Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.768
Filtrer
1.
Cell Rep ; 43(7): 114428, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38996073

RÉSUMÉ

To achieve the functional polarization that underlies brain computation, neurons sort protein material into distinct compartments. Ion channel composition, for example, differs between axons and dendrites, but the molecular determinants for their polarized trafficking remain obscure. Here, we identify mechanisms that target voltage-gated Ca2+ channels (CaVs) to distinct subcellular compartments. In hippocampal neurons, CaV2s trigger neurotransmitter release at the presynaptic active zone, and CaV1s localize somatodendritically. After knockout of all three CaV2s, expression of CaV2.1, but not CaV1.3, restores neurotransmitter release. We find that chimeric CaV1.3s with CaV2.1 intracellular C-termini localize to the active zone, mediate synaptic vesicle exocytosis, and render release sensitive to CaV1 blockers. This dominant targeting function of the CaV2.1 C-terminus requires the first EF hand in its proximal segment, and replacement of the CaV2.1 C-terminus with that of CaV1.3 abolishes CaV2.1 active zone localization and function. We conclude that CaV intracellular C-termini mediate compartment-specific targeting.


Sujet(s)
Hippocampe , Animaux , Hippocampe/métabolisme , Canaux calciques de type L/métabolisme , Canaux calciques de type L/génétique , Canaux calciques/métabolisme , Canaux calciques de type N/métabolisme , Canaux calciques de type N/génétique , Neurones/métabolisme , Humains , Souris , Rats , Vésicules synaptiques/métabolisme , Exocytose , Cellules HEK293
2.
Mikrochim Acta ; 191(7): 435, 2024 06 29.
Article de Anglais | MEDLINE | ID: mdl-38949689

RÉSUMÉ

A novel scaffold for in situ electrochemical detection of cell biomarkers was developed using electrospun nanofibers and commercial adhesive polymeric membranes. The electrochemical sensing of cell biomarkers requires the cultivation of the cells on/near the (bio)sensor surface in a manner to preserve an appropriate electroactive available surface and to avoid the surface passivation and sensor damage. This can be achieved by employing biocompatible nanofiber meshes that allow the cells to have a normal behavior and do not alter the electrochemical detection. For a better mechanical stability and ease of handling, nylon 6/6 nanofibers were collected on commercial polymeric membranes, at an optimal fiber density, obtaining a double-layered platform. To demonstrate the functionality of the fabricated scaffold, the screening of cellular stress has been achieved integrating melanoma B16-F10 cells and the (bio)sensor components on the transducer whereas the melanin exocytosis was successfully quantified using a commercial electrode. Either directly on the surface of the (bio)sensor or spatially detached from it, the integration of cell cultures in biosensing platforms based on electrospun nanofibers represents a powerful bioanalytical tool able to provide real-time information about the biomarker release, enzyme activity or inhibition, and monitoring of various cellular events.


Sujet(s)
Techniques de biocapteur , Techniques électrochimiques , Nanofibres , Nanofibres/composition chimique , Animaux , Souris , Techniques électrochimiques/méthodes , Techniques électrochimiques/instrumentation , Techniques de biocapteur/méthodes , Lignée cellulaire tumorale , Mélanines , Marqueurs biologiques/analyse , Structures d'échafaudage tissulaires/composition chimique , Exocytose , Mélanome expérimental/anatomopathologie , Mélanome expérimental/diagnostic
3.
Biochemistry ; 63(14): 1837-1857, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38953497

RÉSUMÉ

Munc18-1 is an SM (sec1/munc-like) family protein involved in vesicle fusion and neuronal exocytosis. Munc18-1 is known to regulate the exocytosis process by binding with closed- and open-state conformations of Syntaxin1, a protein belonging to the SNARE family established to be central to the exocytosis process. Our previous work studied peptide p5 as a promising drug candidate for CDK5-p25 complex, an Alzheimer's disease (AD) pathological target. Experimental in vivo and in vitro studies suggest that Munc18-1 promotes p5 to selectively inhibit the CDK5-p25 complex without affecting the endogenous CDK5 activity, a characteristic of remarkable therapeutic implications. In this paper, we identify several binding modes of p5 with Munc18-1 that could potentially affect the Munc18-1 binding with SNARE proteins and lead to off-target effects on neuronal communication using molecular dynamics simulations. Recent studies indicate that disruption of Munc18-1 function not only disrupts neurotransmitter release but also results in neurodegeneration, exhibiting clinical resemblance to other neurodegenerative conditions such as AD, causing diagnostic and treatment challenges. We characterize such interactions between p5 and Munc18-1, define the corresponding pharmacophores, and provide guidance for the in vitro validation of our findings to improve therapeutic efficacy and safety of p5.


Sujet(s)
Exocytose , Simulation de dynamique moléculaire , Protéines Munc18 , Neurones , Protéines Munc18/métabolisme , Protéines Munc18/composition chimique , Protéines Munc18/génétique , Exocytose/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Humains , Kinase-5 cycline-dépendante/métabolisme , Kinase-5 cycline-dépendante/composition chimique , Liaison aux protéines , Peptides/composition chimique , Peptides/pharmacologie , Peptides/métabolisme , Animaux
4.
J Physiol ; 602(15): 3793-3814, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39004870

RÉSUMÉ

High voltage-gated Ca2+ channels (HVCCs) shape the electrical activity and control hormone release in most endocrine cells. HVCCs are multi-subunit protein complexes formed by the pore-forming α1 and the auxiliary ß, α2δ and γ subunits. Four genes code for the α2δ isoforms. At the mRNA level, mouse chromaffin cells (MCCs) express predominantly the CACNA2D1 gene coding for the α2δ-1 isoform. Here we show that α2δ-1 deletion led to ∼60% reduced HVCC Ca2+ influx with slower inactivation kinetics. Pharmacological dissection showed that HVCC composition remained similar in α2δ-1-/- MCCs compared to wild-type (WT), demonstrating that α2δ-1 exerts similar functional effects on all HVCC isoforms. Consistent with reduced HVCC Ca2+ influx, α2δ-1-/- MCCs showed reduced spontaneous electrical activity with action potentials (APs) having a shorter half-maximal duration caused by faster rising and decay slopes. However, the induced electrical activity showed opposite effects with α2δ-1-/- MCCs displaying significantly higher AP frequency in the tonic firing mode as well as an increase in the number of cells firing AP bursts compared to WT. This gain-of-function phenotype was caused by reduced functional activation of Ca2+-dependent K+ currents. Additionally, despite the reduced HVCC Ca2+ influx, the intracellular Ca2+ transients and vesicle exocytosis or endocytosis were unaltered in α2δ-1-/- MCCs compared to WT during sustained stimulation. In conclusion, our study shows that α2δ-1 genetic deletion reduces Ca2+ influx in cultured MCCs but leads to a paradoxical increase in catecholamine secretion due to increased excitability. KEY POINTS: Deletion of the α2δ-1 high voltage-gated Ca2+ channel (HVCC) subunit reduces mouse chromaffin cell (MCC) Ca2+ influx by ∼60% but causes a paradoxical increase in induced excitability. MCC intracellular Ca2+ transients are unaffected by the reduced HVCC Ca2+ influx. Deletion of α2δ-1 reduces the immediately releasable pool vesicle exocytosis but has no effect on catecholamine (CA) release in response to sustained stimuli. The increased electrical activity and CA release from MCCs might contribute to the previously reported cardiovascular phenotype of patients carrying α2δ-1 loss-of-function mutations.


Sujet(s)
Potentiels d'action , Canaux calciques , Cellules chromaffines , Animaux , Cellules chromaffines/métabolisme , Cellules chromaffines/physiologie , Souris , Canaux calciques/génétique , Canaux calciques/métabolisme , Souris knockout , Cellules cultivées , Calcium/métabolisme , Exocytose/physiologie , Souris de lignée C57BL , Mâle
5.
Sci Rep ; 14(1): 17469, 2024 07 29.
Article de Anglais | MEDLINE | ID: mdl-39080379

RÉSUMÉ

Mutations in the lysosomal membrane protein CLN3 cause Juvenile Neuronal Ceroid Lipofuscinosis (JNCL). Activation of the lysosomal ion channel TRPML1 has previously been shown to be beneficial in several neurodegenerative disease models. Here, we tested whether TRPML1 activation rescues disease-associated phenotypes in CLN3-deficient retinal pigment epithelial (ARPE-19 CLN3-KO) cells. ARPE-19 CLN3-KO cells accumulate LAMP1 positive organelles and show lysosomal storage of mitochondrial ATPase subunit C (SubC), globotriaosylceramide (Gb3), and glycerophosphodiesters (GPDs), whereas lysosomal bis(monoacylglycero)phosphate (BMP/LBPA) lipid levels were significantly decreased. Activation of TRPML1 reduced lysosomal storage of Gb3 and SubC but failed to restore BMP levels in CLN3-KO cells. TRPML1-mediated decrease of storage was TFEB-independent, and we identified TRPML1-mediated enhanced lysosomal exocytosis as a likely mechanism for clearing storage including GPDs. Therefore, ARPE-19 CLN3-KO cells represent a human cell model for CLN3 disease showing many of the described core lysosomal deficits, some of which can be improved using TRPML1 agonists.


Sujet(s)
Lysosomes , Glycoprotéines membranaires , Chaperons moléculaires , Céroïdes-lipofuscinoses neuronales , Épithélium pigmentaire de la rétine , Canaux cationiques TRP , Lysosomes/métabolisme , Humains , Épithélium pigmentaire de la rétine/métabolisme , Chaperons moléculaires/métabolisme , Chaperons moléculaires/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Céroïdes-lipofuscinoses neuronales/métabolisme , Céroïdes-lipofuscinoses neuronales/génétique , Céroïdes-lipofuscinoses neuronales/anatomopathologie , Canaux cationiques TRP/métabolisme , Canaux cationiques TRP/génétique , Phénotype , Lignée cellulaire , Exocytose , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Lysophospholipides , Monoglycérides
6.
Curr Opin Cell Biol ; 89: 102401, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39018789

RÉSUMÉ

Synthesizing the recent progresses, we present our perspectives on how local modulations of membrane curvature, tension, and bending energy define the feedback controls over membrane traffic processes. We speculate the potential mechanisms of, and the control logic behind, the different membrane mechanics-mediated feedback in endocytosis and exo-endocytosis coupling. We elaborate the path forward with the open questions for theoretical considerations and the grand challenges for experimental validations.


Sujet(s)
Membrane cellulaire , Endocytose , Membrane cellulaire/métabolisme , Endocytose/physiologie , Humains , Animaux , Phénomènes biomécaniques , Rétrocontrôle physiologique , Exocytose/physiologie
7.
J Neurosci ; 44(31)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-38951039

RÉSUMÉ

The release of neurotransmitters (NTs) at central synapses is dependent on a cascade of protein interactions, specific to the presynaptic compartment. Among those dedicated molecules, the cytosolic complexins play an incompletely defined role as synaptic transmission regulators. Complexins are multidomain proteins that bind soluble N-ethylmaleimide sensitive factor attachment protein receptor complexes, conferring both inhibitory and stimulatory functions. Using systematic mutagenesis and comparing reconstituted in vitro membrane fusion assays with electrophysiology in cultured neurons from mice of either sex, we deciphered the function of the N-terminus of complexin (Cpx) II. The N-terminus (amino acid 1-27) starts with a region enriched in hydrophobic amino acids (1-12), which binds lipids. Mutants maintaining this hydrophobic character retained the stimulatory function of Cpx, whereas exchanges introducing charged residues perturbed both spontaneous and evoked exocytosis. Mutants in the more distal region of the N-terminal domain (amino acid 11-18) showed a spectrum of effects. On the one hand, mutation of residue A12 increased spontaneous release without affecting evoked release. On the other hand, replacing D15 with amino acids of different shapes or hydrophobic properties (but not charge) not only increased spontaneous release but also impaired evoked release. Most surprising, this substitution reduced the size of the readily releasable pool, a novel function for Cpx at mammalian synapses. Thus, the exact amino acid composition of the Cpx N-terminus fine-tunes the degree of spontaneous and evoked NT release.


Sujet(s)
Protéines de tissu nerveux , Vésicules synaptiques , Animaux , Vésicules synaptiques/métabolisme , Vésicules synaptiques/génétique , Souris , Mâle , Femelle , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/composition chimique , Mutation , Protéines adaptatrices du transport vésiculaire/génétique , Protéines adaptatrices du transport vésiculaire/métabolisme , Protéines adaptatrices du transport vésiculaire/composition chimique , Fusion membranaire/physiologie , Fusion membranaire/génétique , Cellules cultivées , Phénotype , Neurones/métabolisme , Transmission synaptique/génétique , Transmission synaptique/physiologie , Souris de lignée C57BL , Exocytose/physiologie , Exocytose/génétique
8.
Biosci Rep ; 44(7)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39011584

RÉSUMÉ

Botulinum neurotoxins (BoNTs) are valuable tools to unveil molecular mechanisms of exocytosis in neuronal and non-neuronal cells due to their peptidase activity on exocytic isoforms of SNARE proteins. They are produced by Clostridia as single-chain polypeptides that are proteolytically cleaved into light, catalytic domains covalently linked via disulfide bonds to heavy, targeting domains. This format of two subunits linked by disulfide bonds is required for the full neurotoxicity of BoNTs. We have generated a recombinant version of BoNT/B that consists of the light chain of the toxin fused to the protein transduction domain of the human immunodeficiency virus-1 (TAT peptide) and a hexahistidine tag. His6-TAT-BoNT/B-LC, expressed in Escherichia coli and purified by affinity chromatography, penetrated membranes and exhibited strong enzymatic activity, as evidenced by cleavage of the SNARE synaptobrevin from rat brain synaptosomes and human sperm cells. Proteolytic attack of synaptobrevin hindered exocytosis triggered by a calcium ionophore in the latter. The novel tool reported herein disrupts the function of a SNARE protein within minutes in cells that may or may not express the receptors for the BoNT/B heavy chain, and without the need for transient transfection or permeabilization.


Sujet(s)
Toxines botuliniques de type A , Exocytose , Animaux , Humains , Rats , Toxines botuliniques de type A/métabolisme , Toxines botuliniques de type A/génétique , Toxines botuliniques de type A/isolement et purification , Protéines SNARE/métabolisme , Protéines SNARE/génétique , Mâle , Synaptosomes/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Protéines recombinantes/isolement et purification , Protéines recombinantes/génétique , Perméabilité des membranes cellulaires/effets des médicaments et des substances chimiques , Toxines botuliniques/métabolisme , Toxines botuliniques/génétique , Toxines botuliniques/composition chimique , Toxines botuliniques/isolement et purification
9.
Cell Physiol Biochem ; 58(3): 212-225, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38852193

RÉSUMÉ

BACKGROUND/AIMS: Adrenaline quickly inhibits the release of histamine from mast cells. Besides ß2-adrenergic receptors, several in vitro studies also indicate the involvement of α-adrenergic receptors in the process of exocytosis. Since exocytosis in mast cells can be detected electrophysiologically by the changes in the membrane capacitance (Cm), its continuous monitoring in the presence of drugs would determine their mast cell-stabilizing properties. METHODS: Employing the whole-cell patch-clamp technique in rat peritoneal mast cells, we examined the effects of adrenaline on the degranulation of mast cells and the increase in the Cm during exocytosis. We also examined the degranulation of mast cells in the presence or absence of α-adrenergic receptor agonists or antagonists. RESULTS: Adrenaline dose-dependently suppressed the GTP-γ-S-induced increase in the Cm and inhibited the degranulation from mast cells, which was almost completely erased in the presence of butoxamine, a ß2-adrenergic receptor antagonist. Among α-adrenergic receptor agonists or antagonists, high dose prazosin, a selective α1-adrenergic receptor antagonist, significantly reduced the ratio of degranulating mast cells and suppressed the increase in the Cm. Additionally, prazosin augmented the inhibitory effects of adrenaline on the degranulation of mast cells. CONCLUSION: This study provided electrophysiological evidence for the first time that adrenaline dose-dependently inhibited the process of exocytosis, confirming its usefulness as a potent mast cell-stabilizer. The pharmacological blockade of α1-adrenergic receptor by prazosin synergistically potentiated such mast cell-stabilizing property of adrenaline, which is primarily mediated by ß2-adrenergic receptors.


Sujet(s)
Dégranulation cellulaire , Épinéphrine , Exocytose , Mastocytes , Prazosine , Animaux , Mastocytes/effets des médicaments et des substances chimiques , Mastocytes/métabolisme , Mastocytes/cytologie , Épinéphrine/pharmacologie , Rats , Prazosine/pharmacologie , Dégranulation cellulaire/effets des médicaments et des substances chimiques , Mâle , Exocytose/effets des médicaments et des substances chimiques , Techniques de patch-clamp , Antagonistes des récepteurs alpha-1 adrénergiques/pharmacologie , Rat Wistar
11.
J Cell Biol ; 223(10)2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-38935075

RÉSUMÉ

Regulated cell shape change requires the induction of cortical cytoskeletal domains. Often, local changes to plasma membrane (PM) topography are involved. Centrosomes organize cortical domains and can affect PM topography by locally pulling the PM inward. Are these centrosome effects coupled? At the syncytial Drosophila embryo cortex, centrosome-induced actin caps grow into dome-like compartments for mitoses. We found the nascent cap to be a collection of PM folds and tubules formed over the astral centrosomal MT array. The localized infoldings require centrosome and dynein activities, and myosin-based surface tension prevents them elsewhere. Centrosome-engaged PM infoldings become specifically enriched with an Arp2/3 induction pathway. Arp2/3 actin network growth between the infoldings counterbalances centrosomal pulling forces and disperses the folds for actin cap expansion. Abnormal domain topography with either centrosome or Arp2/3 disruption correlates with decreased exocytic vesicle association. Together, our data implicate centrosome-organized PM infoldings in coordinating Arp2/3 network growth and exocytosis for cortical domain assembly.


Sujet(s)
Complexe Arp-2-3 , Actines , Membrane cellulaire , Centrosome , Protéines de Drosophila , Drosophila melanogaster , Animaux , Complexe Arp-2-3/métabolisme , Complexe Arp-2-3/génétique , Actines/métabolisme , Membrane cellulaire/métabolisme , Centrosome/métabolisme , Drosophila melanogaster/cytologie , Drosophila melanogaster/croissance et développement , Drosophila melanogaster/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Dynéines/métabolisme , Exocytose , Microtubules/métabolisme
12.
J Cell Sci ; 137(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38899547

RÉSUMÉ

The Rho family of GTPases plays a crucial role in cellular mechanics by regulating actomyosin contractility through the parallel induction of actin and myosin assembly and function. Using exocytosis of large vesicles in the Drosophila larval salivary gland as a model, we followed the spatiotemporal regulation of Rho1, which in turn creates distinct organization patterns of actin and myosin. After vesicle fusion, low levels of activated Rho1 reach the vesicle membrane and drive actin nucleation in an uneven, spread-out pattern. Subsequently, the Rho1 activator RhoGEF2 distributes as an irregular meshwork on the vesicle membrane, activating Rho1 in a corresponding punctate pattern and driving local myosin II recruitment, resulting in vesicle constriction. Vesicle membrane buckling and subsequent crumpling occur at local sites of high myosin II concentrations. These findings indicate that distinct thresholds for activated Rho1 create a biphasic mode of actomyosin assembly, inducing anisotropic membrane crumpling during exocrine secretion.


Sujet(s)
Protéines de Drosophila , Exocytose , Myosine de type II , Protéines G rho , Animaux , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Myosine de type II/métabolisme , Protéines G rho/métabolisme , Protéines G rho/génétique , Exocytose/physiologie , Drosophila melanogaster/métabolisme , Actines/métabolisme , Actomyosine/métabolisme , Larve/métabolisme , Glandes salivaires/métabolisme , Glandes salivaires/cytologie , Facteurs d'échange de nucléotides guanyliques/métabolisme , Facteurs d'échange de nucléotides guanyliques/génétique , Vésicules de sécrétion/métabolisme
13.
J Am Chem Soc ; 146(26): 17747-17756, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38889317

RÉSUMÉ

Unveiling molecular mechanisms that dominate protein phase dynamics has been a pressing need for deciphering the intricate intracellular modulation machinery. While ions and biomacromolecules have been widely recognized for modulating protein phase separations, effects of small molecules that essentially constitute the cytosolic chemical atmosphere on the protein phase behaviors are rarely understood. Herein, we report that vitamin C (VC), a key small molecule for maintaining a reductive intracellular atmosphere, drives reentrant phase transitions of myosin II/F-actin (actomyosin) cytoskeletons. The actomyosin bundle condensates dissemble in the low-VC regime and assemble in the high-VC regime in vitro or inside neuronal cells, through a concurrent myosin II protein aggregation-dissociation process with monotonic VC concentration increase. Based on this finding, we employ in situ single-cell and single-vesicle electrochemistry to demonstrate the quantitative modulation of catecholamine transmitter vesicle exocytosis by intracellular VC atmosphere, i.e., exocytotic release amount increases in the low-VC regime and decreases in the high-VC regime. Furthermore, we show how VC regulates cytomembrane-vesicle fusion pore dynamics through counteractive or synergistic effects of actomyosin phase transitions and the intracellular free calcium level on membrane tensions. Our work uncovers the small molecule-based reversive protein phase regulatory mechanism, paving a new way to chemical neuromodulation and therapeutic repertoire expansion.


Sujet(s)
Actines , Acide ascorbique , Exocytose , Acide ascorbique/composition chimique , Acide ascorbique/pharmacologie , Exocytose/effets des médicaments et des substances chimiques , Actines/métabolisme , Actines/composition chimique , Transition de phase , Animaux , Myosine de type II/métabolisme , Myosine de type II/antagonistes et inhibiteurs , Techniques électrochimiques , Actomyosine/métabolisme , Actomyosine/composition chimique , Rats
14.
Cancer Lett ; 597: 217024, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38871244

RÉSUMÉ

Lysosomes are single membrane bounded group of acidic organelles that can be involved in a process called lysosomal exocytosis which leads to the extracellular release of their content. Lysosomal exocytosis is required for plasma membrane repair or remodeling events such as bone resorption, antigen presentation or mitosis, and for protection against toxic agents such as heavy metals. Recently, it has been showed that to fulfill this protective role, lysosomal exocytosis needs some autophagic proteins, in an autophagy-independent manner. In addition to these crucial physiological roles, lysosomal exocytosis plays a major protumoral role in various cancers. This effect is exerted through tumor microenvironment modifications, including extracellular matrix remodeling, acidosis, oncogenic and profibrogenic signals. This review provides a comprehensive overview of the different elements released in the microenvironment during lysosomal exocytosis, i.e. proteases, exosomes, and protons, and their effects in the context of tumor development and treatment.


Sujet(s)
Exocytose , Lysosomes , Tumeurs , Microenvironnement tumoral , Humains , Lysosomes/métabolisme , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Animaux , Autophagie , Exosomes/métabolisme
15.
Elife ; 122024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38829367

RÉSUMÉ

After exocytosis, release sites are cleared of vesicular residues to replenish with transmitter-filled vesicles. Endocytic and scaffold proteins are thought to underlie this site-clearance mechanism. However, the physiological significance of this mechanism at diverse mammalian central synapses remains unknown. Here, we tested this in a physiologically optimized condition using action potential evoked EPSCs at fast calyx synapse and relatively slow hippocampal CA1 synapse, in post-hearing mice brain slices at 37°C and in 1.3 mM [Ca2+]. Pharmacological block of endocytosis enhanced synaptic depression at the calyx synapse, whereas it attenuated synaptic facilitation at the hippocampal synapse. Block of scaffold protein activity likewise enhanced synaptic depression at the calyx but had no effect at the hippocampal synapse. At the fast calyx synapse, block of endocytosis or scaffold protein activity significantly enhanced synaptic depression as early as 10 ms after the stimulation onset. Unlike previous reports, neither endocytic blockers nor scaffold protein inhibitors prolonged the recovery from short-term depression. We conclude that the release-site clearance by endocytosis can be a universal phenomenon supporting vesicle replenishment at both fast and slow synapses, whereas the presynaptic scaffold mechanism likely plays a specialized role in vesicle replenishment predominantly at fast synapses.


Sujet(s)
Endocytose , Vésicules synaptiques , Endocytose/physiologie , Animaux , Souris , Vésicules synaptiques/métabolisme , Vésicules synaptiques/physiologie , Synapses/physiologie , Hippocampe/physiologie , Exocytose , Région CA1 de l'hippocampe/physiologie
16.
Anal Chem ; 96(25): 10228-10236, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38867346

RÉSUMÉ

Exocytosis of a single cell has been extensively researched in recent years due to its close association with numerous diseases. However, current methods only investigate exocytosis at either the single-cell or multiple-cell level, and a method for simultaneously studying exocytosis at both levels has yet to be established. In this study, a combined device incorporating ultramicroelectrode (UME) electrochemistry and surface plasmon resonance (SPR) was developed, enabling the simultaneous monitoring of single-cell and multiple-cell exocytosis. PC12 cells were cultured directly on the SPR sensing Au film, with a carboxylated carbon nanopipette (c-CNP) electrode employed for electrochemical detection in the SPR reaction cell. Upon exocytosis, the released dopamine diffuses onto the inner wall of c-CNP, undergoing an electrochemical reaction to generate a current peak. Concurrently, exocytosis can also induce changes in the refractive index of the Au film surface, leading to the SPR signal. Consequently, the device enables real-time monitoring of exocytosis from both single and multiple cells with a high spatiotemporal resolution. The c-CNP electrode exhibited excellent resistance to protein contamination, high sensitivity for dopamine detection, and the capability to continuously monitor dopamine exocytosis over an extended period. Analysis of both SPR and electrochemical signals revealed a positive correlation between changes in the SPR signal and the frequency of exocytosis. This study introduces a novel method and platform for the simultaneous investigation of single-cell and multiple-cell exocytosis.


Sujet(s)
Dopamine , Techniques électrochimiques , Exocytose , Microélectrodes , Résonance plasmonique de surface , Cellules PC12 , Animaux , Rats , Techniques électrochimiques/méthodes , Techniques électrochimiques/instrumentation , Dopamine/analyse , Dopamine/métabolisme , Or/composition chimique , Analyse sur cellule unique/instrumentation
17.
Acta Histochem ; 126(4): 152170, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38936136

RÉSUMÉ

We previously reported the presence of P2X3 purinoceptors (P2X3)-expressing subserosal afferent nerve endings consisting of net- and basket-like nerve endings in the rat gastric antrum. These nerve endings may morphologically be vagal mechanoreceptors activated by antral peristalsis. The present study investigated immunoreactivities for vesicular glutamate transporter (VGLUT) 1 and VGLUT2 as well as exocytosis-related proteins, i.e., core components of the SNARE complex (SNAP25, Stx1, and VAMP2) and synaptotagmin-1 (Syt1), in whole-mount preparations of the rat gastric antrum using double immunofluorescence. VGLUT1 immunoreactivity was not detected, whereas VGLUT2 immunoreactivity was observed in P2X3-immunoreactive subserosal nerve endings composed of both net- and basket-like endings. In net-like nerve endings, intense VGLUT2 immunoreactivity was localized in polygonal bulges of reticular nerve fibers and peripheral axon terminals. Furthermore, intense immunoreactivities for SNAP25, Stx1, and VAMP2 were localized in net-like nerve endings. Intense immunoreactivities for VAMP2 and Syt1 were observed in VGLUT2-immunoreactive net-like nerve endings. In basket-like nerve endings, VGLUT2 immunoreactivity was localized in pleomorphic terminal structures and small bulges surrounding the subserosal ganglion, whereas immunoreactivities for SNAP25, Stx1, and VAMP2 were weak in these nerve endings. VGLUT2-immunoreactive basket-like nerve endings were weakly immunoreactive for VAMP2 and Syt1. These results suggest that subserosal afferent nerve endings release glutamate by exocytosis mainly from net-like nerve endings to modulate their mechanoreceptor function.


Sujet(s)
Exocytose , Acide glutamique , Terminaisons nerveuses , Antre pylorique , Récepteurs purinergiques P2X3 , Transporteur vésiculaire-2 du glutamate , Animaux , Mâle , Rats , Acide glutamique/métabolisme , Immunohistochimie , Terminaisons nerveuses/métabolisme , Antre pylorique/innervation , Antre pylorique/métabolisme , Rat Wistar , Récepteurs purinergiques P2X3/métabolisme , Protéine SNAP-25/métabolisme , Synaptotagmine I/métabolisme , Syntaxine-1/métabolisme , Synaptobrévine-2/métabolisme , Transporteur vésiculaire-1 du glutamate/métabolisme , Transporteur vésiculaire-2 du glutamate/métabolisme
18.
EMBO J ; 43(15): 3141-3174, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38877304

RÉSUMÉ

Migrating cells preferentially breach and integrate epithelial and endothelial monolayers at multicellular vertices. These sites are amenable to forces produced by the migrating cell and subsequent opening of the junctions. However, the cues that guide migrating cells to these entry portals, and eventually drive the transmigration process, are poorly understood. Here, we show that lymphatic endothelium multicellular junctions are the preferred sites of dendritic cell transmigration in both primary cell co-cultures and in mouse dermal explants. Dendritic cell guidance to multicellular junctions was dependent on the dendritic cell receptor CCR7, whose ligand, lymphatic endothelial chemokine CCL21, was exocytosed at multicellular junctions. Characterization of lymphatic endothelial secretory routes indicated Golgi-derived RAB6+ vesicles and RAB3+/27+ dense core secretory granules as intracellular CCL21 storage vesicles. Of these, RAB6+ vesicles trafficked CCL21 to the multicellular junctions, which were enriched with RAB6 docking factor ELKS (ERC1). Importantly, inhibition of RAB6 vesicle exocytosis attenuated dendritic cell transmigration. These data exemplify how spatially-restricted exocytosis of guidance cues helps to determine where dendritic cells transmigrate.


Sujet(s)
Chimiokine CCL21 , Cellules dendritiques , Exocytose , Récepteurs CCR7 , Protéines G rab , Animaux , Souris , Chimiokine CCL21/métabolisme , Protéines G rab/métabolisme , Protéines G rab/génétique , Cellules dendritiques/métabolisme , Récepteurs CCR7/métabolisme , Récepteurs CCR7/génétique , Jonctions intercellulaires/métabolisme , Migration transendothéliale et transépithéliale , Endothélium lymphatique/métabolisme , Endothélium lymphatique/cytologie , Cellules endothéliales/métabolisme , Souris de lignée C57BL , Humains , Techniques de coculture , Cellules cultivées , Mouvement cellulaire
19.
J Cell Sci ; 137(20)2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-38690758

RÉSUMÉ

Exocytosis is a fundamental process used by eukaryotes to regulate the composition of the plasma membrane and facilitate cell-cell communication. To investigate exocytosis in neuronal morphogenesis, previously we developed computational tools with a graphical user interface to enable the automatic detection and analysis of exocytic events from fluorescence timelapse images. Although these tools were useful, we found the code was brittle and not easily adapted to different experimental conditions. Here, we developed and validated a robust and versatile toolkit, named pHusion, for the analysis of exocytosis, written in ImageTank, a graphical programming language that combines image visualization and numerical methods. We tested pHusion using a variety of imaging modalities and pH-sensitive fluorophores, diverse cell types and various exocytic markers, to generate a flexible and intuitive package. Using this system, we show that VAMP3-mediated exocytosis occurs 30-times more frequently in melanoma cells compared with primary oligodendrocytes, that VAMP2-mediated fusion events in mature rat hippocampal neurons are longer lasting than those in immature murine cortical neurons, and that exocytic events are clustered in space yet random in time in developing cortical neurons.


Sujet(s)
Exocytose , Animaux , Rats , Souris , Neurones/métabolisme , Neurones/cytologie , Humains , Concentration en ions d'hydrogène , Logiciel , Hippocampe/métabolisme , Hippocampe/cytologie
20.
Sci Rep ; 14(1): 12546, 2024 05 31.
Article de Anglais | MEDLINE | ID: mdl-38822068

RÉSUMÉ

Nanosecond pulsed electric field (nsPEF) has emerged as a promising approach for inducing cell death in melanoma, either as a standalone treatment or in combination with chemotherapeutics. However, to date, there has been a shortage of studies exploring the impact of nsPEF on the expression of cancer-specific molecules. In this investigation, we sought to assess the effects of nsPEF on melanoma-specific MAGE (Melanoma Antigen Gene Protein Family) expression. To achieve this, melanoma cells were exposed to nsPEF with parameters set at 8 kV/cm, 200 ns duration, 100 pulses, and a frequency of 10 kHz. We also aimed to comprehensively describe the consequences of this electric field on melanoma cells' invasion and proliferation potential. Our findings reveal that following exposure to nsPEF, melanoma cells release microvesicles containing MAGE antigens, leading to a simultaneous increase in the expression and mRNA content of membrane-associated antigens such as MAGE-A1. Notably, we observed an unexpected increase in the expression of PD-1 as well. While we did not observe significant differences in the cells' proliferation or invasion potential, a remarkable alteration in the cells' metabolomic and lipidomic profiles towards a less aggressive phenotype was evident. Furthermore, we validated these results using ex vivo tissue cultures and 3D melanoma culture models. Our study demonstrates that nsPEF can elevate the expression of membrane-associated proteins, including melanoma-specific antigens. The mechanism underlying the overexpression of MAGE antigens involves the initial release of microvesicles containing MAGE antigens, followed by a gradual increase in mRNA levels, ultimately resulting in elevated expression of MAGE antigens post-experiment. These findings shed light on a novel method for modulating cancer cells to overexpress cancer-specific molecules, thereby potentially enhancing their sensitivity to targeted anticancer therapy.


Sujet(s)
Exocytose , Antigènes spécifiques du mélanome , Mélanome , Humains , Mélanome/métabolisme , Mélanome/anatomopathologie , Mélanome/génétique , Mélanome/immunologie , Lignée cellulaire tumorale , Antigènes spécifiques du mélanome/métabolisme , Antigènes spécifiques du mélanome/génétique , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Antigènes néoplasiques/métabolisme , Antigènes néoplasiques/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE