Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 35(3): ar12, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38117594

RESUMO

Insulin secretion depends on the Ca2+-regulated fusion of granules with the plasma membrane. A recent model of Ca2+-triggered exocytosis in secretory cells proposes that lipids in the plasma membrane couple the calcium sensor Syt1 to the membrane fusion machinery (Kiessling et al., 2018). Specifically, Ca2+-mediated binding of Syt1's C2 domains to the cell membrane shifts the membrane-anchored SNARE syntaxin-1a to a more fusogenic conformation, straightening its juxtamembrane linker. To test this model in live cells and extend it to insulin secretion, we enriched INS1 cells with a panel of lipids with different acyl chain compositions. Fluorescence lifetime measurements demonstrate that cells with more disordered membranes show an increase in fusion efficiency, and vice versa. Experiments with granules purified from INS1 cells and recombinant SNARE proteins reconstituted in supported membranes confirmed that lipid acyl chain composition determines SNARE conformation and that lipid disordering correlates with increased fusion. Addition of Syt1's C2AB domains significantly decreased lipid order in target membranes and increased SNARE-mediated fusion probability. Strikingly, Syt's action on both fusion and lipid order could be partially bypassed by artificially increasing unsaturated phosphatidylserines in the target membrane. Thus, plasma membrane lipids actively participate in coupling Ca2+/synaptotagmin-sensing to the SNARE fusion machinery in cells.


Assuntos
Células Secretoras de Insulina , Fusão de Membrana , Lipídeos de Membrana/metabolismo , Proteínas SNARE/metabolismo , Células Secretoras de Insulina/metabolismo , Membrana Celular/metabolismo , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo , Exocitose , Proteínas Recombinantes/metabolismo , Cálcio/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(38): e2208337119, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36103579

RESUMO

Synchronous release at neuronal synapses is accomplished by a machinery that senses calcium influx and fuses the synaptic vesicle and plasma membranes to release neurotransmitters. Previous studies suggested the calcium sensor synaptotagmin (Syt) is a facilitator of vesicle docking and both a facilitator and inhibitor of fusion. On phospholipid monolayers, the Syt C2AB domain spontaneously oligomerized into rings that are disassembled by Ca2+, suggesting Syt rings may clamp fusion as membrane-separating "washers" until Ca2+-mediated disassembly triggers fusion and release [J. Wang et al., Proc. Natl. Acad. Sci. U.S.A. 111, 13966-13971 (2014)].). Here, we combined mathematical modeling with experiment to measure the mechanical properties of Syt rings and to test this mechanism. Consistent with experimental results, the model quantitatively recapitulates observed Syt ring-induced dome and volcano shapes on phospholipid monolayers and predicts rings are stabilized by anionic phospholipid bilayers or bulk solution with ATP. The selected ring conformation is highly sensitive to membrane composition and bulk ATP levels, a property that may regulate vesicle docking and fusion in ATP-rich synaptic terminals. We find the Syt molecules hosted by a synaptic vesicle oligomerize into a halo, unbound from the vesicle, but in proximity to sufficiently phosphatidylinositol 4,5-bisphosphate (PIP2)-rich plasma membrane (PM) domains, the PM-bound trans Syt ring conformation is preferred. Thus, the Syt halo serves as landing gear for spatially directed docking at PIP2-rich sites that define the active zones of exocytotic release, positioning the Syt ring to clamp fusion and await calcium. Our results suggest the Syt ring is both a Ca2+-sensitive fusion clamp and a high-fidelity sensor for directed docking.


Assuntos
Vesículas Sinápticas , Sinaptotagmina I , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Fosfatidilinositol 4,5-Difosfato/química , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/química
3.
Biophys J ; 121(18): 3370-3380, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36016497

RESUMO

Complexin-1 is an essential protein for neuronal exocytosis that acts to depress spontaneous fusion events while enhancing evoked neurotransmitter release. In addition to binding soluble N-ethylmaleimide-sensitive factor attachment protein receptors, it is well established that complexin associates with membranes in a manner that depends upon membrane curvature. In the present work, we examine the membrane binding of complexin using electron paramagnetic resonance spectroscopy, fluorescence anisotropy, and total internal reflection fluorescence microscopy. The apparent membrane affinity of complexin is found to strongly depend upon the concentration of protein used in the binding assay, and this is a result of a limited number of binding sites for complexin on the membrane interface. Although both the N- and C-terminal regions of complexin associate with the membrane interface, membrane affinity is driven by its C-terminus. Complexin prefers to bind liquid-disordered membrane phases and shows an enhanced affinity toward membranes containing phosphatidylinositol 4-5-bisphosphate (PI(4,5)P2). In the presence of PI(4,5)P2, complexin is displaced from the membrane surface by proteins that bind to or sequester PI(4,5)P2. In particular, the neuronal calcium sensor synaptotagmin-1 displaces complexin from the membrane but only when PI(4,5)P2 is present. Complexin and synaptotagmin compete on the membrane interface in the presence of PI(4,5)P2, and this interaction may play a role in calcium-triggered exocytosis by displacing complexin from its fusion-inhibiting state.


Assuntos
Cálcio , Fosfatidilinositol 4,5-Difosfato , Proteínas Adaptadoras de Transporte Vesicular/química , Sítios de Ligação , Cálcio/metabolismo , Exocitose , Proteínas do Tecido Nervoso/química , Neurotransmissores , Proteínas SNARE/metabolismo , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Sinaptotagmina I/química
4.
Nano Lett ; 22(3): 1449-1455, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34855407

RESUMO

A mechanism for full-length synaptotagmin-1 (syt-1) to interact with anionic bilayers and to promote fusion in the presence of SNAREs is proposed. Colloidal probe force spectroscopy in conjunction with tethered particle motion monitoring showed that in the absence of Ca2+ the binding of syt-1 to membranes depends on the presence and content of PI(4,5)P2. Addition of Ca2+ switches the interaction forces from weak to strong, eventually exceeding the cohesion of the C2A domain of syt-1 leading to partial unfolding of the protein. Fusion of single unilamellar vesicles equipped with syt-1 and synaptobrevin 2 with planar pore-spanning target membranes containing PS and PI(4,5)P2 shows an almost complete suppression of stalled intermediate fusion states and an accelerated fusion kinetics in the presence of Ca2+, which is further enhanced upon addition of ATP.


Assuntos
Cálcio , Fosfatidilinositol 4,5-Difosfato , Proteínas SNARE , Sinaptotagmina I , Cálcio/química , Cálcio/metabolismo , Cinética , Fusão de Membrana , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Desdobramento de Proteína , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo
5.
Life Sci Alliance ; 4(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34408000

RESUMO

Non-vesicular lipid transfer at ER and plasma membrane (PM) contact sites (CS) is crucial for the maintenance of membrane lipid homeostasis. Extended synaptotagmins (E-Syts) play a central role in this process as they act as molecular tethers of ER and PM and as lipid transfer proteins between these organelles. E-Syts are proteins constitutively anchored to the ER through an N-terminal hydrophobic segment and bind the PM via a variable number of C-terminal C2 domains. Synaptotagmins (SYTs) are the plant orthologous of E-Syts and regulate the ER-PM communication in response to abiotic stress. Combining different structural and biochemical techniques, we demonstrate that the binding of SYT1 to lipids occurs through a Ca2+-dependent lipid-binding site and by a site for phosphorylated forms of phosphatidylinositol, thus integrating two different molecular signals in response to stress. In addition, we show that SYT1 displays three highly flexible hinge points that provide conformational freedom to facilitate lipid extraction, protein loading, and subsequent transfer between PM and ER.


Assuntos
Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Membrana Celular , Modelos Moleculares , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo , Sequência de Aminoácidos , Arabidopsis/fisiologia , Proteínas de Arabidopsis/genética , Sítios de Ligação , Cálcio/química , Cálcio/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Lipídeos/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas Mutantes , Ligação Proteica , Relação Estrutura-Atividade , Sinaptotagmina I/genética
6.
Mol Cell Neurosci ; 112: 103613, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33753311

RESUMO

Presynaptic neurotransmitter release is strictly regulated by SNARE proteins, Ca2+ and a number of Ca2+ sensors including synaptotagmins (Syts) and Double C2 domain proteins (Doc2s). More than seventy years after the original description of spontaneous release, the mechanism that regulates this process is still poorly understood. Syt-1, Syt7 and Doc2 proteins contribute predominantly, but not exclusively, to synchronous, asynchronous and spontaneous phases of release. The proteins share a conserved tandem C2 domain architecture, but are functionally diverse in their subcellular location, Ca2+-binding properties and protein interactions. In absence of Syt-1, Doc2a and -b, neurons still exhibit spontaneous vesicle fusion which remains Ca2+-sensitive, suggesting the existence of additional sensors. Here, we selected Doc2c, rabphilin-3a and Syt-7 as three potential Ca2+ sensors for their sequence homology with Syt-1 and Doc2b. We genetically ablated each candidate gene in absence of Doc2a and -b and investigated spontaneous and evoked release in glutamatergic hippocampal neurons, cultured either in networks or on microglial islands (autapses). The removal of Doc2c had no effect on spontaneous or evoked release. Syt-7 removal also did not affect spontaneous release, although it altered short-term plasticity by accentuating short-term depression. The removal of rabphilin caused an increased spontaneous release frequency in network cultures, an effect that was not observed in autapses. Taken together, we conclude that Doc2c and Syt-7 do not affect spontaneous release of glutamate in hippocampal neurons, while our results suggest a possible regulatory role of rabphilin-3a in neuronal networks. These findings importantly narrow down the repertoire of synaptic Ca2+ sensors that may be implicated in the spontaneous release of glutamate.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Cálcio/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Sinaptotagmina I/fisiologia , Proteínas de Transporte Vesicular/fisiologia , Potenciais de Ação , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Sequência Conservada , Ácido Glutâmico/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Domínios Proteicos , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sinaptotagmina I/química , Sinaptotagmina I/deficiência , Sinaptotagmina I/genética , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/deficiência , Proteínas de Transporte Vesicular/genética , Rabfilina-3A
7.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33622785

RESUMO

Maintaining the balance between neuronal excitation and inhibition is essential for proper function of the central nervous system. Inhibitory synaptic transmission plays an important role in maintaining this balance. Although inhibitory transmission has higher kinetic demands compared to excitatory transmission, its properties are poorly understood. In particular, the dynamics and exocytosis of single inhibitory vesicles have not been investigated, due largely to both technical and practical limitations. Using a combination of quantum dots (QDs) conjugated to antibodies against the luminal domain of the vesicular GABA transporter to selectively label GABAergic (i.e., predominantly inhibitory) vesicles together with dual-focus imaging optics, we tracked the real-time three-dimensional position of single GABAergic vesicles up to the moment of exocytosis (i.e., fusion). Using three-dimensional trajectories, we found that GABAergic synaptic vesicles traveled a shorter distance prior to fusion and had a shorter time to fusion compared to synaptotagmin-1 (Syt1)-labeled vesicles, which were mostly from excitatory neurons. Moreover, our analysis revealed that GABAergic synaptic vesicles move more straightly to their release sites than Syt1-labeled vesicles. Finally, we found that GABAergic vesicles have a higher prevalence of kiss-and-run fusion than Syt1-labeled vesicles. These results indicate that inhibitory synaptic vesicles have a unique set of dynamics and exocytosis properties to support rapid synaptic inhibition, thereby maintaining a tightly regulated coordination between excitation and inhibition in the central nervous system.


Assuntos
Exocitose/fisiologia , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Neurônios GABAérgicos/metabolismo , Coloração e Rotulagem/métodos , Vesículas Sinápticas/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos/química , Cálcio/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/química , Neurônios GABAérgicos/citologia , Hipocampo/citologia , Hipocampo/metabolismo , Imageamento Tridimensional , Imunoconjugados/química , Transporte de Íons , Fusão de Membrana/fisiologia , Cultura Primária de Células , Pontos Quânticos/química , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo
8.
Nat Commun ; 12(1): 761, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33536412

RESUMO

Synaptotagmin 1 is a vesicle-anchored membrane protein that functions as the Ca2+ sensor for synchronous neurotransmitter release. In this work, an arginine containing region in the second C2 domain of synaptotagmin 1 (C2B) is shown to control the expansion of the fusion pore and thereby the concentration of neurotransmitter released. This arginine apex, which is opposite the Ca2+ binding sites, interacts with membranes or membrane reconstituted SNAREs; however, only the membrane interactions occur under the conditions in which fusion takes place. Other regions of C2B influence the fusion probability and kinetics but do not control the expansion of the fusion pore. These data indicate that the C2B domain has at least two distinct molecular roles in the fusion event, and the data are consistent with a model where the arginine apex of C2B positions the domain at the curved membrane surface of the expanding fusion pore.


Assuntos
Arginina/metabolismo , Membrana Celular/metabolismo , Fusão de Membrana , Proteínas SNARE/metabolismo , Sinaptotagmina I/metabolismo , Animais , Arginina/química , Sítios de Ligação , Cálcio/metabolismo , Neurotransmissores/metabolismo , Ligação Proteica , Domínios Proteicos , Ratos , Proteínas SNARE/química , Sinaptotagmina I/química
9.
J Chem Theory Comput ; 16(12): 7840-7851, 2020 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-33166466

RESUMO

Fusion pores serve as an effective mechanism to connect intracellular organelles and release vesicle contents during exocytosis. A complex lipid rearrangement takes place as membranes approximate, bend, fuse, and establish a traversing water channel to define the fusion pore, linking initially isolated chambers. Thermodynamically, the process is unfavorable and thought to be mediated by specialized proteins. In this work, we have developed a reaction coordinate to induce fusion pores from initially flat and parallel lipid bilayers and we have used it to describe the effects of the synaptotagmin-1 C2B domain during the process. We have obtained free-energy profiles of the whole lipid reorganization in biologically realistic membranes, going from planar and parallel bilayers through stalk hemifusion to water channel formation. Our results point to a lysine-rich polybasic region on synaptotagmin-1 C2B as the key to lipid reorganization control through the formation of phosphatidylinositol bisphosphate clusters that stabilize the fusion pore.


Assuntos
Bicamadas Lipídicas/química , Fosfatidilinositol 4,5-Difosfato/química , Sinaptotagmina I/química , Humanos , Domínios Proteicos , Estabilidade Proteica , Termodinâmica
10.
Toxins (Basel) ; 12(9)2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32957706

RESUMO

Botulinum neurotoxins (BoNTs) can be used therapeutically to treat a wide range of neuromuscular and neurological conditions. A collection of natural BoNT variants exists which can be classified into serologically distinct serotypes (BoNT/B), and further divided into subtypes (BoNT/B1, B2, …). BoNT subtypes share a high degree of sequence identity within the same serotype yet can display large variation in toxicity. One such example is BoNT/B2, which was isolated from Clostridium botulinum strain 111 in a clinical case of botulism, and presents a 10-fold lower toxicity than BoNT/B1. In an effort to understand the molecular mechanisms behind this difference in potency, we here present the crystal structures of BoNT/B2 in complex with the ganglioside receptor GD1a, and with the human synaptotagmin I protein receptor. We show, using receptor-binding assays, that BoNT/B2 has a slightly higher affinity for GD1a than BoNT/B1, and confirm its considerably weaker affinity for its protein receptors. Although the overall receptor-binding mechanism is conserved for both receptors, structural analysis suggests the lower affinity of BoNT/B2 is the result of key substitutions, where hydrophobic interactions important for synaptotagmin-binding are replaced by polar residues. This study provides a template to drive the development of future BoNT therapeutic molecules centered on assessing the natural subtype variations in receptor-binding that appears to be one of the principal stages driving toxicity.


Assuntos
Toxinas Botulínicas Tipo A/metabolismo , Gangliosídeos/metabolismo , Sinaptotagmina I/metabolismo , Sítios de Ligação , Toxinas Botulínicas Tipo A/química , Configuração de Carboidratos , Gangliosídeos/química , Interações Hidrofóbicas e Hidrofílicas , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Sinaptotagmina I/química
11.
Elife ; 92020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32515733

RESUMO

The success of comparative cell biology for determining protein function relies on quality disruption techniques. Long-lived proteins, in postmitotic cells, are particularly difficult to eliminate. Moreover, cellular processes are notoriously adaptive; for example, neuronal synapses exhibit a high degree of plasticity. Ideally, protein disruption techniques should be both rapid and complete. Here, we describe knockoff, a generalizable method for the druggable control of membrane protein stability. We developed knockoff for neuronal use but show it also works in other cell types. Applying knockoff to synaptotagmin 1 (SYT1) results in acute disruption of this protein, resulting in loss of synchronous neurotransmitter release with a concomitant increase in the spontaneous release rate, measured optically. Thus, SYT1 is not only the proximal Ca2+ sensor for fast neurotransmitter release but also serves to clamp spontaneous release. Additionally, knockoff can be applied to protein domains as we show for another synaptic vesicle protein, synaptophysin 1.


Assuntos
Hipocampo/citologia , Neurônios/metabolismo , Sinaptotagmina I , Animais , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Neurotransmissores/metabolismo , Ratos , Ratos Sprague-Dawley , Sinaptotagmina I/química , Sinaptotagmina I/genética , Sinaptotagmina I/metabolismo
12.
PLoS One ; 15(5): e0232991, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32407359

RESUMO

Following nerve stimulation, there are two distinct phases of Ca2+-dependent neurotransmitter release: a fast, synchronous release phase, and a prolonged, asynchronous release phase. Each of these phases is tightly regulated and mediated by distinct mechanisms. Synaptotagmin 1 is the major Ca2+ sensor that triggers fast, synchronous neurotransmitter release upon Ca2+ binding by its C2A and C2B domains. It has also been implicated in the inhibition of asynchronous neurotransmitter release, as blocking Ca2+ binding by the C2A domain of synaptotagmin 1 results in increased asynchronous release. However, the mutation used to block Ca2+ binding in the previous experiments (aspartate to asparagine mutations, sytD-N) had the unintended side effect of mimicking Ca2+ binding, raising the possibility that the increase in asynchronous release was directly caused by ostensibly constitutive Ca2+ binding. Thus, rather than modulating an asynchronous sensor, sytD-N may be mimicking one. To directly test the C2A inhibition hypothesis, we utilized an alternate C2A mutation that we designed to block Ca2+ binding without mimicking it (an aspartate to glutamate mutation, sytD-E). Analysis of both the original sytD-N mutation and our alternate sytD-E mutation at the Drosophila neuromuscular junction showed differential effects on asynchronous release, as well as on synchronous release and the frequency of spontaneous release. Importantly, we found that asynchronous release is not increased in the sytD-E mutant. Thus, our work provides new mechanistic insight into synaptotagmin 1 function during Ca2+-evoked synaptic transmission and demonstrates that Ca2+ binding by the C2A domain of synaptotagmin 1 does not inhibit asynchronous neurotransmitter release in vivo.


Assuntos
Proteínas de Drosophila/metabolismo , Neurotransmissores/metabolismo , Sinaptotagmina I/metabolismo , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados , Sítios de Ligação/genética , Cálcio/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Genes de Insetos , Mutagênese Sítio-Dirigida , Domínios Proteicos , Transmissão Sináptica , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/química , Sinaptotagmina I/genética
13.
Neuron ; 107(1): 52-64.e7, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32362337

RESUMO

At neuronal synapses, synaptotagmin-1 (syt1) acts as a Ca2+ sensor that synchronizes neurotransmitter release with Ca2+ influx during action potential firing. Heterozygous missense mutations in syt1 have recently been associated with a severe but heterogeneous developmental syndrome, termed syt1-associated neurodevelopmental disorder. Well-defined pathogenic mechanisms, and the basis for phenotypic heterogeneity in this disorder, remain unknown. Here, we report the clinical, physiological, and biophysical characterization of three syt1 mutations from human patients. Synaptic transmission was impaired in neurons expressing mutant variants, which demonstrated potent, graded dominant-negative effects. Biophysical interrogation of the mutant variants revealed novel mechanistic features concerning the cooperative action, and functional specialization, of the tandem Ca2+-sensing domains of syt1. These mechanistic studies led to the discovery that a clinically approved K+ channel antagonist is able to rescue the dominant-negative heterozygous phenotype. Our results establish a molecular cause, basis for phenotypic heterogeneity, and potential treatment approach for syt1-associated neurodevelopmental disorder.


Assuntos
Transtornos do Neurodesenvolvimento/genética , Neurônios/fisiologia , Transmissão Sináptica/genética , Sinaptotagmina I/genética , 4-Aminopiridina/farmacologia , Animais , Células Cultivadas , Humanos , Camundongos , Transtornos do Neurodesenvolvimento/fisiopatologia , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Sinaptotagmina I/química
14.
Metallomics ; 12(2): 164-172, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32051983

RESUMO

Pb2+ is a xenobiotic metal ion that competes for Ca2+-binding sites in proteins. Using the peripheral Ca2+-sensing domains of Syt1, we show that the chelating pH buffer Bis-Tris enables identification and functional characterization of high-affinity Pb2+ sites that are likely to be targeted by bioavailable Pb2+.


Assuntos
Chumbo/química , Sinaptotagmina I/química , Xenobióticos/química , Sítios de Ligação , Ligação Competitiva , Soluções Tampão , Cálcio/química , Membrana Celular/química , Concentração de Íons de Hidrogênio , Ligação Proteica , Domínios Proteicos
15.
Biochem Biophys Res Commun ; 521(1): 145-151, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31629470

RESUMO

Despite decades of intensive studies, the failure to identify plasmodesmata (PD) localization sequences has constrained our understanding of Tobacco mosaic virus (TMV) movement. Recently, we identified the first PD localization signal (major PLS) in the TMV movement protein (MP), which encompasses the first 50 amino acid residues of the MP. Although the major PLS is sufficient for PD targeting, the efficiency is lower than the full-length TMV MP. To address this efficiency gap, we identified two additional PLS domains encompassing amino acid residues 61 to 80, and 147 to 170 of the MP and showed that these two domains target to PD, but do not transit to adjacent cells. We also demonstrated that the MP61-80 fragment interacts with Arabidopsis synaptotagmin A, which was also shown to interact with the major TMV MP PLS. Therefore, our findings have provided new insights to more fully understand the mechanism underlying plasmodesmal targeting of TMV MP.


Assuntos
Proteínas do Movimento Viral em Plantas/metabolismo , Plasmodesmos/química , Vírus do Mosaico do Tabaco/química , Arabidopsis/química , Arabidopsis/crescimento & desenvolvimento , Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Proteínas do Movimento Viral em Plantas/química , Plasmodesmos/metabolismo , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo , Vírus do Mosaico do Tabaco/metabolismo
16.
Nat Commun ; 10(1): 4076, 2019 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-31501440

RESUMO

Synaptic vesicle (SV) exocytosis is mediated by SNARE proteins. Reconstituted SNAREs are constitutively active, so a major focus has been to identify fusion clamps that regulate their activity in synapses: the primary candidates are synaptotagmin (syt) 1 and complexin I/II. Syt1 is a Ca2+ sensor for SV release that binds Ca2+ via tandem C2-domains, C2A and C2B. Here, we first determined whether these C2-domains execute distinct functions. Remarkably, the C2B domain profoundly clamped all forms of SV fusion, despite synchronizing residual evoked release and rescuing the readily-releasable pool. Release was strongly enhanced by an adjacent C2A domain, and by the concurrent binding of complexin to trans-SNARE complexes. Knockdown of complexin had no impact on C2B-mediated clamping of fusion. We postulate that the C2B domain of syt1, independent of complexin, is the molecular clamp that arrests SVs prior to Ca2+-triggered fusion.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Mamíferos/metabolismo , Fusão de Membrana , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/metabolismo , Animais , Cálcio/metabolismo , Camundongos Knockout , Mutagênese , Domínios Proteicos , Proteínas SNARE/metabolismo , Transmissão Sináptica , Sinaptotagmina I/química
17.
Proc Natl Acad Sci U S A ; 116(36): 18098-18108, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31431523

RESUMO

Botulinum neurotoxin type B (BoNT/B) recognizes nerve terminals by binding to 2 receptor components: a polysialoganglioside, predominantly GT1b, and synaptotagmin 1/2. It is widely thought that BoNT/B initially binds to GT1b then diffuses in the plane of the membrane to interact with synaptotagmin. We have addressed the hypothesis that a GT1b-synaptotagmin cis complex forms the BoNT/B receptor. We identified a consensus glycosphingolipid-binding motif in the extracellular juxtamembrane domain of synaptotagmins 1/2 and confirmed by Langmuir monolayer, surface plasmon resonance, and circular dichroism that GT1b interacts with synaptotagmin peptides containing this sequence, inducing α-helical structure. Molecular modeling and tryptophan fluorescence spectroscopy were consistent with the intertwining of GT1b and synaptotagmin, involving cis interactions between the oligosaccharide and ceramide moieties of GT1b and the juxtamembrane and transmembrane domains of synaptotagmin, respectively. Furthermore, a point mutation on synaptotagmin, located outside of the BoNT/B-binding segment, inhibited GT1b binding and blocked GT1b-induced potentiation of BoNT/B binding to synaptotagmin-expressing cells. Our findings are consistent with a model in which a preassembled GT1b-synaptotagmin complex constitutes the high-affinity BoNT/B receptor.


Assuntos
Toxinas Botulínicas Tipo A , Gangliosídeos , Sinaptotagmina I , Animais , Sítios de Ligação , Toxinas Botulínicas Tipo A/química , Toxinas Botulínicas Tipo A/metabolismo , Gangliosídeos/química , Gangliosídeos/farmacologia , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Ratos , Sinaptotagmina I/química , Sinaptotagmina I/genética , Sinaptotagmina I/metabolismo , Sinaptotagmina II/química , Sinaptotagmina II/genética , Sinaptotagmina II/metabolismo
18.
Biophys J ; 117(2): 247-257, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31301806

RESUMO

Synaptotagmin 1 acts as the Ca2+ sensor for synchronous neurotransmitter release; however, the mechanism by which it functions is not understood and is presently a topic of considerable interest. Here, we describe measurements on full-length membrane-reconstituted synaptotagmin 1 using site-directed spin labeling in which we characterize the linker region as well as the cis (vesicle membrane) and trans (cytoplasmic membrane) binding of its two C2 domains. In the full-length protein, the C2A domain does not undergo membrane insertion in the absence of Ca2+; however, the C2B domain will bind to and penetrate in trans to a membrane containing phosphatidylinositol 4,5 bisphosphate, even if phosphatidylserine (PS) is present in the cis membrane. In the presence of Ca2+, the Ca2+ binding loops of C2A and C2B both insert into the membrane interface; moreover, C2A preferentially inserts into PS-containing bilayers and will bind in a cis configuration to membranes containing PS even if a phosphatidylinositol 4,5 bisphosphate membrane is presented in trans. The data are consistent with a bridging activity for synaptotagmin 1 in which the two domains bind to opposing vesicle and plasma membranes. The failure of C2A to bind membranes in the absence of Ca2+ and the long unstructured segment linking C2A to the vesicle membrane indicates that synaptotagmin 1 could act to significantly shorten the vesicle-plasma membrane distance with increasing levels of Ca2+.


Assuntos
Fosfatidilinositol 4,5-Difosfato/metabolismo , Sinaptotagmina I/metabolismo , Animais , Cálcio/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Bicamadas Lipídicas/metabolismo , Modelos Biológicos , Modelos Moleculares , Polieletrólitos/química , Domínios Proteicos , Multimerização Proteica , Ratos , Eletricidade Estática , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/química
19.
Acta Crystallogr D Struct Biol ; 75(Pt 2): 234-241, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30821711

RESUMO

Processing X-ray free-electron laser (XFEL) diffraction images poses challenges, as an XFEL pulse is powerful enough to destroy or damage the diffracting volume and thereby yields only one diffraction image per volume. Moreover, the crystal is stationary during the femtosecond pulse, so reflections are generally only partially recorded. Therefore, each XFEL diffraction image must be scaled individually and, ideally, corrected for partiality prior to merging. An additional complication may arise owing to indexing ambiguities when the symmetry of the Bravais lattice is higher than that of the space group, or when the unit-cell dimensions are similar to each other. Here, an automated method is presented that diagnoses these indexing ambiguities based on the Brehm-Diederichs algorithm [Brehm & Diederichs (2014), Acta Cryst. D70, 101-109] and produces a consistent indexing choice for the large majority of diffraction images. This method was applied to an XFEL diffraction data set measured from crystals of the neuronal SNARE-complexin-1-synaptotagmin-1 complex. After correcting the indexing ambiguities, substantial improvements were observed in the merging statistics and the atomic model refinement R values. This method should be a useful addition to the arsenal of tools for the processing of XFEL diffraction data sets.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/química , Coleta de Dados/métodos , Proteínas do Tecido Nervoso/química , Proteínas SNARE/química , Sinaptotagmina I/química , Difração de Raios X/métodos , Algoritmos , Animais , Elétrons , Lasers , Modelos Moleculares , Ratos , Síncrotrons
20.
Biophys J ; 116(6): 1025-1036, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30795874

RESUMO

Synaptotagmin-1 (Syt-1) and synaptotagmin-7 (Syt-7) contain analogous tandem C2 domains, C2A and C2B, which together sense Ca2+ to bind membranes and promote the stabilization of exocytotic fusion pores. Syt-1 triggers fast release of neurotransmitters, whereas Syt-7 functions in processes that involve lower Ca2+ concentrations such as hormone secretion. Syt-1 C2 domains are reported to bind membranes cooperatively, based on the observation that they penetrate farther into membranes as the C2AB tandem than as individual C2 domains. In contrast, we previously suggested that the two C2 domains of Syt-7 bind membranes independently, based in part on measurements of their liposome dissociation kinetics. Here, we investigated C2A-C2B interdomain cooperativity with Syt-1 and Syt-7 using directly comparable measurements. Equilibrium Ca2+ titrations demonstrate that the Syt-7 C2AB tandem binds liposomes lacking phosphatidylinositol-4,5-bisphosphate (PIP2) with greater Ca2+ sensitivity than either of its individual domains and binds to membranes containing PIP2 even in the absence of Ca2+. Stopped-flow kinetic measurements show differences in cooperativity between Syt-1 and Syt-7: Syt-1 C2AB dissociates from PIP2-free liposomes much more slowly than either of its individual C2 domains, indicating cooperativity, whereas the major population of Syt-7 C2AB has a dissociation rate comparable to its C2A domain, suggesting a lack of cooperativity. A minor subpopulation of Syt-7 C2AB dissociates at a slower rate, which could be due to a small cooperative component and/or liposome clustering. Measurements using an environment-sensitive fluorescent probe indicate that the Syt-7 C2B domain inserts deeply into membranes as part of the C2AB tandem, similar to the coinsertion previously reported for Syt-1. Overall, coinsertion of C2A and C2B domains is coupled to cooperative energetic effects in Syt-1 to a much greater extent than in Syt-7. The difference can be understood in terms of the relative contributions of C2A and C2B domains toward membrane binding in the two proteins.


Assuntos
Membrana Celular/metabolismo , Sinaptotagmina I/química , Sinaptotagmina I/metabolismo , Sinaptotagminas/química , Sinaptotagminas/metabolismo , Cálcio/metabolismo , Humanos , Cinética , Lipossomos/metabolismo , Ligação Proteica , Domínios Proteicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...