Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
iScience ; 26(6): 106826, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37250768

RESUMEN

Synaptic vesicle (SV) clusters, which reportedly result from synapsin's capacity to undergo liquid-liquid phase separation (LLPS), constitute the structural basis for neurotransmission. Although these clusters contain various endocytic accessory proteins, how endocytic proteins accumulate in SV clusters remains unknown. Here, we report that endophilin A1 (EndoA1), the endocytic scaffold protein, undergoes LLPS under physiologically relevant concentrations at presynaptic terminals. On heterologous expression, EndoA1 facilitates the formation of synapsin condensates and accumulates in SV-like vesicle clusters via synapsin. Moreover, EndoA1 condensates recruit endocytic proteins such as dynamin 1, amphiphysin, and intersectin 1, none of which are recruited in vesicle clusters by synapsin. In cultured neurons, like synapsin, EndoA1 is compartmentalized in SV clusters through LLPS, exhibiting activity-dependent dispersion/reassembly cycles. Thus, beyond its essential function in SV endocytosis, EndoA1 serves an additional structural function by undergoing LLPS, thereby accumulating various endocytic proteins in dynamic SV clusters in concert with synapsin.

2.
Methods Mol Biol ; 2417: 45-58, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35099790

RESUMEN

Newly generated synaptic vesicles (SVs) are re-acidified by the activity of the vacuolar-type H+-ATPases. Since H+ gradient across SV membrane drives neurotransmitter uptake into SVs, precise measurements of steady-state vesicular pH and dynamics of re-acidification process will provide important information concerning the H+-driven neurotransmitter uptake. Indeed, we recently demonstrated distinct features of steady state and dynamics of vesicular pH between glutamatergic vesicles and GABAergic vesicles in cultured hippocampal neurons. In this article, we focus on an experimental protocol and setup required to determine steady-state luminal pH of SVs in living neurons. This protocol is composed of efficient expression of a pH-sensitive fluorescent protein in the lumen of SVs in cultured neurons, and recordings of its fluorescence changes under a conventional fluorescent microscope during local applications of acidic buffer and ionophores-containing solution at a given pH. The method described here can be easily applied for measuring luminal pH of different types of secretory organelles and other acidic organelles such as lysosomes and endosomes in cultured cell preparations.


Asunto(s)
Vesículas Sinápticas , ATPasas de Translocación de Protón Vacuolares , Células Cultivadas , Hipocampo/metabolismo , Concentración de Iones de Hidrógeno , Neuronas/metabolismo , Vesículas Sinápticas/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
3.
Elife ; 112022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35014951

RESUMEN

Neurotransmission is based on the exocytic fusion of synaptic vesicles (SVs) followed by endocytic membrane retrieval and the reformation of SVs. Conflicting models have been proposed regarding the mechanisms of SV endocytosis, most notably clathrin/adaptor protein complex 2 (AP-2)-mediated endocytosis and clathrin-independent ultrafast endocytosis. Partitioning between these pathways has been suggested to be controlled by temperature and stimulus paradigm. We report on the comprehensive survey of six major SV proteins to show that SV endocytosis in mouse hippocampal neurons at physiological temperature occurs independent of clathrin while the endocytic retrieval of a subset of SV proteins including the vesicular transporters for glutamate and GABA depend on sorting by the clathrin adaptor AP-2. Our findings highlight a clathrin-independent role of the clathrin adaptor AP-2 in the endocytic retrieval of select SV cargos from the presynaptic cell surface and suggest a revised model for the endocytosis of SV membranes at mammalian central synapses.


Asunto(s)
Complejo 2 de Proteína Adaptadora/genética , Clatrina/metabolismo , Endocitosis , Sinapsis/fisiología , Complejo 2 de Proteína Adaptadora/metabolismo , Animales , Ratones
4.
Commun Biol ; 4(1): 981, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408265

RESUMEN

Upon the arrival of repetitive stimulation at the presynaptic terminals of neurons, replenishment of readily releasable synaptic vesicles (SVs) with vesicles in the recycling pool is important for sustained neurotransmitter release. Kinetics of replenishment and the available pool size define synaptic performance. However, whether all SVs in the recycling pool are recruited for release with equal probability and speed is unknown. Here, based on comprehensive optical imaging of various presynaptic endosomal SNARE proteins in cultured hippocampal neurons, all of which are implicated in organellar membrane fusion in non-neuronal cells, we show that part of the recycling pool bearing the endosomal Q-SNARE, syntaxin 7 (Stx7), is preferentially mobilized for release during high-frequency repetitive stimulation. Recruitment of the SV pool marked with an Stx7-reporter requires actin polymerization, as well as activation of the Ca2+/calmodulin signaling pathway, reminiscent of rapidly replenishing SVs characterized previously in calyx of Held synapses. Furthermore, disruption of Stx7 function by overexpressing its N-terminal domain selectively abolished this pool. Thus, our data indicate that endosomal membrane fusion involving Stx7 forms rapidly replenishing vesicles essential for synaptic responses to high-frequency repetitive stimulation, and also highlight functional diversities of endosomal SNAREs in generating distinct exocytic vesicles in the presynaptic terminals.


Asunto(s)
Endosomas/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Proteínas Qa-SNARE/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Ratones , Ratones Endogámicos ICR
5.
Front Cell Neurosci ; 15: 811892, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095427

RESUMEN

Accumulation of glutamate, the primary excitatory neurotransmitter in the mammalian central nervous system, into presynaptic synaptic vesicles (SVs) depends upon three vesicular glutamate transporters (VGLUTs). Since VGLUTs are driven by a proton electrochemical gradient across the SV membrane generated by vacuolar-type H+-ATPases (V-ATPases), the rate of glutamate transport into SVs, as well as the amount of glutamate in SVs at equilibrium, are influenced by activities of both VGLUTs and V-ATPase. Despite emerging evidence that suggests various factors influencing glutamate transport by VGLUTs in vitro, little has been reported in physiological or pathological contexts to date. Historically, this was partially due to a lack of appropriate methods to monitor glutamate loading into SVs in living synapses. Furthermore, whether or not glutamate refilling of SVs can be rate-limiting for synaptic transmission is not well understood, primarily due to a lack of knowledge concerning the time required for vesicle reuse and refilling during repetitive stimulation. In this review, we first introduce a unique electrophysiological method to monitor glutamate refilling by VGLUTs in a giant model synapse from the calyx of Held in rodent brainstem slices, and we discuss the advantages and limitations of the method. We then introduce the current understanding of factors that potentially alter the amount and rate of glutamate refilling of SVs in this synapse, and discuss open questions from physiological viewpoints.

6.
Proc Natl Acad Sci U S A ; 117(52): 33586-33596, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33376223

RESUMEN

Current proteomic studies clarified canonical synaptic proteins that are common to many types of synapses. However, proteins of diversified functions in a subset of synapses are largely hidden because of their low abundance or structural similarities to abundant proteins. To overcome this limitation, we have developed an "ultra-definition" (UD) subcellular proteomic workflow. Using purified synaptic vesicle (SV) fraction from rat brain, we identified 1,466 proteins, three times more than reported previously. This refined proteome includes all canonical SV proteins, as well as numerous proteins of low abundance, many of which were hitherto undetected. Comparison of UD quantifications between SV and synaptosomal fractions has enabled us to distinguish SV-resident proteins from potential SV-visitor proteins. We found 134 SV residents, of which 86 are present in an average copy number per SV of less than one, including vesicular transporters of nonubiquitous neurotransmitters in the brain. We provide a fully annotated resource of all categorized SV-resident and potential SV-visitor proteins, which can be utilized to drive novel functional studies, as we characterized here Aak1 as a regulator of synaptic transmission. Moreover, proteins in the SV fraction are associated with more than 200 distinct brain diseases. Remarkably, a majority of these proteins was found in the low-abundance proteome range, highlighting its pathological significance. Our deep SV proteome will provide a fundamental resource for a variety of future investigations on the function of synapses in health and disease.


Asunto(s)
Encéfalo/metabolismo , Mamíferos/metabolismo , Proteoma/metabolismo , Vesículas Sinápticas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Péptidos/metabolismo , Proteoma/química , Proteómica , Ratas Sprague-Dawley , Transmisión Sináptica , Vesículas Sinápticas/ultraestructura , Sinaptosomas/metabolismo
7.
Cell Rep ; 32(7): 108040, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814044

RESUMEN

Recycling of synaptic vesicles (SVs) at presynaptic terminals is required for sustained neurotransmitter release. Although SV endocytosis is a rate-limiting step for synaptic transmission, it is unclear whether the rate of the subsequent SV refilling with neurotransmitter also influences synaptic transmission. By analyzing vesicular glutamate transporter 1 (VGLUT1)-deficient calyx of Held synapses, in which both VGLUT1 and VGLUT2 are co-expressed in wild-type situation, we found that VGLUT1 loss causes a drastic reduction in SV refilling rate down to ∼25% of wild-type values, with only subtle changes in basic synaptic parameters. Strikingly, VGLUT1-deficient synapses exhibited abnormal synaptic failures within a few seconds during high-frequency repetitive firing, which was recapitulated by manipulating presynaptic Cl- concentrations to retard SV refilling. Our data show that the speed of SV refilling can be rate limiting for synaptic transmission under certain conditions that entail reduced VGLUT levels during development as well as various neuropathological processes.


Asunto(s)
Sinapsis/metabolismo , Transmisión Sináptica/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Animales , Humanos , Ratones
8.
Sci Rep ; 9(1): 4289, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862855

RESUMEN

Ca2+ transport into synaptic vesicles (SVs) at the presynaptic terminals has been proposed to be an important process for regulating presynaptic [Ca2+] during stimulation as well as at rest. However, the molecular identity of the transport system remains elusive. Previous studies have demonstrated that isolated SVs exhibit two distinct Ca2+ transport systems depending on extra-vesicular (cytosolic) pH; one is mediated by a high affinity Ca2+ transporter which is active at neutral pH and the other is mediated by a low affinity Ca2+/H+ antiporter which is maximally active at alkaline pH of 8.5. In addition, synaptic vesicle glycoprotein 2 s (SV2s), a major SV component, have been proposed to contribute to Ca2+ clearance from the presynaptic cytoplasm. Here, we show that at physiological pH, the plasma membrane Ca2+ ATPases (PMCAs) are responsible for both the Ca2+/H+ exchange activity and Ca2+ uptake into SVs. The Ca2+/H+ exchange activity monitored by acidification assay exhibited high affinity for Ca2+ (Km ~ 400 nM) and characteristic divalent cation selectivity for the PMCAs. Both activities were remarkably reduced by PMCA blockers, but not by a blocker of the ATPase that transfers Ca2+ from the cytosol to the lumen of sarcoplasmic endoplasmic reticulum (SERCA) at physiological pH. Furthermore, we rule out the contribution of SV2s, putative Ca2+ transporters on SVs, since both Ca2+/H+ exchange activity and Ca2+ transport were unaffected in isolated vesicles derived from SV2-deficient brains. Finally, using a PMCA1-pHluorin construct that enabled us to monitor cellular distribution and recycling properties in living neurons, we demonstrated that PMCA1-pHluorin localized to intracellular acidic compartments and recycled at presynaptic terminals in an activity-dependent manner. Collectively, our results imply that vesicular PMCAs may play pivotal roles in both presynaptic Ca2+ homeostasis and the modulation of H+ gradient in SVs.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Membrana Celular/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Animales , Transporte Biológico/fisiología , Calcio/metabolismo , Citosol/metabolismo , Femenino , Concentración de Iones de Hidrógeno , Ratones Endogámicos C57BL , Ratones Noqueados , Vesículas Sinápticas/metabolismo
9.
Sci Rep ; 8(1): 15156, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30310105

RESUMEN

Targeting gene expression to a particular subset of neurons helps study the cellular function of the nervous system. Although neuron-specific promoters, such as the synapsin I promoter and the α-CaMKII promoter, are known to exhibit selectivity for excitatory glutamatergic neurons in vivo, the cell type-specificity of these promoters has not been thoroughly tested in culture preparations. Here, by using hippocampal culture preparation from the VGAT-Venus transgenic mice, we examined the ability of five putative promoter sequences of glutamatergic-selective markers including synapsin I, α-CaMKII, the vesicular glutamate transporter 1 (VGLUT1), Dock10 and Prox1. Among these, a genomic fragment containing a 2.1 kb segment upstream of the translation start site (TSS) of the VGLUT1 implemented in a lentiviral vector with the Tet-Off inducible system achieved the highest preferential gene expression in glutamatergic neurons. Analysis of various lengths of the VGLUT1 promoter regions identified a segment between -2.1 kb and -1.4 kb from the TSS as a responsible element for the glutamatergic selectivity. Consistently, expression of channelrhodopsin under this promoter sequence allowed for selective light-evoked activation of excitatory neurons. Thus, the lentiviral system carrying the VGLUT1 promoter fragment can be used to effectively target exogenous gene expression to excitatory glutamatergic neurons in cultures.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Lentivirus/genética , Células Piramidales/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/genética , Potenciales de Acción , Animales , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Orden Génico , Ratones , Ratones Transgénicos , Neuroglía/metabolismo , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Transgenes
10.
Front Cell Neurosci ; 11: 422, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29379416

RESUMEN

Efficient retrieval of the synaptic vesicle (SV) membrane from the presynaptic plasma membrane, a process called endocytosis, is crucial for the fidelity of neurotransmission, particularly during sustained neural activity. Although multiple modes of endocytosis have been identified, it is clear that the efficient retrieval of the major SV cargos into newly formed SVs during any of these modes is fundamental for synaptic transmission. It is currently believed that SVs are eventually reformed via a clathrin-dependent pathway. Various adaptor proteins recognize SV cargos and link them to clathrin, ensuring the efficient retrieval of the cargos into newly formed SVs. Here, we summarize our current knowledge of the molecular signatures within individual SV cargos that underlie efficient retrieval into SV membranes, as well as discuss possible contributions of the mechanisms under physiological conditions.

11.
Proc Natl Acad Sci U S A ; 113(38): 10702-7, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27601664

RESUMEN

GABA acts as the major inhibitory neurotransmitter in the mammalian brain, shaping neuronal and circuit activity. For sustained synaptic transmission, synaptic vesicles (SVs) are required to be recycled and refilled with neurotransmitters using an H(+) electrochemical gradient. However, neither the mechanism underlying vesicular GABA uptake nor the kinetics of GABA loading in living neurons have been fully elucidated. To characterize the process of GABA uptake into SVs in functional synapses, we monitored luminal pH of GABAergic SVs separately from that of excitatory glutamatergic SVs in cultured hippocampal neurons. By using a pH sensor optimal for the SV lumen, we found that GABAergic SVs exhibited an unexpectedly higher resting pH (∼6.4) than glutamatergic SVs (pH ∼5.8). Moreover, unlike glutamatergic SVs, GABAergic SVs displayed unique pH dynamics after endocytosis that involved initial overacidification and subsequent alkalization that restored their resting pH. GABAergic SVs that lacked the vesicular GABA transporter (VGAT) did not show the pH overshoot and acidified further to ∼6.0. Comparison of luminal pH dynamics in the presence or absence of VGAT showed that VGAT operates as a GABA/H(+) exchanger, which is continuously required to offset GABA leakage. Furthermore, the kinetics of GABA transport was slower (τ > 20 s at physiological temperature) than that of glutamate uptake and may exceed the time required for reuse of exocytosed SVs, allowing reuse of incompletely filled vesicles in the presence of high demand for inhibitory transmission.


Asunto(s)
Neuronas/metabolismo , Neurotransmisores/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Exocitosis/genética , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Concentración de Iones de Hidrógeno , Cinética , Ratones , Neuronas/fisiología , Neurotransmisores/genética , Terminales Presinápticos/metabolismo , Terminales Presinápticos/fisiología , Sinapsis/genética , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética
12.
Artículo en Inglés | MEDLINE | ID: mdl-26903855

RESUMEN

The quantal hypothesis for the release of neurotransmitters at the chemical synapse has gained wide acceptance since it was first worked out at the motor endplate in frog skeletal muscle in the 1950's. Considering the morphological identification of synaptic vesicles (SVs) at the nerve terminals that appeared to be homogeneous in size, the hypothesis proposed that signal transduction at synapses is mediated by the release of neurotransmitters packed in SVs that are individually uniform in size; the amount of transmitter in a synaptic vesicle is called a quantum. Although quantal size-the amplitude of the postsynaptic response elicited by the release of neurotransmitters from a single vesicle-clearly depends on the number and sensitivity of the postsynaptic receptors, accumulating evidence has also indicated that the amount of neurotransmitters stored in SVs can be altered by various presynaptic factors. Here, I provide an overview of the concepts and underlying presynaptic molecular underpinnings that may regulate quantal size.

13.
Pflugers Arch ; 468(3): 513-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26577586

RESUMEN

Since the discovery of a putative Cl(-) channel on synaptic vesicle (SV) membranes, attempts to establish its molecular identity have proven surprisingly difficult. Recent evidence has emerged to support the idea that the vesicular glutamate transporter (VGLUT), whose main function is to concentrate the excitatory neurotransmitter glutamate into SVs, can also act as the Cl(-) permeation pathway. Here, I summarize studies investigating the putative Cl(-) channel on SVs and discuss the possible roles of VGLUT-mediated Cl(-) transport on glutamate loading.


Asunto(s)
Canales de Cloruro/metabolismo , Vesículas Sinápticas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Animales , Cloruros/metabolismo , Humanos , Transporte Iónico
14.
Cell Rep ; 12(11): 1887-901, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26344767

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder caused by MECP2 mutations. Although emerging evidence suggests that MeCP2 deficiency is associated with dysregulation of mechanistic target of rapamycin (mTOR), which functions as a hub for various signaling pathways, the mechanism underlying this association and the molecular pathophysiology of RTT remain elusive. We show here that MeCP2 promotes the posttranscriptional processing of particular microRNAs (miRNAs) as a component of the microprocessor Drosha complex. Among the MeCP2-regulated miRNAs, we found that miR-199a positively controls mTOR signaling by targeting inhibitors for mTOR signaling. miR-199a and its targets have opposite effects on mTOR activity, ameliorating and inducing RTT neuronal phenotypes, respectively. Furthermore, genetic deletion of miR-199a-2 led to a reduction of mTOR activity in the brain and recapitulated numerous RTT phenotypes in mice. Together, these findings establish miR-199a as a critical downstream target of MeCP2 in RTT pathogenesis by linking MeCP2 with mTOR signaling.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/metabolismo , MicroARNs/metabolismo , Síndrome de Rett/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Modelos Animales de Enfermedad , Proteína 2 de Unión a Metil-CpG/antagonistas & inhibidores , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Fenotipo , Síndrome de Rett/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Regulación hacia Arriba
15.
J Neurosci ; 35(8): 3701-10, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716867

RESUMEN

During synaptic vesicle (SV) recycling, the vacuolar-type H(+) ATPase creates a proton electrochemical gradient (ΔµH(+)) that drives neurotransmitter loading into SVs. Given the low estimates of free luminal protons, it has been envisioned that the influx of a limited number of protons suffices to establish ΔµH(+). Consistent with this, the time constant of SV re-acidification was reported to be <5 s, much faster than glutamate loading (τ of ∼ 15 s) and thus unlikely to be rate limiting for neurotransmitter loading. However, such estimates have relied on pHluorin-based probes that lack sensitivity in the lower luminal pH range. Here, we reexamined re-acidification kinetics using the mOrange2-based probe that should report the SV pH more accurately. In recordings from cultured mouse hippocampal neurons, we found that re-acidification took substantially longer (τ of ∼ 15 s) than estimated previously. In addition, we found that the SV lumen exhibited a large buffering capacity (∼ 57 mm/pH), corresponding to an accumulation of ∼ 1200 protons during re-acidification. Together, our results uncover hitherto unrecognized robust proton influx and storage in SVs that can restrict the rate of neurotransmitter refilling.


Asunto(s)
Protones , Vesículas Sinápticas/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Células Cultivadas , Femenino , Ácido Glutámico/metabolismo , Concentración de Iones de Hidrógeno , Transporte Iónico , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR
16.
Neurosci Lett ; 569: 142-7, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24708928

RESUMEN

Valproic acid (VPA) has been used to treat epileptic patients because of its ability to potentiate GABA signaling in the brain. Despite its clinical significance, VPA administration during pregnancy increases the risk of congenital abnormalities, such as neural tube defects and neurodevelopmental disorders including autism. Furthermore, recent studies revealed that early postnatal administration of VPA also leads to neurodevelopmental deficits in rodents. Here, using cultured cortical neurons derived from postnatal day 1 rats, we examined whether exposure to VPA would affect synapse formation. When neurons were exposed to 1mM VPA during early development, expression of the vesicular GABA transporter (VGAT) was selectively reduced, whereas other synaptic markers, including the vesicular glutamate transporters 1 and 2 (VGLUT1 and 2), were not affected. This VPA effect was mediated through inhibition of histone deacetylases (HDACs), since the effects were mostly recapitulated by an HDAC inhibitor, trichostatin A, but not by a VPA derivative, valpromide, which lacks HDAC inhibitor activity. Immunocytochemical analysis demonstrated that VPA exposure resulted in a retardation of axonal growth specific to GABAergic neurons and a decrease in VGAT-positive synapses. Since disturbance of the excitatory and inhibitory (E-I) balance has been implicated as a potential cause of multiple psychiatric disorders, our results may account for one of the cellular mechanisms underlying the pathogenesis of VPA-induced neurodevelopmental impairments.


Asunto(s)
Anticonvulsivantes/farmacología , Neuronas/efectos de los fármacos , Sinapsis/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Axones/fisiología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Inhibidores de Histona Desacetilasas/farmacología , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Sinapsis/fisiología , Factores de Tiempo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Ácido gamma-Aminobutírico/metabolismo
18.
Mol Brain ; 3: 40, 2010 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21190592

RESUMEN

BACKGROUND: The vesicular GABA transporter (VGAT) loads GABA and glycine from the neuronal cytoplasm into synaptic vesicles. To address functional importance of VGAT during embryonic development, we generated global VGAT knockout mice and analyzed them. RESULTS: VGAT knockouts at embryonic day (E) 18.5 exhibited substantial increases in overall GABA and glycine, but not glutamate, contents in the forebrain. Electrophysiological recordings from E17.5-18.5 spinal cord motoneurons demonstrated that VGAT knockouts presented no spontaneous inhibitory postsynaptic currents mediated by GABA and glycine. Histological examination of E18.5 knockout fetuses revealed reductions in the trapezius muscle, hepatic congestion and little alveolar spaces in the lung, indicating that the development of skeletal muscle, liver and lung in these mice was severely affected. CONCLUSION: VGAT is fundamental for the GABA- and/or glycine-mediated transmission that supports embryonic development. VGAT knockout mice will be useful for further investigating the roles of VGAT in normal physiology and pathophysiologic processes.


Asunto(s)
Desarrollo Embrionario , Ratones Noqueados , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Animales , Fisura del Paladar/genética , Femenino , Genotipo , Glutamato Descarboxilasa/genética , Ácido Glutámico/metabolismo , Glicina/metabolismo , Hernia Umbilical/genética , Hígado/citología , Hígado/metabolismo , Hígado/patología , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Técnicas de Placa-Clamp , Embarazo , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo
19.
Nat Neurosci ; 12(2): 156-62, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19169251

RESUMEN

Uptake of glutamate into synaptic vesicles is mediated by vesicular glutamate transporters (VGLUTs). Although glutamate uptake has been shown to depend critically on Cl(-), the precise contribution of this ion to the transport process is unclear. We found that VGLUT1, and not ClC-3 as proposed previously, represents the major Cl(-) permeation pathway in synaptic vesicles. Using reconstituted VGLUT1, we found that the biphasic dependence of glutamate transport on extravesicular Cl(-) is a result of the permeation of this anion through VGLUT1 itself. Moreover, we observed that high luminal Cl(-) concentrations markedly enhanced loading of glutamate by facilitation of membrane potential-driven uptake and discovered a hitherto unrecognized transport mode of VGLUT1. Because a steep Cl(-) gradient across the synaptic vesicle membrane exists in endocytosed synaptic vesicles, our results imply that the transport velocity and the final glutamate content are highly influenced, if not determined, by the extracellular Cl(-) concentration.


Asunto(s)
Cloruros/metabolismo , Ácido Glutámico/metabolismo , Activación del Canal Iónico/fisiología , Neuronas/fisiología , Vesículas Sinápticas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Ácidos/metabolismo , Animales , Encéfalo/citología , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Endocitosis/fisiología , Liposomas/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Proteína 1 de Transporte Vesicular de Glutamato/genética
20.
J Neurosci ; 28(49): 13125-31, 2008 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-19052203

RESUMEN

Neurotransmitter uptake into synaptic vesicles is mediated by vesicular neurotransmitter transporters. Although these transporters belong to different families, they all are thought to share a common overall topology with an even number of transmembrane domains. Using epitope-specific antibodies and mass spectrometry we show that the vesicular GABA transporter (VGAT) possesses an uneven number of transmembrane domains, with the N terminus facing the cytoplasm and the C terminus residing in the synaptic vesicle lumen. Antibodies recognizing the C terminus of VGAT (anti-VGAT-C) selectively label GABAergic nerve terminals of live cultured hippocampal and striatal neurons as confirmed by immunocytochemistry and patch-clamp electrophysiology. Injection of fluorochromated anti-VGAT-C into the hippocampus of mice results in specific labeling of GABAergic synapses in vivo. Overall, our data open the possibility of studying novel GABA release sites, characterizing inhibitory vesicle trafficking, and establishing their contribution to inhibitory neurotransmission at identified GABAergic synapses.


Asunto(s)
Inmunohistoquímica/métodos , Prosencéfalo/metabolismo , Coloración y Etiquetado/métodos , Sinapsis/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/química , Ácido gamma-Aminobutírico/metabolismo , Animales , Especificidad de Anticuerpos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/ultraestructura , Endocitosis/fisiología , Exocitosis/fisiología , Hipocampo/metabolismo , Hipocampo/ultraestructura , Espectrometría de Masas , Ratones , Inhibición Neural/fisiología , Técnicas de Placa-Clamp , Prosencéfalo/ultraestructura , Estructura Terciaria de Proteína/fisiología , Sinapsis/ultraestructura , Membranas Sinápticas/metabolismo , Membranas Sinápticas/ultraestructura , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestructura , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/inmunología , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...