAV-9-A53T-α-synuclein. Taken together, our results demonstrate that exendin-4 treatment relieved behavioral deficits, dopaminergic degeneration, and pathological α-synuclein aggregation in a parkinsonian rat model of α-synucleinopathy and that these effects were mediated by enhanced autophagy via inhibiting the PI3K/Akt/mTOR pathway. In light of the safety and tolerance of exendin-4 administration, our results suggest that exendin-4 may represent a promising disease-modifying treatment for Parkinson's disease.


Assuntos
Autofagia/efeitos dos fármacos , Exenatida/uso terapêutico , Neuroproteção/efeitos dos fármacos , Transtornos Parkinsonianos/prevenção & controle , Sinucleinopatias/prevenção & controle , alfa-Sinucleína/toxicidade , Animais , Autofagia/fisiologia , Linhagem Celular Tumoral , Exenatida/farmacologia , Feminino , Humanos , Neuroproteção/fisiologia , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/patologia , Ratos , Ratos Sprague-Dawley , Sinucleinopatias/induzido quimicamente , Sinucleinopatias/patologia
3.
Acta Neuropathol Commun ; 8(1): 17, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-32059750

RESUMO

Parkinson's disease is a progressive neurodegenerative disorder characterised by the accumulation of misfolded α-synuclein in selected brain regions, including the substantia nigra pars compacta (SNpc), where marked loss of dopaminergic neurons is also observed. Yet, the relationship between misfolded α-synuclein and neurotoxicity currently remains unclear. As the principal route for degradation of misfolded proteins in mammalian cells, the ubiquitin-proteasome system (UPS) is critical for maintenance of cellular proteostasis. Misfolded α-synuclein impairs UPS function and contributes to neuronal death in vitro. Here, we examine its effects in vivo using adeno-associated viruses to co-express A53T α-synuclein and the ubiquitinated reporter protein UbG76V-GFP in rat SNpc. We found that α-synuclein over-expression leads to early-onset catalytic impairment of the 26S proteasome with associated UPS dysfunction, preceding the onset of behavioural deficits and dopaminergic neurodegeneration. UPS failure in dopaminergic neurons was also associated with selective accumulation of α-synuclein phosphorylated at the serine 129 residue, which has previously been linked to increased neurotoxicity. Our study highlights a role for α-synuclein in disturbing proteostasis which may contribute to neurodegeneration in vivo.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Parte Compacta da Substância Negra/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , alfa-Sinucleína/metabolismo , Animais , Feminino , Células HEK293 , Humanos , Ratos Sprague-Dawley
4.
Sci Rep ; 9(1): 8362, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31182727

RESUMO

While GM1 may interact with α-synuclein in vitro to inhibit aggregation, the ability of GM1 to protect against α-synuclein toxicity in vivo has not been investigated. We used targeted adeno-associated viral vector (AAV) overexpression of human mutant α-synuclein (A53T) in the rat substantia nigra (SN) to produce degeneration of SN dopamine neurons, loss of striatal dopamine levels, and behavioral impairment. Some animals received daily GM1 ganglioside administration for 6 weeks, beginning 24 hours after AAV-A53T administration or delayed start GM1 administration for 5 weeks beginning 3 weeks after AAV-A53T administration. Both types of GM1 administration protected against loss of SN dopamine neurons and striatal dopamine levels, reduced α-synuclein aggregation, and delayed start administration of GM1 reversed early appearing behavioral deficits. These results extend prior positive results in MPTP models, are consistent with the results of a small clinical study of GM1 in PD patients that showed slowing of symptom progression with chronic use, and argue for the continued refinement and development of GM1 as a potential disease modifying therapy for PD.


Assuntos
Gangliosídeo G(M1)/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/genética , Animais , Comportamento Animal/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dependovirus/efeitos dos fármacos , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Humanos , Doença de Parkinson/genética , Doença de Parkinson/patologia , Ratos , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
5.
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
6.
Neurobiol Dis ; 106: 133-146, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28673739

RESUMO

Parkinson's disease (PD) is a neurodegenerative movement disorder, which affects approximately 1-2% of the population over 60years of age. Current treatments for PD are symptomatic, and the pathology of the disease continues to progresses over time until palliative care is required. Mitochondria are key players in the pathology of PD. Genetic and post mortem studies have shown a large number of mitochondrial abnormalities in the substantia nigra pars compacta (SNc) of the parkinsonian brain. Furthermore, physiologically, mitochondria of nigral neurons are constantly under unusually high levels of metabolic stress because of the excitatory properties and architecture of these neurons. The protein deacetylase, Sirtuin 3 (SIRT3) reduces the impact subcellular stresses on mitochondria, by stabilising the electron transport chain (ETC), and reducing oxidative stress. We hypothesised that viral overexpression of myc-tagged SIRT3 (SIRT3-myc) would slow the progression of PD pathology, by enhancing the functional capacity of mitochondria. For this study, SIRT3-myc was administered both before and after viral induction of parkinsonism with the AAV-expressing mutant (A53T) α-synuclein. SIRT3-myc corrected behavioural abnormalities, as well as changes in striatal dopamine turnover. SIRT3-myc also prevented degeneration of dopaminergic neurons in the SNc. These effects were apparent, even when SIRT3-myc was transduced after the induction of parkinsonism, at a time point when cell stress and behavioural abnormalities are already observed. Furthermore, in an isolated mitochondria nigral homogenate prepared from parkinsonian SIRT3-myc infected animals, SIRT3 targeted the mitochondria, to reduce protein acetylation levels. Our results demonstrate that transduction of SIRT3 has the potential to be an effective disease-modifying strategy for patients with PD. This study also provides potential mechanisms for the protective effects of SIRT3-myc.


Assuntos
Mitocôndrias/metabolismo , Neurônios/metabolismo , Neuroproteção/fisiologia , Transtornos Parkinsonianos/metabolismo , Sirtuína 3/metabolismo , alfa-Sinucleína/metabolismo , Acetilação , Animais , Linhagem Celular Tumoral , Dependovirus/genética , Feminino , Vetores Genéticos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/patologia , Mutação , Neurônios/patologia , Biogênese de Organelas , Transtornos Parkinsonianos/patologia , Ratos Sprague-Dawley , Sirtuína 3/genética , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/genética
7.
Ann Neurol ; 81(6): 825-836, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28470693

RESUMO

OBJECTIVE: Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is a highly effective symptomatic therapy for motor deficits in Parkinson's disease (PD). An additional, disease-modifying effect has been suspected from studies in toxin-based PD animal models, but these models do not reflect the molecular pathology and progressive nature of PD that would be required to evaluate a disease-modifying action. Defining a disease-modifying effect could radically change the way in which DBS is used in PD. METHODS: We applied STN-DBS in an adeno-associated virus (AAV) 1/2-driven human mutated A53T α-synuclein (aSyn)-overexpressing PD rat model (AAV1/2-A53T-aSyn). Rats were injected unilaterally, in the substantia nigra (SN), with AAV1/2-A53T-aSyn or control vector. Three weeks later, after behavioral and nigrostriatal dopaminergic deficits had developed, rats underwent STN-DBS electrode implantation ipsilateral to the vector-injected SN. Stimulation lasted for 3 weeks. Control groups remained OFF stimulation. Animals were sacrificed at 6 weeks. RESULTS: Motor performance in the single pellet reaching task was impaired in the AAV1/2-A53T-aSyn-injected stim-OFF group, 6 weeks after AAV1/2-A53T-aSyn injection, compared to preoperative levels (-82%; p < 0.01). Deficits were reversed in AAV1/2-A53T-aSyn, stim-ON rats after 3 weeks of active stimulation, compared to the AAV1/2-A53T-aSyn stim-OFF rats (an increase of ∼400%; p < 0.05), demonstrating a beneficial effect of DBS. This motor improvement was maintained when the stimulation was turned off and was accompanied by a higher number of tyrosine hydroxylase+ SN neurons (increase of ∼29%), compared to AAV1/2-A53T-aSyn stim-OFF rats (p < 0.05). INTERPRETATION: Our data support the putative neuroprotective and disease-modifying effect of STN-DBS in a mechanistically relevant model of PD. Ann Neurol 2017;81:825-836.


Assuntos
Estimulação Encefálica Profunda/métodos , Doença de Parkinson/terapia , Núcleo Subtalâmico , alfa-Sinucleína/administração & dosagem , Animais , Comportamento Animal , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Masculino , Mutação , Ratos , Ratos Sprague-Dawley , Núcleo Subtalâmico/citologia , Núcleo Subtalâmico/metabolismo , Núcleo Subtalâmico/fisiopatologia
8.
Acta Neuropathol Commun ; 5(1): 11, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28143577

RESUMO

α-Synuclein is a protein implicated in the etiopathogenesis of Parkinson's disease (PD). AAV1/2-driven overexpression of human mutated A53T-α-synuclein in rat and monkey substantia nigra (SN) induces degeneration of nigral dopaminergic neurons and decreases striatal dopamine and tyrosine hydroxylase (TH). Given certain advantages of the mouse, especially it being amendable to genetic manipulation, translating the AAV1/2-A53T α-synuclein model to mice would be of significant value. AAV1/2-A53T α-synuclein or AAV1/2 empty vector (EV) at a concentration of 5.16 x 1012 gp/ml were unilaterally injected into the right SN of male adult C57BL/6 mice. Post-mortem examinations included immunohistochemistry to analyze nigral α-synuclein, Ser129 phosphorylated α-synuclein and TH expression, striatal dopamine transporter (DAT) levels by autoradiography and dopamine levels by high performance liquid chromatography. At 10 weeks, in AAV1/2-A53T α-synuclein mice there was a 33% reduction in TH+ dopaminergic nigral neurons (P < 0.001), 29% deficit in striatal DAT binding (P < 0.05), 38% and 33% reductions in dopamine (P < 0.001) and DOPAC (P < 0.01) levels and a 60% increase in dopamine turnover (homovanilic acid/dopamine ratio; P < 0.001). Immunofluorescence showed that the AAV1/2-A53T α-synuclein injected mice had widespread nigral and striatal expression of vector-delivered A53T-α-synuclein. Concurrent staining with human PD SN samples using gold standard histological methodology for Lewy pathology detection by proteinase K digestion and application of specific antibody raised against human Lewy body α-synuclein (LB509) and Ser129 phosphorylated α-synuclein (81A) revealed insoluble α-synuclein aggregates in AAV1/2-A53T α-synuclein mice resembling Lewy-like neurites and bodies. In the cylinder test, we observed significant paw use asymmetry in the AAV1/2-A53T α-synuclein group when compared to EV controls at 5 and 9 weeks post injection (P < 0.001; P < 0.05). These data show that unilateral injection of AAV1/2-A53T α-synuclein into the mouse SN leads to persistent motor deficits, neurodegeneration of the nigrostriatal dopaminergic system and development of Lewy-like pathology, thereby reflecting clinical and pathological hallmarks of human PD.


Assuntos
Atividade Motora/fisiologia , Mutação , Transtornos Parkinsonianos/metabolismo , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Dependovirus/genética , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Vetores Genéticos , Ácido Homovanílico/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Transtornos Parkinsonianos/patologia , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/genética
9.
PLoS One ; 11(11): e0167235, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27902767

RESUMO

Recent failures in clinical trials for disease modification in Parkinson's disease have highlighted the need for a non-human primate model of the synucleinopathy underpinning dopaminergic neuron degeneration. The present study was defined to begin the development of such a model in cynomolgus macaque. We have validated surgical and vector parameters to define a means to provide a robust over-expression of alpha-synuclein which is associated with Lewy-like pathology and robust degeneration of the nigrostriatal pathway. Thus, an AAV1/2 vector incorporating strong transcription and transduction regulatory elements was used to deliver the gene for the human A53T mutation of alpha-synuclein. When injected into 4 sites within each substantia nigra (7 µl per site, 1.7 x 1012 gp/ml), this vector provided expression lasting at least 4 months, and a 50% loss of nigral dopaminergic neurons and a 60% reduction in striatal dopamine. Further studies will be required to develop this methodology into a validated model of value as a drug development platform.


Assuntos
Dependovirus/genética , Neurônios Dopaminérgicos/patologia , Terapia Genética/métodos , Doença de Parkinson/terapia , alfa-Sinucleína/genética , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Vetores Genéticos/genética , Humanos , Macaca , Neostriado/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
10.
Mol Neurobiol ; 53(4): 2258-68, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-25972237

RESUMO

The accumulation of misfolded α-synuclein in dopamine (DA) neurons is believed to be of major importance in the pathogenesis of Parkinson's disease (PD). Animal models of PD, based on viral-vector-mediated over-expression of α-synuclein, have been developed and show evidence of dopaminergic toxicity, providing us a good tool to investigate potential therapies to interfere with α-synuclein-mediated pathology. An efficient disease-modifying therapeutic molecule should be able to interfere with the neurotoxicity of α-synuclein aggregation. Our study highlighted the ability of an autophagy enhancer, trehalose (at concentrations of 5 and 2% in drinking water), to protect against A53T α-synuclein-mediated DA degeneration in an adeno-associated virus serotype 1/2 (AAV1/2)-based rat model of PD. Behavioral tests and neurochemical analysis demonstrated a significant attenuation in α-synuclein-mediated deficits in motor asymmetry and DA neurodegeneration including impaired DA neuronal survival and DA turnover, as well as α-synuclein accumulation and aggregation in the nigrostriatal system by commencing 5 and 2% trehalose at the same time as delivery of AAV. Trehalose (0.5%) was ineffective on the above behavioral and neurochemical deficits. Further investigation showed that trehalose enhanced autophagy in the striatum by increasing formation of LC3-II. This study supports the concept of using trehalose as a novel therapeutic strategy that might prevent/reverse α-synuclein aggregation for the treatment of PD.


Assuntos
Comportamento Animal/efeitos dos fármacos , Dependovirus/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Trealose/uso terapêutico , alfa-Sinucleína/metabolismo , Animais , Autofagia/efeitos dos fármacos , Glicemia/metabolismo , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Extremidades/patologia , Feminino , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Doença de Parkinson/sangue , Ratos Sprague-Dawley , Solubilidade , Substância Negra/efeitos dos fármacos , Substância Negra/patologia , Trealose/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Pharmacol Rev ; 65(1): 171-222, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23319549

RESUMO

L-3,4-Dihydroxyphenylalanine (L-DOPA) remains the most effective symptomatic treatment of Parkinson's disease (PD). However, long-term administration of L-DOPA is marred by the emergence of abnormal involuntary movements, i.e., L-DOPA-induced dyskinesia (LID). Years of intensive research have yielded significant progress in the quest to elucidate the mechanisms leading to the development and expression of dyskinesia and maintenance of the dyskinetic state, but the search for a complete understanding is still ongoing. Herein, we summarize the current knowledge of the pharmacology of LID in PD. Specifically, we review evidence gathered from postmortem and pharmacological studies, both preclinical and clinical, and discuss the involvement of dopaminergic and nondopaminergic systems, including glutamatergic, opioid, serotonergic, γ-aminobutyric acid (GABA)-ergic, adenosine, cannabinoid, adrenergic, histaminergic, and cholinergic systems. Moreover, we discuss changes occurring in transcription factors, intracellular signaling, and gene expression in the dyskinetic phenotype. Inasmuch as a multitude of neurotransmitters and receptors play a role in the etiology of dyskinesia, we propose that to optimally alleviate this motor complication, it may be necessary to develop combined treatment approaches that will target simultaneously more than one neurotransmitter system. This could be achieved via three ways as follows: 1) by developing compounds that will interact simultaneously to a multitude of receptors with the required agonist/antagonist effect at each target, 2) by targeting intracellular signaling cascades where the signals mediated by multiple receptors converge, and/or 3) to regulate gene expression in a manner that has effects on signaling by multiple pathways.


Assuntos
Antiparkinsonianos/efeitos adversos , Dopaminérgicos/efeitos adversos , Discinesia Induzida por Medicamentos/fisiopatologia , Levodopa/efeitos adversos , Doença de Parkinson/fisiopatologia , Animais , Gânglios da Base/fisiologia , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/metabolismo , Humanos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Transmissão Sináptica/fisiologia
12.
PLoS One ; 6(3): e17698, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21408191

RESUMO

The pathological hallmarks of Parkinson's disease (PD) are degeneration of dopamine (DA) neurons of the substantia nigra (SN) and the presence of alpha-synuclein (α-syn)-rich Lewy bodies in DA cells that remain. To model these aspects of the disease, we previously showed that high titer (5.1×10exp12 gp/ml) AAV1/2 driven expression of A53T α-syn in the SN of rats caused nigrostriatal pathology including a loss of DA neurons, but also with toxicity in the GFP control group. In the current study, we evaluate the effects of two lower titers by dilution of the vector (1∶3 [1.7×10exp12] and 1∶10 [5.1×10exp11]) to define a concentration that produced pathology specific for α-syn. In GFP and empty vector groups there were no behavioural or post-mortem changes at 3 or 6 weeks post-administration at either vector dose. Dilution of the AAV1/2 A53T α-syn (1:3) produced significant paw use asymmetry, reductions in striatal tyrosine hydroxylase (TH), and increases in DA turnover at 3 weeks in the absence of overt pathology. By 6 weeks greater evidence of pathology was observed and included, reductions in SN DA neurons, striatal DA, TH and DA-transporter, along with a sustained behavioural deficit. In contrast, the 1:10 AAV1/2 A53T α-syn treated animals showed normalization between 3 and 6 weeks in paw use asymmetry, reductions in striatal TH, and increased DA turnover. Progression of dopaminergic deficits using the 1:3 titer of AAV1/2 A53Tα-syn provides a platform for evaluating treatments directed at preventing and/or reversing synucleinopathy. Use of the 1:10 titer of AAV1/2 A53T α-syn provides an opportunity to study mechanisms of endogenous compensation. Furthermore, these data highlight the need to characterize the titer of vector being utilized, when using AAV to express pathogenic proteins and model disease process, to avoid producing non-specific effects.


Assuntos
Degeneração Neural/patologia , Doença de Parkinson/terapia , alfa-Sinucleína/genética , alfa-Sinucleína/uso terapêutico , Animais , Axônios/patologia , Contagem de Células , Dependovirus/genética , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Membro Anterior , Terapia Genética , Vetores Genéticos , Neostriado/metabolismo , Neostriado/patologia , Doença de Parkinson/patologia , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Substância Negra/enzimologia , Substância Negra/patologia , Transgenes/genética , Tirosina 3-Mono-Oxigenase/metabolismo
13.
J Neurosci ; 29(11): 3365-73, 2009 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-19295143

RESUMO

Little is known about key pathological events preceding overt neuronal degeneration in Parkinson's disease (PD) and alpha-synucleinopathy. Recombinant adeno-associated virus 2-mediated delivery of mutant (A53T) human alpha-synuclein into the substantia nigra (SN) under a neuron-specific synapsin promoter resulted in protracted neurodegeneration with significant dopaminergic (DA) neuron loss by 17 weeks. As early as 4 weeks, there was an increase in a dopamine metabolite, DOPAC and histologically, DA axons in the striatum were dystrophic with degenerative bulbs. Before neuronal loss, significant changes were identified in levels of proteins relevant to synaptic transmission and axonal transport in the striatum and the SN. For example, striatal levels of rabphilin 3A and syntaxin were reduced. Levels of anterograde transport motor proteins (KIF1A, KIF1B, KIF2A, and KIF3A) were decreased in the striatum, whereas retrograde motor proteins (dynein, dynamitin, and dynactin1) were increased. In contrast to reduced levels in the striatum, KIF1A and KIF2A levels were elevated in the SN. There were dramatic changes in cytoskeletal protein levels, with actin levels increased and alpha-/gamma-tubulin levels reduced. In addition to these alterations, a neuroinflammatory response was observed at 8 weeks in the striatum, but not in the SN, demonstrated by increased levels of Iba-1, activated microglia and increased levels of proinflammatory cytokines, including IL-1beta, IFN-gamma and TNF-alpha. These results demonstrate that changes in proteins relevant to synaptic transmission and axonal transport coupled with neuroinflammation, precede alpha-synuclein-mediated neuronal death. These findings can provide ideas for antecedent biomarkers and presymptomatic interventions in PD.


Assuntos
Proteínas de Transporte/metabolismo , Dependovirus , Modelos Animais de Doenças , Dopamina/fisiologia , Doenças do Sistema Nervoso/metabolismo , Terminações Pré-Sinápticas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Transporte Axonal/genética , Proteínas de Transporte/genética , Morte Celular/genética , Dependovirus/genética , Dopamina/genética , Feminino , Vetores Genéticos/genética , Humanos , Inflamação/metabolismo , Inflamação/patologia , Inflamação/virologia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/virologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Terminações Pré-Sinápticas/patologia , Terminações Pré-Sinápticas/virologia , Ratos , Ratos Sprague-Dawley , Substância Negra/metabolismo , Substância Negra/patologia , Substância Negra/virologia , alfa-Sinucleína/genética
14.
J Neuroinflammation ; 5: 8, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18304357

RESUMO

BACKGROUND: The etiology of Parkinson's disease (PD) remains elusive despite identification of several genetic mutations. It is more likely that multiple factors converge to give rise to PD than any single cause. Here we report that inflammation can trigger degeneration of dopamine (DA) neurons in an animal model of Parkinson's disease. METHODS: We examined the effects of inflammation on the progressive 6-OHDA rat model of Parkinson's disease using immunohistochemistry, multiplex ELISA, and cell counting stereology. RESULTS: We show that a non-toxic dose of lipopolysaccharide (LPS) induced secretion of cytokines and predisposed DA neurons to be more vulnerable to a subsequent low dose of 6-hydroxydopamine. Alterations in cytokines, prominently an increase in interleukin-1beta (IL-1beta), were identified as being potential mediators of this effect that was associated with activation of microglia. Administration of an interleukin-1 receptor antagonist resulted in significant reductions in tumor necrosis factor-alpha and interferon-gamma and attenuated the augmented loss of DA neurons caused by the LPS-induced sensitization to dopaminergic degeneration. CONCLUSION: These data provide insight into the etiology of PD and support a role for inflammation as a risk factor for the development of neurodegenerative disease.


Assuntos
Dopamina/fisiologia , Interleucina-1beta/imunologia , Degeneração Neural/imunologia , Neurite (Inflamação)/imunologia , Transtornos Parkinsonianos/imunologia , Animais , Antirreumáticos/farmacologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Lipopolissacarídeos/farmacologia , Microglia/imunologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/epidemiologia , Neurite (Inflamação)/induzido quimicamente , Neurite (Inflamação)/epidemiologia , Neuroimunomodulação/imunologia , Oxidopamina , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/epidemiologia , Ratos , Ratos Sprague-Dawley , Fatores de Risco , Substância Negra/efeitos dos fármacos , Substância Negra/imunologia , Simpatolíticos
15.
Brain Res Dev Brain Res ; 147(1-2): 77-84, 2003 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-14741753

RESUMO

Recent findings suggest that prenatal cocaine exposure results in significant attenuation of uterine and placental blood flow. The extent of blood flow reduction to fetuses positively correlates with reductions in glial-derived neurotrophic factor (GDNF) and dopamine (DA). However, whether such changes in uterine blood flow are sufficient to induce oxidative stress have yet to be determined. In the following experiments, the impact of prenatal cocaine exposure on fetal brain levels of the endogenous antioxidant glutathione (GSH and its oxidized form GSSG) or the exogenous antioxidant alpha-tocopherol (alpha-T and its oxidized quinone form) was investigated. It was hypothesized that cocaine exposure would result in greater oxidation of both GSH and alpha-T. Results indicated that a single injection of cocaine to a drug-naive pregnant dam results in significant (-16.38%) reductions in the levels of GSH. GSSG can be either raised or reduced as a result of fetal uterine position: fetuses at the ovarian extremes show significant increases in GSSG in response to cocaine (+64.73%), whereas cervically situated fetuses show decreased GSSG (-47.91%). Additionally, cocaine significantly decreased the levels of alpha-T (-15.9%) and increased the levels of its oxidative product alpha-Tquinone (alpha-Tq, +34.05%). Levels of alpha-T were not affected by fetal uterine position. These data collectively suggest that cocaine exposure increases the utilization of both endogenous and exogenous anti-oxidants in the fetal rat brain. Along with previous studies, these data support the hypothesis that cocaine-induced vasoconstriction results in oxidative stress in the gestating fetus.


Assuntos
Química Encefálica/efeitos dos fármacos , Cocaína/toxicidade , Glutationa/metabolismo , Vitamina E/análogos & derivados , Vitamina E/metabolismo , Animais , Peso ao Nascer/efeitos dos fármacos , Dieta , Comportamento Alimentar/efeitos dos fármacos , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Masculino , Proteínas do Tecido Nervoso/biossíntese , Oxirredução , Gravidez , Ratos , Ratos Sprague-Dawley , Razão de Masculinidade , Útero/anatomia & histologia , Deficiência de Vitamina E/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA