Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FEBS Open Bio ; 11(2): 354-366, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33301617

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder caused by the selective loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). Lewy bodies (LBs), another histological hallmark of PD, are observed in patients with familial or sporadic PD. The therapeutic potential of reducing the accumulation of α-synuclein, a major LB component, has been investigated, but it remains unknown whether the formation of LBs results in the loss of DA neurons. PARK4 patients exhibit multiplication of the α-synuclein gene (SNCA) without any pathological mutations, but their symptoms develop relatively early. Therefore, study of PARK4 might help elucidate the mechanism of α-synuclein aggregation. In this study, we investigated the dynamics of α-synuclein during the early stage of immature DA neurons, which were differentiated from human-induced pluripotent stem cells (hiPSCs) derived from either a PARK4 patient with SNCA triplication or a healthy donor. We observed increased α-synuclein accumulation in PARK4 hiPSC-derived DA neurons relative to those derived from healthy donor hiPSCs. Interestingly, α-synuclein accumulation disappeared over time in the PARK4 patient-derived DA neurons. Moreover, an SNCA-specific antisense oligonucleotide could reduce α-synuclein levels during the accumulation stage. These observations may help reveal the mechanisms that regulate α-synuclein levels, which may consequently be useful in the development of new therapies for patients with sporadic or familial PD.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Enfermedad de Parkinson/patología , alfa-Sinucleína/deficiencia , Diferenciación Celular , Células Cultivadas , Variaciones en el Número de Copia de ADN , Neuronas Dopaminérgicas/efectos de los fármacos , Duplicación de Gen , Voluntarios Sanos , Humanos , Células Madre Pluripotentes Inducidas , Enfermedad por Cuerpos de Lewy/genética , Enfermedad de Parkinson/genética , Cultivo Primario de Células , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
2.
Sci Rep ; 10(1): 22098, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33328543

RESUMEN

Lysosomal dysfunction is an emerging feature in the pathology of Parkinson's disease and Dementia with Lewy bodies. Mutations in the GBA gene, encoding the enzyme Glucocerebrosidase (GCase), have been identified as a genetic risk factor for these synucleinopathies. As a result, there has been a growing interest in the involvement of GCase in these diseases. This GCase activity assay is based on the catalytic hydrolysis of 4-methylumbelliferyl ß-D-glucopyranoside that releases the highly fluorescent 4-methylumbelliferyl (4-MU). The final assay protocol was tested for the following parameters: Lower limit of quantification (LLOQ), precision, parallelism, linearity, spike recovery, number of freeze-thaw events, and sample handling stability. The GCase activity assay is within acceptable criteria for parallelism, precision and spike recovery. The LLOQ of this assay corresponds to an enzymatic activity of generating 0.26 pmol 4-MU/min/ml. The enzymatic activity was stable when samples were processed and frozen at - 80 °C within 4 h after the lumbar puncture procedure. Repetitive freeze-thaw events significantly decreased enzyme activity. We present the validation of an optimized in vitro GCase activity assay, based on commercially available components, to quantify its enzymatic activity in human cerebrospinal fluid and the assessment of preanalytical factors.


Asunto(s)
Glucosilceramidasa/líquido cefalorraquídeo , Cuerpos de Lewy/enzimología , Enfermedad de Parkinson/líquido cefalorraquídeo , alfa-Sinucleína/genética , Fluorometría/métodos , Glucosilceramidasa/genética , Humanos , Técnicas In Vitro , Cuerpos de Lewy/patología , Lisosomas/genética , Lisosomas/patología , Mutación/genética , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/patología , Factores de Riesgo , alfa-Sinucleína/deficiencia
4.
Toxicology ; 438: 152461, 2020 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-32278788

RESUMEN

The α-Synuclein (α-syn) and tau have synergistic effects on neurodegenerative diseases induced by environmental factors or genetic mutation. Thus, we investigated the role of α-syn and tau in neurodegeneration induced by chronic methamphetamine (METH) exposure (1.0∼20.0 mg/kg/d body weight, for 14 consecutive days). Here, we present a mice model with evidences of α-syn and tau participating in toxicology in chronic METH. METH increased α-syn level in the stratum oriens, pyramidal layer, stratum radiatum and stratum moleculare of hippocampal CA1, CA2 and CA3, polymorph layer of hippocampal dentate gyrus (DG), and substantia nigra (SN). The subcellular locations of the upregulated α-syn were mainly found in mitochondria and axons. The METH upregulated α-syn may directly induce mitochondrial damage, myelin sheath destruction, and synaptic failure. Also, the excess α-syn might indirectly promote tau phosphorylation through tau kinase GSK3ß and CDK5, leading to microtubule depolymerization and eventually fusion deficit of autophagosome and lysosome. In the in vitro experiment, the autophagic vacuoles failed to fuse with the lysosome. The neuropathology induced by both the direct and indirect effects of α-syn could be alleviated by α-syn knockout. Taking together, these results indicate that the α-syn mediates the neurodegenerative process induced by chronic METH and that reducing α-syn might be a potential approach to protect the toxic effects of METH and also be, to a broader view, of therapeutic value in neurodegenerative diseases.


Asunto(s)
Hipocampo/metabolismo , Degeneración Nerviosa , Neuronas/metabolismo , Síndromes de Neurotoxicidad/prevención & control , Sustancia Negra/metabolismo , alfa-Sinucleína/deficiencia , Animales , Autofagia , Axones/metabolismo , Axones/ultraestructura , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/ultraestructura , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Metanfetamina , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Neuronas/ultraestructura , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Fosforilación , Sustancia Negra/ultraestructura , Factores de Tiempo , alfa-Sinucleína/genética , Proteínas tau/metabolismo
5.
J Neurochem ; 152(1): 61-71, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31520492

RESUMEN

It has been suggested that extracellular alpha synuclein (αSyn) can mediate neuroinflammation in Parkinson's disease, and that αSyn affects B-cell maturation. However, the function of αSyn in T cells is poorly understood. We hypothesized that αSyn can affect CD4+ T-cell proliferation and activity. We found that αSyn deficiency exacerbates disease progression in 8 weeks old C57BL6/J EAE-induced mice, and that αSyn-deficient CD4+ T cells have increased pro-inflammatory response to myelin antigen relative to wild-type cells, as measured by cytokine secretion of interleukin IL-17 and interferon gamma. Furthermore, expression of αSyn on a background of αSyn knockout mitigates the inflammatory responses in CD4+ T cells. We discovered that elevated levels of Nurr1, a transcription factor belonging to the orphan nuclear receptor family, are associated with the pro-inflammatory profile of αSyn-deficient CD4+ T cells. In addition, we demonstrated that silencing of Nurr1 expression using an siRNA reduces IL-17 levels and increases the levels of IL-10, an anti-inflammatory cytokine. Study of αSyn-mediated cellular pathways in CD4+ T cells may provide useful insights into the development of pro-inflammatory responses in immunity, providing future avenues for therapeutic intervention.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Activación de Linfocitos/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , alfa-Sinucleína/deficiencia , Animales , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Inflamación/inmunología , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/inmunología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Células TH1/inmunología , alfa-Sinucleína/genética , alfa-Sinucleína/fisiología
6.
J Neurochem ; 150(5): 475-486, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31269263

RESUMEN

The protein α-synuclein has a central role in the pathogenesis of Parkinson's disease (PD). In this review, we discuss recent results concerning its primary function, which appears to be on cell membranes. The pre-synaptic location of synuclein has suggested a role in neurotransmitter release and it apparently associates with synaptic vesicles because of their high curvature. Indeed, synuclein over-expression inhibits synaptic vesicle exocytosis. However, loss of synuclein has not yet been shown to have a major effect on synaptic transmission. Consistent with work showing that synuclein can promote as well as sense membrane curvature, recent analysis of synuclein triple knockout mice now shows that synuclein accelerates dilation of the exocytic fusion pore. This form of regulation affects primarily the release of slowly discharged lumenal cargo such as neural peptides, but presumably also contributes to maintenance of the release site. This article is part of the Special Issue "Synuclein".


Asunto(s)
Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/fisiología , Animales , Axones/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Exocitosis/fisiología , Humanos , Fusión de Membrana/fisiología , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/patología , Mutación Missense , Terminales Presinápticos/química , Dominios Proteicos , Pliegue de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiología , Proteínas Recombinantes/metabolismo , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , alfa-Sinucleína/química , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética
7.
Neuron ; 103(4): 627-641.e7, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31255487

RESUMEN

Analysis of human pathology led Braak to postulate that α-synuclein (α-syn) pathology could spread from the gut to brain via the vagus nerve. Here, we test this postulate by assessing α-synucleinopathy in the brain in a novel gut-to-brain α-syn transmission mouse model, where pathological α-syn preformed fibrils were injected into the duodenal and pyloric muscularis layer. Spread of pathologic α-syn in brain, as assessed by phosphorylation of serine 129 of α-syn, was observed first in the dorsal motor nucleus, then in caudal portions of the hindbrain, including the locus coeruleus, and much later in basolateral amygdala, dorsal raphe nucleus, and the substantia nigra pars compacta. Moreover, loss of dopaminergic neurons and motor and non-motor symptoms were observed in a similar temporal manner. Truncal vagotomy and α-syn deficiency prevented the gut-to-brain spread of α-synucleinopathy and associated neurodegeneration and behavioral deficits. This study supports the Braak hypothesis in the etiology of idiopathic Parkinson's disease (PD).


Asunto(s)
Transporte Axonal , Trastornos Parkinsonianos/etiología , Agregado de Proteínas , Nervio Vago/metabolismo , alfa-Sinucleína/farmacocinética , Animales , Química Encefálica , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Duodeno/inervación , Duodeno/metabolismo , Humanos , Inyecciones Intramusculares , Cuerpos de Lewy/metabolismo , Aprendizaje por Laberinto , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Neurológicos , Músculo Liso/inervación , Músculo Liso/metabolismo , Comportamiento de Nidificación/fisiología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/prevención & control , Trastornos Parkinsonianos/psicología , Fosforilación , Procesamiento Proteico-Postraduccional , Píloro/inervación , Píloro/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Vagotomía , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/deficiencia , alfa-Sinucleína/toxicidad
8.
J Neurochem ; 150(5): 591-604, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31165472

RESUMEN

Accumulation of alpha-synuclein protein aggregates is the hallmark neuropathologic feature of synucleinopathies such as Parkinson's disease. Rare point mutations and multiplications in SNCA, the gene encoding alpha-synuclein, as well as other genetic alterations are linked to familial Parkinson's disease cases with high penetrance and hence constitute major genetic risk factors for Parkinson's disease. However, the preponderance of cases seems sporadic, most likely based on a complex interplay between genetic predispositions, aging processes and environmental influences. Deciphering the impact of these environmental factors and their interactions with the individual genetic background in humans is challenging and often requires large cohorts, complicated study designs, and longitudinal set-ups. In contrast, rodent models offer an ideal system to study the influence of individual environmental aspects under controlled genetic background and standardized conditions. In this review, we highlight findings from studies examining effects of environmental enrichment mimicking stimulation of the brain by its physical and social surroundings as well as of environmental stressors on brain health in the context of Parkinson's disease. We discuss possible internal molecular transducers of such environmental cues in Parkinson's disease rodent models and emphasize their potential in developing novel avenues to much-needed therapies for this still incurable disease. This article is part of the Special Issue "Synuclein".


Asunto(s)
Encéfalo/metabolismo , Interacción Gen-Ambiente , Trastornos Parkinsonianos/etiología , Sinucleinopatías/etiología , alfa-Sinucleína/fisiología , Animales , Encéfalo/patología , Enfermedades en Gemelos/genética , Epigénesis Genética , Humanos , Cuerpos de Lewy/metabolismo , Ratones , Ratones Noqueados , Actividad Motora , Enfermedad de Parkinson/genética , Trastornos Parkinsonianos/genética , Plaguicidas/toxicidad , Estimulación Física , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Factores de Riesgo , Estrés Fisiológico , Estrés Psicológico/complicaciones , Sinucleinopatías/genética , Sinucleinopatías/metabolismo , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética
9.
Neuromolecular Med ; 21(3): 239-249, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31079293

RESUMEN

Emerging findings suggest that Parkinson's disease (PD) pathology (α-synuclein accumulation) and neuronal dysfunction may occur first in peripheral neurons of the autonomic nervous system including the enteric branches of the vagus nerve. The risk of PD increases greatly in people over the age of 65, a period of life in which chronic inflammation is common in many organ systems including the gut. Here we report that chronic mild focal intestinal inflammation accelerates the age of disease onset in α-synuclein mutant PD mice. Wild-type and PD mice treated with 0.5% dextran sodium sulfate (DSS) in their drinking water for 12 weeks beginning at 3 months of age exhibited histological and biochemical features of mild gut inflammation. The age of onset of motor dysfunction, evaluated using a rotarod test, gait analysis, and grip strength measurements, was significantly earlier in DSS-treated PD mice compared to control PD mice. Levels of the dopaminergic neuron marker tyrosine hydroxylase in the striatum and numbers of dopaminergic neurons in the substantia nigra were reduced in PD mice with gut inflammation. Levels of total and phosphorylated α-synuclein were elevated in enteric and brain neurons in DSS-treated PD mice, suggesting that mild gut inflammation accelerates α-synuclein pathology. Markers of inflammation in the colon and brain, but not in the blood, were elevated in DSS-treated PD mice, consistent with retrograde transneuronal propagation of α-synuclein pathology and neuroinflammation from the gut to the brain. Our findings suggest that interventions that reduce gut inflammation may prove beneficial in the prevention and treatment of PD.


Asunto(s)
Encéfalo/patología , Colitis/complicaciones , Enteritis/complicaciones , Trastornos Parkinsonianos/etiología , Sinucleinopatías/etiología , alfa-Sinucleína/deficiencia , Animales , Colitis/inducido químicamente , Colon/patología , Cuerpo Estriado/enzimología , Cuerpo Estriado/patología , Citocinas/sangre , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Enteritis/inducido químicamente , Femenino , Trastornos Neurológicos de la Marcha/etiología , Fuerza de la Mano , Humanos , Intestino Delgado/patología , Masculino , Ratones , Ratones Noqueados , Mutación Missense , Proteínas del Tejido Nervioso/análisis , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Prueba de Desempeño de Rotación con Aceleración Constante , Sustancia Negra/enzimología , Sustancia Negra/patología , Sinucleinopatías/genética , Sinucleinopatías/patología , Tirosina 3-Monooxigenasa/análisis , alfa-Sinucleína/genética
10.
Mol Neurobiol ; 56(11): 7448-7457, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31041657

RESUMEN

Prion diseases are a group of neurodegenerative disorders associated with the conformational conversion of the cellular prion protein (PrPC) into an abnormal misfolded form named PrPSc. Other than accumulating in the brain, PrPSc can bind PrPC and force it to change conformation to PrPSc. The exact mechanism which underlies the process of PrPC/PrPSc conversion still needs to be defined and many molecules or cofactors might be involved. Several studies have documented an important role of PrPC to act as receptor for abnormally folded forms of α-synuclein which are responsible of a group of diseases known as synucleinopathies. The presence of PrPC was required to promote efficient internalization and spreading of abnormal α-synuclein between cells. In this work, we have assessed whether α-synuclein exerts any role in PrPSc conversion and propagation either in vitro or in vivo. Indeed, understanding the mechanism of PrPC/PrPSc conversion and the identification of cofactors involved in this process is crucial for developing new therapeutic strategies. Our results showed that PrPSc was able to efficiently propagate in the brain of animals even in the absence of α-synuclein thus suggesting that this protein did not act as key modulator of prion propagation. Thus, α-synuclein might take part in this process but is not specifically required for sustaining prion conversion and propagation.


Asunto(s)
Priones/metabolismo , alfa-Sinucleína/deficiencia , Animales , Encéfalo/metabolismo , Endopeptidasa K/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Pliegue de Proteína , alfa-Sinucleína/metabolismo
11.
Nat Commun ; 10(1): 973, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30846695

RESUMEN

In Parkinson's disease (PD) there is a selective degeneration of neuromelanin-containing neurons, especially substantia nigra dopaminergic neurons. In humans, neuromelanin accumulates with age, the latter being the main risk factor for PD. The contribution of neuromelanin to PD pathogenesis remains unknown because, unlike humans, common laboratory animals lack neuromelanin. Synthesis of peripheral melanins is mediated by tyrosinase, an enzyme also present at low levels in the brain. Here we report that overexpression of human tyrosinase in rat substantia nigra results in age-dependent production of human-like neuromelanin within nigral dopaminergic neurons, up to levels reached in elderly humans. In these animals, intracellular neuromelanin accumulation above a specific threshold is associated to an age-dependent PD phenotype, including hypokinesia, Lewy body-like formation and nigrostriatal neurodegeneration. Enhancing lysosomal proteostasis reduces intracellular neuromelanin and prevents neurodegeneration in tyrosinase-overexpressing animals. Our results suggest that intracellular neuromelanin levels may set the threshold for the initiation of PD.


Asunto(s)
Encéfalo/metabolismo , Melaninas/biosíntesis , Monofenol Monooxigenasa/metabolismo , Enfermedad de Parkinson/metabolismo , Envejecimiento/metabolismo , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Humanos , Cuerpos de Lewy/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monofenol Monooxigenasa/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sustancia Negra/metabolismo , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
12.
Blood Cells Mol Dis ; 73: 33-37, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30195626

RESUMEN

Effects of α-synuclein deficiency on cellular blood components have not been extensively investigated. This study evaluated ultrastructural changes of leukocytes in α-synuclein knockout (KO) mice using electron microscopy (EM). The following ultrastructural characteristics were quantified in leukocytes: mitochondria, primary granules, specific granules (SG), Golgi apparatus (GA), inclusions, rough-endoplasmic reticulum (RER), smooth-endoplasmic reticulum (SER), and cellular projections (CP). EM showed increased numbers or amounts of SG, inclusions, and SER in KO group (5.3 ±â€¯4.5 in WT vs. 14.1 ±â€¯10.3 in KO, p = 0.02; 0.4 ±â€¯0.9 in WT vs. 3.2 ±â€¯2.8 in KO, p = 0.007; and 7.7 ±â€¯6.7 in WT vs. 17.7 ±â€¯12.2 in KO, p = 0.03, respectively). Although CP number was not significantly different between the two groups (13.4 ±â€¯5.3 in WT vs. 16.3 ±â€¯7.5 in KO, p = 0.32), their size and shapes were altered in KO mice. Notably, findings occurred in the setting of significant lymphopenia. α-Synuclein deficiency leads to changes in size and shape of secretory particles and increases in SER, SG, and inclusions, indicating a potential role for α-synuclein in vesicular trafficking in leukocytes. Further studies are needed to elucidate functions mediated by α-synuclein.


Asunto(s)
Leucocitos/ultraestructura , alfa-Sinucleína/deficiencia , Animales , Forma de la Célula , Extensiones de la Superficie Celular , Ratones , Ratones Noqueados , Microscopía Electrónica , Vesículas Transportadoras , alfa-Sinucleína/genética , alfa-Sinucleína/farmacología
13.
Mol Genet Metab ; 122(4): 198-208, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29173981

RESUMEN

Mutations in GBA1 encountered in Gaucher disease are a leading risk factor for Parkinson disease and associated Lewy body disorders. Many GBA1 mutation carriers, especially those with severe or null GBA1 alleles, have earlier and more progressive parkinsonism. To model the effect of partial glucocerebrosidase deficiency on neurological progression in vivo, mice with a human A53T α-synuclein (SNCAA53T) transgene were crossed with heterozygous null gba mice (gba+/-). Survival analysis of 84 mice showed that in gba+/-//SNCAA53T hemizygotes and homozygotes, the symptom onset was significantly earlier than in gba+/+//SNCAA53T mice (p-values 0.023-0.0030), with exacerbated disease progression (p-value <0.0001). Over-expression of SNCAA53T had no effect on glucocerebrosidase levels or activity. Immunoblotting demonstrated that gba haploinsufficiency did not lead to increased levels of either monomeric SNCA or insoluble high molecular weight SNCA in this model. Immunohistochemical analyses demonstrated that the abundance and distribution of SNCA pathology was also unaltered by gba haploinsufficiency. Thus, while the underlying mechanism is not clear, this model shows that gba deficiency impacts the age of onset and disease duration in aged SNCAA53T mice, providing a valuable resource to identify modifiers, pathways and possible moonlighting roles of glucocerebrosidase in Parkinson pathogenesis.


Asunto(s)
Enfermedad de Gaucher/genética , Glucosilceramidasa/genética , Haploinsuficiencia , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Edad de Inicio , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Enfermedad de Gaucher/complicaciones , Glucosilceramidasa/deficiencia , Glucosilceramidas/análisis , Heterocigoto , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Enfermedad de Parkinson/etiología , Psicosina/análogos & derivados , Psicosina/análisis , Transgenes , alfa-Sinucleína/análisis , alfa-Sinucleína/deficiencia , alfa-Sinucleína/metabolismo , beta-Glucosidasa/deficiencia , beta-Glucosidasa/genética
14.
Acta Neuropathol Commun ; 5(1): 37, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28482862

RESUMEN

α-Synuclein is a protein involved in the pathogenesis of synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). We investigated the role of neuronal α-Syn in myelin composition and abnormalities. The phospholipid content of purified myelin was determined by 31P NMR in two mouse lines modeling PD, PrP-A53T α-Syn and Thy-1 wt-α-Syn. Significantly higher levels of phospholipids were detected in myelin purified from brains of these α-Syn transgenic mouse models than in control mice. Nevertheless, myelin ultrastructure appeared intact. To further investigate the effect of α-Syn on myelin abnormalities, we systematically analyzed the striatum, a brain region associated with neurodegeneration in PD. An age and disease-dependent loss of myelin basic protein (MBP) signal was detected by immunohistochemistry in striatal striosomes (patches). The age-dependent loss of MBP signal was associated with lower P25α levels in oligodendrocytes. In addition, we found that α-Syn inhibited oligodendrocyte maturation and the formation of membranous sheets in vitro. Based on these results we concluded that neuronal α-Syn is involved in the regulation and/or maintenance of myelin phospholipid. However, axonal hypomyelination in the PD models is evident only in progressive stages of the disease and associated with α-Syn toxicity.


Asunto(s)
Encéfalo/metabolismo , Vaina de Mielina/metabolismo , Neuronas/metabolismo , Fosfolípidos/metabolismo , alfa-Sinucleína/metabolismo , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/patología , Encéfalo/ultraestructura , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Neuronas/patología , Neuronas/ultraestructura , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Presenilina-1/genética , Presenilina-1/metabolismo , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética
15.
eNeuro ; 4(2)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28462393

RESUMEN

Increased α-synuclein (αsyn) and mitochondrial dysfunction play central roles in the pathogenesis of Parkinson's disease (PD), and lowering αsyn is under intensive investigation as a therapeutic strategy for PD. Increased αsyn levels disrupt mitochondria and impair respiration, while reduced αsyn protects against mitochondrial toxins, suggesting that interactions between αsyn and mitochondria influences the pathologic and physiologic functions of αsyn. However, we do not know if αsyn affects normal mitochondrial function or if lowering αsyn levels impacts bioenergetic function, especially at the nerve terminal where αsyn is enriched. To determine if αsyn is required for normal mitochondrial function in neurons, we comprehensively evaluated how lowering αsyn affects mitochondrial function. We found that αsyn knockout (KO) does not affect the respiration of cultured hippocampal neurons or cortical and dopaminergic synaptosomes, and that neither loss of αsyn nor all three (α, ß and γ) syn isoforms decreased mitochondria-derived ATP levels at the synapse. Similarly, neither αsyn KO nor knockdown altered the capacity of synaptic mitochondria to meet the energy requirements of synaptic vesicle cycling or influenced the localization of mitochondria to dopamine (DA) synapses in vivo. Finally, αsyn KO did not affect overall energy metabolism in mice assessed with a Comprehensive Lab Animal Monitoring System. These studies suggest either that αsyn has little or no significant physiological effect on mitochondrial bioenergetic function, or that any such functions are fully compensated for when lost. These results implicate that αsyn levels can be reduced in neurons without impairing (or improving) mitochondrial bioenergetics or distribution.


Asunto(s)
Mitocondrias/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , alfa-Sinucleína/metabolismo , Animales , Dopamina/metabolismo , Hipocampo/metabolismo , Ratones Noqueados , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética
17.
Acta Neuropathol Commun ; 5(1): 7, 2017 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-28086964

RESUMEN

Neurodegenerative disorders such as Parkinson's Disease (PD), PD dementia (PDD) and Dementia with Lewy bodies (DLB) are characterized by progressive accumulation of α-synuclein (α-syn) in neurons. Recent studies have proposed that neuron-to-neuron propagation of α-syn plays a role in the pathogenesis of these disorders. We have previously shown that antibodies against the C-terminus of α-syn reduce the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy, probably by abrogating the axonal transport and accumulation of α-syn in in vivo models. Here, we assessed the effect of passive immunization against α-syn in a new mouse model of axonal transport and accumulation of α-syn. For these purpose, non-transgenic, α-syn knock-out and mThy1-α-syn tg (line 61) mice received unilateral intra-cerebral injections with a lentiviral (LV)-α-syn vector construct followed by systemic administration of the monoclonal antibody 1H7 (recognizes amino acids 91-99) or control IgG for 3 months. Cerebral α-syn accumulation and axonopathy was assessed by immunohistochemistry and effects on behavior were assessed by Morris water maze. Unilateral LV-α-syn injection resulted in axonal propagation of α-syn in the contra-lateral site with subsequent behavioral deficits and axonal degeneration. Passive immunization with 1H7 antibody reduced the axonal accumulation of α-syn in the contra-lateral side and ameliorated the behavioral deficits. Together this study supports the notion that immunotherapy might improve the deficits in models of synucleinopathy by reducing the axonal propagation and accumulation of α-syn. This represents a potential new mode of action through which α-syn immunization might work.


Asunto(s)
Axones/patología , Encéfalo/patología , Inmunización Pasiva , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , alfa-Sinucleína/inmunología , alfa-Sinucleína/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Transporte Axonal , Axones/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Lateralidad Funcional , Vectores Genéticos , Humanos , Lentivirus , Aprendizaje por Laberinto/fisiología , Ratones Transgénicos , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/psicología , alfa-Sinucleína/deficiencia , alfa-Sinucleína/genética
18.
Dis Model Mech ; 10(5): 619-631, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28108469

RESUMEN

Parkinson's disease (PD) is a frequent neurodegenerative process in old age. Accumulation and aggregation of the lipid-binding SNARE complex component α-synuclein (SNCA) underlies this vulnerability and defines stages of disease progression. Determinants of SNCA levels and mechanisms of SNCA neurotoxicity have been intensely investigated. In view of the physiological roles of SNCA in blood to modulate vesicle release, we studied blood samples from a new large pedigree with SNCA gene duplication (PARK4 mutation) to identify effects of SNCA gain of function as potential disease biomarkers. Downregulation of complexin 1 (CPLX1) mRNA was correlated with genotype, but the expression of other Parkinson's disease genes was not. In global RNA-seq profiling of blood from presymptomatic PARK4 indviduals, bioinformatics detected significant upregulations for platelet activation, hemostasis, lipoproteins, endocytosis, lysosome, cytokine, Toll-like receptor signaling and extracellular pathways. In PARK4 platelets, stimulus-triggered degranulation was impaired. Strong SPP1, GZMH and PLTP mRNA upregulations were validated in PARK4. When analysing individuals with rapid eye movement sleep behavior disorder, the most specific known prodromal stage of general PD, only blood CPLX1 levels were altered. Validation experiments confirmed an inverse mutual regulation of SNCA and CPLX1 mRNA levels. In the 3'-UTR of the CPLX1 gene we identified a single nucleotide polymorphism that is significantly associated with PD risk. In summary, our data define CPLX1 as a PD risk factor and provide functional insights into the role and regulation of blood SNCA levels. The new blood biomarkers of PARK4 in this Turkish family might become useful for PD prediction.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Biomarcadores/sangre , Predisposición Genética a la Enfermedad , Enfermedad por Cuerpos de Lewy/sangre , Proteínas del Tejido Nervioso/genética , Enfermedad de Parkinson/genética , Trastorno de la Conducta del Sueño REM/sangre , ARN/sangre , alfa-Sinucleína/deficiencia , Femenino , Heterocigoto , Humanos , Enfermedad por Cuerpos de Lewy/genética , Persona de Mediana Edad , Enfermedad de Parkinson/sangre , Trastorno de la Conducta del Sueño REM/fisiopatología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , alfa-Sinucleína/sangre , alfa-Sinucleína/genética
20.
J Neuroinflammation ; 13(1): 201, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27565429

RESUMEN

BACKGROUND: Increased α-synuclein immunoreactivity has been associated with inflammatory activity in multiple sclerosis (MS) lesions, but the function of α-synuclein in neuroinflammation remains unknown. The aim of this study was to examine the role of α-synuclein in immunological processes in murine experimental autoimmune encephalomyelitis (EAE) as a model of MS. FINDINGS: We studied EAE in wildtype (aSyn(+/+)) and α-synuclein knockout (aSyn(-/-)) mice on a C57BL/6N background. In the spleen and spinal cord of aSyn(+/+) mice, we observed a gradual reduction of α-synuclein expression during EAE, starting already in the pre-symptomatic disease phase. Compared to aSyn(+/+) mice, aSyn(-/-) mice showed an earlier onset of symptoms but no differences in symptom severity at the peak of disease. Earlier symptom onset was accompanied by increased spinal cord infiltration of CD4(+) T cells, predominantly of interferon-γ-producing T helper 1 (Th1) cells, and reduced infiltration of regulatory T cells, whereas antigen-presenting cells were unaltered. Pre-symptomatically, aSyn(-/-) mice exhibited hyperproliferative CD4(+) splenocytes consistent with increased splenic interleukin-2 mRNA expression, resulting in increased numbers of Th1 cells in the spleen at the onset of symptoms. CONCLUSIONS: Our findings indicate a functional role of α-synuclein in early EAE by increasing Th1 cell-mediated immune response.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células TH1/patología , alfa-Sinucleína/deficiencia , Análisis de Varianza , Animales , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , ARN Mensajero , Médula Espinal/patología , alfa-Sinucleína/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...