Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 139
1.
Alzheimers Res Ther ; 15(1): 186, 2023 10 28.
Article En | MEDLINE | ID: mdl-37898760

BACKGROUND: Synapse loss is an early event that precedes neuronal death and symptom onset and is considered the best neuropathological correlate of cognitive decline in Alzheimer's disease (AD). Vesicle-associated membrane protein 2 (VAMP-2) has emerged as a promising biomarker of AD-related synapse degeneration in cerebrospinal fluid (CSF). The aim of this study was to explore the CSF profile of VAMP-2 across the AD continuum in relation to core AD biomarkers, other synaptic proteins, neurogranin (Ng) and synaptosomal-associated Protein-25 kDa (SNAP-25) and cognitive performance. METHODS: We developed a digital immunoassay on the Single Molecule Array platform to quantify VAMP-2 in CSF and used existing immunoassays to quantify Ng, SNAP-25 and core CSF AD biomarkers. The clinical study included 62 cognitively unimpaired AD biomarker-negative subjects and 152 participants across the AD continuum from the SPIN cohort (Sant Pau Initiative on Neurodegeneration). Cognitive measures of episodic, semantic, executive and visuospatial domains and global cognition were included. Statistical methods included χ2 tests, spearman correlation, and ANCOVA analyses. RESULTS: The VAMP-2 assay had a good analytical performance (repeatability 8.9%, intermediate precision 10.3%). Assay antibodies detected native VAMP-2 protein in human brain homogenates. CSF concentrations of VAMP-2, neurogranin and SNAP-25 were lower in preclinical AD stage 1 compared to controls and higher at later AD stages compared to AD stage 1 and were associated with core AD biomarkers, particularly total tau (adj. r2 = 0.62 to 0.78, p < 0.001). All three synaptic proteins were associated with all cognitive domains in individuals on the AD continuum (adj. r2 = 0.04 to 0.19, p < 0.05). CONCLUSIONS: Our novel digital immunoassay accurately measures VAMP-2 changes in CSF, which reflect AD biomarkers and cognitive performance across multiple domains.


Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cognitive Dysfunction/diagnosis , Neurogranin/cerebrospinal fluid , Synaptic Vesicles/pathology , tau Proteins/cerebrospinal fluid , Vesicle-Associated Membrane Protein 2
2.
J Biol Chem ; 299(11): 105282, 2023 11.
Article En | MEDLINE | ID: mdl-37742923

The intracellular domains of connexins are essential for the assembly of gap junctions. For connexin 36 (Cx36), the major neuronal connexin, it has been shown that a dysfunctional PDZ-binding motif interferes with electrical synapse formation. However, it is still unknown how this motif coordinates the transport of Cx36. In the present study, we characterize a phenotype of Cx36 mutants that lack a functional PDZ-binding motif using HEK293T cells as an expression system. We provide evidence that an intact PDZ-binding motif is critical for proper endoplasmic reticulum (ER) export of Cx36. Removing the PDZ-binding motif of Cx36 results in ER retention and the formation of multimembrane vesicles containing gap junction-like connexin aggregates. Using a combination of site-directed mutagenesis and electron micrographs, we reveal that these vesicles consist of Cx36 channels that docked prematurely in the ER. Our data suggest a model in which ER-retained Cx36 channels reshape the ER membrane into concentric whorls that are released into the cytoplasm.


Connexins , Endoplasmic Reticulum , Gap Junctions , Humans , Connexins/genetics , Connexins/metabolism , Endoplasmic Reticulum/metabolism , Gap Junctions/metabolism , HEK293 Cells , Protein Domains , Amino Acid Motifs , Electrical Synapses/physiology , Mutation , Protein Transport/genetics , Synaptic Vesicles/pathology , Synaptic Vesicles/ultrastructure , Microscopy, Electron, Scanning , Gap Junction delta-2 Protein
3.
J Neurochem ; 160(3): 412-425, 2022 02.
Article En | MEDLINE | ID: mdl-34855215

Mutations in the ESCRT-III subunit CHMP2B cause frontotemporal dementia (FTD) and lead to impaired endolysosomal trafficking and lysosomal storage pathology in neurons. We investigated the effect of mutant CHMP2B on synaptic pathology, as ESCRT function was recently implicated in the degradation of synaptic vesicle (SV) proteins. We report here that expression of C-terminally truncated mutant CHMP2B results in a novel synaptopathy. This unique synaptic pathology is characterised by selective retention of presynaptic SV trafficking proteins in aged mutant CHMP2B transgenic mice, despite significant loss of postsynaptic proteins. Furthermore, ultrastructural analysis of primary cortical cultures from transgenic CHMP2B mice revealed a significant increase in the number of presynaptic endosomes, while neurons expressing mutant CHMP2B display defective SV recycling and alterations to functional SV pools. Therefore, we reveal how mutations in CHMP2B affect specific presynaptic proteins and SV recycling, identifying CHMP2B FTD as a novel synaptopathy. This novel synaptopathic mechanism of impaired SV physiology may be a key early event in multiple forms of FTD, since proteins that mediate the most common genetic forms of FTD all localise at the presynapse.


Endosomal Sorting Complexes Required for Transport/genetics , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Nerve Tissue Proteins/genetics , Synapses/pathology , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology , Aging/metabolism , Aging/pathology , Animals , Cerebral Cortex/pathology , Disease Models, Animal , Frontotemporal Dementia/pathology , Mice , Mice, Knockout , Primary Cell Culture , Receptors, Presynaptic/metabolism
4.
Int J Mol Sci ; 22(19)2021 Oct 06.
Article En | MEDLINE | ID: mdl-34639129

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system that finally leads to demyelination. Demyelinating optic neuritis is a frequent symptom in MS. Recent studies also revealed synapse dysfunctions in MS patients and MS mouse models. We previously reported alterations of photoreceptor ribbon synapses in the experimental auto-immune encephalomyelitis (EAE) mouse model of MS. In the present study, we found that the previously observed decreased imunosignals of photoreceptor ribbons in early EAE resulted from a decrease in synaptic ribbon size, whereas the number/density of ribbons in photoreceptor synapses remained unchanged. Smaller photoreceptor ribbons are associated with fewer docked and ribbon-associated vesicles. At a functional level, depolarization-evoked exocytosis as monitored by optical recording was diminished even as early as on day 7 after EAE induction. Moreover compensatory, post-depolarization endocytosis was decreased. Decreased post-depolarization endocytosis in early EAE correlated with diminished synaptic enrichment of dynamin3. In contrast, basal endocytosis in photoreceptor synapses of resting non-depolarized retinal slices was increased in early EAE. Increased basal endocytosis correlated with increased de-phosphorylation of dynamin1. Thus, multiple endocytic pathways in photoreceptor synapse are differentially affected in early EAE and likely contribute to the observed synapse pathology in early EAE.


Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Endocytosis , Exocytosis , Multiple Sclerosis/pathology , Retinal Rod Photoreceptor Cells/pathology , Synapses/pathology , Animals , Dynamins/metabolism , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Phosphorylation , Retina/metabolism , Retina/pathology , Retinal Rod Photoreceptor Cells/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology
5.
Cell Rep ; 36(1): 109333, 2021 07 06.
Article En | MEDLINE | ID: mdl-34233191

While misfolding of alpha-synuclein (αSyn) is central to the pathogenesis of Parkinson's disease (PD), fundamental questions about its structure and function at the synapse remain unanswered. We examine synaptosomes from non-transgenic and transgenic mice expressing wild-type human αSyn, the E46K fPD-causing mutation, or an amplified form of E46K ("3K"). Synaptosomes from mice expressing the 3K mutant show reduced Ca2+-dependent vesicle exocytosis, altered synaptic vesicle ultrastructure, decreased SNARE complexes, and abnormal levels of certain synaptic proteins. With our intra-synaptosomal nuclear magnetic resonance (NMR) method, we reveal that WT αSyn participates in heterogeneous interactions with synaptic components dependent on endogenous αSyn and synaptosomal integrity. The 3K mutation markedly alters these interactions. The synaptic microenvironment is necessary for αSyn to reach its native conformations and establish a physiological interaction network. Its inability to populate diverse conformational ensembles likely represents an early step in αSyn dysfunction that contributes to the synaptotoxicity observed in synucleinopathies.


Parkinson Disease/metabolism , Parkinson Disease/pathology , Synaptic Vesicles/pathology , Synaptosomes/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Animals , Brain/pathology , Calcium/metabolism , Disease Models, Animal , Exocytosis , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Biological , Protein Conformation , Protein Folding , Protein Multimerization , Recombinant Proteins/metabolism , SNARE Proteins/metabolism , Solubility , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomes/ultrastructure
6.
Neuropathol Appl Neurobiol ; 47(7): 1092-1108, 2021 12.
Article En | MEDLINE | ID: mdl-33955002

AIM: To delineate the neurogenetic profiles of brain degeneration patterns in myotonic dystrophy type I (DM1). METHODS: In two cohorts of DM1 patients, brain maps of volume loss (VL) and neuropsychological deficits (NDs) were intersected to large-scale transcriptome maps provided by the Allen Human Brain Atlas (AHBA). For validation, neuropathological and RNA analyses were performed in a small series of DM1 brain samples. RESULTS: Twofold: (1) From a list of preselected hypothesis-driven genes, confirmatory analyses found that three genes play a major role in brain degeneration: dystrophin (DMD), alpha-synuclein (SNCA) and the microtubule-associated protein tau (MAPT). Neuropathological analyses confirmed a highly heterogeneous Tau-pathology in DM1, different to the one in Alzheimer's disease. (2) Exploratory analyses revealed gene clusters enriched for key biological processes in the central nervous system, such as synaptic vesicle recycling, localization, endocytosis and exocytosis, and the serotonin and dopamine neurotransmitter pathways. RNA analyses confirmed synaptic vesicle dysfunction. CONCLUSIONS: The combination of large-scale transcriptome interactions with brain imaging and cognitive function sheds light on the neurobiological mechanisms of brain degeneration in DM1 that might help define future therapeutic strategies and research into this condition.


Brain/pathology , Dystrophin/metabolism , Myotonic Dystrophy/pathology , Synaptic Vesicles/pathology , tau Proteins/metabolism , Adult , Alzheimer Disease/pathology , Brain/metabolism , Central Nervous System/pathology , Female , Humans , Male , Middle Aged , Myotonic Dystrophy/genetics , Synaptic Vesicles/metabolism
7.
Nat Commun ; 12(1): 2107, 2021 04 08.
Article En | MEDLINE | ID: mdl-33833240

Vacuolar H+-ATPases (V-ATPases) transport protons across cellular membranes to acidify various organelles. ATP6V0A1 encodes the a1-subunit of the V0 domain of V-ATPases, which is strongly expressed in neurons. However, its role in brain development is unknown. Here we report four individuals with developmental and epileptic encephalopathy with ATP6V0A1 variants: two individuals with a de novo missense variant (R741Q) and the other two individuals with biallelic variants comprising one almost complete loss-of-function variant and one missense variant (A512P and N534D). Lysosomal acidification is significantly impaired in cell lines expressing three missense ATP6V0A1 mutants. Homozygous mutant mice harboring human R741Q (Atp6v0a1R741Q) and A512P (Atp6v0a1A512P) variants show embryonic lethality and early postnatal mortality, respectively, suggesting that R741Q affects V-ATPase function more severely. Lysosomal dysfunction resulting in cell death, accumulated autophagosomes and lysosomes, reduced mTORC1 signaling and synaptic connectivity, and lowered neurotransmitter contents of synaptic vesicles are observed in the brains of Atp6v0a1A512P/A512P mice. These findings demonstrate the essential roles of ATP6V0A1/Atp6v0a1 in neuronal development in terms of integrity and connectivity of neurons in both humans and mice.


Brain Diseases/genetics , Brain/growth & development , Neurons/physiology , Neurotransmitter Agents/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Animals , Autophagosomes/pathology , Brain Mapping/methods , Cathepsin D/metabolism , Cell Line , HEK293 Cells , Humans , Loss of Function Mutation/genetics , Lysosomes/pathology , Magnetic Resonance Imaging/methods , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mutation, Missense/genetics , Neurons/cytology , Synaptic Vesicles/pathology
8.
Pharmacol Res ; 165: 105469, 2021 03.
Article En | MEDLINE | ID: mdl-33524541

The communication between neurons constitutes the basis of all neural activities, and synaptic vesicle exocytosis is the fundamental biological event that mediates most communication between neurons in the central nervous system. The SNARE complex is the core component of the protein machinery that facilitates the fusion of synaptic vesicles with presynaptic terminals and thereby the release of neurotransmitters. In synapses, each release event is dependent on the assembly of the SNARE complex. In recent years, basic research on the SNARE complex has provided a clearer understanding of the mechanism underlying the formation of the SNARE complex and its role in vesicle formation. Emerging evidence indicates that abnormal expression or dysfunction of the SNARE complex in synapse physiology might contribute to abnormal neurotransmission and ultimately to synaptic dysfunction. Clinical research using postmortem tissues suggests that SNARE complex dysfunction is correlated with various neurological diseases, and some basic research has also confirmed the important role of the SNARE complex in the pathology of these diseases. Genetic and pharmacogenetic studies suggest that the SNARE complex and individual proteins might represent important molecular targets in neurological disease. In this review, we summarize the recent progress toward understanding the SNARE complex in regulating membrane fusion events and provide an update of the recent discoveries from clinical and basic research on the SNARE complex in neurodegenerative, neuropsychiatric, and neurodevelopmental diseases.


Mental Disorders/metabolism , Nervous System Diseases/metabolism , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Animals , Exocytosis/physiology , Humans , Mental Disorders/diagnosis , Mental Disorders/genetics , Nervous System Diseases/diagnosis , Nervous System Diseases/genetics , SNARE Proteins/genetics , Synaptic Vesicles/genetics , Synaptic Vesicles/pathology
9.
J Biol Chem ; 296: 100166, 2021.
Article En | MEDLINE | ID: mdl-33478937

ATP-binding cassette subfamily A member 13 (ABCA13) is predicted to be the largest ABC protein, consisting of 5058 amino acids and a long N-terminal region. Mutations in the ABCA13 gene were reported to increase the susceptibility to schizophrenia, bipolar disorder, and major depression. However, little is known about the molecular functions of ABCA13 or how they associate with psychiatric disorders. Here, we examined the biochemical activity of ABCA13 using HEK293 cells transfected with mouse ABCA13. The expression of ABCA13 induced the internalization of cholesterol and gangliosides from the plasma membrane to intracellular vesicles. Cholesterol internalization by ABCA13 required the long N-terminal region and ATP hydrolysis. To examine the physiological roles of ABCA13, we generated Abca13 KO mice using CRISPR/Cas and found that these mice exhibited deficits of prepulse inhibition. Vesicular cholesterol accumulation and synaptic vesicle endocytosis were impaired in primary cultures of Abca13 KO cortical neurons. Furthermore, mutations in ABCA13 gene associated with psychiatric disorders disrupted the protein's subcellular localization and impaired cholesterol trafficking. These findings suggest that ABCA13 accelerates cholesterol internalization by endocytic retrograde transport in neurons and that loss of this function is associated with the pathophysiology of psychiatric disorders.


ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Endocytosis/genetics , Neurons/metabolism , Prepulse Inhibition , ATP-Binding Cassette Transporters/deficiency , Adenosine Triphosphate/metabolism , Animals , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Disease Models, Animal , Gangliosides/metabolism , Gene Expression , HEK293 Cells , Humans , Hydrolysis , Mice , Mice, Knockout , Mutation , Neurons/pathology , Primary Cell Culture , Protein Transport , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology , Transgenes
10.
Int J Mol Sci ; 21(15)2020 Aug 03.
Article En | MEDLINE | ID: mdl-32756522

Previously, we demonstrated increased calcium levels and synaptic vesicle densities in the motor axon terminals (MATs) of sporadic amyotrophic lateral sclerosis (ALS) patients. Such alterations could be conferred to mice with an intraperitoneal injection of sera from these patients or with purified immunoglobulin G. Later, we confirmed the presence of similar alterations in the superoxide dismutase 1 G93A transgenic mouse strain model of familial ALS. These consistent observations suggested that calcium plays a central role in the pathomechanism of ALS. This may be further reinforced by completing a similar analytical study of the MATs of ALS patients with identified mutations. However, due to the low yield of muscle biopsy samples containing MATs, and the low incidence of ALS patients with the identified mutations, these examinations are not technically feasible. Alternatively, a passive transfer of sera from ALS patients with known mutations was used, and the MATs of the inoculated mice were tested for alterations in their calcium homeostasis and synaptic activity. Patients with 11 different ALS-related mutations participated in the study. Intraperitoneal injection of sera from these patients on two consecutive days resulted in elevated intracellular calcium levels and increased vesicle densities in the MATs of mice, which is comparable to the effect of the passive transfer from sporadic patients. Our results support the idea that the pathomechanism underlying the identical manifestation of the disease with or without identified mutations is based on a common final pathway, in which increasing calcium levels play a central role.


Amyotrophic Lateral Sclerosis/genetics , Axons/metabolism , Motor Neurons/metabolism , Superoxide Dismutase/genetics , Synaptic Vesicles/genetics , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Calcium/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Motor Neurons/pathology , Mutation/genetics , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Synaptic Vesicles/pathology
11.
Sci Rep ; 10(1): 12372, 2020 07 23.
Article En | MEDLINE | ID: mdl-32704004

Diabetes impairs enteric nervous system functions; however, ultrastructural changes underlying the pathophysiology of the myenteric plexus and the effects of sodium-glucose co-transporter (SGLT) inhibitors are poorly understood. This study aimed to investigate three-dimensional ultrastructural changes in axonal varicosities in the myenteric plexus and the effect thereon of the SGLT inhibitor phlorizin in mice fed a high-fat diet (HFD). Three-dimensional ultrastructural analysis using serial block-face imaging revealed that non-treated HFD-fed mice had fewer axonal varicosities and synaptic vesicles in the myenteric plexus than did normal diet-fed control mice. Furthermore, mitochondrial volume was increased and lysosome number decreased in the axons of non-treated HFD-fed mice when compared to those of control mice. Phlorizin treatment restored the axonal varicosities and organelles in HFD-fed mice. Although HFD did not affect the immunolocalisation of PGP9.5, it reduced synaptophysin immunostaining in the myenteric plexus, which was restored by phlorizin treatment. These results suggest that impairment of the axonal varicosities and their synaptic vesicles underlies the damage to the enteric neurons caused by HFD feeding. SGLT inhibitor treatment could restore axonal varicosities and organelles, which may lead to improved gastrointestinal functions in HFD-induced obesity as well as diabetes.


Axons/metabolism , Dietary Fats/adverse effects , Myenteric Plexus/metabolism , Obesity , Phlorhizin/pharmacology , Synaptic Vesicles/metabolism , Animals , Axons/pathology , Dietary Fats/pharmacology , Mice , Myenteric Plexus/pathology , Obesity/chemically induced , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Synaptic Vesicles/pathology , Ubiquitin Thiolesterase/metabolism
12.
Molecules ; 25(10)2020 May 14.
Article En | MEDLINE | ID: mdl-32422902

Many neurological disorders are related to synaptic loss or pathologies. Before the boom of positrons emission tomography (PET) imaging of synapses, synaptic quantification could only be achieved in vitro on brain samples after autopsy or surgical resections. Until the mid-2010s, electron microscopy and immunohistochemical labelling of synaptic proteins were the gold-standard methods for such analyses. Over the last decade, several PET radiotracers for the synaptic vesicle 2A protein have been developed to achieve in vivo synapses visualization and quantification. Different strategies were used, namely radiolabelling with either 11C or 18F, preclinical development in rodent and non-human primates, and binding quantification with different kinetic modelling methods. This review provides an overview of these PET tracers and underlines their perspectives and limitations by focusing on radiochemical aspects, as well as preclinical proof-of-concept and the main clinical outcomes described so far.


Brain/diagnostic imaging , Neurodegenerative Diseases/diagnostic imaging , Positron-Emission Tomography/methods , Pyridines/pharmacokinetics , Pyrrolidines/pharmacokinetics , Pyrrolidinones/pharmacokinetics , Synaptic Vesicles/pathology , Animals , Brain/metabolism , Carbon Radioisotopes , Clinical Trials as Topic , Drug Evaluation, Preclinical , Fluorine Radioisotopes , Humans , Macaca mulatta , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Positron-Emission Tomography/standards , Proof of Concept Study , Pyridines/chemical synthesis , Pyrrolidines/chemical synthesis , Pyrrolidinones/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rodentia , Synaptic Vesicles/metabolism
13.
ACS Chem Neurosci ; 11(4): 592-603, 2020 02 19.
Article En | MEDLINE | ID: mdl-31961649

Synaptic vesicle glycoprotein 2A (SV2A) is a 12-pass transmembrane glycoprotein ubiquitously expressed in presynaptic vesicles. In vivo imaging of SV2A using PET has potential applications in the diagnosis and prognosis of a variety of neuropsychiatric diseases, e.g., Alzheimer's disease, Parkinson's disease, schizophrenia, multiple sclerosis, autism, epilepsy, stroke, traumatic brain injury, post-traumatic stress disorder, depression, etc. Herein, we report the synthesis and evaluation of a new 18F-labeled SV2A PET imaging probe, [18F]SynVesT-2, which possesses fast in vivo binding kinetics and high specific binding signals in non-human primate brain.


Alzheimer Disease/pathology , Epilepsy/pathology , Membrane Glycoproteins/metabolism , Synaptic Vesicles/pathology , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Epilepsy/diagnosis , Humans , Nerve Tissue Proteins/metabolism , Primates/metabolism , Synaptic Vesicles/metabolism
14.
Sci Rep ; 9(1): 19616, 2019 12 23.
Article En | MEDLINE | ID: mdl-31873156

Aging is associated with functional alterations of synapses thought to contribute to age-dependent memory impairment (AMI). While therapeutic avenues to protect from AMI are largely elusive, supplementation of spermidine, a polyamine normally declining with age, has been shown to restore defective proteostasis and to protect from AMI in Drosophila. Here we demonstrate that dietary spermidine protects from age-related synaptic alterations at hippocampal mossy fiber (MF)-CA3 synapses and prevents the aging-induced loss of neuronal mitochondria. Dietary spermidine rescued age-dependent decreases in synaptic vesicle density and largely restored defective presynaptic MF-CA3 long-term potentiation (LTP) at MF-CA3 synapses (MF-CA3) in aged animals. In contrast, spermidine failed to protect CA3-CA1 hippocampal synapses characterized by postsynaptic LTP from age-related changes in function and morphology. Our data demonstrate that dietary spermidine attenuates age-associated deterioration of MF-CA3 synaptic transmission and plasticity. These findings provide a physiological and molecular basis for the future therapeutic usage of spermidine.


Aging/metabolism , CA3 Region, Hippocampal/metabolism , Long-Term Potentiation/drug effects , Mossy Fibers, Hippocampal/metabolism , Spermidine/pharmacology , Synaptic Transmission/drug effects , Synaptic Vesicles/metabolism , Aging/drug effects , Aging/pathology , Animals , CA3 Region, Hippocampal/pathology , Mice , Mossy Fibers, Hippocampal/pathology , Synaptic Vesicles/pathology
15.
Int J Mol Sci ; 20(21)2019 Oct 25.
Article En | MEDLINE | ID: mdl-31731450

Parkinson's disease (PD) is the second most common neurodegenerative disorder worldwide, mainly affecting the elderly. The disease progresses gradually, with core motor presentations and a multitude of non-motor manifestations. There are two neuropathological hallmarks of PD, the dopaminergic neuronal loss and the alpha-synuclein-containing Lewy body inclusions in the substantia nigra. While the exact pathomechanisms of PD remain unclear, genetic investigations have revealed evidence of the involvement of mitochondrial function, alpha-synuclein (α-syn) aggregation, and the endo-lysosomal system, in disease pathogenesis. Due to the high energy demand of dopaminergic neurons, mitochondria are of special importance acting as the cellular powerhouse. Mitochondrial dynamic fusion and fission, and autophagy quality control keep the mitochondrial network in a healthy state. Should defects of the organelle occur, a variety of reactions would ensue at the cellular level, including disrupted mitochondrial respiratory network and perturbed calcium homeostasis, possibly resulting in cellular death. Meanwhile, α-syn is a presynaptic protein that helps regulate synaptic vesicle transportation and endocytosis. Its misfolding into oligomeric sheets and fibrillation is toxic to the mitochondria and neurons. Increased cellular oxidative stress leads to α-syn accumulation, causing mitochondrial dysfunction. The proteasome and endo-lysosomal systems function to regulate damage and unwanted waste management within the cell while facilitating the quality control of mitochondria and α-syn. This review will analyze the biological functions and interactions between mitochondria, α-syn, and the endo-lysosomal system in the pathogenesis of PD.


Dopaminergic Neurons/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Biological Transport, Active/genetics , Dopaminergic Neurons/pathology , Electron Transport/genetics , Endosomes/genetics , Endosomes/pathology , Humans , Lysosomes/genetics , Lysosomes/pathology , Mitochondria/genetics , Mitochondria/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology , alpha-Synuclein/genetics
16.
J Alzheimers Dis ; 72(1): 1-14, 2019.
Article En | MEDLINE | ID: mdl-31561377

It is now more than two decades since amyloid-ß (Aß), the proteolytic product of the amyloid-ß protein precursor (AßPP), was first demonstrated to be a normal and soluble product of neuronal metabolism. To date, despite a growing body of evidence suggests its regulatory role on synaptic function, the exact cellular and molecular pathways involved in Aß-driven synaptic effects remain elusive. This review provides an overview of the mounting evidence showing Aß-mediated effects on presynaptic functions and neurotransmitter release from axon terminals, focusing on its interaction with synaptic vesicle cycle. Indeed, Aß peptides have been found to interact with key presynaptic scaffold proteins and kinases affecting the consequential steps of the synaptic vesicle dynamics (e.g., synaptic vesicles exocytosis, endocytosis, and trafficking). Defects in the fine-tuning of synaptic vesicle cycle by Aß and deregulation of key molecules and kinases, which orchestrate synaptic vesicle availability, may alter synaptic homeostasis, possibly contributing to synaptic loss and cognitive decline. Elucidating the presynaptic mechanisms by which Aß regulate synaptic transmission is fundamental for a deeper comprehension of the biology of presynaptic terminals as well as of Aß-driven early synaptic defects occurring in prodromal stage of AD. Moreover, a better understating of Aß involvement in cellular signal pathways may allow to set up more effective therapeutic interventions by detecting relevant molecular mechanisms, whose imbalance might ultimately lead to synaptic impairment in AD.


Amyloid beta-Peptides/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Animals , Endocytosis/physiology , Exocytosis/physiology , Humans , Presynaptic Terminals/pathology , Synapses/pathology , Synaptic Vesicles/pathology
17.
ACS Chem Neurosci ; 10(9): 3927-3938, 2019 09 18.
Article En | MEDLINE | ID: mdl-31394034

The synaptic vesicle glycoprotein 2 (SV2) family is comprised of three paralogues: SV2A, SV2B, and SV2C. In vertebrates, SV2s are 12-transmembrane proteins present on every secretory vesicle, including synaptic vesicles, and are critical to neurotransmission. Structural and functional studies suggest that SV2 proteins may play several roles to promote proper vesicular function. Among these roles are their potential to stabilize the transmitter content of vesicles, to maintain and orient the releasable pool of vesicles, and to regulate vesicular calcium sensitivity to ensure efficient, coordinated release of the transmitter. The SV2 family is highly relevant to human health in a number of ways. First, SV2A plays a role in neuronal excitability and as such is the specific target for the antiepileptic drug levetiracetam. SV2 proteins also act as the target by which potent neurotoxins, particularly botulinum, gain access to neurons and exert their toxicity. Both SV2B and SV2C are increasingly implicated in diseases such as Alzheimer's disease and Parkinson's disease. Interestingly, despite decades of intensive research, their exact function remains elusive. Thus, SV2 proteins are intriguing in their potentially diverse roles within the presynaptic terminal, and several recent developments have enhanced our understanding and appreciation of the protein family. Here, we review the structure and function of SV2 proteins as well as their relevance to disease and therapeutic development.


Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Nervous System Diseases/metabolism , Synaptic Vesicles/metabolism , Animals , Humans , Nervous System Diseases/pathology , Protein Transport/physiology , Synaptic Vesicles/chemistry , Synaptic Vesicles/pathology
18.
Sci Rep ; 9(1): 377, 2019 01 23.
Article En | MEDLINE | ID: mdl-30674958

In the present study, we measured the spontaneous post synaptic currents (sPSCs) at the post synaptic principle cells of the medial nucleus of the trapezoid body (MNTB) in early postnatal mice after exposure to 1850 MHz radiofrequency electromagnetic fields (RF-EMF). sPSC frequencies and amplitudes were significantly increased in the RF-EMF exposed group. Moreover, the number of synaptic vesicles in the calyx of Held was significantly increased in presynaptic nerve terminals. Following RF-EMF exposure, the number of docking synaptic vesicles in the active zone increased, thereby expanding the total length of the presynaptic active zone in the calyx of Held. These data suggest that the increased sPSCs are a result of greater synaptic vesicle release from presynaptic nerves. However, we found no morphological changes in the inner hair cell ribbon synapses. Further, there were no significant changes in the hearing threshold of the auditory brainstem response at postnatal day 15. Our results indicate that exposure to RF-EMF at an early postnatal stage might directly affect brainstem auditory circuits, but it does not seem to alter general sound perception.


Evoked Potentials, Auditory, Brain Stem , Radio Waves/adverse effects , Synaptic Transmission , Synaptic Vesicles/metabolism , Trapezoid Body/metabolism , Acebutolol , Animals , Animals, Newborn , Mice, Inbred ICR , Synaptic Vesicles/pathology , Trapezoid Body/pathology
19.
Mol Neurodegener ; 14(1): 7, 2019 01 22.
Article En | MEDLINE | ID: mdl-30670054

BACKGROUND: Identifying effective strategies to prevent memory loss in AD has eluded researchers to date, and likely reflects insufficient understanding of early pathogenic mechanisms directly affecting memory encoding. As synaptic loss best correlates with memory loss in AD, refocusing efforts to identify factors driving synaptic impairments may provide the critical insight needed to advance the field. In this study, we reveal a previously undescribed cascade of events underlying pre and postsynaptic hippocampal signaling deficits linked to cognitive decline in AD. These profound alterations in synaptic plasticity, intracellular Ca2+ signaling, and network propagation are observed in 3-4 month old 3xTg-AD mice, an age which does not yet show overt histopathology or major behavioral deficits. METHODS: In this study, we examined hippocampal synaptic structure and function from the ultrastructural level to the network level using a range of techniques including electron microscopy (EM), patch clamp and field potential electrophysiology, synaptic immunolabeling, spine morphology analyses, 2-photon Ca2+ imaging, and voltage-sensitive dye-based imaging of hippocampal network function in 3-4 month old 3xTg-AD and age/background strain control mice. RESULTS: In 3xTg-AD mice, short-term plasticity at the CA1-CA3 Schaffer collateral synapse is profoundly impaired; this has broader implications for setting long-term plasticity thresholds. Alterations in spontaneous vesicle release and paired-pulse facilitation implicated presynaptic signaling abnormalities, and EM analysis revealed a reduction in the ready-releasable and reserve pools of presynaptic vesicles in CA3 terminals; this is an entirely new finding in the field. Concurrently, increased synaptically-evoked Ca2+ in CA1 spines triggered by LTP-inducing tetani is further enhanced during PTP and E-LTP epochs, and is accompanied by impaired synaptic structure and spine morphology. Notably, vesicle stores, synaptic structure and short-term plasticity are restored by normalizing intracellular Ca2+ signaling in the AD mice. CONCLUSIONS: These findings suggest the Ca2+ dyshomeostasis within synaptic compartments has an early and fundamental role in driving synaptic pathophysiology in early stages of AD, and may thus reflect a foundational disease feature driving later cognitive impairment. The overall significance is the identification of previously unidentified defects in pre and postsynaptic compartments affecting synaptic vesicle stores, synaptic plasticity, and network propagation, which directly impact memory encoding.


Alzheimer Disease/pathology , Hippocampus/physiopathology , Neuronal Plasticity/physiology , Synaptic Vesicles/pathology , Alzheimer Disease/metabolism , Animals , Calcium Signaling/physiology , Disease Models, Animal , Female , Hippocampus/metabolism , Male , Mice , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism
20.
Neurosci Lett ; 697: 59-65, 2019 04 01.
Article En | MEDLINE | ID: mdl-29627340

Parkinson's disease (PD) is a common neurodegenerative disease characterized pathologically by the selective loss of dopaminergic neurons in the substantia nigra and the intracellular accumulation of α-synuclein in the Lewy bodies. While the pathogenic mechanisms of PD are poorly understood, many lines of evidence point to a role of altered autophagy and membrane trafficking in the development of the disease. Emerging studies show that connections between the deregulation of autophagy and synaptic vesicle (SV) trafficking may contribute to PD. Here we review the evidence that many PD related-genes have roles in both autophagy and SV trafficking and examine how deregulation of these pathways contributes to PD pathogenesis. This review also discusses recent studies aimed at uncovering the role of PD-linked genes in autophagy-lysosome function.


Parkinson Disease/metabolism , Parkinson Disease/pathology , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology , Animals , Autophagy/physiology , Endocytosis , Humans , Lysosomes/metabolism , Lysosomes/pathology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Protein Transport
...