Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
EMBO Rep ; 25(3): 1233-1255, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38413732

ABSTRACT

Accumulation of amyloid-beta (Aß) can lead to the formation of aggregates that contribute to neurodegeneration in Alzheimer's disease (AD). Despite globally reduced neural activity during AD onset, recent studies have suggested that Aß induces hyperexcitability and seizure-like activity during the early stages of the disease that ultimately exacerbate cognitive decline. However, the underlying mechanism is unknown. Here, we reveal an Aß-induced elevation of postsynaptic density protein 95 (PSD-95) in cultured neurons in vitro and in an in vivo AD model using APP/PS1 mice at 8 weeks of age. Elevation of PSD-95 occurs as a result of reduced ubiquitination caused by Akt-dependent phosphorylation of E3 ubiquitin ligase murine-double-minute 2 (Mdm2). The elevation of PSD-95 is consistent with the facilitation of excitatory synapses and the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors induced by Aß. Inhibition of PSD-95 corrects these Aß-induced synaptic defects and reduces seizure activity in APP/PS1 mice. Our results demonstrate a mechanism underlying elevated seizure activity during early-stage Aß pathology and suggest that PSD-95 could be an early biomarker and novel therapeutic target for AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Animals , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Disease Models, Animal , Mice, Transgenic , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Receptors, AMPA/metabolism , Seizures
2.
Cell Death Dis ; 13(5): 437, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35513389

ABSTRACT

Aberrantly synchronized neuronal discharges in the brain lead to epilepsy, a devastating neurological disease whose pathogenesis and mechanism are unclear. SAPAP3, a cytoskeletal protein expressed at high levels in the postsynaptic density (PSD) of excitatory synapses, has been well studied in the striatum, but the role of SAPAP3 in epilepsy remains elusive. In this study, we sought to investigate the molecular, cellular, electrophysiological and behavioral consequences of SAPAP3 perturbations in the mouse hippocampus. We identified a significant increase in the SAPAP3 levels in patients with temporal lobe epilepsy (TLE) and in mouse models of epilepsy. In addition, behavioral studies showed that the downregulation of SAPAP3 by shRNA decreased the seizure severity and that the overexpression of SAPAP3 by recombinant SAPAP3 yielded the opposite effect. Moreover, SAPAP3 affected action potentials (APs), miniature excitatory postsynaptic currents (mEPSCs) and N-methyl-D-aspartate receptor (NMDAR)-mediated currents in the CA1 region, which indicated that SAPAP3 plays an important role in excitatory synaptic transmission. Additionally, the levels of the GluN2A protein, which is involved in synaptic function, were perturbed in the hippocampal PSD, and this perturbation was accompanied by ultrastructural morphological changes. These results revealed a previously unknown function of SAPAP3 in epileptogenesis and showed that SAPAP3 may represent a novel target for the treatment of epilepsy.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Animals , Epilepsy/metabolism , Epilepsy, Temporal Lobe/pathology , Hippocampus/metabolism , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/metabolism , Synapses/metabolism
3.
Anesthesiology ; 133(4): 812-823, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32773681

ABSTRACT

BACKGROUND: Experimental evidence shows postnatal exposure to anesthesia negatively affects brain development. The PDZ2 domain, mediating protein-protein interactions of the postsynaptic density-95 protein, serves as a molecular target for several inhaled anesthetics. The authors hypothesized that early postnatal disruption of postsynaptic density-95 PDZ2 domain interactions has persistent effects on dendritic spines and cognitive function. METHODS: One-week-old mice were exposed to 1.5% isoflurane for 4 h or injected with 8 mg/kg active postsynaptic density-95 wild-type PDZ2 peptide along with their respective controls. A subset of these mice also received 4 mg/kg of the nitric oxide donor molsidomine. Hippocampal spine density, long-term potentiation, novel object recognition memory, and fear learning and memory were evaluated in mice. RESULTS: Exposure of 7-day-old mice to isoflurane or postsynaptic density-95 wild-type PDZ2 peptide relative to controls causes: (1) a long-term decrease in mushroom spines at 7 weeks (mean ± SD [spines per micrometer]): control (0.8 ± 0.2) versus isoflurane (0.4 ± 0.2), P < 0.0001, and PDZ2MUT (0.7 ± 0.2) versus PDZ2WT (0.4 ± 0.2), P < 0.001; (2) deficits in object recognition at 6 weeks (mean ± SD [recognition index]): naïve (70 ± 8) versus isoflurane (55 ± 14), P = 0.010, and control (65 ± 13) versus isoflurane (55 ± 14), P = 0.045, and PDZ2MUT (64 ±11) versus PDZ2WT (53 ± 18), P = 0.045; and (3) deficits in fear learning at 7 weeks and memory at 8 weeks (mean ± SD [% freezing duration]): Learning, control (69 ± 12) versus isoflurane (52 ± 13), P < 0.0001, and PDZ2MUT (65 ± 14) versus PDZ2WT (55 ± 14) P = 0.011, and Memory, control (80 ± 17) versus isoflurane (56 ± 23), P < 0.0001 and PDZ2MUT (73 ± 18) versus PDZ2WT (44 ± 19) P < 0.0001. Impairment in long-term potentiation has fully recovered here at 7 weeks (mean ± SD [% baseline]): control (140 ± 3) versus isoflurane (137 ± 8), P = 0.560, and PDZ2MUT (136 ± 17) versus PDZ2WT (128 ± 11), P = 0.512. The isoflurane induced decrease in mushroom spines was preventable by introduction of a nitric oxide donor. CONCLUSIONS: Early disruption of PDZ2 domain-mediated protein-protein interactions mimics isoflurane in decreasing mushroom spine density and causing learning and memory deficits in mice. Prevention of the decrease in mushroom spine density with a nitric oxide donor supports a role for neuronal nitric oxide synthase pathway in mediating this cellular change associated with cognitive impairment.


Subject(s)
Anesthetics, Inhalation/toxicity , Cognition/drug effects , Dendritic Spines/drug effects , Disks Large Homolog 4 Protein/antagonists & inhibitors , Isoflurane/toxicity , Animals , Animals, Newborn , Cognition/physiology , Dendritic Spines/pathology , Dendritic Spines/physiology , Disks Large Homolog 4 Protein/physiology , Female , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Peptides/pharmacology , Post-Synaptic Density/drug effects , Post-Synaptic Density/pathology , Post-Synaptic Density/physiology
4.
Biomed Res Int ; 2019: 5249675, 2019.
Article in English | MEDLINE | ID: mdl-31687391

ABSTRACT

The broad spectrum of disabilities caused by white matter injury (WMI) cannot be explained simply by hypomyelination. Synaptic injury in the thalamus may be related to disabilities in WMI survivors. Neuronal injury in the thalamus has been found most commonly in autopsy cases of preterm WMI. We hypothesized that hypoxia/ischemia (HI) in neonatal rats results in synaptic abnormalities in the thalamus that contribute to disabilities in WMI survivors. We examined changes in synapses in a neonatal rat model of HI-induced WMI. Right common carotid artery ligation and hypoxia (8% oxygen for 2.5 hours (h)) were performed in three-day-old Sprague-Dawley rats. We found HI rats performed worse in the Morris water maze test than sham rats, suggesting long-term cognition impairment after HI injury. A loss of synapses in the thalamus accompanied by hypomyelination and oligodendrocytes (OLs) reduction was observed. At the ultrastructural level, reductions in active zone (AZ) length and postsynaptic density (PSD) thickness were detected at 2 weeks after HI exposure. Furthermore, increased expression of synaptophysin and PSD-95 in both groups was observed from 3 days (d) to 21 d after hypoxic/ischemic (HI) injury. PSD-95 expression was significantly lower in HI rats than in sham rats from 14 d to 21 d after HI injury, and synaptophysin expression was significantly lower in HI rats from 7 d to 14 d after HI injury. However, no significant difference in synaptophysin expression was observed between HI rats and sham rats at 21 d after HI injury. The results demonstrated synaptic abnormalities in the thalamus accompanied by hypomyelination in WMI in response to HI exposure, which may contribute to the diverse neurological defects observed in WMI patients. Although synaptic reorganization occurred as a compensatory response to HI injury, the impairments in synaptic transmission were not reversed.


Subject(s)
Brain Injuries/pathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia/pathology , Synapses/pathology , Thalamus/pathology , White Matter/pathology , Animals , Animals, Newborn , Disease Models, Animal , Maze Learning/physiology , Oligodendroglia/pathology , Post-Synaptic Density/pathology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
5.
Sci Rep ; 9(1): 14837, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619689

ABSTRACT

Alzheimer's disease (AD)-associated synaptic dysfunction drives the progression of pathology from its earliest stages. Amyloid ß (Aß) species, both soluble and in plaque deposits, have been causally related to the progressive, structural and functional impairments observed in AD. It is, however, still unclear how Aß plaques develop over time and how they progressively affect local synapse density and turnover. Here we observed, in a mouse model of AD, that Aß plaques grow faster in the earlier stages of the disease and if their initial area is >500 µm2; this may be due to deposition occurring in the outer regions of the plaque, the plaque cloud. In addition, synaptic turnover is higher in the presence of amyloid pathology and this is paralleled by a reduction in pre- but not post-synaptic densities. Plaque proximity does not appear to have an impact on synaptic dynamics. These observations indicate an imbalance in the response of the pre- and post-synaptic terminals and that therapeutics, alongside targeting the underlying pathology, need to address changes in synapse dynamics.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Plaque, Amyloid/pathology , Post-Synaptic Density/pathology , Presynaptic Terminals/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Transgenic , Mutation
6.
J Clin Invest ; 129(6): 2390-2403, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063986

ABSTRACT

A disintegrine and metalloproteinase 10 (ADAM10) is implicated in synaptic function through its interaction with postsynaptic receptors and adhesion molecules. Here, we report that levels of active ADAM10 are increased in Huntington's disease (HD) mouse cortices and striata and in human postmortem caudate. We show that, in the presence of polyglutamine-expanded (polyQ-expanded) huntingtin (HTT), ADAM10 accumulates at the postsynaptic densities (PSDs) and causes excessive cleavage of the synaptic protein N-cadherin (N-CAD). This aberrant phenotype is also detected in neurons from HD patients where it can be reverted by selective silencing of mutant HTT. Consistently, ex vivo delivery of an ADAM10 synthetic inhibitor reduces N-CAD proteolysis and corrects electrophysiological alterations in striatal medium-sized spiny neurons (MSNs) of 2 HD mouse models. Moreover, we show that heterozygous conditional deletion of ADAM10 or delivery of a competitive TAT-Pro-ADAM10709-729 peptide in R6/2 mice prevents N-CAD proteolysis and ameliorates cognitive deficits in the mice. Reduction in synapse loss was also found in R6/2 mice conditionally deleted for ADAM10. Taken together, these results point to a detrimental role of hyperactive ADAM10 at the HD synapse and provide preclinical evidence of the therapeutic potential of ADAM10 inhibition in HD.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Cognitive Dysfunction/enzymology , Huntington Disease/enzymology , Membrane Proteins/metabolism , Post-Synaptic Density/enzymology , ADAM10 Protein/genetics , Adult , Aged , Amyloid Precursor Protein Secretases/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Disease Models, Animal , Female , HEK293 Cells , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Male , Membrane Proteins/genetics , Mice, Transgenic , Middle Aged , Post-Synaptic Density/genetics , Post-Synaptic Density/pathology
7.
J Clin Invest ; 129(2): 820-833, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30667373

ABSTRACT

Molecular signaling mechanisms underlying Alzheimer's disease (AD) remain unclear. Maintenance of memory and synaptic plasticity depend on de novo protein synthesis, dysregulation of which is implicated in AD. Recent studies showed AD-associated hyperphosphorylation of mRNA translation factor eukaryotic elongation factor 2 (eEF2), which results in inhibition of protein synthesis. We tested to determine whether suppression of eEF2 phosphorylation could improve protein synthesis capacity and AD-associated cognitive and synaptic impairments. Genetic reduction of the eEF2 kinase (eEF2K) in 2 AD mouse models suppressed AD-associated eEF2 hyperphosphorylation and improved memory deficits and hippocampal long-term potentiation (LTP) impairments without altering brain amyloid ß (Aß) pathology. Furthermore, eEF2K reduction alleviated AD-associated defects in dendritic spine morphology, postsynaptic density formation, de novo protein synthesis, and dendritic polyribosome assembly. Our results link eEF2K/eEF2 signaling dysregulation to AD pathophysiology and therefore offer a feasible therapeutic target.


Subject(s)
Alzheimer Disease , Dendritic Spines , Elongation Factor 2 Kinase , Long-Term Potentiation , Post-Synaptic Density , Signal Transduction/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Dendritic Spines/genetics , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Peptide Elongation Factor 2/genetics , Peptide Elongation Factor 2/metabolism , Phosphorylation/genetics , Post-Synaptic Density/genetics , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology
8.
Neuron ; 100(6): 1322-1336.e7, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30392797

ABSTRACT

Synapse loss and Tau pathology are hallmarks of Alzheimer's disease (AD) and other tauopathies, but how Tau pathology causes synapse loss is unclear. We used unbiased proteomic analysis of postsynaptic densities (PSDs) in Tau-P301S transgenic mice to identify Tau-dependent alterations in synapses prior to overt neurodegeneration. Multiple proteins and pathways were altered in Tau-P301S PSDs, including depletion of a set of GTPase-regulatory proteins that leads to actin cytoskeletal defects and loss of dendritic spines. Furthermore, we found striking accumulation of complement C1q in the PSDs of Tau-P301S mice and AD patients. At synapses, C1q decorated perisynaptic membranes, accumulated in correlation with phospho-Tau, and was associated with augmented microglial engulfment of synapses and decline of synapse density. A C1q-blocking antibody inhibited microglial synapse removal in cultured neurons and in Tau-P301S mice, rescuing synapse density. Thus, inhibiting complement-mediated synapse removal by microglia could be a potential therapeutic target for Tau-associated neurodegeneration.


Subject(s)
Antibodies/therapeutic use , Complement C1q/immunology , Synapses/metabolism , Tauopathies/drug therapy , Tauopathies/pathology , tau Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Complement C1q/metabolism , Complement C1q/ultrastructure , Embryo, Mammalian , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Post-Synaptic Density/ultrastructure , Presenilin-2/genetics , Presenilin-2/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Proteome/metabolism , Rats , Synapses/drug effects , Synapses/ultrastructure , Tauopathies/diagnostic imaging , Tauopathies/genetics , tau Proteins/genetics
9.
J Alzheimers Dis ; 65(2): 659-682, 2018.
Article in English | MEDLINE | ID: mdl-30103319

ABSTRACT

Some individuals, here referred to as Non-Demented with Alzheimer's Neuropathology (NDAN), retain their cognitive function despite the presence of amyloid plaques and tau tangles typical of symptomatic Alzheimer's disease (AD). In NDAN, unlike AD, toxic amyloid-ß oligomers do not localize to the postsynaptic densities (PSDs). Synaptic resistance to amyloid-ß in NDAN may thus enable these individuals to remain cognitively intact despite the AD-like pathology. The mechanism(s) responsible for this resistance remains unresolved and understanding such protective biological processes could reveal novel targets for the development of effective treatments for AD. The present study uses a proteomic approach to compare the hippocampal postsynaptic densities of NDAN, AD, and healthy age-matched persons to identify protein signatures characteristic for these groups. Subcellular fractionation followed by 2D gel electrophoresis and mass spectrometry were used to analyze the PSDs. We describe fifteen proteins which comprise the unique proteomic signature of NDAN PSDs, thus setting them apart from control subjects and AD patients.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Post-Synaptic Density/metabolism , Proteome/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Electrophoresis, Gel, Two-Dimensional , Female , Hippocampus/pathology , Humans , Male , Mass Spectrometry , Post-Synaptic Density/pathology , Proteomics , Subcellular Fractions
10.
Biochemistry ; 57(27): 4005-4009, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29913061

ABSTRACT

The strength of each excitatory synapse in the central nervous system is regulated by its prior activity in a process called synaptic plasticity. The initiation of synaptic plasticity occurs when calcium ions enter the postsynaptic compartment and encounter a subcellular structure called the postsynaptic density (PSD). The PSD is attached to the postsynaptic membrane just underneath the concentrated plaque of neurotransmitter receptors. It is comprised of a core set of 30-60 proteins, approximately 20 of which are scaffold proteins. The rest include protein kinases and phosphatases, some of which respond to calcium ion; small GTPases and their regulators; chaperones; ubiquitins; and proteases. The assembly of the PSD involves competitive binding among a variety of specific protein binding sites to form a dynamic network. A biochemical challenge for the future is to understand how the dynamic regulation of the structure, composition, and activity of the PSD mediates synaptic plasticity and how mutations in PSD proteins lead to mental and neurodegenerative diseases.


Subject(s)
Glutamic Acid/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Post-Synaptic Density/metabolism , Animals , Humans , Long-Term Potentiation , Mental Disorders/genetics , Mental Disorders/metabolism , Mental Disorders/pathology , Mutation , Nerve Net/cytology , Nerve Net/pathology , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Post-Synaptic Density/genetics , Post-Synaptic Density/pathology
11.
Sci Rep ; 8(1): 2281, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396406

ABSTRACT

The scaffold protein DLGAP1 is localized at the post-synaptic density (PSD) of glutamatergic neurons and is a component of supramolecular protein complexes organized by PSD95. Gain-of-function variants of DLGAP1 have been associated with obsessive-compulsive disorder (OCD), while haploinsufficient variants have been linked to autism spectrum disorder (ASD) and schizophrenia in human genetic studies. We tested male and female Dlgap1 wild type (WT), heterozygous (HT), and knockout (KO) mice in a battery of behavioral tests: open field, dig, splash, prepulse inhibition, forced swim, nest building, social approach, and sucrose preference. We also used biochemical approaches to examine the role of DLGAP1 in the organization of PSD protein complexes. Dlgap1 KO mice were most notable for disruption of protein interactions in the PSD, and deficits in sociability. Other behavioral measures were largely unaffected. Our data suggest that Dlgap1 knockout leads to PSD disruption and reduced sociability, consistent with reports of DLGAP1 haploinsufficient variants in schizophrenia and ASD.


Subject(s)
Mice, Knockout , Neurons/pathology , Post-Synaptic Density/pathology , SAP90-PSD95 Associated Proteins/deficiency , Social Behavior , Animals , Behavior, Animal , Female , Male , Protein Binding
12.
Mol Psychiatry ; 23(3): 735-746, 2018 03.
Article in English | MEDLINE | ID: mdl-28948968

ABSTRACT

Shank3 is a structural protein found predominantly at the postsynaptic density. Mutations in the SHANK3 gene have been associated with risk for autism spectrum disorder (ASD). We generated induced pluripotent stem cells (iPSCs) from control individuals and from human donors with ASD carrying microdeletions of SHANK3. In addition, we used Zinc finger nucleases to generate isogenic SHANK3 knockout human embryonic stem (ES) cell lines. We differentiated pluripotent cells into either cortical or olfactory placodal neurons. We show that patient-derived placodal neurons make fewer synapses than control cells. Moreover, patient-derived cells display a developmental phenotype: young postmitotic neurons have smaller cell bodies, more extensively branched neurites, and reduced motility compared with controls. These phenotypes were mimicked by SHANK3-edited ES cells and rescued by transduction with a Shank3 expression construct. This developmental phenotype is not observed in the same iPSC lines differentiated into cortical neurons. Therefore, we suggest that SHANK3 has a critical role in neuronal morphogenesis in placodal neurons and that early defects are associated with ASD-associated mutations.


Subject(s)
Autism Spectrum Disorder/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/pathology , Autism Spectrum Disorder/pathology , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Chromosome Deletion , Excitatory Postsynaptic Potentials/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mutation , Neural Stem Cells/metabolism , Neurons/metabolism , Neurons/pathology , Post-Synaptic Density/pathology , Synapses/metabolism , Synapses/pathology , Synaptic Transmission
13.
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
14.
Neurobiol Dis ; 106: 158-170, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28688852

ABSTRACT

Mutations in the Cyclin-dependent kinase-like 5 (CDKL5) gene cause severe neurodevelopmental disorders accompanied by intractable epilepsies, i.e. West syndrome or atypical Rett syndrome. Here we report generation of the Cdkl5 knockout mouse and show that CDKL5 controls postsynaptic localization of GluN2B-containing N-methyl-d-aspartate (NMDA) receptors in the hippocampus and regulates seizure susceptibility. Cdkl5 -/Y mice showed normal sensitivity to kainic acid; however, they displayed significant hyperexcitability to NMDA. In concordance with this result, electrophysiological analysis in the hippocampal CA1 region disclosed an increased ratio of NMDA/α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated excitatory postsynaptic currents (EPSCs) and a significantly larger decay time constant of NMDA receptor-mediated EPSCs (NMDA-EPSCs) as well as a stronger inhibition of the NMDA-EPSCs by the GluN2B-selective antagonist ifenprodil in Cdkl5 -/Y mice. Subcellular fractionation of the hippocampus from Cdkl5 -/Y mice revealed a significant increase of GluN2B and SAP102 in the PSD (postsynaptic density)-1T fraction, without changes in the S1 (post-nuclear) fraction or mRNA transcripts, indicating an intracellular distribution shift of these proteins to the PSD. Immunoelectron microscopic analysis of the hippocampal CA1 region further confirmed postsynaptic overaccumulation of GluN2B and SAP102 in Cdkl5 -/Y mice. Furthermore, ifenprodil abrogated the NMDA-induced hyperexcitability in Cdkl5 -/Y mice, suggesting that upregulation of GluN2B accounts for the enhanced seizure susceptibility. These data indicate that CDKL5 plays an important role in controlling postsynaptic localization of the GluN2B-SAP102 complex in the hippocampus and thereby regulates seizure susceptibility, and that aberrant NMDA receptor-mediated synaptic transmission underlies the pathological mechanisms of the CDKL5 loss-of-function.


Subject(s)
Hippocampus/metabolism , Post-Synaptic Density/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/metabolism , Animals , Disease Models, Animal , Disease Susceptibility/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Guanylate Kinases/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Kainic Acid , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate , Piperidines/pharmacology , Post-Synaptic Density/drug effects , Post-Synaptic Density/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/pathology , Tissue Culture Techniques
15.
Neural Plast ; 2017: 3270725, 2017.
Article in English | MEDLINE | ID: mdl-28458925

ABSTRACT

Synaptic loss is the structural basis for memory impairment in Alzheimer's disease (AD). While the underlying pathological mechanism remains elusive, it is known that misfolded proteins accumulate as ß-amyloid (Aß) plaques and hyperphosphorylated Tau tangles decades before the onset of clinical disease. The loss of Pin1 facilitates the formation of these misfolded proteins in AD. Pin1 protein controls cell-cycle progression and determines the fate of proteins by the ubiquitin proteasome system. The activity of the ubiquitin proteasome system directly affects the functional and structural plasticity of the synapse. We localized Pin1 to dendritic rafts and postsynaptic density (PSD) and found the pathological loss of Pin1 within the synapses of AD brain cortical tissues. The loss of Pin1 activity may alter the ubiquitin-regulated modification of PSD proteins and decrease levels of Shank protein, resulting in aberrant synaptic structure. The loss of Pin1 activity, induced by oxidative stress, may also render neurons more susceptible to the toxicity of oligomers of Aß and to excitation, thereby inhibiting NMDA receptor-mediated synaptic plasticity and exacerbating NMDA receptor-mediated synaptic degeneration. These results suggest that loss of Pin1 activity could lead to the loss of synaptic plasticity in the development of AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neuronal Plasticity , Post-Synaptic Density/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/pathology , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disks Large Homolog 4 Protein/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Post-Synaptic Density/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Ubiquitin/metabolism , tau Proteins/metabolism
16.
Biochem Biophys Res Commun ; 486(3): 671-678, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28336441

ABSTRACT

Chronic stress is a precipitating factor for disorders including depression. The basolateral amygdala (BLA) is a critical substrate that interconnects with stress-modulated neural networks to generate emotion- and mood-related behaviors. The current study shows that 3 h per day of restraint stress for 14 days caused mice to exhibit long-term depressive behaviors, manifested by disrupted sociality and despair levels, which were rescued by fluoxetine. These behavioral changes corresponded with morphological and molecular changes in BLA neurons, including chronic stress-elicited increases in arborization, dendritic length, and spine density of BLA principal neurons. At the molecular level, calcium-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (CP-AMPARs) within the synaptosome exhibited an increased GluR1:GluR2 subunit ratio. We also observed increased GluR1 phosphorylation at Ser 845 and enhanced cyclic AMP-dependent protein kinase (PKA) activity in the BLA. These molecular changes reverted to the basal state post-treatment with fluoxetine. The expression of synaptophysin (SYP) and postsynaptic density protein 95 (PSD-95) at BLA neuronal synapses was also enhanced by chronic stress, which was reversed post-treatment. Finally, chronic stress-provoked depressive behavior was overcome by local blockage of CP-AMPARs in the BLA via stereotaxic injection (IEM-1460). Chronic stress-elicited depressive behavior may be due to hypertrophy of BLA neuronal dendrites and increased of PKA-dependent CP-AMPAR levels in BLA neurons. Furthermore, fluoxetine can reverse chronic stress-triggered cytoarchitectural and functional changes of BLA neurons. These findings provide insights into depression-linked structural and functional changes in BLA neurons.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Depression/genetics , Post-Synaptic Density/metabolism , Receptors, AMPA/genetics , Stress, Psychological/genetics , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Antidepressive Agents/pharmacology , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Depression/metabolism , Depression/physiopathology , Depression/prevention & control , Disease Models, Animal , Disks Large Homolog 4 Protein , Fluoxetine/pharmacology , Gene Expression Regulation , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phosphorylation , Post-Synaptic Density/drug effects , Post-Synaptic Density/pathology , Receptors, AMPA/metabolism , Signal Transduction , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Stress, Psychological/prevention & control , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Synaptophysin/genetics , Synaptophysin/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/pathology
17.
Dev Neurosci ; 39(1-4): 66-81, 2017.
Article in English | MEDLINE | ID: mdl-28315865

ABSTRACT

Proteomics of the synapses and postsynaptic densities (PSDs) have provided a deep understanding of protein composition and signal networks in the adult brain, which underlie neuronal plasticity and neurodegenerative or psychiatric disorders. However, there is a paucity of knowledge about the architecture and organization of PSDs in the immature brain, and how it is modified by brain injury in an early developing stage. Mass spectrometry (MS)-based proteomic analysis was performed on PSDs prepared from cortices of postnatal day 9 naïve mice or pups which had suffered hypoxic-ischemic (HI) brain injury. 512 proteins of different functional groups were identified from PSDs collected 1 h after HI injury, among which 60 have not been reported previously. Seven newly identified proteins involved in neural development were highlighted. HI injury increased the yield of PSDs at early time points upon reperfusion, and multiple proteins were recruited into PSDs following the insult. Quantitative analysis was performed using spectral counting, and proteins whose relative expression was more than 50% up- or downregulated compared to the sham animals 1 h after HI insult were reported. Validation with Western blotting demonstrated changes in expression and phosphorylation of the N-methyl-D-aspartate receptor, activation of a series of postsynaptic protein kinases and dysregulation of scaffold and adaptor proteins in response to neonatal HI insult. This work, along with other recent studies of synaptic protein profiling in the immature brain, builds a foundation for future investigation on the molecular mechanisms underlying developing plasticity. Furthermore, it provides insights into the biochemical changes of PSDs following early brain hypoxia-ischemia, which is helpful for understanding not only the injury mechanisms, but also the process of repair or replenishment of neuronal circuits during recovery from brain damage.


Subject(s)
Asphyxia Neonatorum/pathology , Cerebral Cortex/pathology , Hypoxia-Ischemia, Brain/pathology , Post-Synaptic Density/pathology , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Cerebral Cortex/metabolism , Female , Humans , Hypoxia-Ischemia, Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Post-Synaptic Density/metabolism , Proteomics
18.
Brain Res ; 1657: 232-244, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28027875

ABSTRACT

Inherited mitochondrial complex 1 deficiency causes Leber's hereditary Optic Neuropathy (LHON) and retinal ganglion cell (RGC) degeneration, and optic neuropathies are common in many inherited mitochondrial diseases. How mitochondrial defects pathomechanistically trigger optic neuropathy remains unclear. We observe that complex 1-deficient Ndufs4-/- mice present with acute vision loss around p30, and this vision loss is coincident with an 'inflammatory wave'. In order to understand what causes the inflammatory wave we explored retinal pathology that occurs from p20-p30. The results indicated that in the period p20-p30 in Ndufs4-/- retinas, there is: significant reduction in bipolar cells, RGC dendritic atrophy, reduced PSD95, increased oxidative stress as manifested by increased 4HNE, HO1 and Cuzn-SOD, increased mitochondrial biogenesis and increased apoptosis. These precede the major induction of 'inflammatory wave' at p30 shown previously, but occur earlier than frank RGC loss at p42. In general, complex 1 deficiency in retina triggers oxidative stress and mitochondrial respiratory dysfunction that causes death of the most sensitive cells, including bipolar cells and their synaptic contacts and amacrine cells in the early period, 20-24days. The early death of these cells is the likely precursor to the sharp rise in inflammatory molecules that occurs at day 30 and coincides with vision loss, and greatly precedes the death of RGCs that occurs at p42. These data suggest that metabolic antioxidant support of the most sensitive cells in the retina, or anti-inflammatory suppression of the consequences of their death, are both rational strategies for mitochondrial blinding disease.


Subject(s)
Electron Transport Complex I/deficiency , Mitochondria/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Amacrine Cells/metabolism , Amacrine Cells/pathology , Animals , Apoptosis/physiology , Atrophy , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Disease Progression , Electron Transport Complex I/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Optic Atrophy, Hereditary, Leber/metabolism , Optic Atrophy, Hereditary, Leber/pathology , Organelle Biogenesis , Oxidative Stress/physiology , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Retinal Bipolar Cells/pathology , Retinal Ganglion Cells/pathology
19.
Biochim Biophys Acta ; 1862(10): 1918-25, 2016 10.
Article in English | MEDLINE | ID: mdl-27460706

ABSTRACT

Autism is a severe neurodevelopmental disorder with a large population prevalence, characterized by abnormal reciprocal social interactions, communication deficits, and repetitive behaviors with restricted interests. The BTBR T(+)Itpr3(tf) (BTBR) mice have emerged as strong candidates to serve as models of a range of autism-relevant behaviors. Increasing evidences suggest that interleukin (IL)-6, one of the most important neuroimmune factors, was involved in the pathophysiology of autism. It is of great importance to further investigate whether therapeutic interventions in autism can be achieved through the manipulation of IL-6. Our previous studies showed that IL-6 elevation in the brain could mediate autistic-like behaviors, possibly through the imbalances of neural circuitry and impairments of synaptic plasticity. In this study, we evaluate whether inhibiting IL-6 signaling in the brain is sufficient to modulate the autism-like behaviors on the BTBR mice. The results showed that chronic infusion of an analog of the endogenous IL-6 trans-signaling blocker sgp130Fc protein increased the sociability in BTBR mice. Furthermore, no change was observed in the number of excitatory synapse, level of synaptic proteins, density of dentitic spine and postsynaptic density in BTBR cortices after inhibiting IL-6 trans-signaling. However, inhibition of IL-6 trans-signaling increased the evoked glutamate release in synaptoneurosomes from the cerebral cortex of BTBR mice. Our findings suggest that inhibition of excessive production of IL-6 may have selective therapeutic efficacy in treating abnormal social behaviors in autism.


Subject(s)
Autistic Disorder/metabolism , Behavior, Animal , Cerebral Cortex/metabolism , Interleukin-6/metabolism , Neuronal Plasticity , Animals , Autistic Disorder/drug therapy , Autistic Disorder/genetics , Autistic Disorder/pathology , Cerebral Cortex/pathology , Cytokine Receptor gp130/therapeutic use , Disease Models, Animal , Humans , Immunoglobulin Fc Fragments/pharmacology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Mice , Mice, Transgenic , Post-Synaptic Density/genetics , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Recombinant Proteins/pharmacology , Signal Transduction
20.
J Pineal Res ; 59(2): 163-77, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25988948

ABSTRACT

Translational research on Alzheimer's disease (AD) has often focused on reducing the high cerebral levels of amyloid-ß (Aß) as a key characteristic of AD pathogenesis. There is, however, a growing body of evidence that synaptic dysfunction may be crucial for the development of the most common (sporadic) form of AD. The applicability of melatonin (mainly produced by the pineal gland) to the treatment of AD is actively evaluated, but usually, such studies are based on animal models of early-onset AD, which is responsible for only ~5% of AD cases. We have shown previously that in OXYS rats (an established model of sporadic AD), accumulation of toxic forms of Aß in the brain occurs later than does the development of signs of neurodegenerative changes and synaptic failure. In this regard, recently, we uncovered beneficial neuroprotective effects of melatonin (prophylactic dietary supplementation) in OXYS rats. Our aim here was to evaluate, starting at the age of active progression of AD-like pathology in OXYS rats, the effects of long-term oral administration of melatonin on the structure of synapses and on neuronal and glial cells of the hippocampus. Melatonin significantly increased hippocampal synaptic density and the number of excitatory synapses, decreased the number of inhibitory synapses, and upregulated pre- and postsynaptic proteins (synapsin I and PSD-95, respectively). Furthermore, melatonin improved the ultrastructure of neuronal and glial cells and reduced glial density. Based on our past and present results, the repair of neuroplasticity by melatonin is a promising strategy against AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Melatonin/metabolism , Neuronal Plasticity , Post-Synaptic Density/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Male , Post-Synaptic Density/pathology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL