Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Trends Biochem Sci ; 39(11): 510-6, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25440714

RESUMEN

Pore-forming proteins (PFPs) interact with lipid bilayers to compromise membrane integrity. Many PFPs function by inserting a ring of oligomerized subunits into the bilayer to form a protein-lined hydrophilic channel. However, mounting evidence suggests that PFPs can also generate 'proteolipidic' pores by contributing to the fusion of inner and outer bilayer leaflets to form a toroidal structure. We discuss here toroidal pore formation by peptides including melittin, protegrin, and Alzheimer's Aß1-41, as well as by PFPs from several evolutionarily unrelated families: the colicin/Bcl-2 grouping including the pro-apoptotic protein Bax, actinoporins derived from sea anemones, and the membrane attack complex-perforin/cholesterol dependent cytolysin (MACPF/CDC) set of proteins. We also explore how the structure and biological role of toroidal pores might be investigated further.


Asunto(s)
Membrana Celular/química , Membrana Dobles de Lípidos/química , Lípidos de la Membrana/química , Proteínas Citotóxicas Formadoras de Poros/química , Membrana Celular/metabolismo , Colicinas/química , Colicinas/metabolismo , Membrana Dobles de Lípidos/metabolismo , Meliteno/química , Meliteno/metabolismo , Lípidos de la Membrana/metabolismo , Modelos Moleculares , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
2.
Biochim Biophys Acta ; 1858(3): 446-56, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26351738

RESUMEN

Actinoporins (APs) from sea anemones are ~20 kDa pore forming toxins with a ß-sandwich structure flanked by two α-helices. The molecular mechanism of APs pore formation is composed of several well-defined steps. APs bind to membrane by interfacial binding site composed of several aromatic amino acid residues that allow binding to phosphatidylcholine and specific recognition of sphingomyelin. Subsequently, the N-terminal α-helix from the ß-sandwich has to be inserted into the lipid/water interphase in order to form a functional pore. Functional studies and single molecule imaging revealed that only several monomers, 3-4, oligomerise to form a functional pore. In this model the α-helices and surrounding lipid molecules build toroidal pore. In agreement, AP pores are transient and electrically heterogeneous. On the contrary, crystallized oligomers of actinoporin fragaceatoxin C were found to be composed of eight monomers with no lipids present between the adjacent α-helices. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Maur Dalla Serra and Franco Gambale.


Asunto(s)
Venenos de Cnidarios/química , Perforina/química , Porinas/química , Anémonas de Mar/química , Animales , Venenos de Cnidarios/metabolismo , Perforina/metabolismo , Porinas/metabolismo , Estructura Secundaria de Proteína , Anémonas de Mar/metabolismo
3.
Cell Microbiol ; 17(8): 1241-57, 2015 08.
Artículo en Inglés | MEDLINE | ID: mdl-25737084

RESUMEN

A growing number of receptors, often associated with the innate immune response, are being identified as targets for bacterial toxins of the beta-stranded pore-forming family. These findings raise the new question of whether the receptors are activated or merely used as docking points facilitating the formation of a pore. To elucidate whether the Staphylococcus aureus Panton-Valentine leukocidin and the leukotoxin HlgC/HlgB act through the C5a receptor (C5aR) as agonists, antagonists or differ from the C5a complement-derived peptide, their activity is explored on C5aR-expressing cells. Both leukotoxins equally bound C5aR in neutrophils and in stable transfected U937 cells and initiated mobilization of intracellular Ca(2+) . HlgC/HlgB requires the presence of robust intracellular acidic Ca(2+) stores in order to evoke a rise in free [Ca(2+) ]i , while the LukS-PV/LukF-PV directly altered reticular Ca(2+) stores. Intracellular target specificity is conferred by the F-subunit associated to the S-subunit binding the receptor. Furthermore, internalization of the two leukotoxin components (S- and F-subunits) associated to C5aR is required for the initiation of [Ca(2+) ]i mobilization. Electrophysiological recordings on living cells demonstrated that LukS-PV/LukF-PV does not alter the membrane resistance of C5aR-expressing cells. The present observations suggest that part of the pore-forming process occurs in distinct intracellular compartments rather than at the plasma membrane.


Asunto(s)
Toxinas Bacterianas/metabolismo , Calcio/metabolismo , Exotoxinas/metabolismo , Leucocidinas/metabolismo , Neutrófilos/microbiología , Neutrófilos/fisiología , Receptor de Anafilatoxina C5a/metabolismo , Staphylococcus aureus/inmunología , Células Cultivadas , Fenómenos Electrofisiológicos , Interacciones Huésped-Patógeno , Humanos , Monocitos/microbiología , Monocitos/fisiología , Unión Proteica
4.
Biochem J ; 472(1): 83-95, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26371376

RESUMEN

Genome-wide analyses of translation can provide major contributions in our understanding of the complex interplay between virulent factors and host cells. So far, the activation of host translational control mechanisms by bacterial toxins, owing to specific recruitment of mRNAs, RNA-binding proteins (RBPs) and ncRNAs (non-coding RNAs), are far from being understood. In the present study, we characterize for the first time the changes experienced by the translational control system of host cells in response to the well-known Staphylococcus aureus α-haemolysin (AHL) under both sublytic and lytic conditions. By comparing variations occurring in the cellular transcriptome and translatome, we give evidence that global gene expression is primarily rewired at the translational level, with the contribution of the RBP ELAVL1 (HuR) in the sublytic response. These results reveal the importance of translational control during host-pathogen interaction, opening new approaches for AHL-induced diseases.


Asunto(s)
Toxinas Bacterianas/farmacología , Variación Genética/efectos de los fármacos , Proteínas Hemolisinas/farmacología , Biosíntesis de Proteínas/genética , Transcriptoma/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo , Perfilación de la Expresión Génica/métodos , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Immunoblotting , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Anal Chem ; 87(17): 8818-26, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26258431

RESUMEN

α-Synuclein oligomers can be toxic to cells and may be responsible for cell death in Parkinson's disease. Their typically low abundance and highly heterogeneous nature, however, make such species challenging to study using traditional biochemical techniques. By combining fast-flow microfluidics with single-molecule fluorescence, we are able to rapidly follow the process by which oligomers of αS are formed and to characterize the species themselves. We have used the technique to show that populations of oligomers with different FRET efficiencies have varying stabilities when diluted into low ionic strength solutions. Interestingly, we have found that oligomers formed early in the aggregation pathway have electrostatic repulsions that are shielded in the high ionic strength buffer and therefore dissociate when diluted into lower ionic strength solutions. This property can be used to isolate different structural groups of αS oligomers and can help to rationalize some aspects of αS amyloid fibril formation.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Fluorescencia , Técnicas Analíticas Microfluídicas , alfa-Sinucleína/análisis , Rayos Láser , Técnicas Analíticas Microfluídicas/instrumentación , Electricidad Estática
6.
Subcell Biochem ; 80: 271-91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24798017

RESUMEN

Proteins with membrane-attack complex/perforin (MACPF) domains are found in almost all kingdoms of life, and they have a variety of biological roles, including defence and attack, organism development, and cell adhesion and signalling. The distribution of these proteins in fungi appears to be restricted to some Pezizomycotina and Basidiomycota species only, in correlation with another group of proteins with unknown biological function, known as aegerolysins. These two protein groups coincide in only a few species, and they might operate in concert as cytolytic bi-component pore-forming agents. Representative proteins here include pleurotolysin B, which has a MACPF domain, and the aegerolysin-like protein pleurotolysin A, and the very similar ostreolysin A, which have been purified from oyster mushroom (Pleurotus ostreatus). These have been shown to act in concert to perforate natural and artificial lipid membranes with high cholesterol and sphingomyelin content. The aegerolysin-like proteins provide the membrane cholesterol/sphingomyelin selectivity and recruit oligomerised pleurotolysin B molecules, to create a membrane-inserted pore complex. The resulting protein structure has been imaged with electron microscopy, and it has a 13-meric rosette-like structure, with a central lumen that is ~4-5 nm in diameter. The opened transmembrane pore is non-selectively permeable for ions and smaller neutral solutes, and is a cause of cytolysis of a colloid-osmotic type. The biological significance of these proteins for the fungal life-style is discussed.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Proteínas Fúngicas/fisiología , Proteínas Hemolisinas/fisiología , Perforina/fisiología , Proteínas Citotóxicas Formadoras de Poros/fisiología , Secuencia de Aminoácidos , Animales , Complejo de Ataque a Membrana del Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Humanos , Datos de Secuencia Molecular , Perforina/química , Filogenia , Pleurotus/genética , Pleurotus/patogenicidad , Proteínas Citotóxicas Formadoras de Poros/química , Multimerización de Proteína/fisiología , Homología de Secuencia de Aminoácido
7.
J Biol Chem ; 288(33): 23704-15, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23803608

RESUMEN

Actinoporin equinatoxin II (EqtII) is an archetypal example of α-helical pore-forming toxins that porate cellular membranes by the use of α-helices. Previous studies proposed several steps in the pore formation: binding of monomeric protein onto the membrane, followed by oligomerization and insertion of the N-terminal α-helix into the lipid bilayer. We studied these separate steps with an EqtII triple cysteine mutant. The mutant was engineered to monitor the insertion of the N terminus into the lipid bilayer by labeling Cys-18 with a fluorescence probe and at the same time to control the flexibility of the N-terminal region by the disulfide bond formed between cysteines introduced at positions 8 and 69. The insertion of the N terminus into the membrane proceeded shortly after the toxin binding and was followed by oligomerization. The oxidized, non-lytic, form of the mutant was still able to bind to membranes and oligomerize at the same level as the wild-type or the reduced form. However, the kinetics of the N-terminal helix insertion, the release of calcein from erythrocyte ghosts, and hemolysis of erythrocytes was much slower when membrane-bound oxidized mutant was reduced by the addition of the reductant. Results show that the N-terminal region needs to be inserted in the lipid membrane before the oligomerization into the final pore and imply that there is no need for a stable prepore formation. This is different from ß-pore-forming toxins that often form ß-barrel pores via a stable prepore complex.


Asunto(s)
Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Venenos de Cnidarios/química , Venenos de Cnidarios/farmacología , Animales , Bovinos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cisteína/genética , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Cinética , Lípidos de la Membrana/química , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Ingeniería de Proteínas , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia
8.
Biochim Biophys Acta ; 1828(2): 405-11, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23036932

RESUMEN

γ-Hemolysins are bicomponent ß-barrel pore forming toxins produced by Staphylococcus aureus as water-soluble monomers, which assemble into oligomeric pores on the surface of lipid bilayers. Here, after investigating the oligomeric structure of γ-hemolysins on supported lipid bilayers (SLBs) by atomic force microscopy (AFM), we studied the effect produced by this toxin on the structure of SLBs. We found that oligomeric structures with different number of monomers can assemble on the lipid bilayer being the octameric form the stablest one. Moreover, in this membrane model we found that γ-hemolysins can form clusters of oligomers inducing a curvature in the lipid bilayer, which could probably enhance the aggressiveness of these toxins at high concentrations.


Asunto(s)
Proteínas Hemolisinas/química , Membrana Dobles de Lípidos/química , Microscopía de Fuerza Atómica/métodos , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/química , Biofisica/métodos , Cristalografía por Rayos X/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Procesamiento de Imagen Asistido por Computador , Lípidos/química , Liposomas/química , Modelos Moleculares , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Factores de Tiempo
9.
Cell Mol Life Sci ; 70(12): 2083-98, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22983385

RESUMEN

Recent work on the MACPF/CDC superfamily of pore-forming proteins has focused on the structural analysis of monomers and pore-forming oligomeric complexes. We set the family of proteins in context and highlight aspects of their function which the direct and exclusive equation of oligomers with pores fails to explain. Starting with a description of the distribution of MACPF/CDC proteins across the domains of life, we proceed to show how their evolutionary relationships can be understood on the basis of their structural homology and re-evaluate models for pore formation by perforin, in particular. We furthermore highlight data showing the role of incomplete oligomeric rings (arcs) in pore formation and how this can explain small pores generated by oligomers of proteins belonging to the family. We set this in the context of cell biological and biophysical data on the proteins' function and discuss how this helps in the development of an understanding of how they act in processes such as apicomplexan parasites gliding through cells and exiting from cells.


Asunto(s)
Membrana Celular/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Citotoxinas/metabolismo , Modelos Moleculares , Perforina/metabolismo , Filogenia , Conformación Proteica , Secuencia de Aminoácidos , Apicomplexa , Bacterias , Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/genética , Citotoxinas/química , Datos de Secuencia Molecular , Perforina/química , Polimerizacion , Alineación de Secuencia
10.
Biochem J ; 450(3): 559-71, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23282185

RESUMEN

PVL (Panton-Valentine leukocidin) and other Staphylococcus aureus ß-stranded pore-forming toxins are important virulence factors involved in various pathologies that are often necrotizing. The present study characterized leukotoxin inhibition by selected SCns (p-sulfonato-calix[n]arenes): SC4, SC6 and SC8. These chemicals have no toxic effects on human erythrocytes or neutrophils, and some are able to inhibit both the activity of and the cell lysis by leukotoxins in a dose-dependent manner. Depending on the type of leukotoxins and SCns, flow cytometry revealed IC50 values of 6-22 µM for Ca2+ activation and of 2-50 µM for cell lysis. SCns were observed to affect membrane binding of class S proteins responsible for cell specificity. Electrospray MS and surface plasmon resonance established supramolecular interactions (1:1 stoichiometry) between SCns and class S proteins in solution, but not class F proteins. The membrane-binding affinity of S proteins was Kd=0.07-6.2 nM. The binding ability was completely abolished by SCns at different concentrations according to the number of benzenes (30-300 µM; SC8>SC6≫SC4). The inhibitory properties of SCns were also observed in vivo in a rabbit model of PVL-induced endophthalmitis. These calixarenes may represent new therapeutic avenues aimed at minimizing inflammatory reactions and necrosis due to certain virulence factors.


Asunto(s)
Calixarenos/farmacología , Exotoxinas/antagonistas & inhibidores , Exotoxinas/metabolismo , Staphylococcus aureus/metabolismo , Animales , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/metabolismo , Calixarenos/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Proteínas Hemolisinas/antagonistas & inhibidores , Proteínas Hemolisinas/metabolismo , Humanos , Sustancias Macromoleculares/metabolismo , Modelos Biológicos , Fenoles/metabolismo , Fenoles/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Conejos , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(52): 21016-21, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22173634

RESUMEN

The cytotoxic cell granule secretory pathway is essential for immune defence. How the pore-forming protein perforin (PFN) facilitates the cytosolic delivery of granule-associated proteases (granzymes) remains enigmatic. Here we show that PFN is able to induce invaginations and formation of complete internal vesicles in giant unilamellar vesicles. Formation of internal vesicles depends on native PFN and calcium and antibody labeling shows the localization of PFN at the invaginations. This vesiculation is recapitulated in large unilamellar vesicles and in this case PFN oligomers can be seen associated with the necks of the invaginations. Capacitance measurements show PFN is able to increase a planar lipid membrane surface area in the absence of pore formation, in agreement with the ability to induce invaginations. Finally, addition of PFN to Jurkat cells causes the formation of internal vesicles prior to pore formation. PFN is capable of triggering an endocytosis-like event in addition to pore formation, suggesting a new paradigm for its role in delivering apoptosis-inducing granzymes into target cells.


Asunto(s)
Membrana Celular/metabolismo , Endocitosis/fisiología , Granzimas/metabolismo , Inmunidad Innata/fisiología , Perforina/metabolismo , Vesículas Secretoras/metabolismo , Microscopía por Crioelectrón , Humanos , Células Jurkat , Microscopía Fluorescente , Perforina/inmunología , Perforina/fisiología
12.
Pharmaceutics ; 16(3)2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38543324

RESUMEN

The intestine is essential for the modulation of nutrient absorption and the removal of waste. Gut pathologies, such as cancer, inflammatory bowel diseases (IBD), irritable bowel syndrome (IBS), and celiac disease, which extensively impact gut functions, are thus critical for human health. Targeted drug delivery is essential to tackle these diseases, improve therapy efficacy, and minimize side effects. Recent strategies have taken advantage of both active and passive nanocarriers, which are designed to protect the drug until it reaches the correct delivery site and to modulate drug release via the use of different physical-chemical strategies. In this systematic review, we present a literature overview of the different nanocarriers used for drug delivery in a set of chronic intestinal pathologies, highlighting the rationale behind the controlled release of intestinal therapies. The overall aim is to provide the reader with useful information on the current approaches for gut targeting in novel therapeutic strategies.

13.
Biochim Biophys Acta ; 1818(11): 2876-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22820150

RESUMEN

Alpha-synuclein is a natively unfolded protein widely expressed in neurons at the presynaptic level. It is linked to Parkinson's disease by two lines of evidence: amyloid fibrils of the protein accumulate in patients' brains and three genetic mutants cause autosomal dominant forms of the disease. The biological role of the protein and the mechanisms involved in the etiopathogenesis of Parkinson's disease are still unknown. Membrane binding causes the formation of an amphipathic alpha-helix, which lies on the surface without crossing the bilayer. Recent observations however reported that the application of a voltage induces a pore-like activity of alpha-synuclein. This study aims to characterize the pore forming activity of the protein starting from its monomeric form. In particular, experiments with planar lipid membranes allowed recording of conductance activity bursts with a defined and reproducible fingerprint. Additional experiments with deletion mutants and covalently bound alpha-synuclein dimers were performed to understand both pore assembly and stoichiometry. The information acquired allowed formulation of a model for pore formation at different conductance levels.


Asunto(s)
Membrana Celular , alfa-Sinucleína/metabolismo , Membrana Celular/metabolismo , Dimerización , Membrana Dobles de Lípidos
15.
J Biol Chem ; 286(4): 2946-55, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20889983

RESUMEN

Perforin (PFN) is a pore-forming protein produced by cytotoxic lymphocytes that aids in the clearance of tumor or virus-infected cells by a mechanism that involves the formation of transmembrane pores. The properties of PFN pores and the mechanism of their assembly remain unclear. Here, we studied pore characteristics by functional and structural methods to show that perforin forms pores more heterogeneous than anticipated. Planar lipid bilayer experiments indicate that perforin pores exhibit a broad range of conductances, from 0.15 to 21 nanosiemens. In comparison with large pores that possessed low noise and remained stably open, small pores exhibited high noise and were very unstable. Furthermore, the opening step and the pore size were dependent on the lipid composition of the membrane. The heterogeneity in pore sizes was confirmed with cryo-electron microscopy and showed a range of sizes matching that observed in the conductance measurements. Furthermore, two different membrane-bound PFN conformations were observed, interpreted as pre-pore and pore states of the protein. The results collectively indicate that PFN forms heterogeneous pores through a multistep mechanism and provide a new paradigm for understanding the range of different effects of PFN and related membrane attack complex/perforin domain proteins observed in vivo and in vitro.


Asunto(s)
Membrana Celular/química , Membrana Dobles de Lípidos/química , Perforina/química , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Humanos , Membrana Dobles de Lípidos/metabolismo , Perforina/metabolismo
17.
Mol Plant Microbe Interact ; 25(8): 1118-31, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22550957

RESUMEN

Defensins are a class of small and diverse cysteine-rich proteins found in plants, insects, and vertebrates, which share a common tertiary structure and usually exert broad-spectrum antimicrobial activities. We used a bioinformatic approach to scan the Vitis vinifera genome and identified 79 defensin-like sequences (DEFL) corresponding to 46 genes and allelic variants, plus 33 pseudogenes and gene fragments. Expansion and diversification of grapevine DEFL has occurred after the split from the last common ancestor with the genera Medicago and Arabidopsis. Grapevine DEFL localization on the 'Pinot Noir' genome revealed the presence of several clusters likely evolved through local duplications. By sequencing reverse-transcription polymerase chain reaction products, we could demonstrate the expression of grapevine DEFL with no previously reported record of expression. Many of these genes are predominantly or exclusively expressed in tissues linked to plant reproduction, consistent with findings in other plant species, and some of them accumulated at fruit ripening. The transcripts of five DEFL were also significantly upregulated in tissues infected with Botrytis cinerea, a necrotrophic mold, suggesting a role of these genes in defense against this pathogen. Finally, three novel defensins were discovered among the identified DEFL. They inhibit B. cinerea conidia germination when expressed as recombinant proteins.


Asunto(s)
Defensinas/genética , Familia de Multigenes , Vitis/genética , Secuencia de Aminoácidos , Botrytis/patogenicidad , Resistencia a la Enfermedad/genética , Regulación de la Expresión Génica de las Plantas , Genoma de Planta , Datos de Secuencia Molecular , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vitis/microbiología
18.
Eur Biophys J ; 46(8): 689-690, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29128893

Asunto(s)
Biofisica
19.
Biochim Biophys Acta ; 1798(5): 891-902, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20114031

RESUMEN

Ostreolysin is a cytolytic protein from the edible oyster mushroom (Pleurotus ostreatus), which recognizes specifically and binds to raft-like sterol-enriched membrane domains that exist in the liquid-ordered phase. Its binding can be abolished by micromolar concentrations of lysophospholipids and fatty acids. The membrane activity of ostreolysin, however, does not completely correlate with the ability of a certain sterol to induce the formation of a liquid-ordered phase, suggesting that the protein requires an additional structural organization of the membrane to exert its activity. The aim of this study was to further characterize the lipid membranes that facilitate ostreolysin binding by analyzing their lipid phase domain structure. Fourier-transformed infrared spectroscopy (FTIR) and electron paramagnetic resonance (EPR) were used to analyze the ordering and dynamics of membrane lipids and the membrane domain structure of a series of unilamellar liposomes prepared by systematically changing the lipid components and their ratios. Our results corroborate the earlier conclusion that the average membrane fluidity of ostreolysin-susceptible liposomes alone cannot account for the membrane activity of the protein. Combined with previous data computer-aided interpretation of EPR spectra strongly suggests that chemical properties of membrane constituents, their specific distribution, and physical characteristics of membrane nanodomains, resulting from the presence of sterol and sphingomyelin (or a highly ordered phospholipid, dipalmitoylphosphatidylcholine), are essential prerequisites for ostreolysin membrane binding and pore-formation.


Asunto(s)
Membrana Celular/química , Proteínas Hemolisinas/química , Esteroles/química , Animales , Espectroscopía de Resonancia por Spin del Electrón , Proteínas Fúngicas/química , Liposomas/química , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Esfingomielinas/química , Porcinos
20.
Nat Cell Biol ; 22(10): 1239-1251, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32958857

RESUMEN

The contribution of ribosome heterogeneity and ribosome-associated proteins to the molecular control of proteomes in health and disease remains unclear. Here, we demonstrate that survival motor neuron (SMN) protein-the loss of which causes the neuromuscular disease spinal muscular atrophy (SMA)-binds to ribosomes and that this interaction is tissue-dependent. SMN-primed ribosomes are preferentially positioned within the first five codons of a set of mRNAs that are enriched for translational enhancer sequences in the 5' untranslated region (UTR) and rare codons at the beginning of their coding sequence. These SMN-specific mRNAs are associated with neurogenesis, lipid metabolism, ubiquitination, chromatin regulation and translation. Loss of SMN induces ribosome depletion, especially at the beginning of the coding sequence of SMN-specific mRNAs, leading to impairment of proteins that are involved in motor neuron function and stability, including acetylcholinesterase. Thus, SMN plays a crucial role in the regulation of ribosome fluxes along mRNAs encoding proteins that are relevant to SMA pathogenesis.


Asunto(s)
Neuronas Motoras/patología , Atrofia Muscular Espinal/patología , Biosíntesis de Proteínas , Proteoma/análisis , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ratones , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , ARN Mensajero/genética , Ribosomas/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA