1
|
Tomishige N, Takahashi K, Pollet B, Richert L, Mély Y, Kobayashi T. Using lipid binding proteins and advanced microscopy to study lipid domains. Methods Enzymol 2024; 700:217-234. [PMID: 38971601 DOI: 10.1016/bs.mie.2024.03.026] [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] [Indexed: 07/08/2024]
Abstract
Sphingomyelin is postulated to form clusters with glycosphingolipids, cholesterol and other sphingomyelin molecules in biomembranes through hydrophobic interaction and hydrogen bonds. These clusters form submicron size lipid domains. Proteins that selectively binds sphingomyelin and/or cholesterol are useful to visualize the lipid domains. Due to their small size, visualization of lipid domains requires advanced microscopy techniques in addition to lipid binding proteins. This Chapter describes the method to characterize plasma membrane sphingomyelin-rich and cholesterol-rich lipid domains by quantitative microscopy. This Chapter also compares different permeabilization methods to visualize intracellular lipid domains.
Collapse
Affiliation(s)
| | | | | | | | - Yves Mély
- Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
2
|
Liu MH, Liu ZK, Liu F. An anti-tumor protein PFAP specifically interacts with cholesterol-enriched membrane domains of A549 cells and induces paraptosis and endoplasmic reticulum stress. Int J Biol Macromol 2024; 264:130690. [PMID: 38458297 DOI: 10.1016/j.ijbiomac.2024.130690] [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: 01/09/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Nowadays, non-small cell lung cancer (NSCLC) is still one of the most life-threatening diseases in the world. In previous studies, a fungal protein PFAP with anti-NSCLC properties was isolated and identified from Pleurotus ferulae lanzi. In this study, the amino acid sequence of PFAP was analyzed and found to be highly homologous to the aegerolysin family. PFAP, like other members of the aegerolysin family, specifically recognizes lipid raft domains rich in cholesterol and sphingomyelin, which is probably its specific anti-tumor mechanism. Previous studies have shown that PFAP can induce AMPK-mediated autophagy and G1-phase cell cycle arrest in A549 lung cancer cells. This study further revealed that PFAP can also induce paraptosis and endoplasmic reticulum stress (ERS) in A549 cells in vitro by targeting AMPK. PFAP induces multi-pathway death of A549 cells, and thus demonstrates its potential value for developing new drugs for NSCLC.
Collapse
Affiliation(s)
- Meng-Han Liu
- Department of Microbiology, The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Zhao-Kun Liu
- Research Institute of Public Health, School of Medicine, Nankai University, Tianjin 300071, China.
| | - Fang Liu
- Department of Microbiology, The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
3
|
Yan B, Ma A. PriA is involved in Pleurotus ostreatus development and defense against Pseudomonas tolaasii. Antonie Van Leeuwenhoek 2023; 117:1. [PMID: 38095768 DOI: 10.1007/s10482-023-01900-6] [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: 08/25/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023]
Abstract
Pleurotus ostreatus is a crucial commercial mushroom widely cultivated for diverse uses. Scientists have worked on breeding disease-resistant and high-yielding varieties to secure food supply. Studies on the molecular genetic mechanism of growth and development can provide valuable information to facilitate crop breeding programs by genetic engineering. Aegerolysins are pore-forming proteins widely distributed in both prokaryotes and eukaryotes, which are reported to have haemolytic activity and be involved in the early stages of fructification. The present study aimed to explore biological function of a differential expressed aegerolysin gene PriA in P. ostreatus. The expression level of PriA gene was higher in primordium and fruiting body than that in mycelium. The PriA expression in overexpression (OE) and RNAi interference (RNAi) strains was detected by qRT-PCR. The RNAi strains grew at slightly slower rates and advanced producing yellow pigments than the wild type, while OE strains showed no prominent phenotypic characteristics. Furthermore, Pseudomonas tolaasii infection assays showed that the PriA OE strains could enhance mycelia and caps resistance to P. tolaasii. These data demonstrate PriA from P. ostreatus play an essential role in mycelial development and increase antagonism against P. tolaasii. Our study provides some reference information on interactions between edible fungi and pathogenic bacteria and offers a new resistance-conferring gene for breeding.
Collapse
Affiliation(s)
- Biyun Yan
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Aimin Ma
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
4
|
Yilmaz N, Panevska A, Tomishige N, Richert L, Mély Y, Sepčić K, Greimel P, Kobayashi T. Assembly dynamics and structure of an aegerolysin, ostreolysin A6. J Biol Chem 2023; 299:104940. [PMID: 37343702 PMCID: PMC10366546 DOI: 10.1016/j.jbc.2023.104940] [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: 10/21/2022] [Revised: 05/08/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
Ostreolysin A6 (OlyA6) is an oyster mushroom-derived membrane-binding protein that, upon recruitment of its partner protein, pleurotolysin B, forms a cytolytic membrane pore complex. OlyA6 itself is not cytolytic but has been reported to exhibit pro-apoptotic activities in cell culture. Here we report the formation dynamics and the structure of OlyA6 assembly on a lipid membrane containing an OlyA6 high-affinity receptor, ceramide phosphoethanolamine, and cholesterol. High-speed atomic force microscopy revealed the reorganization of OlyA6 dimers from initial random surface coverage to 2D protein crystals composed of hexameric OlyA6 repeat units. Crystal growth took place predominantly in the longitudinal direction by the association of OlyA6 dimers, forming a hexameric unit cell. Molecular-level examination of the OlyA6 crystal elucidated the arrangement of dimers within the unit cell and the structure of the dimer that recruits pleurotolysin B for pore formation.
Collapse
Affiliation(s)
- Neval Yilmaz
- Lipid Biology Laboratory, RIKEN, 2-1, Wako, Saitama, Japan; NanoLSI, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, Japan.
| | - Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nario Tomishige
- Lipid Biology Laboratory, RIKEN, 2-1, Wako, Saitama, Japan; Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Ludovic Richert
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Yves Mély
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Greimel
- Lipid Biology Laboratory, RIKEN, 2-1, Wako, Saitama, Japan.
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Wako, Saitama, Japan; Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France.
| |
Collapse
|
5
|
Panevska A, Čegovnik N, Fortuna K, Vukovič A, Grundner M, Modic Š, Bajc G, Skočaj M, Mravinec Bohte M, Popošek LL, Žigon P, Razinger J, Veranič P, Resnik N, Sepčić K. A single point mutation expands the applicability of ostreolysin A6 in biomedicine. Sci Rep 2023; 13:2149. [PMID: 36750638 PMCID: PMC9905591 DOI: 10.1038/s41598-023-28949-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
An aegerolysin protein ostreolysin A6 (OlyA6) binds to cholesterol-complexed sphingomyelin and can be used for specific labelling of lipid rafts. In addition, OlyA6 interacts with even higher affinity with ceramide phosphoethanolamine (CPE), a sphingolipid that dominates in invertebrate cell membranes. In the presence of pleurotolysin B, a protein bearing the membrane-attack complex/perforin domain, OlyA6 forms pores in insect midgut cell membranes and acts as a potent bioinsecticide. It has been shown that a point mutation of glutamate 69 to alanine (E69A) allows OlyA6 to bind to cholesterol-free sphingomyelin. Using artificial lipid membranes and mammalian MDCK cells, we show that this mutation significantly enhances the interaction of OlyA6 with sphingomyelin and CPE, and allows recognition of these sphingolipids even in the absence of cholesterol. Our results suggest that OlyA6 mutant E69A could serve as complementary tool to detect and study cholesterol-associated and free sphingomyelin or CPE in membranes. However, the mutation does not improve the membrane-permeabilizing activity after addition of pleurotolysin B, which was confirmed in toxicity tests on insect and mammalian cell lines, and on Colorado potato beetle larvae.
Collapse
Affiliation(s)
- Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Nastja Čegovnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Klavdija Fortuna
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Alen Vukovič
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Maja Grundner
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Špela Modic
- Agricultural Institute of Slovenia, Hacquetova Ulica 17, 1000, Ljubljana, Slovenia
| | - Gregor Bajc
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Martina Mravinec Bohte
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Lara Larisa Popošek
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Primož Žigon
- Agricultural Institute of Slovenia, Hacquetova Ulica 17, 1000, Ljubljana, Slovenia
| | - Jaka Razinger
- Agricultural Institute of Slovenia, Hacquetova Ulica 17, 1000, Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia
| | - Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia.
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia.
| |
Collapse
|
6
|
Mapping trasmembrane distribution of sphingomyelin. Emerg Top Life Sci 2023; 7:31-45. [PMID: 36692108 DOI: 10.1042/etls20220086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 01/25/2023]
Abstract
Our knowledge on the asymmetric distribution of sphingomyelin (SM) in the plasma membrane is largely based on the biochemical analysis of erythrocytes using sphingomyelinase (SMase). However, recent studies showed that the product of SMase, ceramide, disturbs transmembrane lipid distribution. This led to the development of the complimentary histochemical method, which combines electron microscopy and SM-binding proteins. This review discusses the advantages and caveats of published methods of measuring transbilayer distribution of SM. Recent finding of the proteins involved in the transbilayer movement of SM will also be summarized.
Collapse
|
7
|
Critical Sites on Ostreolysin Are Responsible for Interaction with Cytoskeletal Proteins. Biomedicines 2022; 10:biomedicines10102442. [PMID: 36289704 PMCID: PMC9598724 DOI: 10.3390/biomedicines10102442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
We explored the structural features of recombinant ostreolysin A (rOlyA), a protein produced by Pleurotus ostreatus and responsible for binding to α/β-tubulin. We found that rOlyA cell internalization is essential for the induction of adipocyte-associated activity, which is mediated by the interaction of rOlyA and microtubule proteins. We created different point mutations at conserved tryptophan (W) sites in rOlyA and analyzed their biological activity in HIB-1B preadipocytes. We demonstrated that the protein’s cell-internalization ability and the differentiated phenotype induced, such as small lipid-droplet formation and gene expression of mitogenesis activity, were impaired in point-mutated proteins W96A and W28A, where W was converted to alanine (A). We also showed that an rOlyA homologue, OlyA6 complexed with mCherry, cannot bind to β-tubulin and does not induce mitochondrial biosynthesis-associated markers, suggesting that the OlyA6 region masked by mCherry is involved in β-tubulin binding. Protein–protein docking simulations were carried out to investigate the binding mode of rOlyA with β-tubulin. Taken together, we identified functional sites in rOlyA that are essential for its binding to β-tubulin and its adipocyte-associated biological activity.
Collapse
|
8
|
Towards Understanding the Function of Aegerolysins. Toxins (Basel) 2022; 14:toxins14090629. [PMID: 36136567 PMCID: PMC9505663 DOI: 10.3390/toxins14090629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Aegerolysins are remarkable proteins. They are distributed over the tree of life, being relatively widespread in bacteria and fungi, but also present in some insects, plants, protozoa, and viruses. Despite their abundance in cells of certain developmental stages and their presence in secretomes, only a few aegerolysins have been studied in detail. Their function, in particular, is intriguing. Here, we summarize previously published findings on the distribution, molecular interactions, and function of these versatile aegerolysins. They have very diverse protein sequences but a common fold. The machine learning approach of the AlphaFold2 algorithm, which incorporates physical and biological knowledge of protein structures and multisequence alignments, provides us new insights into the aegerolysins and their pore-forming partners, complemented by additional genomic support. We hypothesize that aegerolysins are involved in the mechanisms of competitive exclusion in the niche.
Collapse
|
9
|
Kunduri G, Le SH, Baena V, Vijaykrishna N, Harned A, Nagashima K, Blankenberg D, Yoshihiro I, Narayan K, Bamba T, Acharya U, Acharya JK. Delivery of ceramide phosphoethanolamine lipids to the cleavage furrow through the endocytic pathway is essential for male meiotic cytokinesis. PLoS Biol 2022; 20:e3001599. [PMID: 36170207 PMCID: PMC9550178 DOI: 10.1371/journal.pbio.3001599] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/10/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
Cell division, wherein 1 cell divides into 2 daughter cells, is fundamental to all living organisms. Cytokinesis, the final step in cell division, begins with the formation of an actomyosin contractile ring, positioned midway between the segregated chromosomes. Constriction of the ring with concomitant membrane deposition in a specified spatiotemporal manner generates a cleavage furrow that physically separates the cytoplasm. Unique lipids with specific biophysical properties have been shown to localize to intercellular bridges (also called midbody) connecting the 2 dividing cells; however, their biological roles and delivery mechanisms remain largely unknown. In this study, we show that ceramide phosphoethanolamine (CPE), the structural analog of sphingomyelin, has unique acyl chain anchors in Drosophila spermatocytes and is essential for meiotic cytokinesis. The head group of CPE is also important for spermatogenesis. We find that aberrant central spindle and contractile ring behavior but not mislocalization of phosphatidylinositol phosphates (PIPs) at the plasma membrane is responsible for the male meiotic cytokinesis defect in CPE-deficient animals. Further, we demonstrate the enrichment of CPE in multivesicular bodies marked by Rab7, which in turn localize to cleavage furrow. Volume electron microscopy analysis using correlative light and focused ion beam scanning electron microscopy shows that CPE-enriched Rab7 positive endosomes are juxtaposed on contractile ring material. Correlative light and transmission electron microscopy reveal Rab7 positive endosomes as a multivesicular body-like organelle that releases its intraluminal vesicles in the vicinity of ingressing furrows. Genetic ablation of Rab7 or Rab35 or expression of dominant negative Rab11 results in significant meiotic cytokinesis defects. Further, we show that Rab11 function is required for localization of CPE positive endosomes to the cleavage furrow. Our results imply that endosomal delivery of CPE to ingressing membranes is crucial for meiotic cytokinesis.
Collapse
Affiliation(s)
- Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, Maryland, United States of America
| | - Si-Hung Le
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Valentina Baena
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Nagampalli Vijaykrishna
- Genomic Medicine Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Adam Harned
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Kunio Nagashima
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Daniel Blankenberg
- Genomic Medicine Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Izumi Yoshihiro
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Usha Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, Maryland, United States of America
| | - Jairaj K. Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, Maryland, United States of America
| |
Collapse
|
10
|
Yamaji-Hasegawa A, Murate M, Inaba T, Dohmae N, Sato M, Fujimori F, Sako Y, Greimel P, Kobayashi T. A novel sterol-binding protein reveals heterogeneous cholesterol distribution in neurite outgrowth and in late endosomes/lysosomes. Cell Mol Life Sci 2022; 79:324. [PMID: 35644822 PMCID: PMC11072113 DOI: 10.1007/s00018-022-04339-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
Abstract
We identified a mushroom-derived protein, maistero-2 that specifically binds 3-hydroxy sterol including cholesterol (Chol). Maistero-2 bound lipid mixture in Chol-dependent manner with a binding threshold of around 30%. Changing lipid composition did not significantly affect the threshold concentration. EGFP-maistero-2 labeled cell surface and intracellular organelle Chol with higher sensitivity than that of well-established Chol probe, D4 fragment of perfringolysin O. EGFP-maistero-2 revealed increase of cell surface Chol during neurite outgrowth and heterogeneous Chol distribution between CD63-positive and LAMP1-positive late endosomes/lysosomes. The absence of strictly conserved Thr-Leu pair present in Chol-dependent cytolysins suggests a distinct Chol-binding mechanism for maistero-2.
Collapse
Affiliation(s)
| | - Motohide Murate
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- UMR 7021, CNRS, Université de Strasbourg, 67401, Illkirch, France
| | - Takehiko Inaba
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN CSRS, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Masayuki Sato
- Yukiguni Maitake Co, Ltd. Yokawa 89, Minamiuonuma, Niigata, 949-6695, Japan
| | - Fumihiro Fujimori
- Laboratory of Biological Science and Technology, Tokyo Kasei University, 1-18-1 Kaga, Itabashi, Tokyo, 173-8062, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Peter Greimel
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- Cellular Informatics Laboratory, RIKEN CPR, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan.
- UMR 7021, CNRS, Université de Strasbourg, 67401, Illkirch, France.
| |
Collapse
|
11
|
Panevska A, Glavan G, Jemec Kokalj A, Kukuljan V, Trobec T, Žužek MC, Vrecl M, Drobne D, Frangež R, Sepčić K. Effects of Bioinsecticidal Aegerolysin-Based Cytolytic Complexes on Non-Target Organisms. Toxins (Basel) 2021; 13:457. [PMID: 34208927 PMCID: PMC8310125 DOI: 10.3390/toxins13070457] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/05/2023] Open
Abstract
Aegerolysin proteins ostreolysin A6 (OlyA6), pleurotolysin A2 (PlyA2) and erylysin A (EryA) produced by the mushroom genus Pleurotus bind strongly to an invertebrate-specific membrane sphingolipid, and together with a protein partner pleurotolysin B (PlyB), form transmembrane pore complexes. This pore formation is the basis for the selective insecticidal activity of aegerolysin/PlyB complexes against two economically important coleopteran pests: the Colorado potato beetle and the western corn rootworm. In this study, we evaluated the toxicities of these aegerolysin/PlyB complexes using feeding tests with two ecologically important non-target arthropod species: the woodlouse and the honey bee. The mammalian toxicity of the EryA/PlyB complex was also evaluated after intravenous administration to mice. None of the aegerolysin/PlyB complexes were toxic against woodlice, but OlyA6/PlyB and PlyA2/PlyB were toxic to honeybees, with 48 h mean lethal concentrations (LC50) of 0.22 and 0.39 mg/mL, respectively, in their food. EryA/PlyB was also tested intravenously in mice up to 3 mg/kg body mass, without showing toxicity. With no toxicity seen for EryA/PlyB for environmentally beneficial arthropods and mammals at the tested concentrations, these EryA/PlyB complexes are of particular interest for development of new bioinsecticides for control of selected coleopteran pests.
Collapse
Affiliation(s)
- Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (A.P.); (G.G.); (A.J.K.); (D.D.)
| | - Gordana Glavan
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (A.P.); (G.G.); (A.J.K.); (D.D.)
| | - Anita Jemec Kokalj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (A.P.); (G.G.); (A.J.K.); (D.D.)
| | - Veronika Kukuljan
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia;
| | - Tomaž Trobec
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.); (M.V.)
| | - Monika Cecilija Žužek
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.); (M.V.)
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.); (M.V.)
| | - Damjana Drobne
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (A.P.); (G.G.); (A.J.K.); (D.D.)
| | - Robert Frangež
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.); (M.V.)
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (A.P.); (G.G.); (A.J.K.); (D.D.)
| |
Collapse
|
12
|
Milijaš Jotić M, Panevska A, Iacovache I, Kostanjšek R, Mravinec M, Skočaj M, Zuber B, Pavšič A, Razinger J, Modic Š, Trenti F, Guella G, Sepčić K. Dissecting Out the Molecular Mechanism of Insecticidal Activity of Ostreolysin A6/Pleurotolysin B Complexes on Western Corn Rootworm. Toxins (Basel) 2021; 13:toxins13070455. [PMID: 34209983 PMCID: PMC8310357 DOI: 10.3390/toxins13070455] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 02/04/2023] Open
Abstract
Ostreolysin A6 (OlyA6) is a protein produced by the oyster mushroom (Pleurotus ostreatus). It binds to membrane sphingomyelin/cholesterol domains, and together with its protein partner, pleurotolysin B (PlyB), it forms 13-meric transmembrane pore complexes. Further, OlyA6 binds 1000 times more strongly to the insect-specific membrane sphingolipid, ceramide phosphoethanolamine (CPE). In concert with PlyB, OlyA6 has potent and selective insecticidal activity against the western corn rootworm. We analysed the histological alterations of the midgut wall columnar epithelium of western corn rootworm larvae fed with OlyA6/PlyB, which showed vacuolisation of the cell cytoplasm, swelling of the apical cell surface into the gut lumen, and delamination of the basal lamina underlying the epithelium. Additionally, cryo-electron microscopy was used to explore the membrane interactions of the OlyA6/PlyB complex using lipid vesicles composed of artificial lipids containing CPE, and western corn rootworm brush border membrane vesicles. Multimeric transmembrane pores were formed in both vesicle preparations, similar to those described for sphingomyelin/cholesterol membranes. These results strongly suggest that the molecular mechanism of insecticidal action of OlyA6/PlyB arises from specific interactions of OlyA6 with CPE, and the consequent formation of transmembrane pores in the insect midgut.
Collapse
Affiliation(s)
- Matej Milijaš Jotić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
| | - Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
| | - Ioan Iacovache
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (I.I.); (B.Z.)
| | - Rok Kostanjšek
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
| | - Martina Mravinec
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
| | - Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
| | - Benoît Zuber
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (I.I.); (B.Z.)
| | - Ana Pavšič
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
| | - Jaka Razinger
- Agricultural Institute of Slovenia, 1000 Ljubljana, Slovenia; (J.R.); (Š.M.)
| | - Špela Modic
- Agricultural Institute of Slovenia, 1000 Ljubljana, Slovenia; (J.R.); (Š.M.)
| | - Francesco Trenti
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, 38123 Trento, Italy; (F.T.); (G.G.)
| | - Graziano Guella
- Bioorganic Chemistry Laboratory, Department of Physics, University of Trento, 38123 Trento, Italy; (F.T.); (G.G.)
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (M.M.J.); (A.P.); (R.K.); (M.M.); (M.S.); (A.P.)
- Correspondence: ; Tel.: +386-1-320-3419
| |
Collapse
|
13
|
What Can Mushroom Proteins Teach Us about Lipid Rafts? MEMBRANES 2021; 11:membranes11040264. [PMID: 33917311 PMCID: PMC8067419 DOI: 10.3390/membranes11040264] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/25/2022]
Abstract
The lipid raft hypothesis emerged as a need to explain the lateral organization and behavior of lipids in the environment of biological membranes. The idea, that lipids segregate in biological membranes to form liquid-disordered and liquid-ordered states, was faced with a challenge: to show that lipid-ordered domains, enriched in sphingomyelin and cholesterol, actually exist in vivo. A great deal of indirect evidence and the use of lipid-binding probes supported this idea, but there was a lack of tools to demonstrate the existence of such domains in living cells. A whole new toolbox had to be invented to biochemically characterize lipid rafts and to define how they are involved in several cellular functions. A potential solution came from basic biochemical experiments in the late 1970s, showing that some mushroom extracts exert hemolytic activities. These activities were later assigned to aegerolysin-based sphingomyelin/cholesterol-specific cytolytic protein complexes. Recently, six sphingomyelin/cholesterol binding proteins from different mushrooms have been identified and have provided some insight into the nature of sphingomyelin/cholesterol-rich domains in living vertebrate cells. In this review, we dissect the accumulated knowledge and introduce the mushroom lipid raft binding proteins as molecules of choice to study the dynamics and origins of these liquid-ordered domains in mammalian cells.
Collapse
|
14
|
Tomishige N, Murate M, Didier P, Richert L, Mély Y, Kobayashi T. The use of pore-forming toxins to image lipids and lipid domains. Methods Enzymol 2021; 649:503-542. [PMID: 33712198 DOI: 10.1016/bs.mie.2021.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Very few proteins are reported to bind specific lipids. Because of the high selectivity and strong binding to specific lipids, lipid-targeting pore forming toxins (PFTs) have been employed to study the distribution of lipids in cell- and model-membranes. Non-toxic and monomeric PFT-derivatives are especially useful to study living cells. In this chapter we highlight sphingomyelin (SM)-binding PFT, lysenin (Lys), its derivatives, and newly identified SM/cholesterol binding protein, nakanori. We describe the preparation of non-toxic mutant of Lys (NT-Lys) and its application in optical and super resolution microscopy. We also discuss the observation of nanometer scale lipid domains labeled with nakanori and maltose-binding protein (MBP)-Lys in electron microscopy.
Collapse
Affiliation(s)
| | | | - Pascal Didier
- UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | | | - Yves Mély
- UMR 7021 CNRS, Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
15
|
Johnson KA, Radhakrishnan A. The use of anthrolysin O and ostreolysin A to study cholesterol in cell membranes. Methods Enzymol 2021; 649:543-566. [PMID: 33712199 DOI: 10.1016/bs.mie.2021.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cholesterol is a major component of the plasma membranes (PMs) of animal cells, comprising 35-40mol% of total PM lipids. Recent studies using cholesterol-binding bacterial toxins such as domain 4 of Anthrolysin O (ALOD4) and fungal toxins such as Ostreolysin A (OlyA) have revealed new insights into the organization of PM cholesterol. These studies have defined three distinct pools of PM cholesterol-a fixed pool that is essential for membrane integrity, a sphingomyelin (SM)-sequestered pool that can be detected by OlyA, and a third pool that is accessible and can be detected by ALOD4. Accessible cholesterol is available to interact with proteins and transport to the endoplasmic reticulum (ER), and controls many cellular signaling processes including cholesterol homeostasis, Hedgehog signaling, and bacterial and viral infection. Here, we provide detailed descriptions for the use of ALOD4 and OlyA, both of which are soluble and non-lytic proteins, to study cholesterol organization in the PMs of animal cells. Furthermore, we describe two new versions of ALOD4 that we have developed to increase the versatility of this probe in cellular studies. One is a dual His6 and FLAG epitope-tagged version and the other is a fluorescent version where ALOD4 is fused to Neon, a monomeric fluorescent protein. These new forms of ALOD4 together with previously described OlyA provide an expanded collection of tools to sense, visualize, and modulate levels of accessible and SM-sequestered cholesterol on PMs and study the role of these cholesterol pools in diverse membrane signaling events.
Collapse
Affiliation(s)
- Kristen A Johnson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
16
|
Abe M, Kobayashi T. Imaging Sphingomyelin- and Cholesterol-Enriched Domains in the Plasma Membrane Using a Novel Probe and Super-Resolution Microscopy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1310:81-90. [PMID: 33834433 DOI: 10.1007/978-981-33-6064-8_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this chapter, we show the visualization of lipid domains using a specific lipid-binding protein and super-resolution microscopy. Lipid rafts are plasma membrane domains enriched in both sphingolipids and sterols that play key roles in various physiological events. We identified a novel protein that specifically binds to a complex of sphingomyelin (SM) and cholesterol (Chol). The isolated protein, nakanori, labels the SM/Chol complex at the outer leaflet of the plasma membrane in mammalian cells. Structured illumination microscopic images suggested that the influenza virus buds from the edges of the SM/Chol domains in MDCK cells. Furthermore, a photoactivated localization microscopy analysis indicated that the SM/Chol complex forms domains in the outer leaflet, just above the phosphatidylinositol 4,5-bisphosphate domains in the inner leaflet. These observations provide significant insight into the structure and function of lipid rafts.
Collapse
Affiliation(s)
- Mitsuhiro Abe
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama, Japan.
| | - Toshihide Kobayashi
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama, Japan.,UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| |
Collapse
|
17
|
Kobayashi T, Tomishige N, Inaba T, Makino A, Murata M, Yamaji-Hasegawa A, Murate M. Impact of Intrinsic and Extrinsic Factors on Cellular Sphingomyelin Imaging with Specific Reporter Proteins. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2021; 4:25152564211042456. [PMID: 37366372 PMCID: PMC10259817 DOI: 10.1177/25152564211042456] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Sphingomyelin (SM) is a major sphingolipid in mammalian cells. Although SM is enriched in the outer leaflet of the cell plasma membrane, lipids are also observed in the inner leaflet of the plasma membrane and intracellular organelles such as endolysosomes, the Golgi apparatus and nuclei. SM is postulated to form clusters with glycosphingolipids (GSLs), cholesterol (Chol), and other SM molecules through hydrophobic interactions and hydrogen bonding. Thus, different clusters composed of SM, SM/Chol, SM/GSL and SM/GSL/Chol with different stoichiometries may exist in biomembranes. In addition, SM monomers may be located in the glycerophospholipid-rich areas of membranes. Recently developed SM-binding proteins (SBPs) distinguish these different SM assemblies. Here, we summarize the effects of intrinsic factors regulating the lipid-binding specificity of SBPs and extrinsic factors, such as the lipid phase and lipid density, on SM recognition by SBPs. The combination of different SBPs revealed the heterogeneity of SM domains in biomembranes.
Collapse
Affiliation(s)
- Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan
- Cellular Informatics Laboratory, RIKEN
CPR, Wako, Saitama, Japan
- Laboratoire de Bioimagerie et
Pathologies, Faculté de Pharmacie, UMR 7021 CNRS, Université de Strasbourg,
Illkirch, France
| | - Nario Tomishige
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan
- Cellular Informatics Laboratory, RIKEN
CPR, Wako, Saitama, Japan
- Laboratoire de Bioimagerie et
Pathologies, Faculté de Pharmacie, UMR 7021 CNRS, Université de Strasbourg,
Illkirch, France
| | | | - Asami Makino
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan
| | - Michio Murata
- Department of Chemistry, Graduate
School of Science, Osaka University, Toyonaka, Osaka, Japan
- ERATO, Lipid Active Structure Project,
Japan Science and Technology Agency, Graduate School of Science, Osaka University,
Osaka, Japan
| | | | - Motohide Murate
- Lipid Biology Laboratory, RIKEN, Wako, Saitama, Japan
- Cellular Informatics Laboratory, RIKEN
CPR, Wako, Saitama, Japan
- Laboratoire de Bioimagerie et
Pathologies, Faculté de Pharmacie, UMR 7021 CNRS, Université de Strasbourg,
Illkirch, France
| |
Collapse
|
18
|
Kraševec N, Novak M, Barat S, Skočaj M, Sepčić K, Anderluh G. Unconventional Secretion of Nigerolysins A from Aspergillus Species. Microorganisms 2020; 8:E1973. [PMID: 33322461 PMCID: PMC7763983 DOI: 10.3390/microorganisms8121973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 01/05/2023] Open
Abstract
Aegerolysins are small lipid-binding proteins particularly abundant in fungi. Aegerolysins from oyster mushrooms interact with an insect-specific membrane lipid and, together with MACPF proteins produced by the same organism, form pesticidal pore-forming complexes. The specific interaction with the same membrane lipid was recently demonstrated for nigerolysin A2 (NigA2), an aegerolysin from Aspergillus niger. In Aspergillus species, the aegerolysins were frequently found as secreted proteins, indicating their function in fungal defense. Using immunocytochemistry and live-cell imaging we investigated the subcellular localization of the nigerolysins A in A. niger, while their secretion was addressed by secretion prediction and Western blotting. We show that both nigerolysins A are leaderless proteins that reach the cell exterior by an unconventional protein secretion. NigA proteins are evenly distributed in the cytoplasm of fungal hyphae. A detailed bioinformatics analysis of Aspergillus aegerolysins suggests that the same function occurs only in a limited number of aegerolysins. From alignment, analysis of chromosomal loci, orthology, synteny, and phylogeny it follows that the same or a similar function described for pairs of pesticidal proteins of Pleurotus sp. can be expected in species of the subgenus Circumdati, section Nigri, series Nigri, and some other species with adjacent pairs of putative pesticidal proteins.
Collapse
Affiliation(s)
- Nada Kraševec
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (S.B.); (G.A.)
| | - Maruša Novak
- Department of Biology, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (M.N.); (M.S.); (K.S.)
| | - Simona Barat
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (S.B.); (G.A.)
- Department of Biology, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (M.N.); (M.S.); (K.S.)
| | - Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (M.N.); (M.S.); (K.S.)
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (M.N.); (M.S.); (K.S.)
| | - Gregor Anderluh
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (S.B.); (G.A.)
| |
Collapse
|
19
|
Panevska A, Skočaj M, Modic Š, Razinger J, Sepčić K. Aegerolysins from the fungal genus Pleurotus - Bioinsecticidal proteins with multiple potential applications. J Invertebr Pathol 2020; 186:107474. [PMID: 32971130 DOI: 10.1016/j.jip.2020.107474] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022]
Abstract
The aegerolysin proteins ostreolysin A6, pleurotolysin A2 and erylysin A are produced by mushrooms of the genus Pleurotus. These aegerolysins can interact specifically with sphingolipid-enriched membranes. In particular, they strongly bind insect cells and to artificial lipid membranes that contain physiologically relevant concentrations of the main invertebrate-specific sphingolipid, ceramide phosphoethanolamine. Moreover, the aegerolysins permeabilise these membranes when combined with their protein partner pleurotolysin B, which contains a membrane-attack-complex/perforin domain. These aegerolysin/ pleurotolysin B complexes show strong and selective toxicity towards western corn rootworm larvae and adults and Colorado potato beetle larvae. Their insecticidal activities arise through aegerolysin binding to ceramide phosphoethanolamine in the insect midgut. This mode of membrane binding is different from those described for similar aegerolysin-based complexes of bacterial origin (e.g., Cry34Ab1/Cry35Ab1), or other Bacillus thuringiensis proteinaceous crystal toxins, which associate with protein receptors. The ability of Pleurotus aegerolysins to specifically interact with sphingolipid-enriched domains in mammalian cells can be further exploited to visualize lipid rafts in living cells, and to treat certain types of tumours and metabolic disorders. Finally, these proteins can strongly enhance fruiting initiation of P. ostreatus even when applied externally. In this review, we summarise the current knowledge of the potential biotechnological and biomedical applications of the Pleurotus aegerolysins, either alone or when complexed with pleurotolysin B, with special emphasis on their bioinsecticidal effects.
Collapse
Affiliation(s)
- Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| | - Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| | - Špela Modic
- Plant Protection Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia.
| | - Jaka Razinger
- Plant Protection Department, Agricultural Institute of Slovenia, Hacquetova 17, 1000 Ljubljana, Slovenia.
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia.
| |
Collapse
|
20
|
Binding specificity of ostreolysin A6 towards Sf9 insect cell lipids. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183307. [PMID: 32298680 DOI: 10.1016/j.bbamem.2020.183307] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/24/2020] [Accepted: 04/07/2020] [Indexed: 01/05/2023]
Abstract
Oyster mushrooms (Pleurotus spp.) have recently been shown to produce insecticidal bi-component protein complexes based on the aegerolysin proteins. A role for these proteins is thus indicated for defence and protection of the mushroom, and we propose their use as new environmentally friendly bioinsecticides. These aegerolysin-based protein complexes permeabilise artificial lipid vesicles through aegerolysin binding to an insect-specific sphingolipid, ceramide phosphoethanolamine (CPE), and they are cytotoxic for the Spodoptera frugiferda (Sf9) insect cell line. Tandem mass spectrometry analysis of the Sf9 lipidome uncovered lipids not previously reported in the literature, including in particular C14 sphingosine-based CPE molecular species, which comprised ~4 mol% of the whole lipidome. Further analysis of the lipid binding specificity of an aegerolysin from P. ostreatus, ostreolysin A6 (OlyA6), to lipid vesicles composed of commercial lipids, to lipid vesicles composed of the total lipid extract from Sf9 cells, and to HPLC-separated Sf9 cell lipid fractions containing ceramides, confirmed CPE as the main OlyA6 receptor, but also highlighted the importance of membrane cholesterol for formation of strong and stable interactions of OlyA6 with artificial and natural lipid membranes. Binding assays performed with glycan arrays and surface plasmon resonance, which included invertebrate-specific glycans, excluded these saccharides as potential additional OlyA6 receptors.
Collapse
|
21
|
Sun L, Su Y, Wang JG, Xia F, Xu Y, Li D. DNA nanotweezers for stabilizing and dynamically lighting up a lipid raft on living cell membranes and the activation of T cells. Chem Sci 2020; 11:1581-1586. [PMID: 34084389 PMCID: PMC8148038 DOI: 10.1039/c9sc06203c] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lipid rafts are generally considered as nanodomains on cell membranes and play important roles in signaling, viral infection, and membrane trafficking. However, the raft hypothesis is still debated with many inconsistencies because the nanoscale and transient heterogeneous raft structure creates difficulties in its location and functional analysis. In the present study, we report a DNA nanotweezer composed of a cholesterol-functionalized DNA duplex that stabilizes transient lipid rafts, which facilitate the further analysis of the raft component and its functions via other spectroscopy tools. The proposed DNA nanotweezer can induce clustering of raft-associated components (saturated lipids, membrane protein and possibly endogenous cholesterol), leading to the T cell proliferation through clustering of a T-cell antigen receptor (TCR). The flexibility of random sequence noncoding DNA provides versatile possibilities of manipulating lipid rafts and activating T cells, and thus opens new ways in a future T cell therapy. We report a DNA nanotweezer that recruits raft-associated lipids, proteins and possibly endogenous cholesterol on living cell membrane. The DNA nanotweezers could activate T cell proliferation in a nonspecific activation manner.![]()
Collapse
Affiliation(s)
- Lele Sun
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai
- China
- Institute of Functional Nano & Soft Materials (FUNSOM)
| | - Yingying Su
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai
- China
| | - Jun-Gang Wang
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai
- China
| | - Fei Xia
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai
- China
| | - Ying Xu
- Department of Pathophysiology
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education
- Shanghai Jiao-Tong University School of Medicine
- Shanghai
- China
| | - Di Li
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai
- China
| |
Collapse
|
22
|
Novak M, Čepin U, Hodnik V, Narat M, Jamnik M, Kraševec N, Sepčić K, Anderluh G. Functional studies of aegerolysin and MACPF-like proteins in Aspergillus niger. Mol Microbiol 2019; 112:1253-1269. [PMID: 31376198 DOI: 10.1111/mmi.14360] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2019] [Indexed: 12/21/2022]
Abstract
Proteins of the aegerolysin family have a high abundance in Fungi. Due to their specific binding to membrane lipids, and their membrane-permeabilization potential in concert with protein partner(s) belonging to a membrane-attack-complex/perforin (MACPF) superfamily, they were proposed as useful tools in different biotechnological and biomedical applications. In this work, we performed functional studies on expression of the genes encoding aegerolysin and MACPF-like proteins in Aspergillus niger. Our results suggest the sporulation process being crucial for strong induction of the expression of all these genes. However, deletion of either of the aegerolysin genes did not influence the growth, development, sporulation efficiency and phenotype of the mutants, indicating that aegerolysins are not key factors in the sporulation process. In all our expression studies we noticed a strong correlation in the expression of one aegerolysin and MACPF-like gene. Aegerolysins were confirmed to be secreted from the fungus. We also showed the specific interaction of a recombinant A. niger aegerolysin with an invertebrate-specific membrane sphingolipid. Moreover, using this protein labelled with mCherry we successfully stained insect cells membranes containing this particular sphingolipid. Our combined results suggest, that aegerolysins in this species, and probably also in other aspergilli, could be involved in defence against predators.
Collapse
Affiliation(s)
- Maruša Novak
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Urška Čepin
- BioSistemika Ltd and National Institute of Biology, Ljubljana, Slovenia
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Narat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Jamnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Nada Kraševec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| |
Collapse
|
23
|
Israeli E, Adler Berken N, Gover O, Waechtershaeuser E, Graeve L, Schwartz B. Recombinant ostreolysin (rOly) inhibits the anti-adipogenic Hedgehog (Hh) signaling pathway in 3T3-L1 cells. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
24
|
Ceramide phosphoethanolamine, an enigmatic cellular membrane sphingolipid. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1284-1292. [DOI: 10.1016/j.bbamem.2019.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/14/2022]
|
25
|
Functional link between plasma membrane spatiotemporal dynamics, cancer biology, and dietary membrane-altering agents. Cancer Metastasis Rev 2019; 37:519-544. [PMID: 29860560 DOI: 10.1007/s10555-018-9733-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The cell plasma membrane serves as a nexus integrating extra- and intracellular components, which together enable many of the fundamental cellular signaling processes that sustain life. In order to perform this key function, plasma membrane components assemble into well-defined domains exhibiting distinct biochemical and biophysical properties that modulate various signaling events. Dysregulation of these highly dynamic membrane domains can promote oncogenic signaling. Recently, it has been demonstrated that select membrane-targeted dietary bioactives (MTDBs) have the ability to remodel plasma membrane domains and subsequently reduce cancer risk. In this review, we focus on the importance of plasma membrane domain structural and signaling functionalities as well as how loss of membrane homeostasis can drive aberrant signaling. Additionally, we discuss the intricacies associated with the investigation of these membrane domain features and their associations with cancer biology. Lastly, we describe the current literature focusing on MTDBs, including mechanisms of chemoprevention and therapeutics in order to establish a functional link between these membrane-altering biomolecules, tuning of plasma membrane hierarchal organization, and their implications in cancer prevention.
Collapse
|
26
|
Panevska A, Hodnik V, Skočaj M, Novak M, Modic Š, Pavlic I, Podržaj S, Zarić M, Resnik N, Maček P, Veranič P, Razinger J, Sepčić K. Pore-forming protein complexes from Pleurotus mushrooms kill western corn rootworm and Colorado potato beetle through targeting membrane ceramide phosphoethanolamine. Sci Rep 2019; 9:5073. [PMID: 30911026 PMCID: PMC6433908 DOI: 10.1038/s41598-019-41450-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 03/08/2019] [Indexed: 01/13/2023] Open
Abstract
Aegerolysins ostreolysin A (OlyA) and pleurotolysin A (PlyA), and pleurotolysin B (PlyB) with the membrane-attack-complex/perforin domain are proteins from the mushroom genus Pleurotus. Upon binding to sphingomyelin/cholesterol-enriched membranes, OlyA and PlyA can recruit PlyB to form multimeric bi-component transmembrane pores. Recently, Pleurotus aegerolysins OlyA, PlyA2 and erylysin A (EryA) were demonstrated to preferentially bind to artificial lipid membranes containing 50 mol% ceramide phosphoethanolamine (CPE), the main sphingolipid in invertebrate cell membranes. In this study, we demonstrate that OlyA6, PlyA2 and EryA bind to insect cells and to artificial lipid membranes with physiologically relevant CPE concentrations. Moreover, these aegerolysins permeabilize these membranes when combined with PlyB. These aegerolysin/PlyB complexes show selective toxicity toward western corn rootworm larvae and adults and Colorado potato beetle larvae. These data strongly suggest that these aegerolysin/PlyB complexes recognize CPE as their receptor molecule in the insect midgut. This mode of binding is different from those described for similar aegerolysin-based bacterial complexes, or other Bacillus thuringiensis Cry toxins, which have protein receptors. Targeting of Pleurotus aegerolysins to CPE and formation of transmembrane pores in concert with PlyB suggest the use of aegerolysin/PlyB complexes as novel biopesticides for the control of western corn rootworm and Colorado potato beetle.
Collapse
Affiliation(s)
- Anastasija Panevska
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Maruša Novak
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Špela Modic
- Agricultural Institute of Slovenia, Hacquetova 17, 1000, Ljubljana, Slovenia
| | - Ivana Pavlic
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia.,Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia
| | - Sara Podržaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Miki Zarić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Peter Maček
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Jaka Razinger
- Agricultural Institute of Slovenia, Hacquetova 17, 1000, Ljubljana, Slovenia.
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000, Ljubljana, Slovenia.
| |
Collapse
|
27
|
Oda Y, Saito K, Nakata M. Structural analyses of a hemolytic compound found in an extract of Hypsizygus marmoreus fruiting bodies at a low pH. Biosci Trends 2019; 13:86-90. [PMID: 30700653 DOI: 10.5582/bst.2019.01008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The current study determined the structure of a hemolytic compound found in an extract from the fruiting bodies of the edible mushroom Hypsizygus marmoreus when its pH was lowered. The hemolytic compound was purified using the modified Bligh and Dyer method followed by chromatography using reversed phase and silica gel columns. Structural analyses of the purified hemolytic compound were performed using NMR and ESI-MS. The deduced structure indicated a trans,trans-5,8-docosadienoic acid calcium salt. Although numerous proteinous hemolysins from various mushrooms have been described, the current study is the first to report on a low-molecular-weight hemolytic compound derived from an H. marmoreus extract.
Collapse
Affiliation(s)
- Yoshiki Oda
- Technology Joint Management Office, Research Promotion Division, Tokai University
| | - Kohsuke Saito
- Department of Applied Biochemistry, Tokai University
| | | |
Collapse
|
28
|
Endapally S, Frias D, Grzemska M, Gay A, Tomchick DR, Radhakrishnan A. Molecular Discrimination between Two Conformations of Sphingomyelin in Plasma Membranes. Cell 2019; 176:1040-1053.e17. [PMID: 30712872 DOI: 10.1016/j.cell.2018.12.042] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/19/2018] [Accepted: 12/21/2018] [Indexed: 12/14/2022]
Abstract
Sphingomyelin and cholesterol are essential lipids that are enriched in plasma membranes of animal cells, where they interact to regulate membrane properties and many intracellular signaling processes. Despite intense study, the interaction between these lipids in membranes is not well understood. Here, structural and biochemical analyses of ostreolysin A (OlyA), a protein that binds to membranes only when they contain both sphingomyelin and cholesterol, reveal that sphingomyelin adopts two distinct conformations in membranes when cholesterol is present. One conformation, bound by OlyA, is induced by stoichiometric, exothermic interactions with cholesterol, properties that are consistent with sphingomyelin/cholesterol complexes. In its second conformation, sphingomyelin is free from cholesterol and does not bind OlyA. A point mutation abolishes OlyA's ability to discriminate between these two conformations. In cells, levels of sphingomyelin/cholesterol complexes are held constant over a wide range of plasma membrane cholesterol concentrations, enabling precise regulation of the chemical activity of cholesterol.
Collapse
Affiliation(s)
- Shreya Endapally
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Donna Frias
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Magdalena Grzemska
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Austin Gay
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Diana R Tomchick
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
29
|
Hullin-Matsuda F, Murate M, Kobayashi T. Protein probes to visualize sphingomyelin and ceramide phosphoethanolamine. Chem Phys Lipids 2018; 216:132-141. [DOI: 10.1016/j.chemphyslip.2018.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/29/2018] [Accepted: 09/03/2018] [Indexed: 01/05/2023]
|
30
|
Sezgin E, Levental I, Mayor S, Eggeling C. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol 2017; 18:361-374. [PMID: 28356571 PMCID: PMC5500228 DOI: 10.1038/nrm.2017.16] [Citation(s) in RCA: 1288] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cellular plasma membranes are laterally heterogeneous, featuring a variety of distinct subcompartments that differ in their biophysical properties and composition. A large number of studies have focused on understanding the basis for this heterogeneity and its physiological relevance. The membrane raft hypothesis formalized a physicochemical principle for a subtype of such lateral membrane heterogeneity, in which the preferential associations between cholesterol and saturated lipids drive the formation of relatively packed (or ordered) membrane domains that selectively recruit certain lipids and proteins. Recent studies have yielded new insights into this mechanism and its relevance in vivo, owing primarily to the development of improved biochemical and biophysical technologies.
Collapse
Affiliation(s)
- Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| | - Ilya Levental
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, 6431 Fannin Street, Houston, Texas 77030, USA
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bellary Road, Bangalore 560065, India
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| |
Collapse
|
31
|
Butala M, Novak M, Kraševec N, Skočaj M, Veranič P, Maček P, Sepčić K. Aegerolysins: Lipid-binding proteins with versatile functions. Semin Cell Dev Biol 2017; 72:142-151. [PMID: 28506897 DOI: 10.1016/j.semcdb.2017.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 04/13/2017] [Accepted: 05/11/2017] [Indexed: 01/21/2023]
Abstract
Proteins of the aegerolysin family span many kingdoms of life. They are relatively widely distributed in bacteria and fungi, but also appear in plants, protozoa and insects. Despite being produced in abundance in cells at specific developmental stages and present in secretomes, only a few aegerolysins have been studied in detail. In particular, their organism-specific physiological roles are intriguing. Here, we review published findings to date on the distribution, molecular interactions and biological activities of this family of structurally and functionally versatile proteins, the aegerolysins.
Collapse
Affiliation(s)
- Matej Butala
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Maruša Novak
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Nada Kraševec
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Matej Skočaj
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Peter Maček
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia.
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, 1000 Ljubljana, Slovenia.
| |
Collapse
|
32
|
Skočaj M, Yu Y, Grundner M, Resnik N, Bedina Zavec A, Leonardi A, Križaj I, Guella G, Maček P, Kreft ME, Frangež R, Veranič P, Sepčić K. Characterisation of plasmalemmal shedding of vesicles induced by the cholesterol/sphingomyelin binding protein, ostreolysin A-mCherry. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1858:2882-2893. [PMID: 27591807 DOI: 10.1016/j.bbamem.2016.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 08/10/2016] [Accepted: 08/30/2016] [Indexed: 12/27/2022]
Abstract
Ostreolysin A (OlyA) is a 15-kDa protein that binds selectively to cholesterol/sphingomyelin membrane nanodomains. This binding induces the production of extracellular vesicles (EVs) that comprise both microvesicles with diameters between 100nm and 1μm, and larger vesicles of around 10-μm diameter in Madin-Darby canine kidney cells. In this study, we show that vesiculation of these cells by the fluorescent fusion protein OlyA-mCherry is not affected by temperature, is not mediated via intracellular Ca2+ signalling, and does not compromise cell viability and ultrastructure. Seventy-one proteins that are mostly of cytosolic and nuclear origin were detected in these shed EVs using mass spectroscopy. In the cells and EVs, 218 and 84 lipid species were identified, respectively, and the EVs were significantly enriched in lysophosphatidylcholines and cholesterol. Our collected data suggest that OlyA-mCherry binding to cholesterol/sphingomyelin membrane nanodomains induces specific lipid sorting into discrete patches, which promotes plasmalemmal blebbing and EV shedding from the cells. We hypothesize that these effects are accounted for by changes of local membrane curvature upon the OlyA-mCherry-plasmalemma interaction. We suggest that the shed EVs are a potentially interesting model for biophysical and biochemical studies of cell membranes, and larger vesicles could represent tools for non-invasive sampling of cytosolic proteins from cells and thus metabolic fingerprinting.
Collapse
Affiliation(s)
- Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana, Slovenia; Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia.
| | - Yang Yu
- Bioorganic Chemistry Laboratory, Department of Physics, Via Sommarive 14, University of Trento, Povo (TN), Italy.
| | - Maja Grundner
- Institute of Biophysics, Faculty of Medicine, Vrazov trg 2, University of Ljubljana, Ljubljana, Slovenia.
| | - Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia.
| | - Apolonija Bedina Zavec
- Laboratory of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia.
| | - Adrijana Leonardi
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia.
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Jamova 39, Ljubljana, Slovenia; Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, Večna pot 113, University of Ljubljana, Ljubljana, Slovenia.
| | - Graziano Guella
- Bioorganic Chemistry Laboratory, Department of Physics, Via Sommarive 14, University of Trento, Povo (TN), Italy.
| | - Peter Maček
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana, Slovenia.
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia.
| | - Robert Frangež
- Institute of Physiology, Pharmacology and Toxicology, Veterinary Faculty, Gerbičeva 60, University of Ljubljana, Ljubljana, Slovenia.
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia.
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, Ljubljana, Slovenia.
| |
Collapse
|
33
|
Probing phosphoethanolamine-containing lipids in membranes with duramycin/cinnamycin and aegerolysin proteins. Biochimie 2016; 130:81-90. [DOI: 10.1016/j.biochi.2016.09.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023]
|
34
|
Makino A, Abe M, Ishitsuka R, Murate M, Kishimoto T, Sakai S, Hullin-Matsuda F, Shimada Y, Inaba T, Miyatake H, Tanaka H, Kurahashi A, Pack CG, Kasai RS, Kubo S, Schieber NL, Dohmae N, Tochio N, Hagiwara K, Sasaki Y, Aida Y, Fujimori F, Kigawa T, Nishibori K, Parton RG, Kusumi A, Sako Y, Anderluh G, Yamashita M, Kobayashi T, Greimel P, Kobayashi T. A novel sphingomyelin/cholesterol domain-specific probe reveals the dynamics of the membrane domains during virus release and in Niemann-Pick type C. FASEB J 2016; 31:1301-1322. [PMID: 27492925 DOI: 10.1096/fj.201500075r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/18/2016] [Indexed: 01/07/2023]
Abstract
We identified a novel, nontoxic mushroom protein that specifically binds to a complex of sphingomyelin (SM), a major sphingolipid in mammalian cells, and cholesterol (Chol). The purified protein, termed nakanori, labeled cell surface domains in an SM- and Chol-dependent manner and decorated specific lipid domains that colocalized with inner leaflet small GTPase H-Ras, but not K-Ras. The use of nakanori as a lipid-domain-specific probe revealed altered distribution and dynamics of SM/Chol on the cell surface of Niemann-Pick type C fibroblasts, possibly explaining some of the disease phenotype. In addition, that nakanori treatment of epithelial cells after influenza virus infection potently inhibited virus release demonstrates the therapeutic value of targeting specific lipid domains for anti-viral treatment.-Makino, A., Abe, M., Ishitsuka, R., Murate, M., Kishimoto, T., Sakai, S., Hullin-Matsuda, F., Shimada, Y., Inaba, T., Miyatake, H., Tanaka, H., Kurahashi, A., Pack, C.-G., Kasai, R. S., Kubo, S., Schieber, N. L., Dohmae, N., Tochio, N., Hagiwara, K., Sasaki, Y., Aida, Y., Fujimori, F., Kigawa, T., Nishibori, K., Parton, R. G., Kusumi, A., Sako, Y., Anderluh, G., Yamashita, M., Kobayashi, T., Greimel, P., Kobayashi, T. A novel sphingomyelin/cholesterol domain-specific probe reveals the dynamics of the membrane domains during virus release and in Niemann-Pick type C.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Françoise Hullin-Matsuda
- Rikagaku Kenkyūsho (RIKEN), Saitama, Japan.,Université Lyon 1, INSERM, Unité 1060, Villeurbanne, France
| | | | | | | | - Hideko Tanaka
- Faculty of Core Research, Natural Science Division, Ochanomizu University, Tokyo, Japan
| | | | | | - Rinshi S Kasai
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shuku Kubo
- Daiichi Sankyo Co., Limited, Tokyo, Japan
| | - Nicole L Schieber
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | | | | | | | | | - Yoko Aida
- Rikagaku Kenkyūsho (RIKEN), Saitama, Japan
| | - Fumihiro Fujimori
- Graduate School of Humanities and Life Sciences, Tokyo Kasei University, Tokyo, Japan
| | | | | | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.,Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, Queensland, Australia
| | - Akihiro Kusumi
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | | | - Gregor Anderluh
- National Institute of Chemistry, Ljubljana, Slovenia.,Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia; and
| | | | - Tetsuyuki Kobayashi
- Faculty of Core Research, Natural Science Division, Ochanomizu University, Tokyo, Japan
| | | | - Toshihide Kobayashi
- Rikagaku Kenkyūsho (RIKEN), Saitama, Japan; .,Unité Mixte de Recherche 7213, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France
| |
Collapse
|
35
|
Phospholipase Cβ1 induces membrane tubulation and is involved in caveolae formation. Proc Natl Acad Sci U S A 2016; 113:7834-9. [PMID: 27342861 DOI: 10.1073/pnas.1603513113] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Lipid membrane curvature plays important roles in various physiological phenomena. Curvature-regulated dynamic membrane remodeling is achieved by the interaction between lipids and proteins. So far, several membrane sensing/sculpting proteins, such as Bin/amphiphysin/Rvs (BAR) proteins, are reported, but there remains the possibility of the existence of unidentified membrane-deforming proteins that have not been uncovered by sequence homology. To identify new lipid membrane deformation proteins, we applied liposome-based microscopic screening, using unbiased-darkfield microscopy. Using this method, we identified phospholipase Cβ1 (PLCβ1) as a new candidate. PLCβ1 is well characterized as an enzyme catalyzing the hydrolysis of phosphatidylinositol-4,5-bisphosphate (PIP2). In addition to lipase activity, our results indicate that PLCβ1 possessed the ability of membrane tubulation. Lipase domains and inositol phospholipids binding the pleckstrin homology (PH) domain of PLCβ1 were not involved, but the C-terminal sequence was responsible for this tubulation activity. Computational modeling revealed that the C terminus displays the structural homology to the BAR domains, which is well known as a membrane sensing/sculpting domain. Overexpression of PLCβ1 caused plasma membrane tubulation, whereas knockdown of the protein reduced the number of caveolae and induced the evagination of caveolin-rich membrane domains. Taken together, our results suggest a new function of PLCβ1: plasma membrane remodeling, and in particular, caveolae formation.
Collapse
|
36
|
Abe M, Kobayashi T. Dynamics of sphingomyelin- and cholesterol-enriched lipid domains during cytokinesis. Methods Cell Biol 2016; 137:15-24. [PMID: 28065303 DOI: 10.1016/bs.mcb.2016.03.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sphingomyelin (SM) and cholesterol (Chol) are the major lipids in the mammalian cells, which are mainly localized to the plasma membrane. Multiple lines of evidence suggest that these lipids form local lipid domains in the plasma membrane, playing functional roles in the cell. Several observations have suggested that these lipid domains are required for cytokinesis. In this chapter, we show the methods for visualizing SM-rich and/or Chol-rich membrane domains at cytokinesis by using specific lipid-binding proteins. Lysenin, equinatoxin II, perfringolysin O, and pleurotolysin A2 bind specifically to clustered SM-rich domain, dispersed SM-rich domain, Chol-rich domain, and SM/Chol mixtures, respectively. Nontoxic forms of these lipid-binding proteins fused to fluorescent proteins are used for imaging lipid domains in biological membranes at cytokinesis. The image analysis reveals the structures and functions of SM-rich and/or Chol-rich domains at the time of cytokinesis.
Collapse
Affiliation(s)
- M Abe
- RIKEN, Wako, Saitama, Japan
| | - T Kobayashi
- RIKEN, Wako, Saitama, Japan; CNRS, Illkirch, France
| |
Collapse
|
37
|
Kishimoto T, Ishitsuka R, Kobayashi T. Detectors for evaluating the cellular landscape of sphingomyelin- and cholesterol-rich membrane domains. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:812-829. [PMID: 26993577 DOI: 10.1016/j.bbalip.2016.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/09/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
Abstract
Although sphingomyelin and cholesterol are major lipids of mammalian cells, the detailed distribution of these lipids in cellular membranes remains still obscure. However, the recent development of protein probes that specifically bind sphingomyelin and/or cholesterol provides new information about the landscape of the lipid domains that are enriched with sphingomyelin or cholesterol or both. Here, we critically summarize the tools to study distribution and dynamics of sphingomyelin and cholesterol. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
Affiliation(s)
| | - Reiko Ishitsuka
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan; INSERM U1060, Université Lyon 1, Villeurbanne 69621, France.
| |
Collapse
|
38
|
Abstract
Pore forming toxins (PFTs) evolved to permeate the plasma membrane of target cells. This is achieved in a multistep mechanism that usually involves binding of soluble protein monomer to the lipid membrane, oligomerization at the plane of the membrane, and insertion of part of the polypeptide chain across the lipid membrane to form a conductive channel. Introduced pores allow uncontrolled transport of solutes across the membrane, inflicting damage to the target cell. PFTs are usually studied from the perspective of structure-function relationships, often neglecting the important role of the bulk membrane properties on the PFT mechanism of action. In this Account, we discuss how membrane lateral heterogeneity, thickness, and fluidity influence the pore forming process of PFTs. In general, lipid molecules are more accessible for binding in fluid membranes due to steric reasons. When PFT specifically binds ordered domains, it usually recognizes a specific lipid distribution pattern, like sphingomyelin (SM) clusters or SM/cholesterol complexes, and not individual lipid species. Lipid domains were also suggested to act as an additional concentration platform facilitating PFT oligomerization, but this is yet to be shown. The last stage in PFT action is the insertion of the transmembrane segment across the membranes to build the transmembrane pore walls. Conformational changes are a spontaneous process, and sufficient free energy has to be available for efficient membrane penetration. Therefore, fluid bilayers are permeabilized more readily in comparison to highly ordered and thicker liquid ordered lipid phase (Lo). Energetically more costly insertion into the Lo phase can be driven by the hydrophobic mismatch between the thinner liquid disordered phase (Ld) and large protein complexes, which are unable to tilt like single transmembrane segments. In the case of proteolipid pores, membrane properties can directly modulate pore size, stability, and even selectivity. Finally, events associated with pore formation can modulate properties of the lipid membrane and affect its organization. Model membranes do not necessarily reproduce the physicochemical properties of the native cellular membrane, and caution is needed when transferring results from model to native lipid membranes. In this context, the utilization of novel approaches that enable studying PFTs on living cells at a single molecule level should reveal complex protein-lipid membrane interactions in greater detail.
Collapse
Affiliation(s)
- Nejc Rojko
- 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
| |
Collapse
|
39
|
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
|
40
|
Ogiso H, Taniguchi M, Okazaki T. Analysis of lipid-composition changes in plasma membrane microdomains. J Lipid Res 2015; 56:1594-605. [PMID: 26116739 DOI: 10.1194/jlr.m059972] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Indexed: 11/20/2022] Open
Abstract
Sphingolipids accumulate in plasma membrane microdomain sites, such as caveolae or lipid rafts. Such microdomains are considered to be important nexuses for signal transduction, although changes in the microdomain lipid components brought about by signaling are poorly understood. Here, we applied a cationic colloidal silica bead method to analyze plasma membrane lipids from monolayer cells cultured in a 10 cm dish. The detergent-resistant fraction from the silica bead-coated membrane was analyzed by LC-MS/MS to evaluate the microdomain lipids. This method revealed that glycosphingolipids composed the microdomains as a substitute for sphingomyelin (SM) in mouse embryonic fibroblasts (tMEFs) from an SM synthase 1/2 double KO (DKO) mouse. The rate of formation of the detergent-resistant region was unchanged compared with that of WT-tMEFs. C2-ceramide (Cer) stimulation caused greater elevations in diacylglycerol and phosphatidic acid levels than in Cer levels within the microdomains of WT-tMEFs. We also found that lipid changes in the microdomains of SM-deficient DKO-tMEFs caused by serum stimulation occurred in the same manner as that of WT-tMEFs. This practical method for analyzing membrane lipids will facilitate future comprehensive analyses of membrane microdomain-associated responses.
Collapse
Affiliation(s)
- Hideo Ogiso
- Department of Hematology/Immunology Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan
| | - Makoto Taniguchi
- Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan
| | - Toshiro Okazaki
- Department of Hematology/Immunology Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan
| |
Collapse
|
41
|
Bhat HB, Ishitsuka R, Inaba T, Murate M, Abe M, Makino A, Kohyama-Koganeya A, Nagao K, Kurahashi A, Kishimoto T, Tahara M, Yamano A, Nagamune K, Hirabayashi Y, Juni N, Umeda M, Fujimori F, Nishibori K, Yamaji-Hasegawa A, Greimel P, Kobayashi T. Evaluation of aegerolysins as novel tools to detect and visualize ceramide phosphoethanolamine, a major sphingolipid in invertebrates. FASEB J 2015; 29:3920-34. [PMID: 26060215 DOI: 10.1096/fj.15-272112] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/26/2015] [Indexed: 12/13/2022]
Abstract
Ceramide phosphoethanolamine (CPE), a sphingomyelin analog, is a major sphingolipid in invertebrates and parasites, whereas only trace amounts are present in mammalian cells. In this study, mushroom-derived proteins of the aegerolysin family—pleurotolysin A2 (PlyA2; K(D) = 12 nM), ostreolysin (Oly; K(D) = 1.3 nM), and erylysin A (EryA; K(D) = 1.3 nM)—strongly associated with CPE/cholesterol (Chol)-containing membranes, whereas their low affinity to sphingomyelin/Chol precluded establishment of the binding kinetics. Binding specificity was determined by multilamellar liposome binding assays, supported bilayer assays, and solid-phase studies against a series of neutral and negatively charged lipid classes mixed 1:1 with Chol or phosphatidylcholine. No cross-reactivity was detected with phosphatidylethanolamine. Only PlyA2 also associated with CPE, independent of Chol content (K(D) = 41 μM), rendering it a suitable tool for visualizing CPE in lipid-blotting experiments and biologic samples from sterol auxotrophic organisms. Visualization of CPE enrichment in the CNS of Drosophila larvae (by PlyA2) and in the bloodstream form of the parasite Trypanosoma brucei (by EryA) by fluorescence imaging demonstrated the versatility of aegerolysin family proteins as efficient tools for detecting and visualizing CPE.
Collapse
Affiliation(s)
- Hema Balakrishna Bhat
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Reiko Ishitsuka
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Takehiko Inaba
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Motohide Murate
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Mitsuhiro Abe
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Asami Makino
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Ayako Kohyama-Koganeya
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Kohjiro Nagao
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Atsushi Kurahashi
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Takuma Kishimoto
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Michiru Tahara
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Akinori Yamano
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Kisaburo Nagamune
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Yoshio Hirabayashi
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Naoto Juni
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Masato Umeda
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Fumihiro Fujimori
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Kozo Nishibori
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Akiko Yamaji-Hasegawa
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Peter Greimel
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| | - Toshihide Kobayashi
- *Lipid Biology Laboratory, RIKEN, and Laboratory of Molecular Membrane Neuroscience, Brain Science Institute, RIKEN, Saitama, Japan; Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Yukiguni Maitake Co. Ltd., Niigata, Japan; Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan; Graduate School of Life and Environmental Sciences and Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan; **Department of Environmental Science and Education, Faculty of Human Life Science, Tokyo Kasei University, Tokyo, Japan; and Institut National de la Santé et de la Recherche Médicale Unité 1060, Université Lyon 1, Villeurbanne, France
| |
Collapse
|
42
|
Morante K, Caaveiro JMM, Tanaka K, González-Mañas JM, Tsumoto K. A pore-forming toxin requires a specific residue for its activity in membranes with particular physicochemical properties. J Biol Chem 2015; 290:10850-61. [PMID: 25759390 DOI: 10.1074/jbc.m114.615211] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Indexed: 12/29/2022] Open
Abstract
The physicochemical landscape of the bilayer modulates membrane protein function. Actinoporins are a family of potent hemolytic proteins from sea anemones acting at the membrane level. This family of cytolysins preferentially binds to target membranes containing sphingomyelin, where they form lytic pores giving rise to cell death. Although the cytolytic activity of the actinoporin fragaceatoxin C (FraC) is sensitive to vesicles made of various lipid compositions, it is far from clear how this toxin adjusts its mechanism of action to a broad range of physiochemical landscapes. Herein, we show that the conserved residue Phe-16 of FraC is critical for pore formation in cholesterol-rich membranes such as those of red blood cells. The interaction of a panel of muteins of Phe-16 with model membranes composed of raft-like lipid domains is inactivated in cholesterol-rich membranes but not in cholesterol-depleted membranes. These results indicate that actinoporins recognize different membrane environments, resulting in a wider repertoire of susceptible target membranes (and preys) for sea anemones. In addition, this study has unveiled promising candidates for the development of protein-based biosensors highly sensitive to the concentration of cholesterol within the membrane.
Collapse
Affiliation(s)
- Koldo Morante
- From the Department of Bioengineering, Graduate School of Engineering and the Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940 Leioa, Spain, and
| | - Jose M M Caaveiro
- From the Department of Bioengineering, Graduate School of Engineering and
| | - Koji Tanaka
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Juan Manuel González-Mañas
- the Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940 Leioa, Spain, and
| | - Kouhei Tsumoto
- From the Department of Bioengineering, Graduate School of Engineering and Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan, the Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo, Minato-ku, 108-8639 Tokyo, Japan
| |
Collapse
|
43
|
Novak M, Kraševec N, Skočaj M, Maček P, Anderluh G, Sepčić K. Fungal aegerolysin-like proteins: distribution, activities, and applications. Appl Microbiol Biotechnol 2014; 99:601-10. [PMID: 25476018 DOI: 10.1007/s00253-014-6239-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 12/15/2022]
Abstract
The aegerolysin protein family (from aegerolysin of the mushroom Agrocybe aegerita) comprises proteins of ∼15-20 kDa from various eukaryotic and bacterial taxa. Aegerolysins are inconsistently distributed among fungal species, and variable numbers of homologs have been reported for species within the same genus. As such noncore proteins, without a member of a protein family in each of the sequenced fungi, they can give insight into different species-specific processes. Some aegerolysins have been reported to be hemolytically active against mammalian erythrocytes. However, some function as bi-component proteins that have membrane activity in concert with another protein that contains a membrane attack complex/perforin domain. The function of most of aegerolysins is unknown, although some have been suggested to have a role in development of the organism. Potential biotechnological applications of aegerolysins are already evident, despite the limited scientific knowledge available at present. Some mushroom aegerolysins, for example, can be used as markers to detect and label specific membrane lipids. Others can be used as biomarkers of fungal exposure, where their genes can serve as targets for detection of fungi and their progression during infectious diseases. Antibodies against aegerolysins can also be raised as immuno-diagnostic tools. Aegerolysins have been shown to serve as a species determination tool for fungal phytopathogen isolates in terms of some closely related species, where commonly used internal transcribed spacer barcoding has failed. Moreover, strong promoters that regulate aegerolysin genes can promote secretion of heterologous proteins from fungi and have been successfully applied in simultaneous multi-gene expression techniques.
Collapse
Affiliation(s)
- Maruša Novak
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Večna pot 111, SI-1000, Ljubljana, Slovenia
| | | | | | | | | | | |
Collapse
|
44
|
Makino A, Abe M, Murate M, Inaba T, Yilmaz N, Hullin‐Matsuda F, Kishimoto T, Schieber NL, Taguchi T, Arai H, Anderluh G, Parton RG, Kobayashi T. Visualization of the heterogeneous membrane distribution of sphingomyelin associated with cytokinesis, cell polarity, and sphingolipidosis. FASEB J 2014; 29:477-93. [DOI: 10.1096/fj.13-247585] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Asami Makino
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
| | - Mitsuhiro Abe
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
| | - Motohide Murate
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
| | - Takehiko Inaba
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
| | - Neval Yilmaz
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
| | - Françoise Hullin‐Matsuda
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
- TNSERM U1060‐Université LyonVilleurbanneFrance
| | | | - Nicole L. Schieber
- Institute for Molecular BioscienceUniversity of QueenslandSt. LuciaQueenslandAustralia
| | - Tomohiko Taguchi
- Graduate School of Pharmaceutical SciencesUniversity of TokyoTokyoJapan
| | - Hiroyuki Arai
- Graduate School of Pharmaceutical SciencesUniversity of TokyoTokyoJapan
| | - Gregor Anderluh
- National Institute of ChemistryLjubljanaSlovenia
- Department of Biology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Robert G. Parton
- Institute for Molecular BioscienceUniversity of QueenslandSt. LuciaQueenslandAustralia
- Centre for Microscopy and MicroanalysisUniversity of QueenslandSt. LuciaQueenslandAustralia
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, HirosawaWako‐shiSaitamaJapan
- TNSERM U1060‐Université LyonVilleurbanneFrance
| |
Collapse
|
45
|
Skočaj M, Resnik N, Grundner M, Ota K, Rojko N, Hodnik V, Anderluh G, Sobota A, Maček P, Veranič P, Sepčić K. Tracking cholesterol/sphingomyelin-rich membrane domains with the ostreolysin A-mCherry protein. PLoS One 2014; 9:e92783. [PMID: 24664106 PMCID: PMC3963934 DOI: 10.1371/journal.pone.0092783] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/25/2014] [Indexed: 01/01/2023] Open
Abstract
Ostreolysin A (OlyA) is an ∼15-kDa protein that has been shown to bind selectively to membranes rich in cholesterol and sphingomyelin. In this study, we investigated whether OlyA fluorescently tagged at the C-terminal with mCherry (OlyA-mCherry) labels cholesterol/sphingomyelin domains in artificial membrane systems and in membranes of Madin-Darby canine kidney (MDCK) epithelial cells. OlyA-mCherry showed similar lipid binding characteristics to non-tagged OlyA. OlyA-mCherry also stained cholesterol/sphingomyelin domains in the plasma membranes of both fixed and living MDCK cells, and in the living cells, this staining was abolished by pretreatment with either methyl-β-cyclodextrin or sphingomyelinase. Double labelling of MDCK cells with OlyA-mCherry and the sphingomyelin-specific markers equinatoxin II-Alexa488 and GST-lysenin, the cholera toxin B subunit as a probe that binds to the ganglioside GM1, or the cholesterol-specific D4 domain of perfringolysin O fused with EGFP, showed different patterns of binding and distribution of OlyA-mCherry in comparison with these other proteins. Furthermore, we show that OlyA-mCherry is internalised in living MDCK cells, and within 90 min it reaches the juxtanuclear region via caveolin-1-positive structures. No binding to membranes could be seen when OlyA-mCherry was expressed in MDCK cells. Altogether, these data clearly indicate that OlyA-mCherry is a promising tool for labelling a distinct pool of cholesterol/sphingomyelin membrane domains in living and fixed cells, and for following these domains when they are apparently internalised by the cell.
Collapse
Affiliation(s)
- Matej Skočaj
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Grundner
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Ota
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nejc Rojko
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Vesna Hodnik
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
- National Institute of Chemistry, Ljubljana, Slovenia
| | - Andrzej Sobota
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Peter Maček
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
46
|
Abstract
Proteins with membrane-attack complex/perforin (MACPF) domains are found in almost all kingdoms of life, and they have a variety of biological roles, including defence and attack, organism development, and cell adhesion and signalling. The distribution of these proteins in fungi appears to be restricted to some Pezizomycotina and Basidiomycota species only, in correlation with another group of proteins with unknown biological function, known as aegerolysins. These two protein groups coincide in only a few species, and they might operate in concert as cytolytic bi-component pore-forming agents. Representative proteins here include pleurotolysin B, which has a MACPF domain, and the aegerolysin-like protein pleurotolysin A, and the very similar ostreolysin A, which have been purified from oyster mushroom (Pleurotus ostreatus). These have been shown to act in concert to perforate natural and artificial lipid membranes with high cholesterol and sphingomyelin content. The aegerolysin-like proteins provide the membrane cholesterol/sphingomyelin selectivity and recruit oligomerised pleurotolysin B molecules, to create a membrane-inserted pore complex. The resulting protein structure has been imaged with electron microscopy, and it has a 13-meric rosette-like structure, with a central lumen that is ~4-5 nm in diameter. The opened transmembrane pore is non-selectively permeable for ions and smaller neutral solutes, and is a cause of cytolysis of a colloid-osmotic type. The biological significance of these proteins for the fungal life-style is discussed.
Collapse
|