Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Glia ; 71(4): 974-990, 2023 04.
Article in English | MEDLINE | ID: mdl-36480007

ABSTRACT

Triggering receptor on myeloid cells 2 (TREM2) is an innate immune receptor, upregulated on the surface of microglia associated with amyloid plaques in Alzheimer's disease (AD). Individuals heterozygous for the R47H variant of TREM2 have greatly increased risk of developing AD. We examined the effects of wild-type (WT), R47H and knock-out (KO) of human TREM2 expression in three microglial cell systems. Addition of mouse BV-2 microglia expressing R47H TREM2 to primary mouse neuronal cultures caused neuronal loss, not observed with WT TREM2. Neuronal loss was prevented by using annexin V to block exposed phosphatidylserine, an eat-me signal and ligand of TREM2, suggesting loss was mediated by microglial phagocytosis of neurons exposing phosphatidylserine. Addition of human CHME-3 microglia expressing R47H TREM2 to LUHMES neuronal-like cells also caused loss compared to WT TREM2. Expression of R47H TREM2 in BV-2 and CHME-3 microglia increased their uptake of phosphatidylserine-beads and synaptosomes versus WT TREM2. Human iPSC-derived microglia with heterozygous R47H TREM2 had increased phagocytosis of synaptosomes vs common-variant TREM2. Additionally, phosphatidylserine liposomes increased activation of human iPSC-derived microglia expressing homozygous R47H TREM2 versus common-variant TREM2. Finally, overexpression of TREM2 in CHME-3 microglia caused increased expression of cystatin F, a cysteine protease inhibitor, and knock-down of cystatin F increased CHME-3 uptake of phosphatidylserine-beads. Together, these data suggest that R47H TREM2 may increase AD risk by increasing phagocytosis of synapses and neurons via greater activation by phosphatidylserine and that WT TREM2 may decrease microglial phagocytosis of synapses and neurons via cystatin F.


Subject(s)
Alzheimer Disease , Synaptosomes , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cystatins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microglia/metabolism , Neurons/pathology , Phagocytosis/genetics , Phosphatidylserines/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Synaptosomes/metabolism , Synaptosomes/pathology
2.
J Neurochem ; 159(3): 525-542, 2021 11.
Article in English | MEDLINE | ID: mdl-34379806

ABSTRACT

Sepsis-associated encephalopathy (SAE) represents diverse cerebral dysfunctions in response to pathogen-induced systemic inflammation. Peripheral exposure to lipopolysaccharide (LPS), a component of the gram-negative bacterial cell wall, has been extensively used to model systemic inflammation. Our previous studies suggested that LPS led to hippocampal neuron death and synaptic destruction in vivo. However, the underlying roles of activated microglia in these neuronal changes remained unclear. Here, LPS from two different bacterial strains (Salmonella enterica or E. coli) were compared and injected in 14- to 16-month-old mice and evaluated for neuroinflammation and neuronal integrity in the hippocampus at 7 or 63 days post-injection (dpi). LPS injection resulted in persistent neuroinflammation lasting for seven days and a subsequent normalisation by 63 dpi. Of note, increases in proinflammatory cytokines, microglial morphology and microglial mean lysosome volume were more pronounced after E. coli LPS injection than Salmonella LPS at 7 dpi. While inhibitory synaptic puncta density remained normal, excitatory synaptic puncta were locally reduced in the CA3 region of the hippocampus at 63 dpi. Finally, we provide evidence that excitatory synapses coated with complement factor 3 (C3) decreased between 7 dpi and 63 dpi. Although we did not find an increase of synaptic pruning by microglia, it is plausible that microglia recognised and eliminated these C3-tagged synapses between the two time points of investigation. Since a region-specific decline of CA3 synapses has previously been reported during normal ageing, we postulate that systemic inflammation may have accelerated or worsened the CA3 synaptic changes in the ageing brain.


Subject(s)
Aging/pathology , CA3 Region, Hippocampal/pathology , Inflammation/pathology , Synapses/pathology , Animals , Female , Immunohistochemistry , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Microglia/pathology , Salmonella , Sepsis/pathology , Synaptosomes/pathology
3.
Cell Mol Life Sci ; 77(24): 5243-5258, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32034429

ABSTRACT

Synaptic functional disturbances with concomitant synapse loss represent central pathological hallmarks of Alzheimer's disease. Excessive accumulation of cytotoxic amyloid oligomers is widely recognized as a key event that underlies neurodegeneration. Certain complement components are crucial instruments of widespread synapse loss because they can tag synapses with functional impairments leading to their engulfment by microglia. However, an exact understanding of the affected synaptic functions that predispose to complement-mediated synapse elimination is lacking. Therefore, we conducted systematic proteomic examinations on synaptosomes prepared from an amyloidogenic mouse model of Alzheimer's disease (APP/PS1). Synaptic fractions were separated according to the presence of the C1q-tag using fluorescence-activated synaptosome sorting and subjected to proteomic comparisons. The results raised the decline of mitochondrial functions in the C1q-tagged synapses of APP/PS1 mice based on enrichment analyses, which was verified using flow cytometry. Additionally, proteomics results revealed extensive alterations in the level of septin protein family members, which are known to dynamically form highly organized pre- and postsynaptic supramolecular structures, thereby affecting synaptic transmission. High-resolution microscopy investigations demonstrated that synapses with considerable amounts of septin-3 and septin-5 show increased accumulation of C1q in APP/PS1 mice compared to the wild-type ones. Moreover, a strong positive correlation was apparent between synaptic septin-3 levels and C1q deposition as revealed via flow cytometry and confocal microscopy examinations. In sum, our results imply that deterioration of synaptic mitochondrial functions and alterations in the organization of synaptic septins are associated with complement-dependent synapse loss in Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Amyloid/metabolism , Proteome/genetics , Synapses/genetics , Alzheimer Disease/pathology , Amyloid/toxicity , Amyloidogenic Proteins/genetics , Animals , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Microglia/metabolism , Microglia/pathology , Mitochondria/genetics , Mitochondria/pathology , Oligopeptides/genetics , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Septins/genetics , Synapses/metabolism , Synapses/pathology , Synaptosomes/metabolism , Synaptosomes/pathology
4.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502320

ABSTRACT

Retinitis pigmentosa (RP) is a family of inherited disorders caused by the progressive degeneration of retinal photoreceptors. There is no cure for RP, but recent research advances have provided promising results from many clinical trials. All these therapeutic strategies are focused on preserving existing photoreceptors or substituting light-responsive elements. Vision recovery, however, strongly relies on the anatomical and functional integrity of the visual system beyond photoreceptors. Although the retinal structure and optic pathway are substantially preserved at least in early stages of RP, studies describing the visual cortex status are missing. Using a well-established mouse model of RP, we analyzed the response of visual cortical circuits to the progressive degeneration of photoreceptors. We demonstrated that the visual cortex goes through a transient and previously undescribed alteration in the local excitation/inhibition balance, with a net shift towards increased intracortical inhibition leading to improved filtering and decoding of corrupted visual inputs. These results suggest a compensatory action of the visual cortex that increases the range of residual visual sensitivity in RP.


Subject(s)
Neurotransmitter Agents/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinitis Pigmentosa/pathology , Synaptosomes/pathology , Visual Cortex/physiopathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Retinitis Pigmentosa/etiology , Retinitis Pigmentosa/metabolism , Synaptosomes/metabolism
5.
Am J Pathol ; 189(8): 1621-1636, 2019 08.
Article in English | MEDLINE | ID: mdl-31108099

ABSTRACT

Apolipoprotein E (apoE) colocalizes with amyloid-ß (Aß) in Alzheimer disease (AD) plaques and in synapses, and evidence suggests that direct interactions between apoE and Aß are important for apoE's effects in AD. The present work examines the hypothesis that apoE receptors mediate uptake of apoE/Aß complex into synaptic terminals. Western blot analysis shows multiple SDS-stable assemblies in synaptosomes from human AD cortex; apoE/Aß complex was markedly increased in AD compared with aged control samples. Complex formation between apoE and Aß was confirmed by coimmunoprecipitation experiments. The apoE receptors low-density lipoprotein receptor (LDLR) and LDLR-related protein 1 (LRP1) were quantified in synaptosomes using flow cytometry, revealing up-regulation of LRP1 in early- and late-stage AD. Dual-labeling flow cytometry analysis of LRP1- and LDLR positives indicate most (approximately 65%) of LDLR and LRP1 is associated with postsynaptic density-95 (PSD-95)-positive synaptosomes, indicating that remaining LRP1 and LDLR receptors are exclusively presynaptic. Flow cytometry analysis of Nile red labeling revealed a reduction in cholesterol esters in AD synaptosomes. Dual-labeling experiments showed apoE and Aß concentration into LDLR and LRP1-positive synaptosomes, along with free and esterified cholesterol. Synaptic Aß was increased by apoE4 in control and AD samples. These results are consistent with uptake of apoE/Aß complex and associated lipids into synaptic terminals, with subsequent Aß clearance in control synapses and accumulation in AD synapses.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/metabolism , Apolipoproteins E/metabolism , Cerebral Cortex/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Receptors, LDL/metabolism , Synapses/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Cerebral Cortex/pathology , Disks Large Homolog 4 Protein/metabolism , Female , Humans , Male , Middle Aged , Synapses/pathology , Synaptosomes/metabolism , Synaptosomes/pathology
6.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33227902

ABSTRACT

Synaptic aging has been associated with neuronal circuit dysfunction and cognitive decline. Reduced mitochondrial function may be an early event that compromises synaptic integrity and neurotransmission in vulnerable brain regions during physiological and pathological aging. Thus, we aimed to measure mitochondrial function in synapses from three brain regions at two different ages in the 3xTg-AD mouse model and in wild mice. We found that aging is the main factor associated with the decline in synaptic mitochondrial function, particularly in synapses isolated from the cerebellum. Accumulation of toxic compounds, such as tau and Aß, that occurred in the 3xTg-AD mouse model seemed to participate in the worsening of this decline in the hippocampus. The changes in synaptic bioenergetics were also associated with increased activation of the mitochondrial fission protein Drp1. These results suggest the presence of altered mechanisms of synaptic mitochondrial dynamics and their quality control during aging and in the 3xTg-AD mouse model; they also point to bioenergetic restoration as a useful therapeutic strategy to preserve synaptic function during aging and at the early stages of Alzheimer's disease (AD).


Subject(s)
Aging/genetics , Cognitive Dysfunction/genetics , Dynamins/genetics , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Aging/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Cerebellum/metabolism , Cerebellum/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Dynamins/metabolism , Female , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Transgenic , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Organ Specificity , Synapses/metabolism , Synapses/pathology , Synaptosomes/metabolism , Synaptosomes/pathology , tau Proteins/genetics , tau Proteins/metabolism
7.
J Proteome Res ; 18(8): 3042-3051, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31262178

ABSTRACT

Spinal muscular atrophy (SMA) is a human genetic disorder characterized by muscle weakness, muscle atrophy, and death of motor neurons. SMA is caused by mutations or deletions in a gene called survival motor neuron 1 (SMN1). SMN1 is a housekeeping gene, but the most prominent pathologies in SMA are atrophy of myofibers and death of motor neurons. Further, degeneration of neuromuscular junctions, of synapses, and of axonal regions are features of SMA disease. Here, we have investigated the proteome dynamics of central synapses in P14 Smn2B/- mice, a model of SMA. Label-free quantitative proteomics on isolated synaptosomes from spinal cords of these animals identified 2030 protein groups. Statistical data analysis revealed 65 specific alterations in the proteome of the central synapses at the early onset stage of disease. Functional analysis of the dysregulated proteins indicated a significant enrichment of proteins associated with mitochondrial dynamics, cholesterol biogenesis, and protein clearance. These pathways represent potential targets for therapy development with the goal of providing stability to the central synapses, thereby preserving neuronal integrity in the context of SMA disease. Data are available via ProteomeXchange with identifier PXD012850.


Subject(s)
Muscular Atrophy, Spinal/genetics , Proteome/genetics , Proteomics , Synaptosomes/metabolism , Animals , Disease Models, Animal , Humans , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Synapses/genetics , Synapses/pathology , Synaptosomes/pathology
8.
J Biol Chem ; 292(50): 20644-20654, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29074619

ABSTRACT

Chemotherapeutic drugs such as paclitaxel cause painful peripheral neuropathy in many cancer patients and survivors. Although NMDA receptors (NMDARs) at primary afferent terminals are known to be critically involved in chemotherapy-induced chronic pain, the upstream signaling mechanism that leads to presynaptic NMDAR activation is unclear. Group I metabotropic glutamate receptors (mGluRs) play a role in synaptic plasticity and NMDAR regulation. Here we report that the Group I mGluR agonist (S)-3,5-dihydroxyphenylglycine (DHPG) significantly increased the frequency of miniature excitatory postsynaptic currents (EPSCs) and the amplitude of monosynaptic EPSCs evoked from the dorsal root. DHPG also reduced the paired-pulse ratio of evoked EPSCs in spinal dorsal horn neurons. These effects were blocked by the selective mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP), but not by an mGluR1 antagonist. MPEP normalized the frequency of miniature EPSCs and the amplitude of evoked EPSCs in paclitaxel-treated rats but had no effect in vehicle-treated rats. Furthermore, mGluR5 protein levels in the dorsal root ganglion and spinal cord synaptosomes were significantly higher in paclitaxel- than in vehicle-treated rats. Inhibiting protein kinase C (PKC) or blocking NMDARs abolished DHPG-induced increases in the miniature EPSC frequency of spinal dorsal horn neurons in vehicle- and paclitaxel-treated rats. Moreover, intrathecal administration of MPEP reversed pain hypersensitivity caused by paclitaxel treatment. Our findings suggest that paclitaxel-induced painful neuropathy is associated with increased presynaptic mGluR5 activity at the spinal cord level, which serves as upstream signaling for PKC-mediated tonic activation of NMDARs. mGluR5 is therefore a promising target for reducing chemotherapy-induced neuropathic pain.


Subject(s)
Nerve Tissue Proteins/metabolism , Neuralgia/metabolism , Neurons, Afferent/metabolism , Protein Kinase C/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord Dorsal Horn/metabolism , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Behavior, Animal/drug effects , Cells, Cultured , Evoked Potentials/drug effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Glycine/analogs & derivatives , Glycine/pharmacology , Injections, Spinal , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/pathology , Neurons, Afferent/drug effects , Neurons, Afferent/pathology , Paclitaxel/adverse effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Pyridines/therapeutic use , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Resorcinols/pharmacology , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/pathology , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/pathology
9.
Biochim Biophys Acta Proteins Proteom ; 1866(2): 307-315, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29174846

ABSTRACT

Bartha, the pseudorabies virus (PRV) vaccine strain, is widely used in studies of neuronal circuit-tracing, due to its attenuated virulence and retrograde spreading. However, we know little regarding the molecular mechanisms of PRV infection and spreading between structurally connected neurons. In this study, we systematically analyzed the host brain proteomes after acute infection with PRV, attempting to identified the proteins involved in the processes. Mice were injected with PRV-Bartha and PRV-Becker (PRV-Bartha's wild-type parent strain) in the olfactory system, the proteomes of the brain and synaptosome were analyzed and compared at various infection intervals using mass spectrometry-based proteomics techniques. In all, we identified >100 PRV-infection regulated proteins at the whole-tissue level and the synaptosome level. While at whole-tissue level, bioinformatics analyses mapped most of the regulations to the inflammation pathways, at the synaptosome level, most of those to synaptic transmission, cargo transport and cytoskeleton organization. We established regulated protein networks demonstrating distinct cellular regulation pattern between the global and the synaptosome levels. Moreover, we identified a series of potentially PRV-strain-specific regulated proteins with diverse biological functions. This study may provide new clues for molecular mechanisms for PRV infection and spread.


Subject(s)
Brain/metabolism , Herpesvirus 1, Suid/metabolism , Nerve Tissue Proteins/metabolism , Proteomics , Pseudorabies/metabolism , Synaptosomes/metabolism , Animals , Brain/pathology , Brain/virology , Male , Mice , Pseudorabies/pathology , Synaptosomes/pathology , Synaptosomes/virology
10.
Mol Cell Proteomics ; 15(2): 506-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26598648

ABSTRACT

Synapse disruption takes place in many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the mechanistic understanding of this process is still limited. We set out to study a possible role for dynein in synapse integrity. Cytoplasmic dynein is a multisubunit intracellular molecule responsible for diverse cellular functions, including long-distance transport of vesicles, organelles, and signaling factors toward the cell center. A less well-characterized role dynein may play is the spatial clustering and anchoring of various factors including mRNAs in distinct cellular domains such as the neuronal synapse. Here, in order to gain insight into dynein functions in synapse integrity and disruption, we performed a screen for novel dynein interactors at the synapse. Dynein immunoprecipitation from synaptic fractions of the ALS model mSOD1(G93A) and wild-type controls, followed by mass spectrometry analysis on synaptic fractions of the ALS model mSOD1(G93A) and wild-type controls, was performed. Using advanced network analysis, we identified Staufen1, an RNA-binding protein required for the transport and localization of neuronal RNAs, as a major mediator of dynein interactions via its interaction with protein phosphatase 1-beta (PP1B). Both in vitro and in vivo validation assays demonstrate the interactions of Staufen1 and PP1B with dynein, and their colocalization with synaptic markers was altered as a result of two separate ALS-linked mutations: mSOD1(G93A) and TDP43(A315T). Taken together, we suggest a model in which dynein's interaction with Staufen1 regulates mRNA localization along the axon and the synapses, and alterations in this process may correlate with synapse disruption and ALS toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cytoplasmic Dyneins/genetics , Proteomics , RNA-Binding Proteins/biosynthesis , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/metabolism , Axons/pathology , Cytoplasmic Dyneins/metabolism , Disease Models, Animal , Humans , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation , RNA-Binding Proteins/genetics , Synapses/genetics , Synapses/metabolism , Synapses/pathology , Synaptosomes/metabolism , Synaptosomes/pathology
11.
J Biol Chem ; 291(9): 4374-85, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26719332

ABSTRACT

Although trace levels of phosphorylated α-synuclein (α-syn) are detectable in normal brains, nearly all α-syn accumulated within Lewy bodies in Parkinson disease brains is phosphorylated on serine 129 (Ser-129). The role of the phosphoserine residue and its effects on α-syn structure, function, and intracellular accumulation are poorly understood. Here, co-expression of α-syn and polo-like kinase 2 (PLK2), a kinase that targets Ser-129, was used to generate phosphorylated α-syn for biophysical and biological characterization. Misfolding and fibril formation of phosphorylated α-syn isoforms were detected earlier, although the fibrils remained phosphatase- and protease-sensitive. Membrane binding of α-syn monomers was differentially affected by phosphorylation depending on the Parkinson disease-linked mutation. WT α-syn binding to presynaptic membranes was not affected by phosphorylation, whereas A30P α-syn binding was greatly increased, and A53T α-syn was slightly lower, implicating distal effects of the carboxyl- on amino-terminal membrane binding. Endocytic vesicle-mediated internalization of pre-formed fibrils into non-neuronal cells and dopaminergic neurons matched the efficacy of α-syn membrane binding. Finally, the disruption of internalized vesicle membranes was enhanced by the phosphorylated α-syn isoforms, a potential means for misfolded extracellular or lumenal α-syn to access cytosolic α-syn. Our results suggest that the threshold for vesicle permeabilization is evident even at low levels of α-syn internalization and are relevant to therapeutic strategies to reduce intercellular propagation of α-syn misfolding.


Subject(s)
Endocytosis , Parkinson Disease/genetics , Protein Aggregation, Pathological/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Synaptosomes/metabolism , alpha-Synuclein/metabolism , Amino Acid Substitution , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Mesencephalon/cytology , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mutation , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Folding , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Synaptosomes/pathology , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
12.
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
13.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28621878

ABSTRACT

Arthropod venoms are sources of molecules that may be useful tools to investigate molecular mechanisms of putative new medicines and laboratory drugs. Here we show the effects of the compound agelaiatoxin-8 (AVTx8), isolated from Agelaia vicina venom, on γ-aminobutyric acid (GABA) neurotransmission in rat brain synaptosomes. Analysis reveals that AvTx8 is composed by 14 amino acid residues with a molecular weight (MW) of 1567 Da. AvTx8 increased GABA release and inhibited GABA uptake in synaptosomes from rat cerebral cortex. AvTx8 inhibited GABA uptake and increased GABA release in the presence of Ca+ , Na+ , and K+ channel blockers, suggesting that it acts directly on GABA transporters. In addition, AvTx8 significantly decreases GABA binding in synaptic membranes from rat brain cortex, suggesting that it also modulates the activity of GABA receptors. Moreover, AvTx8 decreased GAT-1- and GAT-3-mediated GABA uptake in transfected COS-7 cells. Accordingly, we suggest that AvTx8 modulates GABA neurotransmission and might provide a novel entry point for identifying a new class of GABA-modulating neuroprotective drugs.


Subject(s)
Synaptic Membranes/metabolism , Synaptic Transmission/drug effects , Synaptosomes/metabolism , Wasp Venoms , Wasps/chemistry , gamma-Aminobutyric Acid/metabolism , Animals , COS Cells , Chlorocebus aethiops , Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , Rats , Rats, Wistar , Synaptic Membranes/pathology , Synaptosomes/pathology , Wasp Venoms/chemistry , Wasp Venoms/isolation & purification , Wasp Venoms/toxicity
14.
Brain Behav Immun ; 56: 289-309, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27058163

ABSTRACT

An increasing number of studies have revealed associations between pre- and perinatal immune activation and the development of schizophrenia and autism spectrum disorders (ASDs). Accordingly, neuroimmune crosstalk has a considerably large impact on brain development during early ontogenesis. While a plethora of heterogeneous abnormalities have already been described in established maternal immune activation (MIA) rodent and primate animal models, which highly correlate to those found in human diseases, the underlying molecular background remains obscure. In the current study, we describe the long-term effects of MIA on the neocortical pre- and postsynaptic proteome of adolescent rat offspring in detail. Molecular differences were revealed in sub-synaptic fractions, which were first thoroughly characterized using independent methods. The widespread proteomic examination of cortical samples from offspring exposed to maternal lipopolysaccharide administration at embryonic day 13.5 was conducted via combinations of different gel-based proteomic techniques and tandem mass spectrometry. Our experimentally validated proteomic data revealed more pre- than postsynaptic protein level changes in the offspring. The results propose the relevance of altered synaptic vesicle recycling, cytoskeletal structure and energy metabolism in the presynaptic region in addition to alterations in vesicle trafficking, the cytoskeleton and signal transduction in the postsynaptic compartment in MIA offspring. Differing levels of the prominent signaling regulator molecule calcium/calmodulin-dependent protein kinase II in the postsynapse was validated and identified specifically in the prefrontal cortex. Finally, several potential common molecular regulators of these altered proteins, which are already known to be implicated in schizophrenia and ASD, were identified and assessed. In summary, unexpectedly widespread changes in the synaptic molecular machinery in MIA rats were demonstrated which might underlie the pathological cortical functions that are characteristic of schizophrenia and ASD.


Subject(s)
Prefrontal Cortex/metabolism , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Proteome/metabolism , Synapses/metabolism , Synaptosomes/metabolism , Animals , Autism Spectrum Disorder/etiology , Disease Models, Animal , Female , Lipopolysaccharides/pharmacology , Male , Pregnancy , Proteomics/methods , Rats , Rats, Wistar , Schizophrenia/etiology , Synapses/pathology , Synaptosomes/pathology
15.
Brain ; 138(Pt 7): 2005-19, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25981964

ABSTRACT

Endophilin-B1, also known as Bax-interacting factor 1 (Bif-1, and encoded by SH3GLB1), is a multifunctional protein involved in apoptosis, autophagy and mitochondrial function. We recently described a unique neuroprotective role for neuron-specific alternatively spliced isoforms of endophilin-B1. To examine whether endophilin-B1-mediated neuroprotection could be a novel therapeutic target for Alzheimer's disease we used a double mutant amyloid precursor protein and presenilin 1 (APPswe/PSEN1dE9) mouse model of Alzheimer's disease and observed that expression of neuron-specific endophilin-B1 isoforms declined with disease progression. To determine if this reduction in endophilin-B1 has a functional role in Alzheimer's disease pathogenesis, we crossed endophilin-B1(-/-) mice with APPswe/PSEN1dE9 mice. Deletion of endophilin-B1 accelerated disease onset and progression in 6-month-old APPswe/PSEN1dE9/endophilin-B1(-/-) mice, which showed more plaques, astrogliosis, synaptic degeneration, cognitive impairment and mortality than APPswe/PSEN1dE9 mice. In mouse primary cortical neuron cultures, overexpression of neuron-specific endophilin-B1 isoforms protected against amyloid-ß-induced apoptosis and mitochondrial dysfunction. Additionally, protein and mRNA levels of neuron-specific endophilin-B1 isoforms were also selectively decreased in the cerebral cortex and in the synaptic compartment of patients with Alzheimer's disease. Flow sorting of synaptosomes from patients with Alzheimer's disease demonstrated a negative correlation between amyloid-ß and endophilin-B1 levels. The importance of endophilin-B1 in neuronal function was further underscored by the development of synaptic degeneration and cognitive and motor impairment in endophilin-B1(-/-) mice by 12 months. Our findings suggest that endophilin-B1 is a key mediator of a feed-forward mechanism of Alzheimer's disease pathogenesis where amyloid-ß reduces neuron-specific endophilin-B1, which in turn enhances amyloid-ß accumulation and neuronal vulnerability to stress.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Neurons/pathology , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoblotting , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synaptosomes/metabolism , Synaptosomes/pathology
16.
IUBMB Life ; 67(4): 306-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25914026

ABSTRACT

Advanced grades of glioblastoma are highly aggressive, especially in terms of multisite spread within the brain or even to distant sites at the spinal cord. In advanced grades of glioblastoma, glutamate and glutamine are reported to be increased in concentration in the extracellular fluid. It has been reported that glutamate acts as an extracellular signaling molecule for facilitating local spread of advanced grades of glioblastoma. In the present study, we aimed to examine whether glutamate uptake mechanisms is impaired in advanced glioblastoma. The possible downregulated mechanisms of glutamate uptake would facilitate persistence of glutamate in the extracellular environment, rather than intracellular uptake. We obtained biobanked human specimens of glioblastoma and tested expression of proteins belonging to the solute carrier families of proteins that are known to function as membrane-located excitatory amino acid like glutamate transporters. The present study provides preliminary evidence of the downregulation of membrane expression of excitatory amino acid transporters solute carrier family 1 member 3 (SLC1A3) and its palmitoylated form in gliosomes, as well as SLC1A2 in the glio-synaptosomes. Compounds like riluzole used in the treatment of amyotrophic lateral sclerosis and the antibiotic ceftriaxone have the potential to facilitate glutamate uptake. These medications may be examined as adjunct chemotherapy in the massively aggressive tumor glioblastoma multiforme.


Subject(s)
Brain Neoplasms/metabolism , Down-Regulation , Excitatory Amino Acid Transporter 1/metabolism , Glioblastoma/metabolism , Glutamates/metabolism , Neoplasm Metastasis , Synaptosomes/metabolism , Aged , Brain Neoplasms/pathology , Case-Control Studies , Glioblastoma/pathology , Humans , Lipoylation , Male , Middle Aged , Synaptosomes/pathology
17.
J Neurochem ; 128(5): 764-75, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24151877

ABSTRACT

Psychostimulant methamphetamine (METH) is toxic to striatal dopaminergic and serotonergic nerve terminals in adult, but not in the adolescent, brain. Betulinic acid (BA) and its derivatives are promising anti-HIV agents with some toxic properties. Many METH users, particularly young men, are HIV-positive; therefore, they might be treated with BA or its derivative for HIV infection. It is not known whether BA, or any of its derivatives, are neurotoxic in combination with METH in the adolescent brain. The present study investigated the effects of BA and binge METH in the striatum of late adolescent rats. BA or METH alone did not decrease the levels of dopaminergic or serotonergic markers in the striatum whereas BA and METH together decreased these markers in a BA dose-dependent manner. BA+METH also caused decreases in the levels of mitochondrial complex I in the same manner; BA alone only slightly decreased the levels of this enzyme in striatal synaptosomes. BA or METH alone increased cytochrome c. METH alone decreased parkin, increased complex II and striatal BA levels. These results suggest that METH in combination with BA can be neurotoxic to striatal dopaminergic and serotonergic nerve terminals in the late adolescent brain via mitochondrial dysfunction and parkin deficit. We report a synergistic neurotoxicity of betulinic acid (BA) and methamphetamine (METH) to monoaminergic terminals in the striatum of male late adolescent rats. BA contribution to the neurotoxicity is decreasing mitochondrial complex I whereas METH contribution is decreasing parkin and increasing brain concentration of BA. We propose that clinical use of BA in young male METH users can be neurotoxic.


Subject(s)
Anti-HIV Agents/toxicity , Dopamine Agents/toxicity , Dopaminergic Neurons/drug effects , Methamphetamine/toxicity , Neostriatum/pathology , Nerve Endings/drug effects , Serotonergic Neurons/drug effects , Triterpenes/toxicity , Animals , Blotting, Western , Brain Chemistry/drug effects , Caspase 3/metabolism , Chymotrypsin/metabolism , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Electron Transport Complex II/drug effects , Electron Transport Complex II/metabolism , Fever/chemically induced , Fever/physiopathology , Male , Mitochondria/drug effects , Mitochondria/pathology , Neostriatum/drug effects , Pentacyclic Triterpenes , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/pathology , Ubiquitin-Protein Ligases/metabolism , Betulinic Acid
18.
J Neuroinflammation ; 11: 39, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24593993

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) enhances pro-inflammatory responses, neuronal loss and long-term behavioral deficits. Caveolins (Cavs) are regulators of neuronal and glial survival signaling. Previously we showed that astrocyte and microglial activation is increased in Cav-1 knock-out (KO) mice and that Cav-1 and Cav-3 modulate microglial morphology. We hypothesized that Cavs may regulate cytokine production after TBI. METHODS: Controlled cortical impact (CCI) model of TBI (3 m/second; 1.0 mm depth; parietal cortex) was performed on wild-type (WT; C57Bl/6), Cav-1 KO, and Cav-3 KO mice. Histology and immunofluorescence microscopy (lesion volume, glia activation), behavioral tests (open field, balance beam, wire grip, T-maze), electrophysiology, electron paramagnetic resonance, membrane fractionation, and multiplex assays were performed. Data were analyzed by unpaired t tests or analysis of variance (ANOVA) with post-hoc Bonferroni's multiple comparison. RESULTS: CCI increased cortical and hippocampal injury and decreased expression of MLR-localized synaptic proteins (24 hours), enhanced NADPH oxidase (Nox) activity (24 hours and 1 week), enhanced polysynaptic responses (1 week), and caused hippocampal-dependent learning deficits (3 months). CCI increased brain lesion volume in both Cav-3 and Cav-1 KO mice after 24 hours (P < 0.0001, n = 4; one-way ANOVA). Multiplex array revealed a significant increase in expression of IL-1ß, IL-9, IL-10, KC (keratinocyte chemoattractant), and monocyte chemoattractant protein 1 (MCP-1) in ipsilateral hemisphere and IL-9, IL-10, IL-17, and macrophage inflammatory protein 1 alpha (MIP-1α) in contralateral hemisphere of WT mice after 4 hours. CCI increased IL-2, IL-6, KC and MCP-1 in ipsilateral and IL-6, IL-9, IL-17 and KC in contralateral hemispheres in Cav-1 KO and increased all 10 cytokines/chemokines in both hemispheres except for IL-17 (ipsilateral) and MIP-1α (contralateral) in Cav-3 KO (versus WT CCI). Cav-3 KO CCI showed increased IL-1ß, IL-9, KC, MCP-1, MIP-1α, and granulocyte-macrophage colony-stimulating factor in ipsilateral and IL-1ß, IL-2, IL-9, IL-10, and IL-17 in contralateral hemispheres (P = 0.0005, n = 6; two-way ANOVA) compared to Cav-1 KO CCI. CONCLUSION: CCI caused astrocyte and microglial activation and hippocampal neuronal injury. Cav-1 and Cav-3 KO exhibited enhanced lesion volume and cytokine/chemokine production after CCI. These findings suggest that Cav isoforms may regulate neuroinflammatory responses and neuroprotection following TBI.


Subject(s)
Brain Injuries/complications , Brain Injuries/pathology , Brain/pathology , Caveolin 1/deficiency , Caveolin 3/deficiency , Encephalitis/complications , Animals , Caveolin 1/genetics , Caveolin 3/genetics , Cells, Cultured , Cognition Disorders/etiology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/genetics , Functional Laterality , Hippocampus/cytology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Movement Disorders/etiology , NADPH Oxidases/metabolism , Neurons/physiology , Synaptosomes/metabolism , Synaptosomes/pathology
19.
Mol Psychiatry ; 18(11): 1225-34, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23399914

ABSTRACT

Genome-wide association studies (GWAS) have identified a region upstream the BIN1 gene as the most important genetic susceptibility locus in Alzheimer's disease (AD) after APOE. We report that BIN1 transcript levels were increased in AD brains and identified a novel 3 bp insertion allele ∼28 kb upstream of BIN1, which increased (i) transcriptional activity in vitro, (ii) BIN1 expression levels in human brain and (iii) AD risk in three independent case-control cohorts (Meta-analysed Odds ratio of 1.20 (1.14-1.26) (P=3.8 × 10(-11))). Interestingly, decreased expression of the Drosophila BIN1 ortholog Amph suppressed Tau-mediated neurotoxicity in three different assays. Accordingly, Tau and BIN1 colocalized and interacted in human neuroblastoma cells and in mouse brain. Finally, the 3 bp insertion was associated with Tau but not Amyloid loads in AD brains. We propose that BIN1 mediates AD risk by modulating Tau pathology.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Genetic Predisposition to Disease/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , tau Proteins/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Endophenotypes , Gene Expression/genetics , Humans , Mice , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nuclear Proteins/biosynthesis , Plaque, Amyloid/pathology , Polymorphism, Single Nucleotide/genetics , Synaptosomes/pathology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/biosynthesis , tau Proteins/antagonists & inhibitors
20.
Cell Biochem Funct ; 32(7): 557-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25111857

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia and is characterized by the presence of senile plaques and neurofibrillary tangles, along with synaptic loss. The underlying mechanisms of AD are not clarified yet, but oxidative stress and mitochondrial dysfunction are important factors. Overactivation of poly(adenosine diphosphate ribose) polymerase-1 (PARP-1) enzyme has been known to cause neuroinflammation and cell death in neurodegenerative processes. The aim of the present study was to investigate the protective effects of the PARP-1 inhibitors, 3-aminobenzamide (3-AB) and nicotinamide (NA), against amyloid ß peptide (1-42) (Aß(1-42))-induced oxidative damage and mitochondrial reduction capacity on isolated synaptosomes. Rats were injected intraperitoneally with 3-AB (30-100 mg kg(-1)), NA (100-500 mg kg(-1)) or with saline for 7 days. Synaptosomes were incubated with 10-30 µM Aß(1-42) or saline for 6 h at 37 °C. Ex vivo Aß(1-42) treatment significantly induced oxidative stress and mitochondrial dysfunction in synaptosomes of the saline group, while synaptosomes of 3-AB and NA groups showed significant decreases in lipid peroxidation, reactive oxygen species production and protein oxidation. Moreover, both NA and 3-AB were able to improve the mitochondrial reduction capacity against Aß(1-42). These data suggest that NA and 3-AB may have protective effects in neurodegenerative processes because of the reduced levels of oxidative stress and the improvement of mitochondrial function.


Subject(s)
Amyloid beta-Peptides/metabolism , Benzamides/pharmacology , Brain/drug effects , Neuroprotective Agents/pharmacology , Niacinamide/pharmacology , Peptide Fragments/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Synaptosomes/drug effects , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Brain/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Rats, Sprague-Dawley , Synaptosomes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL