Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Eur J Neurosci ; 59(6): 1079-1098, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37667848

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by progressive dysfunction and loss of dopaminergic neurons of the substantia nigra pars compacta (SNc). Several pathways of programmed cell death are likely to play a role in dopaminergic neuron death, such as apoptosis, necrosis, pyroptosis and ferroptosis, as well as cell death associated with proteasomal and mitochondrial dysfunction. A better understanding of the molecular mechanisms underlying dopaminergic neuron death could inform the design of drugs that promote neuron survival. Necroptosis is a recently characterized regulated cell death mechanism that exhibits morphological features common to both apoptosis and necrosis. It requires activation of an intracellular pathway involving receptor-interacting protein 1 kinase (RIP1 kinase, RIPK1), receptor-interacting protein 3 kinase (RIP3 kinase, RIPK3) and mixed lineage kinase domain-like pseudokinase (MLKL). The potential involvement of this programmed cell death pathway in the pathogenesis of PD has been studied by analysing biomarkers for necroptosis, such as the levels and oligomerization of phosphorylated RIPK3 (pRIPK3) and phosphorylated MLKL (pMLKL), in several PD preclinical models and in PD human tissue. Although there is evidence that other types of cell death also have a role in DA neuron death, most studies support the hypothesis that this cell death mechanism is activated in PD tissues. Drugs that prevent or reduce necroptosis may provide neuroprotection for PD. In this review, we summarize the findings from these studies. We also discuss how manipulating necroptosis might open a novel therapeutic approach to reduce neuronal degeneration in PD.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Humans , Dopaminergic Neurons/metabolism , Parkinson Disease/metabolism , Necroptosis , Cell Death , Apoptosis , Necrosis/metabolism , Necrosis/pathology , Dopamine/metabolism
2.
Pharmacol Res ; 198: 106993, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37972722

ABSTRACT

The treatment of bipolar disorder (BD) still remains a challenge. Melatonin (MLT), acting through its two receptors MT1 and MT2, plays a key role in regulating circadian rhythms which are dysfunctional in BD. Using a translational approach, we examined the implication and potential of MT1 receptors in the pathophysiology and psychopharmacology of BD. We employed a murine model of the manic phase of BD (Clock mutant (ClockΔ19) mice) to study the activation of MT1 receptors by UCM871, a selective partial agonist, in behavioral pharmacology tests and in-vivo electrophysiology. We then performed a high-resolution Nuclear Magnetic Resonance study on isolated membranes to characterize the molecular mechanism of interaction of UCM871. Finally, in a cohort of BD patients, we investigated the link between clinical measures of BD and genetic variants located in the MT1 receptor and CLOCK genes. We demonstrated that: 1) UCM871 can revert behavioral and electrophysiological abnormalities of ClockΔ19 mice; 2) UCM871 promotes the activation state of MT1 receptors; 3) there is a significant association between the number of severe manic episodes and MLT levels, depending on the genetic configuration of the MT1 rs2165666 variant. Overall, this work lends support to the potentiality of MT1 receptors as target for the treatment of BD.


Subject(s)
Bipolar Disorder , Melatonin , Psychopharmacology , Humans , Mice , Animals , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Melatonin/therapeutic use , Melatonin/pharmacology , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/agonists
3.
Brain Behav Immun ; 89: 175-183, 2020 10.
Article in English | MEDLINE | ID: mdl-32531426

ABSTRACT

Depression and anxiety symptoms are highly prevalent among women during pregnancy and post-partum. Previous studies suggest that one of the pathophysiological underpinnings could be an enhanced metabolism of tryptophan (Trp) into kynurenine (Kyn) due to increased inflammation. However, the longitudinal changes in the Kyn pathway and the complex interplay with inflammation and stress in women with perinatal depressive or anxiety symptoms are incompletely understood. We examined a cohort of healthy women at 34-36 gestational weeks. One hundred and ten women were assessed for salivary cortisol and 97 participants were also assessed for serum levels of Trp, Kyn and Interleukin 6 (IL-6). Women filled in two screening questionnaires for depressive (Edinburgh Postnatal Depression Scale (EPDS)) and anxiety (State Trait Anxiety Inventory subscale (STAI-S)) symptoms at 34-36 gestational weeks, delivery, 3 and 12 months postpartum. Unexpectedly, lower prenatal Kyn levels were associated with higher depressive symptoms in late pregnancy. Furthermore, prenatal Trp levels and the Kyn/Trp ratio moderate the association between IL-6 levels and depressive symptoms during the perinatal and the post-partum period. We found no interactions between Trp and Kyn biomarkers and cortisol on depressive symptoms. The observed associations were more robustly found for depressive symptoms, whereas weak and non-significant effects were found for the trajectory of anxiety symptoms. Overall, our data support the involvement of the Trp to Kyn pathway and inflammation in the course of depressive but not anxiety symptoms in women from late pregnancy until one-year post-partum, providing new evidence on the mechanisms regulating emotions during pregnancy and after delivery in a low-risk sample.


Subject(s)
Interleukin-6 , Kynurenine , Anxiety , Female , Humans , Postpartum Period , Pregnancy , Tryptophan
4.
Cereb Cortex ; 29(5): 2010-2033, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29912316

ABSTRACT

Mutations in PRoline-Rich Transmembrane protein 2 (PRRT2) underlie a group of paroxysmal disorders including epilepsy, kinesigenic dyskinesia and migraine. Most of the mutations lead to impaired PRRT2 expression and/or function, emphasizing the pathogenic role of the PRRT2 deficiency. In this work, we investigated the phenotype of primary hippocampal neurons obtained from mouse embryos in which the PRRT2 gene was constitutively inactivated. Although PRRT2 is expressed by both excitatory and inhibitory neurons, its deletion decreases the number of excitatory synapses without significantly affecting the number of inhibitory synapses or the nerve terminal ultrastructure. Analysis of synaptic function in primary PRRT2 knockout excitatory neurons by live imaging and electrophysiology showed slowdown of the kinetics of exocytosis, weakened spontaneous and evoked synaptic transmission and markedly increased facilitation. Inhibitory neurons showed strengthening of basal synaptic transmission, accompanied by faster depression. At the network level these complex synaptic effects resulted in a state of heightened spontaneous and evoked activity that was associated with increased excitability of excitatory neurons in both PRRT2 knockout primary cultures and acute hippocampal slices. The data indicate the existence of network instability/hyperexcitability as the possible basis of the paroxysmal phenotypes associated with PRRT2 mutations.


Subject(s)
Hippocampus/physiology , Membrane Proteins/physiology , Neuronal Plasticity , Neurons/physiology , Synaptic Transmission , Animals , Cells, Cultured , Exocytosis , Male , Membrane Potentials , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/physiology , Synapses/physiology , Synapses/ultrastructure
5.
Hum Mol Genet ; 26(23): 4699-4714, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973667

ABSTRACT

Intellectual Disability is a common and heterogeneous disorder characterized by limitations in intellectual functioning and adaptive behaviour, whose molecular mechanisms remain largely unknown. Among the numerous genes found to be involved in the pathogenesis of intellectual disability, 10% are located on the X-chromosome. We identified a missense mutation (c.236 C > G; p.S79W) in the SYN1 gene coding for synapsin I in the MRX50 family, affected by non-syndromic X-linked intellectual disability. Synapsin I is a neuronal phosphoprotein involved in the regulation of neurotransmitter release and neuronal development. Several mutations in SYN1 have been identified in patients affected by epilepsy and/or autism. The S79W mutation segregates with the disease in the MRX50 family and all affected members display intellectual disability as sole clinical manifestation. At the protein level, the S79W Synapsin I mutation is located in the region of the B-domain involved in recognition of highly curved membranes. Expression of human S79W Synapsin I in Syn1 knockout hippocampal neurons causes aberrant accumulation of small clear vesicles in the soma, increased clustering of synaptic vesicles at presynaptic terminals and increased frequency of excitatory spontaneous release events. In addition, the presence of S79W Synapsin I strongly reduces the mobility of synaptic vesicles, with possible implications for the regulation of neurotransmitter release and synaptic plasticity. These results implicate SYN1 in the pathogenesis of non-syndromic intellectual disability, showing that alterations of synaptic vesicle trafficking are one possible cause of this disease, and suggest that distinct mutations in SYN1 may lead to distinct brain pathologies.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation, Missense , Synapsins/genetics , Synaptic Vesicles/genetics , Animals , Base Sequence , Humans , Mental Retardation, X-Linked/metabolism , Mice , Mice, Knockout , Mutation , Neurogenesis/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , Pedigree , Presynaptic Terminals/metabolism , Primary Cell Culture , Protein Transport , Synapsins/metabolism , Synaptic Transmission/genetics , Synaptic Vesicles/metabolism
6.
Brain Behav Immun ; 68: 197-210, 2018 02.
Article in English | MEDLINE | ID: mdl-29066310

ABSTRACT

The classical view of multiple sclerosis (MS) pathogenesis states that inflammation-mediated demyelination is responsible for neuronal damage and loss. However, recent findings show that impairment of neuronal functions and demyelination can be independent events, suggesting the coexistence of other pathogenic mechanisms. Due to the inflammatory milieu, subtle alterations in synaptic function occur, which are probably at the basis of the early cognitive decline that often precedes the neurodegenerative phases in MS patients. In particular, it has been reported that inflammation enhances excitatory synaptic transmission while it decreases GABAergic transmission in vitro and ex vivo. This evidence points to the idea that an excitation/inhibition imbalance occurs in the inflamed MS brain, even though the exact molecular mechanisms leading to this synaptic dysfunction are as yet not completely clear. Along this line, we observed that acute treatment of primary hippocampal neurons in culture with pro-inflammatory cytokines leads to an increased phosphorylation of synapsin I (SynI) by ERK1/2 kinase and to an increase in the frequency of spontaneous synaptic vesicle release events, which is prevented by SynI deletion. In vivo, the ablation of SynI expression is protective in terms of disease progression and neuronal damage in the experimental autoimmune encephalomyelitis mouse model of MS. Our results point to a possible key role in MS pathogenesis of the neuronal protein SynI, a regulator of excitation/inhibition balance in neuronal networks.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Synapsins/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Disease Progression , Hippocampus/metabolism , Inflammation/metabolism , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Neurons/metabolism , Neuroprotective Agents/metabolism , Phosphorylation , Synapses/metabolism , Synapsins/genetics , Synaptic Vesicles/metabolism
7.
Brain ; 140(9): 2265-2272, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28335015

ABSTRACT

Loss of function mutations in the gene PARK2, which encodes the protein parkin, cause autosomal recessive juvenile parkinsonism, a neurodegenerative disease characterized by degeneration of the dopaminergic neurons localized in the substantia nigra pars compacta. No therapy is effective in slowing disease progression mostly because the pathogenesis of the disease is yet to be understood. From accruing evidence suggesting that the protein parkin directly regulates synapses it can be hypothesized that PARK2 gene mutations lead to early synaptic damage that results in dopaminergic neuron loss over time. We review evidence that supports the role of parkin in modulating excitatory and dopaminergic synapse functions. We also discuss how these findings underpin the concept that autosomal recessive juvenile parkinsonism can be primarily a synaptopathy. Investigation into the molecular interactions between parkin and synaptic proteins may yield novel targets for pharmacologic interventions.


Subject(s)
Dopaminergic Neurons/physiology , Parkinson Disease/physiopathology , Synaptic Transmission/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Humans , Mutation , Nerve Degeneration/genetics , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics
8.
Cereb Cortex ; 27(3): 2226-2248, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27005990

ABSTRACT

Alterations in the balance of inhibitory and excitatory synaptic transmission have been implicated in the pathogenesis of neurological disorders such as epilepsy. Eukaryotic elongation factor 2 kinase (eEF2K) is a highly regulated, ubiquitous kinase involved in the control of protein translation. Here, we show that eEF2K activity negatively regulates GABAergic synaptic transmission. Indeed, loss of eEF2K increases GABAergic synaptic transmission by upregulating the presynaptic protein Synapsin 2b and α5-containing GABAA receptors and thus interferes with the excitation/inhibition balance. This cellular phenotype is accompanied by an increased resistance to epilepsy and an impairment of only a specific hippocampal-dependent fear conditioning. From a clinical perspective, our results identify eEF2K as a potential novel target for antiepileptic drugs, since pharmacological and genetic inhibition of eEF2K can revert the epileptic phenotype in a mouse model of human epilepsy.


Subject(s)
Elongation Factor 2 Kinase/metabolism , Epilepsy/enzymology , Neurons/enzymology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Conditioning, Psychological/physiology , Disease Models, Animal , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/genetics , Epilepsy/pathology , Fear/physiology , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Neurons/pathology , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Synapsins/genetics , Synapsins/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
9.
J Neurosci ; 36(16): 4624-34, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27098703

ABSTRACT

Growing evidence indicates that sphingosine-1-P (S1P) upregulates glutamate secretion in hippocampal neurons. However, the molecular mechanisms through which S1P enhances excitatory activity remain largely undefined. The aim of this study was to identify presynaptic targets of S1P action controlling exocytosis. Confocal analysis of rat hippocampal neurons showed that S1P applied at nanomolar concentration alters the distribution of Synapsin I (SynI), a presynaptic phosphoprotein that controls the availability of synaptic vesicles for exocytosis. S1P induced SynI relocation to extrasynaptic regions of mature neurons, as well as SynI dispersion from synaptic vesicle clusters present at axonal growth cones of developing neurons. S1P-induced SynI relocation occurred in a Ca(2+)-independent but ERK-dependent manner, likely through the activation of S1P3 receptors, as it was prevented by the S1P3 receptor selective antagonist CAY1044 and in neurons in which S1P3 receptor was silenced. Our recent evidence indicates that microvesicles (MVs) released by microglia enhance the metabolism of endogenous sphingolipids in neurons and stimulate excitatory transmission. We therefore investigated whether MVs affect SynI distribution and whether endogenous S1P could be involved in the process. Analysis of SynI immunoreactivity showed that exposure to microglial MVs induces SynI mobilization at presynaptic sites and growth cones, whereas the use of inhibitors of sphingolipid cascade identified S1P as the sphingolipid mediating SynI redistribution. Our data represent the first demonstration that S1P induces SynI mobilization from synapses, thereby indicating the phosphoprotein as a novel target through which S1P controls exocytosis. SIGNIFICANCE STATEMENT: Growing evidence indicates that the bioactive lipid sphingosine and its metabolite sphingosine-1-P (S1P) stimulate excitatory transmission. While it has been recently clarified that sphingosine influences directly the exocytotic machinery by activating the synaptic vesicle protein VAMP2 to form SNARE fusion complexes, the molecular mechanism by which S1P promotes neurotransmission remained largely undefined. In this study, we identify Synapsin I, a presynaptic phosphoprotein involved in the control of availability of synaptic vesicles for exocytosis, as the key target of S1P action. In addition, we provide evidence that S1P can be produced at mature axon terminals as well as at immature growth cones in response to microglia-derived signals, which may be important to stabilize nascent synapses and to restore or potentiate transmission.


Subject(s)
Lysophospholipids/physiology , Presynaptic Terminals/metabolism , Sphingosine/analogs & derivatives , Synapses/metabolism , Synapsins/biosynthesis , Animals , Cells, Cultured , Female , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/metabolism , Lysophospholipids/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Presynaptic Terminals/chemistry , Rats , Rats, Sprague-Dawley , Sphingosine/analysis , Sphingosine/physiology , Synapses/chemistry , Synapsins/analysis
10.
J Biol Chem ; 291(12): 6111-23, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26797119

ABSTRACT

Proline-rich transmembrane protein 2 (PRRT2) has been identified as the single causative gene for a group of paroxysmal syndromes of infancy, including epilepsy, paroxysmal movement disorders, and migraine. On the basis of topology predictions, PRRT2 has been assigned to the recently characterized family of Dispanins, whose members share the two-transmembrane domain topology with a large N terminus and short C terminus oriented toward the outside of the cell. Because PRRT2 plays a role at the synapse, it is important to confirm the exact orientation of its N and C termini with respect to the plasma membrane to get clues regarding its possible function. Using a combination of different experimental approaches, including live immunolabeling, immunogold electron microscopy, surface biotinylation and computational modeling, we demonstrate a novel topology for this protein. PRRT2 is a type II transmembrane protein in which only the second hydrophobic segment spans the plasma membrane, whereas the first one is associated with the internal surface of the membrane and forms a helix-loop-helix structure without crossing it. Most importantly, the large proline-rich N-terminal domain is not exposed to the extracellular space but is localized intracellularly, and only the short C terminus is extracellular (N cyt/C exo topology). Accordingly, we show that PRRT2 interacts with the Src homology 3 domain-bearing protein Intersectin 1, an intracellular protein involved in synaptic vesicle cycling. These findings will contribute to the clarification of the role of PRRT2 at the synapse and the understanding of pathogenic mechanisms on the basis of PRRT2-related neurological disorders.


Subject(s)
Membrane Proteins/metabolism , Synapses/metabolism , Animals , Biotinylation , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Membrane Proteins/chemistry , Mice , Molecular Dynamics Simulation , Protein Processing, Post-Translational , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Synaptosomes/metabolism
11.
Neurobiol Dis ; 99: 66-83, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28007585

ABSTRACT

Heterozygous and rare homozygous mutations in PRoline-Rich Transmembrane protein 2 (PRRT2) underlie a group of paroxysmal disorders including epilepsy, kinesigenic dyskinesia episodic ataxia and migraine. Most of the mutations lead to impaired PRRT2 expression and/or function. Recently, an important role for PRTT2 in the neurotransmitter release machinery, brain development and synapse formation has been uncovered. In this work, we have characterized the phenotype of a mouse in which the PRRT2 gene has been constitutively inactivated (PRRT2 KO). ß-galactosidase staining allowed to map the regional expression of PRRT2 that was more intense in the cerebellum, hindbrain and spinal cord, while it was localized to restricted areas in the forebrain. PRRT2 KO mice are normal at birth, but display paroxysmal movements at the onset of locomotion that persist in the adulthood. In addition, adult PRRT2 KO mice present abnormal motor behaviors characterized by wild running and jumping in response to audiogenic stimuli that are ineffective in wild type mice and an increased sensitivity to the convulsive effects of pentylentetrazol. Patch-clamp electrophysiology in hippocampal and cerebellar slices revealed specific effects in the cerebellum, where PRRT2 is highly expressed, consisting in a higher excitatory strength at parallel fiber-Purkinje cell synapses during high frequency stimulation. The results show that the PRRT2 KO mouse reproduces the motor paroxysms present in the human PRRT2-linked pathology and can be proposed as an experimental model for the study of the pathogenesis of the disease as well as for testing personalized therapeutic approaches.


Subject(s)
Brain/physiopathology , Membrane Proteins/deficiency , Motor Activity/physiology , Motor Disorders/physiopathology , Seizures/physiopathology , Animals , Animals, Newborn , Brain/growth & development , Brain/pathology , Cognition/physiology , Disease Models, Animal , Female , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Motor Disorders/pathology , Mutation , Nerve Tissue Proteins/genetics , Pentylenetetrazole , Phenotype , Seizures/pathology , Spinal Cord/growth & development , Spinal Cord/pathology , Spinal Cord/physiopathology , Synapses/pathology , Synapses/physiology , Tissue Culture Techniques
12.
EMBO J ; 32(22): 2994-3007, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24149584

ABSTRACT

Intrinsic homeostasis enables neuronal circuits to maintain activity levels within an appropriate range by modulating neuronal voltage-gated conductances, but the signalling pathways involved in this process are largely unknown. We characterized the process of intrinsic homeostasis induced by sustained electrical activity in cultured hippocampal neurons based on the activation of the Repressor Element-1 Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). We showed that 4-aminopyridine-induced hyperactivity enhances the expression of REST/NRSF, which in turn, reduces the expression of voltage-gated Na(+) channels, thereby decreasing the neuronal Na(+) current density. This mechanism plays an important role in the downregulation of the firing activity at the single-cell level, re-establishing a physiological spiking activity in the entire neuronal network. Conversely, interfering with REST/NRSF expression impaired this homeostatic response. Our results identify REST/NRSF as a critical factor linking neuronal activity to the activation of intrinsic homeostasis and restoring a physiological level of activity in the entire neuronal network.


Subject(s)
Homeostasis/physiology , Repressor Proteins/physiology , 4-Aminopyridine/pharmacology , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/physiology , Homeostasis/drug effects , Mice , Mice, Inbred C57BL , Nerve Net , Neurons/physiology
13.
Hum Mol Genet ; 23(1): 90-103, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23956174

ABSTRACT

An increasing number of genes predisposing to autism spectrum disorders (ASDs) has been identified, many of which are implicated in synaptic function. This 'synaptic autism pathway' notably includes disruption of SYN1 that is associated with epilepsy, autism and abnormal behavior in both human and mice models. Synapsins constitute a multigene family of neuron-specific phosphoproteins (SYN1-3) present in the majority of synapses where they are implicated in the regulation of neurotransmitter release and synaptogenesis. Synapsins I and II, the major Syn isoforms in the adult brain, display partially overlapping functions and defects in both isoforms are associated with epilepsy and autistic-like behavior in mice. In this study, we show that nonsense (A94fs199X) and missense (Y236S and G464R) mutations in SYN2 are associated with ASD in humans. The phenotype is apparent in males. Female carriers of SYN2 mutations are unaffected, suggesting that SYN2 is another example of autosomal sex-limited expression in ASD. When expressed in SYN2  knockout neurons, wild-type human Syn II fully rescues the SYN2 knockout phenotype, whereas the nonsense mutant is not expressed and the missense mutants are virtually unable to modify the SYN2 knockout phenotype. These results identify for the first time SYN2  as a novel predisposing gene for ASD and strengthen the hypothesis that a disturbance of synaptic homeostasis underlies ASD.


Subject(s)
Axons/metabolism , Axons/pathology , Child Development Disorders, Pervasive/genetics , Synapsins/genetics , Synaptic Vesicles/pathology , Animals , Child Development Disorders, Pervasive/metabolism , Codon, Nonsense , Female , Genetic Predisposition to Disease , HeLa Cells , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense , Neurons/metabolism , Synaptic Vesicles/metabolism
14.
EMBO J ; 31(8): 1893-903, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22354041

ABSTRACT

The precise polarization and orientation of developing neurons is essential for the correct wiring of the brain. In pyramidal excitatory neurons, polarization begins with the sprouting of opposite neurites, which later define directed migration and axo-dendritic domains. We here show that endogenous N-cadherin concentrates at one pole of the newborn neuron, from where the first neurite subsequently emerges. Ectopic N-cadherin is sufficient to favour the place of appearance of the first neurite. The Golgi and centrosome move towards this newly formed morphological pole in a second step, which is regulated by PI3K and the actin/microtubule cytoskeleton. Moreover, loss of function experiments in vivo showed that developing neurons with a non-functional N-cadherin misorient their cell axis. These results show that polarization of N-cadherin in the immediate post-mitotic stage is an early and crucial mechanism in neuronal polarity.


Subject(s)
Cadherins/metabolism , Cell Division , Cell Polarity , Nerve Tissue Proteins/metabolism , Neurons/physiology , Animals , Centrosome/metabolism , Cytoskeleton/metabolism , Golgi Apparatus/metabolism , Neurites/physiology , Phosphatidylinositol 3-Kinase/metabolism , Rats
15.
J Cell Sci ; 127(Pt 20): 4409-19, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128563

ABSTRACT

Final morphological polarization of neurons, with the development of a distinct axon and several dendrites, is preceded by phases where they have a non-polarized architecture. The earliest of these phases is that of the round neuron arising from the last mitosis. A second non-polarized stage corresponds to the bipolar neuron, with two morphologically identical neurites. Both phases have their distinctive relevance in the establishment of neuronal polarity. During the round cell stage, a decision is made as to where from the cell periphery a first neurite will form, thus creating the first sign of asymmetry. At the bipolar stage a decision is made as to which of the two neurites becomes the axon in neurons polarizing in vitro, and the leading edge in neurons in situ. In this study, we analysed cytoskeletal and membrane dynamics in cells at these two 'pre-polarity' stages. By means of time lapse imaging in dissociated hippocampal neurons and ex vivo cortical slices, we show that both stages are characterized by polarized intracellular arrangements. However, the stages have distinct temporal hierarchies: polarized actin dynamics marks the site of first polarization in round cells, whereas polarized membrane dynamics precedes asymmetric growth in the bipolar stage.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Cell Polarity , Cytoskeleton/metabolism , Hippocampus/growth & development , Neurogenesis , Neurons/physiology , Animals , Cells, Cultured , Female , Hippocampus/cytology , Mice , Organ Culture Techniques , Pregnancy , Protein Transport , Rats , Rats, Inbred Strains , Time-Lapse Imaging
16.
J Neurosci ; 34(21): 7266-80, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24849359

ABSTRACT

Cyclin-dependent kinase-5 (Cdk5) was reported to downscale neurotransmission by sequestering synaptic vesicles (SVs) in the release-reluctant resting pool, but the molecular targets mediating this activity remain unknown. Synapsin I (SynI), a major SV phosphoprotein involved in the regulation of SV trafficking and neurotransmitter release, is one of the presynaptic substrates of Cdk5, which phosphorylates it in its C-terminal region at Ser(549) (site 6) and Ser(551) (site 7). Here we demonstrate that Cdk5 phosphorylation of SynI fine tunes the recruitment of SVs to the active recycling pool and contributes to the Cdk5-mediated homeostatic responses. Phosphorylation of SynI by Cdk5 is physiologically regulated and enhances its binding to F-actin. The effects of Cdk5 inhibition on the size and depletion kinetics of the recycling pool, as well as on SV distribution within the nerve terminal, are virtually abolished in mouse SynI knock-out (KO) neurons or in KO neurons expressing the dephosphomimetic SynI mutants at sites 6,7 or site 7 only. The observation that the single site-7 mutant phenocopies the effects of the deletion of SynI identifies this site as the central switch in mediating the synaptic effects of Cdk5 and demonstrates that SynI is necessary and sufficient for achieving the effects of the kinase on SV trafficking. The phosphorylation state of SynI by Cdk5 at site 7 is regulated during chronic modification of neuronal activity and is an essential downstream effector for the Cdk5-mediated homeostatic scaling.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Hippocampus/cytology , Synapses/ultrastructure , Synapsins/metabolism , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/pharmacology , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology , Pregnancy , Protein Binding/drug effects , Sodium Channel Blockers/pharmacology , Synapsins/deficiency , Synaptic Vesicles/drug effects , Synaptic Vesicles/ultrastructure , Tetrodotoxin/pharmacology
17.
Hum Mol Genet ; 22(11): 2186-99, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23406870

ABSTRACT

Synapsin I (SynI) is a synaptic vesicle (SV) phosphoprotein playing multiple roles in synaptic transmission and plasticity by differentially affecting crucial steps of SV trafficking in excitatory and inhibitory synapses. SynI knockout (KO) mice are epileptic, and nonsense and missense mutations in the human SYN1 gene have a causal role in idiopathic epilepsy and autism. To get insights into the mechanisms of epileptogenesis linked to SYN1 mutations, we analyzed the effects of the recently identified Q555X mutation on neurotransmitter release dynamics and short-term plasticity (STP) in excitatory and inhibitory synapses. We used patch-clamp electrophysiology coupled to electron microscopy and multi-electrode arrays to dissect synaptic transmission of primary SynI KO hippocampal neurons in which the human wild-type and mutant SynI were expressed by lentiviral transduction. A parallel decrease in the SV readily releasable pool in inhibitory synapses and in the release probability in excitatory synapses caused a marked reduction in the evoked synchronous release. This effect was accompanied by an increase in asynchronous release that was much more intense in excitatory synapses and associated with an increased total charge transfer. Q555X-hSynI induced larger facilitation and post-tetanic potentiation in excitatory synapses and stronger depression after long trains in inhibitory synapses. These changes were associated with higher network excitability and firing/bursting activity. Our data indicate that imbalances in STP and release dynamics of inhibitory and excitatory synapses trigger network hyperexcitability potentially leading to epilepsy/autism manifestations.


Subject(s)
Epilepsy/genetics , Epilepsy/metabolism , Neuronal Plasticity/genetics , Synapses/metabolism , Synapsins/genetics , Synapsins/metabolism , Animals , Female , Gene Expression , Hippocampus/metabolism , Humans , Intracellular Space/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Patch-Clamp Techniques , Phenotype , Protein Multimerization , Protein Transport , Synapsins/chemistry , Synaptic Potentials , Synaptic Vesicles/metabolism
19.
Cereb Cortex ; 23(3): 581-93, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22368083

ABSTRACT

Synapsins (Syn I, Syn II, and Syn III) are a family of synaptic vesicle phosphoproteins regulating synaptic transmission and plasticity. SYN1/2 genes have been identified as major epilepsy susceptibility genes in humans and synapsin I/II/III triple knockout (TKO) mice are epileptic. However, excitatory and inhibitory synaptic transmission and short-term plasticity have never been analyzed in intact neuronal circuits of TKO mice. To clarify the generation and expression of the epileptic phenotype, we performed patch-clamp recordings in the CA1 region of acute hippocampal slices from 1-month-old presymptomatic and 6-month-old epileptic TKO mice and age-matched controls. We found a strong imbalance between basal glutamatergic and γ-aminobutyric acid (GABA)ergic transmission with increased evoked excitatory postsynaptic current and impaired evoked inhibitory postsynaptic current amplitude. This imbalance was accompanied by a parallel derangement of short-term plasticity paradigms, with enhanced facilitation of glutamatergic transmission in the presymptomatic phase and milder depression of inhibitory synapses in the symptomatic phase. Interestingly, a lower tonic GABA(A) current due to the impaired GABA release is responsible for the more depolarized resting potential found in TKO CA1 neurons, which makes them more susceptible to fire. All these changes preceded the appearance of epilepsy, indicating that the distinct changes in excitatory and inhibitory transmission due to the absence of Syns initiate the epileptogenic process.


Subject(s)
CA1 Region, Hippocampal/physiology , Epilepsy/physiopathology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Epilepsy/genetics , Excitatory Postsynaptic Potentials/physiology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Theoretical , Patch-Clamp Techniques , Synapses/physiology , Synapsins/deficiency , Synapsins/genetics
20.
Cereb Cortex ; 23(3): 531-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22402347

ABSTRACT

Activation of protein kinase A (PKA) pathway at presynaptic terminals plays a crucial role in the supply of synaptic vesicles (SVs) from the reserve pool, affecting the steady-state level of activity and the reconstitution of the readily releasable pool after intense stimulation. However, the identity of the stimuli activating this pathway is undefined. Using fluorescence resonance energy transfer and molecular genetic, we show that kainate, through the activation of presynaptic kainate receptors, induces PKA activation and enhances synapsin I phosphorylation at PKA-specific residues. This leads to a dispersion of synapsin I immunoreactivity, which is accompanied by a PKA-dependent increase in the rate of SV recycling at the growth cone and by an enhanced miniature excitatory postsynaptic currents frequency in mature networks. Selective activation of this pathway is induced by the native neurotransmitter glutamate, when applied in the high nanomolar range. These data identify glutamate, specifically acting on KARs, as one of the stimuli able to induce phosphorylation of synapsin at PKA sites, both at the axonal growth cone and at the mature synapse, thus increasing SV availability and contributing to plasticity phenomena.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Growth Cones/physiology , Neuronal Plasticity/physiology , Receptors, Kainic Acid/metabolism , Synaptic Vesicles/physiology , Animals , Cells, Cultured , Enzyme Activation/physiology , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/physiology , Female , Fluorescence Resonance Energy Transfer , Glutamic Acid/metabolism , Hippocampus/physiology , Immunohistochemistry , Kainic Acid/metabolism , Kainic Acid/pharmacology , Neurons/physiology , Patch-Clamp Techniques , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Synapsins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL