Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 148(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34081130

RESUMEN

Epigenetic factors have been shown to play a crucial role in X-linked intellectual disability (XLID). Here, we investigate the contribution of the XLID-associated histone demethylase PHF8 to astrocyte differentiation and function. Using genome-wide analyses and biochemical assays in mouse astrocytic cultures, we reveal a regulatory crosstalk between PHF8 and the Notch signaling pathway that balances the expression of the master astrocytic gene Nfia. Moreover, PHF8 regulates key synaptic genes in astrocytes by maintaining low levels of H4K20me3. Accordingly, astrocytic-PHF8 depletion has a striking effect on neuronal synapse formation and maturation in vitro. These data reveal that PHF8 is crucial in astrocyte development to maintain chromatin homeostasis and limit heterochromatin formation at synaptogenic genes. Our studies provide insights into the involvement of epigenetics in intellectual disability.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular , Regulación de la Expresión Génica , Histona Demetilasas/genética , Factores de Transcripción/genética , Animales , Astrocitos/citología , Sitios de Unión , Biomarcadores , Diferenciación Celular/genética , Proliferación Celular , Perfilación de la Expresión Génica , Histona Demetilasas/metabolismo , Histonas/metabolismo , Ratones , Modelos Biológicos , Neurogénesis , Neuronas/metabolismo , Unión Proteica , Sinapsis/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
2.
Brain ; 145(8): 2849-2868, 2022 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-35254410

RESUMEN

Synaptic dysfunction is an early mechanism in Alzheimer's disease that involves progressively larger areas of the brain over time. However, how it starts and propagates is unknown. Here we show that amyloid-ß released by microglia in association with large extracellular vesicles (Aß-EVs) alters dendritic spine morphology in vitro, at the site of neuron interaction, and impairs synaptic plasticity both in vitro and in vivo in the entorhinal cortex-dentate gyrus circuitry. One hour after Aß-EV injection into the mouse entorhinal cortex, long-term potentiation was impaired in the entorhinal cortex but not in the dentate gyrus, its main target region, while 24 h later it was also impaired in the dentate gyrus, revealing a spreading of long-term potentiation deficit between the two regions. Similar results were obtained upon injection of extracellular vesicles carrying Aß naturally secreted by CHO7PA2 cells, while neither Aß42 alone nor inflammatory extracellular vesicles devoid of Aß were able to propagate long-term potentiation impairment. Using optical tweezers combined to time-lapse imaging to study Aß-EV-neuron interaction, we show that Aß-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aß-EV motility was inhibited, no propagation of long-term potentiation deficit occurred along the entorhinal-hippocampal circuit, implicating large extracellular vesicle motion at the neuron surface in the spreading of long-term potentiation impairment. Our data indicate the involvement of large microglial extracellular vesicles in the rise and propagation of early synaptic dysfunction in Alzheimer's disease and suggest a new mechanism controlling the diffusion of large extracellular vesicles and their pathogenic signals in the brain parenchyma, paving the way for novel therapeutic strategies to delay the disease.


Asunto(s)
Enfermedad de Alzheimer , Vesículas Extracelulares , Péptidos beta-Amiloides , Animales , Hipocampo , Potenciación a Largo Plazo , Ratones , Microglía
3.
Int J Mol Sci ; 24(12)2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37372953

RESUMEN

Extensive evidence indicates that the activation of the P2X7 receptor (P2X7R), an ATP-gated ion channel highly expressed in immune and brain cells, is strictly associated with the release of extracellular vesicles. Through this process, P2X7R-expressing cells regulate non-classical protein secretion and transfer bioactive components to other cells, including misfolded proteins, participating in inflammatory and neurodegenerative diseases. In this review, we summarize and discuss the studies addressing the impact of P2X7R activation on extracellular vesicle release and their activities.


Asunto(s)
Encéfalo , Vesículas Extracelulares , Receptores Purinérgicos P2X7 , Adenosina Trifosfato
4.
Pharmacol Res ; 178: 106149, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35240272

RESUMEN

Neural tissue has high metabolic requirements. Following spinal cord injury (SCI), the damaged tissue suffers from a severe metabolic impairment, which aggravates axonal degeneration and neuronal loss. Impaired cellular energetic, tricarboxylic acid (TCA) cycle and oxidative phosphorylation metabolism in neuronal cells has been demonstrated to be a major cause of neural tissue death and regeneration failure following SCI. Therefore, rewiring the spinal cord cell metabolism may be an innovative therapeutic strategy for the treatment of SCI. In this study, we evaluated the therapeutic effect of the recovery of oxidative metabolism in a mouse model of severe contusive SCI. Oral administration of TCA cycle intermediates, co-factors, essential amino acids, and branched-chain amino acids was started 3 days post-injury and continued until the end of the experimental procedures. Metabolomic, immunohistological, and biochemical analyses were performed on the injured spinal cord sections. Administration of metabolic precursors enhanced spinal cord oxidative metabolism. In line with this metabolic shift, we observed the activation of the mTORC1 anabolic pathway, the increase in mitochondrial mass, and ROS defense which effectively prevented the injury-induced neural cell apoptosis in treated animals. Consistently, we found more choline acetyltransferase (ChAT)-expressing motor neurons and increased neurofilament-positive corticospinal axons in the spinal cord parenchyma of the treated mice. Interestingly, oral administration of the metabolic precursors increased the number of activated microglia expressing the CD206 marker suggestive of a pro-resolutive, M2-like phenotype. These molecular and histological modifications observed in treated animals ultimately led to a significant, although partial, improvement of the motor functions. Our data demonstrate that rewiring the cellular metabolism can represent an effective strategy to treat SCI.


Asunto(s)
Microglía , Traumatismos de la Médula Espinal , Animales , Axones/fisiología , Metabolismo Energético , Ratones , Microglía/metabolismo , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología
5.
Mol Ther ; 29(4): 1439-1458, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33309882

RESUMEN

Contrasting myelin damage through the generation of new myelinating oligodendrocytes represents a promising approach to promote functional recovery after stroke. Here, we asked whether activation of microglia and monocyte-derived macrophages affects the regenerative process sustained by G protein-coupled receptor 17 (GPR17)-expressing oligodendrocyte precursor cells (OPCs), a subpopulation of OPCs specifically reacting to ischemic injury. GPR17-iCreERT2:CAG-eGFP reporter mice were employed to trace the fate of GPR17-expressing OPCs, labeled by the green fluorescent protein (GFP), after permanent middle cerebral artery occlusion. By microglia/macrophages pharmacological depletion studies, we show that innate immune cells favor GFP+ OPC reaction and limit myelin damage early after injury, whereas they lose their pro-resolving capacity and acquire a dystrophic "senescent-like" phenotype at later stages. Intracerebral infusion of regenerative microglia-derived extracellular vesicles (EVs) restores protective microglia/macrophages functions, limiting their senescence during the post-stroke phase, and enhances the maturation of GFP+ OPCs at lesion borders, resulting in ameliorated neurological functionality. In vitro experiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.


Asunto(s)
Senescencia Celular/genética , Vaina de Mielina/genética , Receptores Acoplados a Proteínas G/genética , Accidente Cerebrovascular/terapia , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/terapia , Macrófagos/metabolismo , Macrófagos/trasplante , Masculino , Ratones , Microglía/metabolismo , Microglía/trasplante , Oligodendroglía/trasplante , Medicina Regenerativa/métodos , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/genética
6.
Acta Neuropathol ; 138(6): 987-1012, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31363836

RESUMEN

Microglia are highly plastic immune cells which exist in a continuum of activation states. By shaping the function of oligodendrocyte precursor cells (OPCs), the brain cells which differentiate to myelin-forming cells, microglia participate in both myelin injury and remyelination during multiple sclerosis. However, the mode(s) of action of microglia in supporting or inhibiting myelin repair is still largely unclear. Here, we analysed the effects of extracellular vesicles (EVs) produced in vitro by either pro-inflammatory or pro-regenerative microglia on OPCs at demyelinated lesions caused by lysolecithin injection in the mouse corpus callosum. Immunolabelling for myelin proteins and electron microscopy showed that EVs released by pro-inflammatory microglia blocked remyelination, whereas EVs produced by microglia co-cultured with immunosuppressive mesenchymal stem cells promoted OPC recruitment and myelin repair. The molecular mechanisms responsible for the harmful and beneficial EV actions were dissected in primary OPC cultures. By exposing OPCs, cultured either alone or with astrocytes, to inflammatory EVs, we observed a blockade of OPC maturation only in the presence of astrocytes, implicating these cells in remyelination failure. Biochemical fractionation revealed that astrocytes may be converted into harmful cells by the inflammatory EV cargo, as indicated by immunohistochemical and qPCR analyses, whereas surface lipid components of EVs promote OPC migration and/or differentiation, linking EV lipids to myelin repair. Although the mechanisms through which the lipid species enhance OPC maturation still remain to be fully defined, we provide the first demonstration that vesicular sphingosine 1 phosphate stimulates OPC migration, the first fundamental step in myelin repair. From this study, microglial EVs emerge as multimodal and multitarget signalling mediators able to influence both OPCs and astrocytes around myelin lesions, which may be exploited to develop novel approaches for myelin repair not only in multiple sclerosis, but also in neurological and neuropsychiatric diseases characterized by demyelination.


Asunto(s)
Astrocitos/fisiología , Enfermedades Desmielinizantes/fisiopatología , Vesículas Extracelulares/fisiología , Microglía/fisiología , Vaina de Mielina/fisiología , Remielinización/fisiología , Animales , Astrocitos/patología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Técnicas de Cocultivo , Cuerpo Calloso/patología , Cuerpo Calloso/fisiopatología , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Vesículas Extracelulares/patología , Inflamación/patología , Inflamación/fisiopatología , Lisofosfatidilcolinas , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones Endogámicos C57BL , Microglía/patología , Vaina de Mielina/patología , Neuroprotección/fisiología , Células Precursoras de Oligodendrocitos/patología , Células Precursoras de Oligodendrocitos/fisiología , Ratas Sprague-Dawley
7.
J Am Soc Nephrol ; 29(3): 880-905, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29382685

RESUMEN

Increased export of transglutaminase-2 (TG2) by tubular epithelial cells (TECs) into the surrounding interstitium modifies the extracellular homeostatic balance, leading to fibrotic membrane expansion. Although silencing of extracellular TG2 ameliorates progressive kidney scarring in animal models of CKD, the pathway through which TG2 is secreted from TECs and contributes to disease progression has not been elucidated. In this study, we developed a global proteomic approach to identify binding partners of TG2 responsible for TG2 externalization in kidneys subjected to unilateral ureteric obstruction (UUO) using TG2 knockout kidneys as negative controls. We report a robust and unbiased analysis of the membrane interactome of TG2 in fibrotic kidneys relative to the entire proteome after UUO, detected by SWATH mass spectrometry. The data have been deposited to the ProteomeXchange with identifier PXD008173. Clusters of exosomal proteins in the TG2 interactome supported the hypothesis that TG2 is secreted by extracellular membrane vesicles during fibrosis progression. In established TEC lines, we found TG2 in vesicles of both endosomal (exosomes) and plasma membrane origin (microvesicles/ectosomes), and TGF-ß1 stimulated TG2 secretion. Knockout of syndecan-4 (SDC4) greatly impaired TG2 exosomal secretion. TG2 coprecipitated with SDC4 from exosome lysate but not ectosome lysate. Ex vivo, EGFP-tagged TG2 accumulated in globular elements (blebs) protruding/retracting from the plasma membrane of primary cortical TECs, and SDC4 knockout impaired bleb formation, affecting TG2 release. Through this combined in vivo and in vitro approach, we have dissected the pathway through which TG2 is secreted from TECs in CKD.


Asunto(s)
Células Epiteliales/metabolismo , Exosomas/enzimología , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Riñón/patología , Insuficiencia Renal Crónica/metabolismo , Transglutaminasas/genética , Transglutaminasas/metabolismo , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Línea Celular , Micropartículas Derivadas de Células/enzimología , Inhibidores Enzimáticos/farmacología , Fibrosis , Humanos , Túbulos Renales/citología , Ratones , Ratones Noqueados , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteómica , Ratas , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/orina , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Sindecano-4/genética , Sindecano-4/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones
8.
J Lipid Res ; 59(8): 1325-1340, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29853528

RESUMEN

Extracellular vesicles (EVs) are membrane vesicles released by both eukaryotic and prokaryotic cells; they not only serve physiological functions, such as disposal of cellular components, but also play pathophysiologic roles in inflammatory and degenerative diseases. Common molecular mechanisms for EV biogenesis are evident in different cell biological contexts across eukaryotic phyla, and inhibition of this biogenesis may provide an avenue for therapeutic research. The involvement of sphingolipids (SLs) and their enzymes on EV biogenesis and release has not received much attention in current research. Here, we review how SLs participate in EV biogenesis by shaping membrane curvature and how they contribute to EV action in target cells. First, we describe how acid and neutral SMases, by generating the constitutive SL, ceramide, facilitate biogenesis of EVs at the plasma membrane and inside the endocytic compartment. We then discuss the involvement of other SLs, such as sphingosine-1-phosphate and galactosyl-sphingosine, in EV formation and cargo sorting. Last, we look ahead at some biological effects of EVs mediated by changes in SL levels in recipient cells.


Asunto(s)
Vesículas Extracelulares/metabolismo , Esfingolípidos/metabolismo , Animales , Biomarcadores/metabolismo , Membrana Celular/metabolismo , Endocannabinoides/metabolismo , Humanos
9.
Glia ; 66(12): 2531-2549, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30195261

RESUMEN

Microglia, brain cells of nonneural origin, orchestrate the inflammatory response to diverse insults, including hypoxia/ischemia or maternal/fetal infection in the perinatal brain. Experimental studies have demonstrated the capacity of microglia to recognize pathogens or damaged cells activating a cytotoxic response that can exacerbate brain damage. However, microglia display an enormous plasticity in their responses to injury and may also promote resolution stages of inflammation and tissue regeneration. Despite the critical role of microglia in brain pathologies, the cellular mechanisms that govern the diverse phenotypes of microglia are just beginning to be defined. Here we review emerging strategies to drive microglia toward beneficial functions, selectively reporting the studies which provide insights into molecular mechanisms underlying the phenotypic switch. A variety of approaches have been proposed which rely on microglia treatment with pharmacological agents, cytokines, lipid messengers, or microRNAs, as well on nutritional approaches or therapies with immunomodulatory cells. Analysis of the molecular mechanisms relevant for microglia reprogramming toward pro-regenerative functions points to a central role of energy metabolism in shaping microglial functions. Manipulation of metabolic pathways may thus provide new therapeutic opportunities to prevent the deleterious effects of inflammatory microglia and to control excessive inflammation in brain disorders.


Asunto(s)
Reprogramación Celular/fisiología , Encefalitis/terapia , Microglía/fisiología , Animales , Humanos
10.
Acta Neuropathol ; 135(4): 529-550, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302779

RESUMEN

Recent evidence indicates synaptic dysfunction as an early mechanism affected in neuroinflammatory diseases, such as multiple sclerosis, which are characterized by chronic microglia activation. However, the mode(s) of action of reactive microglia in causing synaptic defects are not fully understood. In this study, we show that inflammatory microglia produce extracellular vesicles (EVs) which are enriched in a set of miRNAs that regulate the expression of key synaptic proteins. Among them, miR-146a-5p, a microglia-specific miRNA not present in hippocampal neurons, controls the expression of presynaptic synaptotagmin1 (Syt1) and postsynaptic neuroligin1 (Nlg1), an adhesion protein which play a crucial role in dendritic spine formation and synaptic stability. Using a Renilla-based sensor, we provide formal proof that inflammatory EVs transfer their miR-146a-5p cargo to neuron. By western blot and immunofluorescence analysis we show that vesicular miR-146a-5p suppresses Syt1 and Nlg1 expression in receiving neurons. Microglia-to-neuron miR-146a-5p transfer and Syt1 and Nlg1 downregulation do not occur when EV-neuron contact is inhibited by cloaking vesicular phosphatidylserine residues and when neurons are exposed to EVs either depleted of miR-146a-5p, produced by pro-regenerative microglia, or storing inactive miR-146a-5p, produced by cells transfected with an anti-miR-146a-5p. Morphological analysis reveals that prolonged exposure to inflammatory EVs leads to significant decrease in dendritic spine density in hippocampal neurons in vivo and in primary culture, which is rescued in vitro by transfection of a miR-insensitive Nlg1 form. Dendritic spine loss is accompanied by a decrease in the density and strength of excitatory synapses, as indicated by reduced mEPSC frequency and amplitude. These findings link inflammatory microglia and enhanced EV production to loss of excitatory synapses, uncovering a previously unrecognized role for microglia-enriched miRNAs, released in association to EVs, in silencing of key synaptic genes.


Asunto(s)
Vesículas Extracelulares/inmunología , Inflamación/metabolismo , MicroARNs/metabolismo , Neuroglía/inmunología , Neuronas/inmunología , Sinapsis/inmunología , Animales , Células Cultivadas , Líquido Cefalorraquídeo/metabolismo , Técnicas de Cocultivo , Vesículas Extracelulares/patología , Femenino , Hipocampo/inmunología , Hipocampo/patología , Humanos , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Neuroglía/patología , Plasticidad Neuronal/fisiología , Neuronas/patología , Cultivo Primario de Células , Ratas Sprague-Dawley , Sinapsis/patología
12.
J Neurosci ; 36(16): 4624-34, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27098703

RESUMEN

Growing evidence indicates that sphingosine-1-P (S1P) upregulates glutamate secretion in hippocampal neurons. However, the molecular mechanisms through which S1P enhances excitatory activity remain largely undefined. The aim of this study was to identify presynaptic targets of S1P action controlling exocytosis. Confocal analysis of rat hippocampal neurons showed that S1P applied at nanomolar concentration alters the distribution of Synapsin I (SynI), a presynaptic phosphoprotein that controls the availability of synaptic vesicles for exocytosis. S1P induced SynI relocation to extrasynaptic regions of mature neurons, as well as SynI dispersion from synaptic vesicle clusters present at axonal growth cones of developing neurons. S1P-induced SynI relocation occurred in a Ca(2+)-independent but ERK-dependent manner, likely through the activation of S1P3 receptors, as it was prevented by the S1P3 receptor selective antagonist CAY1044 and in neurons in which S1P3 receptor was silenced. Our recent evidence indicates that microvesicles (MVs) released by microglia enhance the metabolism of endogenous sphingolipids in neurons and stimulate excitatory transmission. We therefore investigated whether MVs affect SynI distribution and whether endogenous S1P could be involved in the process. Analysis of SynI immunoreactivity showed that exposure to microglial MVs induces SynI mobilization at presynaptic sites and growth cones, whereas the use of inhibitors of sphingolipid cascade identified S1P as the sphingolipid mediating SynI redistribution. Our data represent the first demonstration that S1P induces SynI mobilization from synapses, thereby indicating the phosphoprotein as a novel target through which S1P controls exocytosis. SIGNIFICANCE STATEMENT: Growing evidence indicates that the bioactive lipid sphingosine and its metabolite sphingosine-1-P (S1P) stimulate excitatory transmission. While it has been recently clarified that sphingosine influences directly the exocytotic machinery by activating the synaptic vesicle protein VAMP2 to form SNARE fusion complexes, the molecular mechanism by which S1P promotes neurotransmission remained largely undefined. In this study, we identify Synapsin I, a presynaptic phosphoprotein involved in the control of availability of synaptic vesicles for exocytosis, as the key target of S1P action. In addition, we provide evidence that S1P can be produced at mature axon terminals as well as at immature growth cones in response to microglia-derived signals, which may be important to stabilize nascent synapses and to restore or potentiate transmission.


Asunto(s)
Lisofosfolípidos/fisiología , Terminales Presinápticos/metabolismo , Esfingosina/análogos & derivados , Sinapsis/metabolismo , Sinapsinas/biosíntesis , Animales , Células Cultivadas , Femenino , Hipocampo/química , Hipocampo/citología , Hipocampo/metabolismo , Lisofosfolípidos/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terminales Presinápticos/química , Ratas , Ratas Sprague-Dawley , Esfingosina/análisis , Esfingosina/fisiología , Sinapsis/química , Sinapsinas/análisis
13.
EMBO Rep ; 16(2): 213-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25568329

RESUMEN

Endocannabinoids primarily influence neuronal synaptic communication within the nervous system. To exert their function, endocannabinoids need to travel across the intercellular space. However, how hydrophobic endocannabinoids cross cell membranes and move extracellularly remains an unresolved problem. Here, we show that endocannabinoids are secreted through extracellular membrane vesicles produced by microglial cells. We demonstrate that microglial extracellular vesicles carry on their surface N-arachidonoylethanolamine (AEA), which is able to stimulate type-1 cannabinoid receptors (CB1), and inhibit presynaptic transmission, in target GABAergic neurons. This is the first demonstration of a functional role of extracellular vesicular transport of endocannabinoids.


Asunto(s)
Endocannabinoides/metabolismo , Microglía/metabolismo , Animales , Ácidos Araquidónicos , Células Cultivadas , Exosomas/metabolismo , Alcamidas Poliinsaturadas , Terminales Presinápticos/metabolismo , Ratas , Receptores de Cannabinoides/metabolismo , Transmisión Sináptica/fisiología
14.
J Neurochem ; 137(1): 88-100, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26710111

RESUMEN

Alzheimer's disease is a common neurodegenerative, progressive, and fatal disorder. Generation and deposition of amyloid beta (Aß) peptides associate with its pathogenesis and small soluble Aß oligomers show the most pronounced neurotoxic effects and correlate with disease initiation and progression. Recent findings showed that Aß oligomers bind to the cellular prion protein (PrP(C) ) eliciting neurotoxic effects. The role of exosomes, small extracellular vesicles of endosomal origin, in Alzheimer's disease is only poorly understood. Besides serving as disease biomarkers they may promote Aß plaque formation, decrease Aß-mediated synaptotoxicity, and enhance Aß clearance. Here, we explore how exosomal PrP(C) connects to protective functions attributed to exosomes in Alzheimer's disease. To achieve this, we generated a mouse neuroblastoma PrP(C) knockout cell line using transcription activator-like effector nucleases. Using these, as well as SH-SY5Y human neuroblastoma cells, we show that PrP(C) is highly enriched on exosomes and that exosomes bind amyloid beta via PrP(C) . Exosomes showed highest binding affinity for dimeric, pentameric, and oligomeric Aß species. Thioflavin T assays revealed that exosomal PrP(C) accelerates fibrillization of amyloid beta, thereby reducing neurotoxic effects imparted by oligomeric Aß. Our study provides further evidence for a protective role of exosomes in Aß-mediated neurodegeneration and highlights the importance of exosomal PrP(C) in molecular mechanisms of Alzheimer's disease. We show that the prion protein (PrP(C) ) on exosomes captures neurotoxic species of amyloid beta (Aß) promoting its fibrillization. Our study provides evidence for a protective role of exosomes in Alzheimer`s disease and suggests that, depending on its membrane topology, PrP(C) holds a dual function: when expressed at the neuronal surface it acts as receptor for Aß leading to neurotoxic signaling, whereas it detoxifies Aß when present on exosomes. This provides further support for key roles of PrP(C) in Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Exosomas/fisiología , Proteínas PrPC/fisiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Ratones , Proteínas de Neoplasias/metabolismo , Neuroblastoma/patología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Solubilidad , Transfección
15.
EMBO J ; 31(5): 1231-40, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22246184

RESUMEN

Microvesicles (MVs) released into the brain microenvironment are emerging as a novel way of cell-to-cell communication. We have recently shown that microglia, the immune cells of the brain, shed MVs upon activation but their possible role in microglia-to-neuron communication has never been explored. To investigate whether MVs affect neurotransmission, we analysed spontaneous release of glutamate in neurons exposed to MVs and found a dose-dependent increase in miniature excitatory postsynaptic current (mEPSC) frequency without changes in mEPSC amplitude. Paired-pulse recording analysis of evoked neurotransmission showed that MVs mainly act at the presynaptic site, by increasing release probability. In line with the enhancement of excitatory transmission in vitro, injection of MVs into the rat visual cortex caused an acute increase in the amplitude of field potentials evoked by visual stimuli. Stimulation of synaptic activity occurred via enhanced sphingolipid metabolism. Indeed, MVs promoted ceramide and sphingosine production in neurons, while the increase of excitatory transmission induced by MVs was prevented by pharmacological or genetic inhibition of sphingosine synthesis. These data identify microglia-derived MVs as a new mechanism by which microglia influence synaptic activity and highlight the involvement of neuronal sphingosine in this microglia-to-neuron signalling pathway.


Asunto(s)
Microglía/metabolismo , Neuronas/fisiología , Vesículas Secretoras/metabolismo , Esfingolípidos/metabolismo , Sinapsis/metabolismo , Animales , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Neuronas/metabolismo , Ratas
16.
Glia ; 63(7): 1256-69, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25779655

RESUMEN

Trigeminal (TG) pain often lacks a satisfactory pharmacological control. A better understanding of the molecular cross-talk between TG neurons and surrounding satellite glial cells (SGCs) could help identifying innovative targets for the development of more effective analgesics. We have previously demonstrated that neuronal pro-algogenic mediators upregulate G protein-coupled nucleotide P2Y receptors (P2YRs) expressed by TG SGCs in vitro. Here, we have identified the specific P2YR subtypes involved (i.e., the ADP-sensitive P2Y1 R and the UTP-responsive P2Y2 R subtypes), and demonstrated the contribution of neuron-derived prostaglandins to their upregulation. Next, we have translated these data to an in vivo model of TG pain (namely, rats injected with Complete Freund's adjuvant in the temporomandibular joint), by demonstrating activation of SGCs and upregulation of P2Y1 R and P2Y2 R in the ipsi-lateral TG. To unequivocally link P2YRs to the development of facial allodynia, we treated animals with various purinergic antagonists. The selective P2Y2 R antagonist AR-C118925 completely inhibited SGCs activation, exerted a potent anti-allodynic effect that lasted over time, and was still effective when administration was started 6-days post induction of allodynia, i.e. under subchronic pain conditions. Conversely, the selective P2Y1 R antagonist MRS2179 was completely ineffective. Moreover, similarly to the anti-inflammatory drug acetylsalicylic acid and the known anti-migraine agent sumatriptan, the P2X/P2Y nonselective antagonist PPADS was only partially effective, and completely lost its activity under sub-chronic conditions. Taken together, our results highlight glial P2Y2 Rs as potential "druggable" targets for the successful management of TG-related pain.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Dolor Facial/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2Y/farmacología , Células Satélites Perineuronales/efectos de los fármacos , Ganglio del Trigémino/efectos de los fármacos , Enfermedad Aguda , Animales , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/fisiopatología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Dolor Facial/fisiopatología , Adyuvante de Freund , Hiperalgesia/fisiopatología , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Células Satélites Perineuronales/fisiología , Articulación Temporomandibular , Ganglio del Trigémino/fisiopatología
17.
Ann Neurol ; 76(6): 813-25, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25087695

RESUMEN

OBJECTIVES: We have described cerebrospinal fluid (CSF) myeloid microvesicles (MVs) as a marker of microglia activation during neuroinflammation in Alzheimer disease (AD), and characterized their ability to produce toxic amyloid ß1-42 (Aß1-42 ) oligomers from aggregated or soluble substrate. The aim of this study is to investigate the association of CSF myeloid MVs with neuroimaging, clinical, and paraclinical data in AD and mild cognitive impairment (MCI). METHODS: We collected CSF from 106 AD patients, 51 MCI patients, and 29 neurologically healthy controls. We examined CSF myeloid MV content and AD markers. A subgroup of 34 AD and 21 MCI patients underwent structural and diffusion tensor MRI. RESULTS: Higher levels of myeloid MVs were found in the CSF of AD patients and MCI patients converting within 3 years relative to controls, but also, at a lower level, in MCI patients not converting to AD. CSF myeloid MVs were associated with Tau but not with Aß1-42 CSF levels. CSF MVs levels correlated with white matter (WM) tract damage in MCI, and with hippocampal atrophy in AD. INTERPRETATION: Microglial MVs are neurotoxic and myelinotoxic in the presence of Aß1-42 . CSF myeloid MVs, mirroring microglia activation and MV release, are associated with WM damage in MCI and hippocampal atrophy in AD. This suggests that hippocampal microglia activation, in the presence of Aß1-42 in excess, produces neurotoxic and oligodendrotoxic oligomers that, through WM tract damage, spread disease to neighboring and connected areas, causing local microglia activation and propagation of disease through the same sequence of events. Ann Neurol 2014;76:813-825.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Disfunción Cognitiva/líquido cefalorraquídeo , Vaina de Mielina/metabolismo , Células Mieloides/metabolismo , Neuronas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Biomarcadores/líquido cefalorraquídeo , Recuento de Células/métodos , Disfunción Cognitiva/diagnóstico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Vaina de Mielina/patología , Células Mieloides/patología , Neuronas/patología
18.
EMBO Rep ; 14(7): 645-51, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23732542

RESUMEN

SNAP-25 is a key component of the synaptic-vesicle fusion machinery, involved in several psychiatric diseases including schizophrenia and ADHD. SNAP-25 protein expression is lower in different brain areas of schizophrenic patients and in ADHD mouse models. How the reduced expression of SNAP-25 alters the properties of synaptic transmission, leading to a pathological phenotype, is unknown. We show that, unexpectedly, halved SNAP-25 levels at 13-14 DIV not only fail to impair synaptic transmission but instead enhance evoked glutamatergic neurotransmission. This effect is possibly dependent on presynaptic voltage-gated calcium channel activity and is not accompanied by changes in spontaneous quantal events or in the pool of readily releasable synaptic vesicles. Notably, synapses of 13-14 DIV neurons with reduced SNAP-25 expression show paired-pulse depression as opposed to paired-pulse facilitation occurring in their wild-type counterparts. This phenotype disappears with synapse maturation. As alterations in short-term plasticity represent a new mechanism contributing to cognitive impairments in intellectual disabilities, our data provide mechanistic clues for neuronal circuit alterations in psychiatric diseases characterized by reduced expression of SNAP-25.


Asunto(s)
Ácido Glutámico/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Transmisión Sináptica/fisiología , Proteína 25 Asociada a Sinaptosomas/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Ácido Glutámico/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Humanos , Ratones , Plasticidad Neuronal/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transmisión Sináptica/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos , Vesículas Sinápticas/fisiología , Proteína 25 Asociada a Sinaptosomas/antagonistas & inhibidores , Proteína 25 Asociada a Sinaptosomas/genética , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
19.
Cereb Cortex ; 24(2): 364-76, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23064108

RESUMEN

Synaptosomal-associated protein of 25 kDa (SNAP-25) is a protein that participates in the regulation of synaptic vesicle exocytosis through the formation of the soluble NSF attachment protein receptor complex and modulates voltage-gated calcium channels activity. The Snap25 gene has been associated with schizophrenia, attention deficit hyperactivity disorder, and bipolar disorder, and lower levels of SNAP-25 have been described in patients with schizophrenia. We used SNAP-25 heterozygous (SNAP-25(+/-)) mice to investigate at which extent the reduction of the protein levels affects neuronal network function and mouse behavior. As interactions of genotype with the specific laboratory conditions may impact behavioral results, the study was performed through a multilaboratory study in which behavioral tests were replicated in at least 2 of 3 distinct European laboratories. Reductions of SNAP-25 levels were associated with a moderate hyperactivity, which disappeared in the adult animals, and with impaired associative learning and memory. Electroencephalographic recordings revealed the occurrence of frequent spikes, suggesting a diffuse network hyperexcitability. Consistently, SNAP-25(+/-) mice displayed higher susceptibility to kainate-induced seizures, paralleled by degeneration of hilar neurons. Notably, both EEG profile and cognitive defects were improved by antiepileptic drugs. These results indicate that reduction of SNAP-25 expression is associated to generation of epileptiform discharges and cognitive dysfunctions, which can be effectively treated by antiepileptic drugs.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Epilepsia/tratamiento farmacológico , Proteína 25 Asociada a Sinaptosomas/metabolismo , Animales , Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Carbamazepina/uso terapéutico , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Epilepsia/patología , Epilepsia/fisiopatología , Etosuximida/uso terapéutico , Hipercinesia/tratamiento farmacológico , Hipercinesia/patología , Hipercinesia/fisiopatología , Ácido Kaínico , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nimodipina/uso terapéutico , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Proteína 25 Asociada a Sinaptosomas/genética , Ácido Valproico/uso terapéutico
20.
Int J Mol Sci ; 16(3): 4800-13, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25741766

RESUMEN

The intercellular transfer of amyloid-ß (Aß) and tau proteins has received increasing attention in Alzheimer's disease (AD). Among other transfer modes, Aß and tau dissemination has been suggested to occur through release of Extracellular Vesicles (EVs), which may facilitate delivery of pathogenic proteins over large distances. Recent evidence indicates that EVs carry on their surface, specific molecules which bind to extracellular Aß, opening the possibility that EVs may also influence Aß assembly and synaptotoxicity. In this review we focus on studies which investigated the impact of EVs in Aß-mediated neurodegeneration and showed either detrimental or protective role for EVs in the pathology.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Micropartículas Derivadas de Células/metabolismo , Enfermedad de Alzheimer/metabolismo , Exosomas/metabolismo , Humanos , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA