510 results on '"Verderio, Claudia"'
Search Results
152. Astrocytes are required for the oscillatory activity in cultured hippocampal neurons
- Author
-
Verderio, Claudia, primary, Bacci, Alberto, additional, Coco, Silvia, additional, Pravettoni, Elena, additional, Fumagalli, Guido, additional, and Matteoli, Michela, additional
- Published
- 1999
- Full Text
- View/download PDF
153. Synaptic and intrinsic mechanisms shape synchronous oscillations in hippocampal neurons in culture
- Author
-
Bacci, Alberto, primary, Verderio, Claudia, additional, Pravettoni, Elena, additional, and Matteoli, Michela, additional
- Published
- 1999
- Full Text
- View/download PDF
154. Non-synaptic Localization of the Glutamate Transporter EAACI in Cultured Hippocampal Neurons
- Author
-
Coco, Silvia, primary, Verderio, Claudia, additional, Trotti, Davide, additional, Rothstein, Jeffrey D., additional, Volterra, Andrea, additional, and Matteoli, Michela, additional
- Published
- 1997
- Full Text
- View/download PDF
155. Nanostructured TiO2 surfaces promote polarized activation of microglia, but not astrocytes, toward a proinflammatory profile.
- Author
-
De Astis, Silvia, Corradini, Irene, Morini, Raffaella, Rodighiero, Simona, Tomasoni, Romana, Lenardi, Cristina, Verderio, Claudia, Milani, Paolo, and Matteoli, Michela
- Published
- 2013
- Full Text
- View/download PDF
156. P2X 7 Receptor and Extracellular Vesicle Release.
- Author
-
Golia, Maria Teresa, Gabrielli, Martina, and Verderio, Claudia
- Subjects
- *
EXTRACELLULAR vesicles , *CELL anatomy , *ION channels , *NEURODEGENERATION , *BIOACTIVE compounds , *PURINERGIC receptors - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
157. Impact of biofluid viscosity on size and sedimentation efficiency of the isolated microvesicles.
- Author
-
Momen-Heravi, Fatemeh, Balaj, Leonora, Alian, Sara, Trachtenberg1, Alexander J., Hochberg2, Fred H., Skog, Johan, Kuo, Winston Patrick, Verderio, Claudia, Graner, Michael, and Chiesi, Antonio
- Subjects
VESICLES (Cytology) ,LIPIDS ,BLOOD plasma ,SERUM ,CEREBROSPINAL fluid ,URINE - Abstract
Microvesicles are nano-sized lipid vesicles released by all cells in vivo and in vitro. They are released physiologically under normal conditions but their rate of release is higher under pathological conditions such as tumors. Once released they end up in the systemic circulation and have been found and characterized in all biofluids such as plasma, serum, cerebrospinal fluid, breast milk, ascites, and urine. Microvesicles represent the status of the donor cell they are released from and they are currently under intense investigation as a potential source for disease biomarkers. Currently, the "gold standard" for isolating microvesicles is ultracentrifugation, although alternative techniques such as affinity purification have been explored. Viscosity is the resistance of a fluid to a deforming force by either shear or tensile stress. The different chemical and molecular compositions of biofluids have an effect on its viscosity and this could affect movements of the particles inside the fluid. In this manuscript we addressed the issue of whether viscosity has an effect on sedimentation efficiency of microvesicles using ultracentrifugation. We used different biofluids and spiked them with polystyrene beads and assessed their recovery using the Nanoparticle Tracking Analysis. We demonstrate that MVs recovery inversely correlates with viscosity and as a result, sample dilutions should be considered prior to ultracentrifugation when processing any biofluids. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
158. Microglial microvesicle secretion and intercellular signaling.
- Author
-
Turola, Elena, Furlan, Roberto, Bianco, Fabio, Matteoli, Michela, and Verderio, Claudia
- Subjects
VESICLES (Cytology) ,NEUROGLIA ,MYELOID metaplasia ,CYTOKINES ,EXTRACELLULAR space ,SPHINGOMYELINASE ,CERAMIDES ,EXOSOMES - Abstract
Microvesicles (MVs) are released from almost all cell brain types into the microenvironment and are emerging as a novel way of cell-to-cell communication. This review focuses on MVs discharged by microglial cells, the brain resident myeloid cells, which comprise ∼10-12% of brain population. We summarize first evidence indicating that MV shedding is a process activated by the ATP receptor P2X
7 and that shed MVs represent a secretory pathway for the inflammatory cytokine IL-β. We then discuss subsequent findings which clarify how IL-1 β can be locally processed and released from MVs into the extracellular environment. In addition, we describe the current understanding about the mechanism of P2X7 -dependent MV formation and membrane abscission, which, by involving sphingomyelinase activity and ceramide formation, may share similarities with exosome biogenesis. Finally we report our recent results which show that microglia-derived MVs can stimulate neuronal activity and participate to the propagation of inflammatory signals, and suggest new areas for future investigation. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
159. Cracking Down on Inhibition: Selective Removal of GABAergic Interneurons from Hippocampal Networks.
- Author
-
Antonucci, Flavia, Alpár, Alán, Kacza, Johannes, Caleo, Matteo, Verderio, Claudia, Giani, Alice, Martens, Henrik, Chaudhry, Farrukh A., Allegra, Manuela, Grosche, Jens, Michalski, Dominik, Erck, Christian, Hoffmann, Anke, Harkany, Tibor, Matteoli, Michela, and Härtig, Wolfgang
- Subjects
GABA ,INTERNEURONS ,HIPPOCAMPUS (Brain) ,SPASMS ,LABORATORY rats ,NEURAL transmission - Abstract
Inhibitory(GABAergic)interneuronsentrainassembliesofexcitatoryprincipalneuronstoorchestrateinformationprocessinginthehippocampus. Disrupting the dynamic recruitment as well as the temporally precise activity of interneurons in hippocampal circuitries can manifest in epileptiform seizures, and impact specific behavioral traits. Despite the importance of GABAergic interneurons during information encoding in the brain, experimental tools to selectively manipulate GABAergic neurotransmission are limited. Here, we report the selective elimination of GABAergic interneurons by a ribosome inactivation approach through delivery of saporin-conjugated anti-vesicular GABA transporter antibodies (SAVAs) in vitro as well as in the mouse and rat hippocampus in vivo. We demonstrate the selective loss of GABAergic-but not glutamatergic-synapses, reduced GABA release, and a shift in excitation/inhibition balance in mixed cultures of hippocampal neurons exposed to SAVAs.Wealso show the focal and indiscriminate loss of calbindin
+ , calretinin+ , parvalbumin/systemAtransporter 1+ , somatostatin+ , vesicular glutamate transporter 3 (VGLUT3)/cholecystokinin/CB1 cannabinoid receptor+ and neuropeptide Y+ local-circuit interneurons upon SAVA microlesions to the CA1 subfield of the rodent hippocampus, with interneuron debris phagocytosed by infiltrating microglia. SAVA microlesions did not affect VGLUT1+ excitatory afferents. Yet SAVA-induced rearrangement of the hippocampal circuitry triggered network hyperexcitability associated with the progressive loss of CA1 pyramidal cells and the dispersion of dentate granule cells. Overall, our data identifySAVAsas an effective tool to eliminate GABAergic neurons from neuronal circuits underpinning high-order behaviors and cognition, and whose manipulation can recapitulate pathogenic cascades of epilepsy and other neuropsychiatric illnesses. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
160. Selective activation of the transcription factor ATF6 mediates endoplasmic reticulum proliferation triggered by a membrane protein.
- Author
-
Maiuolo, Jessica, Bulotta, Stefania, Verderio, Claudia, Benfante, Roberta, and Borgese, Nica
- Subjects
ENDOPLASMIC reticulum ,TRANSCRIPTION factors ,MEMBRANE proteins ,CELL lines ,CELL proliferation ,LECITHIN - Abstract
It is well known that the endoplasmic reticulum (ER) is capable of expanding its surface area in response both to cargo load and to increased expression of resident membrane proteins. Although the response to increased cargo load, known as the unfolded protein response (UPR), is well characterized, the mechanism of the response to membrane protein load has been unclear. As a model system to investigate this phenomenon, we have used a HeLa-TetOff cell line inducibly expressing a tail-anchored construct consisting of an N-terminal cytosolic GFP moiety anchored to the ER membrane by the tail of cytochrome b5 [GFP-b(5)tail]. After removal of doxycycline, GFP-b(5)tail is expressed at moderate levels (1-2% of total ER protein) that, nevertheless, induce ER proliferation, as assessed both by EM and by a three- to fourfold increase in phosphatidylcholine synthesis. We investigated possible participation of each of the three arms of the UPR and found that only the activating transcription factor 6 (ATF6) arm was selectively activated after induction of GFP-b(5)tail expression; peak ATF6a activation preceded the increase in phosphatidylcholine synthesis. Surprisingly, up-regulation of known ATF6 target genes was not observed under these conditions. Silencing of ATF6a abolished the ER proliferation response, whereas knockdown of Ire1 was without effect. Because GFP-b(5)tail lacks a luminal domain, the response we observe is unlikely to originate from the ER lumen. Instead, we propose that a sensing mechanism operates within the lipid bilayer to trigger the selective activation of ATF6. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
161. Calcitonin Gene-Related Peptide-Mediated Enhancement of Purinergic Neuron/Glia Communication by the Algogenic Factor Bradykinin in Mouse Trigeminal Ganglia from Wild-Type and R192Q Cav2.1 Knock-In Mice: Implications for Basic Mechanisms of Migraine Pain
- Author
-
Ceruti, Stefania, Villa, Giovanni, Fumagalli, Marta, Colombo, Laura, Magni, Giulia, Zanardelli, Matteo, Fabbretti, Elsa, Verderio, Claudia, van den Maagdenberg, Arn M. J. M., Nistri, Andrea, and Abbracchio, Maria P.
- Subjects
CALCITONIN gene-related peptide ,BRADYKININ ,GANGLIA ,MIGRAINE ,GENETIC mutation ,LABORATORY mice - Abstract
Within the trigeminal ganglion, crosstalk between neurons and satellite glial cells (SGCs) contributes to neuronal sensitization and transduction of painful stimuli, including migraine pain, at least partly through activation of purinergic receptor mechanisms. We previously showed that the algogenic mediator bradykinin (BK) potentiates purinergic P2Y receptors on SGCs in primary trigeminal cultures. Our present study investigated the molecular basis of this effect in wild-type (WT) mice and CaV2.1α1 R192Q mutant knock-in (KI) mice expressing a human mutation causing familial hemiplegic migraine type 1. Single-cell calcium imaging ofWTcultures revealed functional BK receptors in neurons only, suggesting a paracrine action by BK to release a soluble mediator responsible for its effects on SGCs. We identified this mediator as the neuropeptide calcitonin gene-related peptide (CGRP), whose levels were markedly increased by BK, while the CGRP antagonist CGRP8-37 and the anti-migraine drug sumatriptan inhibited BK actions. Unlike CGRP, BK was ineffective in neuron-free SGC cultures, confirming the CGRP neuronal source. P2Y receptor potentiation induced by CGRP in SGCs was mediated via activation of the extracellular signal-regulated kinase 1/2 pathways, and after exposure to CGRP, a significant release of several cytokines was detected. Interestingly, both basal and BK-stimulated CGRP release was higher in KI mouse cultures, where BK significantly upregulated the number of SGCs showing functional UTP-sensitive P2Y receptors. Our findings suggest that P2Y receptors on glial cells might be considered as novel players in the cellular processes underlying migraine pathophysiology and might represent new targets for the development of innovative therapeutic agents against migraine pain. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
162. ATP in neuron–glia bidirectional signalling
- Author
-
Verderio, Claudia and Matteoli, Michela
- Subjects
- *
ADENOSINE triphosphate , *CELLULAR signal transduction , *NEUROTRANSMITTERS , *NEURONS , *ASTROCYTES , *NERVOUS system , *CELL populations - Abstract
Abstract: ATP accomplishes important roles in brain, where it functions as neurotransmitter or co-transmitter, being stored and released either as single mediator or together with other neuromodulators. In the last years, the purinergic system has emerged as the most relevant mechanism for intercellular signalling in the nervous system, affecting communication between many types of neurons and all types of glia. In this review, we will focus on recently reported data which describe the role of ATP in bidirectional signalling between neurons and different populations of glial cells, in both peripheral and central system. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
163. Intrinsic calcium dynamics control botulinum toxin A susceptibility in distinct neuronal populations.
- Author
-
Grumelli, Carlotta, Corradini, Irene, Matteoli, Michela, and Verderio, Claudia
- Subjects
BOTULINUM toxin ,NEURAL physiology ,EXOCYTOSIS ,CALCIUM channels ,NEUROMUSCULAR depolarizing agents ,GABA ,SYNAPTIC vesicles ,CELL populations - Abstract
Abstract: SNAP-25 is a SNARE protein implicated in exocytosis and in the negative modulation of voltage-gated calcium channels. We have previously shown that GABAergic synapses, which express SNAP-25 at much lower levels relative to glutamatergic ones, are characterized by a higher calcium responsiveness to depolarization and are largely resistant to botulinum toxin A. We show here that silencing of SNAP-25 in glutamatergic neurons, a procedure which increases KCl-induced calcium elevations, confers these synapses with toxin resistance. Since it is known that calcium reverts the efficacy of botulinum A, we investigated whether the lower effectiveness of the toxin in inhibiting GABAergic vesicle cycling might be attributable to higher evoked calcium transients of inhibitory neurons. We demonstrate that either expression of SNAP-25
1–197 or BAPTA/AM treatment, both inhibiting calcium dynamics, facilitate block of GABAergic vesicle exocytosis upon toxin treatment. These data indicate that intrinsic calcium dynamics control botulinum A susceptibility in distinct neuronal populations. [Copyright &y& Elsevier]- Published
- 2010
- Full Text
- View/download PDF
164. Different properties of P2X7 receptor in hippocampal and cortical astrocytes.
- Author
-
Bianco, Fabio, Colombo, Alessio, Saglietti, Laura, Lecca, Davide, Abbracchio, Maria, Matteoli, Michela, and Verderio, Claudia
- Abstract
P2X
7 receptor is a ligand-gated ion channel, which can induce the opening of large membrane pores. Here, we provide evidence that the receptor induces pore formation in astrocytes cultured from cortex, but not from the hippocampus. Furthermore, P2X7 receptor activation promptly induces p38 mitogen-activated protein kinase (MAPK) phosphorylation in cortical but not in hippocampal astrocytes. Given the role of p38 MAPK activation in pore opening, these data suggest that defective coupling of the receptor to the enzyme could occur in hippocampal cultures. The different capabilities of the receptor to open membrane pores cause relevant functional consequences. Upon pore formation, caspase-1 is activated and pro-IL1-β is cleaved and released extracellularly. The receptor stimulation does not result in interleukin-1beta secretion from hippocampal astrocytes, although the pro-cytokine is present in the cytosol of lipopolysaccharide-primed cultures. These results open the possibility that activation of P2X7 receptors differently influences the neuroinflammatory processes in distinct brain regions. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
165. Plasma Small Extracellular Vesicles with Complement Alterations in GRN / C9orf72 and Sporadic Frontotemporal Lobar Degeneration.
- Author
-
Bellini, Sonia, Saraceno, Claudia, Benussi, Luisa, Squitti, Rosanna, Cimini, Sara, Ricci, Martina, Canafoglia, Laura, Coppola, Cinzia, Puoti, Gianfranco, Ferrari, Clarissa, Longobardi, Antonio, Nicsanu, Roland, Lombardi, Marta, D'Arrigo, Giulia, Verderio, Claudia, Binetti, Giuliano, Rossi, Giacomina, and Ghidoni, Roberta
- Subjects
FRONTOTEMPORAL lobar degeneration ,EXTRACELLULAR vesicles ,EXOSOMES ,NEURONAL ceroid-lipofuscinosis - Abstract
Cutting-edge research suggests endosomal/immune dysregulation in GRN/C9orf72-associated frontotemporal lobar degeneration (FTLD). In this retrospective study, we investigated plasma small extracellular vesicles (sEVs) and complement proteins in 172 subjects (40 Sporadic FTLD, 40 Intermediate/Pathological C9orf72 expansion carriers, and 49 Heterozygous/Homozygous GRN mutation carriers, 43 controls). Plasma sEVs (concentration, size) were analyzed by nanoparticle tracking analysis; plasma and sEVs C1q, C4, C3 proteins were quantified by multiplex assay. We demonstrated that genetic/sporadic FTLD share lower sEV concentrations and higher sEV sizes. The diagnostic performance of the two most predictive variables (sEV concentration/size ratio) was high (AUC = 0.91, sensitivity 85.3%, specificity 81.4%). C1q, C4, and C3 cargo per sEV is increased in genetic and sporadic FTLD. C4 (cargo per sEV, total sEV concentration) is increased in Sporadic FTLD and reduced in GRN+ Homozygous, suggesting its specific unbalance compared with Heterozygous cases. C3 plasma level was increased in genetic vs. sporadic FTLD. Looking at complement protein compartmentalization, in control subjects, the C3 and C4 sEV concentrations were roughly half that in respect to those measured in plasma; interestingly, this compartmentalization was altered in different ways in patients. These results suggest sEVs and complement proteins as potential therapeutic targets to mitigate neurodegeneration in FTLD. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
166. Activity-dependent phosphorylation of Ser187 is required for SNAP-25-negative modulation of neuronal voltage-gated calcium channels.
- Author
-
Pozzi, Davide, Condliffe, Steven, Bozzi, Yuri, Chikhiadze, Maia, Grumelli, Carlotta, Proux-Gillardeaux, Véronique, Takahashiti, Masami, Franceschetti, Silvana, Verderio, Claudia, and Matteoli, Michela
- Subjects
PHOSPHORYLATION ,CALCIUM channels ,NEURAL transmission ,NEUROPLASTICITY ,LABORATORY mice ,NEURONS ,NEUROSCIENCES - Abstract
Synaptosomal-associated protein of 25 kDa (SNAP-25) is a SNARE protein that regulates neurotransmission by the formation of a complex with syntaxin 1 and synaptobrevin/VAMP2. SNAP-25 also reduces neuronal calcium responses to stimuli, but neither the functional relevance nor the molecular mechanisms of this modulation have been clarified. In this study, we demonstrate that hippocampal slices from Snap25
+/- mice display a significantly larger facilitation and that higher calcium peaks are reached after depolarization by Snap25-/- and Snap25+/- cultured neurons compared with wild type. We also show that SNAP-25b modulates calcium dynamics by inhibiting voltage-gated calcium channels (VGCCs) and that PKC phosphorylation of SNAP-25 at ser187 is essential for this process, as indicated by the use of phosphomimetic ($187E) or nonphosphorylated ($187A) mutants. Neuronal activity is the trigger that induces the transient phosphorylation of SNAP-25 at ser187. Indeed, enhancement of network activity increases the levels of phosphorylated SNAP-25, whereas network inhibition reduces the extent of protein phosphorylation. A transient peak of SNAP-25 phosphorylation also is detectable in rat hippocampus in vivo after i.p. injection with kainate to induce seizures. These findings demonstrate that differences in the expression levels of SNAP-25 impact on calcium dynamics and neuronal plasticity, and that SNAP-25 phosphorylation, by promoting inhibition of VGCCs, may mediate a negative feedback modulation of neuronal activity during intense activation. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
167. A role for P2X7 in microglial proliferation.
- Author
-
Bianco, Fabio, Ceruti, Stefania, Colombo, Alessio, Fumagalli, Marta, Ferrari, Davide, Pizzirani, Cinzia, Matteoli, Michela, Di Virgilio, Francesco, Abbracchio, Maria P., and Verderio, Claudia
- Subjects
MICROGLIA ,NEUROGLIA ,ENDOTOXINS ,BACTERIAL diseases ,RNA ,CELL proliferation ,ADENOSINE triphosphate - Abstract
Microglia, glial cells with an immunocompetent role in the CNS, react to stimuli from the surrounding environment with alterations of their phenotypic response. Amongst other activating signals, the endotoxin lipopolysaccharide (LPS) is widely used as a tool to mimic bacterial infection in the CNS. LPS-activated microglia undergo dramatic changes in cell morphology/activity; in particular, they stop proliferating and differentiate from resting to effector cells. Activated microglia also show modifications of purinoreceptor signalling with a significant decrease in P2X
7 expression. In this study, we demonstrate that the down-regulation of the P2X7 receptor in activated microglia may play an important role in the antiproliferative effect of LPS. Indeed, chronic blockade of the P2X7 receptor by antagonists (oxidized ATP, KN62 and Brilliant Blue G), or treatment with the ATP-hydrolase apyrase, severely decreases microglial proliferation, down-regulation of P2X7 receptor expression by small RNA interference (siRNA) decreases cell proliferation, and the proliferation of P2X7 -deficient N9 clones and primary microglia, in which P2X7 expression is down-regulated by siRNA, is unaffected by either LPS or P2X7 antagonists. Furthermore, flow cytometric analysis indicates that exposure to oxidized ATP or treatment with LPS reversibly decreases cell cycle progression, without increasing the percentage of apoptotic cells. Overall, our data show that the P2X7 receptor plays an important role in controlling microglial proliferation by supporting cell cycle progression. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
168. Activated macrophages release microvesicles containing polarized M1 or M2 mRNAs
- Author
-
Garzetti, Livia, Menon, Ramesh, Finardi, Annamaria, Bergami, Alessandra, Sica, Antonio, Martino, Gianvito, Comi, Giancarlo, Verderio, Claudia, Farina, Cinthia, and Furlan, Roberto
- Abstract
Microvesicles released by activated macrophages, and detected in biological fluids, may constitute a valuable biomarker to identify myeloid cell activation phenotype during disease. MVs are known vehicles of horizontal communication among cells, currently under scrutiny as powerful biomarkers in several pathological processes. The potential advantage of MVs relies on the assumption that their content reflects processes ongoing in pathologically relevant cell types. We have described that MVs of myeloid origin in the CSF are a marker of microglia/macrophage activation. Myeloid cells have different activation types, resulting in diverse functional phenotypes. Knowledge on the activation type of myeloid cells during disease would be of paramount importance for the understanding of ongoing pathogenic processes. We show here that macrophages activated in vitro in different ways all release increased amounts of MVs compared with NS cells. Moreover, we show that macrophage‐derived MVs contain a repertoire of mRNAs that is not the result of casual sampling from the parental cells, as it is characterized by distinct mRNA enrichments and species. Nevertheless, mRNA content of MVs clearly allows identification in vivo of the activated phenotype of the cell of origin, indicating carryover of functional macrophage traits. We propose that detection of mRNAs in myeloid MVs permits identification of myeloid cell activation type during disease, allowing for further stratification of pathological processes.
- Published
- 2014
- Full Text
- View/download PDF
169. Role of extracellular vesicles in early synaptic dysfunction in AD.
- Author
-
Gabrielli, Martina, D'Arrigo, Giulia, Falcicchia, Chiara, Origlia, Nicola, Arancio, Ottavio, Verderio, Claudia, and Prada, Ilaria
- Abstract
Background: Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with amyloid‐β (Aβ) and tau protein accumulation. Synaptic dysfunction is an early mechanism in AD which involves progressively larger areas of the brain over time. However how synaptic dysfunction starts and propagates is unknown. The hypothesis we are testing is that large extracellular vesicles (EVs) released by microglia exposed to and carrying Aβ42 (Aβ‐EVs) may be responsible for these early events in AD. Method: Combining optical manipulation and time lapse imaging to place single large EVs on RFP‐positive cultured neuron dendrites or axons, we tested both their effects on the synapse and the dynamics of their interaction with neurons compared to ctrl‐EVs (released by microglia not exposed to Aβ42). Then, ctrl‐EVs, Aβ‐EVs or large EVs released by microglia exposed to CHO7PA2 cell supernatant containing naturally secreted Aβ oligomers (CHO‐EVs) were stereotaxically injected into the mouse entorhinal cortex (EC), one of the most vulnerable regions in AD, and long‐term potentiation (LTP) was measured in the EC and in his main target region, the dentate gyrus of the hippocampus (DG), through field potential extracellular recordings in cortico‐hippocampal brain slices. Result: Aβ‐EVs rapidly altered dendritic spine morphology, locally at the site of interaction, increasing the number of immature protrusions. After contact to the neuronal surface, Aβ‐EVs were able to move along the axonal surface more efficiently than ctrl‐EVs and predominantly in an anterograde direction. Furthermore, 1h after injection in the EC, LTP was impaired in the EC of brains injected with AβEVs, but not ctrl‐EVs. While 24h after AβEV injection, LTP was impaired also in the DG, indicating a spreading of synaptic dysfunction between the two connected regions. Importantly, we could reproduce these results by injection of CHO‐EVs. Conclusion: Our data indicate that Aβ‐EVs affect the synapse both in vitro and in vivo, and are able to propagate synaptic dysfunction among synaptically connected regions in vivo, providing evidence of the involvement of microglial large EVs in the onset and propagation of early synaptic dysfunction in AD, thus paving the way for novel therapeutic strategies. Funding: NIH1R56AG056108‐01 to OA and CV 2018‐AARF‐588984 to IP. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
170. Synaptic vesicle endocytosis mediates the entry of tetanus neurotoxin into hippocampal neurons.
- Author
-
Matteoli, Michela and Verderio, Claudia
- Subjects
- *
TETANUS toxin , *PHYSIOLOGY - Abstract
Shows that the tetanus neurotoxin (TeNT) enters hippocampal neurons by means of the physiological process of synaptic vesicle (SV) recycling and that vesicle acidification is essential for the toxin translocation in the cytosol. TeNT is internalized at nerve terminals; TeNT uptake in developing hippocampal neurons; TeNT is internalized inside small SV.
- Published
- 1996
- Full Text
- View/download PDF
171. Spatial changes in calcium signaling during the establishment of neuronal polarity and...
- Author
-
Verderio, Claudia and Coco, Silvia
- Subjects
- *
NEURON development , *NEUROTRANSMITTER receptors - Abstract
Studies the possible changes in glutamate receptor distribution during development and synaptogenesis of hippocampal neurons in culture. Use of calcium imaging approach; Parallel redistribution during the establishment of neuronal polarity; Synapse formation.
- Published
- 1994
- Full Text
- View/download PDF
172. The role of glial cells in synaptic function.
- Author
-
Bacci, Alberto, Verderio, Claudia, Pravettoni, Elena, and Matteoli, Michela
- Published
- 1999
- Full Text
- View/download PDF
173. Emerging Roles of Extracellular Vesicles in Alzheimer's Disease: Focus on Synaptic Dysfunction and Vesicle–Neuron Interaction.
- Author
-
Gabrielli, Martina, Tozzi, Francesca, Verderio, Claudia, and Origlia, Nicola
- Subjects
- *
ALZHEIMER'S disease , *EXTRACELLULAR vesicles , *TAU proteins - Abstract
Alzheimer's disease (AD) is considered by many to be a synaptic failure. Synaptic function is in fact deeply affected in the very early disease phases and recognized as the main cause of AD-related cognitive impairment. While the reciprocal involvement of amyloid beta (Aβ) and tau peptides in these processes is under intense investigation, the crucial role of extracellular vesicles (EVs) released by different brain cells as vehicles for these molecules and as mediators of early synaptic alterations is gaining more and more ground in the field. In this review, we will summarize the current literature on the contribution of EVs derived from distinct brain cells to neuronal alterations and build a working model for EV-mediated propagation of synaptic dysfunction in early AD. A deeper understanding of EV–neuron interaction will provide useful targets for the development of novel therapeutic approaches aimed at hampering AD progression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
174. Human Umbilical Cord-Mesenchymal Stem Cells Promote Extracellular Matrix Remodeling in Microglia.
- Author
-
Lombardo, Marta Tiffany, Gabrielli, Martina, Julien-Marsollier, Florence, Faivre, Valérie, Le Charpentier, Tifenn, Bokobza, Cindy, D'Aliberti, Deborah, Pelizzi, Nicola, Halimi, Camilla, Spinelli, Silvia, Van Steenwinckel, Juliette, Verderio, Elisabetta A. M., Gressens, Pierre, Piazza, Rocco, and Verderio, Claudia
- Subjects
- *
MESENCHYMAL stem cells , *HUMAN stem cells , *EXTRACELLULAR matrix , *STEM cells , *CELL migration - Abstract
Human mesenchymal stem cells modulate the immune response and are good candidates for cell therapy in neuroinflammatory brain disorders affecting both adult and premature infants. Recent evidence indicates that through their secretome, mesenchymal stem cells direct microglia, brain-resident immune cells, toward pro-regenerative functions, but the mechanisms underlying microglial phenotypic transition are still under investigation. Using an in vitro coculture approach combined with transcriptomic analysis, we identified the extracellular matrix as the most relevant pathway altered by the human mesenchymal stem cell secretome in the response of microglia to inflammatory cytokines. We confirmed extracellular matrix remodeling in microglia exposed to the mesenchymal stem cell secretome via immunofluorescence analysis of the matrix component fibronectin and the extracellular crosslinking enzyme transglutaminase-2. Furthermore, an analysis of hallmark microglial functions revealed that changes in the extracellular matrix enhance ruffle formation by microglia and cell motility. These findings point to extracellular matrix changes, associated plasma membrane remodeling, and enhanced microglial migration as novel mechanisms by which mesenchymal stem cells contribute to the pro-regenerative microglial transition. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
175. Microglial vesicles improve post-stroke recovery by preventing immune cell senescence and favoring oligodendrogenesis
- Author
-
Raffaele, Stefano, Gelosa, Paolo, Bonfanti, Elisabetta, Lombardi, Marta, Castiglioni, Laura, Cimino, Mauro, Sironi, Luigi, Abbracchio, Maria P., Verderio, Claudia, and Fumagalli, Marta
- Abstract
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 vitroexperiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.
- Published
- 2021
- Full Text
- View/download PDF
176. Epileptiform Activity and Cognitive Deficits in SNAP-25+/− Mice are Normalized by Antiepileptic Drugs
- Author
-
Corradini, Irene, Donzelli, Andrea, Antonucci, Flavia, Welzl, Hans, Loos, Maarten, Martucci, Roberta, De Astis, Silvia, Pattini, Linda, Inverardi, Francesca, Wolfer, David Paul, Caleo, Matteo, Bozzi, Yuri, Verderio, Claudia, Frassoni, Carolina, Braida, Daniela, Clerici, Mario, Lipp, Hans-Peter, Sala, Mariaelvina, and Matteoli, Michela
- Subjects
Valproate ,Epilepsy ,Memory ,SNAP-25 ,3. Good health - Abstract
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., Cerebral Cortex, 24 (2), ISSN:1047-3211, ISSN:1460-2199
177. Ectonucleotidase activity and immunosuppression in astrocyte-CD4 T cell bidirectional signaling
- Author
-
Filipello, Fabia, Pozzi, Davide, Proietti, Michele, Romagnani, Andrea, Mazzitelli, Sonia, Matteoli, Michela, Verderio, Claudia, Grassi, Fabio, Filipello, Fabia, Pozzi, Davide, Proietti, Michele, Romagnani, Andrea, Mazzitelli, Sonia, Matteoli, Michela, Verderio, Claudia, and Grassi, Fabio
178. Epileptiform Activity and Cognitive Deficits in SNAP-25+/− Mice are Normalized by Antiepileptic Drugs
- Author
-
Corradini, Irene, Donzelli, Andrea, Antonucci, Flavia, Welzl, Hans, Loos, Maarten, Martucci, Roberta, De Astis, Silvia, Pattini, Linda, Inverardi, Francesca, Wolfer, David, Caleo, Matteo, Bozzi, Yuri, Verderio, Claudia, Frassoni, Carolina, Braida, Daniela, Clerici, Mario, Lipp, Hans-Peter, Sala, Mariaelvina, Matteoli, Michela, Corradini, Irene, Donzelli, Andrea, Antonucci, Flavia, Welzl, Hans, Loos, Maarten, Martucci, Roberta, De Astis, Silvia, Pattini, Linda, Inverardi, Francesca, Wolfer, David, Caleo, Matteo, Bozzi, Yuri, Verderio, Claudia, Frassoni, Carolina, Braida, Daniela, Clerici, Mario, Lipp, Hans-Peter, Sala, Mariaelvina, and Matteoli, Michela
- Abstract
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
179. TRPV1 channels are critical brain inflammation detectors and neuropathic pain biomarkers in mice.
- Author
-
Marrone, Maria Cristina, Morabito, Annunziato, Giustizieri, Michela, Chiurchiù, Valerio, Leuti, Alessandro, Mattioli, Marzia, Marinelli, Sara, Riganti, Loredana, Lombardi, Marta, Murana, Emanuele, Totaro, Antonio, Piomelli, Daniele, Ragozzino, Davide, Oddi, Sergio, Maccarrone, Mauro, Verderio, Claudia, and Marinelli, Silvia
- Abstract
The capsaicin receptor TRPV1 has been widely characterized in the sensory system as a key component of pain and inflammation. A large amount of evidence shows that TRPV1 is also functional in the brain although its role is still debated. Here we report that TRPV1 is highly expressed in microglial cells rather than neurons of the anterior cingulate cortex and other brain areas. We found that stimulation of microglial TRPV1 controls cortical microglia activation per se and indirectly enhances glutamatergic transmission in neurons by promoting extracellular microglial microvesicles shedding. Conversely, in the cortex of mice suffering from neuropathic pain, TRPV1 is also present in neurons affecting their intrinsic electrical properties and synaptic strength. Altogether, these findings identify brain TRPV1 as potential detector of harmful stimuli and a key player of microglia to neuron communication. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
180. Microglial large extracellular vesicles propagate early synaptic dysfunction in Alzheimer's disease.
- Author
-
Gabrielli, Martina, Prada, Ilaria, Joshi, Pooja, Falcicchia, Chiara, D'Arrigo, Giulia, Rutigliano, Grazia, Battocchio, Elisabetta, Zenatelli, Rossella, Tozzi, Francesca, Radeghieri, Annalisa, Arancio, Ottavio, Origlia, Nicola, and Verderio, Claudia
- Abstract
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-β released by microglia in association with large extracellular vesicles (Aβ-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 Aβ-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 Aβ naturally secreted by CHO7PA2 cells, while neither Aβ42 alone nor inflammatory extracellular vesicles devoid of Aβ were able to propagate long-term potentiation impairment. Using optical tweezers combined to time-lapse imaging to study Aβ-EV-neuron interaction, we show that Aβ-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aβ-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. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
181. Mutant PrP Suppresses Glutamatergic Neurotransmission in Cerebellar Granule Neurons by Impairing Membrane Delivery of VGCC α2δ-1 Subunit
- Author
-
Senatore, Assunta, Colleoni, Simona, Verderio, Claudia, Restelli, Elena, Morini, Raffaella, Condliffe, Steven B., Bertani, Ilaria, Mantovani, Susanna, Canovi, Mara, Micotti, Edoardo, Forloni, Gianluigi, Dolphin, Annette C., Matteoli, Michela, Gobbi, Marco, and Chiesa, Roberto
- Subjects
- *
NEURAL transmission , *GLUTAMIC acid , *NEURONS , *NEURODEGENERATION , *BEHAVIOR disorders , *EXOCYTOSIS - Abstract
Summary: How mutant prion protein (PrP) leads to neurological dysfunction in genetic prion diseases is unknown. Tg(PG14) mice synthesize a misfolded mutant PrP which is partially retained in the neuronal endoplasmic reticulum (ER). As these mice age, they develop ataxia and massive degeneration of cerebellar granule neurons (CGNs). Here, we report that motor behavioral deficits in Tg(PG14) mice emerge before neurodegeneration and are associated with defective glutamate exocytosis from granule neurons due to impaired calcium dynamics. We found that mutant PrP interacts with the voltage-gated calcium channel α2δ-1 subunit, which promotes the anterograde trafficking of the channel. Owing to ER retention of mutant PrP, α2δ-1 accumulates intracellularly, impairing delivery of the channel complex to the cell surface. Thus, mutant PrP disrupts cerebellar glutamatergic neurotransmission by reducing the number of functional channels in CGNs. These results link intracellular PrP retention to synaptic dysfunction, indicating new modalities of neurotoxicity and potential therapeutic strategies. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
182. miR-150-5p and let-7b-5p in Blood Myeloid Extracellular Vesicles Track Cognitive Symptoms in Patients with Multiple Sclerosis.
- Author
-
Scaroni, Federica, Visconte, Caterina, Serpente, Maria, Golia, Maria Teresa, Gabrielli, Martina, Huiskamp, Marijn, Hulst, Hanneke E., Carandini, Tiziana, De Riz, Milena, Pietroboni, Anna, Rotondo, Emanuela, Scarpini, Elio, Galimberti, Daniela, Teunissen, Charlotte E., van Dam, Maureen, de Jong, Brigit A., Fenoglio, Chiara, and Verderio, Claudia
- Subjects
- *
EXTRACELLULAR vesicles , *MULTIPLE sclerosis , *VESICLES (Cytology) , *SYMPTOMS , *GENE targeting , *DISEASE progression - Abstract
Cognitive deficits strongly affect the quality of life of patients with multiple sclerosis (MS). However, no cognitive MS biomarkers are currently available. Extracellular vesicles (EVs) contain markers of parental cells and are able to pass from the brain into blood, representing a source of disease biomarkers. The aim of this study was to investigate whether small non-coding microRNAs (miRNAs) targeting synaptic genes and packaged in plasma EVs may reflect cognitive deficits in MS patients. Total EVs were precipitated by Exoquick from the plasma of twenty-six cognitively preserved (CP) and twenty-three cognitively impaired (CI) MS patients belonging to two independent cohorts. Myeloid EVs were extracted by affinity capture from total EVs using Isolectin B4 (IB4). Fourteen miRNAs targeting synaptic genes were selected and measured by RT-PCR in both total and myeloid EVs. Myeloid EVs from CI patients expressed higher levels of miR-150-5p and lower levels of let-7b-5p compared to CP patients. Stratification for progressive MS (PMS) and relapsing-remitting MS (RRMS) and correlation with clinical parameters suggested that these alterations might be attributable to cognitive deficits rather than disease progression. This study identifies miR-150-5p and let-7b-5p packaged in blood myeloid EVs as possible biomarkers for cognitive deficits in MS. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
183. Towards bio-compatible magnetic nanoparticles: Immune-related effects, in-vitro internalization, and in-vivo bio-distribution of zwitterionic ferrite nanoparticles with unexpected renal clearance.
- Author
-
Ferretti, Anna M., Usseglio, Sandro, Mondini, Sara, Drago, Carmelo, La Mattina, Rosa, Chini, Bice, Verderio, Claudia, Leonzino, Marianna, Cagnoli, Cinzia, Joshi, Pooja, Boraschi, Diana, Italiani, Paola, Li, Yang, Swartzwelter, Benjamin J., Sironi, Luigi, Gelosa, Paolo, Castiglioni, Laura, Guerrini, Uliano, and Ponti, Alessandro
- Subjects
- *
NANOPARTICLES , *MAGNETIC nanoparticles , *NANOPARTICLE size , *RETICULO-endothelial system , *INFLAMMATION , *MAGNETIC properties - Abstract
Iron oxide and other ferrite nanoparticles have not yet found widespread application in the medical field since the translation process faces several big hurdles. The incomplete knowledge of the interactions between nanoparticles and living organisms is an unfavorable factor. This complex subject should be made simpler by synthesizing magnetic nanoparticles with good physical (relaxivity) and chemical (colloidal stability, anti-fouling) properties and no biological activity (no immune-related effects, minimal internalization, fast clearance). Such an innocent scaffold is the main aim of the present paper. We systematically searched for it within the class of small-to-medium size ferrite nanoparticles coated by small (zwitter)ionic ligands. Once established, it can be functionalized to achieve targeting, drug delivery, etc. and the observed biological effects will be traced back to the functional molecules only, as the nanosized scaffold is innocent. We synthesized nine types of magnetic nanoparticles by systematic variation of core composition, size, coating. We investigated their physico-chemical properties and interaction with serum proteins, phagocytic microglial cells, and a human model of inflammation and studied their biodistribution and clearance in healthy mice. The nanoparticles have good magnetic properties and their surface charge is determined by the preferential adsorption of anions. All nanoparticle types can be considered as immunologically safe, an indispensable pre-requisite for medical applications in humans. All but one type display low internalization by microglial BV2 cells, a process strongly affected by the nanoparticle size. Both small (3 nm) and medium size (11 nm) zwitterionic nanoparticles are in part captured by the mononuclear phagocyte system (liver and spleen) and in part rapidly (≈1 h) excreted through the urinary system of mice. The latter result questions the universality of the accepted size threshold for the renal clearance of nanoparticles (5.5 nm). We suggest that it depends on the nature of the circulating particles. Renal filterability of medium-size magnetic nanoparticles is appealing because they share with small nanoparticles the decreased accumulation-related toxicity while performing better as magnetic diagnostic/therapeutic agents thanks to their larger magnetic moment. In conclusion, many of our nanoparticle types are a bio-compatible innocent scaffold with unexpectedly favorable clearance. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
184. Immune and central nervous system-related miRNAs expression profiling in monocytes of multiple sclerosis patients.
- Author
-
Amoruso, Antonella, Blonda, Maria, Gironi, Maira, Grasso, Roberta, Di Francescantonio, Valeria, Scaroni, Federica, Furlan, Roberto, Verderio, Claudia, and Avolio, Carlo
- Subjects
- *
IMMUNE system , *CENTRAL nervous system , *MICRORNA , *GENE expression , *MONOCYTES , *MULTIPLE sclerosis - Abstract
It is widely recognized that monocytes-macrophages adopt a wide variety of phenotypes, influencing the inflammatory activity and demyelination in Multiple Sclerosis (MS). However, how the phenotype of human monocytes evolves in the course of MS is largely unknown. The aim of our preliminary study was to analyse in monocytes of relapsing-remitting and progressive forms of MS patients the expression of a set of miRNAs which impact monocyte-macrophage immune function and their communication with brain cells. Quantitative PCR showed that miRNAs with anti-inflammatory functions, which promote pro-regenerative polarization, are increased in MS patients, while pro-inflammatory miR-155 is downregulated in the same patients. These changes may indicate the attempt of monocytes to counteract neuroinflammation. miR-124, an anti-inflammatory marker but also of myeloid cell quiescence was strongly downregulated, especially in progressive MS patients, suggesting complete loss of homeostatic monocyte function in the progressive disease phase. Profiling of miRNAs that control monocyte polarization may help to define not only the activation state of monocytes in the course of the disease but also novel pathogenic mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
185. Detrimental and protective action of microglial extracellular vesicles on myelin lesions: astrocyte involvement in remyelination failure.
- Author
-
Lombardi, Marta, Parolisi, Roberta, Scaroni, Federica, Bonfanti, Elisabetta, Gualerzi, Alice, Gabrielli, Martina, Kerlero de Rosbo, Nicole, Uccelli, Antonio, Giussani, Paola, Viani, Paola, Garlanda, Cecilia, Abbracchio, Maria P., Chaabane, Linda, Buffo, Annalisa, Fumagalli, Marta, and Verderio, Claudia
- Subjects
- *
OLIGODENDROGLIA , *ASTROCYTES , *MICROGLIA , *SPHINGOSINE-1-phosphate , *MESENCHYMAL stem cells , *CORPUS callosum , *MYELIN proteins , *NEUROLOGICAL disorders - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
186. Microglial extracellular vesicles induce Alzheimer’s disease-related cortico-hippocampal network dysfunction
- Author
-
Chiara Falcicchia, Francesca Tozzi, Martina Gabrielli, Stefano Amoretti, Greta Masini, Gabriele Nardi, Stefano Guglielmo, Gian Michele Ratto, Ottavio Arancio, Claudia Verderio, Nicola Origlia, Falcicchia, Chiara, Tozzi, Francesca, Gabrielli, Martina, Amoretti, Stefano, Masini, Greta, Nardi, Gabriele, Guglielmo, Stefano, Michele Ratto, Gian, Arancio, Ottavio, Verderio, Claudia, and Origlia, Nicola
- Subjects
Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,extracellular vesicles, entorhinal cortex, Alzheimer’s disease, microglia, cortical-hippocampal network ,Neurology ,Settore BIO/09 - Fisiologia ,Biological Psychiatry - Abstract
β-Amyloid is one of the main pathological hallmarks of Alzheimer’s disease and plays a major role in synaptic dysfunction. It has been demonstrated that β-amyloid can elicit aberrant excitatory activity in cortical-hippocampal networks, which is associated with behavioral abnormalities. However, the mechanism of the spreading of β-amyloid action within a specific circuitry has not been elucidated yet. We have previously demonstrated that the motion of microglia-derived large extracellular vesicles carrying β-amyloid, at the neuronal surface, is crucial for the initiation and propagation of synaptic dysfunction along the entorhinal-hippocampal circuit. Here, using chronic EEG recordings, we show that a single injection of extracellular vesicles carrying β-amyloid into the mouse entorhinal cortex could trigger alterations in the cortical and hippocampal activity that are reminiscent of those found in Alzheimer’s disease mouse models and human patients. The development of EEG abnormalities was associated with progressive memory impairment as assessed by an associative (object-place context recognition) and non-associative (object recognition) task. Importantly, when the motility of extracellular vesicles, carrying β-amyloid, was inhibited, the effect on network stability and memory function was significantly reduced. Our model proposes a new biological mechanism based on the extracellular vesicles mediated progression of β-amyloid pathology and offers the opportunity to test pharmacological treatments targeting the early stages of Alzheimer’s disease.
- Published
- 2023
- Full Text
- View/download PDF
187. Effect of fingolimod action on the release of monocyte-derived microvesicles in multiple sclerosis patients.
- Author
-
Amoruso, Antonella, Blonda, Maria, D'arrigo, Giulia, Grasso, Roberta, Di Francescantonio, Valeria, Verderio, Claudia, and Avolio, Carlo
- Subjects
- *
FINGOLIMOD , *MULTIPLE sclerosis , *MONOCYTES , *SPHINGOMYELINASE , *BIOLOGICAL tags , *PATIENTS , *THERAPEUTICS - Abstract
Abstract Recently, microvesicles (MVs) were considered as important mediators of intercellular communication, especially in pathological conditions as Multiple Sclerosis (MS). In myeloid cells, MV shedding is induced by the receptor P2X7 with the involvement of acid sphingomyelinase (A-SMase) and release of the IL-1β. In this study we evaluate how Fingolimod affects MVs production by the monocytes, as well as P2X7R, IL-1β expression and A-SMase activity. Treatment decreased MVs production and IL-1β expression. This effect was associated with the inhibition of A-SMase activity in BzATP-stimulated monocytes from MS patients. These evidences suggest monocyte MVs as a possible disease and drug-efficacy biomarkers. Graphical abstract Unlabelled Image Highlights • Fingolimod reduced MVs production. • Fingolimod affected IL-1β gene expression. • Fingolimod modulated P2X7R gene expression. • Fingolimod reduced A-SMase activity but had no effect on A-SMase gene expression. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
188. Glia-to-neuron transfer of miRNAs via extracellular vesicles: a new mechanism underlying inflammation-induced synaptic alterations.
- Author
-
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, and Verderio, Claudia
- Subjects
- *
MICRORNA , *MICROGLIA - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
189. Exosomal cellular prion protein drives fibrillization of amyloid beta and counteracts amyloid beta-mediated neurotoxicity.
- Author
-
Falker, Clemens, Hartmann, Alexander, Guett, Inga, Dohler, Frank, Altmeppen, Hermann, Betzel, Christian, Schubert, Robin, Thurm, Dana, Wegwitz, Florian, Joshi, Pooja, Verderio, Claudia, Krasemann, Susanne, and Glatzel, Markus
- Subjects
- *
PRIONS , *AMYLOID beta-protein , *NEUROTOXICOLOGY , *ALZHEIMER'S disease , *NEUROBLASTOMA - Abstract
Alzheimer's disease is a common neurodegenerative, progressive, and fatal disorder. Generation and deposition of amyloid beta (Aβ) peptides associate with its pathogenesis and small soluble Aβ oligomers show the most pronounced neurotoxic effects and correlate with disease initiation and progression. Recent findings showed that Aβ oligomers bind to the cellular prion protein (PrPC) 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 Aβ plaque formation, decrease Aβ-mediated synaptotoxicity, and enhance Aβ clearance. Here, we explore how exosomal PrPC connects to protective functions attributed to exosomes in Alzheimer's disease. To achieve this, we generated a mouse neuroblastoma PrPC knockout cell line using transcription activator-like effector nucleases. Using these, as well as SH-SY5Y human neuroblastoma cells, we show that PrPC is highly enriched on exosomes and that exosomes bind amyloid beta via PrPC. Exosomes showed highest binding affinity for dimeric, pentameric, and oligomeric Aβ species. Thioflavin T assays revealed that exosomal PrPC accelerates fibrillization of amyloid beta, thereby reducing neurotoxic effects imparted by oligomeric Aβ. Our study provides further evidence for a protective role of exosomes in Aβ-mediated neurodegeneration and highlights the importance of exosomal PrPC in molecular mechanisms of Alzheimer's disease. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
190. Extracellular transglutaminase-2, nude or associated with astrocytic extracellular vesicles, modulates neuronal calcium homeostasis.
- Author
-
Tonoli, Elisa, Verduci, Ivan, Gabrielli, Martina, Prada, Ilaria, Forcaia, Greta, Coveney, Clare, Savoca, Maria Pia, Boocock, David J., Sancini, Giulio, Mazzanti, Michele, Verderio, Claudia, and Verderio, Elisabetta A.M.
- Subjects
- *
EXTRACELLULAR vesicles , *ENCEPHALITIS , *CALCIUM , *NUDITY , *HOMEOSTASIS - Abstract
We have uncovered a novel role for astrocytes-derived extracellular vesicles (EVs) in controlling intraneuronal Ca2+ concentration ([Ca2+] i) and identified transglutaminase-2 (TG2) as a surface-cargo of astrocytes-derived EVs. Incubation of hippocampal neurons with primed astrocyte-derived EVs have led to an increase in [Ca2+] i , unlike EVs from TG2-knockout astrocytes. Exposure of neurons or brain slices to extracellular TG2 promoted a [Ca2+] i rise, which was reversible upon TG2 removal and was dependent on Ca2+ influx through the plasma membrane. Patch-clamp and calcium imaging recordings revealed TG2-dependent neuronal membrane depolarization and activation of inward currents, due to the Na+/Ca2+-exchanger (NCX) operating in the reverse mode and indirect activation of L -type VOCCs, as indicated by VOCCs/NCX pharmacological inhibitors. A subunit of Na+/K+-ATPase was selected by comparative proteomics and identified as being functionally inhibited by extracellular TG2, implicating Na+/K+-ATPase inhibition in NCX reverse mode-switching leading to Ca2+ influx and higher basal [Ca2+] i. These data suggest that reactive astrocytes control intraneuronal [Ca2+] i through release of EVs with TG2 as responsible cargo, which could have a significant impact on synaptic activity in brain inflammation. [Display omitted] • Primed astrocytes-derived EVs control neuronal [Ca2+] i via TG2 as a surface-cargo. • Extracellular TG2 increases basal [Ca2+] i in hippocampal neurons and brain slices. • TG2 mediates Na+/K+-ATPase inhibition, inducing VOCCs opening and NCX reverse mode. • Astrocytic EVs could affect synaptic activity in brain inflammation via TG2 cargo. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
191. Phenotypic Changes, Signaling Pathway, and Functional Correlates of GPR17-expressing Neural Precursor Cells during Oligodendrocyte Differentiation.
- Author
-
Fumagalli, Marta, Daniele, Simona, Lecca, Davide, Lee, Philip R., Parravicini, Chiara, Fields, R. Douglas, Rosa, Patrizia, Antonucci, Flavia, Verderio, Claudia, Trincavelli, M. Letizia, Bramanti, Placido, Martinis, Claudia, and Abbracchio, Maria P.
- Subjects
- *
NUCLEIC acids , *LEUKOTRIENES , *ARACHIDONIC acid , *MEMBRANE proteins , *MYELIN proteins , *PROTEIN precursors , *PROTEOGLYCANS - Abstract
The developing and mature central nervous system contains neural precursor cells expressing the proteoglycan NG2. Some of these cells continuously differentiate to myelin-forming oligodendrocytes; knowledge of the destiny of NG2+ precursors would benefit from the characterization of new key functional players. In this respect, the G protein-coupled membrane receptor GPR17 has recently emerged as a new timer of oligodendrogliogenesis. Here, we used purified oligodendrocyte precursor cells (OPCs) to fully define the immunophenotype of the GPR17-expressing cells during OPC differentiation, unveil its native signaling pathway, and assess the functional consequences of GPR17 activation by its putative endogenous ligands, uracil nucleotides and cysteinyl leukotrienes (cysLTs). GPR17 presence was restricted to very early differentiation stages and completely segregated from that of mature myehin. Specifically, GPR17 decorated two subsets of slowly proliferating NG2+ OPCs: (i) morphologically immature cells expressing other early proteins like Olig2 and PDGF receptor-a, and (ii) ramified pre- oligodendrocytes already expressing more mature factors, like O4 and O1. Thus, GPR17 is a new marker of these transition stages. In OPCs, GPR17 activation by either uracil nucleotides or cysLTs resulted in potent inhibition of intracellular cAMP formation. This effect was counteracted by GPR17 antagonists and receptor silencing with siRNAs. Finally, uracil nucleotides promoted and GPR17 inhibition, by either antagonists or siRNAs, impaired the normal program of OPC differentiation. These data have implications for the in vivo behavior of NG2+ OPCs and point to uracil nucleotides and cysLTs as main extrinsic local regulators of these cells under physiological conditions and during myelin repair. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
192. Endogenous SNAP-25 Regulates Native Voltage-gated Calcium Channels in Glutamatergic Neurons.
- Author
-
Condliffe, Steven B., Corradini, Irene, Pozzi, Davide, Verderio, Claudia, and Matteoli, Michela
- Subjects
- *
CALCIUM channels , *NEURONS , *HIPPOCAMPUS (Brain) , *LABORATORY rats , *BIOCHEMISTRY - Abstract
In addition to its primary role as a fundamental component of the SNARE complex, SNAP-25 also modulates voltage-gated calcium channels (VGCCs) in various overexpression systems. Although these studies suggest a potential negative regulatory role of SNAP-25 on VGCC activity, the effects of endogenous SNAP-25 on native VGCC function in neurons are unclear. In the present study, we investigated the VGCC properties of cultured glutamatergic and GABAergic rat hippocampal neurons. Glutamatergic currents were dominated by P/Q-type channels, whereas GABAergic cells had a dominant L-type component. Also, glutamatergic VGCC current densities were significantly lower with enhanced inactivation rates and shifts in the voltage dependence of activation and inactivation curves compared with GABAergic cells. Silencing endogenous SNAP-25 in glutamatergic neurons did not alter P/Q-type channel expression or localization but led to increased VGCC current density without changes in the VGCC subtype proportions. Isolation of the P/Q-type component indicated that increased current in the absence of SNAP-25 was correlated with a large depolarizing shift in the voltage dependence of inactivation. Overexpressing SNAP-25 in GABAergic neurons reduced current density without affecting the VGCC subtype proportion. Accordingly, VGCC current densities in glutamatergic neurons from Snap-25+/- mice were significantly elevated compared with wild type glutamatergic neurons. Overall, this study demonstrates that endogenous SNAP-25 negatively regulates native VGCCs in glutamatergic neurons which could have important implications for neurological diseases associated with altered SNAP-25 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
193. Abscisic Acid Activates the Murine Microglial Cell Line N9 through the Second Messenger Cyclic ADP-ribose.
- Author
-
Bodrato, Nicoletta, Franco, Luisa, Fresia, Chiara, Gujda, Lucrezia, Usai, Cesare, Salis, Annalisa, Moreschi, Iliana, Ferraris, Chiara, Verderio, Claudia, Basile, Giovanna, Bruzzone, Santina, Scarfi, Sonia, De Flora, Antonio, and Zocchi, Elena
- Subjects
- *
CENTRAL nervous system , *ALZHEIMER'S disease , *LEUCOCYTES , *CYTOKINES , *CELL culture , *PLANT hormones , *ABSCISIC acid , *CELL physiology , *PHYSIOLOGY - Abstract
Abscisic acid (ABA) is a phytohormone regulating important functions in higher plants, notably responses to abiotic stress. Recently, chemical or physical stimulation of human granulocytes was shown to induce production and release of endogenous ABA, which activates specific cell functions. Here we provide evidence that ABA stimulates several functional activities of the murine microglial cell line N9 (NO and tumor necrosis factor-a production, cell migration) through the second messenger cyclic ADP-ribose and an increase of intracellular calcium. ABA production and release occur in N9 cells stimulated with bacterial lipopolysaccharide, phorbol myristate acetate, the chemoattractant peptide f-MLP, or β-amyloid, the primary plaque component in Alzheimer disease. Finally, ABA priming stimulates N9 cell migration toward β-amyloid. These results indicate that ABA is a pro-inflammatory hormone inducing autocrine microglial activation, potentially representing a new target for anti-inflammatory therapies aimed at limiting microglia-induced tissue damage in the central nervous system. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
194. Therapeutic induction of energy metabolism reduces neural tissue damage and increases microglia activation in severe spinal cord injury.
- Author
-
Dolci, Sissi, Mannino, Loris, Bottani, Emanuela, Campanelli, Alessandra, Di Chio, Marzia, Zorzin, Stefania, D'Arrigo, Giulia, Amenta, Alessia, Segala, Agnese, Paglia, Giuseppe, Denti, Vanna, Fumagalli, Guido, Nisoli, Enzo, Valerio, Alessandra, Verderio, Claudia, Martano, Giuseppe, Bifari, Francesco, and Decimo, Ilaria
- Subjects
- *
NERVE tissue , *SPINAL cord injuries , *ENERGY metabolism , *ESSENTIAL amino acids , *CELL metabolism , *CELL death , *MOTOR neurons - Abstract
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. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
195. Uptake and recycling of pro-BDNF for transmitter-induced secretion by cortical astrocytes.
- Author
-
Bergami, Matteo, Santi, Spartaco, Formaggio, Elena, Cagnoli, Cinzia, Verderio, Claudia, Blum, Robert, Berninger, Benedikt, Matteoli, Michela, and Canossa, Marco
- Subjects
- *
NEUROSECRETION , *NEUROTRANSMITTERS , *ASTROCYTES , *NEUROPLASTICITY , *NEURAL transmission - Abstract
Activity-dependent secretion of brain-derived neurotrophic factor (BDNF) is thought to enhance synaptic plasticity, but the mechanisms controlling extracellular availability and clearance of secreted BDNF are poorly understood. We show that BDNF is secreted in its precursor form (pro-BDNF) and is then cleared from the extracellular space through rapid uptake by nearby astrocytes after θ-burst stimulation in layer II/III of cortical slices, a paradigm resulting in long-term potentiation of synaptic transmission. Internalization of pro-BDNF occurs via the formation of a complex with the pan-neurotrophin receptor p75 and subsequent clathrin-dependent endocytosis. Fluorescence-tagged pro-BDNF and real-time total internal reflection fluorescence microscopy in cultured astrocytes is used to monitor single endocytic vesicles in response to the neurotransmitter glutamate. We find that endocytosed pro-BDNF is routed into a fast recycling pathway for subsequent soluble NSF attachment protein receptor-dependent secretion. Thus, astrocytes contain an endocytic compartment competent for pro-BDNF recycling, suggesting a specialized form of bidirectional communication between neurons and glia. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
196. Extracellular proteasome-osteopontin circuit regulates cell migration with implications in multiple sclerosis
- Author
-
Christin Keller, Filippo Martinelli Boneschi, Loredana Riganti, Elena Bellavista, Chiara Dianzani, Klemens Ruprecht, Katharina Janek, Annalisa Chiocchetti, Chiara Fenoglio, Morena Martucci, Daniela Galimberti, Friedemann Paul, Casimiro Luca Gigliotti, Benedetta Ferrara, Claudia Verderio, Elena Boggio, Michael P. H. Stumpf, Agathe Niewienda, Aurelia Santoro, Juliane Liepe, Peter M. Kloetzel, Roberto Cantello, Melissa Sorosina, Michele Mishto, Umberto Dianzani, Cristoforo Comi, Dianzani, Chiara, Bellavista, Elena, Liepe, Juliane, Verderio, Claudia, Martucci, Morena, Santoro, Aurelia, Chiocchetti, Annalisa, Gigliotti, Casimiro Luca, Boggio, Elena, Ferrara, Benedetta, Riganti, Loredana, Keller, Christin, Janek, Katharina, Niewienda, Agathe, Fenoglio, Chiara, Sorosina, Melissa, Cantello, Roberto, Kloetzel, Peter M, Stumpf, Michael P H, Paul, Friedemann, Ruprecht, Klemen, Galimberti, Daniela, Martinelli Boneschi, Filippo, Comi, Cristoforo, Dianzani, Umberto, and Mishto, Michele
- Subjects
Models, Molecular ,0301 basic medicine ,Proteasome Endopeptidase Complex ,Chemokine ,Proteases ,Multiple Sclerosis ,Protein Conformation ,medicine.medical_treatment ,Inflammation ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit ,Article ,Extracellular Vesicles ,Structure-Activity Relationship ,03 medical and health sciences ,Journal Article ,Extracellular ,medicine ,Humans ,Amino Acid Sequence ,Lymphocytes ,Osteopontin ,Multidisciplinary ,biology ,Chemotaxis ,Endothelial Cells ,Cell migration ,Cell biology ,Proteasome, Osteopontin, Multiple Sclerosis ,030104 developmental biology ,Cytokine ,Proteasome ,biology.protein ,medicine.symptom ,Chemokines ,Extracellular Space ,Function and Dysfunction of the Nervous System - Abstract
Osteopontin is a pleiotropic cytokine that is involved in several diseases including multiple sclerosis. Secreted osteopontin is cleaved by few known proteases, modulating its pro-inflammatory activities. Here we show by in vitro experiments that secreted osteopontin can be processed by extracellular proteasomes, thereby producing fragments with novel chemotactic activity. Furthermore, osteopontin reduces the release of proteasomes in the extracellular space. The latter phenomenon seems to occur in vivo in multiple sclerosis, where it reflects the remission/relapse alternation. The extracellular proteasome-mediated inflammatory pathway may represent a general mechanism to control inflammation in inflammatory diseases.
- Published
- 2017
- Full Text
- View/download PDF
197. Sphingosine-1-phosphate (S1P) impacts presynaptic functions by regulating synapsin i localization in the presynaptic compartment
- Author
-
Claudia Verderio, Flavia Antonucci, Paola Giussani, Michela Matteoli, Loredana Riganti, Flavia Valtorta, Elisabetta Menna, Martina Gabrielli, Paola Viani, Ilaria Prada, Riganti, Loredana, Antonucci, Flavia, Gabrielli, Martina, Prada, Ilaria, Giussani, Paola, Viani, Paola, Valtorta, Flavia, Menna, Elisabetta, Matteoli, Michela, and Verderio, Claudia
- Subjects
0301 basic medicine ,Male ,Hippocampus ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Sphingosine ,Axon ,Cells, Cultured ,Lysophospholipid ,Mice, Knockout ,Glutamate secretion ,S-FTY720-vinylphosphonate ,General Neuroscience ,Articles ,Synapse ,medicine.anatomical_structure ,lipids (amino acids, peptides, and proteins) ,Female ,Microglia ,Presynaptic Terminal ,Synapsin I ,S1P3 receptor ,Sphingosine-1-phosphate ,Presynaptic Terminals ,Neurotransmission ,Biology ,Synaptic vesicle ,Exocytosis ,03 medical and health sciences ,Hippocampu ,medicine ,Animals ,Growth cone ,Neuroscience (all) ,Animal ,Neurotransmitter release ,Synapsins ,Rats ,Synapsin ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Synapses ,Rat ,Lysophospholipids ,Neuroscience ,030217 neurology & neurosurgery - Abstract
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 Ca2+-independent but ERK-dependent manner, likely through the activation of S1P3receptors, as it was prevented by the S1P3receptor selective antagonist CAY1044 and in neurons in which S1P3receptor 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 STATEMENTGrowing 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.
- Published
- 2016
198. Extracellular Vesicle-Mediated Neuron-Glia Communications in the Central Nervous System.
- Author
-
Ikezu T, Yang Y, Verderio C, and Krämer-Albers EM
- Subjects
- Humans, Animals, Extracellular Vesicles physiology, Extracellular Vesicles metabolism, Neurons physiology, Neuroglia physiology, Cell Communication physiology, Central Nervous System physiology, Central Nervous System cytology
- Abstract
Communication between neurons and glia significantly influences the development maturation, plasticity, and disease progressions of the nervous system. As a new signaling modality, extracellular vesicles display a diverse role for robust functional regulation of neurons through their protein and nucleic acid cargoes. This review highlights recent breakthroughs in the research of signaling mechanisms between glia and neurons mediated by extracellular vesicles that are important for neural development, axonal maintenance, synaptic functions, and disease progression in the mammalian nervous system. We will discuss the biological roles of extracellular vesicles released from neurons, astroglia, microglia, and oligodendroglia in the nervous system and their implications in neurodegenerative disorders., Competing Interests: The authors declare no competing financial interests., (Copyright © 2024 the authors.)
- Published
- 2024
- Full Text
- View/download PDF
199. Paired plasma lipidomics and proteomics analysis in the conversion from mild cognitive impairment to Alzheimer's disease.
- Author
-
Gómez-Pascual A, Naccache T, Xu J, Hooshmand K, Wretlind A, Gabrielli M, Lombardo MT, Shi L, Buckley NJ, Tijms BM, Vos SJB, Ten Kate M, Engelborghs S, Sleegers K, Frisoni GB, Wallin A, Lleó A, Popp J, Martinez-Lage P, Streffer J, Barkhof F, Zetterberg H, Visser PJ, Lovestone S, Bertram L, Nevado-Holgado AJ, Gualerzi A, Picciolini S, Proitsi P, Verderio C, Botía JA, and Legido-Quigley C
- Subjects
- Humans, Male, Aged, Female, Biomarkers blood, Biomarkers metabolism, Animals, Disease Progression, Machine Learning, Aged, 80 and over, Alzheimer Disease blood, Alzheimer Disease metabolism, Cognitive Dysfunction blood, Cognitive Dysfunction metabolism, Proteomics methods, Lipidomics methods
- Abstract
Background: Alzheimer's disease (AD) is a neurodegenerative condition for which there is currently no available medication that can stop its progression. Previous studies suggest that mild cognitive impairment (MCI) is a phase that precedes the disease. Therefore, a better understanding of the molecular mechanisms behind MCI conversion to AD is needed., Method: Here, we propose a machine learning-based approach to detect the key metabolites and proteins involved in MCI progression to AD using data from the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery Study. Proteins and metabolites were evaluated separately in multiclass models (controls, MCI and AD) and together in MCI conversion models (MCI stable vs converter). Only features selected as relevant by 3/4 algorithms proposed were kept for downstream analysis., Results: Multiclass models of metabolites highlighted nine features further validated in an independent cohort (0.726 mean balanced accuracy). Among these features, one metabolite, oleamide, was selected by all the algorithms. Further in-vitro experiments in rodents showed that disease-associated microglia excreted oleamide in vesicles. Multiclass models of proteins stood out with nine features, validated in an independent cohort (0.720 mean balanced accuracy). However, none of the proteins was selected by all the algorithms. Besides, to distinguish between MCI stable and converters, 14 key features were selected (0.872 AUC), including tTau, alpha-synuclein (SNCA), junctophilin-3 (JPH3), properdin (CFP) and peptidase inhibitor 15 (PI15) among others., Conclusions: This omics integration approach highlighted a set of molecules associated with MCI conversion important in neuronal and glia inflammation pathways., Competing Interests: Declaration of competing interest SL is named as an inventor on biomarker intellectual property protected by Proteome Sciences and Kings College London unrelated to the current study and within the past five years has advised for Optum labs, Merck, SomaLogic and been the recipient of funding from AstraZeneca and other companies via the IMI funding scheme. HZ has served at scientific advisory boards and/or as a consultant for Abbvie, Alector, ALZPath, Annexon, Apellis, Artery Therapeutics, AZTherapies, CogRx, Denali, Eisai, Nervgen, Novo Nordisk, Pinteon Therapeutics, Red Abbey Labs, reMYND, Passage Bio, Roche, Samumed, Siemens Healthineers, Triplet Therapeutics, and Wave, has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure, Biogen, and Roche, and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). AL has served at scientific advisory boards of Fujirebio Europe, Eli Lilly, Novartis, Nutricia and Otsuka and is the inventor of a patent on synaptic markers in CSF (all unrelated to this study). JP has served at scientific advisory boards of Fujirebio Europe, Eli Lilly and Nestle Institute of Health Sciences, all unrelated to this study. SE has received unrestricted research grants from Janssen Pharmaceutica and ADx Neurosciences and has served at scientific advisory boards of Biogen, Eisai, Novartis, Nutricia/Danone, all unrelated to this study. FB is a steering committee or iDMC member for Biogen, Merck, Roche, EISAI and Prothena. Consultant for Roche, Biogen, Merck, IXICO, Jansen, Combinostics. Research agreements with Merck, Biogen, GE Healthcare, Roche. Co-founder and shareholder of Queen Square Analytics LTD, all unrelated to this study. CLQ has received funding related to this study from Pfizer and via the IMI funding scheme, in the past five years has been the recipient of funding from Novo Nordisk, unrelated to this study., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
200. Changes in glial cell activation and extracellular vesicles production precede the onset of disease symptoms in transgenic hSOD1 G93A pigs.
- Author
-
Golia MT, Frigerio R, Pucci S, Sironi F, Margotta C, Pasetto L, Testori C, Berrone E, Ingravalle F, Chiari M, Gori A, Duchi R, Perota A, Bergamaschi L, D'Angelo A, Cagnotti G, Galli C, Corona C, Bonetto V, Bendotti C, Cretich M, Colombo SF, and Verderio C
- Subjects
- Mice, Animals, Humans, Swine, Superoxide Dismutase-1 genetics, Motor Neurons metabolism, Superoxide Dismutase genetics, Mice, Transgenic, Spinal Cord pathology, Neuroglia pathology, Biomarkers metabolism, Peptides metabolism, Disease Models, Animal, Amyotrophic Lateral Sclerosis pathology, Extracellular Vesicles
- Abstract
SOD1 gene is associated with progressive motor neuron degeneration in the familiar forms of amyotrophic lateral sclerosis. Although studies on mutant human SOD1 transgenic rodent models have provided important insights into disease pathogenesis, they have not led to the discovery of early biomarkers or effective therapies in human disease. The recent generation of a transgenic swine model expressing the human pathological hSOD1
G93A gene, which recapitulates the course of human disease, represents an interesting tool for the identification of early disease mechanisms and diagnostic biomarkers. Here, we analyze the activation state of CNS cells in transgenic pigs during the disease course and investigate whether changes in neuronal and glial cell activation state can be reflected by the amount of extracellular vesicles they release in biological fluids. To assess the activation state of neural cells, we performed a biochemical characterization of neurons and glial cells in the spinal cords of hSOD1G93A pigs during the disease course. Quantification of EVs of CNS cell origin was performed in cerebrospinal fluid and plasma of transgenic pigs at different disease stages by Western blot and peptide microarray analyses. We report an early activation of oligodendrocytes in hSOD1G93A transgenic tissue followed by astrocyte and microglia activation, especially in animals with motor symptoms. At late asymptomatic stage, EV production from astrocytes and microglia is increased in the cerebrospinal fluid, but not in the plasma, of transgenic pigs reflecting donor cell activation in the spinal cord. Estimation of EV production by biochemical analyses is corroborated by direct quantification of neuron- and microglia-derived EVs in the cerebrospinal fluid by a Membrane Sensing Peptide enabled on-chip analysis that provides fast results and low sample consumption. Collectively, our data indicate that alteration in astrocytic EV production precedes the onset of disease symptoms in the hSODG93A swine model, mirroring donor cell activation in the spinal cord, and suggest that EV measurements from the cells first activated in the ALS pig model, i.e. OPCs, may further improve early disease detection., Competing Interests: Declaration of competing interest The authors declare no conflict of interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.