Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 42(6): 1001-1019, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34969867

RESUMO

Using postsynaptically tethered calcium sensor GCaMP, we investigated spontaneous synaptic transmission at individual active zones (AZs) at the Drosophila (both sexes) neuromuscular junction. Optical monitoring of GCaMP events coupled with focal electrical recordings of synaptic currents revealed "hot spots" of spontaneous transmission, which corresponded to transient states of elevated activity at selected AZs. The elevated spontaneous activity had two temporal components, one at a timescale of minutes and the other at a subsecond timescale. We developed a three-state model of AZ preparedness for spontaneous transmission and performed Monte Carlo simulations of the release process, which produced an accurate quantitative description of the variability and time course of spontaneous transmission at individual AZs. To investigate the mechanisms of elevated activity, we first focused on the protein complexin, which binds the SNARE protein complex and serves to clamp spontaneous fusion. Overexpression of Drosophila complexin largely abolished the high-activity states of AZs, while complexin deletion drastically promoted it. A mutation in the SNARE protein Syntaxin-1A had an effect similar to complexin deficiency, promoting the high-activity state. We next tested how presynaptic Ca2+ transients affect the states of elevated activity at individual AZs. We either blocked or promoted Ca2+ influx pharmacologically, and also promoted Ca2+ release from internal stores. These experiments coupled with computations revealed that Ca2+ transients can trigger bursts of spontaneous events from individual AZs or AZ clusters at a subsecond timescale. Together, our results demonstrated that spontaneous transmission is highly heterogeneous, with transient hot spots being regulated by the SNARE machinery and Ca2+SIGNIFICANCE STATEMENT Spontaneous synaptic transmission is a vital component of neuronal communication, since it regulates the neuronal development and plasticity. Our study demonstrated that spontaneous transmission is highly heterogeneous and that nerve terminals create transient "hot spots" of spontaneous release of neuronal transmitters. We show that these hot spots are regulated by the protein machinery mediating the release process and by calcium ions. These results contribute to our understanding of spontaneous synaptic transmission as a dynamic, plastic, and tightly regulated signaling mechanism and unravel fundamental biophysical properties of neuronal communication.


Assuntos
Junção Neuromuscular/fisiologia , Transmissão Sináptica/fisiologia , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Masculino , Proteínas do Tecido Nervoso/metabolismo , Proteínas SNARE/metabolismo , Processos Estocásticos
2.
Biochem J ; 478(2): 407-422, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33393983

RESUMO

Insulin stimulates glucose uptake in muscle cells by rapidly redistributing vesicles containing GLUT4 glucose transporters from intracellular compartments to the plasma membrane (PM). GLUT4 vesicle fusion requires the formation of SNARE complexes between vesicular VAMP and PM syntaxin4 and SNAP23. SNARE accessory proteins usually regulate vesicle fusion processes. Complexins aide in neuro-secretory vesicle-membrane fusion by stabilizing trans-SNARE complexes but their participation in GLUT4 vesicle fusion is unknown. We report that complexin-2 is expressed and homogeneously distributed in L6 rat skeletal muscle cells. Upon insulin stimulation, a cohort of complexin-2 redistributes to the PM. Complexin-2 knockdown markedly inhibited GLUT4 translocation without affecting proximal insulin signalling of Akt/PKB phosphorylation and actin fiber remodelling. Similarly, complexin-2 overexpression decreased maximal GLUT4 translocation suggesting that the concentration of complexin-2 is finely tuned to vesicle fusion. These findings reveal an insulin-dependent regulation of GLUT4 insertion into the PM involving complexin-2.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Insulina/farmacologia , Mioblastos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Transportador de Glucose Tipo 4/genética , Insulina/genética , Insulina/metabolismo , Músculo Esquelético/citologia , Mioblastos/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo
3.
Biochem J ; 478(7): 1315-1319, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33821970

RESUMO

Loss of the insulin-stimulated glucose uptake in muscle is a crucial event participating in the defect of whole-body metabolism in type 2 diabetes. Therefore, identification by Pavarotti et al. (Biochem. J (2021) 478 (2): 407-422) of complexin-2 as an important contributor to glucose transporter 4 (GLUT4) translocation to muscle cell plasma membrane upon insulin stimulation is essential. The present commentary discusses the biological importance of the findings and proposes future challenges and opportunities.


Assuntos
Diabetes Mellitus Tipo 2 , Membrana Celular/metabolismo , Glucose , Humanos , Insulina/metabolismo , Proteínas de Transporte de Monossacarídeos , Proteínas Musculares
4.
Am J Hum Genet ; 102(3): 427-446, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29499164

RESUMO

Genetic variation modulating risk of sporadic Parkinson disease (PD) has been primarily explored through genome-wide association studies (GWASs). However, like many other common genetic diseases, the impacted genes remain largely unknown. Here, we used single-cell RNA-seq to characterize dopaminergic (DA) neuron populations in the mouse brain at embryonic and early postnatal time points. These data facilitated unbiased identification of DA neuron subpopulations through their unique transcriptional profiles, including a postnatal neuroblast population and substantia nigra (SN) DA neurons. We use these population-specific data to develop a scoring system to prioritize candidate genes in all 49 GWAS intervals implicated in PD risk, including genes with known PD associations and many with extensive supporting literature. As proof of principle, we confirm that the nigrostriatal pathway is compromised in Cplx1-null mice. Ultimately, this systematic approach establishes biologically pertinent candidates and testable hypotheses for sporadic PD, informing a new era of PD genetic research.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Estudos de Associação Genética , Doença de Parkinson/genética , Doença de Parkinson/patologia , Análise de Sequência de RNA , Análise de Célula Única/métodos , Animais , Separação Celular , Redes Reguladoras de Genes , Loci Gênicos , Marcadores Genéticos , Estudo de Associação Genômica Ampla , Camundongos Knockout , Substância Negra/patologia
5.
J Biomol NMR ; 75(8-9): 347-363, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34505210

RESUMO

The development of methyl transverse relaxation optimized spectroscopy has greatly facilitated the study of macromolecular assemblies by solution NMR spectroscopy. However, limited sample solubility and stability has hindered application of this technique to ongoing studies of complexes formed on membranes by the neuronal SNAREs that mediate neurotransmitter release and synaptotagmin-1, the Ca2+ sensor that triggers release. Since the 1H NMR signal of a tBu group attached to a large protein or complex can be observed with high sensitivity if the group retains high mobility, we have explored the use of this strategy to analyze presynaptic complexes involved in neurotransmitter release. For this purpose, we attached tBu groups at single cysteines of fragments of synaptotagmin-1, complexin-1 and the neuronal SNAREs by reaction with 5-(tert-butyldisulfaneyl)-2-nitrobenzoic acid (BDSNB), tBu iodoacetamide or tBu acrylate. The tBu resonances of the tagged proteins were generally sharp and intense, although tBu groups attached with BDSNB had a tendency to exhibit somewhat broader resonances that likely result because of the shorter linkage between the tBu and the tagged cysteine. Incorporation of the tagged proteins into complexes on nanodiscs led to severe broadening of the tBu resonances in some cases. However, sharp tBu resonances could readily be observed for some complexes of more than 200 kDa at low micromolar concentrations. Our results show that tagging of proteins with tBu groups provides a powerful approach to study large biomolecular assemblies of limited stability and/or solubility that may be applicable even at nanomolar concentrations.


Assuntos
Neurônios , Proteínas SNARE , Substâncias Macromoleculares , Espectroscopia de Ressonância Magnética , Ressonância Magnética Nuclear Biomolecular
6.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34360929

RESUMO

Complexins (Cplxs) 1 to 4 are components of the presynaptic compartment of chemical synapses where they regulate important steps in synaptic vesicle exocytosis. In the retina, all four Cplxs are present, and while we know a lot about Cplxs 3 and 4, little is known about Cplxs 1 and 2. Here, we performed in situ hybridization experiments and bioinformatics and exploited Cplx 1 and Cplx 2 single-knockout mice combined with immunocytochemistry and light microscopy to characterize in detail the cell type and synapse-specific distribution of Cplx 1 and Cplx 2. We found that Cplx 2 and not Cplx 1 is the main isoform expressed in normal and displaced amacrine cells and ganglion cells in mouse retinae and that amacrine cells seem to operate with a single Cplx isoform at their conventional chemical synapses. Surprising was the finding that retinal function, determined with electroretinographic recordings, was altered in Cplx 1 but not Cplx 2 single-knockout mice. In summary, the results provide an important basis for future studies on the function of Cplxs 1 and 2 in the processing of visual signals in the mammalian retina.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Células Amácrinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Fotorreceptoras/metabolismo , Células Bipolares da Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Células Horizontais da Retina/metabolismo , Proteínas SNARE/metabolismo , Sinapses/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Células Cultivadas , Biologia Computacional/métodos , Eletrorretinografia/métodos , Feminino , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética
7.
J Biol Chem ; 294(10): 3325-3342, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30700558

RESUMO

The past few years have resulted in an increased awareness and recognition of the prevalence and roles of intrinsically disordered proteins and protein regions (IDPs and IDRs, respectively) in synaptic vesicle trafficking and exocytosis and in overall synaptic organization. IDPs and IDRs constitute a class of proteins and protein regions that lack stable tertiary structure, but nevertheless retain biological function. Their significance in processes such as cell signaling is now well accepted, but their pervasiveness and importance in other areas of biology are not as widely appreciated. Here, we review the prevalence and functional roles of IDPs and IDRs associated with the release and recycling of synaptic vesicles at nerve terminals, as well as with the architecture of these terminals. We hope to promote awareness, especially among neuroscientists, of the importance of this class of proteins in these critical pathways and structures. The examples discussed illustrate some of the ways in which the structural flexibility conferred by intrinsic protein disorder can be functionally advantageous in the context of cellular trafficking and synaptic function.


Assuntos
Exocitose/fisiologia , Proteínas Intrinsicamente Desordenadas/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Transporte Biológico Ativo/fisiologia , Humanos
8.
Proc Natl Acad Sci U S A ; 113(47): E7590-E7599, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27821736

RESUMO

In presynaptic nerve terminals, complexin regulates spontaneous "mini" neurotransmitter release and activates Ca2+-triggered synchronized neurotransmitter release. We studied the role of the C-terminal domain of mammalian complexin in these processes using single-particle optical imaging and electrophysiology. The C-terminal domain is important for regulating spontaneous release in neuronal cultures and suppressing Ca2+-independent fusion in vitro, but it is not essential for evoked release in neuronal cultures and in vitro. This domain interacts with membranes in a curvature-dependent fashion similar to a previous study with worm complexin [Snead D, Wragg RT, Dittman JS, Eliezer D (2014) Membrane curvature sensing by the C-terminal domain of complexin. Nat Commun 5:4955]. The curvature-sensing value of the C-terminal domain is comparable to that of α-synuclein. Upon replacement of the C-terminal domain with membrane-localizing elements, preferential localization to the synaptic vesicle membrane, but not to the plasma membrane, results in suppression of spontaneous release in neurons. Membrane localization had no measurable effect on evoked postsynaptic currents of AMPA-type glutamate receptors, but mislocalization to the plasma membrane increases both the variability and the mean of the synchronous decay time constant of NMDA-type glutamate receptor evoked postsynaptic currents.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Membrana Celular/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Camundongos , Neurônios/citologia , Ratos , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo , Sinaptotagminas/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo
9.
Alcohol Clin Exp Res ; 42(9): 1661-1673, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29944190

RESUMO

BACKGROUND: Recent work with long-term ethanol (EtOH) self-administration in nonhuman primate models has revealed a complex array of behavioral and physiological effects that closely mimic human alcohol abuse. Detailed neurophysiological analysis in these models suggests a myriad of pre- and postsynaptic neurobiological effects that may contribute to the behavioral manifestations of long-term EtOH drinking. The molecular mechanisms regulating presynaptic effects of this chronic EtOH exposure are largely unknown. To this end, we analyzed the effects of long-term EtOH self-administration on the levels of presynaptic SNARE complex proteins in Macaca mulatta basolateral amygdala, a brain region known to regulate both aversive and reward-seeking behaviors. METHODS: Basolateral amygdala samples from control and EtOH-drinking male and female monkeys were processed. Total basolateral amygdala protein was analyzed by Western blotting using antibodies directed against both core SNARE and SNARE-associated proteins. We also performed correlational analyses between protein expression levels and a number of EtOH drinking parameters, including lifetime grams of EtOH consumed, preference, and blood alcohol concentration. RESULTS: Significant interactions or main effects of sex/drinking were seen for a number of SNARE core and SNARE-associated proteins. Across the range of EtOH-drinking phenotypes, SNAP25 and Munc13-1 proteins levels were significantly different between males and females, and Munc13-2 levels were significantly lower in animals with a history of EtOH drinking. A separate analysis of very heavy-drinking individuals revealed significant decreases in Rab3c (females) and complexin 2 (males). CONCLUSIONS: Protein expression analysis of basolateral amygdala total protein from controls and animals following long-term EtOH self-administration suggests a number of alterations in core SNARE or SNARE-associated components that could dramatically alter presynaptic function. A number of proteins or multiprotein components were also correlated with EtOH drinking behavior, which suggest a potentially heritable role for presynaptic SNARE proteins.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/tendências , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/metabolismo , Etanol/administração & dosagem , Proteínas SNARE/biossíntese , Consumo de Bebidas Alcoólicas/efeitos adversos , Animais , Complexo Nuclear Basolateral da Amígdala/química , Etanol/efeitos adversos , Feminino , Macaca mulatta , Masculino , Proteínas SNARE/análise , Autoadministração , Fatores de Tempo
10.
Cereb Cortex ; 27(10): 4759-4768, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27655928

RESUMO

In mammals, subplate neurons (SPNs) are among the first generated cortical neurons. While most SPNs exist only transiently during development, a number of SPNs persist among adult Layer 6b (L6b). During development, SPNs receive thalamic and intra-cortical input, and primarily project to Layer 4 (L4). SPNs are critical for the anatomical and functional development of thalamocortical connections and also pioneer corticothalamic projections. Since SPNs are heterogeneous, SPN subpopulations might serve different roles. Here, we investigate the connectivity of one subpopulation, complexin-3 (Cplx3)-positive SPNs (Cplx3-SPNs), in mouse whisker somatosensory (barrel) cortex (S1). We find that many Cplx3-SPNs survive into adulthood and become a subpopulation of L6b. Cplx3-SPNs axons project to thalamorecipient layers, that is, L4, 5a, and 1. The L4 projections are biased towards the septal regions between barrels in the second postnatal week. Thus, S1 Cplx3-SPN targets co-localize with the eventual projections of the medial posterior thalamic nucleus (POm). In addition to their cortical targets, Cplx3-SPNs also extend long-range axons to several thalamic nuclei, including POm. Thus, Cplx3-SPN/L6b neurons are associated with paralemniscal pathways and can potentially directly link thalamocortical and corticothalamic circuits. This suggests an additional key role for SPNs in the establishment and maintenance of thalamocortical processing.


Assuntos
Vias Neurais/crescimento & desenvolvimento , Neurônios/metabolismo , Tálamo/metabolismo , Animais , Axônios/metabolismo , Córtex Cerebral/crescimento & desenvolvimento , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vibrissas/metabolismo
11.
Proc Natl Acad Sci U S A ; 112(32): E4475-84, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26216953

RESUMO

Brain-derived neurotrophic factor (BDNF) is known to modulate synapse development and plasticity, but the source of synaptic BDNF and molecular mechanisms regulating BDNF release remain unclear. Using exogenous BDNF tagged with quantum dots (BDNF-QDs), we found that endocytosed BDNF-QDs were preferentially localized to postsynaptic sites in the dendrite of cultured hippocampal neurons. Repetitive neuronal spiking induced the release of BDNF-QDs at these sites, and this process required activation of glutamate receptors. Down-regulating complexin 1/2 (Cpx1/2) expression eliminated activity-induced BDNF-QD secretion, although the overall activity-independent secretion was elevated. Among eight synaptotagmin (Syt) isoforms examined, down-regulation of only Syt6 impaired activity-induced BDNF-QD secretion. In contrast, activity-induced release of endogenously synthesized BDNF did not depend on Syt6. Thus, neuronal activity could trigger the release of endosomal BDNF from postsynaptic dendrites in a Cpx- and Syt6-dependent manner, and endosomes containing BDNF may serve as a source of BDNF for activity-dependent synaptic modulation.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Endocitose , Proteínas do Tecido Nervoso/metabolismo , Sinaptotagminas/metabolismo , Animais , Axônios/metabolismo , Cálcio/metabolismo , Compartimento Celular , Células Cultivadas , Dendritos , Regulação para Baixo , Exocitose , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Espaço Intracelular/metabolismo , Camundongos , Modelos Biológicos , Transporte Proteico , Pontos Quânticos/metabolismo , Receptores de Glutamato/metabolismo , Sinapses
12.
Int J Mol Sci ; 19(11)2018 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-30366444

RESUMO

Retinal vein occlusion (RVO) is a common retinal vascular disease. RVO may be complicated by pronounced ischemia that often leads to severe loss of visual function. The present work aimed at studying the retinal proteome of RVO complicated by ischemia. In six Danish Landrace pigs RVO was induced with argon laser in the right eye of each animal. As four retinal veins were occluded, the RVO best corresponded to a central retinal vein occlusion (CRVO). Left control eyes received a similar laser treatment without inducing occlusion. RVO and retinal ischemia were verified by angiography. The retinas were collected 15 days after RVO for proteomic analysis. RVO resulted in a downregulation of proteins involved in visual perception, including rhodopsin, transducin alpha chain, and peripherin-2. RVO also caused a downregulation of proteins involved in neurotransmitter transport, including glutamate decarboxylase 1 (GAD1), glutamate decarboxylase 2 (GAD2), and complexins 2⁻4. RVO lead to increased contents of proteins involved in inflammation, including interleukin-18 (IL-18), S100A12, and annexin A1 (ANXA1). Immunohistochemistry revealed a general retinal upregulation of IL-18 and ANXA1 while S100A12 was highly abundant in retinal ganglion cells in RVO. IL-18 and S100A12 are likely to be driving forces in the inflammatory response of RVO complicated by ischemia. Our findings also suggest that RVO results in compromised neurotransmission and a downregulation of proteins involved in visual perception.


Assuntos
Interleucina-18/metabolismo , Oclusão da Veia Retiniana/metabolismo , Proteína S100A12/metabolismo , Animais , Anexina A1/metabolismo , Western Blotting , Glutamato Descarboxilase/metabolismo , Espectrometria de Massas , Suínos
13.
Biochem J ; 473(14): 2219-24, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27222590

RESUMO

Complexin (Cpx) is thought to be a major regulator of soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE)-dependent membrane fusion. Although the inhibition of membrane fusion by Cpx has been frequently reported, its structural basis has been elusive and an anticipated disruption of the SNARE core has never been observed. In the present study, to mimic the natural environment, we assembled a single SNAREpin between two nanodisc membrane patches. Single-molecule FRET (smFRET) detects a large conformational change, specifically at the C-terminal half, whereas no conformational change is observed at the N-terminal half. Our results suggest that Cpx splits the C-terminal half of the SNARE core at least 10 Å (1 Å=0.1 nm), whereby inhibiting further progression of SNARE zippering and membrane fusion.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas SNARE/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Fusão de Membrana/fisiologia , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Ligação Proteica/fisiologia , Proteínas SNARE/genética
14.
J Neurosci ; 35(9): 4065-70, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25740533

RESUMO

Ribbon synapses of photoreceptor cells and bipolar neurons in the retina signal graded changes in light intensity via sustained release of neurotransmitter. One molecular specialization of retinal ribbon synapses is the expression of complexin protein subtypes Cplx3 and Cplx4, whereas conventional synapses express Cplx1 and Cplx2. Because complexins bind to the molecular machinery for synaptic vesicle fusion (the SNARE complex) and modulate transmitter release at conventional synapses, we examined the roles of ribbon-specific complexin in regulating release at ribbon synapses of ON bipolar neurons from mouse retina. To interfere acutely with the interaction of native complexins with the SNARE complex, a peptide consisting of the highly conserved SNARE-binding domain of Cplx3 was introduced via a whole-cell patch pipette placed directly on the synaptic terminal, and vesicle fusion was monitored using capacitance measurements and FM-dye destaining. The inhibitory peptide, but not control peptides, increased spontaneous synaptic vesicle fusion, partially depleted reserve synaptic vesicles, and reduced fusion triggered by opening voltage-gated calcium channels under voltage clamp, without affecting the number of synaptic vesicles associated with ribbons, as revealed by electron microscopy of recorded terminals. The results are consistent with a dual role for ribbon-specific complexin, acting as a brake on the SNARE complex to prevent spontaneous fusion in the absence of calcium influx, while at the same time facilitating release evoked by depolarization.


Assuntos
Proteínas do Olho/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Neurotransmissores/metabolismo , Células Bipolares da Retina/fisiologia , Sinapses/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Animais , Proteínas do Olho/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Patch-Clamp , Vesículas Sinápticas/fisiologia
15.
J Neurosci ; 35(21): 8272-90, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26019341

RESUMO

Complexins (Cplxs) are small synaptic proteins that cooperate with SNARE-complexes in the control of synaptic vesicle (SV) fusion. Studies involving genetic mutation, knock-down, or knock-out indicated two key functions of Cplx that are not mutually exclusive but cannot easily be reconciled, one in facilitating SV fusion, and one in "clamping" SVs to prevent premature fusion. Most studies on the role of Cplxs in mammalian synapse function have relied on cultured neurons, heterologous expression systems, or membrane fusion assays in vitro, whereas little is known about the function of Cplxs in native synapses. We therefore studied consequences of genetic ablation of Cplx1 in the mouse calyx of Held synapse, and discovered a developmentally exacerbating phenotype of reduced spontaneous and evoked transmission but excessive asynchronous release after stimulation, compatible with combined facilitating and clamping functions of Cplx1. Because action potential waveforms, Ca(2+) influx, readily releasable SV pool size, and quantal size were unaltered, the reduced synaptic strength in the absence of Cplx1 is most likely a consequence of a decreased release probability, which is caused, in part, by less tight coupling between Ca(2+) channels and docked SV. We found further that the excessive asynchronous release in Cplx1-deficient calyces triggered aberrant action potentials in their target neurons, and slowed-down the recovery of EPSCs after depleting stimuli. The augmented asynchronous release had a delayed onset and lasted hundreds of milliseconds, indicating that it predominantly represents fusion of newly recruited SVs, which remain unstable and prone to premature fusion in the absence of Cplx1.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/deficiência , Tronco Encefálico/metabolismo , Proteínas do Tecido Nervoso/deficiência , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Animais , Tronco Encefálico/citologia , Adesão Celular/fisiologia , Exocitose/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/biossíntese
16.
J Biol Chem ; 290(16): 10518-34, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25716318

RESUMO

Sec1/Munc18 (SM) proteins are essential for membrane trafficking, but their molecular mechanism remains unclear. Using a single vesicle-vesicle content-mixing assay with reconstituted neuronal SNAREs, synaptotagmin-1, and complexin-1, we show that the neuronal SM protein Munc18a/nSec1 has no effect on the intrinsic kinetics of both spontaneous fusion and Ca(2+)-triggered fusion between vesicles that mimic synaptic vesicles and the plasma membrane. However, wild type Munc18a reduced vesicle association ∼50% when the vesicles bearing the t-SNAREs syntaxin-1A and SNAP-25 were preincubated with Munc18 for 30 min. Single molecule experiments with labeled SNAP-25 indicate that the reduction of vesicle association is a consequence of sequestration of syntaxin-1A by Munc18a and subsequent release of SNAP-25 (i.e. Munc18a captures syntaxin-1A via its high affinity interaction). Moreover, a phosphorylation mimic mutant of Munc18a with reduced affinity to syntaxin-1A results in less reduction of vesicle association. In summary, Munc18a does not directly affect fusion, although it has an effect on the t-SNARE complex, depending on the presence of other factors and experimental conditions. Our results suggest that Munc18a primarily acts at the prefusion stage.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Cálcio/metabolismo , Fusão de Membrana , Proteínas Munc18/genética , Proteínas do Tecido Nervoso/genética , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Cinética , Proteínas Munc18/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fosforilação , Ligação Proteica , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transmissão Sináptica , Vesículas Sinápticas/química , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Sinaptotagmina I/metabolismo , Termodinâmica , Proteína 2 Associada à Membrana da Vesícula/genética , Proteína 2 Associada à Membrana da Vesícula/metabolismo
17.
Curr Psychiatry Rep ; 18(8): 77, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27371030

RESUMO

Schizophrenia is a serious psychiatric illness which is experienced by about 1 % of individuals worldwide and has a debilitating impact on perception, cognition, and social function. Over the years, several models/hypotheses have been developed which link schizophrenia to dysregulations of the dopamine, glutamate, and serotonin receptor pathways. An important segment of these pathways that have been extensively studied for the pathophysiology of schizophrenia is the presynaptic neurotransmitter release mechanism. This set of molecular events is an evolutionarily well-conserved process that involves vesicle recruitment, docking, membrane fusion, and recycling, leading to efficient neurotransmitter delivery at the synapse. Accumulated evidence indicate dysregulation of this mechanism impacting postsynaptic signal transduction via different neurotransmitters in key brain regions implicated in schizophrenia. In recent years, after ground-breaking work that elucidated the operations of this mechanism, research efforts have focused on the alterations in the messenger RNA (mRNA) and protein expression of presynaptic neurotransmitter release molecules in schizophrenia and other neuropsychiatric conditions. In this review article, we present recent evidence from schizophrenia human postmortem studies that key proteins involved in the presynaptic release mechanism are dysregulated in the disorder. We also discuss the potential impact of dysfunctional presynaptic neurotransmitter release on the various neurotransmitter systems implicated in schizophrenia.


Assuntos
Encéfalo/fisiopatologia , Esquizofrenia/fisiopatologia , Psicologia do Esquizofrênico , Vesículas Sinápticas/fisiologia , Animais , Encéfalo/patologia , Humanos , Proteínas Munc18/fisiologia , Neurotransmissores/metabolismo , Proteínas Qa-SNARE/fisiologia , Proteínas R-SNARE/fisiologia , RNA Mensageiro/genética , Receptores Pré-Sinápticos/fisiologia , Proteínas SNARE/fisiologia , Esquizofrenia/patologia , Transdução de Sinais/fisiologia , Sinapsinas/fisiologia , Vesículas Sinápticas/genética , Sinaptofisina/fisiologia , Proteína 25 Associada a Sinaptossoma/fisiologia
18.
Cell Mol Life Sci ; 72(22): 4221-35, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26245303

RESUMO

Despite intensive research, it is still unclear how an immediate and profound acceleration of exocytosis is triggered by appropriate Ca(2+)-stimuli in presynaptic terminals. This is due to the fact that the molecular mechanisms of "docking" and "priming" reactions, which set up secretory vesicles to fuse at millisecond time scale, are extremely hard to study. Yet, driven by a fruitful combination of in vitro and in vivo analyses, our mechanistic understanding of Ca(2+)-triggered vesicle fusion has certainly advanced in the past few years. In this review, we aim to highlight recent progress and emerging views on the molecular mechanisms, by which constitutively forming SNAREpins are organized in functional, tightly regulated units for synchronized release. In particular, we will focus on the role of the small regulatory factor complexin whose function in Ca(2+)-dependent exocytosis has been controversially discussed for more than a decade. Special emphasis will also be laid on the functional relationship of complexin and synaptotagmin, as both proteins possibly act as allies and/or antagonists to govern SNARE-mediated exocytosis.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Cálcio/metabolismo , Exocitose , Proteínas do Tecido Nervoso/metabolismo , Vesículas Sinápticas/metabolismo , Humanos , Fusão de Membrana , Modelos Biológicos , Ligação Proteica , Proteínas SNARE/metabolismo , Sinaptotagminas/metabolismo
19.
Neurol Sci ; 37(3): 411-6, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26577184

RESUMO

Although complexin 1 (CPLX1) is not known as an inflammation factor, recent identification of a complexin 1 (CPLX1) polymorphism in Behçet's disease (BD) has sparked an interest in the role of this molecule in autoinflammation. DNA samples were isolated from peripheral blood mononuclear cells (PBMC) of BD and neuro-Behçet's disease (NBD) patients and expression levels of CPLX1 and miR-185, a predicted target miRNA for CPLX1 and an inflammation-related miRNA, were investigated by real time PCR assays. PBMC expression levels of CPLX1 were significantly increased in BD and NBD patients. By contrast, levels of miR-185 were reduced in both patient groups. A moderate inverse correlation was found between levels of CPLX1 and miR-185. No correlation could be found between expression levels and clinical features of patients. Significant expression alterations of CPLX1 in BD and NBD patients suggest that this molecule has a proinflammatory action. The putative role of CPLX1 in BD pathogenesis remains to be further studied.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/sangue , Síndrome de Behçet/sangue , MicroRNAs/sangue , Proteínas do Tecido Nervoso/sangue , Doenças do Sistema Nervoso/sangue , Proteínas Adaptadoras de Transporte Vesicular/genética , Adolescente , Adulto , Síndrome de Behçet/genética , Feminino , Técnicas de Genotipagem , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Doenças do Sistema Nervoso/genética , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença , Adulto Jovem
20.
Mol Cell Neurosci ; 56: 244-54, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23769723

RESUMO

The COMPLEXIN (CPX) proteins play a critical role in synaptic vesicle fusion and neurotransmitter release. Previous studies demonstrated that CPX functions in both activation of evoked neurotransmitter release and inhibition/clamping of spontaneous synaptic vesicle fusion. Here we report a new cpx mutant in Drosophila melanogaster, cpx(1257), revealing spatially defined and separable pools of CPX which make distinct contributions to the activation and clamping functions. In cpx(1257), lack of only the last C-terminal amino acid of CPX is predicted to disrupt prenylation and membrane targeting of CPX. Immunocytochemical analysis established localization of wild-type CPX to active zone (AZ) regions containing neurotransmitter release sites as well as broader presynaptic membrane compartments including synaptic vesicles. Parallel biochemical studies confirmed CPX membrane association and demonstrated robust binding interactions of CPX with all three SNAREs. This is in contrast to the cpx(1257) mutant, in which AZ localization of CPX persists but general membrane localization and, surprisingly, the bulk of CPX-SNARE protein interactions are abolished. Furthermore, electrophysiological analysis of neuromuscular synapses revealed interesting differences between cpx(1257) and a cpx null mutant. The cpx null exhibited a marked decrease in the EPSC amplitude, slowed EPSC rise and decay times and an increased mEPSC frequency with respect to wild-type. In contrast, cpx(1257) exhibited a wild-type EPSC with an increased mEPSC frequency and thus a selective failure to clamp spontaneous release. These results indicate that spatially distinct and separable interactions of CPX with presynaptic membranes and SNARE proteins mediate separable activation and clamping functions of CPX in neurotransmitter release.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Drosophila melanogaster/metabolismo , Exocitose , Mutação , Junção Neuromuscular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Sítios de Ligação , Drosophila melanogaster/genética , Potenciais Pós-Sinápticos Excitadores , Junção Neuromuscular/fisiologia , Ligação Proteica , Transporte Proteico , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA