Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.929
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(30): e2313609121, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39012824

RESUMO

Mitofusins (Mfn1 and Mfn2) are the mitochondrial outer-membrane fusion proteins in mammals and belong to the dynamin superfamily of multidomain GTPases. Recent structural studies of truncated variants lacking alpha helical transmembrane domains suggested that Mfns dimerize to promote the approximation and the fusion of the mitochondrial outer membranes upon the hydrolysis of guanine 5'-triphosphate disodium salt (GTP). However, next to the presence of GTP, the fusion activity seems to require multiple regulatory factors that control the dynamics and kinetics of mitochondrial fusion through the formation of Mfn1-Mfn2 heterodimers. Here, we purified and reconstituted the full-length murine Mfn2 protein into giant unilamellar vesicles (GUVs) with different lipid compositions. The incubation with GTP resulted in the fusion of Mfn2-GUVs. High-speed video-microscopy showed that the Mfn2-dependent membrane fusion pathway progressed through a zipper mechanism where the formation and growth of an adhesion patch eventually led to the formation of a membrane opening at the rim of the septum. The presence of physiological concentration (up to 30 mol%) of dioleoyl-phosphatidylethanolamine (DOPE) was shown to be a requisite to observe GTP-induced Mfn2-dependent fusion. Our observations show that Mfn2 alone can promote the fusion of micron-sized DOPE-enriched vesicles without the requirement of regulatory cofactors, such as membrane curvature, or the assistance of other proteins.


Assuntos
GTP Fosfo-Hidrolases , Fusão de Membrana , Animais , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , Camundongos , Fusão de Membrana/fisiologia , Lipossomas Unilamelares/metabolismo , Lipossomas Unilamelares/química , Guanosina Trifosfato/metabolismo , Fosfatidiletanolaminas/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Mitocôndrias/metabolismo
2.
Nano Lett ; 24(28): 8609-8618, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38954738

RESUMO

Although biomacromolecules are promising cytosolic drugs which have attracted tremendous attention, the major obstacles were the cellular membrane hindering the entrance and the endosome entrapment inducing biomacromolecule degradation. How to avoid those limitations to realize directly cytosolic delivery was still a challenge. Here, we prepared oligoarginine modified lipid to assemble a nanovesicle for biomacromolecules delivery, including mRNA (mRNA) and proteins which could be directly delivered into the cytoplasm of dendritic cells through subendocytosis-mediated membrane fusion. We named this membrane fusion lipid nanovesicle as MF-LNV. The mRNA loaded MF-LNV as nanovaccines showed efficient antigen expression to elicit robust immuno responses for cancer therapy. What's more, the antigen protein loaded MF-LNV as nanovaccines elicits much stronger CD8+ T cell specific responses than lipid nanoparticles through normal uptake pathways. This MF-LNV represented a refreshing strategy for intracellular delivery of the biomacromolecule.


Assuntos
Lipídeos , Lipídeos/química , Animais , Humanos , Nanopartículas/química , Células Dendríticas , RNA Mensageiro/genética , RNA Mensageiro/administração & dosagem , Camundongos , Fusão de Membrana , Sistemas de Liberação de Medicamentos , Linfócitos T CD8-Positivos/imunologia
3.
Methods Enzymol ; 701: 175-236, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39025572

RESUMO

Biomembranes and vesicles cover a wide range of length scales. Indeed, small nanovesicles have a diameter of a few tens of nanometers whereas giant vesicles can have diameters up to hundreds of micrometers. The remodeling of giant vesicles on the micron scale can be observed by light microscopy and understood by the theory of curvature elasticity, which represents a top-down approach. The theory predicts the formation of multispherical shapes as recently observed experimentally. On the nanometer scale, much insight has been obtained via coarse-grained molecular dynamics simulations of nanovesicles, which provides a bottom-up approach based on the lipid numbers assembled in the two bilayer leaflets and the resulting leaflet tensions. The remodeling processes discussed here include the shape transformations of vesicles, their morphological responses to the adhesion of condensate droplets, the instabilities of lipid bilayers and nanovesicles, as well as the topological transformations of vesicles by membrane fission and fusion. The latter processes determine the complex topology of the endoplasmic reticulum.


Assuntos
Bicamadas Lipídicas , Simulação de Dinâmica Molecular , Bicamadas Lipídicas/química , Membrana Celular/química , Membrana Celular/metabolismo , Fusão de Membrana/fisiologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Animais , Humanos
4.
J Cell Biol ; 223(10)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-38980206

RESUMO

Synaptotagmin-1 (Syt1) is a calcium sensor that regulates synaptic vesicle fusion in synchronous neurotransmitter release. Syt1 interacts with negatively charged lipids and the SNARE complex to control the fusion event. However, it remains incompletely understood how Syt1 mediates Ca2+-trigged synaptic vesicle fusion. Here, we discovered that Syt1 undergoes liquid-liquid phase separation (LLPS) to form condensates both in vitro and in living cells. Syt1 condensates play a role in vesicle attachment to the PM and efficiently recruit SNAREs and complexin, which may facilitate the downstream synaptic vesicle fusion. We observed that Syt1 condensates undergo a liquid-to-gel-like phase transition, reflecting the formation of Syt1 oligomers. The phase transition can be blocked or reversed by Ca2+, confirming the essential role of Ca2+ in Syt1 oligomer disassembly. Finally, we showed that the Syt1 mutations causing Syt1-associated neurodevelopmental disorder impair the Ca2+-driven phase transition. These findings reveal that Syt1 undergoes LLPS and a Ca2+-sensitive phase transition, providing new insights into Syt1-mediated vesicle fusion.


Assuntos
Cálcio , Vesículas Sinápticas , Sinaptotagmina I , Sinaptotagmina I/metabolismo , Sinaptotagmina I/genética , Cálcio/metabolismo , Humanos , Animais , Vesículas Sinápticas/metabolismo , Multimerização Proteica , Proteínas SNARE/metabolismo , Proteínas SNARE/genética , Transição de Fase , Mutação/genética , Células HEK293 , Fusão de Membrana , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Separação de Fases
5.
Nat Commun ; 15(1): 5606, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961055

RESUMO

Viral mutations frequently outpace technologies used to detect harmful variants. Given the continual emergence of SARS-CoV-2 variants, platforms that can identify the presence of a virus and its propensity for infection are needed. Our electronic biomembrane sensing platform recreates distinct SARS-CoV-2 host cell entry pathways and reports the progression of entry as electrical signals. We focus on two necessary entry processes mediated by the viral Spike protein: virus binding and membrane fusion, which can be distinguished electrically. We find that closely related variants of concern exhibit distinct fusion signatures that correlate with trends in cell-based infectivity assays, allowing us to report quantitative differences in their fusion characteristics and hence their infectivity potentials. We use SARS-CoV-2 as our prototype, but we anticipate that this platform can extend to other enveloped viruses and cell lines to quantifiably assess virus entry.


Assuntos
COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Internalização do Vírus , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , Humanos , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , COVID-19/virologia , Fusão de Membrana , Sistema Livre de Células , Mutação , Ligação Viral
6.
PLoS Biol ; 22(7): e3002671, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38949997

RESUMO

Mitochondrial shape and network formation have been primarily associated with the well-established processes of fission and fusion. However, recent research has unveiled an intricate and multifaceted landscape of mitochondrial morphology that extends far beyond the conventional fission-fusion paradigm. These less-explored dimensions harbor numerous unresolved mysteries. This review navigates through diverse processes influencing mitochondrial shape and network formation, highlighting the intriguing complexities and gaps in our understanding of mitochondrial architecture. The exploration encompasses various scales, from biophysical principles governing membrane dynamics to molecular machineries shaping mitochondria, presenting a roadmap for future research in this evolving field.


Assuntos
Mitocôndrias , Dinâmica Mitocondrial , Dinâmica Mitocondrial/fisiologia , Mitocôndrias/metabolismo , Animais , Humanos , Membranas Mitocondriais/metabolismo , Forma das Organelas , Proteínas Mitocondriais/metabolismo , Fusão de Membrana/fisiologia
7.
Methods Enzymol ; 700: 127-159, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38971598

RESUMO

The natural asymmetry of the lipid bilayer in biological membranes is, in part, a testament to the complexity of the structure and function of this barrier limiting and protecting cells (or organelles). These lipid bilayers consist of two lipid leaflets with different lipid compositions, resulting in unique interactions within each leaflet. These interactions, combined with interactions between the two leaflets, determine the overall behavior of the membrane. Model membranes provide the most suitable option for investigating the fundamental interactions of lipids. This report describes a comprehensive method to make asymmetric giant unilamellar vesicles (aGUVs) using the technique of hemifusion. In this method, calcium ions induce the hemifusion of giant unilamellar vesicles (GUVs) with a supported lipid bilayer (SLB), both having different lipid compositions. During hemifusion, a stalk, or a more commonly seen hemifusion diaphragm, connects the outer leaflets of GUVs and the SLB. The lateral diffusion of lipids naturally promotes the lipid exchange between the connected outer leaflets. After calcium chelation to prevent further fusion, a mechanical shear detaches aGUVs from the SLB. A fluorescence quench assay is employed to test the extent of bilayer asymmetry. A fluorescence quenching assay tests bilayer asymmetry and verifies dye and lipid migration to a GUV's outer leaflet.


Assuntos
Cálcio , Bicamadas Lipídicas , Lipossomas Unilamelares , Lipossomas Unilamelares/química , Bicamadas Lipídicas/química , Cálcio/química , Cálcio/metabolismo , Fusão de Membrana
8.
J Gen Physiol ; 156(8)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38860965

RESUMO

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.


Assuntos
Hipocampo , Neurônios , Vesículas Sinápticas , Animais , Camundongos , Humanos , Neurônios/fisiologia , Neurônios/metabolismo , Células HEK293 , Vesículas Sinápticas/metabolismo , Hipocampo/fisiologia , Hipocampo/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Sinapses/fisiologia , Sinapses/metabolismo , Células Cultivadas , Fusão de Membrana/fisiologia , Potenciais Pós-Sinápticos em Miniatura/fisiologia
9.
Nat Commun ; 15(1): 5227, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898033

RESUMO

Cells depend on their endolysosomal system for nutrient uptake and downregulation of plasma membrane proteins. These processes rely on endosomal maturation, which requires multiple membrane fusion steps. Early endosome fusion is promoted by the Rab5 GTPase and its effector, the hexameric CORVET tethering complex, which is homologous to the lysosomal HOPS. How these related complexes recognize their specific target membranes remains entirely elusive. Here, we solve the structure of CORVET by cryo-electron microscopy and revealed its minimal requirements for membrane tethering. As expected, the core of CORVET and HOPS resembles each other. However, the function-defining subunits show marked structural differences. Notably, we discover that unlike HOPS, CORVET depends not only on Rab5 but also on phosphatidylinositol-3-phosphate (PI3P) and membrane lipid packing defects for tethering, implying that an organelle-specific membrane code enables fusion. Our data suggest that both shape and membrane interactions of CORVET and HOPS are conserved in metazoans, thus providing a paradigm how tethering complexes function.


Assuntos
Microscopia Crioeletrônica , Endossomos , Fosfatos de Fosfatidilinositol , Endossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fusão de Membrana , Proteínas rab5 de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/genética , Humanos , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Membrana Celular/metabolismo , Animais , Lisossomos/metabolismo
10.
Elife ; 132024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38831693

RESUMO

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.


Assuntos
Autofagossomos , Autofagia , Fusão de Membrana , Proteínas SNARE , Autofagia/fisiologia , Autofagossomos/metabolismo , Proteínas SNARE/metabolismo , Humanos , Animais
11.
PLoS One ; 19(6): e0304345, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38857287

RESUMO

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.


Assuntos
Eletroporação , Lipossomas Unilamelares , Lipossomas Unilamelares/química , Cinética , Eletroporação/métodos , Probabilidade , Fusão de Membrana
12.
Microb Pathog ; 192: 106713, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38810765

RESUMO

Newcastle disease virus (NDV) is the pathogen of a zoonosis that is primarily transmitted by poultry and has severe infectivity and a high fatality rate. Many studies have focused on the role of the NDV fusion (F) protein in the cell-cell membrane fusion process. However, little attention has been given to the heptad repeat region, HR4, which is located in the NDV F2 subunit. Here, site-directed mutants were constructed to study the function of the NDV F protein HR4 region and identify the key amino acids in this region. Nine conserved amino acids were substituted with alanine or the corresponding amino acid of other aligned paramyxoviruses. The desired mutants were examined for changes in fusogenic activity through three kinds of membrane fusion assays and expression and proteolysis through IFA, FACS and WB. The results showed that when conserved amino acids (L81, Y84, L88, L91, L92, P94, L95 and I99) were replaced with alanine, the fusogenic activity of the F protein was abolished, possibly because of failed protein expression not only on the cell surface but also inside cells. These data indicated that the conserved amino acids above in NDV F HR4 are critical for normal protein synthesis and expression, possibly for the stability of the F protein monomer, formation of trimer and conformational changes.


Assuntos
Mutagênese Sítio-Dirigida , Vírus da Doença de Newcastle , Proteínas Virais de Fusão , Internalização do Vírus , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Animais , Substituição de Aminoácidos , Linhagem Celular , Mutação , Proteólise , Fusão de Membrana
13.
Biomolecules ; 14(5)2024 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-38786007

RESUMO

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.


Assuntos
Fusão de Membrana , Proteínas SNARE , Vesículas Sinápticas , Vesículas Sinápticas/metabolismo , Proteínas SNARE/metabolismo , Transmissão Sináptica , Animais , Humanos
14.
J Phys Chem B ; 128(20): 4986-4995, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38739415

RESUMO

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.


Assuntos
Colesterol , Fusão de Membrana , Fusão de Membrana/efeitos dos fármacos , Colesterol/química , Peptídeos/química , Peptídeos/farmacologia , Lipossomas Unilamelares/química , Lipossomas Unilamelares/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/química
15.
J Phys Chem B ; 128(20): 4975-4985, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38743687

RESUMO

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.


Assuntos
Bicamadas Lipídicas , Simulação de Dinâmica Molecular , Serotonina , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Serotonina/química , Serotonina/metabolismo , Fusão de Membrana , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/química , Espectrometria de Fluorescência , Interações Hidrofóbicas e Hidrofílicas
16.
Nat Commun ; 15(1): 4267, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769317

RESUMO

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.


Assuntos
Lipossomos , Fusão de Membrana , Lipossomos/metabolismo , Animais , Camundongos , Humanos , Endocitose , Transfecção , Edição de Genes/métodos , Coroa de Proteína/metabolismo , Coroa de Proteína/química , Incrustação Biológica/prevenção & controle , Feminino , Lipídeos/química
17.
Curr Opin Cell Biol ; 88: 102365, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38705050

RESUMO

Vesicle transport at the Golgi apparatus is a well-described process, and the major protein components involved have been identified. This includes the coat proteins that function in cargo sorting and vesicle formation, and the proteins that mediate the downstream events of vesicle tethering and membrane fusion. However, despite this knowledge, there remain significant gaps in our mechanistic understanding of these processes which includes how they are coordinated in space and time. In this review we discuss recent advances that have provided new insights into the mechanisms of Golgi trafficking, focussing on vesicle formation and cargo sorting, and vesicle tethering and fusion. These studies point to a high degree of spatial organisation of trafficking components at the Golgi and indicate an inherent plasticity of trafficking. Going forward, further advancements in technology and more sophisticated functional assays are expected to yield greater understanding of the mechanisms that govern Golgi trafficking events.


Assuntos
Complexo de Golgi , Complexo de Golgi/metabolismo , Humanos , Animais , Transporte Proteico , Transporte Biológico , Vesículas Transportadoras/metabolismo , Fusão de Membrana
18.
Nat Commun ; 15(1): 4132, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755165

RESUMO

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.


Assuntos
Fusão de Membrana , Proteínas Munc18 , Proteínas do Tecido Nervoso , Proteínas SNARE , Proteínas Munc18/metabolismo , Proteínas Munc18/genética , Proteínas SNARE/metabolismo , Proteínas SNARE/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Animais , Humanos , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/genética , Ratos
19.
ACS Nano ; 18(20): 12737-12748, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38717305

RESUMO

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.


Assuntos
Fusão de Membrana , Fosfatidilinositol 4,5-Difosfato , Eletricidade Estática , Água , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/química , Água/química , Lipossomos/química , Proteínas SNARE/metabolismo , Proteínas SNARE/química , Catálise
20.
Nat Commun ; 15(1): 4524, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38806492

RESUMO

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.


Assuntos
Bicamadas Lipídicas , Fusão de Membrana , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Íons/química , Membranas Artificiais , Hidrogéis/química , Pressão , Peptídeos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...