Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(3)2021 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-33494388

RESUMEN

Epidemiological studies associate milk consumption with an increased risk of Parkinson's disease (PD) and type 2 diabetes mellitus (T2D). PD is an α-synucleinopathy associated with mitochondrial dysfunction, oxidative stress, deficient lysosomal clearance of α-synuclein (α-syn) and aggregation of misfolded α-syn. In T2D, α-syn promotes co-aggregation with islet amyloid polypeptide in pancreatic ß-cells. Prion-like vagal nerve-mediated propagation of exosomal α-syn from the gut to the brain and pancreatic islets apparently link both pathologies. Exosomes are critical transmitters of α-syn from cell to cell especially under conditions of compromised autophagy. This review provides translational evidence that milk exosomes (MEX) disturb α-syn homeostasis. MEX are taken up by intestinal epithelial cells and accumulate in the brain after oral administration to mice. The potential uptake of MEX miRNA-148a and miRNA-21 by enteroendocrine cells in the gut, dopaminergic neurons in substantia nigra and pancreatic ß-cells may enhance miRNA-148a/DNMT1-dependent overexpression of α-syn and impair miRNA-148a/PPARGC1A- and miRNA-21/LAMP2A-dependent autophagy driving both diseases. MiRNA-148a- and galactose-induced mitochondrial oxidative stress activate c-Abl-mediated aggregation of α-syn which is exported by exosome release. Via the vagal nerve and/or systemic exosomes, toxic α-syn may spread to dopaminergic neurons and pancreatic ß-cells linking the pathogenesis of PD and T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Exosomas/metabolismo , Galactosa/metabolismo , Leche/metabolismo , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animales , Autofagia , Transporte Biológico , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Galactosa/farmacología , Humanos , Lisosomas/metabolismo , MicroARNs/genética , Estrés Oxidativo , Enfermedad de Parkinson/patología , Transducción de Señal , alfa-Sinucleína/efectos adversos
2.
Ann Neurol ; 86(4): 593-606, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31343083

RESUMEN

OBJECTIVE: Excessive inflammation in the central nervous system (CNS) and the periphery can result in neurodegeneration and parkinsonism. Recent evidence suggests that immune responses in Parkinson disease patients are dysregulated, leading to an increased inflammatory reaction to unspecific triggers. Although α-synuclein pathology is the hallmark of Parkinson disease, it has not been investigated whether pathologic α-synuclein is a specific trigger for excessive inflammatory responses in Parkinson disease. METHODS: We investigated the immune response of primary human monocytes and a microglial cell line to pathologic forms of α-synuclein by assessing cytokine release upon exposure. RESULTS: We show that pathologic α-synuclein (mutations, aggregation) results in a robust inflammatory activation of human monocytes and microglial BV2 cells. The activation is conformation- dependent, with increasing fibrillation and early onset mutations having the strongest effect on immune activation. We also found that activation of immune cells by extracellular α-synuclein is potentiated by extracellular vesicles, possibly by facilitating the uptake of α-synuclein. Blood extracellular vesicles from Parkinson disease patients induce a stronger activation of monocytes than blood extracellular vesicles from healthy controls. Most importantly, monocytes from Parkinson disease patients are dysregulated and hyperactive in response to stimulation with pathologic α-synuclein. Furthermore, we demonstrate that α-synuclein pathology in the CNS is sufficient to induce the monocyte dysregulation in the periphery of a mouse model. INTERPRETATION: Taken together, our data suggest that α-synuclein pathology and dysregulation of monocytes in Parkinson disease can act together to induce excessive inflammatory responses to α-synuclein. ANN NEUROL 2019;86:593-606.


Asunto(s)
Citocinas/metabolismo , Inflamación/metabolismo , Enfermedad de Parkinson/inmunología , alfa-Sinucleína/efectos adversos , Animales , Células Cultivadas , Vesículas Extracelulares/inmunología , Humanos , Inflamación/complicaciones , Ratones , Ratones Transgénicos , Microglía/metabolismo , Monocitos/metabolismo , Mutación , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/genética
3.
Int J Mol Sci ; 21(6)2020 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-32210174

RESUMEN

Oligomerization and/or aggregation of α-synuclein (α-Syn) triggers α-synucleinopathies such as Parkinson's disease and dementia with Lewy bodies. It is known that α-Syn can spread in the brain like prions; however, the mechanism remains unclear. We demonstrated that fatty acid binding protein 3 (FABP3) promotes propagation of α-Syn in mouse brain. Animals were injected with mouse or human α-Syn pre-formed fibrils (PFF) into the bilateral substantia nigra pars compacta (SNpc). Two weeks after injection of mouse α-Syn PFF, wild-type (WT) mice exhibited motor and cognitive deficits, whereas FABP3 knock-out (Fabp3-/-) mice did not. The number of phosphorylated α-Syn (Ser-129)-positive cells was significantly decreased in Fabp3-/- mouse brain compared to that in WT mice. The SNpc was unilaterally infected with AAV-GFP/FABP3 in Fabp3-/- mice to confirm the involvement of FABP3 in the development of α-Syn PFF toxicity. The number of tyrosine hydroxylase (TH)- and phosphorylated α-Syn (Ser-129)-positive cells following α-Syn PFF injection significantly decreased in Fabp3-/- mice and markedly increased by AAV-GFP/FABP3 infection. Finally, we confirmed that the novel FABP3 inhibitor MF1 significantly antagonized motor and cognitive impairments by preventing α-Syn spreading following α-Syn PFF injection. Overall, FABP3 enhances α-Syn spreading in the brain following α-Syn PFF injection, and the FABP3 ligand MF1 represents an attractive therapeutic candidate for α-synucleinopathy.


Asunto(s)
Encéfalo/metabolismo , Proteína 3 de Unión a Ácidos Grasos/metabolismo , alfa-Sinucleína/metabolismo , Animales , Encéfalo/patología , Cognición , Modelos Animales de Enfermedad , Proteína 3 de Unión a Ácidos Grasos/antagonistas & inhibidores , Proteína 3 de Unión a Ácidos Grasos/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fosforilación , Sinucleinopatías/etiología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología , Sinucleinopatías/psicología , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/efectos adversos
4.
Eur J Neurosci ; 49(3): 339-363, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30269383

RESUMEN

Our understanding of the mechanisms underlying Parkinson's disease, the once archetypical nongenetic neurogenerative disorder, has dramatically increased with the identification of α-synuclein and LRRK2 pathogenic mutations. While α-synuclein protein composes the aggregates that can spread through much of the brain in disease, LRRK2 encodes a multidomain dual-enzyme distinct from any other protein linked to neurodegeneration. In this review, we discuss emergent datasets from multiple model systems that suggest these unlikely partners do interact in important ways in disease, both within cells that express both LRRK2 and α-synuclein as well as through more indirect pathways that might involve neuroinflammation. Although the link between LRRK2 and disease can be understood in part through LRRK2 kinase activity (phosphotransferase activity), α-synuclein toxicity is multilayered and plausibly interacts with LRRK2 kinase activity in several ways. We discuss common protein interactors like 14-3-3s that may regulate α-synuclein and LRRK2 in disease. Finally, we examine cellular pathways and outcomes common to both mutant α-synuclein expression and LRRK2 activity and points of intersection. Understanding the interplay between these two unlikely partners in disease may provide new therapeutic avenues for PD.


Asunto(s)
Encéfalo/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/efectos adversos , Degeneración Nerviosa/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/efectos adversos , Animales , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Mutación , Neuronas/metabolismo , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
5.
Acta Neuropathol ; 135(1): 49-63, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28849371

RESUMEN

In multiple system atrophy (MSA), progressive neurodegeneration results from the protein α-synuclein misfolding into a self-templating prion conformation that spreads throughout the brain. MSA prions are transmissible to transgenic (Tg) mice expressing mutated human α-synuclein (TgM83+/-), inducing neurological disease following intracranial inoculation with brain homogenate from deceased patient samples. Noting the similarities between α-synuclein prions and PrP scrapie (PrPSc) prions responsible for Creutzfeldt-Jakob disease (CJD), we investigated MSA transmission under conditions known to result in PrPSc transmission. When peripherally exposed to MSA via the peritoneal cavity, hind leg muscle, and tongue, TgM83+/- mice developed neurological signs accompanied by α-synuclein prions in the brain. Iatrogenic CJD, resulting from PrPSc prion adherence to surgical steel instruments, has been investigated by incubating steel sutures in contaminated brain homogenate before implantation into mouse brain. Mice studied using this model for MSA developed disease, whereas wire incubated in control homogenate had no effect on the animals. Notably, formalin fixation did not inactivate α-synuclein prions. Formalin-fixed MSA patient samples also transmitted disease to TgM83+/- mice, even after incubating in fixative for 244 months. Finally, at least 10% sarkosyl was found to be the concentration necessary to partially inactivate MSA prions. These results demonstrate the robustness of α-synuclein prions to denaturation. Moreover, they establish the parallel characteristics between PrPSc and α-synuclein prions, arguing that clinicians should exercise caution when working with materials that might contain α-synuclein prions to prevent disease.


Asunto(s)
Atrofia de Múltiples Sistemas/metabolismo , Priones/metabolismo , Animales , Transporte Biológico , Encéfalo/metabolismo , Encéfalo/patología , Detergentes/farmacología , Modelos Animales de Enfermedad , Fijadores , Formaldehído , Células HEK293 , Humanos , Ratones Transgénicos , Atrofia de Múltiples Sistemas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación , Priones/administración & dosificación , Agregado de Proteínas , Estabilidad Proteica/efectos de los fármacos , Sarcosina/análogos & derivados , Sarcosina/farmacología , Acero Inoxidable , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/efectos adversos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
7.
Neurobiol Aging ; 106: 12-25, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34225000

RESUMEN

Synucleinopathies are neurodegenerative disorders involving pathological alpha-synuclein (αSyn) protein, including dementia with Lewy bodies, multiple system atrophy and Parkinson's disease (PD). Current in vivo models of synucleinopathy include transgenic mice overexpressing αSyn variants and methods based on administration of aggregated, exogenous αSyn. Combining these techniques offers the ability to study consequences of introducing pathological αSyn into primed neuronal environments likely to develop synucleinopathy. Herein, we characterize the impacts pre-formed fibrils (PFFs) of recombinant, human αSyn have in mice overexpressing human A30P αSyn, a mutation associated with autosomal dominant PD. A30P mouse brain contains detergent insoluble αSyn biochemically similar to PD brain, and these mice develop Lewy-like synucleinopathy with age. Administration of PFFs in A30P mice resulted in regionally-specific accumulations of phosphorylated synuclein, microglial induction and a motor phenotype that differed from PFF-induced effects in wildtype mice. Surprisingly, PFF-induced losses of tyrosine hydroxylase were similar in A30P and wildtype mice. Thus, the PFF-A30P model recapitulates key aspects of synucleinopathy with induction of microglia, creating an appropriate system for evaluating neurodegenerative therapeutics.


Asunto(s)
Microglía/patología , Sinucleinopatías/etiología , Sinucleinopatías/patología , alfa-Sinucleína/efectos adversos , Animales , Modelos Animales de Enfermedad , Expresión Génica , Ratones Transgénicos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sinucleinopatías/genética , alfa-Sinucleína/administración & dosificación , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
8.
Biochemistry ; 49(5): 862-71, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20041693

RESUMEN

Three familial variants of the presynaptic protein alpha-synuclein (alphaS), A30P, E46K, and A53T, correlate with rare inherited Parkinson's disease (PD), while wild-type alphaS is implicated in sporadic PD. The classic manifestation of both familiar and sporadic PD is the formation of fibrillar structures of alphaS which accumulate as the main component in intraneuronal Lewy bodies. At presynaptic termini, the partitioning of alphaS between disordered cytosolic and membrane-bound states likely mediates its proposed role in regulation of reserve pools of synaptic vesicles. Previously, we reported on multiple distinct phospholipid binding modes of alphaS with slow binding kinetics. Here, we report the phospholipid binding properties of the disease variants, viewed by solution NMR in a residue-specific manner. Our results agree qualitatively with previous biophysical studies citing overall decreased lipid affinity for the A30P mutation, comparable affinity for A53T, and an increased level of binding of E46K, relative to wild-type alphaS. Additionally, our NMR results describe the distribution of lipid-bound states for alphaS: the population of the SL1 binding mode (residues 3-25 bound as a helix) is augmented by each of the disease variants, relative to wild-type alphaS. We propose that the SL1 binding mode, which anchors the N-terminus of alphaS in the lipoprotein complex while the hydrophobic NAC region remains dynamically disordered, is prone to intermolecular interactions which progress toward disease-associated oligomers and fibrils. The elevation of the SL1 binding mode, unchecked by a proportionate population of binding modes incorporating the full N-terminal domain, may well account for the increased toxicity of the A30P, E46K, and A53T disease variants of alphaS.


Asunto(s)
Variación Genética , Espectroscopía de Resonancia Magnética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Fosfolípidos/genética , Fosfolípidos/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Alanina/genética , Ácido Glutámico/genética , Humanos , Lisina/genética , Mutagénesis Sitio-Dirigida , Enfermedad de Parkinson/patología , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Prolina/genética , Unión Proteica/genética , Estructura Secundaria de Proteína/genética , Estructura Terciaria de Proteína/genética , Soluciones , Treonina/genética , alfa-Sinucleína/efectos adversos
9.
J Neurochem ; 110(3): 1005-13, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19493164

RESUMEN

Alpha synuclein can be phosphorylated at serine129 (P-S129), and the presence of highly phosphorylated alpha-synuclein in Lewy bodies suggests changes to its phosphorylation status has an important pathological role. We demonstrate that the kinase(s) responsible for alpha-synuclein S129 phosphorylation is constitutively active in SH-SY5Y cells and involves casein kinase 2 activity. Increased oxidative stress or proteasomal inhibition caused significant elevation of P-S129 alpha-synuclein levels. Under these conditions, similar increases in P-S129 alpha-synuclein were found in both sodium dodecyl sulphate lysates and Triton extracts indicating the phosphorylated protein was soluble and did not lead to aggregation. The rate of S129 phosphorylation was increased in response to proteasomal inhibition indicating a higher activity of the relevant kinase. Cells expressing the phosphorylation mimic, S129D alpha-synuclein increased cell death and enhanced sensitivity to epoxomycin exposure. Proteasomal inhibition markedly decreased S129D alpha-synuclein turnover suggesting proteasomal inhibition leads to the accumulation of P-S129 alpha-synuclein through an increase in the kinase activity and a decrease in protein turnover resulting in increased cell death. We conclude that S129 phosphorylation is toxic to dopaminergic cells and both the levels of S129 phosphorylated protein and its toxicity are increased with proteasomal inhibition emphasising the interdependence of these pathways in Parkinson's disease pathogenesis.


Asunto(s)
Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , alfa-Sinucleína/metabolismo , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , Enfermedad de Parkinson/metabolismo , Fosforilación/fisiología , Regulación hacia Arriba , alfa-Sinucleína/efectos adversos , alfa-Sinucleína/biosíntesis
10.
Alzheimer Dis Assoc Disord ; 23(4): 365-70, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19935145

RESUMEN

Parkinson disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB) share alpha-synuclein immunoreactivity. These "synucleinopathies" have overlapping signs and symptoms, but less is known about similarities and differences in their cognitive and neuropsychiatric profiles. We compared the cognitive and neuropsychiatric profiles of individuals with PD, MSA, and DLB. Overall, the DLB group showed the most cognitive impairment, the MSA group demonstrated milder impairment, and the PD group was the least cognitively impaired. The DLB and MSA groups showed worse executive function and visuospatial skills than PD, whereas DLB showed impaired memory relative to both PD and MSA. On the neuropsychiatric screening, all groups endorsed depression and anxiety; the DLB group alone endorsed delusions and disinhibition. Consistent with their greater level of cognitive and neuropsychiatric impairment, the DLB group showed the greatest amount of functional impairment on a measure of instrumental activities of daily living (Functional Activities Questionnaire). We found that MSA subjects had cognitive difficulties that fell between the mild deficits of the PD group and the more severe deficits of the DLB group. PD, MSA, and DLB groups have similar neuropsychiatric profiles of increased depression and anxiety. Similar underlying alpha-synuclein pathology may contribute to these shared features.


Asunto(s)
Trastornos del Conocimiento/diagnóstico , Enfermedad por Cuerpos de Lewy/diagnóstico , Atrofia de Múltiples Sistemas/diagnóstico , Enfermedad de Parkinson/diagnóstico , alfa-Sinucleína/fisiología , Anciano , Trastornos del Conocimiento/clasificación , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/psicología , Estudios de Cohortes , Femenino , Humanos , Enfermedad por Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/psicología , Masculino , Memoria/fisiología , Persona de Mediana Edad , Atrofia de Múltiples Sistemas/patología , Atrofia de Múltiples Sistemas/psicología , Pruebas Neuropsicológicas , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/psicología , alfa-Sinucleína/efectos adversos
11.
Sheng Li Xue Bao ; 61(4): 324-30, 2009 Aug 25.
Artículo en Zh | MEDLINE | ID: mdl-19701583

RESUMEN

α-synuclein (α-SN) has been postulated to play a pivotal role in the pathogenesis of Parkinson's disease (PD). However, the physiological functions of α-SN and the molecular and cellular mechanisms underlying neuronal loss remain unclear. Recent studies suggest that α-SN plays dual roles of neuroprotection and neurotoxicity depending on its concentration or level of expression. In the present study, we explored the potential mechanisms for α-SN to regulate neuronal survival. α-SN at different concentrations (0.1 to 40 mumol/L) with or without 50 mumol/L 6-hydroxydopamine (6-OHDA) were added into the culture medium of the SH-SY5Y dopaminergic neural cells. The cell viability was measured on post-treatment day 1, 2 and 3. The activity of proteasome inhibited by α-SN was tested by a proteasome activity assay system after 2 h of α-SN treatment. According to the activity of proteasome inhibited by α-SN, the correlative dose of proteasome inhibitor--lactacystin (10 nmol/L to 5 mumol/L) with or without 50 mumol/L 6-OHDA were used and the cell viability was assayed on post-treatment day 1, 2 and 3. The results showed that α-SN played dual roles of neuroprotection and neurotoxicity depending on its concentration. At low concentration (0.1 to 5 mumol/L), α-SN promoted the proliferation and protected neurons against the neurotoxicity of 6-OHDA; in contrast, at high concentration (10 to 40 mumol/L), α-SN possessed cytotoxicity. The results of lactacystin treatment implied that the dual roles of α-SN were related to the moderate and strong inhibition of proteasome activity. The MEK1/2 specific inhibitor PD98059 completely blocked the protection of both α-SN and lactacystin, suggesting that MAPK pathway might be involved in the neuroprotection of α-SN.


Asunto(s)
Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxidopamina/efectos adversos , alfa-Sinucleína/farmacología , Acetilcisteína/análogos & derivados , Apoptosis , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Neurotoxinas/efectos adversos , Enfermedad de Parkinson , alfa-Sinucleína/efectos adversos
12.
Cell Death Dis ; 10(2): 80, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30692508

RESUMEN

Progressive accumulation of α-synuclein (α-syn) and exposure to environmental toxins are risk factors that may both concur to Parkinson's disease (PD) pathogenesis. Electrophysiological recordings of field postsynaptic potentials (fEPSPs) and Ca2+ measures in striatal brain slices and differentiated SH-SY5Y cells showed that co-application of α-syn and the neurotoxic pesticide rotenone (Rot) induced Ca2+ dysregulation and alteration of both synaptic transmission and cell function. Interestingly, the presence of the mitochondrial NCX inhibitor CGP-37157 prevented these alterations. The specific involvement of the mitochondrial NCX was confirmed by the inability of the plasma membrane inhibitor SN-6 to counteract such phenomenon. Of note, using a siRNA approach, we found that NCX1 was the isoform specifically involved. These findings suggested that NCX1, operating on the mitochondrial membrane, may have a critical role in the maintenance of ionic Ca2+ homeostasis in PD and that its inhibition most likely exerts a protective effect in the toxicity induced by α-syn and Rot.


Asunto(s)
Cuerpo Estriado/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Rotenona/efectos adversos , Intercambiador de Sodio-Calcio/metabolismo , alfa-Sinucleína/efectos adversos , Animales , Modelos Animales de Enfermedad , Humanos , Enfermedad de Parkinson , Ratas , Ratas Wistar
13.
Neurobiol Aging ; 65: 60-68, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29407467

RESUMEN

Parkinson's disease (PD) is no longer primarily classified as a motor disorder due to increasing recognition of the impact on patients of several nonmotor PD symptoms, including cognitive dysfunction. These nonmotor symptoms are highly prevalent and greatly affect the quality of life of patients with PD, and so, therapeutic interventions to alleviate these symptoms are urgently needed. The aim of this study was to investigate the potential neuroprotective effects of voluntary running on cognitive dysfunction in an adeno-associated virus-α-synuclein rat model of PD. Bilateral intranigral administration of adeno-associated virus-α-synuclein was found to induce motor dysfunction and a significant loss of nigral dopaminergic neurons, neither of which were rescued by voluntary running. Overexpression of α-synuclein also resulted in significant impairment on hippocampal neurogenesis-dependent pattern separation, a cognitive task; this was rescued by voluntary running. This was substantiated by an effect of running on neurogenesis levels in the dorsal dentate gyrus, suggesting that the functional effects of running on pattern separation were mediated via increased neurogenesis.


Asunto(s)
Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Enfermedad de Parkinson/complicaciones , Carrera/fisiología , alfa-Sinucleína/efectos adversos , Animales , Disfunción Cognitiva/patología , Dependovirus , Modelos Animales de Enfermedad , Hipocampo/patología , Masculino , Neurogénesis , Ratas Sprague-Dawley
14.
Autophagy ; 14(11): 1898-1910, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29989488

RESUMEN

The autophagy-lysosome pathway plays a fundamental role in the clearance of aggregated proteins and protection against cellular stress and neurodegenerative conditions. Alterations in autophagy processes, including macroautophagy and chaperone-mediated autophagy (CMA), have been described in Parkinson disease (PD). CMA is a selective autophagic process that depends on LAMP2A (lysosomal-associated membrane protein 2A), a mammal and bird-specific membrane glycoprotein that translocates cytosolic proteins containing a KFERQ-like peptide motif across the lysosomal membrane. Drosophila reportedly lack CMA and use endosomal microautophagy (eMI) as an alternative selective autophagic process. Here we report that neuronal expression of human LAMP2A protected Drosophila against starvation and oxidative stress, and delayed locomotor decline in aging flies without extending their lifespan. LAMP2A also prevented the progressive locomotor and oxidative defects induced by neuronal expression of PD-associated human SNCA (synuclein alpha) with alanine-to-proline mutation at position 30 (SNCAA30P). Using KFERQ-tagged fluorescent biosensors, we observed that LAMP2A expression stimulated selective autophagy in the adult brain and not in the larval fat body, but did not increase this process under starvation conditions. Noteworthy, we found that neurally expressed LAMP2A markedly upregulated levels of Drosophila Atg5, a key macroautophagy initiation protein, and that it increased the density of Atg8a/LC3-positive puncta, which reflects the formation of autophagosomes. Furthermore, LAMP2A efficiently prevented accumulation of the autophagy defect marker Ref(2)P/p62 in the adult brain under acute oxidative stress. These results indicate that LAMP2A can potentiate autophagic flux in the Drosophila brain, leading to enhanced stress resistance and neuroprotection. ABBREVIATIONS: Act5C: actin 5C; a.E.: after eclosion; Atg5: autophagy-related 5; Atg8a/LC3: autophagy-related 8a; CMA: chaperone-mediated autophagy; DHE: dihydroethidium; elav: embryonic lethal abnormal vision; eMI: endosomal microautophagy; ESCRT: endosomal sorting complexes required for transport; GABARAP: GABA typeA receptor-associated protein; Hsc70-4: heat shock protein cognate 4; HSPA8/Hsc70: heat shock protein family A (Hsp70) member 8; LAMP2: lysosomal associated membrane protein 2; MDA: malondialdehyde; PA-mCherry: photoactivable mCherry; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PD: Parkinson disease; Ref(2)P/p62: refractory to sigma P; ROS: reactive oxygen species; RpL32/rp49: ribosomal protein L32; RT-PCR: reverse transcription polymerase chain reaction; SING: startle-induced negative geotaxis; SNCA/α-synuclein: synuclein alpha; SQSTM1/p62: sequestosome 1; TBS: Tris-buffered saline; UAS: upstream activating sequence.


Asunto(s)
Autofagia/genética , Drosophila , Proteína 2 de la Membrana Asociada a los Lisosomas/fisiología , Neuroprotección/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/prevención & control , alfa-Sinucleína/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Drosophila/genética , Drosophila/metabolismo , Humanos , Locomoción/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Estrés Oxidativo/genética , Enfermedad de Parkinson/genética , Fenotipo , Transducción de Señal/genética , alfa-Sinucleína/efectos adversos
16.
PLoS One ; 10(2): e0116841, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25658425

RESUMEN

The protein α-synuclein (α-Syn) has a central role in the pathogenesis of Parkinson's disease (PD) and immunotherapeutic approaches targeting this molecule have shown promising results. In this study, novel antibodies were generated against specific peptides from full length human α-Syn and evaluated for effectiveness in ameliorating α-Syn-induced cell death and behavioral deficits in an AAV-α-Syn expressing rat model of PD. Fisher 344 rats were injected with rAAV vector into the right substantia nigra (SN), while control rats received an AAV vector expressing green fluorescent protein (GFP). Beginning one week after injection of the AAV-α-Syn vectors, rats were treated intraperitoneally with either control IgG or antibodies against the N-terminal (AB1), or central region (AB2) of α-Syn. An unbiased stereological estimation of TH+, NeuN+, and OX6 (MHC-II) immunostaining revealed that the α-Syn peptide antibodies (AB1 and AB2) significantly inhibited α-Syn-induced dopaminergic cell (DA) and NeuN+ cell loss (one-way ANOVA (F (3, 30) = 5.8, p = 0.002 and (F (3, 29) = 7.92, p = 0.002 respectively), as well as decreasing the number of activated microglia in the ipsilateral SN (one-way ANOVA F = 14.09; p = 0.0003). Antibody treated animals also had lower levels of α-Syn in the ipsilateral SN (one-way ANOVA F (7, 37) = 9.786; p = 0.0001) and demonstrated a partial intermediate improvement of the behavioral deficits. Our data suggest that, in particular, an α-Syn peptide antibody against the N-terminal region of the protein can protect against DA neuron loss and, to some extent behavioral deficits. As such, these results may be a potential therapeutic strategy for halting the progression of PD.


Asunto(s)
Anticuerpos/inmunología , Muerte Celular/fisiología , Neuronas Dopaminérgicas/fisiología , Inmunoterapia/métodos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/inmunología , alfa-Sinucleína/inmunología , Análisis de Varianza , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Conducta Animal/fisiología , Muerte Celular/efectos de los fármacos , Dependovirus , Neuronas Dopaminérgicas/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Inyecciones Intraperitoneales , Ratas , Sustancia Negra/metabolismo , alfa-Sinucleína/efectos adversos
18.
Mol Neurobiol ; 47(2): 525-36, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22923367

RESUMEN

The toxicity of α-synuclein in the neuropathology of Parkinson's disease which includes its hallmark aggregation has been studied scrupulously in the last decade. Although little is known regarding the normal functions of α-synuclein, its association with membrane phospholipids suggests its potential role in signaling pathways. Following extensive evidences for its nuclear localization, we and others recently demonstrated DNA binding activity of α-synuclein that modulates its conformation as well as aggregation properties. Furthermore, we also underscored the similarities among various amyloidogenic proteins involved in neurodegenerative diseases including amyloid beta peptides and tau. Our more recent studies show that α-synuclein is glycated and glycosylated both in vitro and in neurons, significantly affecting its folding, oligomeric, and DNA binding properties. Glycated α-synuclein causes increased genome damage both via its direct interaction with DNA and by increased generation of reactive oxygen species as glycation byproduct. In this review, we discuss the mechanisms of glycation and other posttranslational modifications of α-synuclein, including phosphorylation and nitration, and their role in neuronal death in Parkinson's disease.


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/fisiología , Animales , Muerte Celular/fisiología , Glicosilación , Humanos , Enfermedad de Parkinson/patología , Fosforilación/fisiología , Unión Proteica/fisiología , Pliegue de Proteína , alfa-Sinucleína/efectos adversos , alfa-Sinucleína/toxicidad
19.
Mol Neurobiol ; 47(2): 495-508, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22622968

RESUMEN

The histopathological hallmark of Parkinson's disease (PD) is the presence of fibrillar aggregates referred to as Lewy bodies (LBs), in which α-synuclein is a major constituent. Pale bodies, the precursors of LBs, may serve the material for that LBs continue to expand. LBs consist of a heterogeneous mixture of more than 90 molecules, including PD-linked gene products (α-synuclein, DJ-1, LRRK2, parkin, and PINK-1), mitochondria-related proteins, and molecules implicated in the ubiquitin-proteasome system, autophagy, and aggresome formation. LB formation has been considered to be a marker for neuronal degeneration because neuronal loss is found in the predilection sites for LBs. However, recent studies have indicated that nonfibrillar α-synuclein is cytotoxic and that fibrillar aggregates of α-synuclein (LBs and pale bodies) may represent a cytoprotective mechanism in PD.


Asunto(s)
Cuerpos de Lewy/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Animales , Humanos , Cuerpos de Lewy/fisiología , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/prevención & control , Enfermedad de Parkinson/prevención & control , alfa-Sinucleína/efectos adversos , alfa-Sinucleína/toxicidad
20.
Mol Neurobiol ; 47(2): 598-612, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22933040

RESUMEN

Epidemiological, population-based case-control, and experimental studies at the molecular, cellular, and organism levels revealed that exposure to various environmental agents, including a number of structurally different agrochemicals, may contribute to the pathogenesis of Parkinson's disease (PD) and several other neurodegenerative disorders. The role of genetic predisposition in PD has also been increasingly acknowledged, driven by the identification of a number of disease-related genes [e.g., α-synuclein, parkin, DJ-1, ubiquitin C-terminal hydrolase isozyme L1 (UCH-L1), and nuclear receptor-related factor 1]. Therefore, the etiology of this multifactorial disease is likely to involve both genetic and environmental factors. Various neurotoxicants, including agrochemicals, have been shown to elevate the levels of α-synuclein expression in neurons and to promote aggregation of this protein in vivo. Many agrochemicals physically interact with α-synuclein and accelerate the fibrillation and aggregation rates of this protein in vitro. This review analyzes some of the aspects linking α-synuclein to PD, provides brief structural and functional descriptions of this important protein, and represents some data connecting exposure to agrochemicals with α-synuclein aggregation and PD pathogenesis.


Asunto(s)
Agroquímicos/metabolismo , Contaminantes Ambientales/metabolismo , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/metabolismo , alfa-Sinucleína/metabolismo , Agroquímicos/química , Agroquímicos/toxicidad , Animales , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/química , Contaminantes Ambientales/toxicidad , Humanos , Enfermedad de Parkinson Secundaria/etiología , alfa-Sinucleína/efectos adversos , alfa-Sinucleína/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA