Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Rep ; 43(8): 114531, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39058591

RÉSUMÉ

Spontaneous and sensory-evoked activity sculpts developing circuits. Yet, how these activity patterns intersect with cellular programs regulating the differentiation of neuronal subtypes is not well understood. Through electrophysiological and in vivo longitudinal analyses, we show that C-X-C motif chemokine ligand 14 (Cxcl14), a gene previously characterized for its association with tumor invasion, is expressed by single-bouquet cells (SBCs) in layer I (LI) of the somatosensory cortex during development. Sensory deprivation at neonatal stages markedly decreases Cxcl14 expression. Additionally, we report that loss of function of this gene leads to increased intrinsic excitability of SBCs-but not LI neurogliaform cells-and augments neuronal complexity. Furthermore, Cxcl14 loss impairs sensory map formation and compromises the in vivo recruitment of superficial interneurons by sensory inputs. These results indicate that Cxcl14 is required for LI differentiation and demonstrate the emergent role of chemokines as key players in cortical network development.

2.
Nat Cell Biol ; 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951706

RÉSUMÉ

α-Synuclein (α-Syn) aggregation is closely associated with Parkinson's disease neuropathology. Physiologically, α-Syn promotes synaptic vesicle (SV) clustering and soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex assembly. However, the underlying structural and molecular mechanisms are uncertain and it is not known whether this function affects the pathological aggregation of α-Syn. Here we show that the juxtamembrane region of vesicle-associated membrane protein 2 (VAMP2)-a component of the SNARE complex that resides on SVs-directly interacts with the carboxy-terminal region of α-Syn through charged residues to regulate α-Syn's function in clustering SVs and promoting SNARE complex assembly by inducing a multi-component condensed phase of SVs, α-Syn and other components. Moreover, VAMP2 binding protects α-Syn against forming aggregation-prone oligomers and fibrils in these condensates. Our results suggest a molecular mechanism that maintains α-Syn's function and prevents its pathological amyloid aggregation, the failure of which may lead to Parkinson's disease.

3.
Mov Disord ; 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38946200

RÉSUMÉ

Various forms of Parkinson's disease, including its common sporadic form, are characterized by prominent α-synuclein (αSyn) aggregation in affected brain regions. However, the role of αSyn in the pathogenesis and evolution of the disease remains unclear, despite vast research efforts of more than a quarter century. A better understanding of the role of αSyn, either primary or secondary, is critical for developing disease-modifying therapies. Previous attempts to hone this research have been challenged by experimental limitations, but recent technological advances may facilitate progress. The Scientific Issues Committee of the International Parkinson and Movement Disorder Society (MDS) charged a panel of experts in the field to discuss current scientific priorities and identify research strategies with potential for a breakthrough. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.

5.
Article de Anglais | MEDLINE | ID: mdl-38772708

RÉSUMÉ

Parkinson's disease (PD) involves both the central nervous system (CNS) and enteric nervous system (ENS), and their interaction is important for understanding both the clinical manifestations of the disease and the underlying disease pathophysiology. Although the neuroanatomical distribution of pathology strongly suggests that the ENS is involved in disease pathophysiology, there are significant gaps in knowledge about the underlying mechanisms. In this article, we review the clinical presentation and management of gastrointestinal dysfunction in PD. In addition, we discuss the current understanding of disease pathophysiology in the gut, including controversies about early involvement of the gut in disease pathogenesis. We also review current knowledge about gut α-synuclein and the microbiome, discuss experimental models of PD-linked gastrointestinal pathophysiology, and highlight areas for further research. Finally, we discuss opportunities to use the gut-brain axis for the development of biomarkers and disease-modifying treatments.

6.
bioRxiv ; 2024 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-38496494

RÉSUMÉ

Post-translational modifications (PTMs) of α-synuclein (α-syn) such as acetylation and phosphorylation play important yet distinct roles in regulating α-syn conformation, membrane binding, and amyloid aggregation. However, how PTMs regulate α-syn function in presynaptic terminals remains unclear. Previously, we reported that α-syn clusters synaptic vesicles (SV) 1, and neutral phospholipid lysophosphatidylcholine (LPC) can mediate this clustering 2. Here, based on our previous findings, we further demonstrate that N-terminal acetylation, which occurs under physiological condition and is irreversible in mammalian cells, significantly enhances the functional activity of α-syn in clustering SVs. Mechanistic studies reveal that this enhancement is caused by the N-acetylation-promoted insertion of α-syn's N-terminus and increased intermolecular interactions on the LPC-containing membrane. Our work demonstrates that N-acetylation fine-tunes α-syn-LPC interaction for mediating α-syn's function in SV clustering.

7.
Brain ; 147(6): 2185-2202, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38242640

RÉSUMÉ

Heterozygous de novo mutations in the neuronal protein Munc18-1/STXBP1 cause syndromic neurological symptoms, including severe epilepsy, intellectual disability, developmental delay, ataxia and tremor, summarized as STXBP1 encephalopathies. Although haploinsufficiency is the prevailing disease mechanism, it remains unclear how the reduction in Munc18-1 levels causes synaptic dysfunction in disease as well as how haploinsufficiency alone can account for the significant heterogeneity among patients in terms of the presence, onset and severity of different symptoms. Using biochemical and cell biological readouts on mouse brains, cultured mouse neurons and heterologous cells, we found that the synaptic Munc18-1 interactors Doc2A and Doc2B are unstable in the absence of Munc18-1 and aggregate in the presence of disease-causing Munc18-1 mutants. In haploinsufficiency-mimicking heterozygous knockout neurons, we found a reduction in Doc2A/B levels that is further aggravated by the presence of the disease-causing Munc18-1 mutation G544D as well as an impairment in Doc2A/B synaptic targeting in both genotypes. We also demonstrated that overexpression of Doc2A/B partially rescues synaptic dysfunction in heterozygous knockout neurons but not heterozygous knockout neurons expressing G544D Munc18-1. Our data demonstrate that STXBP1 encephalopathies are not only characterized by the dysfunction of Munc18-1 but also by the dysfunction of the Munc18-1 binding partners Doc2A and Doc2B, and that this dysfunction is exacerbated by the presence of a Munc18-1 missense mutant. These findings may offer a novel explanation for the significant heterogeneity in symptoms observed among STXBP1 encephalopathy patients.


Sujet(s)
Protéines de liaison au calcium , Protéines Munc18 , Mutation , Protéines de tissu nerveux , Neurones , Synapses , Animaux , Humains , Souris , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique , Cellules cultivées , Souris de lignée C57BL , Souris knockout , Protéines Munc18/génétique , Protéines Munc18/métabolisme , Mutation/génétique , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Neurones/métabolisme , Synapses/métabolisme , Synapses/génétique
8.
Proc Natl Acad Sci U S A ; 120(44): e2310174120, 2023 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-37883437

RÉSUMÉ

α-synuclein (α-Syn) is a presynaptic protein that is involved in Parkinson's and other neurodegenerative diseases and binds to negatively charged phospholipids. Previously, we reported that α-Syn clusters synthetic proteoliposomes that mimic synaptic vesicles. This vesicle-clustering activity depends on a specific interaction of α-Syn with anionic phospholipids. Here, we report that α-Syn surprisingly also interacts with the neutral phospholipid lysophosphatidylcholine (lysoPC). Even in the absence of anionic lipids, lysoPC facilitates α-Syn-induced vesicle clustering but has no effect on Ca2+-triggered fusion in a single vesicle-vesicle fusion assay. The A30P mutant of α-Syn that causes familial Parkinson disease has a reduced affinity to lysoPC and does not induce vesicle clustering. Taken together, the α-Syn-lysoPC interaction may play a role in α-Syn function.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Humains , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme , Vésicules synaptiques/métabolisme , Lysolécithine/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Phospholipides/métabolisme
9.
Trends Neurosci ; 46(2): 153-166, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36567199

RÉSUMÉ

α-Synuclein is a neuronal protein that is enriched in presynaptic terminals. Under physiological conditions, it binds to synaptic vesicle membranes and functions in neurotransmitter release, although the molecular details remain unclear, and it is controversial whether α-synuclein inhibits or facilitates neurotransmitter release. Pathologically, in synucleinopathies including Parkinson's disease (PD), α-synuclein forms aggregates that recruit monomeric α-synuclein and spread throughout the brain, which triggers neuronal dysfunction at molecular, cellular, and organ levels. Here, we present an overview of the effects of α-synuclein on SNARE-complex assembly, neurotransmitter release, and synaptic vesicle pool homeostasis, and discuss how the observed divergent effects of α-synuclein on neurotransmitter release can be reconciled. We also discuss how gain-of-function versus loss-of-function of α-synuclein may contribute to pathogenesis in synucleinopathies.


Sujet(s)
Maladie de Parkinson , Synucléinopathies , Humains , alpha-Synucléine/métabolisme , Synucléinopathies/métabolisme , Maladie de Parkinson/métabolisme , Vésicules synaptiques/métabolisme , Agents neuromédiateurs/métabolisme
10.
J Mol Biol ; 435(1): 167714, 2023 01 15.
Article de Anglais | MEDLINE | ID: mdl-35787839

RÉSUMÉ

α-Synuclein is an abundant protein at the neuronal synapse that has been implicated in Parkinson's disease for over 25 years and characterizes the hallmark pathology of a group of neurodegenerative diseases now known as the synucleinopathies. Physiologically, α-synuclein exists in an equilibrium between a synaptic vesicle membrane-bound α-helical multimer and a cytosolic largely unstructured monomer. Through its membrane-bound state, α-synuclein functions in neurotransmitter release by modulating several steps in the synaptic vesicle cycle, including synaptic vesicle clustering and docking, SNARE complex assembly, and homeostasis of synaptic vesicle pools. These functions have been ascribed to α-synuclein's interactions with the synaptic vesicle SNARE protein VAMP2/synaptobrevin-2, the synaptic vesicle-attached synapsins, and the synaptic vesicle membrane itself. How α-synuclein affects these processes, and whether disease is due to loss-of-function or gain-of-toxic-function of α-synuclein remains unclear. In this review, we provide an in-depth summary of the existing literature, discuss possible reasons for the discrepancies in the field, and propose a working model that reconciles the findings in the literature.


Sujet(s)
Maladie de Parkinson , Protéines SNARE , Synapses , alpha-Synucléine , Humains , alpha-Synucléine/métabolisme , Maladie de Parkinson/métabolisme , Protéines SNARE/métabolisme , Synapses/métabolisme , Vésicules synaptiques/métabolisme , Synaptobrévine-2/métabolisme
11.
Nat Commun ; 13(1): 4918, 2022 08 22.
Article de Anglais | MEDLINE | ID: mdl-35995799

RÉSUMÉ

Considerable evidence supports the release of pathogenic aggregates of the neuronal protein α-Synuclein (αSyn) into the extracellular space. While this release is proposed to instigate the neuron-to-neuron transmission and spread of αSyn pathology in synucleinopathies including Parkinson's disease, the molecular-cellular mechanism(s) remain unclear. To study this, we generated a new mouse model to specifically immunoisolate neuronal lysosomes, and established a long-term culture model where αSyn aggregates are produced within neurons without the addition of exogenous fibrils. We show that neuronally generated pathogenic species of αSyn accumulate within neuronal lysosomes in mouse brains and primary neurons. We then find that neurons release these pathogenic αSyn species via SNARE-dependent lysosomal exocytosis. The released aggregates are non-membrane enveloped and seeding-competent. Additionally, we find that this release is dependent on neuronal activity and cytosolic Ca2+. These results propose lysosomal exocytosis as a central mechanism for the release of aggregated and degradation-resistant proteins from neurons.


Sujet(s)
Synucléinopathies , alpha-Synucléine , Animaux , Exocytose , Lysosomes/métabolisme , Souris , Neurones/métabolisme , alpha-Synucléine/métabolisme
12.
Cell Rep ; 39(2): 110675, 2022 04 12.
Article de Anglais | MEDLINE | ID: mdl-35417693

RÉSUMÉ

α-synuclein, ß-synuclein, and γ-synuclein are abundantly expressed proteins in the vertebrate nervous system. α-synuclein functions in neurotransmitter release by binding to and clustering synaptic vesicles and chaperoning SNARE-complex assembly. Pathologically, aggregates originating from soluble pools of α-synuclein are deposited into Lewy bodies in Parkinson's disease and related synucleinopathies. The functions of ß-synuclein and γ-synuclein in presynaptic terminals remain poorly studied. Using in vitro liposome binding studies, circular dichroism spectroscopy, immunoprecipitation, and fluorescence resonance energy transfer (FRET) experiments on isolated synaptic vesicles in combination with subcellular fractionation of brains from synuclein mouse models, we show that ß-synuclein and γ-synuclein have a reduced affinity toward synaptic vesicles compared with α-synuclein, and that heteromerization of ß-synuclein or γ-synuclein with α-synuclein results in reduced synaptic vesicle binding of α-synuclein in a concentration-dependent manner. Our data suggest that ß-synuclein and γ-synuclein are modulators of synaptic vesicle binding of α-synuclein and thereby reduce α-synuclein's physiological activity at the neuronal synapse.


Sujet(s)
Vésicules synaptiques , alpha-Synucléine , Animaux , Souris , Terminaisons présynaptiques/métabolisme , Vésicules synaptiques/métabolisme , alpha-Synucléine/métabolisme , bêta-Synucléine/métabolisme , gamma-Synucléine/métabolisme
14.
J Neurochem ; 157(2): 165-178, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-32643187

RÉSUMÉ

Mutations in Munc18-1/STXBP1 (syntaxin-binding protein 1) are linked to various severe early epileptic encephalopathies and neurodevelopmental disorders. Heterozygous mutations in the STXBP1 gene include missense, nonsense, frameshift, and splice site mutations, as well as intragenic deletions and duplications and whole-gene deletions. No genotype-phenotype correlation has been identified so far, and patients are treated by anti-epileptic drugs because of the lack of a specific disease-modifying therapy. The molecular disease mechanisms underlying STXBP1-linked disorders are yet to be fully understood, but both haploinsufficiency and dominant-negative mechanisms have been proposed. This review focuses on the current understanding of the phenotypic spectrum of STXBP1-linked disorders, as well as discusses disease mechanisms in the context of the numerous pathways in which STXBP1 functions in the brain. We additionally evaluate the available animal models to study these disorders and highlight potential therapeutic approaches for treating these devastating diseases.


Sujet(s)
Anticonvulsivants/usage thérapeutique , Encéphalopathies/métabolisme , Protéines Munc18/métabolisme , Troubles du développement neurologique/traitement médicamenteux , Animaux , Encéphale/métabolisme , Encéphalopathies/génétique , Humains , Protéines Munc18/génétique , Mutation/génétique , Troubles du développement neurologique/génétique
15.
EMBO Mol Med ; 13(1): e12354, 2021 01 11.
Article de Anglais | MEDLINE | ID: mdl-33332765

RÉSUMÉ

Heterozygous de novo mutations in the neuronal protein Munc18-1 cause syndromic neurological symptoms, including severe epilepsy, intellectual disability, developmental delay, ataxia, and tremor. No disease-modifying therapy exists to treat these disorders, and while chemical chaperones have been shown to alleviate neuronal dysfunction caused by missense mutations in Munc18-1, their required high concentrations and potential toxicity necessitate a Munc18-1-targeted therapy. Munc18-1 is essential for neurotransmitter release, and mutations in Munc18-1 have been shown to cause neuronal dysfunction via aggregation and co-aggregation of the wild-type protein, reducing functional Munc18-1 levels well below hemizygous levels. Here, we identify two pharmacological chaperones via structure-based drug design, that bind to wild-type and mutant Munc18-1, and revert Munc18-1 aggregation and neuronal dysfunction in vitro and in vivo, providing the first targeted treatment strategy for these severe pediatric encephalopathies.


Sujet(s)
Encéphalopathies , Épilepsie , Ataxie/traitement médicamenteux , Ataxie/génétique , Enfant , Hétérozygote , Humains , Protéines Munc18/génétique
16.
Nat Struct Mol Biol ; 27(2): 192-201, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-32042150

RÉSUMÉ

Point mutations in cysteine string protein-α (CSPα) cause dominantly inherited adult-onset neuronal ceroid lipofuscinosis (ANCL), a rapidly progressing and lethal neurodegenerative disease with no treatment. ANCL mutations are proposed to trigger CSPα aggregation/oligomerization, but the mechanism of oligomer formation remains unclear. Here we use purified proteins, mouse primary neurons and patient-derived induced neurons to show that the normally palmitoylated cysteine string region of CSPα loses palmitoylation in ANCL mutants. This allows oligomerization of mutant CSPα via ectopic binding of iron-sulfur (Fe-S) clusters. The resulting oligomerization of mutant CSPα causes its mislocalization and consequent loss of its synaptic SNARE-chaperoning function. We then find that pharmacological iron chelation mitigates the oligomerization of mutant CSPα, accompanied by partial rescue of the downstream SNARE defects and the pathological hallmark of lipofuscin accumulation. Thus, the iron chelators deferiprone (L1) and deferoxamine (Dfx), which are already used to treat iron overload in humans, offer a new approach for treating ANCL.


Sujet(s)
Protéines du choc thermique HSP40/génétique , Protéines membranaires/génétique , Céroïdes-lipofuscinoses neuronales/génétique , Mutation ponctuelle , Agrégation pathologique de protéines/génétique , Animaux , Cellules cultivées , Femelle , Cellules HEK293 , Protéines du choc thermique HSP40/métabolisme , Humains , Agents chélateurs du fer/métabolisme , Lipoylation , Protéines membranaires/métabolisme , Souris , Céroïdes-lipofuscinoses neuronales/métabolisme , Neurones/métabolisme , Agrégation pathologique de protéines/métabolisme , Liaison aux protéines , Multimérisation de protéines
17.
Nat Commun ; 9(1): 3986, 2018 09 28.
Article de Anglais | MEDLINE | ID: mdl-30266908

RÉSUMÉ

Heterozygous de novo mutations in the neuronal protein Munc18-1 are linked to epilepsies, intellectual disability, movement disorders, and neurodegeneration. These devastating diseases have a poor prognosis and no known cure, due to lack of understanding of the underlying disease mechanism. To determine how mutations in Munc18-1 cause disease, we use newly generated S. cerevisiae strains, C. elegans models, and conditional Munc18-1 knockout mouse neurons expressing wild-type or mutant Munc18-1, as well as in vitro studies. We find that at least five disease-linked missense mutations of Munc18-1 result in destabilization and aggregation of the mutant protein. Aggregates of mutant Munc18-1 incorporate wild-type Munc18-1, depleting functional Munc18-1 levels beyond hemizygous levels. We demonstrate that the three chemical chaperones 4-phenylbutyrate, sorbitol, and trehalose reverse the deficits caused by mutations in Munc18-1 in vitro and in vivo in multiple models, offering a novel strategy for the treatment of varied encephalopathies.


Sujet(s)
Encéphalopathies/génétique , Protéines Munc18/génétique , Mutation faux-sens , Composés chimiques organiques/pharmacologie , Animaux , Encéphalopathies/métabolisme , Encéphalopathies/prévention et contrôle , Protéines de Caenorhabditis elegans/métabolisme , Lignée cellulaire tumorale , Cellules cultivées , Cellules HEK293 , Humains , Souris knockout , Protéines Munc18/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Phénylbutyrates/pharmacologie , Agrégats de protéines/effets des médicaments et des substances chimiques , Agrégation pathologique de protéines/prévention et contrôle , Protéines de Saccharomyces cerevisiae/métabolisme , Sorbitol/pharmacologie , Tréhalose/pharmacologie
18.
Article de Anglais | MEDLINE | ID: mdl-28108534

RÉSUMÉ

α-Synuclein is an abundant neuronal protein that is highly enriched in presynaptic nerve terminals. Genetics and neuropathology studies link α-synuclein to Parkinson's disease (PD) and other neurodegenerative disorders. Accumulation of misfolded oligomers and larger aggregates of α-synuclein defines multiple neurodegenerative diseases called synucleinopathies, but the mechanisms by which α-synuclein acts in neurodegeneration are unknown. Moreover, the normal cellular function of α-synuclein remains debated. In this perspective, we review the structural characteristics of α-synuclein, its developmental expression pattern, its cellular and subcellular localization, and its function in neurons. We also discuss recent progress on secretion of α-synuclein, which may contribute to its interneuronal spread in a prion-like fashion, and describe the neurotoxic effects of α-synuclein that are thought to be responsible for its role in neurodegeneration.


Sujet(s)
Maladie de Parkinson/physiopathologie , alpha-Synucléine/composition chimique , alpha-Synucléine/physiologie , Animaux , Humains , Souris , Souris transgéniques , Neurones/métabolisme , Synapses/physiologie
20.
PLoS Biol ; 13(10): e1002267, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26437117

RÉSUMÉ

In forebrain neurons, Ca(2+) triggers exocytosis of readily releasable vesicles by binding to synaptotagmin-1 and -7, thereby inducing fast and slow vesicle exocytosis, respectively. Loss-of-function of synaptotagmin-1 or -7 selectively impairs the fast and slow phase of release, respectively, but does not change the size of the readily-releasable pool (RRP) of vesicles as measured by stimulation of release with hypertonic sucrose, or alter the rate of vesicle priming into the RRP. Here we show, however, that simultaneous loss-of-function of both synaptotagmin-1 and -7 dramatically decreased the capacity of the RRP, again without altering the rate of vesicle priming into the RRP. Either synaptotagmin-1 or -7 was sufficient to rescue the RRP size in neurons lacking both synaptotagmin-1 and -7. Although maintenance of RRP size was Ca(2+)-independent, mutations in Ca(2+)-binding sequences of synaptotagmin-1 or synaptotagmin-7--which are contained in flexible top-loop sequences of their C2 domains--blocked the ability of these synaptotagmins to maintain the RRP size. Both synaptotagmins bound to SNARE complexes; SNARE complex binding was reduced by the top-loop mutations that impaired RRP maintenance. Thus, synaptotagmin-1 and -7 perform redundant functions in maintaining the capacity of the RRP in addition to nonredundant functions in the Ca(2+) triggering of different phases of release.


Sujet(s)
Signalisation calcique , Hippocampe/métabolisme , Protéines de tissu nerveux/métabolisme , Neurones/métabolisme , Vésicules synaptiques/métabolisme , Synaptotagmine I/métabolisme , Synaptotagmines/métabolisme , Animaux , Animaux nouveau-nés , Sites de fixation , Cellules cultivées , Potentiels post-synaptiques excitateurs , Cellules HEK293 , Hippocampe/cytologie , Hippocampe/ultrastructure , Humains , Potentiels post-synaptiques inhibiteurs , Souris knockout , Mutation , Protéines de tissu nerveux/antagonistes et inhibiteurs , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/génétique , Neurones/cytologie , Neurones/ultrastructure , Interférence par ARN , Protéines recombinantes/composition chimique , Protéines recombinantes/métabolisme , Protéines SNARE/métabolisme , Vésicules synaptiques/ultrastructure , Synaptotagmine I/composition chimique , Synaptotagmine I/génétique , Synaptotagmines/antagonistes et inhibiteurs , Synaptotagmines/composition chimique , Synaptotagmines/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE