1
|
Rivero-Hernández AL, Hervis YP, Valdés-Tresanco ME, Escalona-Rodríguez FA, Cancelliere R, Relova-Hernández E, Romero-Hernández G, Pérez-Rivera E, Torres-Palacios Y, Cartaya-Quintero P, Ros U, Porchetta A, Micheli L, Fernández LE, Laborde R, Álvarez C, Sagan S, Lanio ME, Pazos Santos IF. Decoupling immunomodulatory properties from lipid binding in the α-pore-forming toxin Sticholysin II. Int J Biol Macromol 2024; 280:136244. [PMID: 39368578 DOI: 10.1016/j.ijbiomac.2024.136244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Sticholysin II (StII), a pore-forming toxin from the marine anemone Stichodactyla helianthus, enhances an antigen-specific cytotoxic T lymphocyte (CTL) response when co-encapsulated in liposomes with a model antigen. This capacity does not depend exclusively on its pore-forming activity and is partially supported by its ability to activate Toll-like receptor 4 (TLR4) in dendritic cells, presumably by interacting with this receptor or by triggering signaling cascades upon binding to lipid membrane. In order to investigate whether the lipid binding capacity of StII is required for immunomodulation, we designed a mutant in which the aromatic amino acids from the interfacial binding site Trp110, Tyr111 and Trp114 were substituted by Ala. In the present work, we demonstrated that StII3A keeps the secondary structure composition and global folding of StII, while it loses its lipid binding and permeabilization abilities. Despite this, StII3A upregulates dendritic cells maturation markers, enhances an antigen-specific effector CD8+ T cells response and confers antitumor protection in a preventive scenario in C57BL/6 mice. Our results indicate that a mechanism independent of its lipid binding ability is involved in the immunomodulatory capacity of StII, pointing to StII3A as a promising candidate to improve the reliability of the Sts-based vaccine platform.
Collapse
Affiliation(s)
- Ada L Rivero-Hernández
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Yadira P Hervis
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; Sorbonne Université, École normale supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
| | - Mario E Valdés-Tresanco
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; Center for Molecular Simulations and Department of Biological Sciences, University of Calgary, Alberta T2N 1N4, Canada.
| | - Felipe A Escalona-Rodríguez
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Rocco Cancelliere
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Rome 00133, Italy.
| | | | - Glenda Romero-Hernández
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Eric Pérez-Rivera
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | - Yusniel Torres-Palacios
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Patricia Cartaya-Quintero
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba
| | - Uris Ros
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany.
| | - Alessandro Porchetta
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Rome 00133, Italy.
| | - Laura Micheli
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, Rome 00133, Italy.
| | | | - Rady Laborde
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Carlos Álvarez
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Sandrine Sagan
- Sorbonne Université, École normale supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France.
| | - Maria Eliana Lanio
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| | - Isabel F Pazos Santos
- Center for Protein Studies/Department of Biochemistry, Faculty of Biology, University of Havana, Havana 10400, Cuba; NanoCancer, Center of Molecular Immunology (CIM), Havana 11600, Cuba.
| |
Collapse
|
2
|
Potrich C, Pedrotti A, Pederzolli C, Lunelli L. Functional surfaces for exosomes capturing and exosomal microRNAs analysis. Colloids Surf B Biointerfaces 2024; 233:113627. [PMID: 37948834 DOI: 10.1016/j.colsurfb.2023.113627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023]
Abstract
Exosomes are small extracellular vesicles well-studied both as cell signaling elements and as source of highly informative biomarkers, in particular microRNAs. Standard techniques for exosome isolation are in general scarcely efficient and give low purity vesicles. New techniques combining microfluidics with suitable functionalized surfaces could overcome these disadvantages. Here, different functional surfaces aimed at exosomes capture are developed thank to the functionalization of silicon oxide substrates. Charged surfaces, both positive and negative, neutral and immunoaffinity surfaces are characterized and tested in functional assays with both exosome mimicking vesicles and exosomes purified from cell supernatants. The different surfaces showed promising properties, in particular the negatively-charged surface could capture more than 4 × 108 exosomes per square centimeter. The captured exosomes could be recovered and their biomarker cargo analyzed. Exosomal microRNAs were successfully analyzed with RT-PCR, confirming the good performances of the negatively-charged surface. The best-performing functionalization could be easily moved to microdevice surfaces for developing modular microfluidic systems for on-chip isolation of exosomes, to be integrated in simple and fast biosensors aimed at biomarker analysis both in clinical settings and in research.
Collapse
Affiliation(s)
- Cristina Potrich
- FBK-Fondazione Bruno Kessler, Center for Sensors and Devices, via Sommarive, 18, I-38123, Trento, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biofisica, via alla Cascata 56/C, I-38123, Trento, Italy.
| | - Anna Pedrotti
- FBK-Fondazione Bruno Kessler, Center for Sensors and Devices, via Sommarive, 18, I-38123, Trento, Italy
| | - Cecilia Pederzolli
- FBK-Fondazione Bruno Kessler, Center for Sensors and Devices, via Sommarive, 18, I-38123, Trento, Italy
| | - Lorenzo Lunelli
- FBK-Fondazione Bruno Kessler, Center for Sensors and Devices, via Sommarive, 18, I-38123, Trento, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biofisica, via alla Cascata 56/C, I-38123, Trento, Italy
| |
Collapse
|
3
|
Donato M, Soto C, Lanio ME, Itri R, Álvarez C. The pore-forming activity of sticholysin I is enhanced by the presence of a phospholipid hydroperoxide in membrane. Toxicon 2021; 204:44-55. [PMID: 34736955 DOI: 10.1016/j.toxicon.2021.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 10/29/2021] [Indexed: 11/19/2022]
Abstract
Sticholysin I (StI) is a pore-forming toxin (PFT) belonging to the actinoporin protein family characterized by high permeabilizing activity in membranes. StI readily associates with sphingomyelin (SM)-containing membranes originating pores that can lead to cell death. Binding and pore-formation are critically dependent on the physicochemical properties of membrane. 1-palmitoyl-2-oleoylphosphatidylcholine hydroperoxide (POPC-OOH) is an oxidized phospholipid (OxPL) containing an -OOH moiety in the unsaturated hydrocarbon chain which orientates towards the bilayer interface. This orientation causes an increase in the lipid molecular area, lateral expansion and decrease in bilayer thickness, elastic and bending modulus, as well as modification of lipid packing. Taking advantage of membrane structural changes promoted by POPC-OOH, we investigated its influence on the permeabilizing ability of StI. Here we report the action of StI on Giant Unilamellar Vesicles (GUVs) made of 1-palmitoyl-2-oleoylphosphatidylcholine (POPC) and SM containing increasing amount of POPC-OOH to assess vesicle permeability changes when compared to OxPL-lacking membranes. Inclusion of POPC-OOH in membranes did not promote spontaneous vesicle leaking but resulted in increased membrane permeability due to StI action. StI activity did not modify the fluid-gel phase coexistence boundaries neither in POPC:SM or POPC-OOH:SM membranes. However, the StI insertion mechanism in membrane seems to differ between POPC:SM and POPC-OOH:SM mixtures as suggested by changes in the time course of monolayer surface tension measurements, even though a preferable binding of the toxin to OxPL-containing systems could not be here demonstrated. In summary, modifications in the membrane imposed by lipid hydroperoxidation favor StI permeabilizing activity.
Collapse
Affiliation(s)
- Maressa Donato
- Instituto de Física, Universidade de São Paulo (USP), São Paulo, SP, Brazil; Center for Laser and Applications, Nuclear and Energy Research Institute, São Paulo, Brazil
| | - Carmen Soto
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana, CP, 10400, La Habana, Cuba
| | - María Eliana Lanio
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana, CP, 10400, La Habana, Cuba
| | - Rosangela Itri
- Instituto de Física, Universidade de São Paulo (USP), São Paulo, SP, Brazil.
| | - Carlos Álvarez
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana, CP, 10400, La Habana, Cuba.
| |
Collapse
|
4
|
Alvarez C, Soto C, Cabezas S, Alvarado-Mesén J, Laborde R, Pazos F, Ros U, Hernández AM, Lanio ME. Panorama of the Intracellular Molecular Concert Orchestrated by Actinoporins, Pore-Forming Toxins from Sea Anemones. Toxins (Basel) 2021; 13:toxins13080567. [PMID: 34437438 PMCID: PMC8402351 DOI: 10.3390/toxins13080567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/25/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Carlos Alvarez
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
- Correspondence:
| | - Carmen Soto
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
| | - Sheila Cabezas
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
| | - Javier Alvarado-Mesén
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
- Escuela de Ciencias Biológicas, Universidad Nacional, Heredia 40101, Costa Rica
| | - Rady Laborde
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
| | - Fabiola Pazos
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
| | - Uris Ros
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-strasse 26, 50931 Cologne, Germany
| | - Ana María Hernández
- Immunobiology Division, Molecular Immunology Institute, Center of Molecular Immunology (CIM), Playa, Havana CP 11600, Cuba;
| | - María Eliana Lanio
- Centro de Estudio de Proteínas, Facultad de Biología, Universidad de La Habana (UH) and Laboratorio UH-Centro de Inmunología Molecular, Havana CP 11600, Cuba; (C.S.); (S.C.); (J.A.-M.); (R.L.); (F.P.); (U.R.); (M.E.L.)
| |
Collapse
|
5
|
Palacios-Ortega J, García-Linares S, Rivera-de-Torre E, Heras-Márquez D, Gavilanes JG, Slotte JP, Martínez-Del-Pozo Á. Structural foundations of sticholysin functionality. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140696. [PMID: 34246789 DOI: 10.1016/j.bbapap.2021.140696] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023]
Abstract
Actinoporins constitute a family of α pore-forming toxins produced by sea anemones. The soluble fold of these proteins consists of a β-sandwich flanked by two α-helices. Actinoporins exert their activity by specifically recognizing sphingomyelin at their target membranes. Once there, they penetrate the membrane with their N-terminal α-helices, a process that leads to the formation of cation-selective pores. These pores kill the target cells by provoking an osmotic shock on them. In this review, we examine the role and relevance of the structural features of actinoporins, down to the residue level. We look at the specific amino acids that play significant roles in the function of actinoporins and their fold. Particular emphasis is given to those residues that display a high degree of conservation across the actinoporin sequences known to date. In light of the latest findings in the field, the membrane requirements for pore formation, the effect of lipid composition, and the process of pore formation are also discussed.
Collapse
Affiliation(s)
- Juan Palacios-Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain; Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| | - Sara García-Linares
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - Esperanza Rivera-de-Torre
- Department of Biochemistry and Biotechnology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Diego Heras-Márquez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - José G Gavilanes
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - J Peter Slotte
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Álvaro Martínez-Del-Pozo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| |
Collapse
|
6
|
MLKL promotes cellular differentiation in myeloid leukemia by facilitating the release of G-CSF. Cell Death Differ 2021; 28:3235-3250. [PMID: 34079078 PMCID: PMC8630008 DOI: 10.1038/s41418-021-00811-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 04/30/2021] [Accepted: 05/19/2021] [Indexed: 11/15/2022] Open
Abstract
The blockade of cellular differentiation represents a hallmark of acute myeloid leukemia (AML), which is largely attributed to the dysfunction of lineage-specific transcription factors controlling cellular differentiation. However, alternative mechanisms of cellular differentiation programs in AML remain largely unexplored. Here we report that mixed lineage kinase domain-like protein (MLKL) contributes to the cellular differentiation of transformed hematopoietic progenitor cells in AML. Using gene-targeted mice, we show that MLKL facilitates the release of granulocyte colony-stimulating factor (G-CSF) by controlling membrane permeabilization in leukemic cells. Mlkl−/− hematopoietic stem and progenitor cells released reduced amounts of G-CSF while retaining their capacity for CSF3 (G-CSF) mRNA expression, G-CSF protein translation, and G-CSF receptor signaling. MLKL associates with early endosomes and controls G-CSF release from intracellular storage by plasma membrane pore formation, whereas cell death remained unaffected by loss of MLKL. Of note, MLKL expression was significantly reduced in AML patients, specifically in those with a poor-risk AML subtype. Our data provide evidence that MLKL controls myeloid differentiation in AML by controlling the release of G-CSF from leukemic progenitor cells.
Collapse
|
7
|
Ferroptotic pores induce Ca 2+ fluxes and ESCRT-III activation to modulate cell death kinetics. Cell Death Differ 2021; 28:1644-1657. [PMID: 33335287 PMCID: PMC8167089 DOI: 10.1038/s41418-020-00691-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 10/30/2020] [Accepted: 11/17/2020] [Indexed: 01/28/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated necrosis associated with lipid peroxidation. Despite its key role in the inflammatory outcome of ferroptosis, little is known about the molecular events leading to the disruption of the plasma membrane during this type of cell death. Here we show that a sustained increase in cytosolic Ca2+ is a hallmark of ferroptosis that precedes complete bursting of the cell. We report that plasma membrane damage leading to ferroptosis is associated with membrane nanopores of a few nanometers in radius and that ferroptosis, but not lipid peroxidation, can be delayed by osmoprotectants. Importantly, Ca2+ fluxes during ferroptosis induce the activation of the ESCRT-III-dependent membrane repair machinery, which counterbalances the kinetics of cell death and modulates the immunological signature of ferroptosis. Our findings with ferroptosis provide a unifying concept that sustained increase of cytosolic Ca2+ prior to plasma membrane rupture is a common feature of regulated types of necrosis and position ESCRT-III activation as a general protective mechanism in these lytic cell death pathways.
Collapse
|
8
|
Chorieva NM, Fayziev DD, Tsiferova NA, Toshtemirova GA, Khamidova OJ, Merzlyak PG, Kurbannazarova RS, Ziyaev KL, Gafurov MB, Sabirov RZ. Lytic and sublytic effects of gossypol on red blood cells and thymocytes. Clin Exp Pharmacol Physiol 2021; 48:227-237. [PMID: 33124084 DOI: 10.1111/1440-1681.13429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 01/21/2023]
Abstract
Gossypol is a natural polyphenol presently considered as a promising biological phytochemical with a range of activities including anticancer. We examined volume regulation-dependent effects of gossypol using erythrocytes and thymic lymphocytes. Gossypol effectively lysed human red blood cells (RBC) with a half-maximal concentration of 67.4 ± 1.6 μmol/L and in a non-colloid osmotic manner. Sublytic gossypol doses of 1-10 μmol/L significantly protected RBC from osmotic hemolysis, but potentiated their sensitivity to the colloid-osmotic lysis induced by a pore-former nystatin. When added to the thymocytes suspension, gossypol caused a strong depression of the ability of cells to restore their volume under hypoosmotic stress with a half-maximal activity at 2.1 ± 0.3 μmol/L. Gossypol suppressed regulatory volume decrease under experimental conditions, when cationic permeability was controlled by gramicidin D, and volume recovery depended mainly on anionic conductance, suggesting that the polyphenol inhibits the swelling-induced anion permeability. In direct patch-clamp experiments, gossypol inhibited the volume-sensitive outwardly rectifying (VSOR) chloride channel in thymocytes and in human HCT116 and HeLa cells, possibly by a mechanism when gossypol molecule with a radius close to the size of channel pore plugs into the narrowest portion of the native VSOR chloride channel. Micromolar gossypol suppressed proliferation of thymocytes, HCT116 and HeLa cells. VSOR blockage may represent new mechanism of anticancer activity of gossypol in addition to its action as a BH3-mimetic.
Collapse
Affiliation(s)
- Nargiza M Chorieva
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Termez State University, Termez, Uzbekistan
| | - Diyor D Fayziev
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Nargiza A Tsiferova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Center for Advanced Technologies, Tashkent, Uzbekistan
| | - Gulnoza A Toshtemirova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Ozoda J Khamidova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Petr G Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Ranokhon Sh Kurbannazarova
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Technical Institute of the National Guard, Tashkent, Uzbekistan
| | - Khayrulla L Ziyaev
- Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan
| | - Makhmud B Gafurov
- Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan
| | - Ravshan Z Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
- Department of Biophysics, National University of Uzbekistan, Tashkent, Uzbekistan
| |
Collapse
|
9
|
Pore-forming proteins: From defense factors to endogenous executors of cell death. Chem Phys Lipids 2020; 234:105026. [PMID: 33309552 DOI: 10.1016/j.chemphyslip.2020.105026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Pore-forming proteins (PFPs) and small antimicrobial peptides (AMPs) represent a large family of molecules with the common ability to punch holes in cell membranes to alter their permeability. They play a fundamental role as infectious bacteria's defensive tools against host's immune system and as executors of endogenous machineries of regulated cell death in eukaryotic cells. Despite being highly divergent in primary sequence and 3D structure, specific folds of pore-forming domains have been conserved. In fact, pore formation is considered an ancient mechanism that takes place through a general multistep process involving: membrane partitioning and insertion, oligomerization and pore formation. However, different PFPs and AMPs assemble and form pores following different mechanisms that could end up either in the formation of protein-lined or protein-lipid pores. In this review, we analyze the current findings in the mechanism of action of different PFPs and AMPs that support a wide role of membrane pore formation in nature. We also provide the newest insights into the development of state-of-art techniques that have facilitated the characterization of membrane pores. To understand the physiological role of these peptides/proteins or develop clinical applications, it is essential to uncover the molecular mechanism of how they perforate membranes.
Collapse
|
10
|
Pore-forming toxins from sea anemones: from protein-membrane interaction to its implications for developing biomedical applications. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2020. [DOI: 10.1016/bs.abl.2020.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Mesa-Galloso H, Valiente PA, Valdés-Tresanco ME, Epand RF, Lanio ME, Epand RM, Alvarez C, Tieleman DP, Ros U. Membrane Remodeling by the Lytic Fragment of SticholysinII: Implications for the Toroidal Pore Model. Biophys J 2019; 117:1563-1576. [PMID: 31587828 DOI: 10.1016/j.bpj.2019.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 01/15/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Haydee Mesa-Galloso
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, Calgary, Alberta, Canada; Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba
| | - Pedro A Valiente
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba
| | - Mario E Valdés-Tresanco
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, Calgary, Alberta, Canada; Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba
| | - Raquel F Epand
- Department of Biochemistry and Biomedical Sciences, Health Science Center, McMaster University, Hamilton, Ontario, Canada
| | - Maria E Lanio
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, Health Science Center, McMaster University, Hamilton, Ontario, Canada
| | - Carlos Alvarez
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba
| | - D Peter Tieleman
- Department of Biological Sciences and Centre for Molecular Simulation, University of Calgary, Calgary, Alberta, Canada.
| | - Uris Ros
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba; Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
12
|
Hervis YP, Valle A, Dunkel S, Klare JP, Canet L, Lanio ME, Alvarez C, Pazos IF, Steinhoff HJ. Architecture of the pore forming toxin sticholysin I in membranes. J Struct Biol 2019; 208:30-42. [PMID: 31330179 DOI: 10.1016/j.jsb.2019.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 12/28/2022]
Abstract
Sticholysin I (StI) is a toxin produced by the sea anemone Stichodactyla helianthus and belonging to the actinoporins family. Upon binding to sphingomyelin-containing membranes StI forms oligomeric pores, thereby leading to cell death. According to recent controversial experimental evidences, the pore architecture of actinoporins is a debated topic. Here, we investigated the StI topology in membranes by site-directed spin labeling and electron paramagnetic resonance spectroscopy. The results reveal that StI in membrane exhibits an oligomeric architecture with heterogeneous stoichiometry of predominantly eight or nine protomers, according to the available structural models. The StI topology resembles the conic pore structure reported for the actinoporin fragaceatoxin C. Our data show that StI coexists in two membrane-associated conformations, with the N-terminal segment either attached to the protein core or inserted in the membrane forming the pore. This finding suggests a 'pre-pore' to 'pore' transition determined by a conformational change that detaches the N-terminal segment.
Collapse
Affiliation(s)
- Yadira P Hervis
- Center for Protein Studies/Department of Biochemistry, University of Havana, Calle 25 #455 e/I y J, Vedado, Plaza de la Revolución, ZIP 10400, Havana, Cuba.
| | - Aisel Valle
- Center for Protein Studies/Department of Biochemistry, University of Havana, Calle 25 #455 e/I y J, Vedado, Plaza de la Revolución, ZIP 10400, Havana, Cuba.
| | - Sabrina Dunkel
- Department of Physics, University of Osnabrueck, Barbarastr. 7, 49076 Osnabrueck, Germany.
| | - Johann P Klare
- Department of Physics, University of Osnabrueck, Barbarastr. 7, 49076 Osnabrueck, Germany.
| | - Liem Canet
- Center for Protein Studies/Department of Biochemistry, University of Havana, Calle 25 #455 e/I y J, Vedado, Plaza de la Revolución, ZIP 10400, Havana, Cuba.
| | - Maria E Lanio
- Center for Protein Studies/Department of Biochemistry, University of Havana, Calle 25 #455 e/I y J, Vedado, Plaza de la Revolución, ZIP 10400, Havana, Cuba.
| | - Carlos Alvarez
- Center for Protein Studies/Department of Biochemistry, University of Havana, Calle 25 #455 e/I y J, Vedado, Plaza de la Revolución, ZIP 10400, Havana, Cuba.
| | - Isabel F Pazos
- Center for Protein Studies/Department of Biochemistry, University of Havana, Calle 25 #455 e/I y J, Vedado, Plaza de la Revolución, ZIP 10400, Havana, Cuba.
| | - Heinz-J Steinhoff
- Department of Physics, University of Osnabrueck, Barbarastr. 7, 49076 Osnabrueck, Germany.
| |
Collapse
|
13
|
Espiritu RA, Pedrera L, Ros U. Tuning the way to die: implications of membrane perturbations in necroptosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/bs.abl.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Ros U, Carretero GPB, Paulino J, Crusca E, Pazos F, Cilli EM, Lanio ME, Schreier S, Alvarez C. Self-association and folding in membrane determine the mode of action of peptides from the lytic segment of sticholysins. Biochimie 2018; 156:109-117. [PMID: 30326255 DOI: 10.1016/j.biochi.2018.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/11/2018] [Indexed: 01/09/2023]
Abstract
Sticholysin I and II (Sts: St I and St II) are proteins of biomedical interest that form pores upon the insertion of their N-terminus in the plasma membrane. Peptides spanning the N-terminal residues of StI (StI1-31) or StII (StII1-30) can mimic the permeabilizing ability of these toxins, emerging as candidates to rationalize their potential biomedical applications. These peptides have different activities that correlate with their hydrophobicity. However, it is not clear how this property contributes to peptide folding in solution or upon binding to membranes. Here we compared the conformational properties of these peptides and shorter versions lacking the hydrophobic segment 1-11 of StI (StI12-31) or 1-10 of StII (StII11-30). Folding of peptides was assessed in solution and in membrane mimetic systems and related with their ability to bind to membranes and to permeabilize lipid vesicles. Our results suggest that the differences in activity among peptides could be ascribed to their different folding propensity and different membrane binding properties. In solution, StII1-30 tends to acquire α-helical conformation coexisting with self-associated structures, while StI1-31 remains structureless. Both peptides fold as α-helix in membrane; but StII1-30 also self-associates in the lipid environment, a process that is favored by its higher affinity for membrane. We stress the contribution of the non-polar/polar balance of the 1-10 amino acid sequence of the peptides as a determining factor for different self-association capabilities. Such difference in hydrophobicity seems to determine the molecular path of peptides folding upon binding to membranes, with an impact in their permeabilizing activity. This study contributes to a better understanding of the molecular mechanisms underlying the permeabilizing activity of Sts N-terminal derived peptides, with connotation for the exploitation of these small molecules as alternative of the full-length toxins in clinical settings.
Collapse
Affiliation(s)
- Uris Ros
- Center for Protein Studies, Biology Faculty, University of Havana, Havana, Cuba
| | - Gustavo P B Carretero
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Joana Paulino
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Edson Crusca
- Department of Biochemistry, Institute of Chemistry, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Fabiola Pazos
- Center for Protein Studies, Biology Faculty, University of Havana, Havana, Cuba
| | - Eduardo M Cilli
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Maria E Lanio
- Center for Protein Studies, Biology Faculty, University of Havana, Havana, Cuba
| | - Shirley Schreier
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Carlos Alvarez
- Center for Protein Studies, Biology Faculty, University of Havana, Havana, Cuba.
| |
Collapse
|
15
|
Carretero GPB, Vicente EF, Cilli EM, Alvarez CM, Jenssen H, Schreier S. Dissecting the mechanism of action of actinoporins. Role of the N-terminal amphipathic α-helix in membrane binding and pore activity of sticholysins I and II. PLoS One 2018; 13:e0202981. [PMID: 30161192 PMCID: PMC6117003 DOI: 10.1371/journal.pone.0202981] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/13/2018] [Indexed: 11/19/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Gustavo P. B. Carretero
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Eduardo F. Vicente
- Faculty of Science and Engineering, State University of São Paulo, Tupã, Brazil
| | - Eduardo M. Cilli
- Institute of Chemistry, State University of São Paulo, Araraquara, Brazil
| | | | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Shirley Schreier
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
16
|
Cloning, purification and characterization of nigrelysin, a novel actinoporin from the sea anemone Anthopleura nigrescens. Biochimie 2018; 156:206-223. [PMID: 30036605 DOI: 10.1016/j.biochi.2018.07.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/19/2018] [Indexed: 12/15/2022]
Abstract
Actinoporins constitute a unique class of pore-forming toxins found in sea anemones that being secreted as soluble monomers are able to bind and permeabilize membranes leading to cell death. The interest in these proteins has risen due to their high cytotoxicity that can be properly used to design immunotoxins against tumor cells and antigen-releasing systems to cell cytosol. In this work we describe a novel actinoporin produced by Anthopleura nigrescens, an anemone found in the Central American Pacific Ocean. Here we report the amino acid sequence of an actinoporin as deduced from cDNA obtained from total body RNA. The synthetic DNA sequence encoding for one cytolysin variant was expressed in BL21 Star (DE3) Escherichia coli and the protein purified by chromatography on CM Sephadex C-25 with more than 97% homogeneity as verified by MS-MS and HPLC analyses. This actinoporin comprises 179 amino acid residues, consistent with its observed isotope-averaged molecular mass of 19 661 Da. The toxin lacks Cys and readily permeabilizes erythrocytes, as well as L1210 cells. CD spectroscopy revealed that its secondary structure is dominated by beta structure (58.5%) with 5.5% of α-helix, and 35% of random structure. Moreover, binding experiments to lipidic monolayers and to liposomes, as well as permeabilization studies in vesicles, revealed that the affinity of this toxin for sphingomyelin-containing membranes is quite similar to sticholysin II (StII). Comparison by spectroscopic techniques and modeling the three-dimensional structure of nigrelysin (Ng) showed a high homology with StII but several differences were also detectable. Taken together, these results reinforce the notion that Ng is a novel member of the actinoporin pore-forming toxin (PFT) family with a HA as high as that of StII, the most potent actinoporin so far described, but with peculiar structural characteristics contributing to expand the understanding of the structure-function relationship in this protein family.
Collapse
|
17
|
Disassembling a cancer puzzle: Cell junctions and plasma membrane as targets for anticancer therapy. J Control Release 2018; 286:125-136. [PMID: 30030181 DOI: 10.1016/j.jconrel.2018.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023]
Abstract
Despite an enhanced permeability and retention effect typical of many solid tumors, drug penetration is not always sufficient. Possible strategies for the drug delivery improvement are a modification of the tumor cell-to-cell junctions and usage of cell membrane permeabilization proteins. In this review we discuss epithelial cell junctions as targets for a combined anticancer therapy and propose new possible sources of such agents. We suggest considering viral and bacterial pathogens disrupting epithelial layers as plentiful sources of new therapeutic agents for increasing tumor permeability for other effector agents. We also observe the application of pore forming proteins and peptides of different origin for cytoplasmic delivery of anti-cancer agents and consider the main obstacles of their use in vivo.
Collapse
|
18
|
Soto C, Bergado G, Blanco R, Griñán T, Rodríguez H, Ros U, Pazos F, Lanio ME, Hernández AM, Álvarez C. Sticholysin II-mediated cytotoxicity involves the activation of regulated intracellular responses that anticipates cell death. Biochimie 2018; 148:18-35. [DOI: 10.1016/j.biochi.2018.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/07/2018] [Indexed: 12/12/2022]
|
19
|
Tanaka K, Caaveiro JMM, Morante K, Tsumoto K. Haemolytic actinoporins interact with carbohydrates using their lipid-binding module. Philos Trans R Soc Lond B Biol Sci 2018. [PMID: 28630155 DOI: 10.1098/rstb.2016.0216] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Pore-forming toxins (PFTs) are proteins endowed with metamorphic properties that enable them to stably fold in water solutions as well as in cellular membranes. PFTs produce lytic pores on the plasma membranes of target cells conducive to lesions, playing key roles in the defensive and offensive molecular systems of living organisms. Actinoporins are a family of potent haemolytic toxins produced by sea anemones vigorously studied as a paradigm of α-helical PFTs, in the context of lipid-protein interactions, and in connection with nanopore technologies. We have recently reported that fragaceatoxin C (FraC), an actinoporin, engages biological membranes with a large adhesive motif allowing the simultaneous attachment of up to four lipid molecules prior to pore formation. Since actinoporins also interact with carbohydrates, we sought to understand the molecular and energetic basis of glycan recognition by FraC. By employing structural and biophysical methodologies, we show that FraC engages glycans with low affinity using its lipid-binding module. Contrary to other PFTs requiring separate domains for glycan and lipid recognition, the small single-domain actinoporins economize resources by achieving dual recognition with a single binding module. This mechanism could enhance the recruitment of actinoporins to the surface of target tissues in their marine environment.This article is part of the themed issue 'Membrane pores: from structure and assembly, to medicine and technology'.
Collapse
Affiliation(s)
- Koji Tanaka
- Department of Chemistry and Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Jose M M Caaveiro
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Koldo Morante
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Chemistry and Biotechnology, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan .,Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan.,The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
20
|
Lima de Oliveira A, Maffud Cilli E, Ros U, Crusca E, Lanio ME, Alvarez C, Schreier S, Aguiar Pertinhez T, Spisni A. Insights on the structure-activity relationship of peptides derived from Sticholysin II. Biopolymers 2018; 110. [PMID: 29359791 DOI: 10.1002/bip.23097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/26/2017] [Accepted: 12/03/2017] [Indexed: 02/06/2023]
Abstract
Sticholysin II (StII) is a pore-forming actinoporin from the sea anemone Stichodactyla helianthus. A mechanistic model of its action has been proposed: proteins bind to cell membrane, insert their N-termini into the lipid core and assemble into homo-tetramer pores responsible for host-cell death. Because very likely the first 10 residues of StII N-terminus are critical for membrane penetration, to dissect the molecular details of that functionality, we studied two synthetic peptides: StII1-30 and StII16-35 . They show diverse haemolytic and candidacidal activity that correlate with distinct orientations in SDS micelles. NMR shows that StII1-30 partly inserts into the micelle, while StII16-35 lays on the micelle surface. These results justify the diverse concentration dependence of their candidacidal activity supposing a different mechanism of action and providing new hints on StII lytic activity at molecular level. Biotechnological application of these peptides, focused on the development of therapeutic immunocomplexes, may be envisaged.
Collapse
Affiliation(s)
- Aline Lima de Oliveira
- Institute of Chemistry, University of Brasilia, Brasilia, DF, Brazil
- Department of Biochemistry and Chemical Technology, Institute of Chemistry, São Paulo State University, Araraquara, Brazil
| | | | - Uris Ros
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Edson Crusca
- Center for Protein Studies, Biology Faculty, University of Havana, Havana, Cuba
| | - María Eliana Lanio
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Carlos Alvarez
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Department of Medicine and Surgery, Unit of Biochemistry, University of Parma, Parma, Italy
| | - Shirley Schreier
- Department of Medicine and Surgery, Unit of Biochemistry, University of Parma, Parma, Italy
| | - Thelma Aguiar Pertinhez
- Department of Biochemistry and Chemical Technology, Institute of Chemistry, São Paulo State University, Araraquara, Brazil
- Transfusion Medicine Unit, AUSL - IRCCS Reggio Emilia, Italy
| | - Alberto Spisni
- Department of Biochemistry and Chemical Technology, Institute of Chemistry, São Paulo State University, Araraquara, Brazil
| |
Collapse
|
21
|
Ros U, Peña-Blanco A, Hänggi K, Kunzendorf U, Krautwald S, Wong WWL, García-Sáez AJ. Necroptosis Execution Is Mediated by Plasma Membrane Nanopores Independent of Calcium. Cell Rep 2017; 19:175-187. [PMID: 28380356 PMCID: PMC5465952 DOI: 10.1016/j.celrep.2017.03.024] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/22/2016] [Accepted: 03/06/2017] [Indexed: 01/14/2023] Open
Abstract
Necroptosis is a form of regulated necrosis that
results in cell death and content release after plasma membrane permeabilization.
However, little is known about the molecular events responsible for the disruption of
the plasma membrane. Here, we find that early increase in cytosolic calcium in
TNF-induced necroptosis is mediated by treatment with a Smac mimetic via the
TNF/RIP1/TAK1 survival pathway. This does not require the activation of the necrosome
and is dispensable for necroptosis. Necroptosis induced by the activation of TLR3/4
pathways does not trigger early calcium flux. We also demonstrate that necroptotic
plasma membrane rupture is mediated by osmotic forces and membrane pores around 4 nm
in diameter. This late permeabilization step represents a hallmark in necroptosis
execution that is cell and treatment independent and requires the RIP1/RIP3/MLKL
core. In support of this, treatment with osmoprotectants reduces cell damage in an
in vivo necroptosis model of ischemia-reperfusion injury. Early calcium signaling in TSZ necroptosis correlates with
cellular levels of cIAP1/2 Calcium flux is induced by a Smac mimetic and is
dispensable for necroptosis execution Pores ∼4 nm in diameter mediate final plasma membrane
disruption in necroptosis
Collapse
Affiliation(s)
- Uris Ros
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany.
| | - Aida Peña-Blanco
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany
| | - Kay Hänggi
- Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Ulrich Kunzendorf
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - W Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Tübingen University, 72076 Tübingen, Germany; Max-Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany.
| |
Collapse
|
22
|
Biophysical and biochemical strategies to understand membrane binding and pore formation by sticholysins, pore-forming proteins from a sea anemone. Biophys Rev 2017; 9:529-544. [PMID: 28853034 DOI: 10.1007/s12551-017-0316-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022] Open
Abstract
Actinoporins constitute a unique class of pore-forming toxins found in sea anemones that are able to bind and oligomerize in membranes, leading to cell swelling, impairment of ionic gradients and, eventually, to cell death. In this review we summarize the knowledge generated from the combination of biochemical and biophysical approaches to the study of sticholysins I and II (Sts, StI/II), two actinoporins largely characterized by the Center of Protein Studies at the University of Havana during the last 20 years. These approaches include strategies for understanding the toxin structure-function relationship, the protein-membrane association process leading to pore formation and the interaction of toxin with cells. The rational combination of experimental and theoretical tools have allowed unraveling, at least partially, of the complex mechanisms involved in toxin-membrane interaction and of the molecular pathways triggered upon this interaction. The study of actinoporins is important not only to gain an understanding of their biological roles in anemone venom but also to investigate basic molecular mechanisms of protein insertion into membranes, protein-lipid interactions and the modulation of protein conformation by lipid binding. A deeper knowledge of the basic molecular mechanisms involved in Sts-cell interaction, as described in this review, will support the current investigations conducted by our group which focus on the design of immunotoxins against tumor cells and antigen-releasing systems to cell cytosol as Sts-based vaccine platforms.
Collapse
|
23
|
Soto C, del Valle A, Valiente PA, Ros U, Lanio ME, Hernández AM, Alvarez C. Differential binding and activity of the pore-forming toxin sticholysin II in model membranes containing diverse ceramide-derived lipids. Biochimie 2017; 138:20-31. [DOI: 10.1016/j.biochi.2017.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/04/2017] [Indexed: 01/07/2023]
|
24
|
Characterization of pore size distribution (PSD) in cellulose triacetate (CTA) and polyamide (PA) thin active layers by positron annihilation lifetime spectroscopy (PALS) and fractional rejection (FR) method. J Memb Sci 2017. [DOI: 10.1016/j.memsci.2016.12.064] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Laborde RJ, Sanchez-Ferras O, Luzardo MC, Cruz-Leal Y, Fernández A, Mesa C, Oliver L, Canet L, Abreu-Butin L, Nogueira CV, Tejuca M, Pazos F, Álvarez C, Alonso ME, Longo-Maugéri IM, Starnbach MN, Higgins DE, Fernández LE, Lanio ME. Novel Adjuvant Based on the Pore-Forming Protein Sticholysin II Encapsulated into Liposomes Effectively Enhances the Antigen-Specific CTL-Mediated Immune Response. THE JOURNAL OF IMMUNOLOGY 2017; 198:2772-2784. [PMID: 28258198 DOI: 10.4049/jimmunol.1600310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022]
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.
Collapse
Affiliation(s)
- Rady J Laborde
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Oraly Sanchez-Ferras
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - María C Luzardo
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Yoelys Cruz-Leal
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Audry Fernández
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Circe Mesa
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Liliana Oliver
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Liem Canet
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Liane Abreu-Butin
- Discipline of Immunology, Department of Microbiology, Immunology, and Parasitology, Paulista Medical School, Federal University of São Paulo, São Paulo 04023-900, Brazil; and
| | - Catarina V Nogueira
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Mayra Tejuca
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Fabiola Pazos
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Carlos Álvarez
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - María E Alonso
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Ieda M Longo-Maugéri
- Discipline of Immunology, Department of Microbiology, Immunology, and Parasitology, Paulista Medical School, Federal University of São Paulo, São Paulo 04023-900, Brazil; and
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Darren E Higgins
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Luis E Fernández
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba;
| | - María E Lanio
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba;
| |
Collapse
|
26
|
Ahumada M, Calderon C, Lissi E, Alvarez C, Lanio M, Pazos F. The pore forming capacity of Sticholysin I in dipalmitoyl phosphatidyl vesicles is tuned by osmotic stress. Chem Phys Lipids 2017; 203:87-93. [DOI: 10.1016/j.chemphyslip.2016.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/28/2016] [Accepted: 12/28/2016] [Indexed: 11/25/2022]
|
27
|
Cabezas S, Ho S, Ros U, Lanio ME, Alvarez C, van der Goot FG. Damage of eukaryotic cells by the pore-forming toxin sticholysin II: Consequences of the potassium efflux. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:982-992. [PMID: 28173991 DOI: 10.1016/j.bbamem.2017.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/06/2017] [Accepted: 02/03/2017] [Indexed: 01/17/2023]
Abstract
Pore-forming toxins (PFTs) form holes in membranes causing one of the most catastrophic damages to a target cell. Target organisms have evolved a regulated response against PFTs damage including cell membrane repair. This ability of cells strongly depends on the toxin concentration and the properties of the pores. It has been hypothesized that there is an inverse correlation between the size of the pores and the time required to repair the membrane, which has been for long a non-intuitive concept and far to be completely understood. Moreover, there is a lack of information about how cells react to the injury triggered by eukaryotic PFTs. Here, we investigated some molecular events related with eukaryotic cells response against the membrane damage caused by sticholysin II (StII), a eukaryotic PFT produced by a sea anemone. We evaluated the change in the cytoplasmic potassium, identified the main MAPK pathways activated after pore-formation by StII, and compared its effect with those from two well-studied bacterial PFTs: aerolysin and listeriolysin O (LLO). Strikingly, we found that membrane recovery upon StII damage takes place in a time scale similar to LLO in spite of the fact that they form pores by far different in size. Furthermore, our data support a common role of the potassium ion, as well as MAPKs in the mechanism that cells use to cope with these toxins injury.
Collapse
Affiliation(s)
- Sheila Cabezas
- Center for Protein Studies, Faculty of Biology, Havana University, Street 25 # 455, CP 10400, Havana, Cuba.
| | - Sylvia Ho
- École Polytechnique Fédérale de Lausanne, Global Health Institution, Faculty of Life Sciences, Station 15, CH 1015 Lausanne, Switzerland.
| | - Uris Ros
- Center for Protein Studies, Faculty of Biology, Havana University, Street 25 # 455, CP 10400, Havana, Cuba; Interfakultäres Institut für Biochemie, Universität Tübingen, Hoppe Seyler Strasse, 4, 72076, Tübingen, Germany.
| | - María E Lanio
- Center for Protein Studies, Faculty of Biology, Havana University, Street 25 # 455, CP 10400, Havana, Cuba.
| | - Carlos Alvarez
- Center for Protein Studies, Faculty of Biology, Havana University, Street 25 # 455, CP 10400, Havana, Cuba.
| | - F Gisou van der Goot
- École Polytechnique Fédérale de Lausanne, Global Health Institution, Faculty of Life Sciences, Station 15, CH 1015 Lausanne, Switzerland.
| |
Collapse
|
28
|
García-Linares S, Rivera-de-Torre E, Palacios-Ortega J, Gavilanes JG, Martínez-del-Pozo Á. The Metamorphic Transformation of a Water-Soluble Monomeric Protein Into an Oligomeric Transmembrane Pore. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2017. [DOI: 10.1016/bs.abl.2017.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Evolution of the Cytolytic Pore-Forming Proteins (Actinoporins) in Sea Anemones. Toxins (Basel) 2016; 8:toxins8120368. [PMID: 27941639 PMCID: PMC5198562 DOI: 10.3390/toxins8120368] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/28/2016] [Accepted: 11/23/2016] [Indexed: 12/27/2022] Open
Abstract
Sea anemones (Cnidaria, Anthozoa, and Actiniaria) use toxic peptides to incapacitate and immobilize prey and to deter potential predators. Their toxin arsenal is complex, targeting a variety of functionally important protein complexes and macromolecules involved in cellular homeostasis. Among these, actinoporins are one of the better characterized toxins; these venom proteins form a pore in cellular membranes containing sphingomyelin. We used a combined bioinformatic and phylogenetic approach to investigate how actinoporins have evolved across three superfamilies of sea anemones (Actinioidea, Metridioidea, and Actinostoloidea). Our analysis identified 90 candidate actinoporins across 20 species. We also found clusters of six actinoporin-like genes in five species of sea anemone (Nematostella vectensis, Stomphia coccinea, Epiactis japonica, Heteractis crispa, and Diadumene leucolena); these actinoporin-like sequences resembled actinoporins but have a higher sequence similarity with toxins from fungi, cone snails, and Hydra. Comparative analysis of the candidate actinoporins highlighted variable and conserved regions within actinoporins that may pertain to functional variation. Although multiple residues are involved in initiating sphingomyelin recognition and membrane binding, there is a high rate of replacement for a specific tryptophan with leucine (W112L) and other hydrophobic residues. Residues thought to be involved with oligomerization were variable, while those forming the phosphocholine (POC) binding site and the N-terminal region involved with cell membrane penetration were highly conserved.
Collapse
|
30
|
Yi Z, Wu FD, Zhou Y, Gao CJ. Novel nanofiltration membranes with tunable permselectivity by polymer mediated phase separation in polyamide selective layer. Chin J Chem Eng 2016. [DOI: 10.1016/j.cjche.2016.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Lv X, Zhang J, Xu R, Dong Y, Sun A, Shen Y, Wei D. Gigantoxin-4-4D5 scFv is a novel recombinant immunotoxin with specific toxicity against HER2/neu-positive ovarian carcinoma cells. Appl Microbiol Biotechnol 2016; 100:6403-6413. [DOI: 10.1007/s00253-016-7487-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/14/2016] [Accepted: 03/17/2016] [Indexed: 12/19/2022]
|
32
|
Wacklin HP, Bremec BB, Moulin M, Rojko N, Haertlein M, Forsyth T, Anderluh G, Norton RS. Neutron reflection study of the interaction of the eukaryotic pore-forming actinoporin equinatoxin II with lipid membranes reveals intermediate states in pore formation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:640-52. [DOI: 10.1016/j.bbamem.2015.12.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 11/02/2015] [Accepted: 12/15/2015] [Indexed: 01/07/2023]
|
33
|
Pore-forming toxins: Properties, diversity, and uses as tools to image sphingomyelin and ceramide phosphoethanolamine. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:576-92. [PMID: 26498396 DOI: 10.1016/j.bbamem.2015.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 09/30/2015] [Accepted: 10/14/2015] [Indexed: 12/27/2022]
Abstract
Pore-forming toxins (PFTs) represent a unique class of highly specific lipid-binding proteins. The cytotoxicity of these compounds has been overcome through crystallographic structure and mutation studies, facilitating the development of non-toxic lipid probes. As a consequence, non-toxic PFTs have been utilized as highly specific probes to visualize the diversity and dynamics of lipid nanostructures in living and fixed cells. This review is focused on the application of PFTs and their non-toxic analogs as tools to visualize sphingomyelin and ceramide phosphoethanolamine, two major phosphosphingolipids in mammalian and insect cells, respectively. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.
Collapse
|
34
|
Pedrera L, Gomide AB, Sánchez RE, Ros U, Wilke N, Pazos F, Lanio ME, Itri R, Fanani ML, Alvarez C. The Presence of Sterols Favors Sticholysin I-Membrane Association and Pore Formation Regardless of Their Ability to Form Laterally Segregated Domains. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:9911-9923. [PMID: 26273899 DOI: 10.1021/acs.langmuir.5b01687] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
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. As for actinoporins, it has been proposed that the presence of cholesterol (Chol) and the coexistence of lipid phases increase binding to the target membrane and pore-forming ability. However, little is known about the role of membrane structure and dynamics (phase state, fluidity, and the presence of lipid domains) on the activity of actinoporins or which regions of the membrane are the most favorable for protein insertion, oligomerization, and eventually pore formation. To gain insight into the role of membrane properties on the functional activity of St I, we studied its binding to monolayers and vesicles of phosphatidylcholine (PC), sphingomyelin (SM), and sterols inducing (ergosterol -Erg and cholesterol -Chol) or not (cholestenone - Cln) membrane phase segregation in liquid ordered (Lo) and liquid disordered (Ld) domains. This study revealed that St I binds and permeabilizes with higher efficiency sterol-containing membranes independently of their ability to form domains. We discuss the results in terms of the relevance of different membrane properties for the actinoporins mechanism of action, namely, molecular heterogeneity, specially potentiated in membranes with sterols inducers of phase separation (Chol or Erg) or Cln, a sterol noninducer of phase separation but with a high propensity to induce nonlamellar phase. The role of the Ld phase is pointed out as the most suitable platform for pore formation. In this regard, such regions in Chol-containing membranes seem to be the most favored due to its increased fluidity; this property promotes toxin insertion, diffusion, and oligomerization leading to pore formation.
Collapse
Affiliation(s)
- Lohans Pedrera
- Centro de Estudio de Proteínas (CEP), Facultad de Biología, Universidad de la Habana , CP 10400, La Habana, Cuba
| | - Andreza B Gomide
- Departamento de Física Aplicada, Instituto de Física, Universidade de São Paulo , 05508090, São Paulo, Brasil
- Centro Universitário Padre Anchieta, Jundiaí , 13207270, São Paulo, Brasil
| | - Rafael E Sánchez
- Centro de Estudio de Proteínas (CEP), Facultad de Biología, Universidad de la Habana , CP 10400, La Habana, Cuba
| | - Uris Ros
- Centro de Estudio de Proteínas (CEP), Facultad de Biología, Universidad de la Habana , CP 10400, La Habana, Cuba
| | - Natalia Wilke
- Departamento de Química Biológica, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Facultad de Ciencias Químicas-CONICET, Universidad Nacional de Córdoba , X5000HUA Córdoba, Argentina
| | - Fabiola Pazos
- Centro de Estudio de Proteínas (CEP), Facultad de Biología, Universidad de la Habana , CP 10400, La Habana, Cuba
| | - María E Lanio
- Centro de Estudio de Proteínas (CEP), Facultad de Biología, Universidad de la Habana , CP 10400, La Habana, Cuba
| | - Rosangela Itri
- Departamento de Física Aplicada, Instituto de Física, Universidade de São Paulo , 05508090, São Paulo, Brasil
| | - María Laura Fanani
- Departamento de Química Biológica, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Facultad de Ciencias Químicas-CONICET, Universidad Nacional de Córdoba , X5000HUA Córdoba, Argentina
| | - Carlos Alvarez
- Centro de Estudio de Proteínas (CEP), Facultad de Biología, Universidad de la Habana , CP 10400, La Habana, Cuba
| |
Collapse
|
35
|
Rojko N, Dalla Serra M, Maček P, Anderluh G. Pore formation by actinoporins, cytolysins from sea anemones. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1858:446-56. [PMID: 26351738 DOI: 10.1016/j.bbamem.2015.09.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/31/2015] [Accepted: 09/02/2015] [Indexed: 11/30/2022]
Abstract
Actinoporins (APs) from sea anemones are ~20 kDa pore forming toxins with a β-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 β-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.
Collapse
Affiliation(s)
- Nejc Rojko
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Mauro Dalla Serra
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche & Fondazione Bruno Kessler, via alla Cascata 56/C, 38123 Trento, Italy
| | - Peter Maček
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Gregor Anderluh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| |
Collapse
|
36
|
Ros U, García-Sáez AJ. More Than a Pore: The Interplay of Pore-Forming Proteins and Lipid Membranes. J Membr Biol 2015; 248:545-61. [PMID: 26087906 DOI: 10.1007/s00232-015-9820-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/12/2015] [Indexed: 01/09/2023]
Abstract
Pore-forming proteins (PFPs) punch holes in their target cell membrane to alter their permeability. Permeabilization of lipid membranes by PFPs has received special attention to study the basic molecular mechanisms of protein insertion into membranes and the development of biotechnological tools. PFPs act through a general multi-step mechanism that involves (i) membrane partitioning, (ii) insertion into the hydrophobic core of the bilayer, (iii) oligomerization, and (iv) pore formation. Interestingly, PFPs and membranes show a dynamic interplay. As PFPs are usually produced as soluble proteins, they require a large conformational change for membrane insertion. Moreover, membrane structure is modified upon PFPs insertion. In this context, the toroidal pore model has been proposed to describe a pore architecture in which not only protein molecules but also lipids are directly involved in the structure. Here, we discuss how PFPs and lipids cooperate and remodel each other to achieve pore formation, and explore new evidences of protein-lipid pore structures.
Collapse
Affiliation(s)
- Uris Ros
- Center for Protein Studies, Faculty of Biology, Calle 25 # 455, Plaza de la Revolución, Havana, Cuba
| | | |
Collapse
|
37
|
Cajnko MM, Marušić M, Kisovec M, Rojko N, Benčina M, Caserman S, Anderluh G. Listeriolysin O Affects the Permeability of Caco-2 Monolayer in a Pore-Dependent and Ca2+-Independent Manner. PLoS One 2015; 10:e0130471. [PMID: 26087154 PMCID: PMC4472510 DOI: 10.1371/journal.pone.0130471] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 05/20/2015] [Indexed: 12/20/2022] Open
Abstract
Listeria monocytogenes is a food and soil-borne pathogen that secretes a pore-forming toxin listeriolysin O (LLO) as its major virulence factor. We tested the effects of LLO on an intestinal epithelial cell line Caco-2 and compared them to an unrelated pore-forming toxin equinatoxin II (EqtII). Results showed that apical application of both toxins causes a significant drop in transepithelial electrical resistance (TEER), with higher LLO concentrations or prolonged exposure time needed to achieve the same magnitude of response than with EqtII. The drop in TEER was due to pore formation and coincided with rearrangement of claudin-1 within tight junctions and associated actin cytoskeleton; however, no significant increase in permeability to fluorescein or 3 kDa FITC-dextran was observed. Influx of calcium after pore formation affected the magnitude of the drop in TEER. Both toxins exhibit similar effects on epithelium morphology and physiology. Importantly, LLO action upon the membrane is much slower and results in compromised epithelium on a longer time scale at lower concentrations than EqtII. This could favor listerial invasion in hosts resistant to E-cadherin related infection.
Collapse
Affiliation(s)
- Miša Mojca Cajnko
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Maja Marušić
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Matic Kisovec
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Nejc Rojko
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Mojca Benčina
- Laboratory for Biotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Simon Caserman
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
38
|
Jouiaei M, Yanagihara AA, Madio B, Nevalainen TJ, Alewood PF, Fry BG. Ancient Venom Systems: A Review on Cnidaria Toxins. Toxins (Basel) 2015; 7:2251-71. [PMID: 26094698 PMCID: PMC4488701 DOI: 10.3390/toxins7062251] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 01/22/2023] Open
Abstract
Cnidarians are the oldest extant lineage of venomous animals. Despite their simple anatomy, they are capable of subduing or repelling prey and predator species that are far more complex and recently evolved. Utilizing specialized penetrating nematocysts, cnidarians inject the nematocyst content or "venom" that initiates toxic and immunological reactions in the envenomated organism. These venoms contain enzymes, potent pore forming toxins, and neurotoxins. Enzymes include lipolytic and proteolytic proteins that catabolize prey tissues. Cnidarian pore forming toxins self-assemble to form robust membrane pores that can cause cell death via osmotic lysis. Neurotoxins exhibit rapid ion channel specific activities. In addition, certain cnidarian venoms contain or induce the release of host vasodilatory biogenic amines such as serotonin, histamine, bunodosine and caissarone accelerating the pathogenic effects of other venom enzymes and porins. The cnidarian attacking/defending mechanism is fast and efficient, and massive envenomation of humans may result in death, in some cases within a few minutes to an hour after sting. The complexity of venom components represents a unique therapeutic challenge and probably reflects the ancient evolutionary history of the cnidarian venom system. Thus, they are invaluable as a therapeutic target for sting treatment or as lead compounds for drug design.
Collapse
Affiliation(s)
- Mahdokht Jouiaei
- Venom Evolution Lab, School of Biological Sciences, the University of Queensland, St. Lucia 4072, QLD, Australia.
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia 4072, QLD, Australia.
| | - Angel A Yanagihara
- Pacific Cnidaria Research Lab, Department of Tropical Medicine, University of Hawaii, Honolulu, HI 96822, USA.
| | - Bruno Madio
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia 4072, QLD, Australia.
| | - Timo J Nevalainen
- Department of Pathology, University of Turku, Turku FIN-20520, Finland.
| | - Paul F Alewood
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia 4072, QLD, Australia.
| | - Bryan G Fry
- Venom Evolution Lab, School of Biological Sciences, the University of Queensland, St. Lucia 4072, QLD, Australia.
- Institute for Molecular Bioscience, the University of Queensland, St. Lucia 4072, QLD, Australia.
| |
Collapse
|
39
|
Subburaj Y, Ros U, Hermann E, Tong R, García-Sáez AJ. Toxicity of an α-pore-forming toxin depends on the assembly mechanism on the target membrane as revealed by single molecule imaging. J Biol Chem 2014; 290:4856-4865. [PMID: 25525270 DOI: 10.1074/jbc.m114.600676] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
α-Pore-forming toxins (α-PFTs) are ubiquitous defense tools that kill cells by opening pores in the target cell membrane. Despite their relevance in host/pathogen interactions, very little is known about the pore stoichiometry and assembly pathway leading to membrane permeabilization. Equinatoxin II (EqtII) is a model α-PFT from sea anemone that oligomerizes and forms pores in sphingomyelin-containing membranes. Here, we determined the spatiotemporal organization of EqtII in living cells by single molecule imaging. Surprisingly, we found that on the cell surface EqtII did not organize into a unique oligomeric form. Instead, it existed as a mixture of oligomeric species mostly including monomers, dimers, tetramers, and hexamers. Mathematical modeling based on our data supported a new model in which toxin clustering happened in seconds and proceeded via condensation of EqtII dimer units formed upon monomer association. Furthermore, altering the pathway of EqtII assembly strongly affected its toxic activity, which highlights the relevance of the assembly mechanism on toxicity.
Collapse
Affiliation(s)
- Yamunadevi Subburaj
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569 Stuttgart, Germany; German Cancer Research Center, Bioquant, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Uris Ros
- Center for Protein Studies, Faculty of Biology, Calle 25 #455, Plaza de la Revolución, La Habana, Cuba
| | - Eduard Hermann
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569 Stuttgart, Germany; German Cancer Research Center, Bioquant, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany,; Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany
| | - Rudi Tong
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569 Stuttgart, Germany
| | - Ana J García-Sáez
- Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, 70569 Stuttgart, Germany; German Cancer Research Center, Bioquant, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany,; Interfaculty Institute of Biochemistry, University of Tübingen, Hoppe-Seyler-Str. 4, 72076 Tübingen, Germany.
| |
Collapse
|
40
|
Antonini V, Pérez-Barzaga V, Bampi S, Pentón D, Martínez D, Serra MD, Tejuca M. Functional characterization of sticholysin I and W111C mutant reveals the sequence of the actinoporin's pore assembly. PLoS One 2014; 9:e110824. [PMID: 25350457 PMCID: PMC4211696 DOI: 10.1371/journal.pone.0110824] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022] Open
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.
Collapse
Affiliation(s)
- Valeria Antonini
- National Research Council of Italy - Institute of Biophysics and Bruno Kessler Foundation, Trento, Italy
| | - Victor Pérez-Barzaga
- Center for Protein Studies, Faculty of Biology, University of Havana, Vedado, Ciudad de La Habana, Cuba
| | - Silvia Bampi
- National Research Council of Italy - Institute of Biophysics and Bruno Kessler Foundation, Trento, Italy
| | - David Pentón
- Center for Protein Studies, Faculty of Biology, University of Havana, Vedado, Ciudad de La Habana, Cuba
| | - Diana Martínez
- Center for Protein Studies, Faculty of Biology, University of Havana, Vedado, Ciudad de La Habana, Cuba
| | - Mauro Dalla Serra
- National Research Council of Italy - Institute of Biophysics and Bruno Kessler Foundation, Trento, Italy
- * E-mail: (MDS); (MT)
| | - Mayra Tejuca
- Center for Protein Studies, Faculty of Biology, University of Havana, Vedado, Ciudad de La Habana, Cuba
- * E-mail: (MDS); (MT)
| |
Collapse
|
41
|
Baker MAB, Rojko N, Cronin B, Anderluh G, Wallace MI. Photobleaching Reveals Heterogeneous Stoichiometry for Equinatoxin II Oligomers. Chembiochem 2014; 15:2139-45. [DOI: 10.1002/cbic.201300799] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Indexed: 01/19/2023]
|
42
|
del Monte-Martínez A, González-Bacerio J, Romero L, Aragón C, Martínez D, Chávez MDLÁ, Álvarez C, Lanio ME, Guisán JM, Díaz J. Improved purification and enzymatic properties of a mixture of Sticholysin I and II: Isotoxins with hemolytic and phospholipase A2 activities from the sea anemone Stichodactyla helianthus. Protein Expr Purif 2014; 95:57-66. [DOI: 10.1016/j.pep.2013.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 11/27/2013] [Accepted: 11/29/2013] [Indexed: 11/28/2022]
|
43
|
Shatursky OY, Romanenko OV, Himmelreich NH. Long open amphotericin channels revealed in cholesterol-containing phospholipid membranes are blocked by thiazole derivative. J Membr Biol 2014; 247:211-29. [PMID: 24402241 DOI: 10.1007/s00232-013-9626-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/26/2013] [Indexed: 10/25/2022]
Abstract
The action of antifungal drug, amphotericin B (AmB), on solvent-containing planar lipid bilayers made of sterols (cholesterol, ergosterol) and synthetic C14-C18 tail phospholipids (PCs) or egg PC has been investigated in a voltage-clamp mode. Within the range of PCs tested, a similar increase was achieved in the lifetime of one-sided AmB channels in cholesterol- and ergosterol-containing membranes with the C16 tail PC, DPhPC at sterol/DPhPC molar ratio ≤1. The AmB channel lifetimes decreased only at sterol/DPhPC molar ratio >1 that occurred with sterol/PC molar ratio of target cell membranes at a pathological state. These data obtained on bilayer membranes two times thicker than one-sided AmB channel length are consistent with the accepted AmB pore-forming mechanism, which is associated with membrane thinning around AmB-sterol complex in the lipid rafts. Our results show that AmB can create cytotoxic (long open) channels in cholesterol membrane with C14-C16 tail PCs and nontoxic (short open) channels with C17-C18 tail PCs as the lifetime of one-sided AmB channel depends on ~2-5 Å difference in the thickness of sterol-containing C16 and C18 tail PC membranes. The reduction in toxic AmB channels efficacy can be required at the drug administration because C16 tails in native membrane PCs occur almost as often as C18 tails. The comparative analysis of AmB channel blocking by tetraethylammonium chloride, tetramethylammonium chloride and thiazole derivative of vitamin B1, 3-decyloxycarbonylmethyl-4-methyl-5-(2-hydroxyethyl) thiazole chloride (DMHT), has proved that DMHT is a comparable substitute for both tetraalkylammonia that exhibits a much higher affinity.
Collapse
Affiliation(s)
- Oleg Ya Shatursky
- Department of Neurochemistry, Palladin Institute of Biochemistry, Leontovich Str., 9, Kiev, 01601, Ukraine,
| | | | | |
Collapse
|
44
|
Celedón G, González G, Gulppi F, Pazos F, Lanio ME, Alvarez C, Calderón C, Montecinos R, Lissi E. Effect of human serum albumin upon the permeabilizing activity of sticholysin II, a pore forming toxin from Stichodactyla heliantus. Protein J 2013; 32:593-600. [PMID: 24197505 DOI: 10.1007/s10930-013-9521-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
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.
Collapse
Affiliation(s)
- Gloria Celedón
- Departamento de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Peptide-induced membrane leakage by lysine derivatives of gramicidin A in liposomes, planar bilayers, and erythrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2428-35. [DOI: 10.1016/j.bbamem.2013.06.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 11/21/2022]
|
46
|
Ros U, Souto ALCF, de Oliveira FJ, Crusca E, Pazos F, Cilli EM, Lanio ME, Schreier S, Alvarez C. Functional and topological studies with Trp-containing analogs of the peptide StII1-30derived from the N-terminus of the pore forming toxin sticholysin II: contribution to understand its orientation in membrane. Biopolymers 2013; 100:337-46. [DOI: 10.1002/bip.22211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/14/2012] [Accepted: 01/14/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Uris Ros
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| | - Ana Lucia C. F. Souto
- Department of Biochemistry; Institute of Chemistry; University of São Paulo (USP); São Paulo; Brazil
| | - Felipe J. de Oliveira
- Department of Biochemistry; Institute of Chemistry; University of São Paulo (USP); São Paulo; Brazil
| | - Edson Crusca
- Department of Biochemistry and Chemical Technology; Institute of Chemistry; São Paulo State University (UNESP); Araraquara; São Paulo; Brazil
| | - Fabiola Pazos
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| | - Eduardo M. Cilli
- Department of Biochemistry and Chemical Technology; Institute of Chemistry; São Paulo State University (UNESP); Araraquara; São Paulo; Brazil
| | - Maria E. Lanio
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| | - Shirley Schreier
- Department of Biochemistry; Institute of Chemistry; University of São Paulo (USP); São Paulo; Brazil
| | - Carlos Alvarez
- Center for Protein Studies; Biology Faculty; University of Havana (UH); Havana; Cuba
| |
Collapse
|
47
|
Rojko N, Kristan KČ, Viero G, Žerovnik E, Maček P, Dalla Serra M, Anderluh G. Membrane damage by an α-helical pore-forming protein, Equinatoxin II, proceeds through a succession of ordered steps. J Biol Chem 2013; 288:23704-15. [PMID: 23803608 DOI: 10.1074/jbc.m113.481572] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
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 β-pore-forming toxins that often form β-barrel pores via a stable prepore complex.
Collapse
Affiliation(s)
- Nejc Rojko
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia
| | | | | | | | | | | | | |
Collapse
|
48
|
Loew M, Forsythe JC, McCarley RL. Lipid nature and their influence on opening of redox-active liposomes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:6615-23. [PMID: 23698020 PMCID: PMC3778659 DOI: 10.1021/la304340e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The pathway for content release from reduction-sensitive liposomes based on a quinone-dioleoylphosphatidylethanolamine lipid conjugate (Q-DOPE) is outlined using results from fluorescent dye content release assays as well as single- and multiple-angle light scattering. Experimental observations are consistent with a shape/size change of the reduced liposomes prior to their aggregation, with subsequent near-quantitative content release achieved only when the lipid membrane experiences conditions favorable to a lamellar to an inverted hexagonal phase transition. Addition of poly(ethyleneglycol)-modified DOPE (PEG-DOPE) to the Q-DOPE liposomal formulation results in stabilization of the lipid bilayer, whereas incorporation of DOPE yields faster content release. At high DOPE concentrations, DOPE/PEG-DOPE/Q-DOPE liposomes exhibit larger content release, indicating a change in pathway for content release. The outcomes here provide a better understanding of the underlying principles of triggered liposomal content release and the potential utility of specific lipid properties for the rational design of drug delivery systems based on the novel Q-DOPE lipid.
Collapse
Affiliation(s)
| | | | - Robin L. McCarley
- CORRESPONDING AUTHOR: Telephone: (225) 578-3239. Facsimile: (225) 578-3458.
| |
Collapse
|
49
|
Monroy-Estrada HI, Chirino YI, Soria-Mercado IE, Sánchez-Rodríguez J. Toxins from the Caribbean sea anemone Bunodeopsis globulifera increase cisplatin-induced cytotoxicity of lung adenocarcinoma cells. J Venom Anim Toxins Incl Trop Dis 2013; 19:12. [PMID: 24499018 PMCID: PMC3710156 DOI: 10.1186/1678-9199-19-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 12/05/2012] [Indexed: 01/06/2023] Open
Abstract
Background Lung cancer causes 1.4 million deaths worldwide while non-small-cell lung cancer (NSCLC) represents 80-85% of the cases. Cisplatin is a standard chemotherapy against this type of cancer; however, tumor cell resistance to this drug limits its efficacy. Sea anemones produce compounds with pharmacological activities that may be useful for augmenting cisplatin efficacy. This study aimed to evaluate the pharmacological activities of crude venom (CV) from the sea anemone Bunodeopsis globulifera and four derived fractions (F1, F2, F3 and F4) to test their increase efficiency cisplatin cytotoxicity in human lung adenocarcinoma cells. Results Pre-exposure to CV, F1 and F2 fractions increases cisplatin cytotoxicity in human lung adenocarcinoma cells under specific conditions. Exposure to CV at 50 μgmL-1 induced a reduction of approximately 50% in cell viability, while a similar cytotoxic effect was observed when cell culture was exposed to F1 at 25 μgmL -1 or F2 at 50 μgmL-1. The cell culture exposure to F1 (10 μgmL-1) fraction combined with cisplatine (25 μM) provoked a decrease in MTT reduction until 65.57% while F2 (25 μgmL-1) fraction combined with cisplatin (10 μM) provoked a decrease in MTT reduction of 72.55%. Conclusions The F1 fraction had the greatest effect on the lung adenocarcinoma cell line compared with CV and F2. The combination of antineoplastic drugs and sea anemone toxins might allow a reduction of chemotherapeutic doses and thus mitigate side effects.
Collapse
Affiliation(s)
| | | | | | - Judith Sánchez-Rodríguez
- Unidad Académica de Sistemas Arrecifales, Puerto Morelos, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México, Puerto Morelos, Quintana Roo State, Mexico.
| |
Collapse
|
50
|
Characteristics of hemolytic activity induced by skin secretions of the frog Kaloula pulchra hainana. J Venom Anim Toxins Incl Trop Dis 2013; 19:9. [PMID: 24499077 PMCID: PMC3710140 DOI: 10.1186/1678-9199-19-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 04/11/2013] [Indexed: 11/29/2022] Open
Abstract
Background The hemolytic activity of skin secretions obtained by stimulating the frog Kaloula pulchra hainana with diethyl ether was tested using human, cattle, rabbit, and chicken erythrocytes. The skin secretions had a significant concentration-dependent hemolytic effect on erythrocytes. The hemolytic activity of the skin secretions was studied in the presence of osmotic protectants (polyethylene glycols and carbohydrates), cations (Mg2+, Ca2+, Ba2+, Cu2+, and K+), or antioxidants (ascorbic acid, reduced glutathione, and cysteine). Results Depending on their molecular mass, osmotic protectants effectively inhibited hemolysis. The inhibition of skin hemolysis was observed after treatment with polyethylene glycols (1000, 3400, and 6000 Da). Among divalent cations, only 1 mM Cu2+ markedly inhibited hemolytic activity. Antioxidant compounds slightly reduced the hemolytic activity. Conclusions The results suggested that skin secretions of K. pulchra hainana induce a pore-forming mechanism to form pores with a diameter of 1.36-2.0 nm rather than causing oxidative damage to the erythrocyte membrane.
Collapse
|