Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 86: 143-155, 2020 02.
Article in English | MEDLINE | ID: mdl-31784278

ABSTRACT

Despite the great effort of the scientific community in the field, the pathogenesis of frontotemporal dementia (FTD) remains elusive. Recently, a role for autoimmunity and altered glutamatergic neurotransmission in triggering disease onset has been put forward. We reported the presence of autoantibodies recognizing the GluA3 subunit of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors in about 25% of FTD cases. In this study, we evaluated the mechanisms involved in anti-GluA3 autoimmunity, through molecular/neurochemical analyses conducted on patients' brain specimens with frontotemporal lobar degeneration-tau neuropathology. We then corroborated these results in vivo in FTD patients with transcranial magnetic stimulation and glutamate, D-serine, and L-serine dosages in the cerebrospinal fluid and serum. We observed that GluA3 autoantibodies affect glutamatergic neurotransmission, decreasing glutamate release and altering GluA3-containing α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor levels. These alterations were accompanied by changes of scaffolding proteins involved in receptor synaptic retention/internalization. The above results were confirmed by transcranial magnetic stimulation, suggesting a significant impairment of indirect measures of glutamatergic neurotransmission in FTD patients compared with controls, with further add-on harmful effect in those FTD patients with anti-GluA3 antibodies. Finally, FTD patients showed a significant increase of glutamate, D-serine, and L-serine levels in the cerebrospinal fluid.


Subject(s)
Autoantibodies , Frontotemporal Dementia/etiology , Frontotemporal Dementia/immunology , Frontotemporal Dementia/physiopathology , Glutamates/cerebrospinal fluid , Receptors, AMPA/immunology , Synapses/physiology , Synaptic Transmission , Adult , Autoimmunity , Female , Humans , Male , Middle Aged
2.
iScience ; 19: 927-939, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31518901

ABSTRACT

NMDA receptor (NMDAR) subunit composition plays a pivotal role in synaptic plasticity at excitatory synapses. Still, the mechanisms responsible for the synaptic retention of NMDARs following induction of plasticity need to be fully elucidated. Rabphilin3A (Rph3A) is involved in the stabilization of NMDARs at synapses through the formation of a complex with GluN2A and PSD-95. Here we used different protocols to induce synaptic plasticity in the presence or absence of agents modulating Rph3A function. The use of Forskolin/Rolipram/Picrotoxin cocktail to induce chemical LTP led to synaptic accumulation of Rph3A and formation of synaptic GluN2A/Rph3A complex. Notably, Rph3A silencing or use of peptides interfering with the GluN2A/Rph3A complex blocked LTP induction. Moreover, in vivo disruption of GluN2A/Rph3A complex led to a profound alteration of spatial memory. Overall, our results demonstrate a molecular mechanism needed for NMDAR stabilization at synapses after plasticity induction and to trigger downstream signaling events necessary for cognitive behavior.

3.
Brain ; 142(5): 1365-1385, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30927362

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder characterized by altered striatal dopaminergic signalling that leads to motor and cognitive deficits. Parkinson's disease is also characterized by abnormal presence of soluble toxic forms of α-synuclein that, when clustered into Lewy bodies, represents one of the pathological hallmarks of the disease. However, α-synuclein oligomers might also directly affect synaptic transmission and plasticity in Parkinson's disease models. Accordingly, by combining electrophysiological, optogenetic, immunofluorescence, molecular and behavioural analyses, here we report that α-synuclein reduces N-methyl-d-aspartate (NMDA) receptor-mediated synaptic currents and impairs corticostriatal long-term potentiation of striatal spiny projection neurons, of both direct (D1-positive) and indirect (putative D2-positive) pathways. Intrastriatal injections of α-synuclein produce deficits in visuospatial learning associated with reduced function of GluN2A NMDA receptor subunit indicating that this protein selectively targets this subunit both in vitro and ex vivo. Interestingly, this effect is observed in spiny projection neurons activated by optical stimulation of either cortical or thalamic glutamatergic afferents. We also found that treatment of striatal slices with antibodies targeting α-synuclein prevents the α-synuclein-induced loss of long-term potentiation and the reduced synaptic localization of GluN2A NMDA receptor subunit suggesting that this strategy might counteract synaptic dysfunction occurring in Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spatial Memory/physiology , Synapses/physiology , Visual Perception/physiology , alpha-Synuclein/toxicity , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Humans , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spatial Memory/drug effects , Synapses/drug effects , Visual Perception/drug effects , alpha-Synuclein/administration & dosage
4.
Neurobiol Dis ; 121: 338-349, 2019 01.
Article in English | MEDLINE | ID: mdl-30261285

ABSTRACT

In the striatum, specific N-methyl-d-aspartate receptor (NMDAR) subtypes are found in different neuronal cells. Spiny projection neurons (SPNs) are characterized by NMDARs expressing GluN2A and GluN2B subunits, while GluN2D is exclusively detected in striatal cholinergic interneurons (ChIs). In Parkinson's disease (PD), dopamine depletion and prolonged treatment with levodopa (L-DOPA) trigger adaptive changes in the glutamatergic transmission from the cortex to the striatum, also resulting in the aberrant function of striatal NMDARs. While modifications of GluN2A- and GluN2B-NMDARs in SPNs have been extensively documented, only few studies report GluN2D dysfunction in PD and no data are available in L-DOPA-induced dyskinesia (LID). Here we investigate the contribution of a specific NMDAR subtype (GluN2D-NMDAR) to PD and LID, and whether this receptor could represent a candidate for future pharmacological interventions. Our results show that GluN2D synaptic abundance is selectively augmented in the striatum of L-DOPA-treated male parkinsonian rats displaying a dyskinetic phenotype. This event is associated to a dramatic increase in GluN2D binding to the postsynaptic protein scaffold PSD-95. Moreover, immunohistochemistry and electrophysiology experiments reveal that GluN2D-NMDARs are expressed not only by striatal ChIs but also by SPNs in dyskinetic rats. Notably, in vivo treatment with a well-characterized GluN2D antagonist ameliorates the severity of established dyskinesia in L-DOPA-treated animals. Our findings support a role for GluN2D-NMDARs in LID, and they confirm that cell-type and subunit specific modifications of NMDARs underlie the pathophysiology of LID.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cholinergic Neurons/metabolism , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Interneurons/metabolism , Levodopa/administration & dosage , Macaca mulatta , Male , Rats, Sprague-Dawley , Synapses/metabolism
5.
J Neural Transm (Vienna) ; 125(8): 1225-1236, 2018 08.
Article in English | MEDLINE | ID: mdl-29387966

ABSTRACT

Overactivation of the glutamatergic synapse leading to maladaptive synaptic plasticity in the basal ganglia is a well-demonstrated process involved in the onset of L-DOPA-induced dyskinesia (LID). Changes in glutamate release are paralleled by compensatory modifications of the expression and/or synaptic localization of both ionotropic and metabotropic glutamate receptors (mGluRs). Accordingly, compounds targeting N-methyl-D-aspartate glutamate receptors (NMDARs) and specific subtypes of metabotropic glutamate receptors (mGluR4 and mGluR5) have been tested both in preclinical and clinical studies. At present, amantadine, a low-affinity non-competitive NMDAR antagonist, represents the only recommended add-on agent with a moderate anti-dyskinetic activity. The present review describes recent advances in basic research, preclinical and early clinical studies in the attempt of identifying innovative strategies for an accurate modulation of both pre- and postsynaptic glutamate receptors to reduce the severity of LID. Even if a complete understanding of LID molecular bases is still lacking, several compounds demonstrated an anti-dyskinetic activity in preclinical and early clinical studies. These results indicate that modulation of the glutamatergic system remains one of the most promising pharmacological strategies in the field.


Subject(s)
Dyskinesia, Drug-Induced/metabolism , Glutamic Acid/metabolism , Receptors, Glutamate/metabolism , Animals , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Humans , Levodopa/adverse effects , Neurons/drug effects , Neurons/metabolism , Receptors, Glutamate/drug effects
6.
Neurobiol Dis ; 108: 54-64, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28823933

ABSTRACT

N-methyl-d-aspartate receptor (NMDAR) subunit composition strictly commands receptor function and pharmacological responses. Changes in NMDAR subunit composition have been documented in brain disorders such as Parkinson's disease (PD) and levodopa (L-DOPA)-induced dyskinesias (LIDs), where an increase of NMDAR GluN2A/GluN2B subunit ratio at striatal synapses has been observed. A therapeutic approach aimed at rebalancing NMDAR synaptic composition represents a valuable strategy for PD and LIDs. To this, the comprehension of the molecular mechanisms regulating the synaptic localization of different NMDAR subtypes is required. We have recently demonstrated that Rabphilin 3A (Rph3A) is a new binding partner of NMDARs containing the GluN2A subunit and that it plays a crucial function in the synaptic stabilization of these receptors. Considering that protein-protein interactions govern the synaptic retention of NMDARs, the purpose of this work was to analyse the role of Rph3A and Rph3A/NMDAR complex in PD and LIDs, and to modulate Rph3A/GluN2A interaction to counteract the aberrant motor behaviour associated to chronic L-DOPA administration. Thus, an array of biochemical, immunohistochemical and pharmacological tools together with electron microscopy were applied in this study. Here we found that Rph3A is localized at the striatal postsynaptic density where it interacts with GluN2A. Notably, Rph3A expression at the synapse and its interaction with GluN2A-containing NMDARs were increased in parkinsonian rats displaying a dyskinetic profile. Acute treatment of dyskinetic animals with a cell-permeable peptide able to interfere with Rph3A/GluN2A binding significantly reduced their abnormal motor behaviour. Altogether, our findings indicate that Rph3A activity is linked to the aberrant synaptic localization of GluN2A-expressing NMDARs characterizing LIDs. Thus, we suggest that Rph3A/GluN2A complex could represent an innovative therapeutic target for those pathological conditions where NMDAR composition is significantly altered.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Nerve Tissue Proteins/metabolism , Parkinsonian Disorders/metabolism , Post-Synaptic Density/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Aged , Aged, 80 and over , Animals , Antiparkinson Agents/therapeutic use , Antiparkinson Agents/toxicity , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Female , Humans , Levodopa/therapeutic use , Macaca mulatta , Male , Nerve Tissue Proteins/antagonists & inhibitors , Oxidopamine , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Post-Synaptic Density/drug effects , Post-Synaptic Density/pathology , Protein Binding/drug effects , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synapses/metabolism , Tissue Culture Techniques , Vesicular Transport Proteins/antagonists & inhibitors , Rabphilin-3A
7.
Mol Brain ; 9(1): 53, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27169991

ABSTRACT

Levodopa (L-DOPA)-induced dyskinesias (LIDs) represent the major side effect in Parkinson's disease (PD) therapy. Leucine-rich repeat kinase 2 (LRRK2) mutations account for up to 13 % of familial cases of PD. LRRK2 N-terminal domain encompasses several serine residues that undergo phosphorylation influencing LRRK2 function. This work aims at investigating whether LRRK2 phosphorylation/function may be involved in the molecular pathways downstream D1 dopamine receptor leading to LIDs. Here we show that LRRK2 phosphorylation level at serine 935 correlates with LIDs induction and that inhibition of LRRK2 induces a significant increase in the dyskinetic score in L-DOPA treated parkinsonian animals. Our findings support a close link between LRKK2 functional state and L-DOPA-induced abnormal motor behaviour and highlight that LRRK2 phosphorylation level may be implicated in LIDs, calling for novel therapeutic strategies.


Subject(s)
Dyskinesias/enzymology , Dyskinesias/physiopathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Motor Activity , Animals , Disease Models, Animal , Dyskinesias/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Levodopa , Male , Motor Activity/drug effects , Oxidopamine , Parkinson Disease/enzymology , Parkinson Disease/physiopathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley
8.
Elife ; 5: e12430, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26977767

ABSTRACT

Synapses and nuclei are connected by bidirectional communication mechanisms that enable information transfer encoded by macromolecules. Here, we identified RNF10 as a novel synaptonuclear protein messenger. RNF10 is activated by calcium signals at the postsynaptic compartment and elicits discrete changes at the transcriptional level. RNF10 is enriched at the excitatory synapse where it associates with the GluN2A subunit of NMDA receptors (NMDARs). Activation of synaptic GluN2A-containing NMDARs and induction of long term potentiation (LTP) lead to the translocation of RNF10 from dendritic segments and dendritic spines to the nucleus. In particular, we provide evidence for importin-dependent long-distance transport from synapto-dendritic compartments to the nucleus. Notably, RNF10 silencing prevents the maintenance of LTP as well as LTP-dependent structural modifications of dendritic spines.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/physiology , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Animals , Cell Nucleus/metabolism , Protein Transport , Rats
9.
Biol Psychiatry ; 79(5): 402-414, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26392130

ABSTRACT

BACKGROUND: Advanced Parkinson's disease (PD) is characterized by massive degeneration of nigral dopaminergic neurons, dramatic motor and cognitive alterations, and presence of nigral Lewy bodies, whose main constituent is α-synuclein (α-syn). However, the synaptic mechanisms underlying behavioral and motor effects induced by early selective overexpression of nigral α-syn are still a matter of debate. METHODS: We performed behavioral, molecular, and immunohistochemical analyses in two transgenic models of PD, mice transgenic for truncated human α-synuclein 1-120 and rats injected with the adeno-associated viral vector carrying wild-type human α-synuclein. We also investigated striatal synaptic plasticity by electrophysiological recordings from spiny projection neurons and cholinergic interneurons. RESULTS: We found that overexpression of truncated or wild-type human α-syn causes partial reduction of striatal dopamine levels and selectively blocks the induction of long-term potentiation in striatal cholinergic interneurons, producing early memory and motor alterations. These effects were dependent on α-syn modulation of the GluN2D-expressing N-methyl-D-aspartate receptors in cholinergic interneurons. Acute in vitro application of human α-syn oligomers mimicked the synaptic effects observed ex vivo in PD models. CONCLUSIONS: We suggest that striatal cholinergic dysfunction, induced by a direct interaction between α-syn and GluN2D-expressing N-methyl-D-aspartate receptors, represents a precocious biological marker of the disease.


Subject(s)
Cholinergic Neurons/drug effects , Dopamine/physiology , Parkinson Disease/drug therapy , Receptors, N-Methyl-D-Aspartate/genetics , alpha-Synuclein/genetics , Animals , Animals, Genetically Modified , Dependovirus , Disease Models, Animal , Female , Humans , Long-Term Potentiation , Male , Mice , Mice, Transgenic , Neostriatum/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Synaptic Transmission
10.
Neurobiol Dis ; 86: 140-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26639853

ABSTRACT

L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesias (LIDs) represent the main side effect of Parkinson's Disease (PD) therapy. Among the various pharmacological targets for novel therapeutic approaches, the serotonergic system represents a promising one. In experimental models of PD and in PD patients the development of abnormal involuntary movements (AIMs) and LIDs, respectively, is accompanied by the impairment of bidirectional synaptic plasticity in key structures such as striatum. Recently, it has been shown that the 5-HT1A/1B receptor agonist, eltoprazine, significantly decreased LIDs in experimental PD and human patients. Despite the fact that several papers have tested this and other serotonergic drugs, nothing is known about the electrophysiological consequences on this combined serotonin receptors modulation at striatal neurons. The present study demonstrates that activation of 5-HT1A/1B receptors reduces AIMs via the restoration of Long-Term Potentiation (LTP) and synaptic depotentiation in a sub-set of striatal spiny projection neurons (SPNs). This recovery is associated with the normalization of D1 receptor-dependent cAMP/PKA and ERK/mTORC signaling pathways, and the recovery of NMDA receptor subunits balance, indicating these events as key elements in AIMs induction. Moreover, we analyzed whether the manipulation of the serotonergic system might affect motor behavior and cognitive performances. We found that a defect in locomotor activity in parkinsonian and L-DOPA-treated rats was reversed by eltoprazine treatment. Conversely, the impairment in the striatal-dependent learning was found exacerbated in L-DOPA-treated rats and eltoprazine failed to recover it.


Subject(s)
Behavior, Animal/drug effects , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Neurons/drug effects , Neurons/physiology , Parkinsonian Disorders/complications , Piperazines/administration & dosage , Serotonin Receptor Agonists/administration & dosage , Animals , Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/psychology , Levodopa , MAP Kinase Signaling System/drug effects , Male , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Neurons/metabolism , Oxidopamine , Parkinsonian Disorders/chemically induced , Rats , Rats, Wistar , Synapses/metabolism , Synaptic Transmission/drug effects , TOR Serine-Threonine Kinases/metabolism
11.
Front Cell Neurosci ; 9: 245, 2015.
Article in English | MEDLINE | ID: mdl-26217176

ABSTRACT

Levodopa-induced dyskinesias (LIDs) are major complications in the pharmacological management of Parkinson's disease (PD). Abnormal glutamatergic transmission in the striatum is considered a key factor in the development of LIDs. This work aims at: (i) characterizing N-methyl-D-aspartate (NMDA) receptor GluN2A/GluN2B subunit ratio as a common synaptic trait in rat and primate models of LIDs as well as in dyskinetic PD patients; and (ii) validating the potential therapeutic effect of a cell-permeable peptide (CPP) interfering with GluN2A synaptic localization on the dyskinetic behavior of these experimental models of LIDs. Here we demonstrate an altered ratio of synaptic GluN2A/GluN2B-containing NMDA receptors in the striatum of levodopa-treated dyskinetic rats and monkeys as well as in post-mortem tissue from dyskinetic PD patients. The modulation of synaptic NMDA receptor composition by a cell-permeable peptide interfering with GluN2A subunit interaction with the scaffolding protein postsynaptic density protein 95 (PSD-95) leads to a reduction in the dyskinetic motor behavior in the two animal models of LIDs. Our results indicate that targeting synaptic NMDA receptor subunit composition may represent an intriguing therapeutic approach aimed at ameliorating levodopa motor side effects.

12.
Prog Neurobiol ; 132: 96-168, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26209473

ABSTRACT

Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.


Subject(s)
Antiparkinson Agents/adverse effects , Central Nervous System/physiopathology , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Animals , Central Nervous System/drug effects , Humans , Parkinson Disease/drug therapy
13.
J Neurochem ; 132(2): 159-68, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319628

ABSTRACT

Zinc (Zn(2+) ) is believed to play a relevant role in the physiology and pathophysiology of the brain. Hence, Zn(2+) homeostasis is critical and involves different classes of molecules, including Zn(2+) transporters. The ubiquitous Zn(2+) transporter-1 (ZNT-1) is a transmembrane protein that pumps cytosolic Zn(2+) to the extracellular space, but its function in the central nervous system is not fully understood. Here, we show that ZNT-1 interacts with GluN2A-containing NMDA receptors, suggesting a role for this transporter at the excitatory glutamatergic synapse. First, we found that ZNT-1 is highly expressed at the hippocampal postsynaptic density (PSD) where NMDA receptors are enriched. Two-hybrid screening, coimmunoprecipitation experiments and clustering assay in COS-7 cells demonstrated that ZNT-1 specifically binds the GluN2A subunit of the NMDA receptor. GluN2A deletion mutants and pull-down assays indicated GluN2A(1390-1464) domain as necessary for the binding to ZNT-1. Most importantly, ZNT-1/GluN2A complex was proved to be dynamic, since it was regulated by induction of synaptic plasticity. Finally, modulation of ZNT-1 expression in hippocampal neurons determined a significant change in dendritic spine morphology, PSD-95 clusters and GluN2A surface levels, supporting the involvement of ZNT-1 in the dynamics of excitatory PSD. Zn(2+) transporter-1 (ZNT-1) pumps cytosolic Zn(2+) to the extracellular space, but its function in the central nervous system is not fully understood. We show that ZNT-1 interacts with GluN2A-containing NMDA receptors at the glutamatergic synapse. Most importantly, ZNT-1/GluN2A complex is regulated by induction of synaptic plasticity. Modulation of ZNT-1 expression in hippocampal neurons determined a shrinkage of dendritic spines and a reduction of GluN2A surface levels supporting the involvement of ZNT-1 in the dynamics of the excitatory synapse.


Subject(s)
Cation Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Post-Synaptic Density/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/cytology , Hippocampus/metabolism , Ion Transport , Male , Neuronal Plasticity , Pregnancy , Primary Cell Culture , Protein Interaction Mapping , Rats , Rats, Sprague-Dawley , Zinc/metabolism
14.
J Neurosci ; 33(46): 18175-89, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24227726

ABSTRACT

Intracellular tau aggregates are the neuropathological hallmark of several neurodegenerative diseases, including Alzheimer's disease, progressive supranuclear palsy, and cases of frontotemporal dementia, but the link between these aggregates and neurodegeneration remains unclear. Neuronal models recapitulating the main features of tau pathology are necessary to investigate the molecular mechanisms of tau malfunction, but current models show little and inconsistent spontaneous tau aggregation. We show that dorsal root ganglion (DRG) neurons in transgenic mice expressing human P301S tau (P301S-htau) develop tau pathology similar to that found in brain and spinal cord and a significant reduction in mechanosensation occurs before detectable fibrillar tau formation. DRG neuronal cultures established from adult P301S-htau mice at different ages retained the pattern of aberrant tau found in vivo. Moreover, htau became progressively hyperphosphorylated over 2 months in vitro beginning with nonsymptomatic neurons, while hyperphosphorylated P301S-htau-positive neurons from 5-month-old mice cultured for 2 months died preferentially. P301S-htau-positive neurons grew aberrant axons, including spheroids, typically found in human tauopathies. Neurons cultured at advanced stages of tau pathology showed a 60% decrease in the fraction of moving mitochondria. SEG28019, a novel O-GlcNAcase inhibitor, reduced steady-state pSer396/pSer404 phosphorylation over 7 weeks in a significant proportion of DRG neurons showing for the first time the possible beneficial effect of prolonged dosing of O-GlcNAcase inhibitor in vitro. Our system is unique in that fibrillar tau forms without external manipulation and provides an important new tool for understanding the mechanisms of tau dysfunction and for screening of compounds for treatment of tauopathies.


Subject(s)
Sensory Receptor Cells/metabolism , Tauopathies/metabolism , beta-N-Acetylhexosaminidases/antagonists & inhibitors , tau Proteins/biosynthesis , Animals , Cells, Cultured , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Tauopathies/drug therapy , Tauopathies/genetics , Tauopathies/pathology , beta-N-Acetylhexosaminidases/metabolism , tau Proteins/genetics
15.
Eur J Pharmacol ; 719(1-3): 75-83, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-23872417

ABSTRACT

There is general agreement that excessive activation of N-methyl-D-aspartate (NMDA) receptors plays a key role in mediating at least some aspects of synaptic dysfunction in several central nervous system disorders. On this view, in the last decades, research focused on the discovery of different compounds able to reduce NMDA receptor activity, such as classical and/or subunit-specific antagonists. However, the increasing body of knowledge on specific signaling pathways downstream NMDA receptors led to the identification of new pharmacological targets for NMDA receptor-related pathological conditions. Moreover, besides over-activation, several studies indicated that also abnormal NMDA receptor trafficking, resulting in the modification of the receptor subunit composition at the synapse, has a major role in the pathogenesis of several brain disorders. For this reason, the discovery of the molecular mechanisms regulating the abundance of synaptic versus extra-synaptic NMDA receptors as well as the activation of the specific signaling pathways downstream the different NMDA receptor subtypes is needed for the development of novel therapeutic approaches for NMDA receptor-dependent synaptic dysfunction.


Subject(s)
Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Molecular Targeted Therapy/methods , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Central Nervous System Diseases/pathology , Humans , Protein Transport/drug effects , Signal Transduction/drug effects , Synapses/drug effects
16.
Apoptosis ; 17(3): 289-304, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22160861

ABSTRACT

Stem cell (SC) transplantation represents a promising tool to treat neurodegenerative disorders, such as Parkinson's disease (PD), but positive therapeutic outcomes require elucidation of the biological mechanisms involved. Therefore, we investigated human Mesenchymal SCs (hMSCs) ability to protect murine differentiated Neural SCs (mdNSCs) against the cytotoxic effects of 6-hydroxydopamine (6-OHDA) in a co-culture model mimicking the in vivo neurovascular niche. The internalization of 6-OHDA mainly relies on its uptake by the dopamine active transporter (DAT), but its toxicity could also involve other pathways. We demonstrated that mdNSCs consistently expressed DAT along the differentiative process. Exposure to 6-OHDA did not affect hMSCs, but induced DAT-independent apoptosis in mdNSCs with generation of reactive oxygen species and caspases 3/7 activation. The potential neuroprotective action of hMSCs on mdNSCs exposed to 6-OHDA was tested in different co-culture conditions, in which hMSCs were added to mdNSCs prior to, simultaneously, or after 6-OHDA treatment. In the presence of the neurotoxin, the majority of mdNSCs acquired an apoptotic phenotype, while co-cultures with hMSCs significantly increased their survival (up to 70%) in all conditions. Multiplex human angiogenic array analysis on the conditioned media demonstrated that cytokine release by hMSCs was finely modulated. Moreover, sole growth factor addition yielded a similar neuroprotective effect on mdNSCs. In conclusion, our findings demonstrate that hMSCs protect mdNSCs against 6-OHDA neurotoxicity, and rescue cells from ongoing neurodegeneration likely through the release of multiple cytokines. Our findings provide novel insights for the development of therapeutic strategies designed to counteract the neurodegenerative processes of PD.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neural Stem Cells/drug effects , Oxidopamine/toxicity , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Parkinson Disease/therapy , Rats
17.
Cell Transplant ; 19(2): 203-17, 2010.
Article in English | MEDLINE | ID: mdl-19906332

ABSTRACT

Stem cells have been increasingly recognized as a potential tool to replace or support cells damaged by the neurodegenerative process that underlies Parkinson's disease (PD). In this frame, human adult mesenchymal stem cells (hMSCs) have been proposed as an attractive alternative to heterologous embryonic or neural precursor cells. To address this issue, in this study we implanted undifferentiated hMSCs into the striatum of rats bearing a lesion of the nigrostriatal pathway induced by local injection of 6-hydroxydopamine (6-OHDA), a widely recognized rodent model of PD. Before grafting, cultured hMSCs expressed markers of both undifferentiated and committed neural cells, including nestin, GAP-43, NSE, beta-tubulin III, and MAP-2, as well as several cytokine mRNAs. No glial or specific neuronal markers were detected. Following transplantation, some hMSCs acquired a glial-like phenotype, as shown by immunoreactivity for glial fibrillary acid protein (GFAP), but only in animals bearing the nigrostriatal lesion. More importantly, rats that received the striatal graft showed increased survival of both cell bodies and terminals of dopaminergic, nigrostriatal neurons, coupled with a reduction of the behavioral abnormalities (apomorphine-induced turning behavior) associated with the lesion. No differentiation of the MSCs toward a neuronal (dopaminergic) phenotype was observed in vivo. In conclusion, our results suggest that grafted hMSCs exert neuroprotective effects against nigrostriatal degeneration induced by 6-OHDA. The mechanisms underlying this effect remain to be clarified, although it is likely that the acquisition of a glial phenotype by grafted hMSCs may lead to the release of prosurvival cytokines within the lesioned striatum.


Subject(s)
Corpus Striatum , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Oxidopamine/toxicity , Substantia Nigra , Animals , Behavior, Animal/physiology , Biomarkers/metabolism , Cell Differentiation/physiology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Male , Mesenchymal Stem Cells/cytology , Neuroprotective Agents/metabolism , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...