Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Toxins (Basel) ; 13(8)2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34437438

RESUMO

Actinoporins (APs) are soluble pore-forming proteins secreted by sea anemones that experience conformational changes originating in pores in the membranes that can lead to cell death. The processes involved in the binding and pore-formation of members of this protein family have been deeply examined in recent years; however, the intracellular responses to APs are only beginning to be understood. Unlike pore formers of bacterial origin, whose intracellular impact has been studied in more detail, currently, we only have knowledge of a few poorly integrated elements of the APs' intracellular action. In this review, we present and discuss an updated landscape of the studies aimed at understanding the intracellular pathways triggered in response to APs attack with particular reference to sticholysin II, the most active isoform produced by the Caribbean Sea anemone Stichodactyla helianthus. To achieve this, we first describe the major alterations these cytolysins elicit on simpler cells, such as non-nucleated mammalian erythrocytes, and then onto more complex eukaryotic cells, including tumor cells. This understanding has provided the basis for the development of novel applications of sticholysins such as the construction of immunotoxins directed against undesirable cells, such as tumor cells, and the design of a cancer vaccine platform. These are among the most interesting potential uses for the members of this toxin family that have been carried out in our laboratory.


Assuntos
Morte Celular/efeitos dos fármacos , Venenos de Cnidários/metabolismo , Venenos de Cnidários/toxicidade , Imunotoxinas/química , Imunotoxinas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Anêmonas-do-Mar/química , Animais
2.
Biophys J ; 117(9): 1563-1576, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31587828

RESUMO

Sticholysins are pore-forming toxins of biomedical interest and represent a prototype of proteins acting through the formation of protein-lipid or toroidal pores. Peptides spanning the N-terminus of sticholysins can mimic their permeabilizing activity and, together with the full-length toxins, have been used as a tool to understand the mechanism of pore formation in membranes. However, the lytic mechanism of these peptides and the lipid shape modulating their activity are not completely clear. In this article, we combine molecular dynamics simulations and experimental biophysical tools to dissect different aspects of the pore-forming mechanism of StII1-30, a peptide derived from the N-terminus of sticholysin II (StII). With this combined approach, membrane curvature induction and flip-flop movement of the lipids were identified as two important membrane remodeling steps mediated by StII1-30. Pore formation by this peptide was enhanced by the presence of the negatively curved lipid phosphatidylethanolamine in membranes. This lipid emerged not only as a facilitator of membrane interactions but also as a structural element of the StII1-30 pore that is recruited to the ring upon its assembly. Collectively, these, to our knowledge, new findings support a toroidal model for the architecture of the pore formed by StII1-30 and provide new molecular insight into the role of phosphatidylethanolamine as a membrane component that can easily integrate into the ring of toroidal pores, thus probably aiding in their stabilization. This study contributes to a better understanding of the molecular mechanism underlying the permeabilizing activity of StII1-30 and peptides or proteins acting via a toroidal pore mechanism and offers an informative framework for the optimization of the biomedical application of this and similar molecules.


Assuntos
Membrana Celular/metabolismo , Venenos de Cnidários/metabolismo , Modelos Moleculares , Sequência de Aminoácidos , Animais , Venenos de Cnidários/química , Bicamadas Lipídicas/química , Simulação de Dinâmica Molecular , Permeabilidade , Fosfatidiletanolaminas/química , Soluções , Suínos
3.
Front Immunol ; 9: 2473, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30455685

RESUMO

Cross-presentation is an important mechanism for the differentiation of effector cytotoxic T lymphocytes (CTL) from naïve CD8+ T-cells, a key response for the clearance of intracellular pathogens and tumors. The liposomal co-encapsulation of the pore-forming protein sticholysin II (StII) with ovalbumin (OVA) (Lp/OVA/StII) induces a powerful OVA-specific CTL activation and an anti-tumor response in vivo. However, the pathway through which the StII contained in this preparation is able to induce antigen cross-presentation and the type of professional antigen presenting cells (APCs) involved have not been elucidated. Here, the ability of mouse bone marrow-derived dendritic cells (BM-DCs) and macrophages (BM-MΦs) stimulated with Lp/OVA/StII to activate SIINFEKL-specific B3Z CD8+ T cells was evaluated in the presence of selected inhibitors. BM-MΦs, but not BM-DCs were able to induce SIINFEKL-specific B3Z CD8+ T cell activation upon stimulation with Lp/OVA/StII. The cross-presentation of OVA was markedly decreased by the lysosome protease inhibitors, leupeptin and cathepsin general inhibitor, while it was unaffected by the proteasome inhibitor epoxomicin. This process was also significantly reduced by phagocytosis and Golgi apparatus function inhibitors, cytochalasin D and brefeldin A, respectively. These results are consistent with the concept that BM-MΦs internalize these liposomes through a phagocytic mechanism resulting in the cross-presentation of the encapsulated OVA by the vacuolar pathway. The contribution of macrophages to the CTL response induced by Lp/OVA/StII in vivo was determined by depleting macrophages with clodronate-containing liposomes. CTL induction was almost completely abrogated in mice depleted of macrophages, demonstrating the relevance of these APCs in the antigen cross-presentation induced by this formulation.


Assuntos
Venenos de Cnidários/metabolismo , Células Dendríticas/fisiologia , Macrófagos/fisiologia , Linfócitos T Citotóxicos/imunologia , Vacúolos/metabolismo , Animais , Antígenos/imunologia , Antígenos CD8/metabolismo , Células Cultivadas , Venenos de Cnidários/química , Apresentação Cruzada , Feminino , Leupeptinas/farmacologia , Lipossomos/química , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia
4.
PLoS One ; 13(8): e0202981, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30161192

RESUMO

Actinoporins sticholysin I and sticholysin II (St I, St II) are proposed to lyse model and biomembranes via toroidal pore formation by their N-terminal domain. Based on the hypothesis that peptide fragments can reproduce the structure and function of this domain, the behavior of peptides containing St I residues 12-31 (StI12-31), St II residues 11-30 (StII11-30), and its TOAC-labeled analogue (N-TOAC-StII11-30) was examined. Molecular modeling showed a good match with experimental structures, indicating amphipathic α-helices in the same regions as in the toxins. CD spectra revealed that the peptides were essentially unstructured in aqueous solution, acquiring α-helical conformation upon interaction with micelles and large unilamellar vesicles (LUV) of variable lipid composition. Fluorescence quenching studies with NBD-containing lipids indicated that N-TOAC-StII11-30's nitroxide moiety is located in the membranes polar head group region. Pyrene-labeled phospholipid inter-leaflet redistribution suggested that the peptides form toroidal pores, according to the mechanism of action proposed for the toxins. Binding occurred only to negatively charged LUV, indicating the importance of electrostatic interactions; in contrast the peptides bound to both negatively charged and zwitterionic micelles, pointing to a lesser influence of these interactions. In addition, differences between bilayers and micelles in head group packing and in curvature led to differences in peptide-membrane interaction. We propose that the peptides topography in micelles resembles that of the toxins in the toroidal pore. The peptides mimicked the toxins permeabilizing activity, St II peptides being more effective than StI12-31. To our knowledge, this is the first demonstration that differences in the toxins N-terminal amphipathic α-helix play a role in the difference between St I and St II activities.


Assuntos
Membrana Celular/metabolismo , Venenos de Cnidários/metabolismo , Animais , Venenos de Cnidários/genética , Venenos de Cnidários/farmacologia , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Hemolíticos/metabolismo , Hemolíticos/farmacologia , Humanos , Bicamadas Lipídicas/química , Micelas , Modelos Moleculares , Compostos Orgânicos/metabolismo , Compostos Orgânicos/farmacologia , Permeabilidade , Conformação Proteica em alfa-Hélice , Anêmonas-do-Mar , Eletricidade Estática
5.
Biochimie ; 138: 20-31, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28396016

RESUMO

Sticholysin II is a pore-forming toxin produced by the sea anemone Stichodactyla helianthus that belongs to the actinoporin protein family. The high affinity of actinoporins for sphingomyelin (SM)-containing membranes has been well documented. However, the molecular determinants that define this affinity have not been fully clarified. Here, we have examined the binding and permeabilizing activity of StII to different single and mixed lipidic systems by combining lipid monolayers, liposomes, and permeabilizing assays. This study characterizes the contribution of ceramide-derived compounds for StII-membrane interaction. Molecular dynamics simulations revealed a differential binding mode of StII with the polar head group of SM and PC. The electrostatic interaction energies were the major energetic contributors to the better affinity of StII for SM compared to PC, while the van der Waals interaction energies were the major driving forces of the better affinity of StII for SM respect to Cer. Furthermore, the presence of sugar residues in glycosphingolipids modulated binding and pore-formation by actinoporins probably by hindering StII to reach relevant structural motifs in membrane for binding or inducing a non-competent adsorption to membrane. Our results demonstrate that StII-membrane interaction, leading to pore formation, may critically respond to changes in lipid head group properties, and the access to SM interfacial structural motif.


Assuntos
Venenos de Cnidários/metabolismo , Simulação de Dinâmica Molecular , Anêmonas-do-Mar/química , Esfingomielinas/metabolismo , Termodinâmica , Animais , Venenos de Cnidários/química , Interações Hidrofóbicas e Hidrofílicas , Bicamadas Lipídicas/química , Lipossomos/química , Esfingomielinas/química
6.
Cent Nerv Syst Agents Med Chem ; 16(3): 173-182, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26201531

RESUMO

Natural products from animal venoms have been used widely in the discovery of novel molecules with particular biological activities that enable their use as potential drug candidates. The phylum Cnidaria (jellyfish, sea anemones, corals zoanthids, hydrozoans, etc.) is the most ancient venomous phylum on earth. Its venoms are composed of a complex mixture of peptidic compounds with neurotoxic and cytolitic properties that have shown activity on mammalian systems despite the fact that they are naturally targeted against fish and invertebrate preys, mainly crustaceans. For this reason, cnidarian venoms are an interesting and vast source of molecules with a remarkable activity on central nervous system, targeting mainly voltage-gated ion channels, ASIC channels, and TRPV1 receptors. In this brief review, we list the amino acid sequences of most cnidarian neurotoxic peptides reported to date. Additionally, we propose the inclusion of a new type of voltage-gated sea anemone sodium channel toxins based on the most recent reports.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Venenos de Cnidários/genética , Venenos de Cnidários/toxicidade , Peptídeos/genética , Peptídeos/toxicidade , Anêmonas-do-Mar , Sequência de Aminoácidos , Animais , Sistema Nervoso Central/metabolismo , Venenos de Cnidários/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Peptídeos/metabolismo
7.
Biochim Biophys Acta ; 1838(7): 1752-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24680653

RESUMO

Sticholysin I (St I) is a pore-forming toxin (PFT) produced by the Caribbean Sea anemone Stichodactyla helianthus belonging to the actinoporin protein family, a unique class of eukaryotic PFT exclusively found in sea anemones. As for actinoporins, it has been proposed that the presence of sphingomyelin (SM) and the coexistence of lipid phases increase binding to the target membrane. However, little is known about the role of membrane structure and dynamics (phase state, fluidity, presence of lipid domains) on actinoporins' activity or which regions of the membrane are the most favorable platforms for protein insertion. To gain insight into the role of SM on the interaction of St I to lipid membranes we studied their binding to monolayers of phosphatidylcholine (PC) and SM in different proportions. Additionally, the effect of acyl chain length and unsaturation, two features related to membrane fluidity, was evaluated on St I binding to monolayers. This study revealed that St I binds and penetrates preferentially and with a faster kinetic to liquid-expanded films with high lateral mobility and moderately enriched in SM. A high content of SM induces a lower lateral diffusion and/or liquid-condensed phases, which hinder St I binding and penetration to the lipid monolayer. Furthermore, the presence of lipid domain borders does not appear as an important factor for St I binding to the lipid monolayer.


Assuntos
Fluidez de Membrana/fisiologia , Lipídeos de Membrana/metabolismo , Esfingomielinas/metabolismo , Animais , Venenos de Cnidários/metabolismo , Cinética , Membranas/metabolismo , Compostos Orgânicos/metabolismo , Fosfatidilcolinas/metabolismo , Anêmonas-do-Mar
8.
Protein J ; 32(8): 593-600, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24197505

RESUMO

Sticholysin II (St II) is a haemolytic toxin isolated from the sea anemone Stichodactyla helianthus. The high haemolytic activity of this toxin is strongly dependent on the red cell status and the macromolecule conformation. In the present communication we evaluate the effect of human serum albumin on St II haemolytic activity and its capacity to form pores in the bilayer of synthetic liposomes. St II retains its pore forming capacity in the presence of large concentrations (up to 500 µM) of human serum albumin. This effect is observed both in its capacity to produce red blood cells haemolysis and to generate functional pores in liposomes. In particular, the capacity of the toxin to lyse red blood cells increases in the presence of human serum albumin (HSA). Regarding the rate of the pore forming process, it is moderately decreased in liposomes and in red blood cells, in spite of an almost total coverage of the interface by albumin. All the data obtained in red cells and model membranes show that St II remains lytically active even in the presence of high HSA concentrations. This stubbornness can explain why the toxin is able to exert its haemolytic activity on membranes immersed in complex plasma matrixes such as those present in living organisms.


Assuntos
Venenos de Cnidários/metabolismo , Hemólise/efeitos dos fármacos , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Anêmonas-do-Mar/química , Albumina Sérica/metabolismo , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Venenos de Cnidários/isolamento & purificação , Eritrócitos/efeitos dos fármacos , Eritrócitos/patologia , Humanos , Lipossomos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/isolamento & purificação
9.
Toxicon ; 54(8): 1135-47, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19268489

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Venenos de Cnidários/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Anêmonas-do-Mar/química , Sequência de Aminoácidos , Animais , Venenos de Cnidários/química , Dados de Sequência Molecular , Compostos Orgânicos/química , Compostos Orgânicos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Anêmonas-do-Mar/metabolismo
10.
Biochim Biophys Acta ; 1760(3): 453-61, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16458433

RESUMO

Two cationic proteins, C1 and C3, were purified to homogeneity from the hemolytic fraction of the venom of Bunodosoma caissarum sea anemone. The purification processes employed gel filtration followed by ion exchange chromatography, being the purity and molecular mass confirmed by SDS-PAGE and mass spectrometry. Protein C1 represented the second major peak of the hemolytic fraction and was previously believed to be a cytolysin belonging to a new class of hemolysins. The C1 protein has a molecular mass of 15495 Da and was assayed for hemolysis, PLA2 activity and acute toxicity in crabs and mice, showing no activity in these assays. It has an amino terminal with no similarity to all known hemolysins and, therefore, should not be considered a toxin, being its function completely unknown. The protein C3 (19757 Da), that also lacks PLA2 activity, was recognized by antiserum against Eqt II and presented high hemolytic activity to human erythrocytes (ED50 of 0.270 microg/ml), being named Caissarolysin I (Bcs I). Its activity was inhibited by pre-incubation with sphingomyelin (SM) and also when in presence of erythrocytes pre-treated with the SMase P2, a phospholipase D from the brown spider Loxosceles intermedia, indicating that SM is the main target of Bcs I. Caissarolysin I is the first hemolysin purified from a sea anemone belonging to the genus Bunodosoma and belongs to the Actinoporin family of sea anemone hemolysins.


Assuntos
Venenos de Cnidários/isolamento & purificação , Anêmonas-do-Mar/química , Sequência de Aminoácidos , Animais , Bioensaio , Cromatografia em Gel , Cromatografia por Troca Iônica , Venenos de Cnidários/metabolismo , Estabilidade de Medicamentos , Proteínas Hemolisinas/efeitos dos fármacos , Proteínas Hemolisinas/isolamento & purificação , Hemólise/efeitos dos fármacos , Temperatura Alta , Humanos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Fosfolipases A/metabolismo , Fosfolipases A2 , Diester Fosfórico Hidrolases/metabolismo , Esfingomielinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA