Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Rep ; 5: 15926, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26639035

RESUMEN

Sphingolipid metabolites are involved in the regulation of autophagy, a degradative recycling process that is required to prevent neuronal degeneration. Drosophila blue cheese mutants neurodegenerate due to perturbations in autophagic flux, and consequent accumulation of ubiquitinated aggregates. Here, we demonstrate that blue cheese mutant brains exhibit an elevation in total ceramide levels; surprisingly, however, degeneration is ameliorated when the pool of available ceramides is further increased, and exacerbated when ceramide levels are decreased by altering sphingolipid catabolism or blocking de novo synthesis. Exogenous ceramide is seen to accumulate in autophagosomes, which are fewer in number and show less efficient clearance in blue cheese mutant neurons. Sphingolipid metabolism is also shifted away from salvage toward de novo pathways, while pro-growth Akt and MAP pathways are down-regulated, and ER stress is increased. All these defects are reversed under genetic rescue conditions that increase ceramide generation from salvage pathways. This constellation of effects suggests a possible mechanism whereby the observed deficit in a potentially ceramide-releasing autophagic pathway impedes survival signaling and exacerbates neuronal death.


Asunto(s)
Autofagia , Ceramidas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/genética , Transducción de Señal , Estrés Fisiológico , Animales , Células Cultivadas , Ceramidasas/metabolismo , Regulación hacia Abajo , Drosophila melanogaster/enzimología , Técnicas de Silenciamiento del Gen , Metabolismo de los Lípidos , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Degeneración Nerviosa/patología , Neuronas/metabolismo , Fagosomas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo
2.
ChemSusChem ; 8(21): 3718-26, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26404512

RESUMEN

Butanol is an ideal biofuel, although poor titers lead to high recovery costs by distillation. Fluidization of microbial membranes by butanol is one of the major factors limiting titers in butanol-producing bioprocesses. Starting with the hypothesis that certain membrane insertion molecules would stabilize the lipid bilayer in the presence of butanol, we applied a combination of in vivo and in vitro techniques within an in silico framework to describe a new approach to achieve solvent tolerance in bacteria. Single-molecule tracking of a model supported bilayer showed that COE1-5C, a five-ringed oligo-polyphenylenevinylene conjugated oligoelectrolyte (COE), reduced the diffusion rate of phospholipids in a microbially derived lipid bilayer to a greater extent than three-ringed and four-ringed COEs. Furthermore, COE1-5C treatment increased the specific growth rate of E. coli K12 relative to a control at inhibitory butanol concentrations. Consequently, to confer butanol tolerance to microbes by exogenous means is complementary to genetic modification of strains in industrial bioprocesses, extends the physiological range of microbes to match favorable bioprocess conditions, and is amenable with complex and undefined microbial consortia for biobutanol production. Molecular dynamics simulations indicated that the π-conjugated aromatic backbone of COE1-5C likely acts as a hydrophobic tether for glycerophospholipid acyl chains by enhancing bilayer integrity in the presence of high butanol concentrations, which thereby counters membrane fluidization. COE1-5C-mitigated E. coli K12 membrane depolarization by butanol is consistent with the hypothesis that improved growth rates in the presence of butanol are a consequence of improved bilayer stability.


Asunto(s)
Butanoles/toxicidad , Membrana Celular/química , Escherichia coli K12/efectos de los fármacos , Microbiología Industrial/métodos , Membrana Dobles de Lípidos/química , Polivinilos/química , Biocombustibles , Butanoles/metabolismo , Membrana Celular/metabolismo , Escherichia coli K12/crecimiento & desarrollo , Escherichia coli K12/metabolismo , Fermentación , Membrana Dobles de Lípidos/metabolismo , Fluidez de la Membrana/efectos de los fármacos , Simulación de Dinámica Molecular
3.
Adv Healthc Mater ; 4(6): 911-7, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25650337

RESUMEN

A non-membrane protein-based nanoparticle agent for the tracking of lipid rafts on live cells is produced by stoichiometric functionalization of gold nanoparticles with a previously characterized sphingolipid- and cell membrane microdomain-binding domain peptide (SBD). The SBD peptide is inserted in a self-assembled monolayer of peptidol and alkane thiol ethylene glycol, on gold nanoparticles surface. The stoichiometric functionalization of nanoparticles with the SBD peptide, essential for single molecule tracking, is achieved by means of non-affinity nanoparticle purification. The SBD-nanoparticles have remarkable long-term resistance to electrolyte-induced aggregation and ligand-exchange and have no detectable non-specific binding to live cells. Binding and diffusion of SBD-nanoparticles bound to the membrane of live cells is measured by real-time photothermal microscopy and shows the dynamics of sphingolipid-enriched microdomains on cells membrane, with evidence for clustering, splitting, and diffusion over time of the SBD-nanoparticle labeled membrane domains. The monofunctionalized SBD-nanoparticle is a promising targeting agent for the tracking of lipid rafts independently of their protein composition and the labelling requires no prior modification of the cells. This approach has potential for further functionalization of the particles to manipulate the organization of, or targeting to microdomains that control signaling events and thereby lead to novel diagnostics and therapeutics.


Asunto(s)
Oro/química , Microdominios de Membrana/metabolismo , Nanopartículas del Metal , Péptidos/metabolismo , Esfingolípidos/metabolismo , Secuencia de Aminoácidos , Microdominios de Membrana/química , Datos de Secuencia Molecular
4.
PLoS One ; 7(12): e51222, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251459

RESUMEN

Organized assembly or aggregation of sphingolipid-binding ligands, such as certain toxins and pathogens, has been suggested to increase binding affinity of the ligand to the cell membrane and cause membrane reorganization or distortion. Here we show that the diffusion behavior of the fluorescently tagged sphingolipid-interacting peptide probe SBD (Sphingolipid Binding Domain) is altered by modifications in the construction of the peptide sequence that both result in a reduction in binding to ganglioside-containing supported lipid membranes, and at the same time increase aggregation on the cell plasma membrane, but that do not change relative amounts of secondary structural features. We tested the effects of modifying the overall charge and construction of the SBD probe on its binding and diffusion behavior, by Surface Plasmon Resonance (SPR; Biacore) analysis on lipid surfaces, and by Fluorescence Correlation Spectroscopy (FCS) on live cells, respectively. SBD binds preferentially to membranes containing the highly sialylated gangliosides GT1b and GD1a. However, simple charge interactions of the peptide with the negative ganglioside do not appear to be a critical determinant of binding. Rather, an aggregation-suppressing amino acid composition and linker between the fluorophore and the peptide are required for optimum binding of the SBD to ganglioside-containing supported lipid bilayer surfaces, as well as for interaction with the membrane. Interestingly, the strength of interactions with ganglioside-containing artificial membranes is mirrored in the diffusion behavior by FCS on cell membranes, with stronger binders displaying similar characteristic diffusion profiles. Our findings indicate that for aggregation-prone peptides, aggregation occurs upon contact with the cell membrane, and rather than giving a stronger interaction with the membrane, aggregation is accompanied by weaker binding and complex diffusion profiles indicative of heterogeneous diffusion behavior in the probe population.


Asunto(s)
Glucolípidos/metabolismo , Péptidos/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Membrana Celular/metabolismo , Dicroismo Circular , Endocitosis , Humanos , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Estructura Secundaria de Proteína , Espectrometría de Fluorescencia/métodos , Resonancia por Plasmón de Superficie
5.
Biophys J ; 97(9): 2630-9, 2009 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-19883607

RESUMEN

Cell membrane organization is dynamic and is assumed to have different characteristic length scales. These length scales, which are influenced by lipid and protein composition as well as by the cytoskeleton, can range from below the optical resolution limit (as with rafts or microdomains) to far above the resolution limit (as with capping phenomena or the formation of lipid "platforms"). The measurement of these membrane features poses a significant problem because membrane dynamics are on the millisecond timescale and are thus beyond the time resolution of conventional imaging approaches. Fluorescence correlation spectroscopy (FCS), a widely used spectroscopic technique to measure membrane dynamics, has the required time resolution but lacks imaging capabilities. A promising solution is the recently introduced method known as imaging total internal reflection (ITIR)-FCS, which can probe diffusion phenomena in lipid membranes with good temporal and spatial resolution. In this work, we extend ITIR-FCS to perform ITIR fluorescence cross-correlation spectroscopy (ITIR-FCCS) between pixel areas of arbitrary shape and derive a generalized expression that is applicable to active transport and diffusion. ITIR-FCCS is applied to model systems exhibiting diffusion, active transport, or a combination of the two. To demonstrate its applicability to live cells, we observe the diffusion of a marker, the sphingolipid-binding domain (SBD) derived from the amyloid peptide Abeta, on live neuroblastoma cells. We investigate the organization and dynamics of SBD-bound lipid microdomains under the conditions of cholesterol removal and cytoskeleton disruption.


Asunto(s)
Biofisica/métodos , Membrana Celular/metabolismo , Espectrometría de Fluorescencia/métodos , Amiloide/química , Anisotropía , Transporte Biológico , Línea Celular Tumoral , Difusión , Humanos , Lípidos/química , Fluidez de la Membrana , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Neuroblastoma/metabolismo , Esfingolípidos/química
6.
J Cell Sci ; 122(Pt 20): 3715-28, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19773361

RESUMEN

Several cholesterol-dependent cellular uptake pathways involving microdomain-resident sphingolipids have been characterized, but little is known about what controls the further intracellular trafficking routes of those domains. Here, we present evidence that the uptake and intracellular trafficking of a recently described sphingolipid-binding probe, the sphingolipid binding domain (SBD) peptide, is mediated by two parallel cooperating mechanisms requiring flotillin, dynamin and cdc42, which act in concert to direct a distinct surface behavior and trafficking itinerary. Diffusion measurements of SBD at the cell surface by fluorescence correlation spectroscopy suggest that cdc42- and flotillin-associated uptake sites both correspond to domains of intermediate mobility, but that they can cooperate to form low-mobility, efficiently internalized domains. Interestingly, we find that the choice of uptake mechanism affects subsequent trafficking of SBD, as does cholesterol content. Interference with one or other uptake pathway acts as a toggle switch for the trafficking of SBD to recycling endosomes or endolysosomes, whereas both of these pathways are bypassed if cholesterol is reduced. The data are in accordance with a scenario in which SBD mirrors the trafficking response of raft-borne lipids towards a degradative or recycling target. In summary, we suggest that both the surface behavior of a cargo and its subsequent trafficking are determined by a combination of endocytic accessory proteins and the cholesterol content of different membrane compartments.


Asunto(s)
Compartimento Celular , Endocitosis , Microdominios de Membrana/metabolismo , Sondas Moleculares/metabolismo , Esfingolípidos/metabolismo , Animales , Transporte Biológico , Línea Celular Tumoral , Toxina del Cólera/metabolismo , Colesterol/metabolismo , Difusión , Dinaminas/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Humanos , Espacio Intracelular/metabolismo , Cinética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Péptidos/química , Péptidos/metabolismo , Estructura Terciaria de Proteína , Ratas , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
7.
J Lipid Res ; 49(5): 1077-89, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18263852

RESUMEN

We have designed a tagged probe [sphingolipid binding domain (SBD)] to facilitate the tracking of intracellular movements of sphingolipids in living neuronal cells. SBD is a small peptide consisting of the SBD of the amyloid precursor protein. It can be conjugated to a fluorophore of choice and exogenously applied to cells, thus allowing for in vivo imaging. Here, we present evidence to describe the characteristics of the SBD association with the plasma membrane. Our experiments demonstrate that SBD binds to isolated raft fractions from human neuroblastomas and insect neuronal cells. In protein-lipid overlay experiments, SBD interacts with a subset of glycosphingolipids and sphingomyelin, consistent with its raft association in neurons. We also provide evidence that SBD is taken up by neuronal cells in a cholesterol- and sphingolipid-dependent manner via detergent-resistant microdomains. Furthermore, using fluorescence correlation spectroscopy to assay the mobility of SBD in live cells, we show that SBD's behavior at the plasma membrane is similar to that of the previously described raft marker cholera toxin B, displaying both a fast and a slow component. Our data suggest that fluorescently tagged SBD can be used to investigate the dynamic nature of glycosphingolipid-rich detergent-resistant microdomains that are cholesterol-dependent.


Asunto(s)
Colesterol/metabolismo , Microdominios de Membrana/metabolismo , Péptidos/metabolismo , Esfingolípidos/metabolismo , Animales , Sitios de Unión , Línea Celular , Colesterol/deficiencia , Drosophila , Cinética , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA