Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 28(8): R417-R420, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29689226

RESUMEN

Cells are largely compartmentalized into numerous interacting organelles with dedicated functions in lipid metabolism, energy generation, or protein turnover. In the past, each organelle has been considered as an isolated unit with an individual proteome, membrane composition, and shape. However, this view is changing rapidly as organelles communicate via contact sites, fuse directly with each other, or correspond via vesicular carriers. Each of these processes disturbs the initial individual character of each organelle and they thus need to be tightly controlled and regulated.


Asunto(s)
Fusión de Membrana/fisiología , Orgánulos/metabolismo , Orgánulos/fisiología , Animales , Humanos , Membranas Intracelulares/metabolismo , Metabolismo de los Lípidos/fisiología , Subunidades de Proteína/metabolismo , Subunidades de Proteína/fisiología , Transporte de Proteínas/fisiología
2.
Nature ; 551(7682): 634-638, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29088698

RESUMEN

Membrane fusion in eukaryotic cells mediates the biogenesis of organelles, vesicular traffic between them, and exo- and endocytosis of important signalling molecules, such as hormones and neurotransmitters. Distinct tasks in intracellular membrane fusion have been assigned to conserved protein systems. Tethering proteins mediate the initial recognition and attachment of membranes, whereas SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) protein complexes are considered as the core fusion engine. SNARE complexes provide mechanical energy to distort membranes and drive them through a hemifusion intermediate towards the formation of a fusion pore. This last step is highly energy-demanding. Here we combine the in vivo and in vitro fusion of yeast vacuoles with molecular simulations to show that tethering proteins are critical for overcoming the final energy barrier to fusion pore formation. SNAREs alone drive vacuoles only into the hemifused state. Tethering proteins greatly increase the volume of SNARE complexes and deform the site of hemifusion, which lowers the energy barrier for pore opening and provides the driving force. Thereby, tethering proteins assume a crucial mechanical role in the terminal stage of membrane fusion that is likely to be conserved at multiple steps of vesicular traffic. We therefore propose that SNAREs and tethering proteins should be considered as a single, non-dissociable device that drives fusion. The core fusion machinery may then be larger and more complex than previously thought.


Asunto(s)
Fusión de Membrana , Proteínas SNARE/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Ligandos , Simulación de Dinámica Molecular , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Vacuolas/metabolismo
3.
Mol Biol Cell ; 28(2): 322-332, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27852901

RESUMEN

Membrane fusion at endomembranes requires cross-talk between Rab GTPases and tethers to drive SNARE-mediated lipid bilayer mixing. Several tethers have multiple Rab-binding sites with largely untested function. Here we dissected the lysosomal HOPS complex as a tethering complex with just two binding sites for the Rab7-like Ypt7 protein to determine their relevance for fusion. Using tethering and fusion assays combined with HOPS mutants, we show that HOPS-dependent fusion requires both Rab-binding sites, with Vps39 being the stronger Ypt7 interactor than Vps41. The intrinsic amphipathic lipid packaging sensor (ALPS) motif within HOPS Vps41, a target of the vacuolar kinase Yck3, is dispensable for tethering and fusion but can affect tethering if phosphorylated. In combination, our data demonstrate that a multivalent tethering complex uses its two Rab bindings to determine the place of SNARE assembly and thus fusion at endomembranes.


Asunto(s)
Fusión de Membrana/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Sitios de Unión , Endosomas/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas/fisiología , Proteínas SNARE/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Vacuolas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/fisiología
4.
J Biol Chem ; 290(9): 5405-13, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25564619

RESUMEN

Membrane fusion at vacuoles requires a consecutive action of the HOPS tethering complex, which is recruited by the Rab GTPase Ypt7, and vacuolar SNAREs to drive membrane fusion. It is assumed that the Sec1/Munc18-like Vps33 within the HOPS complex is largely responsible for SNARE chaperoning. Here, we present direct evidence for HOPS binding to SNAREs and the Habc domain of the Vam3 SNARE protein, which may explain its function during fusion. We show that HOPS interacts strongly with the Vam3 Habc domain, assembled Q-SNAREs, and the R-SNARE Ykt6, but not the Q-SNARE Vti1 or the Vam3 SNARE domain. Electron microscopy combined with Nanogold labeling reveals that the binding sites for vacuolar SNAREs and the Habc domain are located in the large head of the HOPS complex, where Vps16 and Vps33 have been identified before. Competition experiments suggest that HOPS bound to the Habc domain can still interact with assembled Q-SNAREs, whereas Q-SNARE binding prevents recognition of the Habc domain. In agreement, membranes carrying Vam3ΔHabc fuse poorly unless an excess of HOPS is provided. These data suggest that the Habc domain of Vam3 facilitates the assembly of the HOPS/SNARE machinery at fusion sites and thus supports efficient membrane fusion.


Asunto(s)
Proteínas Qa-SNARE/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Vacuolas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Sitios de Unión , Immunoblotting , Fusión de Membrana , Microscopía Electrónica , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Qa-SNARE/química , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Transporte Vesicular/química , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/metabolismo
5.
J Biol Chem ; 289(48): 33503-12, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25324549

RESUMEN

Membrane fusion at the vacuole, the lysosome equivalent in yeast, requires the HOPS tethering complex, which is recruited by the Rab7 GTPase Ypt7. HOPS provides a template for the assembly of SNAREs and thus likely confers fusion at a distinct position on vacuoles. Five of the six subunits in HOPS have a similar domain prediction with strong similarity to COPII subunits and nuclear porins. Here, we show that Vps18 indeed has a seven-bladed ß-propeller as its N-terminal domain by revealing its structure at 2.14 Å. The Vps18 N-terminal domain can interact with the N-terminal part of Vps11 and also binds to lipids. Although deletion of the Vps18 N-terminal domain does not preclude HOPS assembly, as revealed by negative stain electron microscopy, the complex is instable and cannot support membrane fusion in vitro. We thus conclude that the ß-propeller of Vps18 is required for HOPS stability and function and that it can serve as a starting point for further structural analyses of the HOPS tethering complex.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/química , Complejos Multiproteicos/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/química , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Estabilidad Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(10): 3823-8, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23417307

RESUMEN

Membrane fusion along the endocytic pathway occurs in a sequence of tethering, docking, and fusion. At endosomes and vacuoles, the CORVET (class C core vacuole/endosome tethering) and HOPS (homotypic fusion and vacuole protein sorting) tethering complexes require their organelle-specific Rabs for localization and function. Until now, despite the absence of experimental evidence, it has been assumed that CORVET is a membrane-tethering factor. To test this theory and understand the mechanistic analogies with the HOPS complex, we set up an in vitro system, and establish CORVET as a bona-fide tether for Vps21-positive endosome/vacuole membranes. Purified CORVET binds to SNAREs and Rab5/Vps21-GTP. We then demonstrate that purified CORVET can specifically tether Vps21-positive membranes. Tethering via CORVET is dose-dependent, stimulated by the GEF Vps9, and inhibited by Msb3, the Vps21-GAP. Moreover, CORVET supports fusion of isolated membranes containing Vps21. In agreement with its role as a tether, overexpressed CORVET drives Vps21, but not the HOPS-specific Ypt7 into contact sites between vacuoles, which likely represent vacuole-associated endosomes. We therefore conclude that CORVET is a tethering complex that promotes fusion of Rab5-positive membranes and thus facilitates receptor down-regulation and recycling at the late endosome.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/química , Proteínas de Unión al GTP rab5/metabolismo , Endocitosis , Endosomas/metabolismo , Lisosomas/metabolismo , Fusión de Membrana , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas SNARE/química , Proteínas SNARE/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab5/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA