Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 629(8014): 1062-1068, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38720082

RESUMEN

Most chemistry and biology occurs in solution, in which conformational dynamics and complexation underlie behaviour and function. Single-molecule techniques1 are uniquely suited to resolving molecular diversity and new label-free approaches are reshaping the power of single-molecule measurements. A label-free single-molecule method2-16 capable of revealing details of molecular conformation in solution17,18 would allow a new microscopic perspective of unprecedented detail. Here we use the enhanced light-molecule interactions in high-finesse fibre-based Fabry-Pérot microcavities19-21 to detect individual biomolecules as small as 1.2 kDa, a ten-amino-acid peptide, with signal-to-noise ratios (SNRs) >100, even as the molecules are unlabelled and freely diffusing in solution. Our method delivers 2D intensity and temporal profiles, enabling the distinction of subpopulations in mixed samples. Notably, we observe a linear relationship between passage time and molecular radius, unlocking the potential to gather crucial information about diffusion and solution-phase conformation. Furthermore, mixtures of biomolecule isomers of the same molecular weight and composition but different conformation can also be resolved. Detection is based on the creation of a new molecular velocity filter window and a dynamic thermal priming mechanism that make use of the interplay between optical and thermal dynamics22,23 and Pound-Drever-Hall (PDH) cavity locking24 to reveal molecular motion even while suppressing environmental noise. New in vitro ways of revealing molecular conformation, diversity and dynamics can find broad potential for applications in the life and chemical sciences.


Asunto(s)
Péptidos , Imagen Individual de Molécula , Difusión , Isomerismo , Luz , Péptidos/análisis , Péptidos/química , Péptidos/efectos de la radiación , Relación Señal-Ruido , Imagen Individual de Molécula/métodos , Soluciones , Conformación Proteica , Peso Molecular , Movimiento (Física)
2.
Cell ; 147(3): 666-77, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036572

RESUMEN

Synaptic transmission involves a fast synchronous phase and a slower asynchronous phase of neurotransmitter release that are regulated by distinct Ca(2+) sensors. Though the Ca(2+) sensor for rapid exocytosis, synaptotagmin I, has been studied in depth, the sensor for asynchronous release remains unknown. In a screen for neuronal Ca(2+) sensors that respond to changes in [Ca(2+)] with markedly slower kinetics than synaptotagmin I, we observed that Doc2--another Ca(2+), SNARE, and lipid-binding protein--operates on timescales consistent with asynchronous release. Moreover, up- and downregulation of Doc2 expression levels in hippocampal neurons increased or decreased, respectively, the slow phase of synaptic transmission. Synchronous release, when triggered by single action potentials, was unaffected by manipulation of Doc2 but was enhanced during repetitive stimulation in Doc2 knockdown neurons, potentially due to greater vesicle availability. In summary, we propose that Doc2 is a Ca(2+) sensor that is kinetically tuned to regulate asynchronous neurotransmitter release.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo , Transmisión Sináptica , Potenciales de Acción , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Metabolismo de los Lípidos , Ratones , Neuronas/metabolismo , Ratas , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/genética , Sinaptotagmina I/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(40): e2210649119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161950

RESUMEN

Molecular pathways that intrinsically regulate neuronal maintenance are poorly understood, but rare pathogenic mutations that underlie neurodegenerative disease can offer important insights into the mechanisms that facilitate lifelong neuronal function. Here, we leverage a rat model to demonstrate directly that the TFG p.R106C variant implicated previously in complicated forms of hereditary spastic paraplegia (HSP) underlies progressive spastic paraparesis with accompanying ventriculomegaly and thinning of the corpus callosum, consistent with disease phenotypes identified in adolescent patients. Analyses of primary cortical neurons obtained from CRISPR-Cas9-edited animals reveal a kinetic delay in biosynthetic secretory protein transport from the endoplasmic reticulum (ER), in agreement with prior induced pluripotent stem cell-based studies. Moreover, we identify an unexpected role for TFG in the trafficking of Rab4A-positive recycling endosomes specifically within axons and dendrites. Impaired TFG function compromises the transport of at least a subset of endosomal cargoes, which we show results in down-regulated inhibitory receptor signaling that may contribute to excitation-inhibition imbalances. In contrast, the morphology and trafficking of other organelles, including mitochondria and lysosomes, are unaffected by the TFG p.R106C mutation. Our findings demonstrate a multifaceted role for TFG in secretory and endosomal protein sorting that is unique to cells of the central nervous system and highlight the importance of these pathways to maintenance of corticospinal tract motor neurons.


Asunto(s)
Endosomas , Neuronas Motoras , Transporte de Proteínas , Animales , Ratas , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Neuronas Motoras/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas/metabolismo , Paraplejía Espástica Hereditaria/metabolismo
4.
J Neurosci ; 43(19): 3421-3438, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-36997314

RESUMEN

Mitochondria exert powerful control over cellular physiology, contributing to ion homeostasis, energy production, and metabolite biosynthesis. The trafficking and function of these organelles are particularly important in neurons, with impaired mitochondrial function or altered morphology observed in every neurodegenerative disorder studied. While mitochondrial biosynthetic products play a crucial role in maintaining cellular function, their resulting byproducts can have negative consequences. Thus, organelle quality control (QC) mechanisms that maintain mitochondrial function are imperative to restrict destructive signaling cascades in the cell. Axons are particularly sensitive to damage, and there is little consensus regarding the mechanisms that mediate mitochondrial QC in this compartment. Here, we first investigated the unstressed behavior of mitochondria in rat hippocampal neurons of mixed sex, focusing on mitochondrial trafficking and fusion to better understand potential QC mechanisms. We observed size and redox asymmetry of mitochondrial traffic in axons, suggesting an active QC mechanism in this compartment. We also document biochemical complementation upon the fusion and fission of axonal mitochondria. Eliminating fusion by knocking down the neuronal mitochondrial fusion protein mitofusin 2 (MFN2) reduced the rates of axonal mitochondrial trafficking and fusion, decreased the levels of synaptic vesicle (SV) proteins, inhibited exocytosis, and impaired SV recruitment from the reserve pool during extended stimulation. MFN2 knockdown also resulted in presynaptic Ca2+ dyshomeostasis. Remarkably, upon MFN2 knockdown, presynaptic mitochondria sequestered Ca2+ more efficiently, effectively limiting presynaptic Ca2+ transients during stimulation. These results support an active mitochondrial trafficking and fusion-related QC process that supports presynaptic Ca2+ handling and the SV cycle.SIGNIFICANCE STATEMENT Decreased or altered mitochondrial function is observed in many disease states. All neurodegenerative diseases co-present with some sort of mitochondrial abnormality. Therefore, identifying quality control mechanisms that sustain the mitochondrial network in neurons, and particularly in axons, is of significant interest. The response of axonal mitochondria to acutely applied toxins or injury has been studied in detail. Although informative, the response of neurons to these insults might not be physiologically relevant, so it is crucial to also study the basal behavior of axonal mitochondria. Here, we use fluorescent biosensors to investigate the mitochondrial network in neurons and examine the role of mitofusin 2 in maintaining the axonal mitochondrial network and in supporting the synaptic vesicle cycle.


Asunto(s)
Axones , Vesículas Sinápticas , Animales , Ratas , Transporte Axonal/fisiología , Axones/metabolismo , Hipocampo/metabolismo , Homeostasis , Mitocondrias/metabolismo , Vesículas Sinápticas/metabolismo
5.
J Neurosci ; 43(9): 1475-1491, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36732068

RESUMEN

Synaptotagmin 9 (SYT9) is a tandem C2 domain Ca2+ sensor for exocytosis in neuroendocrine cells; its function in neurons remains unclear. Here, we show that, in mixed-sex cultures, SYT9 does not trigger rapid synaptic vesicle exocytosis in mouse cortical, hippocampal, or striatal neurons, unless it is massively overexpressed. In striatal neurons, loss of SYT9 reduced the frequency of spontaneous neurotransmitter release events (minis). We delved into the underlying mechanism and discovered that SYT9 was localized to dense-core vesicles that contain substance P (SP). Loss of SYT9 impaired SP release, causing the observed decrease in mini frequency. This model is further supported by loss of function mutants. Namely, Ca2+ binding to the C2A domain of SYT9 triggered membrane fusion in vitro, and mutations that disrupted this activity abolished the ability of SYT9 to regulate both SP release and mini frequency. We conclude that SYT9 indirectly regulates synaptic transmission in striatal neurons by controlling SP release.SIGNIFICANCE STATEMENT Synaptotagmin 9 (SYT9) has been described as a Ca2+ sensor for dense-core vesicle (DCV) exocytosis in neuroendocrine cells, but its role in neurons remains unclear, despite widespread expression in the brain. This article examines the role of SYT9 in synaptic transmission across cultured cortical, hippocampal, and striatal neuronal preparations. We found that SYT9 regulates spontaneous neurotransmitter release in striatal neurons by serving as a Ca2+ sensor for the release of the neuromodulator substance P from DCVs. This demonstrates a novel role for SYT9 in neurons and uncovers a new field of study into neuromodulation by SYT9, a protein that is widely expressed in the brain.


Asunto(s)
Sustancia P , Vesículas Sinápticas , Animales , Ratones , Sinaptotagminas/metabolismo , Sustancia P/metabolismo , Vesículas Sinápticas/metabolismo , Transmisión Sináptica/fisiología , Neuronas/metabolismo , Exocitosis , Neurotransmisores/metabolismo , Sinaptotagmina I/metabolismo , Calcio/metabolismo
6.
Cell ; 138(4): 709-21, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19703397

RESUMEN

Decades ago it was proposed that exocytosis involves invagination of the target membrane, resulting in a highly localized site of contact between the bilayers destined to fuse. The vesicle protein synaptotagmin-I (syt) bends membranes in response to Ca(2+), but whether this drives localized invagination of the target membrane to accelerate fusion has not been determined. Previous studies relied on reconstituted vesicles that were already highly curved and used mutations in syt that were not selective for membrane-bending activity. Here, we directly address this question by utilizing vesicles with different degrees of curvature. A tubulation-defective syt mutant was able to promote fusion between highly curved SNARE-bearing liposomes but exhibited a marked loss of activity when the membranes were relatively flat. Moreover, bending of flat membranes by adding an N-BAR domain rescued the function of the tubulation-deficient syt mutant. Hence, syt-mediated membrane bending is a critical step in membrane fusion.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Sinaptotagminas/metabolismo , Animales , Química Encefálica , Exocitosis , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Liposomas/metabolismo , Mutación , Proteínas SNARE/metabolismo
7.
Nature ; 554(7691): 260-263, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29420480

RESUMEN

The fusion pore is the first crucial intermediate formed during exocytosis, yet little is known about the mechanisms that determine the size and kinetic properties of these transient structures. Here, we reduced the number of available SNAREs (proteins that mediate vesicle fusion) in neurons and observed changes in transmitter release that are suggestive of alterations in fusion pores. To investigate these changes, we employed reconstituted fusion assays using nanodiscs to trap pores in their initial open state. Optical measurements revealed that increasing the number of SNARE complexes enhanced the rate of release from single pores and enabled the escape of larger cargoes. To determine whether this effect was due to changes in nascent pore size or to changes in stability, we developed an approach that uses nanodiscs and planar lipid bilayer electrophysiology to afford microsecond resolution at the single event level. Both pore size and stability were affected by SNARE copy number. Increasing the number of vesicle (v)-SNAREs per nanodisc from three to five caused a twofold increase in pore size and decreased the rate of pore closure by more than three orders of magnitude. Moreover, pairing of v-SNAREs and target (t)-SNAREs to form trans-SNARE complexes was highly dynamic: flickering nascent pores closed upon addition of a v-SNARE fragment, revealing that the fully assembled, stable SNARE complex does not form at this stage of exocytosis. Finally, a deletion at the base of the SNARE complex, which mimics the action of botulinum neurotoxin A, markedly reduced fusion pore stability. In summary, trans-SNARE complexes are dynamic, and the number of SNAREs recruited to drive fusion determines fundamental properties of individual pores.


Asunto(s)
Membrana Celular/metabolismo , Exocitosis , Fusión de Membrana , Porosidad , Proteínas SNARE/metabolismo , Animales , Toxinas Botulínicas Tipo A/metabolismo , Potenciales Postsinápticos Excitadores , Membrana Dobles de Lípidos/metabolismo , Ratones , Neuronas/citología , Neuronas/metabolismo , Neurotransmisores/metabolismo , Ratas , Ratas Sprague-Dawley , Vesículas Secretoras/metabolismo
8.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34810248

RESUMEN

Synaptotagmin 1 (syt1) is a Ca2+ sensor that regulates synaptic vesicle exocytosis. Cell-based experiments suggest that syt1 functions as a multimer; however, biochemical and electron microscopy studies have yielded contradictory findings regarding putative self-association. Here, we performed dynamic light scattering on syt1 in solution, followed by electron microscopy, and we used atomic force microscopy to study syt1 self-association on supported lipid bilayers under aqueous conditions. Ring-like multimers were clearly observed. Multimerization was enhanced by Ca2+ and required anionic phospholipids. Large ring-like structures (∼180 nm) were reduced to smaller rings (∼30 nm) upon neutralization of a cluster of juxtamembrane lysine residues; further substitution of residues in the second C2-domain completely abolished self-association. When expressed in neurons, syt1 mutants with graded reductions in self-association activity exhibited concomitant reductions in 1) clamping spontaneous release and 2) triggering and synchronizing evoked release. Thus, the juxtamembrane linker of syt1 plays a crucial role in exocytosis by mediating multimerization.


Asunto(s)
Neurotransmisores/metabolismo , Animales , Calcio/metabolismo , Citoplasma/metabolismo , Electrofisiología , Exocitosis , Técnicas In Vitro , Luz , Membrana Dobles de Lípidos/química , Lípidos/química , Lisina/química , Fusión de Membrana , Microscopía de Fuerza Atómica , Neuronas/metabolismo , Fosfolípidos/química , Terminales Presinápticos/metabolismo , Dominios Proteicos , Multimerización de Proteína , Proteínas Recombinantes/metabolismo , Dispersión de Radiación , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/metabolismo
9.
J Neurosci ; 42(30): 5816-5829, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35701163

RESUMEN

Synaptotagmin-1 (Syt1) is a vesicular calcium sensor required for synchronous neurotransmitter release, composed of a single-pass transmembrane domain linked to two C2 domains (C2A and C2B) that bind calcium, acidic lipids, and SNARE proteins that drive fusion of the synaptic vesicle with the plasma membrane. Despite its essential role, how Syt1 couples calcium entry to synchronous release is poorly understood. Calcium binding to C2B is critical for synchronous release, and C2B additionally binds the SNARE complex. The C2A domain is also required for Syt1 function, but it is not clear why. Here, we asked what critical feature of C2A may be responsible for its functional role and compared this to the analogous feature in C2B. We focused on highly conserved poly-lysine patches located on the sides of C2A (K189-192) and C2B (K324-327). We tested effects of charge-neutralization mutations in either region (Syt1K189-192A and Syt1K326-327A) side by side to determine their relative contributions to Syt1 function in cultured cortical neurons from mice of either sex and in single-molecule experiments. Combining electrophysiological recordings and optical tweezers measurements to probe dynamic single C2 domain-membrane interactions, we show that both C2A and C2B polybasic patches contribute to membrane binding, and both are required for evoked release. The size of the readily releasable vesicle pool and the rate of spontaneous release were unaffected, so both patches are likely required specifically for synchronization of release. We suggest these patches contribute to cooperative membrane binding, increasing the overall affinity of Syt1 for negatively charged membranes and facilitating evoked release.SIGNIFICANCE STATEMENT Synaptotagmin-1 is a vesicular calcium sensor required for synchronous neurotransmitter release. Its tandem cytosolic C2 domains (C2A and C2B) bind calcium, acidic lipids, and SNARE proteins that drive fusion of the synaptic vesicle with the plasma membrane. How calcium binding to Synaptotagmin-1 leads to release and the relative contributions of the C2 domains are unclear. Combining electrophysiological recordings from cultured neurons and optical tweezers measurements of single C2 domain-membrane interactions, we show that conserved polybasic regions in both domains contribute to membrane binding cooperatively, and both are required for evoked release, likely by increasing the overall affinity of Synaptotagmin-1 for acidic membranes.


Asunto(s)
Dominios C2 , Calcio , Neurotransmisores , Sinaptotagmina I , Animales , Calcio/metabolismo , Lípidos , Ratones , Neurotransmisores/metabolismo , Proteínas SNARE/metabolismo , Sinaptotagmina I/genética , Sinaptotagmina I/metabolismo
10.
J Neurosci ; 42(17): 3512-3522, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35296545

RESUMEN

Current methods to isolate synaptic vesicles (SVs), the organellar quanta of synaptic transmission, require highly specialized materials and up to 24 h. These technical obstacles have thus far limited the study of SVs in models of synaptic function and pathophysiology. Here, we describe techniques for the rapid isolation of SVs by immunoprecipitation with widely available antibodies conjugated to magnetic beads. We report that the inexpensive rho1D4 monoclonal antibody binds SVs and show that elution with the 1D4 peptide yields native vesicles that are ≥ 10-fold purer than those obtained with classical techniques. These methods substantially widen the accessibility of SVs, enabling their purification in 60-90 min for downstream analyses including mass spectrometry and cryo-electron microscopy. Immunopurified SV preparations from mouse brain contained apolipoprotein E, the LDL receptor Lrp1, and enzymes involved in lipid metabolism, suggesting that SVs may play direct roles in lipid homeostasis and lipoprotein trafficking at the nerve terminal.SIGNIFICANCE STATEMENT SVs are small organelles that form and recycle at nerve terminals to enable synaptic transmission. Much remains unknown about the processes that enable the formation and function of SVs. Moreover, nerve terminals appear to be particularly vulnerable to pathophysiologic processes underlying neurodegenerative diseases and schizophrenia. Although techniques to purify synaptic vesicles thus have the potential to yield significant insights into physiology and pathophysiology of nerve terminals, current methods rely on either esoteric materials or expression of transgenes. This article addresses these problems by establishing robust, efficient methods for SV purification using widely available materials, and it highlights several promising areas of future study arising from proteomic analyses of immunopurified SVs.


Asunto(s)
Proteómica , Vesículas Sinápticas , Animales , Encéfalo/metabolismo , Microscopía por Crioelectrón , Ratones , Transmisión Sináptica , Vesículas Sinápticas/metabolismo
11.
J Physiol ; 600(3): 645-654, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34957569

RESUMEN

SV2A, an essential transporter-like synaptic vesicle protein, is a major target for antiepileptic drugs and a receptor for clostridial neurotoxins including Botox. While SV2A is required for normal levels of evoked neurotransmitter release, the mechanism underlying this role remains unclear. Here, we introduce a new chemogenetic approach for all-optical monitoring of excitation-secretion coupling, and we demonstrate its use in characterizing the SV2A knockout (KO) phenotype in cultured hippocampal neurons. This method employs the HaloTag system to target a robust small-molecule Ca2+ indicator, JF646 -BAPTA, to the presynaptic compartment. The far-red fluorescence of this indicator enables multiplexing with the fluorescent glutamate sensor iGluSnFR for detection of presynaptic Ca2+ influx and glutamate release at the same axonal boutons. Evoked glutamate release probability was reduced in SV2A KO neurons without a change in presynaptic Ca2+ entry, suggesting that SV2A supports vesicle fusion by increasing the functional availability, or efficiency, of the Ca2+ -regulated membrane fusion machinery. KEY POINTS: One of the most prescribed antiepileptic medications, levetiracetam, acts by binding a protein of uncertain molecular function. This transporter-like protein, SV2A, is trafficked to synaptic vesicles and acts to support neurotransmitter release, but the mechanism underlying this function has not been determined In this study, we sought to establish whether SV2A changes Ca2+ signalling at nerve terminals, which is a key regulatory system for synaptic vesicle exocytosis. To do so, we adapted new chemogenetic tools to perform all-optical measurements of presynaptic Ca2+ and glutamate release in neurons lacking SV2A. Our measurements showed that loss of SV2A reduces glutamate release without reducing Ca2+ influx at hippocampal nerve terminals, demonstrating that SV2A increases the likelihood that Ca2+ will trigger synaptic vesicle fusion.


Asunto(s)
Terminales Presinápticos , Vesículas Sinápticas , Exocitosis , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo
12.
Nat Methods ; 16(2): 206, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30602783

RESUMEN

In the version of this paper originally published, important figure labels in Fig. 3d were not visible. An image layer present in the authors' original figure that included two small dashed outlines and text labels indicating ROI 1 and ROI 2, as well as a scale bar and the name of the cell label, was erroneously altered during image processing. The figure has been corrected in the HTML and PDF versions of the paper.

13.
Nat Methods ; 16(4): 351, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30820033

RESUMEN

The version of this paper originally published cited a preprint version of ref. 12 instead of the published version (Proc. Natl. Acad. Sci. USA 115, 5594-5599; 2018), which was available before this Nature Methods paper went to press. The reference information has been updated in the PDF and HTML versions of the article.

14.
Proc Natl Acad Sci U S A ; 116(23): 11113-11115, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31110017

RESUMEN

The function of α-synuclein (α-syn) has been long debated, and two seemingly divergent views have emerged. In one, α-syn binds to VAMP2, acting as a SNARE chaperone-but with no effect on neurotransmission-while another posits that α-syn attenuates neurotransmitter release by restricting synaptic vesicle mobilization and recycling. Here, we show that α-syn-VAMP2 interactions are necessary for α-syn-induced synaptic attenuation. Our data connect divergent views and suggest a unified model of α-syn function.


Asunto(s)
Vesículas Sinápticas/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , alfa-Sinucleína/metabolismo , Transporte Biológico/fisiología , Humanos , Neuronas/metabolismo , Proteínas SNARE/metabolismo , Transmisión Sináptica/inmunología
15.
Biophys J ; 120(8): 1367-1377, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33582136

RESUMEN

SNARE-mediated membrane fusion proceeds via the formation of a fusion pore. This intermediate structure is highly dynamic and can flicker between open and closed states. In cells, cholesterol has been reported to affect SNARE-mediated exocytosis and fusion pore dynamics. Here, we address the question of whether cholesterol directly affects the flickering rate of reconstituted fusion pores in vitro. These experiments were enabled by the recent development of a nanodisc⋅black lipid membrane recording system that monitors dynamic transitions between the open and closed states of nascent recombinant pores with submillisecond time resolution. The fusion pores formed between nanodiscs that bore the vesicular SNARE synaptobrevin 2 and black lipid membranes that harbored the target membrane SNAREs syntaxin 1A and SNAP-25B were markedly affected by cholesterol. These effects include strong reductions in flickering out of the open state, resulting in a significant increase in the open dwell-time. We attributed these effects to the known role of cholesterol in altering the elastic properties of lipid bilayers because manipulation of phospholipids to increase membrane stiffness mirrored the effects of cholesterol. In contrast to the observed effects on pore kinetics, cholesterol had no effect on the current that passed through individual pores and, hence, did not affect pore size. In conclusion, our results show that cholesterol dramatically stabilizes fusion pores in the open state by increasing membrane bending rigidity.


Asunto(s)
Fusión de Membrana , Proteínas SNARE , Colesterol , Exocitosis , Proteína 2 de Membrana Asociada a Vesículas
16.
J Am Chem Soc ; 143(8): 3219-3230, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33611913

RESUMEN

Amphiphilic nylon-3 polymers have been reported to mimic the biological activities of natural antimicrobial peptides, with high potency against bacteria and minimal toxicity toward eukaryotic cells. Amphiphilic balance, determined by the proportions of hydrophilic and lipophilic subunits, is considered one of the most important features for achieving this activity profile for nylon-3 polymers and many other antimicrobial polymers. Insufficient hydrophobicity often correlates with weak activities against bacteria, whereas excessive hydrophobicity correlates with high toxicity toward eukaryotic cells. To ask whether factors beyond amphiphilic balance influence polymer activities, we synthesized and evaluated new nylon-3 polymers with two stereoisomeric subunits, each bearing an ethyl side chain and an aminomethyl side chain. Subunits that differ only in stereochemistry are predicted to contribute equally to amphiphilic balance, but we observed that the stereochemical difference correlates with significant changes in biological activity profile. Antibacterial activities were not strongly affected by subunit stereochemistry, but the ability to disrupt eukaryotic cell membranes varied considerably. Experiments with planar lipid bilayers and synthetic liposomes suggested that eukaryotic membrane disruption results from polymer-mediated formation of large pores. Collectively, our results suggest that factors other than amphiphilic balance influence the membrane activity profile of synthetic polymers. Subunits that differ in stereochemistry are likely to have distinct conformational propensities, which could potentially lead to differences in the average shapes of polymer chains, even when the subunits are heterochiral. These findings highlight a dimension of polymer design that should be considered more broadly in efforts to improve specificity and efficacy of antimicrobial polymers.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Nylons/química , Modelos Moleculares , Conformación Molecular , Porosidad , Estereoisomerismo
17.
Nat Methods ; 15(11): 936-939, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30377363

RESUMEN

Single-wavelength fluorescent reporters allow visualization of specific neurotransmitters with high spatial and temporal resolution. We report variants of intensity-based glutamate-sensing fluorescent reporter (iGluSnFR) that are functionally brighter; detect submicromolar to millimolar amounts of glutamate; and have blue, cyan, green, or yellow emission profiles. These variants could be imaged in vivo in cases where original iGluSnFR was too dim, resolved glutamate transients in dendritic spines and axonal boutons, and allowed imaging at kilohertz rates.


Asunto(s)
Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Fluorescente/métodos , Neuronas/citología , Retina/citología , Corteza Visual/citología , Animales , Color , Femenino , Hurones , Colorantes Fluorescentes , Ácido Glutámico/análisis , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/metabolismo , Corteza Visual/metabolismo
18.
Proc Natl Acad Sci U S A ; 115(24): E5605-E5613, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29844163

RESUMEN

Various forms of synaptic plasticity underlie aspects of learning and memory. Synaptic augmentation is a form of short-term plasticity characterized by synaptic enhancement that persists for seconds following specific patterns of stimulation. The mechanisms underlying this form of plasticity are unclear but are thought to involve residual presynaptic Ca2+ Here, we report that augmentation was reduced in cultured mouse hippocampal neurons lacking the Ca2+ sensor, Doc2; other forms of short-term enhancement were unaffected. Doc2 binds Ca2+ and munc13 and translocates to the plasma membrane to drive augmentation. The underlying mechanism was not associated with changes in readily releasable pool size or Ca2+ dynamics, but rather resulted from superpriming a subset of synaptic vesicles. Hence, Doc2 forms part of the Ca2+-sensing apparatus for synaptic augmentation via a mechanism that is molecularly distinct from other forms of short-term plasticity.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Células HEK293 , Hipocampo/metabolismo , Humanos , Ratones , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Ratas , Transmisión Sináptica/fisiología
19.
Nano Lett ; 20(8): 6032-6037, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32648765

RESUMEN

Nanodiscs (ND) are soluble phospholipid bilayers bounded by membrane scaffold proteins; they have become invaluable in the study of membrane proteins. However, this multifunctional tool has been used individually, and applications involving multiple NDs and their interactions have fallen far behind their counterpart membrane model system: liposomes. One major obstacle is the lack of reliable methods to manage the spatial arrangement of NDs. Here we sought to extend the utility of NDs by organizing them on DNA origami. NDs constructed with DNA-anchor amphiphiles were placed precisely and specifically into these DNA nanostructures via hybridization. Four different tethering strategies were explored and validated. A variety of geometric patterns of NDs were successfully programmed on origami, as evidenced by electron microscopy. The ND ensembles generated in this study provide new and powerful platforms to study protein-lipid or protein-protein interactions with spatial control of membranes.


Asunto(s)
ADN , Nanoestructuras , Membrana Dobles de Lípidos , Liposomas , Proteínas de la Membrana , Fosfolípidos
20.
J Biol Chem ; 294(28): 10942-10953, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31147445

RESUMEN

Exocytosis mediates the release of neurotransmitters and hormones from neurons and neuroendocrine cells. Tandem C2 domain proteins in the synaptotagmin (syt) and double C2 domain (Doc2) families regulate exocytotic membrane fusion via direct interactions with Ca2+ and phospholipid bilayers. Syt1 is a fast-acting, low-affinity Ca2+ sensor that penetrates membranes upon binding Ca2+ to trigger synchronous vesicle fusion. The closely related Doc2ß is a slow-acting, high-affinity Ca2+ sensor that triggers spontaneous and asynchronous vesicle fusion, but whether it also penetrates membranes is unknown. Both syt1 and Doc2ß bind the dynamically regulated plasma membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2), but it is unclear whether PIP2 serves only as a membrane contact or enables specialized membrane-binding modes by these Ca2+ sensors. Furthermore, it has been shown that PIP2 uncaging can trigger rapid, syt1-dependent exocytosis in the absence of Ca2+ influx, suggesting that current models for the action of these Ca2+ sensors are incomplete. Here, using a series of steady-state and time-resolved fluorescence measurements, we show that Doc2ß, like syt1, penetrates membranes in a Ca2+-dependent manner. Unexpectedly, we observed that PIP2 can drive membrane penetration by both syt1 and Doc2ß in the absence of Ca2+, providing a plausible mechanism for Ca2+-independent, PIP2-dependent exocytosis. Quantitative measurements of penetration depth revealed that, in the presence of Ca2+, PIP2 drives Doc2ß, but not syt1, substantially deeper into the membrane, defining a biophysical regulatory mechanism specific to this high-affinity Ca2+ sensor. Our results provide evidence of a novel role for PIP2 in regulating, and under some circumstances triggering, exocytosis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Sinaptotagmina I/metabolismo , Animales , Dominios C2 , Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Membrana Celular/metabolismo , Exocitosis/fisiología , Fusión de Membrana , Lípidos de la Membrana/metabolismo , Proteínas del Tejido Nervioso/fisiología , Neuronas/metabolismo , Neurotransmisores/metabolismo , Fosfatidilinositol 4,5-Difosfato/fisiología , Fosfatidilinositoles/metabolismo , Unión Proteica , Sinapsis/fisiología , Transmisión Sináptica , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA