Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 212: 432-441, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35618088

ABSTRACT

Autophagy is a process in which parts of the eukaryotic cell are selectively degraded in the lysosome. The materials to be catabolized are first surrounded by a double-membrane structure, the autophagosome. Autophagosome generation is a complex event, in which many proteins are involved. Among the latter, yeast Atg8 or its mammalian orthologues are essential in autophagosome membrane elongation, shaping and closure. A subfamily of the human Atg8 orthologues is formed by the proteins LC3A, LC3B, and LC3C. Previous studies suggest that, at variance with the other two, LC3C does not participate in cardiolipin-mediated mitophagy. The present study was devoted to exploring the binding of LC3C to lipid vesicles, bilayers and monolayers, and the ensuing protein-dependent perturbing effects, in the absence of the mitochondrial lipid cardiolipin. All Atg8 orthologues are covalently bound to a phospholipid prior to their involvement in autophagosome elongation. In our case, a mutant in the C-terminal amino acid, LC3C G126C, together with the use of a maleimide-derivatized phosphatidyl ethanolamine, ensured LC3C lipidation, up to 100% under certain conditions. Ultracentrifugation, surface pressure measurements, spectroscopic and cryo-electron microscopic techniques revealed that lipidated LC3C induced vesicle aggregation (5-fold faster in sonicated than in large unilamellar vesicles) and inter-vesicular lipid mixing (up to 82%), including inner-monolayer lipid mixing (up to 32%), consistent with in vitro partial vesicle fusion. LC3C was also able to cause the release of 80-90% vesicular aqueous contents. The data support the idea that LC3C would be able to help in autophagosome elongation/fusion in autophagy phenomena.


Subject(s)
Microtubule-Associated Proteins , Phospholipids , Autophagy , Cardiolipins/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Phospholipids/metabolism , Protein Binding , Saccharomyces cerevisiae/metabolism
2.
Biochim Biophys Acta Biomembr ; 1859(11): 2181-2192, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28803731

ABSTRACT

The immunity proteins against pore-forming colicins represent a family of integral membrane proteins that reside in the inner membrane of producing cells. Cai, the colicin A immunity protein, was characterized here in detergent micelles by circular dichroism (CD), size exclusion chromatography, chemical cross-linking, nuclear magnetic resonance (NMR) spectroscopy, cysteine accessibility, and colicin A binding in detergent micelles. Bile-salt derivatives induced extensive protein polymerization that precluded further investigation. The physical characterization of detergent-solubilized protein indicates that phosphate-containing detergents are more efficient in extracting, solubilizing and maintaining Cai in a monomeric state. Yet, their capacity to ensure protein activity, reconstitution, helix packing, and high-quality NMR spectra was inferior to that of milder detergents. Solvent ionic strength and composition greatly modified the solubilizing capacity of milder detergents. Most importantly, binding to the colicin A pore-forming domain (pf-ColA) occurred almost exclusively in sugar-derived detergents. The relative performance of the different detergents in each experiment depends on their impact not only on Cai structure, solubility and oligomerization state, but also on other reaction components and technical aspects. Thus, proteoliposomes were best obtained from protein in LDAO micelles, possibly also due to indirect effects on the lipidic bilayer. The compatibility of a detergent with Cai/pf-ColA complex formation is influenced by its effect on the conformational landscape of each protein, where detergent-mediated pf-ColA denaturation could also lead to negative results. The NMR spectra were greatly affected by the solubility, monodispersity, fold and dynamics of the protein-detergent complexes, and none of those tested here provided NMR spectra of sufficient quality to allow for peak assignment. Cai function could be proven in alkyl glycosides and not in those detergents that afforded the best solubility, reconstitution efficiency or spectral quality indicating that these criteria cannot be taken as unambiguous proof of nativeness without the support of direct activity measurements.


Subject(s)
Colicins/chemistry , Colicins/isolation & purification , Detergents/chemistry , Micelles , Amino Acid Sequence , Chromatography, Gel , Circular Dichroism , Detergents/pharmacology , Escherichia coli/chemistry , Lipid Bilayers/chemistry , Magnetic Resonance Spectroscopy , Sequence Analysis, Protein , Solubility
3.
Biophys J ; 107(12): 2828-2837, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25517149

ABSTRACT

The origin of resistance to detergent solubilization in certain membranes, or membrane components, is not clearly understood. We have studied the solubilization by Triton X-100 of binary mixtures composed of egg sphingomyelin (SM) and either ceramide, diacylglycerol, or cholesterol. Solubilization has been assayed in the 4-50°C range, and the results are summarized in a novel, to our knowledge, form of plots, that we have called temperature-solubilization diagrams. Despite using a large detergent excess (lipid/detergent 1:20 mol ratio) and extended solubilization times (24-48 h) certain mixtures were not amenable to Triton X-100 solubilization at one or more temperatures. DSC of all the lipid mixtures, and of all the lipid + detergent mixtures revealed that detergent resistance was associated with the presence of gel domains at the assay temperature. Once the system melted down, solubilization could occur. In general adding high-melting lipids limited the solubilization, whereas the addition of low-melting lipids promoted it. Lipidomic analysis of Madin-Darby canine kidney cell membranes and of the corresponding detergent-resistant fraction indicated a large enrichment of the nonsolubilized components in saturated diacylglycerol and ceramide. SM-cholesterol mixtures were special in that detergent solubilization was accompanied, for certain temperatures and compositions, by an independent phenomenon of reassembly of the partially solubilized lipid bilayers. The temperature at which lysis and reassembly prevailed was ∼25°C, thus for some SM-cholesterol mixtures solubilization occurred both above and below 25°C, but not at that temperature. These observations can be at the origin of the detergent resistance effects observed with cell membranes, and they also mean that cholesterol-containing detergent-resistant membrane remnants cannot correspond to structures existing in the native membrane before detergent addition.


Subject(s)
Cell Membrane/chemistry , Ceramides/chemistry , Cholesterol/chemistry , Detergents/chemistry , Diglycerides/chemistry , Liposomes/chemistry , Sphingomyelins/chemistry , Animals , Cell Membrane/drug effects , Detergents/pharmacology , Dogs , Madin Darby Canine Kidney Cells , Membrane Fluidity , Solubility , Transition Temperature
4.
Biophys J ; 106(12): 2577-84, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24940775

ABSTRACT

Sphingosine [(2S, 3R, 4E)-2-amino-4-octadecen-1, 3-diol] is the most common sphingoid long chain base in sphingolipids. It is the precursor of important cell signaling molecules, such as ceramides. In the last decade it has been shown to act itself as a potent metabolic signaling molecule, by activating a number of protein kinases. Moreover, sphingosine has been found to permeabilize phospholipid bilayers, giving rise to vesicle leakage. The present contribution intends to analyze the mechanism by which this bioactive lipid induces vesicle contents release, and the effect of negatively charged bilayers in the release process. Fluorescence lifetime measurements and confocal fluorescence microscopy have been applied to observe the mechanism of sphingosine efflux from large and giant unilamellar vesicles; a graded-release efflux has been detected. Additionally, stopped-flow measurements have shown that the rate of vesicle permeabilization increases with sphingosine concentration. Because at the physiological pH sphingosine has a net positive charge, its interaction with negatively charged phospholipids (e.g., bilayers containing phosphatidic acid together with sphingomyelins, phosphatidylethanolamine, and cholesterol) gives rise to a release of vesicular contents, faster than with electrically neutral bilayers. Furthermore, phosphorous 31-NMR and x-ray data show the capacity of sphingosine to facilitate the formation of nonbilayer (cubic phase) intermediates in negatively charged membranes. The data might explain the pathogenesis of Niemann-Pick type C1 disease.


Subject(s)
Cell Membrane Permeability/drug effects , Lipids/chemistry , Lipids/pharmacology , Sphingosine/pharmacology , Fluorescence , Magnetic Resonance Spectroscopy , Phase Transition/drug effects , Phosphatidic Acids/pharmacology , Scattering, Small Angle , Temperature , Unilamellar Liposomes/chemistry , X-Ray Diffraction
5.
Biochim Biophys Acta ; 1838(8): 2071-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24802275

ABSTRACT

Lipid peroxidation plays a central role in the pathogenesis of many diseases like atherosclerosis and multiple sclerosis. We have analyzed the interaction of sphingosine with peroxidized bilayers in model membranes. Cu(2+) induced peroxidation was checked following UV absorbance at 245nm, and also using the novel Avanti snoopers®. Mass spectrometry confirms the oxidation of phospholipid unsaturated chains. Our results show that sphingosine causes aggregation of Cu(2+)-peroxidized vesicles. We observed that aggregation is facilitated by the presence of negatively-charged phospholipids in the membrane, and inhibited by anti-oxidants e.g. BHT. Interestingly, long-chain alkylamines (C18, C16) but not their short-chain analogues (C10, C6, C1) can substitute sphingosine as promoters of vesicle aggregation. Furthermore, sphinganine but not sphingosine-1-phosphate can mimic this effect. Formation of imines in the membrane upon peroxidation was detected by (1)H-NMR and it appeared to be necessary for the aggregation effect. (31)P-NMR spectroscopy reveals that sphingosine facilitates formation of non-lamellar phase in parallel with vesicle aggregation. The data might suggest a role for sphingosine in the pathogenesis of atherosclerosis.


Subject(s)
Imines/metabolism , Lipid Bilayers/metabolism , Lipid Peroxidation , Liposomes , Sphingosine/metabolism , Copper/pharmacology , Magnetic Resonance Spectroscopy , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
6.
J Membr Biol ; 247(2): 155-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24343571

ABSTRACT

Human phospholipid scramblase 1 (SCR) is a 318 amino acid protein that was originally described as catalyzing phospholipid transbilayer (flip-flop) motion in plasma membranes in a Ca²âº-dependent, ATP-independent way. Further studies have suggested an intranuclear role for this protein in addition. A putative transmembrane domain located at the C terminus (aa 291-309) has been related to the flip-flop catalysis. In order to clarify the role of the C-terminal region of SCR, a mutant was produced (SCRΔ) in which the last 28 amino acid residues were lacking, including the α-helix. SCRΔ had lost the scramblase activity and its affinity for Ca²âº was decreased by one order of magnitude. Fluorescence and IR spectroscopic studies revealed that the C-terminal region of SCR was essential for the proper folding of the protein. Moreover, it was found that Ca²âº exerted an overall destabilizing effect on SCR, which might facilitate its binding to membranes.


Subject(s)
Calcium/metabolism , Phospholipid Transfer Proteins/metabolism , Protein Interaction Domains and Motifs/physiology , Enzyme Activation , Humans , Lipid Metabolism , Mutation , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thermodynamics
7.
Biophys J ; 105(6): 1432-43, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-24047995

ABSTRACT

The colicins are bacteriocins that target Escherichia coli and kill bacterial cells through different mechanisms. Colicin A forms ion channels in the inner membranes of nonimmune bacteria. This activity resides exclusively in its C-terminal fragment (residues 387-592). The soluble free form of this domain is a 10 α-helix bundle. The hydrophobic helical hairpin, H8-H9, is buried inside the structure and shielded by eight amphipathic surface helices. The interaction of the C-terminal colicin A domain and several chimeric variants with lipidic vesicles was examined here by isothermal titration calorimetry. In the mutant constructions, natural sequences of the hydrophobic helices H8 and H9 were either removed or substituted by polyalanine or polyleucine. All the constructions fully associated with DOPG liposomes including the mutant that lacked helices H8 and H9, indicating that amphipathic rather than hydrophobic helices were the major determinants of the exothermic binding reactions. Alanine is not specially favored in the lipid-bound form; the chimeric construct with polyalanine produced lower enthalpy gain. On the other hand, the large negative heat capacities associated with partitioning, a characteristic feature of the hydrophobic effect, were found to be dependent on the sequence hydrophobicity of helices H8 and H9.


Subject(s)
Cell Membrane/metabolism , Colicins/chemistry , Colicins/metabolism , Hydrophobic and Hydrophilic Interactions , Amino Acid Substitution , Colicins/genetics , Hot Temperature , Hydrogen-Ion Concentration , Models, Molecular , Phosphatidylglycerols/metabolism , Porosity , Protein Structure, Secondary , Protein Structure, Tertiary , Thermodynamics
8.
Biomol NMR Assign ; 4(1): 33-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19941091

ABSTRACT

Colicin A protein kills cells by opening voltage-dependent ion channels in the cytoplasmic membrane. The C-terminal domain of colicin A retains the full protein's ability to form membrane pores, making it an excellent model for in vitro studies of protein-membrane interaction. We report here the NMR assignment and backbone dynamics of this domain in solution. The chemical shifts identify ten alpha-helices that match those observed in the crystal structure, while the (15)N{(1)H} NOEs show differential fast mobility for some of the inter-helical loops and the chain ends. This analysis provides the basis for further NMR studies of this channel forming protein and its interactions.


Subject(s)
Colicins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli , Hydrogen/chemistry , Motion , Nitrogen Isotopes/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary , Solutions/chemistry
9.
Arch Virol ; 153(8): 1427-32, 2008.
Article in English | MEDLINE | ID: mdl-18563286

ABSTRACT

Triatoma virus (TrV) is the only entomopathogenic virus found in triatomines. TrV replicates in cells of the midgut epithelium of triatomines, causing a high mortality rate and delayed development of the infected insect. In this work, we report an antigen-capture enzyme-linked immunosorbent assay (AC-ELISA) and a reverse transcription-polymerase chain reaction (RT-PCR) assay for detection of TrV infection. For antiserum production, rabbits and hens where inoculated with purified TrV. Antiserum reactivity was checked by immunodiffusion, and its specificity was confirmed by western blot and AC-ELISA. Totally 90 fecal samples from T. infestans were analysed. AC-ELISA and RT-PCR results correlated well with transmission electron microscopy (EM) observations, which are considered the gold standard, with Kappa values of 0.73 for AC-ELISA and 0.93 for RT-PCR when compared with EM. Applications and complementary uses of the two techniques reported in this work are discussed.


Subject(s)
Antibodies, Viral , Enzyme-Linked Immunosorbent Assay/methods , Picornaviridae Infections/diagnosis , Picornaviridae/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction/methods , Triatoma/virology , Animals , Antibodies, Viral/immunology , Cattle , Hemiptera/virology , Immunoassay , Insect Viruses/isolation & purification , Insect Viruses/pathogenicity , Microscopy, Electron, Transmission , Picornaviridae/isolation & purification , Picornaviridae/physiology , Picornaviridae Infections/immunology , Rabbits
10.
J Biol Chem ; 282(16): 11827-35, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17324923

ABSTRACT

alpha-Hemolysin (HlyA) from Escherichia coli is a protein toxin (1024 amino acids) that targets eukaryotic cell membranes, causing loss of the permeability barrier. HlyA consists of two main regions, an N-terminal domain rich in amphipathic helices, and a C-terminal Ca(2+)-binding domain containing a Gly- and Asp-rich nonapeptide repeated in tandem 11-17 times. The latter is called the RTX domain and gives its name to the RTX protein family. It had been commonly assumed that membrane interaction occurred mainly if not exclusively through the amphipathic helix domain. However, we have cloned and expressed the C-terminal region of HlyA, containing the RTX domain plus a few stabilizing sequences, and found that it is a potent surface-active molecule. The isolated domain binds Ca(2+) with about the same affinity (apparent K(0.5) approximately 150 microM) as the parent protein HlyA, and Ca(2+) binding induces in turn a more compact folding with an increased proportion of beta-sheet structure. Both with and without Ca(2+) the C-terminal region of HlyA can interact with lipid monolayers spread at an air-water interface. However, the C-terminal domain by itself is devoid of membrane lytic properties. The present results can be interpreted in the light of our previous studies that involved in receptor binding a peptide in the C-terminal region of HlyA. We had also shown experimentally the distinction between reversible membrane adsorption and irreversible lytic insertion of the toxin. In this context, the present data allow us to propose that both major domains of HlyA are directly involved in membrane-toxin interaction, the nonapeptide repeat, calcium-binding RTX domain being responsible for the early stages of HlyA docking to the target membrane.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/physiology , Hemolysin Proteins/chemistry , Hemolysin Proteins/physiology , Adsorption , Air , Calcium/chemistry , Cell Membrane/metabolism , Circular Dichroism , Kinetics , Lipids/chemistry , Liposomes/chemistry , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrophotometry , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...