Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Elife ; 122024 Mar 27.
Article En | MEDLINE | ID: mdl-38536730

Despite decades of intense study, the molecular basis of asynchronous neurotransmitter release remains enigmatic. Synaptotagmin (syt) 7 and Doc2 have both been proposed as Ca2+ sensors that trigger this mode of exocytosis, but conflicting findings have led to controversy. Here, we demonstrate that at excitatory mouse hippocampal synapses, Doc2α is the major Ca2+ sensor for asynchronous release, while syt7 supports this process through activity-dependent docking of synaptic vesicles. In synapses lacking Doc2α, asynchronous release after single action potentials is strongly reduced, while deleting syt7 has no effect. However, in the absence of syt7, docked vesicles cannot be replenished on millisecond timescales. Consequently, both synchronous and asynchronous release depress from the second pulse onward during repetitive activity. By contrast, synapses lacking Doc2α have normal activity-dependent docking, but continue to exhibit decreased asynchronous release after multiple stimuli. Moreover, disruption of both Ca2+ sensors is non-additive. These findings result in a new model whereby syt7 drives activity-dependent docking, thus providing synaptic vesicles for synchronous (syt1) and asynchronous (Doc2 and other unidentified sensors) release during ongoing transmission.


Synapses , Synaptic Vesicles , Synaptotagmins , Animals , Mice , Action Potentials , Calcium/metabolism , Exocytosis , Neurotransmitter Agents , Synapses/metabolism , Synaptic Transmission , Synaptic Vesicles/metabolism , Synaptotagmin I/metabolism , Synaptotagmins/metabolism , Calcium-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism
2.
Talanta ; 270: 125526, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38091748

Logic gate-based fluorescent probes are powerful tools for the discriminative sensing of multiple signaling molecules that are expressed in concert during the progression of many diseases such as inflammation, cancer, aging, and other disorders. To achieve logical sensing, multiple functional groups are introduced to the different substitution sites of a single fluorescent dye, which increases the complexity of chemical synthesis. Herein, we report a simple strategy that incorporates just one responsive unit into a hemicyanine dye achieving the logic gate-based sensing of two independent analytes. We introduce boronic acid to hemicyanine to quench the fluorescence, and in the presence of hydrogen peroxide (H2O2), the fluorescence is recovered due to removal of the boronate. Interestingly, the subsequent decrease in pH turned the red fluorescence of hemicyanine to green emissive because of protonation of the phenolic alcohol. This unique feature of the probe enables us to construct "INHIBIT" and "AND" logical gates for the accurate measuring of intracellular H2O2 and acidic pH in tandem. This study offers insight into the simple construction of logic-gate based fluorescent probes for the tandem sensing of multiple analytes that are correlatively produced during disease progression.


Fluorescent Dyes , Hydrogen Peroxide , Fluorescent Dyes/chemistry , Carbocyanines/chemistry , Hydrogen-Ion Concentration
3.
J Neurosci ; 43(9): 1475-1491, 2023 03 01.
Article En | MEDLINE | ID: mdl-36732068

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.


Substance P , Synaptic Vesicles , Animals , Mice , Synaptotagmins/metabolism , Substance P/metabolism , Synaptic Vesicles/metabolism , Synaptic Transmission/physiology , Neurons/metabolism , Exocytosis , Neurotransmitter Agents/metabolism , Synaptotagmin I/metabolism , Calcium/metabolism
4.
Environ Dev Sustain ; : 1-29, 2022 Aug 01.
Article En | MEDLINE | ID: mdl-35935514

Sustainable supply chain management (SSCM) has received extensive attention by academia and industries recently. However, there are increasing yet still scarce studies measuring the social sustainability performance of supply chain and discussing the interrelationship between social and economic sustainability. Further, the measurement does not fully utilize key performance indicators (KPIs) attributing to the lack of understanding of precise quantitative gauge of the supply chain social sustainable performance. To bridge this gap, this study analyses the social and economic sustainability performance in terms of demand planning, innovation, manufacturing, finance, sales and customer relationship, distribution and delivery and compliance. A framework is proposed to locate key metrics to evaluate the social sustainable supply chain (SSC) performance. A hybrid fuzzy-AHP-DEMATEL-VIKOR method is designed to investigate the social sustainability of supply chain. Data analysis and a case study are given to validate and support the feasibility and potency of the proposed approach. The robustness of our proposed model is executed via sensitivity analysis. From the proposed framework, demand planning and distribution and delivery are found to be the most critical criteria in economic and social dimension, respectively.

5.
J Neurosci ; 42(30): 5816-5829, 2022 07 27.
Article En | MEDLINE | ID: mdl-35701163

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.


C2 Domains , Calcium , Neurotransmitter Agents , Synaptotagmin I , Animals , Calcium/metabolism , Lipids , Mice , Neurotransmitter Agents/metabolism , SNARE Proteins/metabolism , Synaptotagmin I/genetics , Synaptotagmin I/metabolism
6.
Nat Struct Mol Biol ; 29(2): 97-107, 2022 02.
Article En | MEDLINE | ID: mdl-35132256

Neurotransmitter release is mediated by proteins that drive synaptic vesicle fusion with the presynaptic plasma membrane. While soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs) form the core of the fusion apparatus, additional proteins play key roles in the fusion pathway. Here, we report that the C-terminal amphipathic helix of the mammalian accessory protein, complexin (Cpx), exerts profound effects on membranes, including the formation of pores and the efficient budding and fission of vesicles. Using nanodisc-black lipid membrane electrophysiology, we demonstrate that the membrane remodeling activity of Cpx modulates the structure and stability of recombinant exocytic fusion pores. Cpx had particularly strong effects on pores formed by small numbers of SNAREs. Under these conditions, Cpx increased the current through individual pores 3.5-fold, and increased the open time fraction from roughly 0.1 to 1.0. We propose that the membrane sculpting activity of Cpx contributes to the phospholipid rearrangements that underlie fusion by stabilizing highly curved membrane fusion intermediates.


Adaptor Proteins, Vesicular Transport/chemistry , Nerve Tissue Proteins/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HEK293 Cells , Humans , Lipid Bilayers/chemistry , Membrane Fusion/physiology , Molecular Dynamics Simulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation, alpha-Helical , Protein Stability , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism
7.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article En | MEDLINE | ID: mdl-34810248

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.


Neurotransmitter Agents/metabolism , Animals , Calcium/metabolism , Cytoplasm/metabolism , Electrophysiology , Exocytosis , In Vitro Techniques , Light , Lipid Bilayers/chemistry , Lipids/chemistry , Lysine/chemistry , Membrane Fusion , Microscopy, Atomic Force , Neurons/metabolism , Phospholipids/chemistry , Presynaptic Terminals/metabolism , Protein Domains , Protein Multimerization , Recombinant Proteins/metabolism , Scattering, Radiation , Synaptic Vesicles/metabolism , Synaptotagmin I/metabolism
8.
Elife ; 102021 06 30.
Article En | MEDLINE | ID: mdl-34190041

All membrane fusion reactions proceed through an initial fusion pore, including calcium-triggered release of neurotransmitters and hormones. Expansion of this small pore to release cargo is energetically costly and regulated by cells, but the mechanisms are poorly understood. Here, we show that the neuronal/exocytic calcium sensor Synaptotagmin-1 (Syt1) promotes expansion of fusion pores induced by SNARE proteins. Pore dilation relied on calcium-induced insertion of the tandem C2 domain hydrophobic loops of Syt1 into the membrane, previously shown to reorient the C2 domain. Mathematical modelling suggests that C2B reorientation rotates a bound SNARE complex so that it exerts force on the membranes in a mechanical lever action that increases the height of the fusion pore, provoking pore dilation to offset the bending energy penalty. We conclude that Syt1 exerts novel non-local calcium-dependent mechanical forces on fusion pores that dilate pores and assist neurotransmitter and hormone release.


SNARE Proteins/metabolism , Synaptotagmin I/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Calcium/metabolism , Cell Fusion , Cell Membrane , Gene Expression Regulation/physiology , HeLa Cells , Humans , Lipoproteins , Models, Biological , Models, Molecular , Nanostructures , Protein Conformation , SNARE Proteins/genetics , Synaptotagmin I/genetics , Vesicle-Associated Membrane Protein 2/genetics
9.
Nat Chem ; 13(4): 335-342, 2021 04.
Article En | MEDLINE | ID: mdl-33785892

In cells, myriad membrane-interacting proteins generate and maintain curved membrane domains with radii of curvature around or below 50 nm. To understand how such highly curved membranes modulate specific protein functions, and vice versa, it is imperative to use small liposomes with precisely defined attributes as model membranes. Here, we report a versatile and scalable sorting technique that uses cholesterol-modified DNA 'nanobricks' to differentiate hetero-sized liposomes by their buoyant densities. This method separates milligrams of liposomes, regardless of their origins and chemical compositions, into six to eight homogeneous populations with mean diameters of 30-130 nm. We show that these uniform, leak-resistant liposomes serve as ideal substrates to study, with an unprecedented resolution, how membrane curvature influences peripheral (ATG3) and integral (SNARE) membrane protein activities. Compared with conventional methods, our sorting technique represents a streamlined process to achieve superior liposome size uniformity, which benefits research in membrane biology and the development of liposomal drug-delivery systems.


Centrifugation/methods , DNA/chemistry , Liposomes/isolation & purification , Autophagy-Related Protein 7/metabolism , Cholesterol/analogs & derivatives , Liposomes/metabolism , Particle Size , SNARE Proteins/metabolism
10.
Methods Mol Biol ; 1860: 263-275, 2019.
Article En | MEDLINE | ID: mdl-30317511

During exocytosis, vesicles fuse with the plasma membrane and release their contents. The fusion pore is the initial, nanometer-sized connection between the plasma membrane and the cargo-laden vesicle. A growing body of evidence points toward the fusion pore being a regulator of exocytosis, but the shortcomings of current experimental techniques to investigate single-fusion pores make it difficult to study factors governing pore behavior. Here we describe an assay that fuses v-SNARE-reconstituted nanodiscs with cells ectopically expressing "flipped" t-SNAREs to monitor dynamics of single fusion pores in a biochemically defined system using electrical recordings. We also describe a fluorescence microscopy-based approach to monitor nanodisc-cell fusion that is much simpler to employ, but cannot resolve single pores.


Biological Assay/methods , Nanostructures/chemistry , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Membrane/chemistry , Cell Membrane/metabolism , Exocytosis , Genetic Engineering , HeLa Cells , Humans , Membrane Fusion , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Secretory Vesicles/chemistry , Secretory Vesicles/metabolism , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/genetics , Syntaxin 1/chemistry , Syntaxin 1/genetics
11.
Elife ; 62017 10 30.
Article En | MEDLINE | ID: mdl-29083305

Many biological processes rely on protein-membrane interactions in the presence of mechanical forces, yet high resolution methods to quantify such interactions are lacking. Here, we describe a single-molecule force spectroscopy approach to quantify membrane binding of C2 domains in Synaptotagmin-1 (Syt1) and Extended Synaptotagmin-2 (E-Syt2). Syts and E-Syts bind the plasma membrane via multiple C2 domains, bridging the plasma membrane with synaptic vesicles or endoplasmic reticulum to regulate membrane fusion or lipid exchange, respectively. In our approach, single proteins attached to membranes supported on silica beads are pulled by optical tweezers, allowing membrane binding and unbinding transitions to be measured with unprecedented spatiotemporal resolution. C2 domains from either protein resisted unbinding forces of 2-7 pN and had binding energies of 4-14 kBT per C2 domain. Regulation by bilayer composition or Ca2+ recapitulated known properties of both proteins. The method can be widely applied to study protein-membrane interactions.


Cell Membrane/metabolism , Single Molecule Imaging/methods , Protein Binding , Synaptotagmin I/metabolism , Synaptotagmins/metabolism
12.
Front Mol Neurosci ; 10: 315, 2017.
Article En | MEDLINE | ID: mdl-29066949

Calcium-triggered exocytotic release of neurotransmitters and hormones from neurons and neuroendocrine cells underlies neuronal communication, motor activity and endocrine functions. The core of the neuronal exocytotic machinery is composed of soluble N-ethyl maleimide sensitive factor attachment protein receptors (SNAREs). Formation of complexes between vesicle-attached v- and plasma-membrane anchored t-SNAREs in a highly regulated fashion brings the membranes into close apposition. Small, soluble proteins called Complexins (Cpx) and calcium-sensing Synaptotagmins cooperate to block fusion at low resting calcium concentrations, but trigger release upon calcium increase. A growing body of evidence suggests that the transmembrane domains (TMDs) of SNARE proteins play important roles in regulating the processes of fusion and release, but the mechanisms involved are only starting to be uncovered. Here we review recent evidence that SNARE TMDs exert influence by regulating the dynamics of the fusion pore, the initial aqueous connection between the vesicular lumen and the extracellular space. Even after the fusion pore is established, hormone release by neuroendocrine cells is tightly controlled, and the same may be true of neurotransmitter release by neurons. The dynamics of the fusion pore can regulate the kinetics of cargo release and the net amount released, and can determine the mode of vesicle recycling. Manipulations of SNARE TMDs were found to affect fusion pore properties profoundly, both during exocytosis and in biochemical reconstitutions. To explain these effects, TMD flexibility, and interactions among TMDs or between TMDs and lipids have been invoked. Exocytosis has provided the best setting in which to unravel the underlying mechanisms, being unique among membrane fusion reactions in that single fusion pores can be probed using high-resolution methods. An important role will likely be played by methods that can probe single fusion pores in a biochemically defined setting which have recently become available. Finally, computer simulations are valuable mechanistic tools because they have the power to access small length scales and very short times that are experimentally inaccessible.

13.
Mol Med Rep ; 16(4): 4247-4252, 2017 Oct.
Article En | MEDLINE | ID: mdl-28731156

It has been previously identified that α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs) are expressed in pancreatic ß cells and regulate exocytosis and insulin release. It is known that protein interacting with C­kinase 1 (PICK1) regulates trafficking and synaptic targeting of AMPARs in the central nervous system. However, it is unknown whether PICK1 regulates glutamate­induced insulin release in ß cells. The present study demonstrated that glutamate­induced exocytosis was increased in ß cells derived from PICK1­knockout mice. In agreement with this result, adding PICK1 in ß cells reduced glutamate­induced exocytosis, whereas adding EVKI, a peptide that interrupts the interaction between AMPARs and PICK1, increased the exocytosis of ß cells with the application of glutamate. Furthermore, the conductance of ATP­sensitive potassium (KATP) channels was reduced in PICK1­knockout mice, which was reversed by the overexpression of PICK1. In addition, PICK1 application reduced voltage oscillation induced by the closure of KATP. Taken together, the results indicate that PICK1 regulates glutamate­induced exocytosis in ß cells.


Carrier Proteins/metabolism , Exocytosis , Glutamic Acid/pharmacology , Insulin-Secreting Cells/metabolism , Ion Channel Gating/drug effects , KATP Channels/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins , Exocytosis/drug effects , Insulin-Secreting Cells/drug effects , Male , Mice , Nuclear Proteins/deficiency
14.
Elife ; 62017 03 27.
Article En | MEDLINE | ID: mdl-28346138

Hormones and neurotransmitters are released through fluctuating exocytotic fusion pores that can flicker open and shut multiple times. Cargo release and vesicle recycling depend on the fate of the pore, which may reseal or dilate irreversibly. Pore nucleation requires zippering between vesicle-associated v-SNAREs and target membrane t-SNAREs, but the mechanisms governing the subsequent pore dilation are not understood. Here, we probed the dilation of single fusion pores using v-SNARE-reconstituted ~23-nm-diameter discoidal nanolipoprotein particles (vNLPs) as fusion partners with cells ectopically expressing cognate, 'flipped' t-SNAREs. Pore nucleation required a minimum of two v-SNAREs per NLP face, and further increases in v-SNARE copy numbers did not affect nucleation rate. By contrast, the probability of pore dilation increased with increasing v-SNARE copies and was far from saturating at 15 v-SNARE copies per face, the NLP capacity. Our experimental and computational results suggest that SNARE availability may be pivotal in determining whether neurotransmitters or hormones are released through a transient ('kiss and run') or an irreversibly dilating pore (full fusion).


Exocytosis , SNARE Proteins/metabolism , Secretory Vesicles/metabolism , HeLa Cells , Hormones/metabolism , Humans , Neurotransmitter Agents/metabolism
15.
Growth Factors ; 34(5-6): 159-165, 2016 12.
Article En | MEDLINE | ID: mdl-27681688

OBJECTIVE: To investigate the role of intrauterine malnourishment in the development and function of pancreatic islet ß-cells. METHODS: Whole-cell patch clamping was used to record voltage-gated calcium channel (VGCC)-mediated currents. Insulin secretion was detected by measuring capacitance using a sequence of sine wave stimuli. VGCC currents and insulin secretion were measured in the small for gestational age (SGA) group treated with human recombinant growth hormone (hGH). RESULTS: The membrane capacitance in the SGA group (6.4 ± 0.9 fF/Pf) was significantly reduced. Calcium current density and peak current density in the SGA group were also markedly decreased, whereas other measurements of calcium channels were unaltered. Treatment with hGH significantly rescued the membrane capacitance, whereas calcium channels were not affected. CONCLUSION: Our data suggest that decreased ß-cell secretion is caused by a decreased expression of calcium channels and reduced calcium currents. hGH restores ß-cell secretion in SGA animals, possibly independently of VGCC.


Calcium Channels/metabolism , Fetal Growth Retardation/metabolism , Insulin-Secreting Cells/metabolism , Action Potentials , Animals , Cells, Cultured , Female , Growth Hormone/pharmacology , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Rats , Rats, Sprague-Dawley
16.
Sci Rep ; 6: 27287, 2016 06 06.
Article En | MEDLINE | ID: mdl-27264104

The initial, nanometer-sized connection between the plasma membrane and a hormone- or neurotransmitter-filled vesicle -the fusion pore- can flicker open and closed repeatedly before dilating or resealing irreversibly. Pore dynamics determine release and vesicle recycling kinetics, but pore properties are poorly known because biochemically defined single-pore assays are lacking. We isolated single flickering pores connecting v-SNARE-reconstituted nanodiscs to cells ectopically expressing cognate, "flipped" t-SNAREs. Conductance through single, voltage-clamped fusion pores directly reported sub-millisecond pore dynamics. Pore currents fluctuated, transiently returned to baseline multiple times, and disappeared ~6 s after initial opening, as if the fusion pore fluctuated in size, flickered, and resealed. We found that interactions between v- and t-SNARE transmembrane domains (TMDs) promote, but are not essential for pore nucleation. Surprisingly, TMD modifications designed to disrupt v- and t-SNARE TMD zippering prolonged pore lifetimes dramatically. We propose that the post-fusion geometry of the proteins contribute to pore stability.


Cell Fusion/methods , Cell Nucleus/metabolism , SNARE Proteins/chemistry , SNARE Proteins/metabolism , Calcium/metabolism , Exocytosis , HeLa Cells , Humans , Membrane Fusion , Neurotransmitter Agents , Protein Binding , Protein Domains , Secretory Vesicles/metabolism
17.
Biophys J ; 110(7): 1538-1550, 2016 Apr 12.
Article En | MEDLINE | ID: mdl-27074679

Flickering of fusion pores during exocytotic release of hormones and neurotransmitters is well documented, but without assays that use biochemically defined components and measure single-pore dynamics, the mechanisms remain poorly understood. We used total internal reflection fluorescence microscopy to quantify fusion-pore dynamics in vitro and to separate the roles of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and lipid bilayer properties. When small unilamellar vesicles bearing neuronal v-SNAREs fused with planar bilayers reconstituted with cognate t-SNARES, lipid and soluble cargo transfer rates were severely reduced, suggesting that pores flickered. From the lipid release times we computed pore openness, the fraction of time the pore is open, which increased dramatically with cholesterol. For most lipid compositions tested, SNARE-mediated and nonspecifically nucleated pores had similar openness, suggesting that pore flickering was controlled by lipid bilayer properties. However, with physiological cholesterol levels, SNAREs substantially increased the fraction of fully open pores and fusion was so accelerated that there was insufficient time to recruit t-SNAREs to the fusion site, consistent with t-SNAREs being preclustered by cholesterol into functional docking and fusion platforms. Our results suggest that cholesterol opens pores directly by reducing the fusion-pore bending energy, and indirectly by concentrating several SNAREs into individual fusion events.


Cholesterol/metabolism , Membrane Fusion , SNARE Proteins/chemistry , SNARE Proteins/metabolism , Kinetics , Microscopy, Fluorescence , Models, Molecular , Protein Conformation , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 23(6): 1692-6, 2015 Dec.
Article Zh | MEDLINE | ID: mdl-26708895

OBJECTIVE: To investigate whether the progesterone can promote fibronection (FN) synthesis by human bone marrow mesenchymal stem cells (MSCs) and to explore the potential underlying mechanism. METHODS: The human bone marrow MSCs were cultured in a serum-free medium with progesterone for 72 hours, the MTT test was performed to observe the proliferation status and adhension ability of the treated cells. Western blot was used to detect the content of FN in MSDs with GAPDH as the internal reference, the phosphorylation of ERK1/2, as well as the FN content in MSC treated by PD98059, a specific inhibitor of ERK1/2. RESULTS: The progesterone at a range of certain doses not effect on the proliferation of human bone marrow MSCs. Progesterone (25 µg/L) treatment enhanced the FN expression and adherent ability of marrow MSCs. Progesterone could induce prompt phosphorylation of ERK 1/2 and its promoting effects on FN synthesis was reversed by PD98059. CONCLUSION: The progesterone can promote FN synthesis by human bone marrow MSCs via ERK 1/2 pathway, and it might be used to culture MSCs in serum-free medium.


Bone Marrow Cells , Fibronectins , MAP Kinase Signaling System , Mesenchymal Stem Cells , Cells, Cultured , Hematopoietic Stem Cells , Humans , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Progesterone
19.
J Membr Biol ; 246(2): 101-7, 2013 Feb.
Article En | MEDLINE | ID: mdl-23296347

Pancreatic beta cells act as glucose sensors, in which intracellular ATP ([ATP](i)) are altered with glucose concentration change. The characterization of voltage-gated sodium channels under different [ATP](i) remains unclear. Here, we demonstrated that increasing [ATP](i) within a certain range of concentrations (2-8 mM) significantly enhanced the voltage-gated sodium channel currents, compared with 2 mM cytosolic ATP. This enhancement was attenuated by even high intracellular ATP (12 mM). Furthermore, elevated ATP modulated the sodium channel kinetics in a dose-dependent manner. Increased [ATP](i) shifted both the current-voltage curve and the voltage-dependent inactivation curve of sodium channel to the right. Finally, the sodium channel recovery from inactivation was significantly faster when the intracellular ATP level was increased, especially in 8 mM [ATP](i), which is an attainable concentration by the high glucose stimulation. In summary, our data suggested that elevated cytosolic ATP enhanced the activity of Na(+) channels, which may play essential roles in modulating ß cell excitability and insulin release when blood glucose concentration increases.


Adenosine Triphosphate/metabolism , Insulin-Secreting Cells/metabolism , Animals , Blood Glucose/metabolism , Electrophysiology , Humans , In Vitro Techniques , Kinetics , Male , Mice , Pancreas/metabolism
20.
PLoS One ; 7(9): e46012, 2012.
Article En | MEDLINE | ID: mdl-23049922

BACKGROUND: Zinc distributes widely in the central nervous system, especially in the hippocampus, amygdala and cortex. The dynamic balance of zinc is critical for neuronal functions. Zinc modulates the activity of N-methyl-D-aspartate receptors (NMDARs) through the direct inhibition and various intracellular signaling pathways. Abnormal NMDAR activities have been implicated in the aetiology of many brain diseases. Sustained zinc accumulation in the extracellular fluid is known to link to pathological conditions. However, the mechanism linking this chronic zinc exposure and NMDAR dysfunction is poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: We reported that chronic zinc exposure reduced the numbers of NR1 and NR2A clusters in cultured hippocampal pyramidal neurons. Whole-cell and synaptic NR2A-mediated currents also decreased. By contrast, zinc did not affect NR2B, suggesting that chronic zinc exposure specifically influences NR2A-containg NMDARs. Surface biotinylation indicated that zinc exposure attenuated the membrane expression of NR1 and NR2A, which might arise from to the dissociation of the NR2A-PSD-95-Src complex. CONCLUSIONS: Chronic zinc exposure perturbs the interaction of NR2A to PSD-95 and causes the disorder of NMDARs in hippocampal neurons, suggesting a novel action of zinc distinct from its acute effects on NMDAR activity.


Hippocampus/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Zinc/pharmacology , Amygdala/metabolism , Animals , Biotinylation , Brain/metabolism , Cell Death , Cell Survival , Cerebral Cortex/metabolism , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Electrophysiology/methods , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Potentials , Membrane Proteins/metabolism , Mitochondria/metabolism , Models, Biological , Nimodipine/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Surface Properties , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology
...