Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Int J Mol Sci ; 24(18)2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37762560

RESUMO

Signals of nerve impulses are transmitted to excitatory cells to induce the action of organs via the activation of Ca2+ entry through voltage-gated Ca2+ channels (VGCC), which are classified based on their activation threshold into high- and low-voltage activated channels, expressed specifically for each organ [...].


Assuntos
Canais de Cálcio , Proteínas de Ligação ao Cálcio , Potenciais de Ação
2.
Int J Mol Sci ; 22(21)2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34769208

RESUMO

An action potential triggers neurotransmitter release from synaptic vesicles docking to a specialized release site of the presynaptic plasma membrane, the active zone. The active zone is a highly organized structure with proteins that serves as a platform for synaptic vesicle exocytosis, mediated by SNAREs complex and Ca2+ sensor proteins, within a sub-millisecond opening of nearby Ca2+ channels with the membrane depolarization. In response to incoming neuronal signals, each active zone protein plays a role in the release-ready site replenishment with synaptic vesicles for sustainable synaptic transmission. The active zone release apparatus provides a possible link between neuronal activity and plasticity. This review summarizes the mostly physiological role of active zone protein interactions that control synaptic strength, presynaptic short-term plasticity, and homeostatic synaptic plasticity.


Assuntos
Canais de Cálcio/metabolismo , Terminações Pré-Sinápticas/metabolismo , Transmissão Sináptica , Potenciais de Ação , Animais , Humanos , Plasticidade Neuronal , Neurotransmissores/metabolismo , Mapas de Interação de Proteínas , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo
3.
Int J Mol Sci ; 22(1)2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33396919

RESUMO

An action potential (AP) triggers neurotransmitter release from synaptic vesicles (SVs) docking to a specialized release site of presynaptic plasma membrane, the active zone (AZ). The AP simultaneously controls the release site replenishment with SV for sustainable synaptic transmission in response to incoming neuronal signals. Although many studies have suggested that the replenishment time is relatively slow, recent studies exploring high speed resolution have revealed SV dynamics with milliseconds timescale after an AP. Accurate regulation is conferred by proteins sensing Ca2+ entering through voltage-gated Ca2+ channels opened by an AP. This review summarizes how millisecond Ca2+ dynamics activate multiple protein cascades for control of the release site replenishment with release-ready SVs that underlie presynaptic short-term plasticity.


Assuntos
Plasticidade Neuronal , Neurotransmissores/metabolismo , Terminações Pré-Sinápticas/metabolismo , Transmissão Sináptica , Animais , Humanos
4.
Int J Mol Sci ; 20(9)2019 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-31064106

RESUMO

Presynaptic Ca2+ entry occurs through voltage-gated Ca2+ (CaV) channels which are activated by membrane depolarization. Depolarization accompanies neuronal firing and elevation of Ca2+ triggers neurotransmitter release from synaptic vesicles. For synchronization of efficient neurotransmitter release, synaptic vesicles are targeted by presynaptic Ca2+ channels forming a large signaling complex in the active zone. The presynaptic CaV2 channel gene family (comprising CaV2.1, CaV2.2, and CaV2.3 isoforms) encode the pore-forming α1 subunit. The cytoplasmic regions are responsible for channel modulation by interacting with regulatory proteins. This article overviews modulation of the activity of CaV2.1 and CaV2.2 channels in the control of synaptic strength and presynaptic plasticity.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Terminações Pré-Sinápticas/metabolismo , Animais , Canais de Cálcio Tipo N/genética , Proteínas de Ligação ao Cálcio/metabolismo , Humanos , Terminações Pré-Sinápticas/fisiologia , Potenciais Sinápticos
5.
Proc Jpn Acad Ser B Phys Biol Sci ; 93(10): 802-820, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29225307

RESUMO

For reliable transmission at chemical synapses, neurotransmitters must be released dynamically in response to neuronal activity in the form of action potentials. Stable synaptic transmission is dependent on the efficacy of transmitter release and the rate of resupplying synaptic vesicles to their release sites. Accurate regulation is conferred by proteins sensing Ca2+ entering through voltage-gated Ca2+ channels opened by an action potential. Presynaptic Ca2+ concentration changes are dynamic functions in space and time, with wide fluctuations associated with different rates of neuronal activity. Thus, regulation of transmitter release includes reactions involving multiple Ca2+-dependent proteins, each operating over a specific time window. Classically, studies of presynaptic proteins function favored large invertebrate presynaptic terminals. I have established a useful mammalian synapse model based on sympathetic neurons in culture. This review summarizes the use of this model synapse to study the roles of presynaptic proteins in neuronal activity for the control of transmitter release efficacy and synaptic vesicle recycling.


Assuntos
Neurotransmissores/metabolismo , Proteínas/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Humanos , Neurônios/metabolismo , Terminações Pré-Sinápticas/metabolismo , Sinapses/metabolismo , Transmissão Sináptica
6.
J Neurosci ; 35(23): 8901-13, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26063922

RESUMO

Presynaptic nerve terminals must maintain stable neurotransmissions via synaptic vesicle (SV) resupply despite encountering wide fluctuations in the number and frequency of incoming action potentials (APs). However, the molecular mechanism linking variation in neural activity to SV resupply is unknown. Myosins II and VI are actin-based cytoskeletal motors that drive dendritic actin dynamics and membrane transport, respectively, at brain synapses. Here we combined genetic knockdown or molecular dysfunction and direct physiological measurement of fast synaptic transmission from paired rat superior cervical ganglion neurons in culture to show that myosins IIB and VI work individually in SV reuse pathways, having distinct dependency and time constants with physiological AP frequency. Myosin VI resupplied the readily releasable pool (RRP) with slow kinetics independently of firing rates but acted quickly within 50 ms after AP. Under high-frequency AP firing, myosin IIB resupplied the RRP with fast kinetics in a slower time window of 200 ms. Knockdown of both myosin and dynamin isoforms by mixed siRNA microinjection revealed that myosin IIB-mediated SV resupply follows amphiphysin/dynamin-1-mediated endocytosis, while myosin VI-mediated SV resupply follows dynamin-3-mediated endocytosis. Collectively, our findings show how distinct myosin isoforms work as vesicle motors in appropriate SV reuse pathways associated with specific firing patterns.


Assuntos
Dinamina I/metabolismo , Neurônios/fisiologia , Miosina não Muscular Tipo IIB/metabolismo , Vesículas Sinápticas/metabolismo , Miosinas Ventriculares/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Células Cultivadas , Dinamina I/genética , Estimulação Elétrica , Endocitose/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Gânglio Cervical Superior/citologia , Fatores de Tempo , Miosinas Ventriculares/genética , Miosinas Ventriculares/farmacologia
7.
J Neurosci ; 34(32): 10603-15, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25100594

RESUMO

α-Synuclein is thought to regulate neurotransmitter release through multiple interactions with presynaptic proteins, cytoskeletal elements, ion channels, and synaptic vesicles membrane. α-Synuclein is abundant in the presynaptic compartment, and its release from neurons and glia has been described as responsible for spreading of α-synuclein-derived pathology. α-Synuclein-dependent dysregulation of neurotransmitter release might occur via its action on surface-exposed calcium channels. Here, we provide electrophysiological and biochemical evidence to show that α-synuclein, applied to rat neurons in culture or striatal slices, selectively activates Cav2.2 channels, and said activation correlates with increased neurotransmitter release. Furthermore, in vivo perfusion of α-synuclein into the striatum also leads to acute dopamine release. We further demonstrate that α-synuclein reduces the amount of plasma membrane cholesterol and alters the partitioning of Cav2.2 channels, which move from raft to cholesterol-poor areas of the plasma membrane. We provide evidence for a novel mechanism through which α-synuclein acts from the extracellular milieu to modulate neurotransmitter release and propose a unifying hypothesis for the mechanism of α-synuclein action on multiple targets: the reorganization of plasma membrane microdomains.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Dopamina/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Neurônios/citologia , alfa-Sinucleína/farmacologia , Compostos de Anilina/metabolismo , Animais , Anticorpos/farmacologia , Canais de Cálcio Tipo N/imunologia , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Bloqueadores dos Canais de Sódio/farmacologia , Gânglio Cervical Superior/citologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Sinaptofisina/metabolismo , Xantenos/metabolismo
8.
Eur J Neurosci ; 41(4): 398-409, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25484265

RESUMO

Synaptic vesicle glycoprotein (SV)2A is a transmembrane protein found in secretory vesicles and is critical for Ca(2+) -dependent exocytosis in central neurons, although its mechanism of action remains uncertain. Previous studies have proposed, variously, a role of SV2 in the maintenance and formation of the readily releasable pool (RRP) or in the regulation of Ca(2+) responsiveness of primed vesicles. Such previous studies have typically used genetic approaches to ablate SV2 levels; here, we used a strategy involving small interference RNA (siRNA) injection to knockdown solely presynaptic SV2A levels in rat superior cervical ganglion (SCG) neuron synapses. Moreover, we investigated the effects of SV2A knockdown on voltage-dependent Ca(2+) channel (VDCC) function in SCG neurons. Thus, we extended the studies of SV2A mechanisms by investigating the effects on vesicular transmitter release and VDCC function in peripheral sympathetic neurons. We first demonstrated an siRNA-mediated SV2A knockdown. We showed that this SV2A knockdown markedly affected presynaptic function, causing an attenuated RRP size, increased paired-pulse depression and delayed RRP recovery after stimulus-dependent depletion. We further demonstrated that the SV2A-siRNA-mediated effects on vesicular release were accompanied by a reduction in VDCC current density in isolated SCG neurons. Together, our data showed that SV2A is required for correct transmitter release at sympathetic neurons. Mechanistically, we demonstrated that presynaptic SV2A: (i) acted to direct normal synaptic transmission by maintaining RRP size, (ii) had a facilitatory role in recovery from synaptic depression, and that (iii) SV2A deficits were associated with aberrant Ca(2+) current density, which may contribute to the secretory phenotype in sympathetic peripheral neurons.


Assuntos
Canais de Cálcio/metabolismo , Exocitose , Glicoproteínas de Membrana/metabolismo , Gânglio Cervical Superior/metabolismo , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Células Cultivadas , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/genética , Ratos , Ratos Wistar , Gânglio Cervical Superior/citologia , Sinapses/fisiologia , Transmissão Sináptica
9.
Proc Natl Acad Sci U S A ; 109(42): 17069-74, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-23027954

RESUMO

Modulation of P/Q-type Ca(2+) currents through presynaptic voltage-gated calcium channels (Ca(V)2.1) by binding of Ca(2+)/calmodulin contributes to short-term synaptic plasticity. Ca(2+)-binding protein-1 (CaBP1) and Visinin-like protein-2 (VILIP-2) are neurospecific calmodulin-like Ca(2+) sensor proteins that differentially modulate Ca(V)2.1 channels, but how they contribute to short-term synaptic plasticity is unknown. Here, we show that activity-dependent modulation of presynaptic Ca(V)2.1 channels by CaBP1 and VILIP-2 has opposing effects on short-term synaptic plasticity in superior cervical ganglion neurons. Expression of CaBP1, which blocks Ca(2+)-dependent facilitation of P/Q-type Ca(2+) current, markedly reduced facilitation of synaptic transmission. VILIP-2, which blocks Ca(2+)-dependent inactivation of P/Q-type Ca(2+) current, reduced synaptic depression and increased facilitation under conditions of high release probability. These results demonstrate that activity-dependent regulation of presynaptic Ca(V)2.1 channels by differentially expressed Ca(2+) sensor proteins can fine-tune synaptic responses to trains of action potentials and thereby contribute to the diversity of short-term synaptic plasticity.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Calmodulina/metabolismo , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Análise de Variância , Humanos , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/metabolismo
10.
Mol Pharmacol ; 86(3): 297-305, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24981043

RESUMO

Calcium regulation of neurotransmitter release is essential for maintenance of synaptic transmission. However, the temporal and spatial organization of Ca(2+) dynamics that regulate synaptic vesicle (SV) release efficacy in sympathetic neurons is poorly understood. Here, we investigate the N-type Ca(2+) channel-mediated kinetic structure of Ca(2+) regulation of cholinergic transmission of sympathetic neurons. We measured the effect of Ca(2+) chelation with fast 1,2-bis(2-aminophenoxy) ethane-tetraacetic acid (BAPTA) and slow ethyleneglycol-tetraacetic acid (EGTA) buffers on exocytosis, synaptic depression, and recovery of the readily releasable vesicle pool (RRP), after both single action potential (AP) and repetitive APs. Surprisingly, postsynaptic potentials peaking at ~12 milliseconds after the AP was inhibited by both rapid and slow Ca(2+) buffers suggests that, in addition to the well known fast Ca(2+) signals at the active zone (AZ), slow Ca(2+) signals at the peak of Ca(2+) entry also contribute to paired-pulse or repetitive AP responses. Following a single AP, discrete Ca(2+) transient increase regulated synaptic depression in rapid (<30-millisecond) and slow (<120-millisecond) phases. In contrast, following prolonged AP trains, synaptic depression was reduced by a slow Ca(2+) signal regulation lasting >200 milliseconds. Finally, after an AP burst, recovery of the RRP was mediated by an AP-dependent rapid Ca(2+) signal, and the expansion of releasable SV number by an AP firing activity-dependent slow Ca(2+) signal. These data indicate that local Ca(2+) signals operating near Ca(2+) sources in the AZ are organized into discrete fast and slow temporal phases that remodel exocytosis and short-term plasticity to ensure long-term stability in acetylcholine release efficacy.


Assuntos
Acetilcolina/metabolismo , Sinalização do Cálcio , Neurônios/metabolismo , Gânglio Cervical Superior/citologia , Vesículas Sinápticas/metabolismo , Potenciais de Ação , Animais , Células Cultivadas , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Plasticidade Neuronal , Ratos Wistar , Gânglio Cervical Superior/metabolismo , Transmissão Sináptica
11.
J Biol Chem ; 288(26): 19050-9, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23687302

RESUMO

Presynaptic nerve terminals must maintain stable neurotransmission via synaptic vesicle membrane recycling despite encountering wide fluctuations in the number and frequency of incoming action potentials (APs). However, the molecular mechanism linking variation in neuronal activity to vesicle trafficking is unknown. Here, we combined genetic knockdown and direct physiological measurements of synaptic transmission from paired neurons to show that three isoforms of dynamin, an essential endocytic protein, work individually to match vesicle reuse pathways, having distinct rate and time constants with physiological AP frequencies. Dynamin 3 resupplied the readily releasable pool with slow kinetics independently of the AP frequency but acted quickly, within 20 ms of the incoming AP. Under high-frequency firing, dynamin 1 regulated recycling to the readily releasable pool with fast kinetics in a slower time window of greater than 50 ms. Dynamin 2 displayed a hybrid response between the other isoforms. Collectively, our findings show how dynamin isoforms select appropriate vesicle reuse pathways associated with specific neuronal firing patterns.


Assuntos
Potenciais de Ação/fisiologia , Dinaminas/fisiologia , Vesículas Sinápticas/fisiologia , Animais , Eletrofisiologia , Endocitose , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Neurônios/fisiologia , Células PC12 , Terminações Pré-Sinápticas/fisiologia , Isoformas de Proteínas/fisiologia , Ratos , Ratos Wistar , Transmissão Sináptica/fisiologia
12.
J Biol Chem ; 288(7): 4637-48, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23255606

RESUMO

Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) forms a major component of the postsynaptic density where its functions in synaptic plasticity are well established, but its presynaptic actions are poorly defined. Here we show that CaMKII binds directly to the C-terminal domain of Ca(V)2.1 channels. Binding is enhanced by autophosphorylation, and the kinase-channel signaling complex persists after dephosphorylation and removal of the Ca(2+)/CaM stimulus. Autophosphorylated CaMKII can bind the Ca(V)2.1 channel and synapsin-1 simultaneously. CaMKII binding to Ca(V)2.1 channels induces Ca(2+)-independent activity of the kinase, which phosphorylates the enzyme itself as well as the neuronal substrate synapsin-1. Facilitation and inactivation of Ca(V)2.1 channels by binding of Ca(2+)/CaM mediates short term synaptic plasticity in transfected superior cervical ganglion neurons, and these regulatory effects are prevented by a competing peptide and the endogenous brain inhibitor CaMKIIN, which blocks binding of CaMKII to Ca(V)2.1 channels. These results define the functional properties of a signaling complex of CaMKII and Ca(V)2.1 channels in which both binding partners are persistently activated by their association, and they further suggest that this complex is important in presynaptic terminals in regulating protein phosphorylation and short term synaptic plasticity.


Assuntos
Canais de Cálcio Tipo N/química , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Regulação da Expressão Gênica , Eletrofisiologia/métodos , Humanos , Modelos Biológicos , Plasticidade Neuronal , Neurotransmissores/metabolismo , Fosforilação , Terminações Pré-Sinápticas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Transfecção
13.
Nature ; 446(7131): 41-5, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17287729

RESUMO

CD38, a transmembrane glycoprotein with ADP-ribosyl cyclase activity, catalyses the formation of Ca2+ signalling molecules, but its role in the neuroendocrine system is unknown. Here we show that adult CD38 knockout (CD38-/-) female and male mice show marked defects in maternal nurturing and social behaviour, respectively, with higher locomotor activity. Consistently, the plasma level of oxytocin (OT), but not vasopressin, was strongly decreased in CD38-/- mice. Replacement of OT by subcutaneous injection or lentiviral-vector-mediated delivery of human CD38 in the hypothalamus rescued social memory and maternal care in CD38-/- mice. Depolarization-induced OT secretion and Ca2+ elevation in oxytocinergic neurohypophysial axon terminals were disrupted in CD38-/- mice; this was mimicked by CD38 metabolite antagonists in CD38+/+ mice. These results reveal that CD38 has a key role in neuropeptide release, thereby critically regulating maternal and social behaviours, and may be an element in neurodevelopmental disorders.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Comportamento Materno/fisiologia , Ocitocina/metabolismo , Comportamento Social , ADP-Ribosil Ciclase 1/deficiência , ADP-Ribosil Ciclase 1/genética , Amnésia/genética , Amnésia/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Feminino , Regulação da Expressão Gênica , Humanos , Injeções , Masculino , Memória/fisiologia , Camundongos , Atividade Motora/fisiologia , Ocitocina/administração & dosagem , Ocitocina/sangue , Ocitocina/farmacologia , Vasopressinas/sangue
14.
Cells ; 12(5)2023 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-36899886

RESUMO

V-ATPase is an important factor in synaptic vesicle acidification and is implicated in synaptic transmission. Rotation in the extra-membranous V1 sector drives proton transfer through the membrane-embedded multi-subunit V0 sector of the V-ATPase. Intra-vesicular protons are then used to drive neurotransmitter uptake by synaptic vesicles. V0a and V0c, two membrane subunits of the V0 sector, have been shown to interact with SNARE proteins, and their photo-inactivation rapidly impairs synaptic transmission. V0d, a soluble subunit of the V0 sector strongly interacts with its membrane-embedded subunits and is crucial for the canonic proton transfer activity of the V-ATPase. Our investigations show that the loop 1.2 of V0c interacts with complexin, a major partner of the SNARE machinery and that V0d1 binding to V0c inhibits this interaction, as well as V0c association with SNARE complex. The injection of recombinant V0d1 in rat superior cervical ganglion neurons rapidly reduced neurotransmission. In chromaffin cells, V0d1 overexpression and V0c silencing modified in a comparable manner several parameters of unitary exocytotic events. Our data suggest that V0c subunit promotes exocytosis via interactions with complexin and SNAREs and that this activity can be antagonized by exogenous V0d.


Assuntos
Proteínas SNARE , ATPases Vacuolares Próton-Translocadoras , Ratos , Animais , Proteínas SNARE/metabolismo , Prótons , Vesículas Sinápticas/metabolismo , Fusão de Membrana , ATPases Vacuolares Próton-Translocadoras/metabolismo
15.
Neuron ; 57(2): 210-6, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18215619

RESUMO

Short-term synaptic plasticity shapes the postsynaptic response to bursts of impulses and is crucial for encoding information in neurons, but the molecular mechanisms are unknown. Here we show that activity-dependent modulation of presynaptic Ca(V)2.1 channels mediated by neuronal Ca(2+) sensor proteins (CaS) induces synaptic plasticity in cultured superior cervical ganglion (SCG) neurons. A mutation of the IQ-like motif in the C terminus that blocks Ca(2+)/CaS-dependent facilitation of the P/Q-type Ca(2+) current markedly reduces facilitation of synaptic transmission. Deletion of the nearby calmodulin-binding domain, which inhibits CaS-dependent inactivation, substantially reduces depression of synaptic transmission. These results demonstrate that residual Ca(2+) in presynaptic terminals can act through CaS-dependent regulation of Ca(V)2.1 channels to induce short-term synaptic facilitation and rapid synaptic depression. Activity-dependent regulation of presynaptic Ca(V)2.1 channels by CaS proteins may therefore be a primary determinant of short-term synaptic plasticity and information-processing in the nervous system.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Proteínas Sensoras de Cálcio Intracelular/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Terminações Pré-Sinápticas/fisiologia , Motivos de Aminoácidos/fisiologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Relação Dose-Resposta à Radiação , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Camundongos , Microinjeções/métodos , Mutação , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Gânglio Cervical Superior/citologia , Fatores de Tempo , ômega-Agatoxina IVA/farmacologia
16.
Mol Pharmacol ; 82(2): 199-208, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22554805

RESUMO

Levetiracetam (LEV) is a prominent antiepileptic drug that binds to neuronal synaptic vesicle glycoprotein 2A protein and has reported effects on ion channels, but with a poorly defined mechanism of action. We investigated inhibition of voltage-dependent Ca(2+) (Ca(V)) channels as a potential mechanism through which LEV exerts effects on neuronal activity. We used electrophysiological methods to investigate the effects of LEV on cholinergic synaptic transmission and Ca(V) channel activity in superior cervical ganglion neurons (SCGNs). In parallel, we investigated the effects of the inactive LEV R-enantiomer, (R)-α-ethyl-2-oxo-1-pyrrolidine acetamide (UCB L060). LEV but not UCB L060 (each at 100 µM) inhibited synaptic transmission between SCGNs in long-term culture in a time-dependent manner, significantly reducing excitatory postsynaptic potentials after a ≥30-min application. In isolated SCGNs, LEV pretreatment (≥1 h) but not short-term application (5 min) significantly inhibited whole-cell Ba(2+) current (I(Ba)) amplitude. In current-clamp recordings, LEV reduced the amplitude of the afterhyperpolarizing potential in a Ca(2+)-dependent manner but also increased the action potential latency in a Ca(2+)-independent manner, which suggests additional mechanisms associated with reduced excitability. Intracellular LEV application (4-5 min) caused rapid inhibition of I(Ba) amplitude, to an extent comparable to that seen with extracellular LEV pretreatment (≥1 h). Neither pretreatment nor intracellular application of UCB L060 produced any inhibitory effects on I(Ba) amplitude. These results identify a stereospecific intracellular pathway through which LEV inhibits presynaptic Ca(V) channels; resultant reductions in neuronal excitability are proposed to contribute to the anticonvulsant effects of LEV.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio , Líquido Intracelular/efeitos dos fármacos , Glicoproteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Piracetam/análogos & derivados , Terminações Pré-Sinápticas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio/metabolismo , Líquido Intracelular/metabolismo , Levetiracetam , Ligantes , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Piracetam/metabolismo , Piracetam/farmacologia , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Gânglio Cervical Superior/efeitos dos fármacos , Gânglio Cervical Superior/metabolismo
17.
Proc Natl Acad Sci U S A ; 106(43): 18385-9, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19822743

RESUMO

Short-term synaptic facilitation plays an important role in information processing in the central nervous system. Although the crucial requirement of presynaptic Ca(2+) in the expression of this plasticity has been known for decades, the molecular mechanisms underlying the plasticity remain controversial. Here, we show that presynaptic metabotropic glutamate receptors (mGluRs) bind and release Munc18-1 (also known as rbSec1/nSec1), an essential protein for synaptic transmission, in a Ca(2+)-dependent manner, whose actions decrease and increase synaptic vesicle release, respectively. We found that mGluR4 bound Munc18-1 with an EC(50) for Ca(2+) of 168 nM, close to the resting Ca(2+) concentration, and that the interaction was disrupted by Ca(2+)-activated calmodulin (CaM) at higher concentrations of Ca(2+). Consistently, the Munc18-1-interacting domain of mGluR4 suppressed both dense-core vesicle secretion from permeabilized PC12 cells and synaptic transmission in neuronal cells. Furthermore, this domain was sufficient to induce paired-pulse facilitation. Obviously, the role of mGluR4 in these processes was independent of its classical function of activation by glutamate. On the basis of these experimental data, we propose the following model: When neurons are not active, Munc18-1 is sequestered by mGluR4, and therefore the basal synaptic transmission is kept low. After the action potential, the increase in the Ca(2+) level activates CaM, which in turn liberates Munc18-1 from mGluR4, causing short-term synaptic facilitation. Our findings unite and provide a new insight into receptor signaling and vesicular transport, which are pivotal activities involved in a variety of cellular processes.


Assuntos
Cálcio/metabolismo , Estriol/análogos & derivados , Proteínas Munc18/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transmissão Sináptica , Animais , Estriol/metabolismo , Camundongos , Proteínas Munc18/genética , Células PC12 , Ligação Proteica , Ratos
18.
Biomedicines ; 10(7)2022 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-35884898

RESUMO

Within 1 millisecond of action potential arrival at presynaptic terminals voltage-gated Ca2+ channels open. The Ca2+ channels are linked to synaptic vesicles which are tethered by active zone proteins. Ca2+ entrance into the active zone triggers: (1) the fusion of the vesicle and exocytosis, (2) the replenishment of the active zone with vesicles for incoming exocytosis, and (3) various types of endocytosis for vesicle reuse, dependent on the pattern of firing. These time-dependent vesicle dynamics are controlled by presynaptic Ca2+ sensor proteins, regulating active zone scaffold proteins, fusion machinery proteins, motor proteins, endocytic proteins, several enzymes, and even Ca2+ channels, following the decay of Ca2+ concentration after the action potential. Here, I summarize the Ca2+-dependent protein controls of synchronous and asynchronous vesicle release, rapid replenishment of the active zone, endocytosis, and short-term plasticity within 100 msec after the action potential. Furthermore, I discuss the contribution of active zone proteins to presynaptic plasticity and to homeostatic readjustment during and after intense activity, in addition to activity-dependent endocytosis.

19.
J Physiol ; 589(Pt 13): 3085-101, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21521766

RESUMO

Modulation of presynaptic voltage-dependent Ca2+ channels is a major means of controlling neurotransmitter release. The CaV2.2Ca2+ channel subunit contains several inhibitory interaction sites for Gßγ subunits, including the amino terminal (NT) and I-II loop. The NT and I-II loop have also been proposed to undergo a G protein-gated inhibitory interaction, whilst the NT itself has also been proposed to suppress CaV2 channel activity. Here, we investigate the effects of an amino terminal (CaV2.2[45-55]) 'NT peptide' and a I-II loop alpha interaction domain (CaV2.2[377-393]) 'AID peptide' on synaptic transmission, Ca2+ channel activity and G protein modulation in superior cervical ganglion neurones (SCGNs). Presynaptic injection of NT or AID peptide into SCGN synapses inhibited synaptic transmission and also attenuated noradrenaline-induced G protein modulation. In isolated SCGNs, NT and AID peptides reduced whole-cell Ca2+ current amplitude, modified voltage dependence of Ca2+ channel activation and attenuated noradrenaline-induced G protein modulation. Co-application of NT and AID peptide negated inhibitory actions. Together, these data favour direct peptide interaction with presynaptic Ca2+ channels, with effects on current amplitude and gating representing likely mechanisms responsible for inhibition of synaptic transmission. Mutations to residues reported as determinants of Ca2+ channel function within the NT peptide negated inhibitory effects on synaptic transmission, Ca2+ current amplitude and gating and G protein modulation. A mutation within the proposed QXXER motif for G protein modulation did not abolish inhibitory effects of the AID peptide. This study suggests that the CaV2.2 amino terminal and I-II loop contribute molecular determinants for Ca2+ channel function; the data favour a direct interaction of peptides with Ca2+ channels to inhibit synaptic transmission and attenuate G protein modulation.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/fisiologia , Inibição Neural/fisiologia , Peptídeos/fisiologia , Transmissão Sináptica/fisiologia , Sequência de Aminoácidos , Animais , Canais de Cálcio Tipo N/síntese química , Potenciais Pós-Sinápticos Excitadores/fisiologia , Dados de Sequência Molecular , Peptídeos/síntese química , Estrutura Terciária de Proteína , Ratos , Ratos Wistar
20.
J Biol Chem ; 285(52): 40943-55, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20978127

RESUMO

Neurotransmitter release is triggered by Ca(2+) binding to a low affinity Ca(2+) sensor, mostly synaptotagmin-1, which catalyzes SNARE-mediated synaptic vesicle fusion. Tomosyn negatively regulates Ca(2+)-dependent neurotransmitter release by sequestering target SNAREs through the C-terminal VAMP-like domain. In addition to the C terminus, the N-terminal WD40 repeats of tomosyn also have potent inhibitory activity toward Ca(2+)-dependent neurotransmitter release, although the molecular mechanism underlying this effect remains elusive. Here, we show that through its N-terminal WD40 repeats tomosyn directly binds to synaptotagmin-1 in a Ca(2+)-dependent manner. The N-terminal WD40 repeats impaired the activities of synaptotagmin-1 to promote SNARE complex-mediated membrane fusion and to bend the lipid bilayers. Decreased acetylcholine release from N-terminal WD40 repeat-microinjected superior cervical ganglion neurons was relieved by microinjection of the cytoplasmic domain of synaptotagmin-1. These results indicate that, upon direct binding, the N-terminal WD40 repeats negatively regulate the synaptotagmin-1-mediated step of Ca(2+)-dependent neurotransmitter release. Furthermore, we show that synaptotagmin-1 binding enhances the target SNARE-sequestering activity of tomosyn. These results suggest that the interplay between tomosyn and synaptotagmin-1 underlies inhibitory control of Ca(2+)-dependent neurotransmitter release.


Assuntos
Cálcio/metabolismo , Fusão de Membrana/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurotransmissores/metabolismo , Proteínas R-SNARE/metabolismo , Gânglio Cervical Superior/metabolismo , Sinaptotagmina I/metabolismo , Animais , Proteínas do Tecido Nervoso/genética , Estrutura Terciária de Proteína , Proteínas R-SNARE/genética , Ratos , Ratos Wistar , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Gânglio Cervical Superior/citologia , Sinaptotagmina I/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA