Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.055
Filtrar
1.
J Neurosci ; 44(38)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39147590

RESUMEN

Ribbon synapses of inner hair cells (IHCs) are uniquely designed for ultrafast and indefatigable neurotransmission of the sound. The molecular machinery ensuring the efficient, compensatory recycling of the synaptic vesicles (SVs), however, remains elusive. This study showed that hair cell knock-out of murine Dmxl2, whose human homolog is responsible for nonsyndromic sensorineural hearing loss DFNA71, resulted in auditory synaptopathy by impairing synaptic endocytosis and recycling. The mutant mice in the C57BL/6J background of either sex had mild hearing loss with severely diminished wave I amplitude of the auditory brainstem response. Membrane capacitance measurements of the IHCs revealed deficiency in sustained synaptic exocytosis and endocytic membrane retrieval. Consistent with the electrophysiological findings, 3D electron microscopy reconstruction showed reduced reserve pool of SVs and endocytic compartments, while the membrane-proximal and ribbon-associated vesicles remain intact. Our results propose an important role of DMXL2 in hair cell endocytosis and recycling of the SVs.


Asunto(s)
Endocitosis , Células Ciliadas Auditivas Internas , Proteínas del Tejido Nervioso , Vesículas Sinápticas , Animales , Femenino , Masculino , Ratones , Endocitosis/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Exocitosis/fisiología , Células Ciliadas Auditivas Internas/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Vesículas Sinápticas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
2.
Prog Neurobiol ; 240: 102658, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39103114

RESUMEN

Our understanding of how otoferlin, the major calcium sensor in inner hair cells (IHCs) synaptic transmission, contributes to the overall dynamics of synaptic vesicle (SV) trafficking remains limited. To address this question, we generated a knock-in mouse model expressing an otoferlin-GFP protein, where GFP was fused to its C-terminal transmembrane domain. Similar to the wild type protein, the GFP-tagged otoferlin showed normal expression and was associated with IHC SV. Surprisingly, while the heterozygote Otof+/GFP mice exhibited a normal hearing function, homozygote OtofGFP/GFP mice were profoundly deaf attributed to severe reduction in SV exocytosis. Fluorescence recovery after photobleaching revealed a markedly increased mobile fraction of the otof-GFP-associated SV in Otof GFP/GFP IHCs. Correspondingly, 3D-electron tomographic of the ribbon synapses indicated a reduced density of SV attached to the ribbon active zone. Collectively, these results indicate that otoferlin requires a free intravesicular C-terminal end for normal SV docking and fusion.


Asunto(s)
Células Ciliadas Auditivas Internas , Proteínas de la Membrana , Sinapsis , Vesículas Sinápticas , Animales , Vesículas Sinápticas/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Sinapsis/metabolismo , Ratones Transgénicos , Exocitosis/fisiología
3.
Cell Mol Life Sci ; 81(1): 342, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39123091

RESUMEN

A Disintegrin And Metalloproteinase 10 (ADAM10) plays a pivotal role in shaping neuronal networks by orchestrating the activity of numerous membrane proteins through the shedding of their extracellular domains. Despite its significance in the brain, the specific cellular localization of ADAM10 remains not well understood due to a lack of appropriate tools. Here, using a specific ADAM10 antibody suitable for immunostainings, we observed that ADAM10 is localized to presynapses and especially enriched at presynaptic vesicles of mossy fiber (MF)-CA3 synapses in the hippocampus. These synapses undergo pronounced frequency facilitation of neurotransmitter release, a process that play critical roles in information transfer and neural computation. We demonstrate, that in conditional ADAM10 knockout mice the ability of MF synapses to undergo this type of synaptic plasticity is greatly reduced. The loss of facilitation depends on the cytosolic domain of ADAM10 and association with the calcium sensor synaptotagmin 7 rather than ADAM10's proteolytic activity. Our findings unveil a new role of ADAM10 in the regulation of synaptic vesicle exocytosis.


Asunto(s)
Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide , Proteínas de la Membrana , Ratones Noqueados , Plasticidad Neuronal , Vesículas Sinápticas , Animales , Proteína ADAM10/metabolismo , Proteína ADAM10/genética , Plasticidad Neuronal/fisiología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Vesículas Sinápticas/metabolismo , Ratones Endogámicos C57BL , Sinapsis/metabolismo , Fibras Musgosas del Hipocampo/metabolismo , Hipocampo/metabolismo , Exocitosis/fisiología , Terminales Presinápticos/metabolismo , Transmisión Sináptica , Sinaptotagminas/metabolismo , Sinaptotagminas/genética
4.
J Neurosci ; 44(31)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-38951039

RESUMEN

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.


Asunto(s)
Proteínas del Tejido Nervioso , Vesículas Sinápticas , Animales , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/genética , Ratones , Masculino , Femenino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/química , Mutación , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/química , Fusión de Membrana/fisiología , Fusión de Membrana/genética , Células Cultivadas , Fenotipo , Neuronas/metabolismo , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Ratones Endogámicos C57BL , Exocitosis/fisiología , Exocitosis/genética
5.
Biochem Soc Trans ; 52(4): 1715-1725, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39082978

RESUMEN

Various cell types release neurotransmitters, hormones and many other compounds that are stored in secretory vesicles by exocytosis via the formation of a fusion pore traversing the vesicular membrane and the plasma membrane. This process of membrane fusion is mediated by the Soluble N-ethylmaleimide-Sensitive Factor Attachment Proteins REceptor (SNARE) protein complex, which in neurons and neuroendocrine cells is composed of the vesicular SNARE protein Synaptobrevin and the plasma membrane proteins Syntaxin and SNAP25 (Synaptosomal-Associated Protein of 25 kDa). Before a vesicle can undergo fusion and release of its contents, it must dock at the plasma membrane and undergo a process named 'priming', which makes it ready for release. The primed vesicles form the readily releasable pool, from which they can be rapidly released in response to stimulation. The stimulus is an increase in Ca2+ concentration near the fusion site, which is sensed primarily by the vesicular Ca2+ sensor Synaptotagmin. Vesicle priming involves at least the SNARE proteins as well as Synaptotagmin and the accessory proteins Munc18, Munc13, and Complexin but additional proteins may also participate in this process. This review discusses the current views of the interactions and the structural changes that occur among the proteins of the vesicle priming machinery.


Asunto(s)
Exocitosis , Fusión de Membrana , Proteínas SNARE , Proteínas SNARE/metabolismo , Humanos , Animales , Exocitosis/fisiología , Vesículas Secretoras/metabolismo , Sinaptotagminas/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo
6.
Curr Opin Cell Biol ; 89: 102401, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39018789

RESUMEN

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.


Asunto(s)
Membrana Celular , Endocitosis , Membrana Celular/metabolismo , Endocitosis/fisiología , Humanos , Animales , Fenómenos Biomecánicos , Retroalimentación Fisiológica , Exocitosis/fisiología
7.
J Physiol ; 602(15): 3793-3814, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39004870

RESUMEN

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.


Asunto(s)
Potenciales de Acción , Canales de Calcio , Células Cromafines , Animales , Células Cromafines/metabolismo , Células Cromafines/fisiología , Ratones , Canales de Calcio/genética , Canales de Calcio/metabolismo , Ratones Noqueados , Células Cultivadas , Calcio/metabolismo , Exocitosis/fisiología , Ratones Endogámicos C57BL , Masculino
8.
Neurosci Bull ; 40(9): 1379-1395, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38976218

RESUMEN

Endocytosis is a fundamental biological process that couples exocytosis to maintain the homeostasis of the plasma membrane and sustained neurotransmission. Super-resolution microscopy enables optical imaging of exocytosis and endocytosis in live cells and makes an essential contribution to understanding molecular mechanisms of endocytosis in neuronal somata and other types of cells. However, visualization of exo-endocytic events at the single vesicular level in a synapse with optical imaging remains a great challenge to reveal mechanisms governing the synaptic exo-endocytotic coupling. In this protocol, we describe the technical details of stimulated emission depletion (STED) imaging of synaptic endocytosis at the single-vesicle level, from sample preparation and microscopy calibration to data acquisition and analysis.


Asunto(s)
Endocitosis , Sinapsis , Vesículas Sinápticas , Endocitosis/fisiología , Animales , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo , Exocitosis/fisiología , Neuronas/metabolismo , Microscopía Fluorescente/métodos
10.
J Cell Sci ; 137(13)2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38899547

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Exocitosis , Miosina Tipo II , Proteínas de Unión al GTP rho , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Miosina Tipo II/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteínas de Unión al GTP rho/genética , Exocitosis/fisiología , Drosophila melanogaster/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Larva/metabolismo , Glándulas Salivales/metabolismo , Glándulas Salivales/citología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Vesículas Secretoras/metabolismo
11.
Cell Rep ; 43(5): 114218, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38758651

RESUMEN

Glucose has long been considered a primary energy source for synaptic function. However, it remains unclear to what extent alternative fuels, such as lactate/pyruvate, contribute to powering synaptic transmission. By detecting individual release events in hippocampal synapses, we find that mitochondrial ATP production regulates basal vesicle release probability and release location within the active zone (AZ), evoked by single action potentials. Mitochondrial inhibition shifts vesicle release closer to the AZ center and alters the efficiency of vesicle retrieval by increasing the occurrence of ultrafast endocytosis. Furthermore, we uncover that terminals can use oxidative fuels to maintain the vesicle cycle during trains of activity. Mitochondria are sparsely distributed along hippocampal axons, and we find that terminals containing mitochondria display enhanced vesicle release and reuptake during high-frequency trains. Our findings suggest that mitochondria not only regulate several fundamental features of synaptic transmission but may also contribute to modulation of short-term synaptic plasticity.


Asunto(s)
Endocitosis , Exocitosis , Hipocampo , Mitocondrias , Sinapsis , Vesículas Sinápticas , Vesículas Sinápticas/metabolismo , Endocitosis/fisiología , Animales , Hipocampo/metabolismo , Sinapsis/metabolismo , Mitocondrias/metabolismo , Exocitosis/fisiología , Transmisión Sináptica/fisiología , Ratas , Adenosina Trifosfato/metabolismo , Masculino , Potenciales de Acción/fisiología
12.
Neurochem Res ; 49(8): 2021-2037, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38814360

RESUMEN

Acetylcholine is the main neurotransmitter at the vertebrate neuromuscular junctions (NMJs). ACh exocytosis is precisely modulated by co-transmitter ATP and its metabolites. It is assumed that ATP/ADP effects on ACh release rely on activation of presynaptic Gi protein-coupled P2Y13 receptors. However, downstream signaling mechanism of ATP/ADP-mediated modulation of neuromuscular transmission remains elusive. Using microelectrode recording and fluorescent indicators, the mechanism underlying purinergic regulation was studied in the mouse diaphragm NMJs. Pharmacological stimulation of purinoceptors with ADP decreased synaptic vesicle exocytosis evoked by both low and higher frequency stimulation. This inhibitory action was suppressed by antagonists of P2Y13 receptors (MRS 2211), Ca2+ mobilization (TMB8), protein kinase C (chelerythrine) and NADPH oxidase (VAS2870) as well as antioxidants. This suggests the participation of Ca2+ and reactive oxygen species (ROS) in the ADP-triggered signaling. Indeed, ADP caused an increase in cytosolic Ca2+ with subsequent elevation of ROS levels. The elevation of [Ca2+]in was blocked by MRS 2211 and TMB8, whereas upregulation of ROS was prevented by pertussis toxin (inhibitor of Gi protein) and VAS2870. Targeting the main components of lipid rafts, cholesterol and sphingomyelin, suppressed P2Y13 receptor-dependent attenuation of exocytosis and ADP-induced enhancement of ROS production. Inhibition of P2Y13 receptors decreased ROS production and increased the rate of exocytosis during intense activity. Thus, suppression of neuromuscular transmission by exogenous ADP or endogenous ATP can rely on P2Y13 receptor/Gi protein/Ca2+/protein kinase C/NADPH oxidase/ROS signaling, which is coordinated in a lipid raft-dependent manner.


Asunto(s)
Microdominios de Membrana , Unión Neuromuscular , Oxidación-Reducción , Transducción de Señal , Transmisión Sináptica , Animales , Unión Neuromuscular/metabolismo , Unión Neuromuscular/efectos de los fármacos , Microdominios de Membrana/metabolismo , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de los fármacos , Ratones , Transducción de Señal/fisiología , Transducción de Señal/efectos de los fármacos , Masculino , Especies Reactivas de Oxígeno/metabolismo , Exocitosis/fisiología , Exocitosis/efectos de los fármacos , Adenosina Difosfato/metabolismo , Adenosina Difosfato/farmacología , Calcio/metabolismo
13.
Commun Biol ; 7(1): 573, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750123

RESUMEN

Vesicles carry out many essential functions within cells through the processes of endocytosis, exocytosis, and passive and active transport. This includes transporting and delivering molecules between different parts of the cell, and storing and releasing neurotransmitters in neurons. To date, computational simulation of these key biological players has been rather limited and has not advanced at the same pace as other aspects of cell modeling, restricting the realism of computational models. We describe a general vesicle modeling tool that has been designed for wide application to a variety of cell models, implemented within our software STochastic Engine for Pathway Simulation (STEPS), a stochastic reaction-diffusion simulator that supports realistic reconstructions of cell tissue in tetrahedral meshes. The implementation is validated in an extensive test suite, parallel performance is demonstrated in a realistic synaptic bouton model, and example models are visualized in a Blender extension module.


Asunto(s)
Simulación por Computador , Difusión , Modelos Biológicos , Programas Informáticos , Vesículas Sinápticas/metabolismo , Exocitosis/fisiología , Animales , Humanos , Endocitosis/fisiología , Neuronas/fisiología , Neuronas/metabolismo , Procesos Estocásticos
14.
J Clin Invest ; 134(7)2024 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557489

RESUMEN

Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.


Asunto(s)
Canales de Calcio , Calcio , Ratones , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Calcio/metabolismo , Páncreas/metabolismo , Exocitosis/fisiología , Vesículas Secretoras/genética
15.
Biomater Sci ; 12(10): 2660-2671, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38592706

RESUMEN

The endo-lysosomal pathway is a major barrier for the trans-epithelial transport of nanoparticles (NPs), but escape strategies could facilitate trans-epithelial delivery. Based on the polarization properties of the epithelium, different escape compartments may result in different exocytosis fates of NPs and further affect the delivery efficiency. Therefore, optimizing the escape sites is critical for trans-epithelial delivery. Here, commonly used PEG-coated-poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles were fabricated as model nanoparticles (MNPs) and the intestinal epithelium was chosen as the polarized epithelium. The MNPs were incubated with different endosomolytic agents for early endosomal escape, late endosomal escape and lysosomal escape, respectively. According to in vitro and in vivo studies, MNPs escaping from early endosomes and late endosomes exhibited stronger capacity for trans-epithelial transport than those escaping from lysosomes. By further probing into the mechanism, we surprisingly found that although MNPs escaping from early endosomes quickly egressed from the apical side of epithelia, they were subsequently followed by "reuptake" via caveolae and trafficked through the endoplasmic reticulum-Golgi apparatus (ER/GA) secretory pathway, achieving efficient trans-epithelial transport; MNPs escaping from late endosomes, which were located near the nucleus, were prone to enter the ER/GA for efficient basolateral exocytosis. However, MNPs escaping from lysosomes were detained within cells by autophagosomes. Collectively, our research suggested that early endosomes and late endosomes were ideal escape sites for trans-epithelial delivery.


Asunto(s)
Endosomas , Exocitosis , Lisosomas , Nanopartículas , Lisosomas/metabolismo , Exocitosis/fisiología , Animales , Nanopartículas/química , Endosomas/metabolismo , Polietilenglicoles/química , Humanos , Ratones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Perros , Mucosa Intestinal/metabolismo
16.
ACS Chem Biol ; 19(4): 953-961, 2024 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-38566504

RESUMEN

Synaptotagmin-1 (Syt-1) is a calcium sensing protein that is resident in synaptic vesicles. It is well established that Syt-1 is essential for fast and synchronous neurotransmitter release. However, the role of Ca2+ and phospholipid binding in the function of Syt-1, and ultimately in neurotransmitter release, is unclear. Here, we investigate the binding of Ca2+ to Syt-1, first in the absence of lipids, using native mass spectrometry to evaluate individual binding affinities. Syt-1 binds to one Ca2+ with a KD ∼ 45 µM. Each subsequent binding affinity (n ≥ 2) is successively unfavorable. Given that Syt-1 has been reported to bind anionic phospholipids to modulate the Ca2+ binding affinity, we explored the extent that Ca2+ binding was mediated by selected anionic phospholipid binding. We found that phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and dioleoylphosphatidylserine (DOPS) positively modulated Ca2+ binding. However, the extent of Syt-1 binding to phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) was reduced with increasing [Ca2+]. Overall, we find that specific lipids differentially modulate Ca2+ binding. Given that these lipids are enriched in different subcellular compartments and therefore may interact with Syt-1 at different stages of the synaptic vesicle cycle, we propose a regulatory mechanism involving Syt-1, Ca2+, and anionic phospholipids that may also control some aspects of vesicular exocytosis.


Asunto(s)
Calcio , Fosfolípidos , Sinaptotagmina I , Calcio/metabolismo , Exocitosis/fisiología , Neurotransmisores/metabolismo , Fosfolípidos/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/metabolismo , Animales , Ratas
17.
Invest Ophthalmol Vis Sci ; 65(4): 32, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38648041

RESUMEN

Purpose: To undertake the first ultrastructural characterization of human retinal pigment epithelial (RPE) differentiation from fetal development to adolescence. Methods: Ten fetal eyes and three eyes aged six, nine, and 17 years were examined in the temporal retina adjacent to the optic nerve head by transmission electron microscopy. The area, number, and distribution of RPE organelles were quantified and interpreted within the context of adjacent photoreceptors, Bruch's membrane, and choriocapillaris maturation. Results: Between eight to 12 weeks' gestation (WG), pseudostratified columnar epithelia with apical tight junctions differentiate to a simple cuboidal epithelium with random distribution of melanosomes and mitochondria. Between 12 to 26 WG, cells enlarge and show long apical microvilli and apicolateral junctional complexes. Coinciding with eye opening at 26 WG, melanosomes migrate apically whereas mitochondria distribute to perinuclear regions, with the first appearance of phagosomes, complex granules, and basolateral extracellular space (BES) formation. Significantly, autophagy and heterophagy, as evidenced by organelle recycling, and the gold standard of ultrastructural evidence for autophagy of double-membrane autophagosomes and mitophagosomes were evident from 32 WG, followed by basal infoldings of RPE cell membrane at 36 WG. Lipofuscin formation and deposition into the BES evident at six years increased at 17 years. Conclusions: We provide compelling ultrastructural evidence that heterophagy and autophagy begins in the third trimester of human fetal development and that deposition of cellular byproducts into the extracellular space of RPE takes place via exocytosis. Transplanted RPE cells must also demonstrate the capacity to subserve autophagic and heterophagic functions for effective disease mitigation.


Asunto(s)
Autofagia , Exocitosis , Lipofuscina , Microscopía Electrónica de Transmisión , Epitelio Pigmentado de la Retina , Humanos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Epitelio Pigmentado de la Retina/embriología , Adolescente , Autofagia/fisiología , Niño , Lipofuscina/metabolismo , Exocitosis/fisiología , Espacio Extracelular/metabolismo , Edad Gestacional , Femenino , Masculino , Desarrollo Fetal/fisiología , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Diferenciación Celular/fisiología
18.
Plant J ; 118(5): 1475-1485, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38402593

RESUMEN

Plant cell walls are essential for defining plant growth and development, providing structural support to the main body and responding to abiotic and biotic cues. Cellulose, the main structural polymer of plant cell walls, is synthesized at the plasma membrane by cellulose synthase complexes (CSCs). The construction and transport of CSCs to and from the plasma membrane is poorly understood but is known to rely on the coordinated activity of cellulose synthase-interactive protein 1 (CSI1), a key regulator of CSC trafficking. In this study, we found that Trs85, a TRAPPIII complex subunit, interacted with CSI1 in vitro. Using functional genetics and live-cell imaging, we have shown that trs85-1 mutants have reduced cellulose content, stimulated CSC delivery, an increased population of static CSCs and deficient clathrin-mediated endocytosis in the primary cell wall. Overall, our findings suggest that Trs85 has a dual role in the trafficking of CSCs, by negatively regulating the exocytosis and clathrin-mediated endocytosis of CSCs.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Pared Celular , Celulosa , Endocitosis , Glucosiltransferasas , Transporte de Proteínas , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/enzimología , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Glucosiltransferasas/metabolismo , Glucosiltransferasas/genética , Pared Celular/metabolismo , Endocitosis/fisiología , Celulosa/metabolismo , Clatrina/metabolismo , Membrana Celular/metabolismo , Exocitosis/fisiología , Mutación , Proteínas Portadoras
19.
ACS Chem Neurosci ; 15(4): 816-826, 2024 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-38344810

RESUMEN

The two essential fatty acids, alpha-linolenic acid and linoleic acid, and the higher unsaturated fatty acids synthesized from them are critical for the development and maintenance of normal brain functions. Deficiencies of these fatty acids have been shown to cause damage to the neuronal development, cognition, and locomotor function. We combined electrochemistry and imaging techniques to examine the effects of the two essential fatty acids on catecholamine release dynamics and the vesicle content as well as on the cell membrane phospholipid composition to understand how they impact exocytosis and by extension neurotransmission at the single-cell level. Incubation of either of the two fatty acids reduces the size of secretory vesicles and enables the incorporation of more double bonds into the cell membrane structure, resulting in higher membrane flexibility. This subsequently affects proteins regulating the dynamics of the exocytotic fusion pore and thereby affects exocytosis. Our data suggest a possible pathway whereby the two essential fatty acids affect the membrane structure to impact exocytosis and provide a potential treatment for diseases and impairments related to catecholamine signaling.


Asunto(s)
Catecolaminas , Lípidos de la Membrana , Catecolaminas/metabolismo , Ácidos Grasos Insaturados , Ácidos Grasos Esenciales/farmacología , Exocitosis/fisiología
20.
J Cell Sci ; 137(6)2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38348894

RESUMEN

Dense core vesicles (DCVs) and synaptic vesicles are specialised secretory vesicles in neurons and neuroendocrine cells, and abnormal release of their cargo is associated with various pathophysiologies. Endoplasmic reticulum (ER) stress and inter-organellar communication are also associated with disease biology. To investigate the functional status of regulated exocytosis arising from the crosstalk of a stressed ER and DCVs, ER stress was modelled in PC12 neuroendocrine cells using thapsigargin. DCV exocytosis was severely compromised in ER-stressed PC12 cells and was reversed to varying magnitudes by ER stress attenuators. Experiments with tunicamycin, an independent ER stressor, yielded similar results. Concurrently, ER stress also caused impaired DCV exocytosis in insulin-secreting INS-1 cells. Molecular analysis revealed blunted SNAP25 expression, potentially attributed to augmented levels of ATF4, an inhibitor of CREB that binds to the CREB-binding site. The effects of loss of function of ATF4 in ER-stressed cells substantiated this attribution. Our studies revealed severe defects in DCV exocytosis in ER-stressed cells for the first time, mediated by reduced levels of key exocytotic and granulogenic switches regulated via the eIF2α (EIF2A)-ATF4 axis.


Asunto(s)
Neuronas , Vesículas Sinápticas , Ratas , Animales , Neuronas/metabolismo , Vesículas Sinápticas/metabolismo , Exocitosis/fisiología , Vesículas Secretoras/metabolismo , Estrés del Retículo Endoplásmico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA