Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(5): 1140-1155.e18, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32814015

RESUMEN

The endosomal sorting complex required for transport-III (ESCRT-III) catalyzes membrane fission from within membrane necks, a process that is essential for many cellular functions, from cell division to lysosome degradation and autophagy. How it breaks membranes, though, remains unknown. Here, we characterize a sequential polymerization of ESCRT-III subunits that, driven by a recruitment cascade and by continuous subunit-turnover powered by the ATPase Vps4, induces membrane deformation and fission. During this process, the exchange of Vps24 for Did2 induces a tilt in the polymer-membrane interface, which triggers transition from flat spiral polymers to helical filament to drive the formation of membrane protrusions, and ends with the formation of a highly constricted Did2-Ist1 co-polymer that we show is competent to promote fission when bound on the inside of membrane necks. Overall, our results suggest a mechanism of stepwise changes in ESCRT-III filament structure and mechanical properties via exchange of the filament subunits to catalyze ESCRT-III activity.


Asunto(s)
Membrana Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Fusión de Membrana/fisiología , Adenosina Trifosfatasas/metabolismo , Línea Celular Tumoral , Endosomas/metabolismo , Células HeLa , Humanos , Polimerizacion , Transporte de Proteínas/fisiología
2.
Nat Rev Mol Cell Biol ; 19(5): 313-326, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29410531

RESUMEN

Clathrin-mediated endocytosis is a key process in vesicular trafficking that transports a wide range of cargo molecules from the cell surface to the interior. Clathrin-mediated endocytosis was first described over 5 decades ago. Since its discovery, over 50 proteins have been shown to be part of the molecular machinery that generates the clathrin-coated endocytic vesicles. These proteins and the different steps of the endocytic process that they mediate have been studied in detail. However, we still lack a good understanding of how all these different components work together in a highly coordinated manner to drive vesicle formation. Nevertheless, studies in recent years have provided several important insights into how endocytic vesicles are built, starting from initiation, cargo loading and the mechanisms governing membrane bending to membrane scission and the release of the vesicle into the cytoplasm.


Asunto(s)
Vesículas Cubiertas por Clatrina/metabolismo , Clatrina/metabolismo , Endocitosis/fisiología , Vesículas Transportadoras/metabolismo , Animales , Transporte Biológico/fisiología , Membrana Celular/metabolismo , Membrana Celular/fisiología , Humanos
3.
Cell ; 163(4): 866-79, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26522593

RESUMEN

ESCRT-III is required for lipid membrane remodeling in many cellular processes, from abscission to viral budding and multi-vesicular body biogenesis. However, how ESCRT-III polymerization generates membrane curvature remains debated. Here, we show that Snf7, the main component of ESCRT-III, polymerizes into spirals at the surface of lipid bilayers. When covering the entire membrane surface, these spirals stopped growing when densely packed: they had a polygonal shape, suggesting that lateral compression could deform them. We reasoned that Snf7 spirals could function as spiral springs. By measuring the polymerization energy and the rigidity of Snf7 filaments, we showed that they were deformed while growing in a confined area. Furthermore, we observed that the elastic expansion of compressed Snf7 spirals generated an area difference between the two sides of the membrane and thus curvature. This spring-like activity underlies the driving force by which ESCRT-III could mediate membrane deformation and fission.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/ultraestructura , Membrana Dobles de Lípidos/química , Modelos Moleculares , Levaduras/metabolismo , Membranas Intracelulares/química , Liberación del Virus , Levaduras/citología
4.
Cell ; 156(5): 882-92, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581490

RESUMEN

Biological membranes undergo constant remodeling by membrane fission and fusion to change their shape and to exchange material between subcellular compartments. During clathrin-mediated endocytosis, the dynamic assembly and disassembly of protein scaffolds comprising members of the bin-amphiphysin-rvs (BAR) domain protein superfamily constrain the membrane into distinct shapes as the pathway progresses toward fission by the GTPase dynamin. In this Review, we discuss how BAR domain protein assembly and disassembly are controlled in space and time and which structural and biochemical features allow the tight regulation of their shape and function to enable dynamin-mediated membrane fission.


Asunto(s)
Membrana Celular/metabolismo , Dinaminas/metabolismo , Animales , Vesículas Cubiertas por Clatrina/metabolismo , Endocitosis , Humanos , Estructura Terciaria de Proteína
5.
EMBO J ; 42(2): e112287, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36644906

RESUMEN

Proteins exit from endosomes through tubular carriers coated by retromer, a complex that impacts cellular signaling, lysosomal biogenesis and numerous diseases. The coat must overcome membrane tension to form tubules. We explored the dynamics and driving force of this process by reconstituting coat formation with yeast retromer and the BAR-domain sorting nexins Vps5 and Vps17 on oriented synthetic lipid tubules. This coat oligomerizes bidirectionally, forming a static tubular structure that does not exchange subunits. High concentrations of sorting nexins alone constrict membrane tubes to an invariant radius of 19 nm. At lower concentrations, oligomers of retromer must bind and interconnect the sorting nexins to drive constriction. Constricting less curved membranes into tubes, which requires more energy, coincides with an increased surface density of retromer on the sorting nexin layer. Retromer-mediated crosslinking of sorting nexins at variable densities may thus tune the energy that the coat can generate to deform the membrane. In line with this, genetic ablation of retromer oligomerization impairs endosomal protein exit in yeast and human cells.


Asunto(s)
Saccharomyces cerevisiae , Nexinas de Clasificación , Humanos , Transporte de Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Nexinas de Clasificación/genética , Nexinas de Clasificación/metabolismo , Constricción , Endosomas/metabolismo
6.
Cell ; 151(3): 619-29, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23101629

RESUMEN

The GTPase dynamin polymerizes into a helical coat that constricts membrane necks of endocytic pits to promote their fission. However, the dynamin mechanism is still debated because constriction is necessary but not sufficient for fission. Here, we show that fission occurs at the interface between the dynamin coat and the uncoated membrane. At this location, the considerable change in membrane curvature increases the local membrane elastic energy, reducing the energy barrier for fission. Fission kinetics depends on tension, bending rigidity, and the dynamin constriction torque. Indeed, we experimentally find that the fission rate depends on membrane tension in vitro and during endocytosis in vivo. By estimating the energy barrier from the increased elastic energy at the edge of dynamin and measuring the dynamin torque, we show that the mechanical energy spent on dynamin constriction can reduce the energy barrier for fission sufficiently to promote spontaneous fission. :


Asunto(s)
Membrana Celular/metabolismo , Dinaminas/metabolismo , Endocitosis , Modelos Biológicos , Animales , Células COS , Chlorocebus aethiops , Guanosina Trifosfato/metabolismo , Proteínas SNARE/metabolismo
7.
Proc Natl Acad Sci U S A ; 120(43): e2309698120, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37844218

RESUMEN

Mutations in Leucine-rich repeat kinase 2 (LRRK2) are responsible for late-onset autosomal dominant Parkinson's disease. LRRK2 has been implicated in a wide range of physiological processes including membrane repair in the endolysosomal system. Here, using cell-free systems, we report that purified LRRK2 directly binds acidic lipid bilayers with a preference for highly curved bilayers. While this binding is nucleotide independent, LRRK2 can also deform low-curvature liposomes into narrow tubules in a guanylnucleotide-dependent but Adenosine 5'-triphosphate-independent way. Moreover, assembly of LRRK2 into scaffolds at the surface of lipid tubules can constrict them. We suggest that an interplay between the membrane remodeling and signaling properties of LRRK2 may be key to its physiological function. LRRK2, via its kinase activity, may achieve its signaling role at sites where membrane remodeling occurs.


Asunto(s)
Enfermedad de Parkinson , Proteínas Serina-Treonina Quinasas , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Fosforilación , Mutación
8.
Proc Natl Acad Sci U S A ; 120(39): e2300416120, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37725653

RESUMEN

The shape of cells is the outcome of the balance of inner forces produced by the actomyosin network and the resistive forces produced by cell adhesion to their environment. The specific contributions of contractile, anchoring and friction forces to network deformation rate and orientation are difficult to disentangle in living cells where they influence each other. Here, we reconstituted contractile actomyosin networks in vitro to study specifically the role of the friction forces between the network and its anchoring substrate. To modulate the magnitude and spatial distribution of friction forces, we used glass or lipids surface micropatterning to control the initial shape of the network. We adapted the concentration of Nucleating Promoting Factor on each surface to induce the assembly of actin networks of similar densities and compare the deformation of the network toward the centroid of the pattern shape upon myosin-induced contraction. We found that actin network deformation was faster and more coordinated on lipid bilayers than on glass, showing the resistance of friction to network contraction. To further study the role of the spatial distribution of these friction forces, we designed heterogeneous micropatterns made of glass and lipids. The deformation upon contraction was no longer symmetric but biased toward the region of higher friction. Furthermore, we showed that the pattern of friction could robustly drive network contraction and dominate the contribution of asymmetric distributions of myosins. Therefore, we demonstrate that during contraction, both the active and resistive forces are essential to direct the actin network deformation.


Asunto(s)
Actinas , Actomiosina , Fricción , Contracción Muscular , Membrana Dobles de Lípidos
9.
Nature ; 571(7765): 429-433, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292547

RESUMEN

Balanced fusion and fission are key for the proper function and physiology of mitochondria1,2. Remodelling of the mitochondrial inner membrane is mediated by the dynamin-like protein mitochondrial genome maintenance 1 (Mgm1) in fungi or the related protein optic atrophy 1 (OPA1) in animals3-5. Mgm1 is required for the preservation of mitochondrial DNA in yeast6, whereas mutations in the OPA1 gene in humans are a common cause of autosomal dominant optic atrophy-a genetic disorder that affects the optic nerve7,8. Mgm1 and OPA1 are present in mitochondria as a membrane-integral long form and a short form that is soluble in the intermembrane space. Yeast strains that express temperature-sensitive mutants of Mgm19,10 or mammalian cells that lack OPA1 display fragmented mitochondria11,12, which suggests that Mgm1 and OPA1 have an important role in inner-membrane fusion. Consistently, only the mitochondrial outer membrane-not the inner membrane-fuses in the absence of functional Mgm113. Mgm1 and OPA1 have also been shown to maintain proper cristae architecture10,14; for example, OPA1 prevents the release of pro-apoptotic factors by tightening crista junctions15. Finally, the short form of OPA1 localizes to mitochondrial constriction sites, where it presumably promotes mitochondrial fission16. How Mgm1 and OPA1 perform their diverse functions in membrane fusion, scission and cristae organization is at present unknown. Here we present crystal and electron cryo-tomography structures of Mgm1 from Chaetomium thermophilum. Mgm1 consists of a GTPase (G) domain, a bundle signalling element domain, a stalk, and a paddle domain that contains a membrane-binding site. Biochemical and cell-based experiments demonstrate that the Mgm1 stalk mediates the assembly of bent tetramers into helical filaments. Electron cryo-tomography studies of Mgm1-decorated lipid tubes and fluorescence microscopy experiments on reconstituted membrane tubes indicate how the tetramers assemble on positively or negatively curved membranes. Our findings convey how Mgm1 and OPA1 filaments dynamically remodel the mitochondrial inner membrane.


Asunto(s)
Chaetomium/química , Microscopía por Crioelectrón , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Proteínas de Unión al GTP/química , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Cristalografía por Rayos X , Proteínas Fúngicas/ultraestructura , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/ultraestructura , Galactosilceramidas/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/ultraestructura , Modelos Moleculares , Dominios Proteicos , Multimerización de Proteína
10.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34785592

RESUMEN

During osmotic changes of their environment, cells actively regulate their volume and plasma membrane tension that can passively change through osmosis. How tension and volume are coupled during osmotic adaptation remains unknown, as their quantitative characterization is lacking. Here, we performed dynamic membrane tension and cell volume measurements during osmotic shocks. During the first few seconds following the shock, cell volume varied to equilibrate osmotic pressures inside and outside the cell, and membrane tension dynamically followed these changes. A theoretical model based on the passive, reversible unfolding of the membrane as it detaches from the actin cortex during volume increase quantitatively describes our data. After the initial response, tension and volume recovered from hypoosmotic shocks but not from hyperosmotic shocks. Using a fluorescent membrane tension probe (fluorescent lipid tension reporter [Flipper-TR]), we investigated the coupling between tension and volume during these asymmetric recoveries. Caveolae depletion and pharmacological inhibition of ion transporters and channels, mTORCs, and the cytoskeleton all affected tension and volume responses. Treatments targeting mTORC2 and specific downstream effectors caused identical changes to both tension and volume responses, their coupling remaining the same. This supports that the coupling of tension and volume responses to osmotic shocks is primarily regulated by mTORC2.


Asunto(s)
Tamaño de la Célula , Membranas/metabolismo , Ósmosis/fisiología , Actinas/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Células HeLa , Humanos , Membranas/efectos de los fármacos , Modelos Teóricos , Presión Osmótica/fisiología
11.
Biophys J ; 122(4): 624-631, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36659849

RESUMEN

In biology, release of Ca2+ ions in the cytosol is essential to trigger or control many cell functions. Calcium signaling acutely depends on lipid membrane permeability to Ca2+. For proper understanding of membrane permeability to Ca2+, both membrane hydration and the structure of the hydrophobic core must be taken into account. Here, we vary the hydrophobic core of bilayer membranes and observe different types of behavior in high-throughput wide-field second harmonic imaging. Ca2+ translocation is observed through mono-unsaturated (DOPC:DOPA) membranes, reduced upon the addition of cholesterol, and completely inhibited for branched (DPhPC:DPhPA) and poly-unsaturated (SLPC:SLPA) lipid membranes. We propose, using molecular dynamics simulations, that ion transport occurs through ion-induced transient pores, which requires nonequilibrium membrane restructuring. This results in different rates at different locations and suggests that the hydrophobic structure of lipids plays a much more sophisticated regulating role than previously thought.


Asunto(s)
Membrana Dobles de Lípidos , Microscopía de Generación del Segundo Armónico , Membrana Dobles de Lípidos/química , Microscopía , Iones , Colesterol/química , Simulación de Dinámica Molecular
12.
Nat Mater ; 21(5): 588-597, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35145258

RESUMEN

Tissues acquire function and shape via differentiation and morphogenesis. Both processes are driven by coordinating cellular forces and shapes at the tissue scale, but general principles governing this interplay remain to be discovered. Here we report that self-organization of myoblasts around integer topological defects, namely spirals and asters, suffices to establish complex multicellular architectures. In particular, these arrangements can trigger localized cell differentiation or, alternatively, when differentiation is inhibited, they can drive the growth of swirling protrusions. Both localized differentiation and growth of cellular vortices require specific stress patterns. By analysing the experimental velocity and orientational fields through active gel theory, we show that integer topological defects can generate force gradients that concentrate compressive stresses. We reveal these gradients by assessing spatial changes in nuclear volume and deformations of elastic pillars. We propose integer topological defects as mechanical organizing centres controlling differentiation and morphogenesis.


Asunto(s)
Citoesqueleto , Diferenciación Celular , Morfogénesis
13.
Phys Rev Lett ; 131(26): 268301, 2023 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-38215373

RESUMEN

Topological defects in active polar fluids can organize spontaneous flows and influence macroscopic density patterns. Both of them play an important role during animal development. Yet the influence of density on active flows is poorly understood. Motivated by experiments on cell monolayers confined to disks, we study the coupling between density and polar order for a compressible active polar fluid in the presence of a +1 topological defect. As in the experiments, we find a density-controlled spiral-to-aster transition. In addition, biphasic orientational phases emerge as a generic outcome of such coupling. Our results highlight the importance of density gradients as a potential mechanism for controlling flow and orientational patterns in biological systems.

14.
PLoS Comput Biol ; 18(10): e1010586, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36251703

RESUMEN

ESCRT-III filaments are composite cytoskeletal polymers that can constrict and cut cell membranes from the inside of the membrane neck. Membrane-bound ESCRT-III filaments undergo a series of dramatic composition and geometry changes in the presence of an ATP-consuming Vps4 enzyme, which causes stepwise changes in the membrane morphology. We set out to understand the physical mechanisms involved in translating the changes in ESCRT-III polymer composition into membrane deformation. We have built a coarse-grained model in which ESCRT-III polymers of different geometries and mechanical properties are allowed to copolymerise and bind to a deformable membrane. By modelling ATP-driven stepwise depolymerisation of specific polymers, we identify mechanical regimes in which changes in filament composition trigger the associated membrane transition from a flat to a buckled state, and then to a tubule state that eventually undergoes scission to release a small cargo-loaded vesicle. We then characterise how the location and kinetics of polymer loss affects the extent of membrane deformation and the efficiency of membrane neck scission. Our results identify the near-minimal mechanical conditions for the operation of shape-shifting composite polymers that sever membrane necks.


Asunto(s)
Citoesqueleto , Complejos de Clasificación Endosomal Requeridos para el Transporte , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Polimerizacion , Citoesqueleto/metabolismo , Membrana Celular/metabolismo , Adenosina Trifosfato/metabolismo , Polímeros
15.
Cell ; 135(7): 1163-5, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-19109885

RESUMEN

During membrane fission, the GTPase dynamin forms helical assemblies at the neck of membrane buds. Although it has been proposed that fission results from the constriction of the dynamin helix, new work by Bashkirov et al. (2008) and Pucadyil and Schmid (2008) unexpectedly shows that helix disassembly is also necessary for membrane fission.


Asunto(s)
Dinaminas/metabolismo , Membranas Intracelulares/metabolismo , Animales , Membrana Celular/metabolismo , Membrana Dobles de Lípidos/metabolismo , Modelos Biológicos
16.
Cell ; 132(5): 807-17, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18329367

RESUMEN

BAR superfamily domains shape membranes through poorly understood mechanisms. We solved structures of F-BAR modules bound to flat and curved bilayers using electron (cryo)microscopy. We show that membrane tubules form when F-BARs polymerize into helical coats that are held together by lateral and tip-to-tip interactions. On gel-state membranes or after mutation of residues along the lateral interaction surface, F-BARs adsorb onto bilayers via surfaces other than their concave face. We conclude that membrane binding is separable from membrane bending, and that imposition of the module's concave surface forces fluid-phase bilayers to bend locally. Furthermore, exposure of the domain's lateral interaction surface through a change in orientation serves as the crucial trigger for assembly of the helical coat and propagation of bilayer bending. The geometric constraints and sequential assembly of the helical lattice explain how F-BAR and classical BAR domains segregate into distinct microdomains, and provide insight into the spatial regulation of membrane invagination.


Asunto(s)
Membrana Celular/química , Membrana Celular/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Animales , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Membrana Celular/ultraestructura , Chlorocebus aethiops , Microscopía por Crioelectrón , Dinaminas/metabolismo , Proteínas de Unión a Ácidos Grasos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Liposomas/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Antígenos de Histocompatibilidad Menor , Modelos Biológicos , Modelos Moleculares , Estructura Terciaria de Proteína , Transfección
17.
J Cell Sci ; 133(8)2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32184262

RESUMEN

The ESCRT-III protein complex executes reverse-topology membrane scission. The scission mechanism is unclear but is linked to remodeling of ESCRT-III complexes at the membrane surface. At endosomes, ESCRT-III mediates the budding of intralumenal vesicles (ILVs). In Saccharomyces cerevisiae, ESCRT-III activity at endosomes is regulated through an unknown mechanism by Doa4, an ubiquitin hydrolase that deubiquitylates transmembrane proteins sorted into ILVs. We report that the non-catalytic N-terminus of Doa4 binds Snf7, the predominant ESCRT-III subunit. Through this interaction, Doa4 overexpression alters Snf7 assembly status and inhibits ILV membrane scission. In vitro, the Doa4 N-terminus inhibits association of Snf7 with Vps2, which functions with Vps24 to arrest Snf7 polymerization and remodel Snf7 polymer structure. In vivo, Doa4 overexpression inhibits Snf7 interaction with Vps2 and also with the ATPase Vps4, which is recruited by Vps2 and Vps24 to remodel ESCRT-III complexes by catalyzing subunit turnover. Our data suggest a mechanism by which the deubiquitylation machinery regulates ILV biogenesis by interfering with ESCRT-III remodeling.


Asunto(s)
Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Ubiquitina Tiolesterasa/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Endosomas/metabolismo , Hidrolasas/metabolismo , Transporte de Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitina/metabolismo
18.
Chembiochem ; 23(15): e202200192, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35535626

RESUMEN

Labeled ammonium cations with pKa ∼7.4 accumulate in acidic organelles because they can be neutralized transiently to cross the membrane at cytosolic pH 7.2 but not at their internal pH<5.5. Retention in early endosomes with less acidic internal pH was achieved recently using weaker acids of up to pKa 9.8. We report here that primary ammonium cations with higher pKa 10.6, label early endosomes more efficiently. This maximized early endosome tracking coincides with increasing labeling of Golgi networks with similarly weak internal acidity. Guanidinium cations with pKa 13.5 cannot cross the plasma membrane in monomeric form and label the plasma membrane with selectivity for vesicles embarking into endocytosis. Self-assembled into micelles, guanidinium cations enter cells like arginine-rich cell-penetrating peptides and, driven by their membrane potential, penetrate mitochondria unidirectionally despite their high inner pH. The resulting tracking rules with an approximated dynamic range of pKa change ∼3.5 are expected to be generally valid, thus enabling the design of chemistry tools for biology research in the broadest sense. From a practical point of view, most relevant are two complementary fluorescent flipper probes that can be used to image the mechanics at the very beginning of endocytosis.


Asunto(s)
Compuestos de Amonio , Endocitosis , Ácidos , Compuestos de Amonio/metabolismo , Cationes/metabolismo , Endosomas/metabolismo , Guanidina , Concentración de Iones de Hidrógeno
19.
Phys Rev Lett ; 129(26): 268101, 2022 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-36608212

RESUMEN

The sequential exchange of filament composition to increase filament curvature was proposed as a mechanism for how some biological polymers deform and cut membranes. The relationship between the filament composition and its mechanical effect is lacking. We develop a kinetic model for the assembly of composite filaments that includes protein-membrane adhesion, filament mechanics and membrane mechanics. We identify the physical conditions for such a membrane remodeling and show this mechanism of sequential polymer assembly lowers the energetic barrier for membrane deformation.


Asunto(s)
Citoesqueleto , Polímeros , Membranas , Polímeros/química
20.
Proc Natl Acad Sci U S A ; 116(26): 12845-12850, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31189604

RESUMEN

Dynamin proteins assemble into characteristic helical structures around necks of clathrin-coated membrane buds. Hydrolysis of dynamin-bound GTP results in both fission of the membrane neck and partial disruption of the dynamin oligomer. Imaging by atomic force microscopy reveals that, on GTP hydrolysis, dynamin oligomers undergo a dynamic remodeling and lose their distinctive helical shape. While breakup of the dynamin helix is a critical stage in clathrin-mediated endocytosis, the mechanism for this remodeling of the oligomer has not been resolved. In this paper, we formulate an analytical, elasticity-based model for the reshaping and disassembly of the dynamin scaffold. We predict that the shape of the oligomer is modulated by the orientation of dynamin's pleckstrin homology (PH) domain relative to the underlying membrane. Our results indicate that tilt of the PH domain drives deformation and fragmentation of the oligomer, in agreement with experimental observations. This model motivated the introduction of the tilted helix: a curve that maintains a fixed angle between its normal and the normal of the embedding surface. Our findings highlight the importance of tilt as a key regulator of size and morphology of membrane-bound oligomers.


Asunto(s)
Dinaminas/química , Elasticidad , Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Dominios Homólogos a Pleckstrina , Conformación Proteica en Hélice alfa , Subunidades de Proteína/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA