Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Brain ; 142(9): 2845-2859, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31312839

RESUMEN

Mutations in lysosomal genes increase the risk of neurodegenerative diseases, as is the case for Parkinson's disease. Here, we found that pathogenic and protective mutations in arylsulfatase A (ARSA), a gene responsible for metachromatic leukodystrophy, a lysosomal storage disorder, are linked to Parkinson's disease. Plasma ARSA protein levels were changed in Parkinson's disease patients. ARSA deficiency caused increases in α-synuclein aggregation and secretion, and increases in α-synuclein propagation in cells and nematodes. Despite being a lysosomal protein, ARSA directly interacts with α-synuclein in the cytosol. The interaction was more extensive with protective ARSA variant and less with pathogenic ARSA variant than wild-type. ARSA inhibited the in vitro fibrillation of α-synuclein in a dose-dependent manner. Ectopic expression of ARSA reversed the α-synuclein phenotypes in both cell and fly models of synucleinopathy, the effects correlating with the extent of the physical interaction between these molecules. Collectively, these results suggest that ARSA is a genetic modifier of Parkinson's disease pathogenesis, acting as a molecular chaperone for α-synuclein.


Asunto(s)
Cerebrósido Sulfatasa/fisiología , Chaperonas Moleculares/metabolismo , Mutación Missense , Enfermedad de Parkinson/metabolismo , Mutación Puntual , alfa-Sinucleína/metabolismo , Adulto , Anciano , Animales , Animales Modificados Genéticamente , Encéfalo/enzimología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Cultivadas , Cerebrósido Sulfatasa/sangre , Cerebrósido Sulfatasa/genética , Demencia/sangre , Demencia/etiología , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Técnicas de Inactivación de Genes , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/psicología , Linaje , Agregación Patológica de Proteínas/genética , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/metabolismo
2.
J Neurosci ; 32(39): 13454-69, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23015436

RESUMEN

Abnormal deposition and intercellular propagation of α-synuclein plays a central role in the pathogenesis of disorders such as Parkinson's Disease (PD) and dementia with Lewy bodies (DLB). Previous studies demonstrated that immunization against α-synuclein resulted in reduced α-synuclein accumulation and synaptic loss in a transgenic (tg) mouse model, highlighting the potential for immunotherapy. However, the mechanism by which immunization prevents synucleinopathy-associated deficits remains unknown. Here, we show that antibodies against α-synuclein specifically target and aid in clearance of extracellular α-synuclein proteins by microglia, thereby preventing their actions on neighboring cells. Antibody-assisted clearance occurs mainly in microglia through the Fcγ receptor, and not in neuronal cells or astrocytes. Stereotaxic administration of antibody into the brains of α-synuclein tg mice prevented neuron-to-astroglia transmission of α-synuclein and led to increased localization of α-synuclein and the antibody in microglia. Furthermore, passive immunization with α-synuclein antibody reduced neuronal and glial accumulation of α-synuclein and ameliorated neurodegeneration and behavioral deficits associated with α-synuclein overexpression. These findings provide an underlying mechanistic basis for immunotherapy for PD/DLB and suggest extracellular forms of α-synuclein as potential therapeutic targets.


Asunto(s)
Anticuerpos/farmacología , Neuroglía/efectos de los fármacos , alfa-Sinucleína/inmunología , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Amiloide/ultraestructura , Análisis de Varianza , Animales , Anticuerpos/metabolismo , Antígenos CD/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Catepsina D/metabolismo , Caveolina 1/metabolismo , Comunicación Celular/fisiología , Línea Celular , Cromatografía en Gel , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/inmunología , Espacio Extracelular/metabolismo , Humanos , Inmunización Pasiva , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/inmunología , Enfermedad por Cuerpos de Lewy/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/genética , Degeneración Nerviosa/inmunología , Neuroglía/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transmisión Sináptica/fisiología , alfa-Sinucleína/genética
3.
Biol Chem ; 394(7): 807-18, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23435096

RESUMEN

Mutations in the gene encoding glucocerebrosidase (GBA1) cause Gaucher disease (GD), a lysosomal storage disease with recessive inheritance. Glucocerebrosidase (GCase) is a lysosomal lipid hydrolase that digests glycolipid substrates, such as glucosylceramide and glucosylsphingosine. GBA1 mutations have been implicated in Lewy body diseases (LBDs), such as Parkinson's disease and dementia with Lewy bodies. Parkinsonism occurs more frequently in certain types of GD, and GBA1 mutation carriers are more likely to have LBDs than non-carriers. Furthermore, GCase is often found in Lewy bodies, which are composed of α-synuclein fibrils as well as a variety of proteins and vesicles. In this review, we discuss potential mechanisms of action of GBA1 mutations in LBDs with particular emphasis on α-synuclein aggregation by reviewing the current literature on the role of GCase in lysosomal functions and glycolipid metabolism.


Asunto(s)
Enfermedad de Gaucher/enzimología , Glucosilceramidasa/metabolismo , Enfermedad por Cuerpos de Lewy/enzimología , alfa-Sinucleína/metabolismo , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/genética , Glucolípidos/metabolismo , Humanos , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/metabolismo , Lisosomas/metabolismo , Mutación
4.
J Mol Biol ; 435(12): 168114, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37085010

RESUMEN

Neurodegenerative diseases, such as Alzheimer's and Parkinson's, are characterized by several pathological features, including selective neuronal loss, aggregation of specific proteins, and chronic inflammation. Aging is the most critical risk factor of these disorders. However, the mechanism by which aging contributes to the pathogenesis of neurodegenerative diseases is not clearly understood. Cellular senescence is a cell state or fate in response to stimuli. It is typically associated with a series of changes in cellular phenotypes such as abnormal cellular metabolism and proteostasis, reactive oxygen species (ROS) production, and increased secretion of certain molecules via senescence-associated secretory phenotype (SASP). In this review, we discuss how cellular senescence contributes to brain aging and neurodegenerative diseases, and the relationship between protein aggregation and cellular senescence. Finally, we discuss the potential of senescence modifiers and senolytics in the treatment of neurodegenerative diseases.


Asunto(s)
Senescencia Celular , Enfermedades Neurodegenerativas , Senoterapéuticos , Humanos , Encéfalo/metabolismo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/fisiología , Enfermedades Neurodegenerativas/patología , Agregado de Proteínas , Senoterapéuticos/farmacología , Senoterapéuticos/uso terapéutico
5.
Proc Natl Acad Sci U S A ; 106(31): 13010-5, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19651612

RESUMEN

Neuronal accumulation of alpha-synuclein and Lewy body formation are characteristic to many neurodegenerative diseases, including Parkinson's disease (PD). This Lewy pathology appears to spread throughout the brain as the disease progresses. Furthermore, recent studies showed the occurrence of Lewy pathology in neurons grafted into the brains of PD patients, suggesting the spread of pathology from the host tissues to the grafts. The mechanism underlying this propagation is unknown. Here, we show that alpha-synuclein is transmitted via endocytosis to neighboring neurons and neuronal precursor cells, forming Lewy-like inclusions. Moreover, alpha-synuclein was transmitted from the affected neurons to engrafted neuronal precursor cells in a transgenic model of PD-like pathology. Failure of the protein quality control systems, especially lysosomes, promoted the accumulation of transmitted alpha-synuclein and inclusion formation. Cells exposed to neuron-derived alpha-synuclein showed signs of apoptosis, such as nuclear fragmentation and caspase 3 activation, both in vitro and in vivo. These findings demonstrate the cell-to-cell transmission of alpha-synuclein aggregates and provide critical insights into the mechanism of pathological progression in PD and other proteinopathies.


Asunto(s)
Apoptosis , Cuerpos de Lewy/fisiología , Neuronas/fisiología , Transmisión Sináptica , alfa-Sinucleína/metabolismo , Animales , Transporte Biológico , Línea Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Trasplante de Células Madre , alfa-Sinucleína/toxicidad
6.
Exp Mol Med ; 54(2): 115-128, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35136202

RESUMEN

α-Synuclein is a crucial element in the pathogenesis of Parkinson's disease (PD) and related neurological diseases. Although numerous studies have presented potential mechanisms underlying its pathogenesis, the understanding of α-synuclein-mediated neurodegeneration remains far from complete. Here, we show that overexpression of α-synuclein leads to impaired DNA repair and cellular senescence. Transcriptome analysis showed that α-synuclein overexpression led to cellular senescence with activation of the p53 pathway and DNA damage responses (DDRs). Chromatin immunoprecipitation analyses using p53 and γH2AX, chromosomal markers of DNA damage, revealed that these proteins bind to promoters and regulate the expression of DDR and cellular senescence genes. Cellular marker analyses confirmed cellular senescence and the accumulation of DNA double-strand breaks. The non-homologous end joining (NHEJ) DNA repair pathway was activated in α-synuclein-overexpressing cells. However, the expression of MRE11, a key component of the DSB repair system, was reduced, suggesting that the repair pathway induction was incomplete. Neuropathological examination of α-synuclein transgenic mice showed increased levels of phospho-α-synuclein and DNA double-strand breaks, as well as markers of cellular senescence, at an early, presymptomatic stage. These results suggest that the accumulation of DNA double-strand breaks (DSBs) and cellular senescence are intermediaries of α-synuclein-induced pathogenesis in PD.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , ADN/genética , Daño del ADN , Reparación del ADN , Ratones , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
7.
Exp Mol Med ; 54(12): 2148-2161, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36473937

RESUMEN

The clinical progression of neurodegenerative diseases correlates with the spread of proteinopathy in the brain. The current understanding of the mechanism of proteinopathy spread is far from complete. Here, we propose that inflammation is fundamental to proteinopathy spread. A sequence variant of α-synuclein (V40G) was much less capable of fibril formation than wild-type α-synuclein (WT-syn) and, when mixed with WT-syn, interfered with its fibrillation. However, when V40G was injected intracerebrally into mice, it induced aggregate spreading even more effectively than WT-syn. Aggregate spreading was preceded by sustained microgliosis and inflammatory responses, which were more robust with V40G than with WT-syn. Oral administration of an anti-inflammatory agent suppressed aggregate spreading, inflammation, and behavioral deficits in mice. Furthermore, exposure of cells to inflammatory cytokines increased the cell-to-cell propagation of α-synuclein. These results suggest that the inflammatory microenvironment is the major driver of the spread of synucleinopathy in the brain.


Asunto(s)
Enfermedades Neurodegenerativas , Sinucleinopatías , Ratones , Animales , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Inflamación , Modelos Animales de Enfermedad
8.
J Biol Chem ; 285(12): 9262-72, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20071342

RESUMEN

Abnormal neuronal aggregation of alpha-synuclein is implicated in the development of many neurological disorders, including Parkinson disease and dementia with Lewy bodies. Glial cells also show extensive alpha-synuclein pathology and may contribute to disease progression. However, the mechanism that produces the glial alpha-synuclein pathology and the interaction between neurons and glia in the disease-inflicted microenvironment remain unknown. Here, we show that alpha-synuclein proteins released from neuronal cells are taken up by astrocytes through endocytosis and form inclusion bodies. The glial accumulation of alpha-synuclein through the transmission of the neuronal protein was also demonstrated in a transgenic mouse model expressing human alpha-synuclein. Furthermore, astrocytes that were exposed to neuronal alpha-synuclein underwent changes in the gene expression profile reflecting an inflammatory response. Induction of pro-inflammatory cytokines and chemokines correlated with the extent of glial accumulation of alpha-synuclein. Together, these results suggest that astroglial alpha-synuclein pathology is produced by direct transmission of neuronal alpha-synuclein aggregates, causing inflammatory responses. This transmission step is thus an important mediator of pathogenic glial responses and could qualify as a new therapeutic target.


Asunto(s)
Astrocitos/metabolismo , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Humanos , Inflamación , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Enfermedades Neurodegenerativas/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Biomol Ther (Seoul) ; 29(1): 83-89, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-32536617

RESUMEN

Multiple system atrophy (MSA) is a neurodegenerative disease characterized by presence of α-synuclein-positive inclusions in the cytoplasm of oligodendrocytes. These glial cytoplasmic inclusions (GCIs) are considered an integral part of the pathogenesis of MSA, leading to demyelination and neuronal demise. What is most puzzling in the research fields of GCIs is the origin of α-synuclein aggregates in GCIs, since adult oligodendrocytes do not express high levels of α-synuclein. The most recent leading hypothesis is that GCIs form via transfer and accumulation of α-synuclein from neurons to oligodendrocytes. However, studies regarding this subject are limited due to the absence of proper human cell models, to demonstrate the entry and accumulation of neuronal α-synuclein in human oligodendrocytes. Here, we generated mature human oligodendrocytes that can take up neuronderived α-synuclein and form GCI-like inclusions. Mature human oligodendrocytes are derived from neural stem cells via "oligosphere" formation and then into oligodendrocytes, treating the cells with the proper differentiation factors at each step. In the final cell preparations, oligodendrocytes consist of the majority population, while some astrocytes and unidentified stem cell-like cells were present as well. When these cells were exposed to α-synuclein proteins secreted from neuron-like human neuroblastoma cells, oligodendrocytes developed perinuclear inclusion bodies with α-synuclein immunoreactivity, resembling GCIs, while the stem cell-like cells showed α-synuclein-positive, scattered puncta in the cytoplasm. In conclusion, we have established a human oligodendrocyte model for the study of GCI formation, and the characterization and use of this model might pave the way for understanding the pathogenesis of MSA.

10.
J Biol Chem ; 284(44): 30583-93, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19721135

RESUMEN

Glial cell line-derived neurotrophic factor (Gdnf) promotes neurite outgrowth and survival of neuronal cells, but its transcriptional regulation is poorly understood. Here, we sought to investigate the mechanism underlying fibroblast growth factor-2 (FGF2) induction of Gdnf expression in astrocytes. We found that FGF2 stimulation of rat astrocytes induced expression of Egr-1 at a high level. Sequence analysis of the rat Gdnf gene identified three overlapping Egr-1-binding sites between positions -185 and -163 of the rat Gdnf promoter. Transfection studies using a series of deleted Gdnf promoters revealed that these Egr-1-binding sites are required for maximal activation of the Gdnf promoter by FGF2. Chromatin immunoprecipitation analysis indicated that Egr-1 binds to the Gdnf promoter. Furthermore, the induction of Gdnf expression by FGF2 is strongly attenuated both in C6 glioma cells stably expressing Egr-1-specific small interfering RNA and in primary cultured astrocytes from the Egr-1 knock-out mouse. Additionally, we found that stimulation of the ERK and JNK pathways by FGF2 is functionally linked to Gdnf expression through the induction of Egr-1. These data demonstrate that FGF2-induced Gdnf expression is mediated by the induction of Egr-1 through activation of the ERK and JNK/Elk-1 signaling pathways.


Asunto(s)
Astrocitos/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Activación Transcripcional , Animales , Sitios de Unión , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Proteína Elk-1 con Dominio ets/metabolismo
11.
J Neurochem ; 113(5): 1263-74, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20345754

RESUMEN

Parkinson's disease is characterized by deposition of misfolded/aggregated alpha-synuclein proteins in multiple regions of the brain. Neurons can release alpha-synuclein; through this release, pathological forms of alpha-synuclein are propagated between neurons, and also cause neuroinflammation. In this study, we demonstrate that release of alpha-synuclein is consistently increased under various protein misfolding stress conditions in both neuroblastoma and primary neuron models. This release is mediated by a non-classical, endoplasmic reticulum (ER)/Golgi-independent exocytosis, and stress-induced release coincides with increased translocation of alpha-synuclein into vesicles. Both vesicle translocation and secretion were blocked by attachment of a highly stable, globular protein to alpha-synuclein, whereas forced protein misfolding resulted in an increase in both of these activities. Mass spectrometry analysis showed a higher degree of oxidative modification in secreted alpha-synuclein than in the cellular protein. Together, these results suggest that structurally abnormal, damaged alpha-synuclein proteins translocate preferentially into vesicles and are released from neuronal cells via exocytosis.


Asunto(s)
Exocitosis/efectos de los fármacos , Pliegue de Proteína , Estrés Fisiológico/fisiología , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Supervivencia Celular/efectos de los fármacos , Centrifugación , Chlorocebus aethiops , Citosol/metabolismo , Endopeptidasa K/química , Humanos , Hidrólisis , L-Lactato Deshidrogenasa/metabolismo , Conformación Proteica , Inhibidores de la Síntesis de la Proteína/farmacología , Transducción de Señal/fisiología , Vesículas Transportadoras/metabolismo , Regulación hacia Arriba/fisiología
12.
Exp Mol Med ; 51(11): 1-10, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31740682

RESUMEN

Multiple system atrophy (MSA) is a neurodegenerative disease with diverse clinical manifestations, including parkinsonism, cerebellar syndrome, and autonomic failure. Pathologically, MSA is characterized by glial cytoplasmic inclusions in oligodendrocytes, which contain fibrillary forms of α-synuclein. MSA is categorized as one of the α-synucleinopathy, and α-synuclein aggregation is thought to be the culprit of the disease pathogenesis. Studies on MSA pathogenesis are scarce relative to studies on the pathogenesis of other synucleinopathies, such as Parkinson's disease and dementia with Lewy bodies. However, recent developments in cellular and animal models of MSA, especially α-synuclein transgenic models, have driven advancements in research on this disease. Here, we review the currently available models of MSA, which include toxicant-induced animal models, α-synuclein-overexpressing cellular models, and mouse models that express α-synuclein specifically in oligodendrocytes through cell type-specific promoters. We will also discuss the results of studies in recently developed transmission mouse models, into which MSA brain extracts were intracerebrally injected. By reviewing the findings obtained from these model systems, we will discuss what we have learned about the disease and describe the strengths and limitations of the models, thereby ultimately providing direction for the design of better models and future research.


Asunto(s)
Modelos Animales de Enfermedad , Atrofia de Múltiples Sistemas/patología , Animales , Humanos , Atrofia de Múltiples Sistemas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
14.
Int J Biochem Cell Biol ; 40(9): 1835-49, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18291704

RESUMEN

Abnormal folding and accumulation of alpha-synuclein is implicated in several neurological disorders including Parkinson's disease. Although alpha-synuclein is a typical cytoplasmic protein, a small amount of both monomeric and aggregated forms is secreted from cells and is present in human body fluids, such as cerebrospinal fluid. Extracellular alpha-synuclein aggregates have been shown to be neurotoxic, posing a challenge to any cell exposed to them. Here, we examine the internalization of various forms of extracellular alpha-synuclein, including fibrils, oligomers, and monomer, into neuronal cells and their subsequent degradation. Internalization of fibrillar alpha-synuclein could be inhibited by low temperature or the expression of a dominant-negative mutant dynamin-1 K44A, suggesting the endocytosis-mediated internalization. The internalized fibrils moved through the endosomal pathway and were degraded in the lysosome, which ultimately resulted in the clearance of the alpha-synuclein aggregates from the culture medium. Non-fibrillar oligomeric aggregates were also internalized via endocytosis and degraded by the lysosome. In contrast to aggregate uptake, the internalization of monomeric alpha-synuclein was unaffected by cold temperature and the expression of dynamin-1 K44A, consistent with direct translocation across the plasma membrane. Internalized monomers rapidly pass the plasma membrane, escaping the cells before being degraded by the cellular proteolytic systems. These results suggest that only aggregated forms of extracellular alpha-synuclein can be cleared by cell-mediated uptake and degradation, and this might represent a mechanism of preventing neurons from exposure to potentially toxic alpha-synuclein.


Asunto(s)
Endocitosis , Espacio Extracelular/metabolismo , alfa-Sinucleína/metabolismo , Línea Celular , Membrana Celular/metabolismo , Difusión , Endosomas/metabolismo , Humanos , Lisosomas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Unión Proteica
15.
Biochem Biophys Res Commun ; 372(3): 423-8, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18492487

RESUMEN

Abnormal deposition of alpha-synuclein in neurons and glia is implicated in many neurological diseases, such as Parkinson's disease and Dementia with Lewy bodies. Recently, evidence has emerged that this protein and its aggregates are secreted from neuronal cells, and this extracellular protein may contribute to the pathogenic process. Here, we show that all the major brain cell types (neurons, astrocytes, and microglia) are capable of clearing the extracellular alpha-synuclein aggregates by internalization and degradation. Among these cell types, microglia showed the highest rate of degradation. Upon activation by lipopolysaccharide, the degradation of the internalized alpha-synuclein aggregates was slowed, causing protein accumulation in the microglial cytoplasm. These results suggest that microglia may be the major scavenger cells for extracellular alpha-synuclein aggregates in brain parenchyma, and that clearance may be regulated by the activation state of these cells.


Asunto(s)
Microglía/metabolismo , alfa-Sinucleína/metabolismo , Línea Celular Tumoral , Humanos
16.
Cell Death Dis ; 9(7): 712, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29907758

RESUMEN

Trehalose is a non-reducing disaccharide with two glucose molecules linked through an α, α-1,1-glucosidic bond. Trehalose has received attention for the past few decades for its role in neuroprotection especially in animal models of various neurodegenerative diseases, such as Parkinson and Huntington diseases. The mechanism underlying the neuroprotective effects of trehalose remains elusive. The prevailing hypothesis is that trehalose protects neurons by inducing autophagy, thereby clearing protein aggregates. Some of the animal studies showed activation of autophagy and reduced protein aggregates after trehalose administration in neurodegenerative disease models, seemingly supporting the autophagy induction hypothesis. However, results from cell studies have been less certain; although many studies claim that trehalose induces autophagy and reduces protein aggregates, the studies have their weaknesses, failing to provide sufficient evidence for the autophagy induction theory. Furthermore, a recent study with a thorough examination of autophagy flux showed that trehalose interfered with the flux from autophagosome to autolysosome, raising controversy on the direct effects of trehalose on autophagy. This review summarizes the fundamental properties of trehalose and the studies on its effects on neurodegenerative diseases. We also discuss the controversy related to the autophagy induction theory and seek to explain how trehalose works in neuroprotection.


Asunto(s)
Autofagia/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Trehalosa/farmacología , Animales , Invertebrados/metabolismo , Trehalosa/química , Trehalosa/metabolismo , Vertebrados/metabolismo
17.
Mol Neurodegener ; 13(1): 43, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30092810

RESUMEN

BACKGROUND: Synucleinopathies of the aging population are an heterogeneous group of neurological disorders that includes Parkinson's disease (PD) and dementia with Lewy bodies (DLB) and are characterized by the progressive accumulation of α-synuclein in neuronal and glial cells. Toll-like receptor 2 (TLR2), a pattern recognition immune receptor, has been implicated in the pathogenesis of synucleinopathies because TLR2 is elevated in the brains of patients with PD and TLR2 is a mediator of the neurotoxic and pro-inflammatory effects of extracellular α-synuclein aggregates. Therefore, blocking TLR2 might alleviate α-synuclein pathological and functional effects. For this purpose, herein, we targeted TLR2 using a functional inhibitory antibody (anti-TLR2). METHODS: Two different human α-synuclein overexpressing transgenic mice were used in this study. α-synuclein low expresser mouse (α-syn-tg, under the PDGFß promoter, D line) was stereotaxically injected with TLR2 overexpressing lentivirus to demonstrate that increment of TLR2 expression triggers neurotoxicity and neuroinflammation. α-synuclein high expresser mouse (α-Syn-tg; under mThy1 promoter, Line 61) was administrated with anti-TLR2 to examine that functional inhibition of TLR2 ameliorates neuropathology and behavioral defect in the synucleinopathy animal model. In vitro α-synuclein transmission live cell monitoring system was used to evaluate the role of TLR2 in α-synuclein cell-to-cell transmission. RESULTS: We demonstrated that administration of anti-TLR2 alleviated α-synuclein accumulation in neuronal and astroglial cells, neuroinflammation, neurodegeneration, and behavioral deficits in an α-synuclein tg mouse model of PD/DLB. Moreover, in vitro studies with neuronal and astroglial cells showed that the neuroprotective effects of anti-TLR2 antibody were mediated by blocking the neuron-to-neuron and neuron-to-astrocyte α-synuclein transmission which otherwise promotes NFκB dependent pro-inflammatory responses. CONCLUSION: This study proposes TLR2 immunotherapy as a novel therapeutic strategy for synucleinopathies of the aging population.


Asunto(s)
Encéfalo/patología , Enfermedad por Cuerpos de Lewy/patología , Trastornos Parkinsonianos/patología , Receptor Toll-Like 2/antagonistas & inhibidores , alfa-Sinucleína/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/métodos , Enfermedad por Cuerpos de Lewy/metabolismo , Ratones , Ratones Transgénicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Trastornos Parkinsonianos/metabolismo
18.
Nat Commun ; 9(1): 3465, 2018 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-30150626

RESUMEN

Propagation of α-synuclein aggregates has been suggested as a contributing factor in Parkinson's disease (PD) progression. However, the molecular mechanisms underlying α-synuclein aggregation are not fully understood. Here, we demonstrate in cell culture, nematode, and rodent models of PD that leucine-rich repeat kinase 2 (LRRK2), a PD-linked kinase, modulates α-synuclein propagation in a kinase activity-dependent manner. The PD-linked G2019S mutation in LRRK2, which increases kinase activity, enhances propagation efficiency. Furthermore, we show that the role of LRRK2 in α-synuclein propagation is mediated by RAB35 phosphorylation. Constitutive activation of RAB35 overrides the reduced α-synuclein propagation phenotype in lrk-1 mutant C. elegans. Finally, in a mouse model of synucleinopathy, administration of an LRRK2 kinase inhibitor reduced α-synuclein aggregation via enhanced interaction of α-synuclein with the lysosomal degradation pathway. These results suggest that LRRK2-mediated RAB35 phosphorylation is a potential therapeutic target for modifying disease progression.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Plásmidos/genética , alfa-Sinucleína/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Western Blotting , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Línea Celular Tumoral , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Microscopía Fluorescente , Fosforilación , Ratas , alfa-Sinucleína/genética , Proteínas de Unión al GTP rab/genética
19.
Cell Death Dis ; 8(10): e3091, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28981090

RESUMEN

Autophagy is a pivotal intracellular process by which cellular macromolecules are degraded upon various stimuli. A failure in the degradation of autophagic substrates such as impaired organelles and protein aggregates leads to their accumulations, which are characteristics of many neurodegenerative diseases. Pharmacological activation of autophagy has thus been considered a prospective therapeutic approach for treating neurodegenerative diseases. Among a number of autophagy-inducing agents, trehalose has received attention for its beneficial effects in different disease models of neurodegeneration. However, how trehalose promotes autophagy has not been fully revealed. We investigated the influence of trehalose and other disaccharides upon autophagic flux and aggregation of α-synuclein, a protein linked to Parkinson's disease. In differentiated human neuroblastoma and primary rat cortical neuron culture models, treatment with trehalose and other disaccharides resulted in accumulation of lipidated LC3 (LC3-II), p62, and autophagosomes, whereas it decreased autolysosomes. On the other hand, addition of Bafilomycin A1 to trehalose treatments had relatively marginal effect, an indicative of autophagic flux blockage. In concordance with these results, the cells treated with trehalose exhibited an incremental tendency in α-synuclein aggregation. Secretion of α-synuclein was also elevated in the culture medium upon trehalose treatment, thereby significantly increasing intercellular transmission of this protein. Despite the substantial increase in α-synuclein aggregation, which normally leads to cell death, cell viability was not affected upon treatment with trehalose, suggesting an autophagy-independent protective function of trehalose against protein aggregates. This study demonstrates that, although trehalose has been widely considered an autophagic inducer, it may be actually a potent blocker of the autophagic flux.


Asunto(s)
Enfermedad de Parkinson/tratamiento farmacológico , Agregación Patológica de Proteínas/tratamiento farmacológico , Trehalosa/administración & dosificación , alfa-Sinucleína/genética , Animales , Autofagosomas/efectos de los fármacos , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Disacáridos/administración & dosificación , Humanos , Lisosomas , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Ratas
20.
Exp Neurobiol ; 26(6): 321-328, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29302199

RESUMEN

Huntington disease (HD) is an inherited neurodegenerative disorder characterized by motor and cognitive dysfunction caused by expansion of polyglutamine (polyQ) repeat in exon 1 of huntingtin (HTT). In patients, the number of glutamine residues in polyQ tracts are over 35, and it is correlated with age of onset, severity, and disease progression. Expansion of polyQ increases the propensity for HTT protein aggregation, process known to be implicated in neurodegeneration. These pathological aggregates can be transmitted from neuron to another neuron, and this process may explain the pathological spreading of polyQ aggregates. Here, we developed an in vivo model for studying transmission of polyQ aggregates in a highly quantitative manner in real time. HTT exon 1 with expanded polyQ was fused with either N-terminal or C-terminal fragments of Venus fluorescence protein and expressed in pharyngeal muscles and associated neurons, respectively, of C. elegans. Transmission of polyQ proteins was detected using bimolecular fluorescence complementation (BiFC). Mutant polyQ (Q97) was transmitted much more efficiently than wild type polyQ (Q25) and forms numerous inclusion bodies as well. The transmission of Q97 was gradually increased with aging of animal. The animals with polyQ transmission exhibited degenerative phenotypes, such as nerve degeneration, impaired pharyngeal pumping behavior, and reduced life span. The C. elegans model presented here would be a useful in vivo model system for the study of polyQ aggregate propagation and might be applied to the screening of genetic and chemical modifiers of the propagation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA