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










Publication year range
1.
J Immunol ; 198(7): 2772-2784, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28258198

ABSTRACT

Vaccine strategies to enhance CD8+ CTL responses remain a current challenge because they should overcome the plasmatic and endosomal membranes for favoring exogenous Ag access to the cytosol of APCs. As a way to avoid this hurdle, sticholysin (St) II, a pore-forming protein from the Caribbean Sea anemone Stichodactyla helianthus, was encapsulated with OVA into liposomes (Lp/OVA/StII) to assess their efficacy to induce a CTL response. OVA-specific CD8+ T cells transferred to mice immunized with Lp/OVA/StII experienced a greater expansion than when the recipients were injected with the vesicles without St, mostly exhibiting a memory phenotype. Consequently, Lp/OVA/StII induced a more potent effector function, as shown by CTLs, in vivo assays. Furthermore, treatment of E.G7-OVA tumor-bearing mice with Lp/OVA/StII significantly reduced tumor growth being more noticeable in the preventive assay. The contribution of CD4+ and CD8+ T cells to CTL and antitumor activity, respectively, was elucidated. Interestingly, the irreversibly inactive variant of the StI mutant StI W111C, encapsulated with OVA into Lp, elicited a similar OVA-specific CTL response to that observed with Lp/OVA/StII or vesicles encapsulating recombinant StI or the reversibly inactive StI W111C dimer. These findings suggest the relative independence between StII pore-forming activity and its immunomodulatory properties. In addition, StII-induced in vitro maturation of dendritic cells might be supporting these properties. These results are the first evidence, to our knowledge, that StII, a pore-forming protein from a marine eukaryotic organism, encapsulated into Lp functions as an adjuvant to induce a robust specific CTL response.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Cnidarian Venoms/administration & dosage , Neoplasms, Experimental/pathology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Cnidarian Venoms/immunology , Female , Flow Cytometry , Liposomes/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
2.
PLoS One ; 9(10): e110824, 2014.
Article in English | MEDLINE | ID: mdl-25350457

ABSTRACT

The use of pore-forming toxins in the construction of immunotoxins against tumour cells is an alternative for cancer therapy. In this protein family one of the most potent toxins are the actinoporins, cytolysins from sea anemones. We work on the construction of tumour proteinase-activated immunotoxins using sticholysin I (StI), an actinoporin isolated from the sea anemone Stichodactyla helianthus. To accomplish this objective, recombinant StI (StIr) with a mutation in the membrane binding region has been employed. In this work, it was evaluated the impact of mutating tryptophan 111 to cysteine on the toxin pore forming capability. StI W111C is still able to permeabilize erythrocytes and liposomes, but at ten-fold higher concentration than StI. This is due to its lower affinity for the membrane, which corroborates the importance of residue 111 for the binding of actinoporins to the lipid bilayer. In agreement, other functional characteristics not directly associated to the binding, are essentially the same for both variants, that is, pores have oligomeric structures with similar radii, conductance, cation-selectivity, and instantaneous current-voltage behavior. In addition, this work provides experimental evidence sustaining the toroidal protein-lipid actinoporins lytic structures, since the toxins provoke the trans-bilayer movement (flip-flop) of a pyrene-labeled analogue of phosphatidylcholine in liposomes, indicating the existence of continuity between the outer and the inner membrane leaflet. Finally, our planar lipid membranes results have also contributed to a better understanding of the actinoporin's pore assembly mechanism. After the toxin binding and the N-terminal insertion in the lipid membrane, the pore assembly occurs by passing through different transient sub-conductance states. These states, usually 3 or 4, are due to the successive incorporation of N-terminal α-helices and lipid heads to the growing pores until a stable toroidal oligomeric structure is formed, which is mainly tetrameric.


Subject(s)
Sea Anemones/chemistry , Adsorption , Animals , Cryoelectron Microscopy , Cysteine/chemistry , Electric Conductivity , Erythrocytes/drug effects , Hemolysis , Humans , Immunotoxins/chemistry , Ions , Lipid Bilayers/chemistry , Lipids/chemistry , Liposomes/chemistry , Mutation , Organic Chemicals/chemistry , Permeability , Phosphatidylcholines/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sea Anemones/genetics
3.
Protein Eng Des Sel ; 24(6): 485-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21296830

ABSTRACT

The use of pore-forming toxins from sea anemones (actinoporins) in the construction of immunotoxins (ITs) against tumour cells is an alternative for cancer therapy. However, the main disadvantage of actinoporin-based ITs obtained so far has been the poor cellular specificity associated with the toxin's ability to bind and exert its activity in almost any cell membrane. Our final goal is the construction of tumour proteinase-activated ITs using a cysteine mutant at the membrane binding region of sticholysin-I (StI), a cytolysin isolated from the sea anemone Stichodactyla helianthus. The mutant and the ligand moiety would be linked by proteinase-sensitive peptides through the StI cysteine residue blocking the toxin binding region and hence the IT non-specific killing activity. To accomplish this objective the first step was to obtain the mutant StI W111C, and to evaluate the impact of mutating tryptophan 111 by cysteine on the toxin pore-forming capacity. After proteolysis of the cleavage sequence, a short peptide would remain attached to the toxin. The next step was to evaluate whether this mutant is able to form pores even with a residual peptide linked to cysteine 111. In this work we demonstrated that (i) StI W111C shows pore-forming capacity in a nanomolar range, although it is 8-fold less active than the wild-type recombinant StI, corroborating the previously reported importance of residue 111 for the binding of StI to membranes, and (ii) the mutant is able to form pores even with a residual seven-residue peptide linked to cysteine 111. In addition, it was demonstrated that binding of a large molecule to cysteine 111 renders an inactive toxin that is no longer able to bind to the membrane. These results validate the mutant StI W111C for its use in the construction of tumour proteinase-activated ITs.


Subject(s)
Immunotoxins/chemistry , Pore Forming Cytotoxic Proteins/chemistry , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Chromatography, Gel , Chromatography, Ion Exchange , Dimerization , Immunotoxins/genetics , Immunotoxins/isolation & purification , Immunotoxins/metabolism , Models, Molecular , Mutation , Organic Chemicals/chemistry , Organic Chemicals/isolation & purification , Organic Chemicals/metabolism , Perforin , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/isolation & purification , Pore Forming Cytotoxic Proteins/metabolism , Protein Binding , Reproducibility of Results , Sea Anemones
4.
Toxicon ; 54(8): 1135-47, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19268489

ABSTRACT

Sticholysins (Sts) I and II (StI/II) are pore-forming toxins (PFTs) produced by the Caribbean Sea anemone Stichodactyla helianthus belonging to the actinoporin family, a unique class of eukaryotic PFTs exclusively found in sea anemones. As for the rest of the members of this family, Sts are cysteine-less proteins, with molecular weights around 20 kDa, high isoelectric points (>9.5), and a preference for sphingomyelin-containing membranes. A three-dimensional structure of StII, solved by X-ray crystallography, showed that it is composed of a hydrophobic beta-sandwich core flanked on the opposite sides by two alpha helices comprising residues 14-23 and 128-135. A variety of experimental results indicate that the first thirty N-terminal residues, which include one of the helices, are directly involved in pore formation. This region contains an amphipathic stretch, well conserved in all actinoporins, which is the only portion of the molecule that can change conformation without perturbing the general protein fold; in fact, binding to model membranes only produces a slight increase in the regular secondary structure content of Sts. Sts are produced in soluble form but they readily bind to different cell and model membrane systems such as lipidic monolayers, micelles, and lipid vesicles. Remarkably, both the binding and pore-formation steps are critically dependent on the physico-chemical nature of the membrane. In fact, a large population of toxin irreversibly binds with high affinity in membranes containing sphingomyelin whereas binding in membranes lacking this sphingolipid is relatively low and reversible. The joint presence of SM and cholesterol largely promotes binding and pore formation. Minor amounts of lipids favoring a non-lamellar organization also augment the efficiency of pore formation. The functional pore formed in cellular and model membranes has a diameter of approximately 2.0 nm and is presumably formed by the N-terminal alpha helices of four monomers tilted 31 degrees in relation to the bilayer normal. Experimental evidence supports the hypothesis that sticholysins, as well as equinatoxin II, another actinoporin, form a toroidal pore in membranes in which the polypeptide chains as well as the polar head groups of phospholipids are involved.


Subject(s)
Cell Membrane/metabolism , Cnidarian Venoms/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Sea Anemones/chemistry , Amino Acid Sequence , Animals , Cnidarian Venoms/chemistry , Molecular Sequence Data , Organic Chemicals/chemistry , Organic Chemicals/metabolism , Pore Forming Cytotoxic Proteins/chemistry , Sea Anemones/metabolism
5.
Toxicon ; 54(8): 1206-14, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19268683

ABSTRACT

The use of membrane active toxins as toxic moieties in the construction of immunotoxins (ITs) is an attractive alternative to overcome some of the problems of classical ITs since these new conjugates are based in the use of a different mechanism of killing undesired cells. Pore-forming cytolysins from sea anemones were used in the construction of ITs targeted to different cell types including tumour cell lines and the parasite Giardia duodenalis. The results obtained support the feasibility of directing these cytolysins to the surface of the cancer cells or the parasite through their conjugation to monoclonal antibodies recognizing tumour-associated or parasite antigens, respectively. However the main problem with the IT constructed in this fashion is the lack of specificity associated with the toxin moiety. An approach designed to overcome this limitation was the construction of inactive cytolysin with built-in biological "trigger" that renders the toxin active in the presence of tumour-specific proteinases. This construction is considered as a proof of concept to demonstrate the feasibility of such activation systems in the construction of ITs based on pore-forming cytolysins from sea anemones with reduced unspecific activity. The future prospects of the use of the N-terminal region of actinoporins for construction of IT is also described.


Subject(s)
Cytotoxins/toxicity , Immunotoxins/chemistry , Immunotoxins/toxicity , Sea Anemones/chemistry , Animals , Cell Membrane/drug effects , Cell Membrane/immunology , Cytotoxins/chemistry , Giardia/drug effects , Giardia/growth & development , Giardia/immunology , Humans , Immunotoxins/immunology , Immunotoxins/metabolism
6.
Toxicon ; 50(6): 731-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17681582

ABSTRACT

The effect of three cationic surfactants bearing the same polar head group and different chain length (cetyltrimethyl ammonium bromide (CTAB); tetradecyltrimethylammonium bromide (TTAB); dodecyltrimethylammonium bromide (DTAB)) on the conformation and function of the sea anemone pore-forming toxins sticholysins I and II (St I and St II) was studied by fluorescence and circular dichroism spectroscopy and evaluation of hemolytic activity (HA). Preincubation of the toxins with the longer chain surfactants CTAB and TTAB at concentrations slightly above their critical micelle concentration (CMC) leads to an enhancement of their HA. Significant increases in the fluorescence intensity with a slightly red shift in lambda(max) were observed at concentrations close to the surfactants' CMC, suggesting changes in the environment of the tryptophan residues. The changes in the fluorescence intensity are more noticeable and take place at lower surfactant concentrations for St I, irrespective of the surfactant alkyl chain length, although the differences between St I and St II increase as the surfactant alkyl chain length increases. This is evinced not only by the higher fluorescence intensity values and the lower surfactant concentrations required to reach them, but also by the higher acrylamide-quenching constant values (Ksv) for St I. However, the surfactant's effects on the toxins' HA were not found to be directly related to the observed changes in fluorescence intensity, as well as near- and far-UV-CD spectra. In particular, the latter spectra indicate that changes in HA and in fluorescence behavior take place without noticeable modifications in St I and St II secondary and tertiary structures. The results suggest that the interaction with the surfactants induces only subtle conformational changes in the toxins that favor the formation of lytic competent structures.


Subject(s)
Cnidarian Venoms/pharmacology , Hemolysis/drug effects , Pore Forming Cytotoxic Proteins/pharmacology , Quaternary Ammonium Compounds/pharmacology , Sea Anemones , Surface-Active Agents/pharmacology , Animals , Cations , Cetrimonium , Cetrimonium Compounds/chemistry , Circular Dichroism , Cnidarian Venoms/chemistry , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Erythrocytes/drug effects , Humans , In Vitro Techniques , Micelles , Organic Chemicals/chemistry , Organic Chemicals/pharmacology , Pore Forming Cytotoxic Proteins/chemistry , Protein Conformation , Quaternary Ammonium Compounds/chemistry , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Surface-Active Agents/chemistry , Trimethyl Ammonium Compounds/chemistry
7.
Toxicon ; 49(1): 68-81, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17113118

ABSTRACT

Sticholysin II (St II) is a cytolysin produced by the sea anemone Stichodactyla helianthus, characterized by forming oligomeric pores in natural and artificial membranes. In the present work the influence of the membrane lipidic components sphingomyelin (SM) and cholesterol (Cho) on binding and functional activity of St II, was evaluated using ELISA, lipid monolayers and liposomes. The aim of this work was to establish the promoting role of Cho and SM, both in St II binding and pore formation efficiency. In general the association (evaluated by ELISA and incorporation to phospholipid monolayers) of St II to lipids mixtures was better than to any one of the single components. Regarding the unique role of SM, it was found that, albeit inefficiently, St II binds to phosphatidylcholine (PC):Cho monolayers and liposomes, and is able to form active pores in these bilayers. The results in monolayers and liposomes show that the presence of SM and large amounts of Cho leads to the highest values of critical pressure and rate of association to monolayers, the most favorable interaction with liposomes, and the fastest rate of pore formation, in spite of the rigidity of the layers as suggested by the high generalized polarization (GP) of Laurdan incorporated to liposomes and FTIR data. Taken together, the present results show that the joint presence of SM and Cho, both in binary and ternary (PC containing) mixtures provide conditions particularly suitable for St II binding and function. We suggest that microdomains present in the bilayers could be important for toxin-membrane association.


Subject(s)
Cholesterol/pharmacology , Cnidarian Venoms/pharmacology , Membrane Lipids/metabolism , Sphingomyelins/pharmacology , Animals , Drug Interactions , Enzyme-Linked Immunosorbent Assay , Liposomes/metabolism , Protein Binding , Sea Anemones
8.
Biopolymers ; 84(2): 169-80, 2006.
Article in English | MEDLINE | ID: mdl-16170802

ABSTRACT

To investigate the role of the N-terminal region in the lytic mechanism of the pore-forming toxin sticholysin II (St II), we studied the conformational and functional properties of peptides encompassing the first 30 residues of the protein. Peptides containing residues 1-30 (P1-30) and 11-30 (P11-30) were synthesized and their conformational properties were examined in aqueous solution as a function of peptide concentration, pH, ionic strength, and addition of the secondary structure-inducing solvent trifluoroethanol (TFE). CD spectra showed that increasing concentration, pH, and ionic strength led to aggregation of P1-30; as a consequence, the peptide acquired beta-sheet conformation. In contrast, P11-30 exhibited practically no conformational changes under the same conditions, remaining essentially structureless. Moreover, this peptide did not undergo aggregation. These differences clearly point to the modulating effect of the first 10 hydrophobic residues on the peptides aggregation and conformational properties. In TFE both the first ten hydrophobic peptides acquired alpha-helical conformation, albeit to a different extent, P11-30 displayed lower alpha-helical content. P1-30 presented a larger fraction of residues in alpha-helical conformation in TFE than that found in St II's crystal structure for that portion of the protein. Since TFE mimics the membrane environment, such increase in helical content could also occur upon toxin binding to membranes and represent a step in the mechanism of pore formation. The peptides conformational properties correlated well with their functional behavior. Thus, P1-30 exhibited much higher hemolytic activity than P11-30. In addition, P11-30 was able to block the toxin's hemolytic activity. The size of pores formed in red blood cells by P1-30 was estimated by measuring the permeability to PEGs of different molecular mass. The pore radius (0.95 +/- 0.01 nm) was very similar to that of the pore formed by the toxin. The results demonstrate that the synthetic peptide P1-30 is a good model of St II conformation and function and emphasize the contribution of the toxin's N-terminal region, and, in particular, the hydrophobic residues 1-10 to pore formation.


Subject(s)
Cnidarian Venoms/chemistry , Cnidarian Venoms/metabolism , Peptides/chemistry , Peptides/metabolism , Amino Acid Sequence , Animals , Cell Membrane Permeability , Circular Dichroism , Cnidarian Venoms/chemical synthesis , Cnidarian Venoms/isolation & purification , Cnidarian Venoms/pharmacology , Cnidarian Venoms/toxicity , Erythrocytes/drug effects , Hemolysin Proteins/toxicity , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Models, Chemical , Molecular Sequence Data , Molecular Weight , Osmolar Concentration , Peptides/chemical synthesis , Polyethylene Glycols/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Sea Anemones/chemistry , Sea Anemones/pathogenicity , Trifluoroethanol/pharmacology , Water/chemistry
9.
Chem Phys Lipids ; 122(1-2): 97-105, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12598041

ABSTRACT

Sticholysins I and II (St I and St II) are water-soluble toxins produced by the sea anemone Stichodactyla helianthus. St I and St II bind to biological and model membranes containing sphingomyelin (SM), forming oligomeric pores that lead to leakage of internal contents. Here we describe functional and structural studies of the toxins aiming at the understanding at a molecular level of their mechanism of binding, as well as their effects on membrane permeabilization. St I and St II caused potassium leakage from red blood cells and temperature-dependent hemolysis, the activation energy of the process being lower for the latter toxin. Protein intrinsic fluorescence measurements provided evidence for toxin binding to model membranes composed of 1:1 (mol:mol) egg phosphatidyl choline (ePC):SM. The fluorescence intensity increased and the maximum emission wavelength decreased as a result of binding. The changes were quantitatively different for both toxins. Circular dichroism spectra showed that both St I and St II exhibit a high content of beta-sheet structure and that binding to model membranes did not alter the toxin's conformation to a large extent. Changing the lipid composition by adding 5 mol% of negatively charged phosphatidic acid (PA) or phosphatidyl glycerol (PG) had small, but detectable, effects on protein conformation. The influence of lipid composition on toxin-induced membrane permeabilization was assessed by means of fluorescence measurements of calcein leakage. The effect was larger for ePC:SM bilayers containing 5 mol% of negative curvature-inducing lipids. Electron paramagnetic resonance (EPR) spectra of intercalated fatty acid spin probes carrying the nitroxide moiety at different carbons (5, 7, 12, and 16) evidenced the occurrence of lipid-protein interaction. Upon addition of the toxins, two-component spectra were observed for the probe labeled at C-12. The broader component, corresponding to a population of strongly immobilized spin probes, was ascribed to boundary lipid. The contribution of this component to the total spectrum was larger for St II than for St I. Moreover, it was clearly detectable for the C-12-labeled probe, but it was absent when the label was at C-16, indicating a lack of lipid-protein interaction close to the lipid terminal methyl group. This effect could be either due to the fact that the toxins do not span the whole bilayer thickness or to the formation of a toroidal pore leading to the preferential interaction with acyl chain carbons closer to the phospholipids head groups.


Subject(s)
Cnidarian Venoms/metabolism , Hemolysin Proteins/metabolism , Lipid Metabolism , Proteins/metabolism , Animals , Circular Dichroism , Electron Spin Resonance Spectroscopy , Hemolysis , Humans , Magnetic Resonance Spectroscopy , Molecular Conformation , Organic Chemicals , Protein Binding , Sea Anemones , Spectrometry, Fluorescence
10.
Toxicon ; 40(8): 1219-221, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12165325

ABSTRACT

Stichodactyla helianthus is a sea anemone relatively abundant along Cuban coasts appearing in two morphos with different colors in their tentacles: green or brownish, probably due to their association with algal symbionts. Traditionally, the brownish morpho has been used as a source of sticholysins I and II, the most characterized cytolysins from this anemone, but the green morpho is the most abundant along the western coasts of Havana. The present work is aimed to establish if the cytolysins purified from the green morpho (StIg and StIIg) are similar to those purified from brownish anemones (StI and StII). Following the same chromatographic procedure used to purify the toxins from morphos, the electrophoretic mobilities, amino acid compositions, amino terminal sequences and molecular masses were practically identical between analogal cytolysins. In conclusion, homologous sticholysins purified from the green and brownish variants of Stichodactyla helianthus are the same molecular entities.


Subject(s)
Cnidarian Venoms/chemistry , Cytotoxins/chemistry , Sea Anemones/chemistry , Amino Acids/analysis , Animals , Caribbean Region , Chromatography, High Pressure Liquid , Cnidarian Venoms/pharmacology , Cytotoxins/pharmacology , Hemolysis , Humans , In Vitro Techniques , Molecular Weight , Peptides/chemistry , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...