Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.940
Filter
1.
Elife ; 132024 Jun 04.
Article in English | MEDLINE | ID: mdl-38831693

ABSTRACT

A change in the electric charge of autophagosome membranes controls the recruitment of SNARE proteins to ensure that membrane fusion occurs at the right time during autophagy.


Subject(s)
Autophagosomes , Autophagy , Membrane Fusion , SNARE Proteins , Autophagy/physiology , Autophagosomes/metabolism , SNARE Proteins/metabolism , Humans , Animals
2.
J Gen Physiol ; 156(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38860965

ABSTRACT

The release of neurotransmitter from a single synaptic vesicle generates a quantal response, which at excitatory synapses in voltage-clamped neurons is referred to as a miniature excitatory postsynaptic current (mEPSC). We analyzed mEPSCs in cultured mouse hippocampal neurons and in HEK cells expressing postsynaptic proteins enabling them to receive synaptic inputs from cocultured neurons. mEPSC amplitudes and rise-times varied widely within and between cells. In neurons, mEPSCs with larger amplitudes had longer rise-times, and this correlation was stronger in neurons with longer mean rise-times. In HEK cells, this correlation was weak and unclear. Standard mechanisms thought to govern mEPSCs cannot account for these results. We therefore developed models to simulate mEPSCs and assess their dependence on different factors. Modeling indicated that longer diffusion times for transmitters released by larger vesicles to reach more distal receptors cannot account for the correlation between rise-time and amplitude. By contrast, incorporating the vesicle size dependence of fusion pore expulsion time recapitulated experimental results well. Larger vesicles produce mEPSCs with larger amplitudes and also take more time to lose their content. Thus, fusion pore flux directly contributes to mEPSC rise-time. Variations in fusion pores account for differences among neurons, between neurons and HEK cells, and the correlation between rise-time and the slope of rise-time versus amplitude plots. Plots of mEPSC amplitude versus rise-time are sensitive to otherwise inaccessible properties of a synapse and offer investigators a means of assessing the role of fusion pores in synaptic release.


Subject(s)
Hippocampus , Neurons , Synaptic Vesicles , Animals , Mice , Humans , Neurons/physiology , Neurons/metabolism , HEK293 Cells , Synaptic Vesicles/metabolism , Hippocampus/physiology , Hippocampus/metabolism , Excitatory Postsynaptic Potentials/physiology , Synapses/physiology , Synapses/metabolism , Cells, Cultured , Membrane Fusion/physiology , Miniature Postsynaptic Potentials/physiology
3.
PLoS One ; 19(6): e0304345, 2024.
Article in English | MEDLINE | ID: mdl-38857287

ABSTRACT

Irreversible electroporation induces permanent permeabilization of lipid membranes of vesicles, resulting in vesicle rupture upon the application of a pulsed electric field. Electrofusion is a phenomenon wherein neighboring vesicles can be induced to fuse by exposing them to a pulsed electric field. We focus how the frequency of direct current (DC) pulses of electric field impacts rupture and electrofusion in cell-sized giant unilamellar vesicles (GUVs) prepared in a physiological buffer. The average time, probability, and kinetics of rupture and electrofusion in GUVs have been explored at frequency 500, 800, 1050, and 1250 Hz. The average time of rupture of many 'single GUVs' decreases with the increase in frequency, whereas electrofusion shows the opposite trend. At 500 Hz, the rupture probability stands at 0.45 ± 0.02, while the electrofusion probability is 0.71 ± 0.01. However, at 1250 Hz, the rupture probability increases to 0.69 ± 0.03, whereas the electrofusion probability decreases to 0.46 ± 0.03. Furthermore, when considering kinetics, at 500 Hz, the rate constant of rupture is (0.8 ± 0.1)×10-2 s-1, and the rate constant of fusion is (2.4 ± 0.1)×10-2 s-1. In contrast, at 1250 Hz, the rate constant of rupture is (2.3 ± 0.8)×10-2 s-1, and the rate constant of electrofusion is (1.0 ± 0.1)×10-2 s-1. These results are discussed by considering the electrical model of the lipid bilayer and the energy barrier of a prepore.


Subject(s)
Electroporation , Unilamellar Liposomes , Unilamellar Liposomes/chemistry , Kinetics , Electroporation/methods , Probability , Membrane Fusion
4.
J Phys Chem B ; 128(20): 4986-4995, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38739415

ABSTRACT

Membrane fusion is considered the first step in the entry of enveloped viruses into the host cell. Several targeted strategies have been implemented to block viral entry by limiting the fusion protein to form a six-helix bundle, which is a prerequisite for fusion. Nonetheless, the development of broad-spectrum fusion inhibitors is essential to combat emerging and re-emerging viral infections. TG-23, a coronin 1, a tryptophan-aspartate-rich phagosomal protein-derived peptide, demonstrated inhibition of fusion between small unilamellar vesicles (SUVs) by modulating the membrane's physical properties. However, its inhibitory efficacy reduces with an increasing concentration of membrane cholesterol. The present work aims to develop a fusion inhibitor whose efficacy would be unaltered in the presence of membrane cholesterol. A stretch of the tryptophan-aspartic acid-containing peptide with a similar secondary structure and hydrophobicity profile of TG-23 from coronin 1 was synthesized, and its ability to inhibit SUV-SUV fusion with varying concentrations of membrane cholesterol was evaluated. Our results demonstrate that the GG-21 peptide inhibits fusion irrespective of the cholesterol content of the membrane. We have further evaluated the peptide-induced change in the membrane organization and dynamics utilizing arrays of steady-state and time-resolved fluorescence measurements and correlated these results with their effect on fusion. Interestingly, GG-21 displays inhibitory efficacy in a wide variety of lipid compositions despite having a secondary structure and physical properties similar to those of TG-23. Overall, our results advocate that the secondary structure and physical properties of the peptide may not be sufficient to predict its inhibitory efficacy.


Subject(s)
Cholesterol , Membrane Fusion , Membrane Fusion/drug effects , Cholesterol/chemistry , Peptides/chemistry , Peptides/pharmacology , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/chemistry
5.
J Phys Chem B ; 128(20): 4975-4985, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38743687

ABSTRACT

The primary event in chemical neurotransmission involves the fusion of a membrane-limited vesicle at the plasma membrane and the subsequent release of its chemical neurotransmitter cargo. The cargo itself is not known to have any effect on the fusion event. However, amphiphilic monoamine neurotransmitters (e.g., serotonin and dopamine) are known to strongly interact with lipid bilayers and to affect their mechanical properties, which can in principle impact membrane-mediated processes. Here, we probe whether serotonin can enhance the association and fusion of artificial lipid vesicles in vitro. We employ fluorescence correlation spectroscopy and total internal reflection fluorescence microscopy to measure the attachment and fusion of vesicles whose lipid compositions mimic the major lipid components of synaptic vesicles. We find that the association between vesicles and supported lipid bilayers is strongly enhanced in a serotonin dose-dependent manner, and this drives an increase in the rate of spontaneous fusion. Molecular dynamics simulations and fluorescence spectroscopy data show that serotonin insertion increases the water content of the hydrophobic part of the bilayer. This suggests that the enhanced membrane association is likely driven by an energetically favorable drying transition. Other monoamines, such as dopamine and norepinephrine, but not other related species, such as tryptophan, show similar effects on membrane association. Our results reveal a lipid bilayer-mediated mechanism by which monoamines can themselves modulate vesicle fusion, potentially adding to the control toolbox for the tightly regulated process of neurotransmission in vivo.


Subject(s)
Lipid Bilayers , Molecular Dynamics Simulation , Serotonin , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Serotonin/chemistry , Serotonin/metabolism , Membrane Fusion , Synaptic Vesicles/metabolism , Synaptic Vesicles/chemistry , Spectrometry, Fluorescence , Hydrophobic and Hydrophilic Interactions
6.
Nat Commun ; 15(1): 4267, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769317

ABSTRACT

The membrane-fusion-based internalization without lysosomal entrapment is advantageous for intracellular delivery over endocytosis. However, protein corona formed on the membrane-fusogenic liposome surface converts its membrane-fusion performance to lysosome-dependent endocytosis, causing poorer delivery efficiency in biological conditions. Herein, we develop an antifouling membrane-fusogenic liposome for effective intracellular delivery in vivo. Leveraging specific lipid composition at an optimized ratio, such antifouling membrane-fusogenic liposome facilitates fusion capacity even in protein-rich conditions, attributed to the copious zwitterionic phosphorylcholine groups for protein-adsorption resistance. Consequently, the antifouling membrane-fusogenic liposome demonstrates robust membrane-fusion-mediated delivery in the medium with up to 38% fetal bovine serum, outclassing two traditional membrane-fusogenic liposomes effective at 4% and 6% concentrations. When injected into mice, antifouling membrane-fusogenic liposomes can keep their membrane-fusion-transportation behaviors, thereby achieving efficient luciferase transfection and enhancing gene-editing-mediated viral inhibition. This study provides a promising tool for effective intracellular delivery under complex physiological environments, enlightening future nanomedicine design.


Subject(s)
Liposomes , Membrane Fusion , Liposomes/metabolism , Animals , Mice , Humans , Endocytosis , Transfection , Gene Editing/methods , Protein Corona/metabolism , Protein Corona/chemistry , Biofouling/prevention & control , Female , Lipids/chemistry
7.
Nat Commun ; 15(1): 4132, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755165

ABSTRACT

The regulated release of chemical messengers is crucial for cell-to-cell communication; abnormalities in which impact coordinated human body function. During vesicular secretion, multiple SNARE complexes assemble at the release site, leading to fusion pore opening. How membrane fusion regulators act on heterogeneous SNARE populations to assemble fusion pores in a timely and synchronized manner, is unknown. Here, we demonstrate the role of SNARE chaperones Munc13-1 and Munc18-1 in rescuing individual nascent fusion pores from their diacylglycerol lipid-mediated inhibitory states. At the onset of membrane fusion, Munc13-1 clusters multiple SNARE complexes at the release site and synchronizes release events, while Munc18-1 stoichiometrically interacts with trans-SNARE complexes to enhance N- to C-terminal zippering. When both Munc proteins are present simultaneously, they differentially access dynamic trans-SNARE complexes to regulate pore properties. Overall, Munc proteins' direct action on fusion pore assembly indicates their role in controlling quantal size during vesicular secretion.


Subject(s)
Membrane Fusion , Munc18 Proteins , Nerve Tissue Proteins , SNARE Proteins , Munc18 Proteins/metabolism , Munc18 Proteins/genetics , SNARE Proteins/metabolism , SNARE Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Animals , Humans , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Rats
8.
ACS Nano ; 18(20): 12737-12748, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38717305

ABSTRACT

Lipids are key factors in regulating membrane fusion. Lipids are not only structural components to form membranes but also active catalysts for vesicle fusion and neurotransmitter release, which are driven by soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. SNARE proteins seem to be partially assembled before fusion, but the mechanisms that arrest vesicle fusion before Ca2+ influx are still not clear. Here, we show that phosphatidylinositol 4,5-bisphosphate (PIP2) electrostatically triggers vesicle fusion as an electrostatic catalyst by lowering the hydration energy and that a myristoylated alanine-rich C-kinase substrate (MARCKS), a PIP2-binding protein, arrests vesicle fusion in a vesicle docking state where the SNARE complex is partially assembled. Vesicle-mimicking liposomes fail to reproduce vesicle fusion arrest by masking PIP2, indicating that native vesicles are essential for the reconstitution of physiological vesicle fusion. PIP2 attracts cations to repel water molecules from membranes, thus lowering the hydration energy barrier.


Subject(s)
Membrane Fusion , Phosphatidylinositol 4,5-Diphosphate , Static Electricity , Water , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Water/chemistry , Liposomes/chemistry , SNARE Proteins/metabolism , SNARE Proteins/chemistry , Catalysis
9.
Nat Commun ; 15(1): 4131, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755250

ABSTRACT

The transition between yeast and hyphae is crucial for regulating the commensalism and pathogenicity in Candida albicans. The mechanisms that affect the invasion of hyphae in solid media, whose deficiency is more related to the pathogenicity of C. albicans, have not been elucidated. Here, we found that the disruption of VAM6 or VPS41 which are components of the homotypic vacuolar fusion and protein sorting (HOPS) complex, or the Rab GTPase YPT72, all responsible for vacuole fusion, led to defects in hyphal growth in both liquid and solid media, but more pronounced on solid agar. The phenotypes of vac8Δ/Δ and GTR1OE-vam6Δ/Δ mutants indicated that these deficiencies are mainly caused by the reduced mechanical forces that drive agar and organs penetration, and confirmed that large vacuoles are required for hyphal mechanical penetration. In summary, our study revealed that large vacuoles generated by vacuolar fusion support hyphal penetration and provided a perspective to refocus attention on the role of solid agar in evaluating C. albicans invasion.


Subject(s)
Candida albicans , Fungal Proteins , Hyphae , Vacuoles , Candida albicans/metabolism , Candida albicans/genetics , Hyphae/metabolism , Hyphae/growth & development , Hyphae/genetics , Vacuoles/metabolism , Fungal Proteins/metabolism , Fungal Proteins/genetics , Animals , Mice , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Candidiasis/microbiology , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Female , Membrane Fusion
10.
Biomolecules ; 14(5)2024 May 19.
Article in English | MEDLINE | ID: mdl-38786007

ABSTRACT

During neurotransmission, neurotransmitters are released less than a millisecond after the arrival of the action potential. To achieve this ultra-fast event, the synaptic vesicle must be pre-docked to the plasma membrane. In this primed state, SNAREpins, the protein-coiled coils whose assembly provides the energy to trigger fusion, are partly zippered and clamped like a hairpin and held open and ready to snap close when the clamp is released. Recently, it was suggested that three types of regulatory factors, synaptophysin, synaptotagmins, and complexins act cooperatively to organize two concentric rings, a central and a peripheral ring, containing up to six SNAREpins each. We used a mechanical model of the SNAREpins with two separate states, half-zippered and fully zippered, and determined the energy landscape according to the number of SNAREpins in each ring. We also performed simulations to estimate the fusion time in each case. The presence of the peripheral SNAREpins generally smoothens the energy landscape and accelerates the fusion time. With the predicted physiological numbers of six central and six peripheral SNAREpins, the fusion time is accelerated at least 100 times by the presence of the peripheral SNAREpins, and fusion occurs in less than 10 µs, which is well within the physiological requirements.


Subject(s)
Membrane Fusion , SNARE Proteins , Synaptic Vesicles , Synaptic Vesicles/metabolism , SNARE Proteins/metabolism , Synaptic Transmission , Animals , Humans
11.
Nat Commun ; 15(1): 4524, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806492

ABSTRACT

Membrane fusion, merging two lipid bilayers, is crucial for fabricating artificial membrane structures. Over the past 40 years, in contrast to precise and controllable membrane fusion in-vivo through specific molecules such as SNAREs, controlling the fusion in-vitro while fabricating artificial membrane structures in physiological ionic solutions without fusion proteins has been a challenge, becoming a significant obstacle to practical applications. We present an approach consisting of an electric field and a few kPa hydraulic pressure as an additional variable to physically control the fusion, enabling tuning of the shape and size of the 3D freestanding lipid bilayers in physiological ionic solutions. Mechanical model analysis reveals that pressure-induced parallel/normal tensions enhance fusion among membranes in the microwell. In-vitro peptide-membrane assay, mimicking vesicular transport via pressure-assisted fusion, and stability of 38 days with in-chip pressure control via pore size-regulated hydrogel highlight the potential for diverse biological applications.


Subject(s)
Lipid Bilayers , Membrane Fusion , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Ions/chemistry , Membranes, Artificial , Hydrogels/chemistry , Pressure , Peptides/chemistry
12.
Proc Natl Acad Sci U S A ; 121(16): e2321447121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38593076

ABSTRACT

The SNAP receptor (SNARE) proteins syntaxin-1, SNAP-25, and synaptobrevin mediate neurotransmitter release by forming tight SNARE complexes that fuse synaptic vesicles with the plasma membranes in microseconds. Membrane fusion is generally explained by the action of proteins on macroscopic membrane properties such as curvature, elastic modulus, and tension, and a widespread model envisions that the SNARE motifs, juxtamembrane linkers, and C-terminal transmembrane regions of synaptobrevin and syntaxin-1 form continuous helices that act mechanically as semirigid rods, squeezing the membranes together as they assemble ("zipper") from the N to the C termini. However, the mechanism underlying fast SNARE-induced membrane fusion remains unknown. We have used all-atom molecular dynamics simulations to investigate this mechanism. Our results need to be interpreted with caution because of the limited number and length of the simulations, but they suggest a model of membrane fusion that has a natural physicochemical basis, emphasizes local molecular events over general membrane properties, and explains extensive experimental data. In this model, the central event that initiates fast (microsecond scale) membrane fusion occurs when the SNARE helices zipper into the juxtamembrane linkers which, together with the adjacent transmembrane regions, promote encounters of acyl chains from both bilayers at the polar interface. The resulting hydrophobic nucleus rapidly expands into stalk-like structures that gradually progress to form a fusion pore, aided by the SNARE transmembrane regions and without clearly discernible intermediates. The propensity of polyunsaturated lipids to participate in encounters that initiate fusion suggests that these lipids may be important for the high speed of neurotransmitter release.


Subject(s)
Membrane Fusion , SNARE Proteins , SNARE Proteins/metabolism , Molecular Dynamics Simulation , R-SNARE Proteins , Syntaxin 1 , Neurotransmitter Agents , Lipids
13.
Proc Natl Acad Sci U S A ; 121(16): e2309211121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38593081

ABSTRACT

Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and ß-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.


Subject(s)
Membrane Fusion , Secretagogins , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Secretagogins/metabolism , Cell Membrane/metabolism , Synaptosomal-Associated Protein 25/metabolism , Exocytosis , Cell Communication , Syntaxin 1/metabolism , Protein Binding
14.
J Phys Chem Lett ; 15(16): 4515-4522, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38634827

ABSTRACT

Cholesterol-rich lipid rafts are found to facilitate membrane fusion, central to processes like viral entry, fertilization, and neurotransmitter release. While the fusion process involves local, transient membrane dehydration, the impact of reduced hydration on cholesterol's structural organization in biological membranes remains unclear. Here, we employ confocal fluorescence microscopy and atomistic molecular dynamics simulations to investigate cholesterol behavior in phase-separated lipid bilayers under controlled hydration. We unveiled that dehydration prompts cholesterol release from raft-like domains into the surrounding fluid phase. Unsaturated phospholipids undergo more significant dehydration-induced structural changes and lose more hydrogen bonds with water than sphingomyelin. The results suggest that cholesterol redistribution is driven by the equalization of biophysical properties between phases and the need to satisfy lipid hydrogen bonds. This underscores the role of cholesterol-phospholipid-water interplay in governing cholesterol affinity for a specific lipid type, providing a new perspective on the regulatory role of cell membrane heterogeneity during membrane fusion.


Subject(s)
Cholesterol , Lipid Bilayers , Molecular Dynamics Simulation , Water , Cholesterol/chemistry , Cholesterol/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Water/chemistry , Water/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Hydrogen Bonding , Sphingomyelins/chemistry , Sphingomyelins/metabolism , Membrane Fusion , Phospholipids/chemistry , Phospholipids/metabolism
15.
J Biol Chem ; 300(5): 107274, 2024 May.
Article in English | MEDLINE | ID: mdl-38588809

ABSTRACT

The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex forms a 4-helix coiled-coil bundle consisting of 16 layers of interacting side chains upon membrane fusion. The central layer (layer 0) is highly conserved and comprises three glutamines (Q) and one arginine (R), and thus SNAREs are classified into Qa-, Qb-, Qc-, and R-SNAREs. Homotypic vacuolar fusion in Saccharomyces cerevisiae requires the SNAREs Vam3 (Qa), Vti1 (Qb), Vam7 (Qc), and Nyv1 (R). However, the yeast strain lacking NYV1 (nyv1Δ) shows no vacuole fragmentation, whereas the vam3Δ and vam7Δ strains display fragmented vacuoles. Here, we provide genetic evidence that the R-SNAREs Ykt6 and Nyv1 are functionally redundant in vacuole homotypic fusion in vivo using a newly isolated ykt6 mutant. We observed the ykt6-104 mutant showed no defect in vacuole morphology, but the ykt6-104 nyv1Δ double mutant had highly fragmented vacuoles. Furthermore, we show the defect in homotypic vacuole fusion caused by the vam7-Q284R mutation was compensated by the nyv1-R192Q or ykt6-R165Q mutations, which maintained the 3Q:1R ratio in the layer 0 of the SNARE complex, indicating that Nyv1 is exchangeable with Ykt6 in the vacuole SNARE complex. Unexpectedly, we found Ykt6 assembled with exocytic Q-SNAREs when the intrinsic exocytic R-SNAREs Snc1 and its paralog Snc2 lose their ability to assemble into the exocytic SNARE complex. These results suggest that Ykt6 may serve as a backup when other R-SNAREs become dysfunctional and that this flexible assembly of SNARE complexes may help cells maintain the robustness of the vesicular transport network.


Subject(s)
R-SNARE Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Vacuoles , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Vacuoles/metabolism , Vacuoles/genetics , R-SNARE Proteins/metabolism , R-SNARE Proteins/genetics , Membrane Fusion , Exocytosis , SNARE Proteins/metabolism , SNARE Proteins/genetics , Mutation
16.
Nanoscale ; 16(21): 10221-10229, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38679949

ABSTRACT

Membrane fusion is crucial for infection of enveloped viruses, cellular transport, and drug delivery via liposomes. Nanoparticles can serve as fusogenic agents facilitating such membrane fusion for direct transmembrane transport. However, the underlying mechanisms of nanoparticle-induced fusion and the ideal properties of such nanoparticles remain largely unknown. Here, we used molecular dynamics simulations to investigate the efficacy of spheroidal nanoparticles with different size, prolateness, and ligand interaction strengths to enhance fusion between vesicles. By systematically varying nanoparticle properties, we identified how each parameter affects the fusion process and determined the optimal parameter range that promotes fusion. These findings provide valuable insights for the design and optimization of fusogenic nanoparticles with potential biotechnological and biomedical applications.


Subject(s)
Membrane Fusion , Molecular Dynamics Simulation , Nanoparticles , Nanoparticles/chemistry , Liposomes/chemistry , Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Membrane Lipids/metabolism
17.
Chemistry ; 30(29): e202304111, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38486422

ABSTRACT

Extracellular vesicles (EVs) carry diverse biomolecules (e. g., nucleic acids, proteins) for intercellular communication, serving as important markers for diseases. Analyzing nucleic acids derived from EVs enables non-invasive disease diagnosis and prognosis evaluation. Membrane fusion, a fundamental cellular process wherein two lipid membranes merge, facilitates cell communication and cargo transport. Building on this natural phenomenon, recent years have witnessed the emergence of membrane fusion-based strategies for the detection of nucleic acids within EVs. These strategies entail the encapsulation of detection probes within either artificial or natural vesicles, followed by the induction of membrane fusion with EVs to deliver probes. This innovative approach not only enables in situ detection of nucleic acids within EVs but also ensures the maintenance of structural integrity of EVs, thus preventing nucleic acid degradation and minimizing the interference from free nucleic acids. This concept categorizes approaches into universal and targeted membrane fusion strategies, and discusses their application potential, and challenges and future prospects.


Subject(s)
Extracellular Vesicles , Membrane Fusion , Nucleic Acids , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Nucleic Acids/analysis , Nucleic Acids/chemistry , Humans
18.
Nat Commun ; 15(1): 2652, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38531902

ABSTRACT

Tomosyns are widely thought to attenuate membrane fusion by competing with synaptobrevin-2/VAMP2 for SNARE-complex assembly. Here, we present evidence against this scenario. In a novel mouse model, tomosyn-1/2 deficiency lowered the fusion barrier and enhanced the probability that synaptic vesicles fuse, resulting in stronger synapses with faster depression and slower recovery. While wild-type tomosyn-1m rescued these phenotypes, substitution of its SNARE motif with that of synaptobrevin-2/VAMP2 did not. Single-molecule force measurements indeed revealed that tomosyn's SNARE motif cannot substitute synaptobrevin-2/VAMP2 to form template complexes with Munc18-1 and syntaxin-1, an essential intermediate for SNARE assembly. Instead, tomosyns extensively bind synaptobrevin-2/VAMP2-containing template complexes and prevent SNAP-25 association. Structure-function analyses indicate that the C-terminal polybasic region contributes to tomosyn's inhibitory function. These results reveal that tomosyns regulate synaptic transmission by cooperating with synaptobrevin-2/VAMP2 to prevent SNAP-25 binding during SNARE assembly, thereby limiting initial synaptic strength and equalizing it during repetitive stimulation.


Subject(s)
SNARE Proteins , Vesicle-Associated Membrane Protein 2 , Animals , Mice , SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Membrane Fusion , Depression , Syntaxin 1/metabolism , Nerve Tissue Proteins/metabolism , R-SNARE Proteins/metabolism
19.
Mol Biol Cell ; 35(5): ar71, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38536444

ABSTRACT

Membrane fusion is regulated by Rab GTPases, their tethering effectors such as HOPS, SNARE proteins on each fusion partner, SM proteins to catalyze SNARE assembly, Sec17 (SNAP), and Sec18 (NSF). Though concentrated HOPS can support fusion without Sec18, we now report that fusion falls off sharply at lower HOPS levels, where direct Sec18 binding to HOPS restores fusion. This Sec18-dependent fusion needs adenine nucleotide but neither ATP hydrolysis nor Sec17. Sec18 enhances HOPS recognition of the Qc-SNARE. With high levels of HOPS, Qc has a Km for fusion of a few nM. Either lower HOPS levels, or substitution of a synthetic tether for HOPS, strikingly increases the Km for Qc to several hundred nM. With dilute HOPS, Sec18 returns the Km for Qc to low nM. In contrast, HOPS concentration and Sec18 have no effect on Qb-SNARE recognition. Just as Qc is required for fusion but not for the initial assembly of SNAREs in trans, impaired Qc recognition by limiting HOPS without Sec18 still allows substantial trans-SNARE assembly. Thus, in addition to the known Sec18 functions of disassembling SNARE complexes, oligomerizing Sec17 for membrane association, and allowing Sec17 to drive fusion without complete SNARE zippering, we report a fourth Sec18 function, the Sec17-independent binding of Sec18 to HOPS to enhance functional Qc-SNARE engagement.


Subject(s)
Membrane Fusion , Saccharomyces cerevisiae Proteins , Vesicular Transport Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Vacuoles/metabolism
20.
J Cell Biol ; 223(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38478017

ABSTRACT

SM proteins including Sly1 are essential cofactors of SNARE-mediated membrane fusion. Using SNARE and Sly1 mutants and chemically defined in vitro assays, we separate and assess proposed mechanisms through which Sly1 augments fusion: (i) opening the closed conformation of the Qa-SNARE Sed5; (ii) close-range tethering of vesicles to target organelles, mediated by the Sly1-specific regulatory loop; and (iii) nucleation of productive trans-SNARE complexes. We show that all three mechanisms are important and operate in parallel, and that close-range tethering promotes trans-complex assembly when cis-SNARE assembly is a competing process. Further, we demonstrate that the autoinhibitory N-terminal Habc domain of Sed5 has at least two positive activities: it is needed for correct Sed5 localization, and it directly promotes Sly1-dependent fusion. "Split Sed5," with Habc presented solely as a soluble fragment, can function both in vitro and in vivo. Habc appears to facilitate events leading to lipid mixing rather than promoting opening or stability of the fusion pore.


Subject(s)
Membrane Fusion , Munc18 Proteins , SNARE Proteins , Saccharomyces cerevisiae Proteins , Munc18 Proteins/metabolism , Protein Binding , Qa-SNARE Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...