Your browser doesn't support javascript.
loading
Glia-to-neuron transfer of miRNAs via extracellular vesicles: a new mechanism underlying inflammation-induced synaptic alterations.
Prada, Ilaria; Gabrielli, Martina; Turola, Elena; Iorio, Alessia; D'Arrigo, Giulia; Parolisi, Roberta; De Luca, Mariacristina; Pacifici, Marco; Bastoni, Mattia; Lombardi, Marta; Legname, Giuseppe; Cojoc, Dan; Buffo, Annalisa; Furlan, Roberto; Peruzzi, Francesca; Verderio, Claudia.
Afiliação
  • Prada I; CNR Institute of Neuroscience, via Vanvitelli 32, 20129, Milan, Italy.
  • Gabrielli M; CNR Institute of Neuroscience, via Vanvitelli 32, 20129, Milan, Italy.
  • Turola E; Gastroenterology Unit, Department of Internal Medicine, University of Modena and Reggio Emilia, 41124, Modena, Italy.
  • Iorio A; CNR Institute of Neuroscience, via Vanvitelli 32, 20129, Milan, Italy.
  • D'Arrigo G; Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), via Bonomea 265, 34136, Trieste, Italy.
  • Parolisi R; Department of Neuroscience Rita Levi-Montalcini and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10126, Turin, Italy.
  • De Luca M; LSU Health Sciences Center School of Medicine and Stanley S. Scott Cancer Center, New Orleans, USA.
  • Pacifici M; LSU Health Sciences Center School of Medicine and Stanley S. Scott Cancer Center, New Orleans, USA.
  • Bastoni M; Clinical Neuroimmunology Unit, Department of Neuroscience, Institute for Experimental Neurology, San Raffaele Scientific Institute, via Olgettina 58, 20132, Milan, Italy.
  • Lombardi M; IRCCS Humanitas, via Manzoni 56, 20089, Rozzano, Italy.
  • Legname G; Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), via Bonomea 265, 34136, Trieste, Italy.
  • Cojoc D; CNR-Institute of Materials, Area Science Park, 34149, Trieste, Basovizza, Italy.
  • Buffo A; Department of Neuroscience Rita Levi-Montalcini and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10126, Turin, Italy.
  • Furlan R; Clinical Neuroimmunology Unit, Department of Neuroscience, Institute for Experimental Neurology, San Raffaele Scientific Institute, via Olgettina 58, 20132, Milan, Italy.
  • Peruzzi F; LSU Health Sciences Center School of Medicine and Stanley S. Scott Cancer Center, New Orleans, USA.
  • Verderio C; CNR Institute of Neuroscience, via Vanvitelli 32, 20129, Milan, Italy. c.verderio@in.cnr.it.
Acta Neuropathol ; 135(4): 529-550, 2018 04.
Article em En | MEDLINE | ID: mdl-29302779
ABSTRACT
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.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Sinapses / Neuroglia / MicroRNAs / Vesículas Extracelulares / Inflamação / Neurônios Limite: Animals / Female / Humans / Male Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Sinapses / Neuroglia / MicroRNAs / Vesículas Extracelulares / Inflamação / Neurônios Limite: Animals / Female / Humans / Male Idioma: En Ano de publicação: 2018 Tipo de documento: Article