Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 79, 2023 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-36945016

RESUMEN

Neuroinflammation has been suggested as a pathogenetic mechanism contributing to Parkinson's disease (PD). However, anti-inflammatory treatment strategies have not yet been established as a therapeutic option for PD patients. We have used a human α-synuclein mouse model of progressive PD to examine the anti-inflammatory and neuroprotective effects of inflammasome inhibition on dopaminergic (DA) neurons in the substantia nigra (SN). As the NLRP3 (NOD-, LRR- and pyrin domain-containing 3)-inflammasome is a core interface for both adaptive and innate inflammation and is also highly druggable, we investigated the implications of its inhibition. Repeat administration of MCC950, an inhibitor of NLRP3, in a PD model with ongoing pathology reduced CD4+ and CD8+ T cell infiltration into the SN. Furthermore, the anti-inflammasome treatment mitigated microglial activation and modified the aggregation of α-synuclein protein in DA neurons. MCC950-treated mice showed significantly less neurodegeneration of DA neurons and a reduction in PD-related motor behavior. In summary, early inflammasome inhibition can reduce neuroinflammation and prevent DA cell death in an α-synuclein mouse model for progressive PD.


Asunto(s)
Inflamasomas , Enfermedad de Parkinson , Humanos , Ratones , Animales , Inflamasomas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , alfa-Sinucleína/metabolismo , Neuronas Dopaminérgicas , Enfermedades Neuroinflamatorias , Microglía/metabolismo , Ratones Endogámicos NOD , Sulfonamidas/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
2.
J Neuroinflammation ; 19(1): 319, 2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36587195

RESUMEN

BACKGROUND: Regulatory CD4+CD25+FoxP3+ T cells (Treg) are a subgroup of T lymphocytes involved in maintaining immune balance. Disturbance of Treg number and impaired suppressive function of Treg correlate with Parkinson's disease severity. Superagonistic anti-CD28 monoclonal antibodies (CD28SA) activate Treg and cause their expansion to create an anti-inflammatory environment. METHODS: Using the AAV1/2-A53T-α-synuclein Parkinson's disease mouse model that overexpresses the pathogenic human A53T-α-synuclein (hαSyn) variant in dopaminergic neurons of the substantia nigra, we assessed the neuroprotective and disease-modifying efficacy of a single intraperitoneal dose of CD28SA given at an early disease stage. RESULTS: CD28SA led to Treg expansion 3 days after delivery in hαSyn Parkinson's disease mice. At this timepoint, an early pro-inflammation was observed in vehicle-treated hαSyn Parkinson's disease mice with elevated percentages of CD8+CD69+ T cells in brain and increased levels of interleukin-2 (IL-2) in the cervical lymph nodes and spleen. These immune responses were suppressed in CD28SA-treated hαSyn Parkinson's disease mice. Early treatment with CD28SA attenuated dopaminergic neurodegeneration in the SN of hαSyn Parkinson's disease mice accompanied with reduced brain numbers of activated CD4+, CD8+ T cells and CD11b+ microglia observed at the late disease-stage 10 weeks after AAV injection. In contrast, a later treatment 4 weeks after AAV delivery failed to reduce dopaminergic neurodegeneration. CONCLUSIONS: Our data indicate that immune modulation by Treg expansion at a timepoint of overt inflammation is effective for treatment of hαSyn Parkinson's disease mice and suggest that the concept of early immune therapy could pose a disease-modifying option for Parkinson's disease patients.


Asunto(s)
Enfermedad de Parkinson , Ratones , Humanos , Animales , Enfermedad de Parkinson/patología , Linfocitos T Reguladores , alfa-Sinucleína/metabolismo , Linfocitos T CD8-positivos/metabolismo , Antígenos CD28 , Anticuerpos/farmacología , Sustancia Negra/metabolismo , Neuronas Dopaminérgicas/metabolismo , Dopamina , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
3.
Nat Rev Neurosci ; 18(9): 515-529, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28747776

RESUMEN

A major challenge in Parkinson disease (PD) will be to turn an emerging and expanding pipeline of novel disease-modifying candidate compounds into therapeutics. Novel targets need in vivo validation, and candidate therapeutics require appropriate preclinical platforms on which to define potential efficacy and target engagement before advancement to clinical development. We propose that α-synuclein (α-syn)-based mammalian models will be crucial for this process. Here, we review α-syn transgenic mouse models, viral vector models of α-syn overexpression and models of 'prion-like' spread of α-syn, and describe how each of these model types may contribute to PD drug discovery. We conclude by presenting our opinion on how to use a combination of these models through the late-stage preclinical, proof-of-principle investigation of novel therapeutics.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Animales , Animales Modificados Genéticamente , Humanos
4.
Brain Behav Immun ; 101: 194-210, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35032575

RESUMEN

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.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Animales , Modelos Animales de Enfermedad , Dopamina , Neuronas Dopaminérgicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad de Parkinson/patología , ARN , Sustancia Negra/metabolismo , Linfocitos T/metabolismo , alfa-Sinucleína/metabolismo
5.
Eur J Neurosci ; 49(3): 399-409, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30269407

RESUMEN

The drive to develop drugs to treat PD starts and ends with the patient. Herein, we discuss how the experience with drug development for LID has led the field in translational studies in PD with advancing ground-breaking science via rigorous clinical trial design, to deliver clinical proof-of-concepts across multiple therapeutic targets. However, issues remain in advancing drugs efficacious preclinically to the clinic, and future studies need to learn from past successes and failures. Such lessons include implementing better early indicators of tolerability, for instance evaluating non-motor symptoms in preclinical models; improving patient-related outcome measures in clinical trials, as well as considering the unique nature of dyskinesia in an individual patient. The field of translational studies needs to become more patient focused to improve successful outcomes.


Asunto(s)
Desarrollo de Medicamentos/métodos , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Levodopa/efectos adversos , Animales , Discinesia Inducida por Medicamentos/complicaciones , Humanos , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/tratamiento farmacológico , Investigación Biomédica Traslacional/normas
6.
J Pharmacol Exp Ther ; 369(3): 364-374, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30918068

RESUMEN

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.


Asunto(s)
Dopamina/metabolismo , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Trehalosa/farmacología , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Macaca fascicularis , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas , Distribución Tisular , Trehalosa/sangre , Trehalosa/farmacocinética , Trehalosa/uso terapéutico
7.
Mov Disord ; 34(5): 708-716, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30575996

RESUMEN

BACKGROUND: Pridopidine, in development for Huntington's disease, may modulate aberrant l-dopa-induced effects including l-dopa-induced dyskinesia (LID). OBJECTIVE: This study investigated whether pridopidine could reduce LID in the MPTP macaque model of Parkinson's disease and characterized the observed behavioral effects in terms of receptor occupancy. METHODS: The pharmacokinetic profile and effects of pridopidine (15-30 mg/kg) on parkinsonism, dyskinesia, and quality of on-time, in combination with l-dopa, were assessed in MPTP macaques with LID. Pridopidine receptor occupancy was estimated using known in vitro binding affinities to σ1 and dopamine D2 receptors, in vivo PET imaging, and pharmacokinetic profiling across different species. RESULTS: Pridopidine produced a dose-dependent reduction in dyskinesia (up to 71%, 30 mg/kg) and decreased the duration of on-time with disabling dyskinesia evoked by l-dopa by 37% (20 mg/kg) and 60% (30 mg/kg). Pridopidine did not compromise the anti-parkinsonian benefit of l-dopa. Plasma exposures following the ineffective dose (15 mg/kg) were associated with full σ1 occupancy (>80%), suggesting that σ1 engagement alone is unlikely to account for the antidyskinetic benefits of pridopidine. Exposures following effective doses (20-30 mg/kg), while providing full σ1 occupancy, provide only modest dopamine D2 occupancy (<40%). However, effective pridopidine doses clearly engage a range of receptors (including adrenergic-α2C , dopamine-D3 , and serotoninergic-5-HT1A sites) to a higher degree than D2 and might contribute to the antidyskinetic actions. CONCLUSIONS: In MPTP macaques, pridopidine produced a significant decrease in LID without compromising the antiparkinsonian benefit of l-dopa. Although the actions of pridopidine were associated with full σ1 occupancy, effective exposures are more likely associated with occupancy of additional, non-sigma receptors. This complex pharmacology may underlie the effectiveness of pridopidine against LID. © 2018 International Parkinson and Movement Disorder Society.


Asunto(s)
Antiparkinsonianos/efectos adversos , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Levodopa/efectos adversos , Intoxicación por MPTP/tratamiento farmacológico , Movimiento/efectos de los fármacos , Trastornos Parkinsonianos/tratamiento farmacológico , Piperidinas/farmacología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Discinesia Inducida por Medicamentos/etiología , Macaca fascicularis , Trastornos Parkinsonianos/inducido químicamente , Tomografía de Emisión de Positrones , Receptor Muscarínico M2/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Receptores Histamínicos H3/metabolismo , Receptores sigma/metabolismo , Receptor Sigma-1
8.
J Pharmacol Exp Ther ; 367(2): 373-381, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30087157

RESUMEN

Dyskinesia is a common motor complication associated with the use of levodopa to treat Parkinson's disease. Numerous animal studies in mice, rats, and nonhuman primates have demonstrated that the N-methyl-d-aspartate antagonist, amantadine, dose dependently reduces levodopa-induced dyskinesia (LID). However, none of these studies characterized the amantadine plasma concentrations required for a therapeutic effect. This study evaluates the pharmacokinetic (PK)/pharmacodynamic (PD) relationship between amantadine plasma concentrations and antidyskinetic efficacy across multiple species to define optimal therapeutic dosing. The PK profile of amantadine was determined in mice, rats, and macaques. Efficacy data from the 6-hydroxydopamine rat and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine macaque model of LID, along with previously published antidyskinetic efficacy data, were used to establish species-specific PK/PD relationships using a direct-effect maximum possible effect model. Results from the PK/PD model were compared with amantadine plasma concentrations and antidyskinetic effect in a phase 2 study in patients with Parkinson's disease treated with ADS-5102, an extended-release amantadine capsule formulation. Outcomes from each of the species evaluated indicate that the EC50 of amantadine for reducing dyskinesia range from 1025 to 1633 ng/ml (1367 ng/ml for an all-species model). These data are consistent with the mean amantadine plasma concentrations observed in patients with Parkinson's disease (∼1500 ng/ml) treated with ADS-5102 at doses that demonstrated a statistically significant reduction in dyskinesia. These results demonstrate that the EC50 of amantadine for reducing dyskinesia is consistent across multiple species and supports a plasma concentration target of ∼1400 ng/ml to achieve therapeutic efficacy.


Asunto(s)
Amantadina/farmacología , Amantadina/farmacocinética , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Levodopa/farmacología , Animales , Antiparkinsonianos/farmacocinética , Antiparkinsonianos/farmacología , Modelos Animales de Enfermedad , Discinesia Inducida por Medicamentos/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Ann Neurol ; 81(6): 825-836, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28470693

RESUMEN

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.


Asunto(s)
Estimulación Encefálica Profunda/métodos , Enfermedad de Parkinson/terapia , Núcleo Subtalámico , alfa-Sinucleína/administración & dosificación , Animales , Conducta Animal , Dependovirus , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Masculino , Mutación , Ratas , Ratas Sprague-Dawley , Núcleo Subtalámico/citología , Núcleo Subtalámico/metabolismo , Núcleo Subtalámico/fisiopatología
10.
Neurobiol Dis ; 106: 133-146, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28673739

RESUMEN

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.


Asunto(s)
Mitocondrias/metabolismo , Neuronas/metabolismo , Neuroprotección/fisiología , Trastornos Parkinsonianos/metabolismo , Sirtuina 3/metabolismo , alfa-Sinucleína/metabolismo , Acetilación , Animales , Línea Celular Tumoral , Dependovirus/genética , Femenino , Vectores Genéticos , Humanos , Masculino , Ratones Endogámicos C57BL , Mitocondrias/patología , Mutación , Neuronas/patología , Biogénesis de Organelos , Trastornos Parkinsonianos/patología , Ratas Sprague-Dawley , Sirtuina 3/genética , Sustancia Negra/metabolismo , Sustancia Negra/patología , alfa-Sinucleína/genética
11.
J Pharmacol Exp Ther ; 357(3): 562-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27190169

RESUMEN

Striatal dopamine deficiency is the core feature of the pathology of Parkinson's disease (PD), and dopamine replacement with l-3,4-dihydroxyphenylalanine (l-DOPA) is the mainstay of PD treatment. Unfortunately, chronic l-DOPA administration is marred by the emergence of dyskinesia and wearing-off. Alternatives to l-DOPA for alleviation of parkinsonism are of interest, although none can match the efficacy of l-DOPA to date. Catechol-O-methyltransferase and monoamine oxidase inhibitors are currently used to alleviate wearing-off, but they do not increase "on-time" without exacerbating dyskinesia. Alternate approaches to dopamine replacement in parkinsonism generally (and to wearing-off and dyskinesia, specifically) are therefore urgently needed. Inasmuch as they increase synaptic dopamine levels, dopamine transporter (DAT) inhibitors, whether they are selective or have actions on noradrenaline or serotonin transporters, theoretically represent an attractive way to alleviate parkinsonism per se and potentially enhance l-DOPA antiparkinsonian action (provided that sufficient dopamine terminals remain within the striatum). Several nonhuman primate studies and clinical trials have been performed to evaluate the potential of DAT inhibitors for PD. In this article, we review nonhuman primate studies and clinical trials, we summarize the current knowledge of DAT inhibitors in PD, and we propose a hypothesis as to how tailoring the selectivity of DAT inhibitors might maximize the benefits of DAT inhibition in PD.


Asunto(s)
Ensayos Clínicos como Asunto/métodos , Inhibidores de Captación de Dopamina/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Primates , Animales , Inhibidores de Captación de Dopamina/uso terapéutico , Humanos , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico
12.
Pharmacol Rev ; 65(1): 171-222, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23319549

RESUMEN

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.


Asunto(s)
Antiparkinsonianos/efectos adversos , Dopaminérgicos/efectos adversos , Discinesia Inducida por Medicamentos/fisiopatología , Levodopa/efectos adversos , Enfermedad de Parkinson/fisiopatología , Animales , Ganglios Basales/fisiología , Discinesia Inducida por Medicamentos/etiología , Discinesia Inducida por Medicamentos/metabolismo , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Transmisión Sináptica/fisiología
13.
Hum Mol Genet ; 22(2): 358-71, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23077217

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder caused primarily by mutations of the X-linked MECP2 gene. Although the loss of MeCP2 function affects many neural systems, impairments of catecholaminergic function have been hypothesized to underlie several of the cardinal behavioral deficits of RTT patients and Mecp2-deficient mice. Although recent Mecp2 reactivation studies indicate that RTT may be a reversible condition, it remains unclear whether specifically preserving Mecp2 function within a specific system will be sufficient to convey beneficial effects. Here, we test whether the selective preservation of Mecp2 within catecholaminergic cells will improve the phenotype of Mecp2-deficient mice. Our results show that this targeted preservation of Mecp2 significantly improves the lifespan, phenotypic severity and cortical epileptiform discharge activity of both male and female Mecp2-deficient mice. Further, we found that the catecholaminergic preservation of Mecp2 also improves the ambulatory rate, rearing activity, motor coordination, anxiety and nest-building performances of Mecp2-deficient mice of each gender. Interestingly, our results also revealed a gender-specific improvement, as specific cortical and hippocampal electroencephalographic abnormalities were significantly improved in male, but not female, rescue mice. Collectively, these results support the role of the catecholaminergic system in the pathogenesis of RTT and provide proof-of-principle that restoring MeCP2 function within this specific system could represent a treatment strategy for RTT.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas/metabolismo , Fenotipo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Animales , Conducta Animal , Encéfalo/metabolismo , Muerte Súbita , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Hipocampo/metabolismo , Hipocampo/fisiopatología , Longevidad/genética , Masculino , Ratones , Ratones Noqueados , Desempeño Psicomotor , Factores Sexuales , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
14.
Mov Disord ; 30(9): 1283-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26377152

RESUMEN

BACKGROUND: We have previously defined a parkinsonism-related metabolic brain network in rhesus macaques using a high-resolution research positron emission tomography camera. This brief article reports a descriptive pilot study to assess the reproducibility of network activity and regional glucose metabolism in independent parkinsonian macaques using a clinical positron emission tomography/CT camera. METHODS: [(18)F]fluorodeoxyglucose PET scans were acquired longitudinally over 3 months in three drug-naïve parkinsonian and three healthy control cynomolgus macaques. Group difference and test-retest stability in network activity and regional glucose metabolism were evaluated graphically, using all brain images from these macaques. RESULTS: Comparing the parkinsonian macaques with the controls, network activity was elevated and remained stable over 3 months. Normalized glucose metabolism increased in putamen/globus pallidus and sensorimotor regions but decreased in posterior parietal cortices. CONCLUSIONS: Parkinsonism-related network activity can be reliably quantified in different macaques with a clinical positron emission tomography/CT scanner and is reproducible over a period typically employed in preclinical intervention studies. This measure can be a useful biomarker of disease process or drug effects in primate models of Parkinson's disease.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Glucosa/metabolismo , Intoxicación por MPTP/diagnóstico por imagen , Intoxicación por MPTP/patología , Priones/metabolismo , Animales , Mapeo Encefálico , Modelos Animales de Enfermedad , Femenino , Fluorodesoxiglucosa F18/farmacocinética , Macaca fascicularis , Proyectos Piloto , Tomografía de Emisión de Positrones , Radiografía , Tomógrafos Computarizados por Rayos X
15.
Mov Disord ; 30(2): 150-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25607799

RESUMEN

Impulse control disorders in Parkinson's disease are a group of impulsive behaviors most often associated with dopaminergic treatment. Presently, there is a lack of high quality evidence available to guide their management. This manuscript reviews current management strategies, before concentrating on the concept of dopamine agonist withdrawal syndrome and its implications for the management of impulse control disorders. Further, we focus on controversies, including the role of more recently available anti-parkinsonian drugs, and potential future approaches involving routes of drug delivery, nonpharmacological treatments (such as cognitive behavioral therapy and deep brain stimulation), and other as yet experimental strategies.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Estimulación Encefálica Profunda , Trastornos Disruptivos, del Control de Impulso y de la Conducta/terapia , Agonistas de Dopamina/uso terapéutico , Enfermedad de Parkinson/terapia , Animales , Estimulación Encefálica Profunda/métodos , Trastornos Disruptivos, del Control de Impulso y de la Conducta/complicaciones , Humanos , Conducta Impulsiva/efectos de los fármacos , Conducta Impulsiva/fisiología , Enfermedad de Parkinson/complicaciones
16.
Synapse ; 69(3): 99-102, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25559284

RESUMEN

In this pilot study, pioglitazone, an agonist of the peroxisome proliferator-activated receptor gamma (PPAR-γ) used in the treatment of diabetes mellitus, was administered to dyskinetic parkinsonian macaques. Pioglitazone alleviated L-DOPA-induced dyskinesia, but impaired L-DOPA anti-parkinsonian efficacy. These results suggest caution when administering pioglitazone to patients with advanced Parkinson's disease.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Levodopa/uso terapéutico , Intoxicación por MPTP/tratamiento farmacológico , Tiazolidinedionas/uso terapéutico , Animales , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/efectos adversos , Combinación de Medicamentos , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Femenino , Levodopa/administración & dosificación , Levodopa/efectos adversos , Macaca fascicularis , Proyectos Piloto , Pioglitazona , Tiazolidinedionas/administración & dosificación
17.
Behav Pharmacol ; 26(1-2): 101-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25303957

RESUMEN

L-3,4-Dihydroxyphenylalanine (L-DOPA) is the most effective treatment for Parkinson's disease, but chronic administration is complicated by the development of dyskinesia. We have previously demonstrated that the dopamine D4 receptor antagonist L-745,870 reduces the severity of L-DOPA-induced dyskinesia in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned macaque without compromising L-DOPA antiparkinsonian benefits. In the current study, we have addressed the effects of L-745,870 on the expression of L-DOPA-induced abnormal involuntary movements (AIMs) in the 6-hydroxydopamine-lesioned rat. Rats were primed with repeated L-DOPA administration, after which acute challenges of L-DOPA/L-745,870 (vehicle, 0.1, 0.3 and 1 mg/kg) were administered, and AIMs were assessed. Rotarod performance and AIMs were assessed. In L-DOPA-primed rats, L-745,870 (1 mg/kg, but not lower doses) alleviated previously established AIMs (by 84%, P<0.001). Whereas rotarod performance was significantly improved by L-DOPA/vehicle treatment, L-DOPA/L-745,870 failed to improve rotarod performance (P>0.05), suggesting that, in contrast to the MPTP-lesioned macaque, L-745,870 reduces L-DOPA antiparkinsonian benefit in the rat model. Overall, these data suggest that L-745,870 may have a narrow therapeutic window as an antidyskinetic agent in advanced Parkinson's disease.


Asunto(s)
Discinesia Inducida por Medicamentos/prevención & control , Levodopa/toxicidad , Trastornos Parkinsonianos/tratamiento farmacológico , Piridinas/farmacología , Pirroles/farmacología , Animales , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/farmacología , Antiparkinsonianos/toxicidad , Modelos Animales de Enfermedad , Antagonistas de Dopamina/administración & dosificación , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Discinesia Inducida por Medicamentos/etiología , Femenino , Levodopa/administración & dosificación , Levodopa/farmacología , Oxidopamina/toxicidad , Trastornos Parkinsonianos/fisiopatología , Piridinas/administración & dosificación , Pirroles/administración & dosificación , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
18.
Neurobiol Dis ; 71: 334-44, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25171793

RESUMEN

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.


Asunto(s)
Antiparkinsonianos/efectos adversos , Cuerpo Estriado/patología , Discinesia Inducida por Medicamentos/patología , Levodopa/efectos adversos , Vías Nerviosas/patología , Plasticidad Neuronal/fisiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , Discinesia Inducida por Medicamentos/etiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Lateralidad Funcional , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Ratones , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Purinérgicos/farmacología , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
19.
Neuropharmacology ; 248: 109880, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38412888

RESUMEN

Repurposing regulatory agency-approved molecules, with proven safety in humans, is an attractive option for developing new treatments for disease. We identified and assessed the efficacy of 3 drugs predicted by an in silico screen as having the potential to treat l-DOPA-induced dyskinesia (LID) in Parkinson's disease. We analysed ∼1.3 million Medline abstracts using natural language processing and ranked 3539 existing drugs based on predicted ability to reduce LID. 3 drugs from the top 5% of the 3539 candidates; lorcaserin, acamprosate and ganaxolone, were prioritized for preclinical testing based on i) having a novel mechanism of action, ii) having not been previously validated for the treatment of LID, iii) being blood-brain-barrier penetrant and orally bioavailable and iv) being clinical trial ready. We assessed the efficacy of acamprosate, ganaxolone and lorcaserin in a rodent model of l-DOPA-induced hyperactivity, with lorcaserin affording a 58% reduction in rotational asymmetry (P < 0.05) compared to vehicle. Acamprosate and ganaxolone failed to demonstrate efficacy. Lorcaserin, a 5HT2C agonist, was then further tested in MPTP lesioned dyskinetic macaques where it afforded an 82% reduction in LID (P < 0.05), unfortunately accompanied by a significant increase in parkinsonian disability. In conclusion, although our data do not support the repurposing of lorcaserin, acamprosate or ganaxolone per se for LID, we demonstrate value of an in silico approach to identify candidate molecules which, in combination with an in vivo screen, can facilitate clinical development decisions. The present study adds to a growing literature in support of this paradigm shifting approach in the repurposing pipeline.


Asunto(s)
Discinesia Inducida por Medicamentos , Levodopa , Humanos , Animales , Levodopa/efectos adversos , Inteligencia Artificial , Reposicionamiento de Medicamentos , Acamprosato/uso terapéutico , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Macaca , Antiparkinsonianos/efectos adversos , Modelos Animales de Enfermedad
20.
J Neurophysiol ; 109(2): 363-74, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23076106

RESUMEN

Deep brain stimulation (DBS) employing high-frequency stimulation (HFS) is commonly used in the globus pallidus interna (GPi) and the subthalamic nucleus (STN) for treating motor symptoms of patients with Parkinson's disease (PD). Although DBS improves motor function in most PD patients, disease progression and stimulation-induced nonmotor complications limit DBS in these areas. In this study, we assessed whether stimulation of the substantia nigra pars reticulata (SNr) improved motor function. Hemiparkinsonian rats predominantly touched with their unimpaired forepaw >90% of the time in the stepping and limb-use asymmetry tests. After SNr-HFS (150 Hz), rats touched equally with both forepaws, similar to naive and sham-lesioned rats. In vivo, SNr-HFS decreased beta oscillations (12-30 Hz) in the SNr of freely moving hemiparkinsonian rats and decreased SNr neuronal spiking activity from 28 ± 1.9 Hz before stimulation to 0.8 ± 1.9 Hz during DBS in anesthetized animals; also, neuronal spiking activity increased from 7 ± 1.6 to 18 ± 1.6 Hz in the ventromedial portion of the thalamus (VM), the primary SNr efferent. In addition, HFS of the SNr in brain slices from normal and reserpine-treated rat pups resulted in a depolarization block of SNr neuronal activity. We demonstrate improvement of forelimb akinesia with SNr-HFS and suggest that this motor effect may have resulted from the attenuation of SNr neuronal activity, decreased SNr beta oscillations, and increased activity of VM thalamic neurons, suggesting that the SNr may be a plausible DBS target for treating motor symptoms of DBS.


Asunto(s)
Estimulación Encefálica Profunda , Hipocinesia/terapia , Enfermedad de Parkinson Secundaria/terapia , Sustancia Negra/fisiopatología , Animales , Antipsicóticos/uso terapéutico , Ritmo beta , Miembro Anterior/inervación , Miembro Anterior/fisiopatología , Masculino , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/fisiopatología , Ratas , Ratas Sprague-Dawley , Reserpina/uso terapéutico , Tálamo/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA