1
|
Mayorova TD, Koch TL, Kachar B, Jung JH, Reese TS, Smith CL. Placozoan secretory cell types implicated in feeding, innate immunity and regulation of behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.18.613768. [PMID: 39372748 PMCID: PMC11452194 DOI: 10.1101/2024.09.18.613768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Placozoa are millimeter-sized, flat, irregularly shaped ciliated animals that crawl on surfaces in warm oceans feeding on biofilms, which they digest externally. They stand out from other animals due to their simple body plans. They lack organs, body cavities, muscles and a nervous system and have only seven broadly defined morphological cell types, each with a unique distribution. Analyses of single cell transcriptomes of four species of placozoans revealed greater diversity of secretory cell types than evident from morphological studies, but the locations of many of these new cell types were unknown and it was unclear which morphological cell types they represent. Furthermore, there were contradictions between the conclusions of previous studies and the single cell RNAseq studies. To address these issues, we used mRNA probes for genes encoding secretory products expressed in different metacells in Trichoplax adhaerens to localize cells in whole mounts and in dissociated cell cultures, where their morphological features could be visualized and identified. The nature and functions of their secretory granules were further investigated with electron microscopic techniques and by imaging secretion in live animals during feeding episodes. We found that two cell types participate in disintegrating prey, one resembling a lytic cell type in mammals and another combining features of zymogen gland cells and enterocytes. We identified secretory epithelial cells expressing glycoproteins or short peptides implicated in defense. We located seven peptidergic cell types and two types of mucocytes. Our findings reveal mechanisms that placozoans use to feed and protect themselves from pathogens and clues about neuropeptidergic signaling. We compare placozoan secretory cell types with cell types in other animal phyla to gain insight about general evolutionary trends in cell type diversification, as well as pathways leading to the emergence of synapomorphies.
Collapse
|
2
|
Rojas-Palomino J, Altuna-Alvarez J, González-Magaña A, Queralt-Martín M, Albesa-Jové D, Alcaraz A. Electrophysiological dissection of the ion channel activity of the Pseudomonas aeruginosa ionophore protein toxin Tse5. Chem Phys Lipids 2025; 267:105472. [PMID: 39778700 DOI: 10.1016/j.chemphyslip.2025.105472] [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: 10/14/2024] [Revised: 12/19/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
We present an in-depth electrophysiological analysis of Tse5, a pore-forming toxin (PFT) delivered by the type VI secretion system (T6SS) of Pseudomonas aeruginosa. The T6SS is a sophisticated bacterial secretion system that injects toxic effector proteins into competing bacteria or host cells, providing a competitive advantage by disabling other microbes and modulating their environment. Our findings highlight the dependency of Tse5 insertion on membrane charge and electrolyte concentration, suggesting an in vivo effect from the periplasmic space. Conductance and selectivity experiments reveal a predominant and reproducible pore architecture of Tse5, characterized by a weak cation selectivity without chemical specificity. pH titration experiments suggest a proteolipidic pore structure influenced by both protein and lipid charges, a hypothesis further supported by experiments involving engineered mutants of Tse5 with altered glycine zippers. These results significantly advance our understanding of Tse5's molecular mechanism of toxicity, paving the way for potential applications in biosensing and macromolecular delivery.
Collapse
Affiliation(s)
- Jessica Rojas-Palomino
- Laboratory of Molecular Biophysics, Department of Physics, University Jaume I, Castellón 12071, Spain
| | - Jon Altuna-Alvarez
- Instituto Biofisika (CSIC, UPV/EHU), Fundación Biofísica Bizkaia/Biofisika Bizkaia Fundazioa (FBB) and Departamento de Bioquímica y Biología Molecular, University of the Basque Country, Leioa 48940, Spain
| | - Amaia González-Magaña
- Instituto Biofisika (CSIC, UPV/EHU), Fundación Biofísica Bizkaia/Biofisika Bizkaia Fundazioa (FBB) and Departamento de Bioquímica y Biología Molecular, University of the Basque Country, Leioa 48940, Spain
| | - María Queralt-Martín
- Laboratory of Molecular Biophysics, Department of Physics, University Jaume I, Castellón 12071, Spain
| | - David Albesa-Jové
- Instituto Biofisika (CSIC, UPV/EHU), Fundación Biofísica Bizkaia/Biofisika Bizkaia Fundazioa (FBB) and Departamento de Bioquímica y Biología Molecular, University of the Basque Country, Leioa 48940, Spain; Ikerbasque, Basque Foundation for Science, Bilbao 48013, Spain.
| | - Antonio Alcaraz
- Laboratory of Molecular Biophysics, Department of Physics, University Jaume I, Castellón 12071, Spain.
| |
Collapse
|
3
|
Sun F, Dong B, Zhang H, Tian M. Permeability-Controlled Probe for Ratiometric Detection of Plasma Membrane Integrity and Late Apoptosis. ACS Sens 2024. [PMID: 39460734 DOI: 10.1021/acssensors.4c01963] [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: 10/28/2024]
Abstract
The destruction of plasma membrane integrity is closely related to immune response, neuronal injury, cell apoptosis, and other pathological events. However, the construction of ratiometric fluorescent probes capable of detecting plasma membrane integrity remains a significant challenge, hindering in-depth studies on related biomedical areas. Herein, a polarity-responsive fluorescent probe was constructed for the ratiometric detection of cell membrane integrity for the first time. The probe targeted intact plasma membranes in healthy cells and relocated into the cytoplasm to give significantly red-shifted fluorescence after plasma membrane damage. Molecular simulations revealed that the high transmembrane barrier and amphipathic nature of the probe were responsible for its targeting ability. With the probe, the ratiometric detection of late apoptosis stage was realized for the first time, and the membrane damage of tumor cells induced by UV irradiation, toxins, and antitumor drugs was visualized. The effect of formaldehyde on membrane integrity was evaluated using a probe, and cysteine was proved to be a potential detoxifier to counteract the toxicity of formaldehyde.
Collapse
Affiliation(s)
- Fengkai Sun
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, People's Republic of China
| | - Baoli Dong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, People's Republic of China
| | - Heng Zhang
- Key Lab of Colloid and Interface Chemistry, Shandong University, Jinan, Shandong 250100, PR China
- Shandong Chambroad Holding Co., Ltd. Binzhou, Shandong 256500, PR China
| | - Minggang Tian
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, People's Republic of China
| |
Collapse
|
4
|
Gorbushin A, Ruparčič M, Anderluh G. Littoporins: Novel actinoporin-like proteins in caenogastropod genus Littorina. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109698. [PMID: 38871141 DOI: 10.1016/j.fsi.2024.109698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
In the course of searching for genes controlling the immune system in caenogastropod mollusks, we characterized and phylogenetically placed five new actinoporin-like cytolysins expressed in periwinkles of the genus Littorina. These newly discovered proteins, named littoporins (LitP), contain a central cytolysin/lectin domain and exhibit a predicted protein fold that is almost identical to the three-dimensional structures of actinoporins. Two of these proteins, LitP-1 and LitP-2, were found to be upregulated in L. littorea kidney tissues and immune cells in response to natural and experimental infection with the trematode Himasthla elongata, suggesting their potential role as perforins in the systemic anti-trematode immune response. The primary sequence divergence of littoporins is hypothesized to be attributed to the taxonomic range of cell membranes they can recognize and permeabilize.
Collapse
Affiliation(s)
- Alexander Gorbushin
- Sechenov Institute of Evolutionary Physiology and Biochemistry (IEPhB RAS), St Petersburg, Russia.
| | - Matija Ruparčič
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| |
Collapse
|
5
|
Xu N, Kahn TW, Jacob T, Liu Y. Graphical models for identifying pore-forming proteins. Proteins 2024; 92:975-983. [PMID: 38618860 DOI: 10.1002/prot.26687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 02/22/2024] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
Pore-forming toxins (PFTs) are proteins that form lesions in biological membranes. Better understanding of the structure and function of these proteins will be beneficial in a number of biotechnological applications, including the development of new pest control methods in agriculture. When searching for new pore formers, existing sequence homology-based methods fail to discover truly novel proteins with low sequence identity to known proteins. Search methodologies based on protein structures would help us move beyond this limitation. As the number of known structures for PFTs is very limited, it's quite challenging to identify new proteins having similar structures using computational approaches like deep learning. In this article, we therefore propose a sample-efficient graphical model, where a protein structure graph is first constructed according to consensus secondary structures. A semi-Markov conditional random fields model is then developed to perform protein sequence segmentation. We demonstrate that our method is able to distinguish structurally similar proteins even in the absence of sequence similarity (pairwise sequence identity < 0.4)-a feat not achievable by traditional approaches like HMMs. To extract proteins of interest from a genome-wide protein database for further study, we also develop an efficient framework for UniRef50 with 43 million proteins.
Collapse
Affiliation(s)
- Nan Xu
- Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Theodore W Kahn
- BASF Corporation, Research Triangle Park, North Carolina, USA
| | - Theju Jacob
- BASF Corporation, Research Triangle Park, North Carolina, USA
| | - Yan Liu
- Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Hashimoto S, Matsuo K. Dynamic Observation of the Membrane Interaction Processes of β-Lactoglobulin by Time-Resolved Vacuum-Ultraviolet Circular Dichroism. Anal Chem 2024; 96:10524-10533. [PMID: 38907695 DOI: 10.1021/acs.analchem.4c00556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
The elucidation of protein-membrane interactions is pivotal for comprehending the mechanisms underlying diverse biological phenomena and membrane-related diseases. In this investigation, vacuum-ultraviolet circular dichroism (VUVCD) spectroscopy, utilizing synchrotron radiation (SR), was employed to dynamically observe membrane interaction processes involving water-soluble proteins at the secondary-structure level. The study utilized a time-resolved (TR) T-shaped microfluidic cell, facilitating the rapid and efficient mixing of protein and membrane solutions. This system was instrumental in acquiring measurements of the time-resolved circular dichroism (TRCD) spectra of β-lactoglobulin (bLG) during its interaction with lysoDMPG micelles. The results indicate that bLG undergoes a β-α conformation change, leading to the formation of the membrane-interacting state (M-state), with structural alterations occurring in more than two steps. Global fitting analysis, employing biexponential functions with all of the TRCD spectral data sets, yielded two distinct rate constants (0.18 ± 0.01 and 0.06 ± 0.003/s) and revealed a unique spectrum corresponding to an intermediate state (I-state). Secondary-structure analysis of bLG in its native (N-, I-, and M-states) highlighted that structural changes from the N- to I-states predominantly occurred in the N- and C-terminal regions, which were prominently exposed to the membrane. Meanwhile, transitions from the I- to M-states extended into the inner barrel regions of bLG. Further examination of the physical properties of α-helical segments, such as effective charge and hydrophobicity, revealed that the N- to I- and I- to M-state transitions, which are ascribed to first- and second-rate constants, respectively, are primarily driven by electrostatic and hydrophobic interactions, respectively. These findings underscore the capability of the TR-VUVCD system as a robust tool for characterizing protein-membrane interactions at the molecular level.
Collapse
Affiliation(s)
- Satoshi Hashimoto
- Graduate School of Advanced Science and Engineering, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8526, Japan
| | - Koichi Matsuo
- Graduate School of Advanced Science and Engineering, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8526, Japan
- Research Institute for Synchrotron Radiation Science, Hiroshima University, 2-313 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM2), Hiroshima University, 2-313 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
| |
Collapse
|
7
|
Lyu L, Jia H, Liu Q, Ma W, Li Z, Pan L, Zhang X. Individualized lipid profile in urine-derived extracellular vesicles from clinical patients with Mycobacterium tuberculosis infections. Front Microbiol 2024; 15:1409552. [PMID: 38873163 PMCID: PMC11169924 DOI: 10.3389/fmicb.2024.1409552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Background Lipids are a key nutrient source for the growth and reproduction of Mycobacterium tuberculosis (Mtb). Urine-derived extracellular vesicles (EVs), because of its non-invasive sampling, lipid enrichment, and specific sorting character, have been recognized as a promising research target for biomarker discovery and pathogenesis elucidation in tuberculosis (TB). We aim to profile lipidome of Mtb-infected individuals, offer novel lipid signatures for the development of urine-based TB testing, and provide new insights into the lipid metabolism after Mtb infection. Methods Urine-derived extracellular vesicles from 41 participants (including healthy, pulmonary tuberculosis, latent tuberculosis patients, and other lung disease groups) were isolated and individually detected using targeted lipidomics and proteomics technology platforms. Biomarkers were screened by multivariate and univariate statistical analysis and evaluated by SPSS software. Correlation analyses were performed on lipids and proteins using the R Hmisc package. Results Overall, we identified 226 lipids belonging to 14 classes. Of these, 7 potential lipid biomarkers for TB and 6 for latent TB infection (LTBI) were identified, all of which were classified into diacylglycerol (DAG), monoacylglycerol (MAG), free fatty acid (FFA), and cholesteryl ester (CE). Among them, FFA (20:1) was the most promising biomarker target in diagnosing TB/LTBI from other compared groups and also have great diagnostic performance in distinguishing TB from LTBI with AUC of 0.952. In addition, enhanced lipolysis happened as early as individuals got latent Mtb infection, and ratio of raft lipids was gradually elevated along TB progression. Conclusion This study demonstrated individualized lipid profile of urinary EVs in patients with Mtb infection, revealed novel potential lipid biomarkers for TB/LTBI diagnosis, and explored mechanisms by which EV lipid raft-dependent bio-processes might affect pathogenesis. It lays a solid foundation for the subsequent diagnosis and therapeutic intervention of TB.
Collapse
Affiliation(s)
- Lingna Lyu
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Hongyan Jia
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Qiuyue Liu
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Wenxia Ma
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zihui Li
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Liping Pan
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital Affiliated to Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiuli Zhang
- The Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, China
| |
Collapse
|
8
|
Nagel AS, Vetrova OS, Rudenko NV, Karatovskaya AP, Zamyatina AV, Andreeva-Kovalevskaya ZI, Salyamov VI, Egorova NA, Siunov AV, Ivanova TD, Boziev KM, Brovko FA, Solonin AS. A High-Homology Region Provides the Possibility of Detecting β-Barrel Pore-Forming Toxins from Various Bacterial Species. Int J Mol Sci 2024; 25:5327. [PMID: 38791367 PMCID: PMC11120785 DOI: 10.3390/ijms25105327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024] Open
Abstract
The pathogenicity of many bacteria, including Bacillus cereus and Staphylococcus aureus, depends on pore-forming toxins (PFTs), which cause the lysis of host cells by forming pores in the membranes of eukaryotic cells. Bioinformatic analysis revealed a region homologous to the Lys171-Gly250 sequence in hemolysin II (HlyII) from B. cereus in over 600 PFTs, which we designated as a "homologous peptide". Three β-barrel PFTs were used for a detailed comparative analysis. Two of them-HlyII and cytotoxin K2 (CytK2)-are synthesized in Bacillus cereus sensu lato; the third, S. aureus α-toxin (Hla), is the most investigated representative of the family. Protein modeling showed certain amino acids of the homologous peptide to be located on the surface of the monomeric forms of these β-barrel PFTs. We obtained monoclonal antibodies against both a cloned homologous peptide and a 14-membered synthetic peptide, DSFNTFYGNQLFMK, as part of the homologous peptide. The HlyII, CytK2, and Hla regions recognized by the obtained antibodies, as well as an antibody capable of suppressing the hemolytic activity of CytK2, were identified in the course of this work. Antibodies capable of recognizing PFTs of various origins can be useful tools for both identification and suppression of the cytolytic activity of PFTs.
Collapse
Affiliation(s)
- Alexey S. Nagel
- FSBIS FRC Pushchino Scientific Centre of Biological Research, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 5 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (A.S.N.); (Z.I.A.-K.); (V.I.S.); (A.V.S.); (T.D.I.); (A.S.S.)
| | - Olesya S. Vetrova
- Pushchino Branch, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (O.S.V.); (A.P.K.); (A.V.Z.); (K.M.B.); (F.A.B.)
| | - Natalia V. Rudenko
- Pushchino Branch, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (O.S.V.); (A.P.K.); (A.V.Z.); (K.M.B.); (F.A.B.)
| | - Anna P. Karatovskaya
- Pushchino Branch, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (O.S.V.); (A.P.K.); (A.V.Z.); (K.M.B.); (F.A.B.)
| | - Anna V. Zamyatina
- Pushchino Branch, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (O.S.V.); (A.P.K.); (A.V.Z.); (K.M.B.); (F.A.B.)
| | - Zhanna I. Andreeva-Kovalevskaya
- FSBIS FRC Pushchino Scientific Centre of Biological Research, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 5 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (A.S.N.); (Z.I.A.-K.); (V.I.S.); (A.V.S.); (T.D.I.); (A.S.S.)
| | - Vadim I. Salyamov
- FSBIS FRC Pushchino Scientific Centre of Biological Research, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 5 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (A.S.N.); (Z.I.A.-K.); (V.I.S.); (A.V.S.); (T.D.I.); (A.S.S.)
| | - Nadezhda A. Egorova
- Federal State Budgetary Educational Institution of Higher Education “Ryazan State University Named for S.A. Yesenin”, 46 st. Svobody, 390000 Ryazan, Ryazan Region, Russia;
| | - Alexander V. Siunov
- FSBIS FRC Pushchino Scientific Centre of Biological Research, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 5 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (A.S.N.); (Z.I.A.-K.); (V.I.S.); (A.V.S.); (T.D.I.); (A.S.S.)
| | - Tatiana D. Ivanova
- FSBIS FRC Pushchino Scientific Centre of Biological Research, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 5 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (A.S.N.); (Z.I.A.-K.); (V.I.S.); (A.V.S.); (T.D.I.); (A.S.S.)
| | - Khanafi M. Boziev
- Pushchino Branch, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (O.S.V.); (A.P.K.); (A.V.Z.); (K.M.B.); (F.A.B.)
| | - Fedor A. Brovko
- Pushchino Branch, Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (O.S.V.); (A.P.K.); (A.V.Z.); (K.M.B.); (F.A.B.)
| | - Alexander S. Solonin
- FSBIS FRC Pushchino Scientific Centre of Biological Research, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Russian Academy of Sciences, 5 Prospekt Nauki, 142290 Pushchino, Moscow Region, Russia; (A.S.N.); (Z.I.A.-K.); (V.I.S.); (A.V.S.); (T.D.I.); (A.S.S.)
| |
Collapse
|
9
|
Peigneur S, Tibery D, Tytgat J. The Helix Ring Peptide U 11 from the Venom of the Ant, Tetramorium bicarinatum, Acts as a Putative Pore-Forming Toxin. MEMBRANES 2024; 14:114. [PMID: 38786948 PMCID: PMC11123039 DOI: 10.3390/membranes14050114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
An insect neuroactive helix ring peptide called U11-MYRTX-Tb1a (abbreviated as U11) from the venom of the ant, Tetramorium bicarinatum. U11 is a 34-amino-acid peptide that is claimed to be one of the most paralytic peptides ever reported from ant venoms acting against blowflies and honeybees. The peptide features a compact triangular ring helix structure stabilized by a single disulfide bond, which is a unique three-dimensional scaffold among animal venoms. Pharmacological assays using Drosophila S2 cells have demonstrated that U11 is not cytotoxic but instead suggest that it may modulate potassium channels via the presence of a functional dyad. In our work described here, we have tested this hypothesis by investigating the action of synthetically made U11 on a wide array of voltage-gated K and Na channels since it is well known that these channels play a crucial role in the phenomenon of paralysis. Using the Xenopus laevis oocyte heterologous expression system and voltage clamp, our results have not shown any modulatory effect of 1 μM U11 on the activity of Kv1.1, Kv1.3, Kv1.4, Kv1.5, Shaker IR, Kv4.2, Kv7.1, Kv10.1, Kv11.1 and KQT1, nor on DmNav and BgNav. Instead, 10 μM U11 caused a quick and irreversible cytolytic effect, identical to the cytotoxic effect caused by Apis mellifera venom, which indicates that U11 can act as a pore-forming peptide. Interestingly, the paralytic dose (PD50) on blowflies and honeybees corresponds with the concentration at which U11 displays clear pore-forming activity. In conclusion, our results indicate that the insecticidal and paralytic effects caused by U11 may be explained by the putative pore formation of the peptide.
Collapse
Affiliation(s)
- Steve Peigneur
- Toxicology and Pharmacology, University of Leuven (KU Leuven), P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium; (S.P.); (D.T.)
| | - Diogo Tibery
- Toxicology and Pharmacology, University of Leuven (KU Leuven), P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium; (S.P.); (D.T.)
- Laboratory of Neuropharmacology, Department of Physiological Sciences, University of Brasília, Distrito Federal, Brasília 70910-900, Brazil
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), P.O. Box 922, Herestraat 49, 3000 Leuven, Belgium; (S.P.); (D.T.)
| |
Collapse
|
10
|
Zafar H, Saier MHH. An Insider's Perspective about the Pathogenic Relevance of Gut Bacterial Transportomes. Microb Physiol 2024; 34:133-141. [PMID: 38636461 PMCID: PMC11283328 DOI: 10.1159/000538779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/04/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND The gut microbiome is integral to host health, hosting complex interactions between the host and numerous microbial species in the gastrointestinal tract. Key among the molecular mechanisms employed by gut bacteria are transportomes, consisting of diverse transport proteins crucial for bacterial adaptation to the dynamic, nutrient-rich environment of the mammalian gut. These transportomes facilitate the movement of a wide array of molecules, impacting both the host and the microbial community. SUMMARY This communication explores the significance of transportomes in gut bacteria, focusing on their role in nutrient acquisition, competitive interactions among microbes, and potential pathogenicity. It delves into the transportomes of key gut bacterial species like E. coli, Salmonella, Bacteroides, Lactobacillus, Clostridia, and Bifidobacterium, examining the functions of predicted transport proteins. The overview synthesizes recent research efforts, highlighting how these transportomes influence host-microbe interactions and contribute to the microbial ecology of the gut. KEY MESSAGES Transportomes are vital for the survival and adaptation of bacteria in the gut, enabling the import and export of various nutrients and molecules. The complex interplay of transport proteins not only supports bacterial growth and competition but also has implications for host health, potentially contributing to pathogenic processes. Understanding the pathogenic potential of transportomes in major gut bacterial species provides insights into gut health and disease, offering avenues for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Hassan Zafar
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Milton H. Herman Saier
- Department of Molecular Biology, School of Biological Sciences, University of California at San Diego, La Jolla, California, 92093-0116, USA
| |
Collapse
|
11
|
Volarić J, van der Heide NJ, Mutter NL, Samplonius DF, Helfrich W, Maglia G, Szymanski W, Feringa BL. Visible Light Control over the Cytolytic Activity of a Toxic Pore-Forming Protein. ACS Chem Biol 2024; 19:451-461. [PMID: 38318850 PMCID: PMC10877574 DOI: 10.1021/acschembio.3c00640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Enabling control over the bioactivity of proteins with light, along with the principles of photopharmacology, has the potential to generate safe and targeted medical treatments. Installing light sensitivity in a protein can be achieved through its covalent modification with a molecular photoswitch. The general challenge in this approach is the need for the use of low energy visible light for the regulation of bioactivity. In this study, we report visible light control over the cytolytic activity of a protein. A water-soluble visible-light-operated tetra-ortho-fluoro-azobenzene photoswitch was synthesized by utilizing the nucleophilic aromatic substitution reaction for installing a solubilizing sulfonate group onto the electron-poor photoswitch structure. The azobenzene was attached to two cysteine mutants of the pore-forming protein fragaceatoxin C (FraC), and their respective activities were evaluated on red blood cells. For both mutants, the green-light-irradiated sample, containing predominantly the cis-azobenzene isomer, was more active compared to the blue-light-irradiated sample. Ultimately, the same modulation of the cytolytic activity pattern was observed toward a hypopharyngeal squamous cell carcinoma. These results constitute the first case of using low energy visible light to control the biological activity of a toxic protein.
Collapse
Affiliation(s)
- Jana Volarić
- Stratingh
Institute for Organic Chemistry, University
of Groningen, 9747 AG Groningen, The Netherlands
| | - Nieck J. van der Heide
- Groningen
Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Natalie L. Mutter
- Groningen
Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Douwe F. Samplonius
- Department
of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Wijnand Helfrich
- Department
of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Giovanni Maglia
- Groningen
Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Wiktor Szymanski
- Stratingh
Institute for Organic Chemistry, University
of Groningen, 9747 AG Groningen, The Netherlands
- Department
of Radiology, Medical Imaging Center, University
of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Ben L. Feringa
- Stratingh
Institute for Organic Chemistry, University
of Groningen, 9747 AG Groningen, The Netherlands
| |
Collapse
|
12
|
Lecaudey LA, Netzer R, Wibberg D, Busche T, Bloecher N. Metatranscriptome analysis reveals the putative venom toxin repertoire of the biofouling hydroid Ectopleura larynx. Toxicon 2024; 237:107556. [PMID: 38072317 DOI: 10.1016/j.toxicon.2023.107556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023]
Abstract
Cnidarians thriving in biofouling communities on aquaculture net pens represent a significant health risk for farmed finfish due to their stinging cells. The toxins coming into contact with the fish, during net cleaning, can adversely affect their behavior, welfare, and survival, with a particularly serious health risk for the gills, causing direct tissue damage such as formation of thrombi and increasing risks of secondary infections. The hydroid Ectopleura larynx is one of the most common fouling organisms in Northern Europe. However, despite its significant economic, environmental, and operational impact on finfish aquaculture, biological information on this species is scarce and its venom composition has never been investigated. In this study, we generated a whole transcriptome of E. larynx, and identified its putative expressed venom toxin proteins (predicted toxin proteins, not functionally characterized) based on in silico transcriptome annotation mining and protein sequence analysis. The results uncovered a broad and diverse repertoire of putative toxin proteins for this hydroid species. Its toxic arsenal appears to include a wide and complex selection of toxin proteins, covering a large panel of potential biological functions that play important roles in envenomation. The putative toxins identified in this species, such as neurotoxins, GTPase toxins, metalloprotease toxins, ion channel impairing toxins, hemorrhagic toxins, serine protease toxins, phospholipase toxins, pore-forming toxins, and multifunction toxins may cause various major deleterious effects in prey, predators, and competitors. These results provide valuable new insights into the venom composition of cnidarians, and venomous marine organisms in general, and offer new opportunities for further research into novel and valuable bioactive molecules for medicine, agronomics and biotechnology.
Collapse
Affiliation(s)
| | - Roman Netzer
- SINTEF Ocean, Aquaculture Department, Brattørkaia 17c, 7010, Trondheim, Norway
| | - Daniel Wibberg
- Center for Biotechnology (CeBiTec), Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Tobias Busche
- Center for Biotechnology (CeBiTec), Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany; Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany
| | - Nina Bloecher
- SINTEF Ocean, Aquaculture Department, Brattørkaia 17c, 7010, Trondheim, Norway
| |
Collapse
|
13
|
Zhu Z, Hu Z, Li S, Fang R, Ono HK, Hu DL. Molecular Characteristics and Pathogenicity of Staphylococcus aureus Exotoxins. Int J Mol Sci 2023; 25:395. [PMID: 38203566 PMCID: PMC10778951 DOI: 10.3390/ijms25010395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Staphylococcus aureus stands as one of the most pervasive pathogens given its morbidity and mortality worldwide due to its roles as an infectious agent that causes a wide variety of diseases ranging from moderately severe skin infections to fatal pneumonia and sepsis. S. aureus produces a variety of exotoxins that serve as important virulence factors in S. aureus-related infectious diseases and food poisoning in both humans and animals. For example, staphylococcal enterotoxins (SEs) produced by S. aureus induce staphylococcal foodborne poisoning; toxic shock syndrome toxin-1 (TSST-1), as a typical superantigen, induces toxic shock syndrome; hemolysins induce cell damage in erythrocytes and leukocytes; and exfoliative toxin induces staphylococcal skin scalded syndrome. Recently, Panton-Valentine leucocidin, a cytotoxin produced by community-associated methicillin-resistant S. aureus (CA-MRSA), has been reported, and new types of SEs and staphylococcal enterotoxin-like toxins (SEls) were discovered and reported successively. This review addresses the progress of and novel insights into the molecular structure, biological activities, and pathogenicity of both the classic and the newly identified exotoxins produced by S. aureus.
Collapse
Affiliation(s)
- Zhihao Zhu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zuo Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Shaowen Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Rendong Fang
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China;
| | - Hisaya K. Ono
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Dong-Liang Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| |
Collapse
|
14
|
Has C, Das SL. The Functionality of Membrane-Inserting Proteins and Peptides: Curvature Sensing, Generation, and Pore Formation. J Membr Biol 2023; 256:343-372. [PMID: 37650909 DOI: 10.1007/s00232-023-00289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
Proteins and peptides with hydrophobic and amphiphilic segments are responsible for many biological functions. The sensing and generation of membrane curvature are the functions of several protein domains or motifs. While some specific membrane proteins play an essential role in controlling the curvature of distinct intracellular membranes, others participate in various cellular processes such as clathrin-mediated endocytosis, where several proteins sort themselves at the neck of the membrane bud. A few membrane-inserting proteins form nanopores that permeate selective ions and water to cross the membrane. In addition, many natural and synthetic small peptides and protein toxins disrupt the membrane by inducing nonspecific pores in the membrane. The pore formation causes cell death through the uncontrolled exchange between interior and exterior cellular contents. In this article, we discuss the insertion depth and orientation of protein/peptide helices, and their role as a sensor and inducer of membrane curvature as well as a pore former in the membrane. We anticipate that this extensive review will assist biophysicists to gain insight into curvature sensing, generation, and pore formation by membrane insertion.
Collapse
Affiliation(s)
- Chandra Has
- Department of Chemical Engineering, GSFC University, Vadodara, 391750, Gujarat, India.
| | - Sovan Lal Das
- Physical and Chemical Biology Laboratory and Department of Mechanical Engineering, Indian Institute of Technology, Palakkad, 678623, Kerala, India
| |
Collapse
|
15
|
Li Y, Chen R, Wang C, Deng J, Luo S. Double-edged functions of hemopexin in hematological related diseases: from basic mechanisms to clinical application. Front Immunol 2023; 14:1274333. [PMID: 38022615 PMCID: PMC10653390 DOI: 10.3389/fimmu.2023.1274333] [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: 08/08/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
It is now understood that hemolysis and the subsequent release of heme into circulation play a critical role in driving the progression of various diseases. Hemopexin (HPX), a heme-binding protein with the highest affinity for heme in plasma, serves as an effective antagonist against heme toxicity resulting from severe acute or chronic hemolysis. In the present study, changes in HPX concentration were characterized at different stages of hemolytic diseases, underscoring its potential as a biomarker for assessing disease progression and prognosis. In many heme overload-driven conditions, such as sickle cell disease, transfusion-induced hemolysis, and sepsis, endogenous HPX levels are often insufficient to provide protection. Consequently, there is growing interest in developing HPX therapeutics to mitigate toxic heme exposure. Strategies include HPX supplementation when endogenous levels are depleted and enhancing HPX's functionality through modifications, offering a potent defense against heme toxicity. It is worth noting that HPX may also exert deleterious effects under certain circumstances. This review aims to provide a comprehensive overview of HPX's roles in the progression and prognosis of hematological diseases. It highlights HPX-based clinical therapies for different hematological disorders, discusses advancements in HPX production and modification technologies, and offers a theoretical basis for the clinical application of HPX.
Collapse
Affiliation(s)
| | | | | | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Jalali E, Bel Y, Maghsoudi S, Noroozian E, Escriche B. Enhancing insecticidal efficacy of Bacillus thuringiensis Cry1Ab through pH-sensitive encapsulation. Appl Microbiol Biotechnol 2023; 107:6407-6419. [PMID: 37632523 PMCID: PMC10560195 DOI: 10.1007/s00253-023-12723-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/14/2023] [Accepted: 08/06/2023] [Indexed: 08/28/2023]
Abstract
Nanotechnology is a promising way to enhance the stability of Bacillus thuringiensis (Bt) insecticidal proteins under environmental conditions. In this work, two emulsions were prepared through the Pickering emulsion technique, stabilized by Cu2+-SQDs/S-CN nanocomposites and by GO nanosheets. In addition, a pH-sensitive polymer was incorporated into these emulsions, allowing the Bt protein, Cry1Ab, to be released in an alkaline pH environment, as it occurs in the lepidopteran pests' gut. The effectiveness of these two nanomaterials in protecting Cry1Ab from degradation, and therefore enhancing its pesticidal activity, was assessed by exposing samples of the purified unprotected protein and encapsulated protein to high-intensity UV light and 40°C temperature treatments. The UV treatment results were evaluated using SDS-PAGE analysis and pointed out that Cry1Ab could be structurally protected by the emulsions. The bioassays with first instar larvae of the lepidopteran pest Ostrinia nubilalis confirm the nanomaterial protection to UV and temperature treatments, i.e., decreasing about half the degradation rate and increasing up to 12-fold the residual activity after UV treatment. Our results indicate that encapsulation could be an effective strategy to improve the effectiveness of Cry1Ab under environmental conditions. KEY POINTS: • Pickering emulsions are effective for solubilized Cry1Ab encapsulation. • Structural and toxicity Cry1Ab properties are enhanced by pH-sensitive encapsulation. • Cu2+-SQDs/S-CN and GO nanomaterials improve the efficacy of Bt insecticides.
Collapse
Affiliation(s)
- Elham Jalali
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, 76169-14111 Iran
- Instituto BioTecMed, Department of Genetics, University of Valencia, 46100 Burjassot, Valencia Spain
| | - Yolanda Bel
- Instituto BioTecMed, Department of Genetics, University of Valencia, 46100 Burjassot, Valencia Spain
| | - Shahab Maghsoudi
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, 76169-14111 Iran
| | - Ebrahim Noroozian
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, 76169-14111 Iran
| | - Baltasar Escriche
- Instituto BioTecMed, Department of Genetics, University of Valencia, 46100 Burjassot, Valencia Spain
| |
Collapse
|
17
|
Yadav GP, Wang H, Ouwendijk J, Cross S, Wang Q, Qin F, Verkade P, Zhu MX, Jiang QX. Chromogranin B (CHGB) is dimorphic and responsible for dominant anion channels delivered to cell surface via regulated secretion. Front Mol Neurosci 2023; 16:1205516. [PMID: 37435575 PMCID: PMC10330821 DOI: 10.3389/fnmol.2023.1205516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/26/2023] [Indexed: 07/13/2023] Open
Abstract
Regulated secretion is conserved in all eukaryotes. In vertebrates granin family proteins function in all key steps of regulated secretion. Phase separation and amyloid-based storage of proteins and small molecules in secretory granules require ion homeostasis to maintain their steady states, and thus need ion conductances in granule membranes. But granular ion channels are still elusive. Here we show that granule exocytosis in neuroendocrine cells delivers to cell surface dominant anion channels, to which chromogranin B (CHGB) is critical. Biochemical fractionation shows that native CHGB distributes nearly equally in soluble and membrane-bound forms, and both reconstitute highly selective anion channels in membrane. Confocal imaging resolves granular membrane components including proton pumps and CHGB in puncta on the cell surface after stimulated exocytosis. High pressure freezing immuno-EM reveals a major fraction of CHGB at granule membranes in rat pancreatic β-cells. A cryo-EM structure of bCHGB dimer of a nominal 3.5 Å resolution delineates a central pore with end openings, physically sufficient for membrane-spanning and large single channel conductance. Together our data support that CHGB-containing (CHGB+) channels are characteristic of regulated secretion, and function in granule ion homeostasis near the plasma membrane or possibly in other intracellular processes.
Collapse
Affiliation(s)
- Gaya P. Yadav
- Departments of Microbiology and Cell Science and of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
- Departments of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, United States
- Laboratory of Molecular Physiology and Biophysics, Hauptman-Woodward Medical Research Institute, Buffalo, NY, United States
| | - Haiyuan Wang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Joke Ouwendijk
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Stephen Cross
- Wolfson Bioimaging facility, University of Bristol, Bristol, United Kingdom
| | - Qiaochu Wang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Feng Qin
- Departments of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, United States
| | - Paul Verkade
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Qiu-Xing Jiang
- Departments of Microbiology and Cell Science and of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
- Departments of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, United States
- Laboratory of Molecular Physiology and Biophysics, Hauptman-Woodward Medical Research Institute, Buffalo, NY, United States
- Cryo-EM Center, Laoshan Laboratory, Qingdao, Shandong, China
| |
Collapse
|
18
|
Liu LZ, Liu L, Shi ZH, Bian XL, Si ZR, Wang QQ, Xiang Y, Zhang Y. Amphibian pore-forming protein βγ-CAT drives extracellular nutrient scavenging under cell nutrient deficiency. iScience 2023; 26:106598. [PMID: 37128610 PMCID: PMC10148134 DOI: 10.1016/j.isci.2023.106598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 02/22/2023] [Accepted: 03/30/2023] [Indexed: 05/03/2023] Open
Abstract
Nutrient acquisition is essential for animal cells. βγ-CAT is a pore-forming protein (PFP) and trefoil factor complex assembled under tight regulation identified in toad Bombina maxima. Here, we reported that B. maxima cells secreted βγ-CAT under glucose, glutamine, and pyruvate deficiency to scavenge extracellular proteins for their nutrient supply and survival. AMPK signaling positively regulated the expression and secretion of βγ-CAT. The PFP complex selectively bound extracellular proteins and promoted proteins uptake through endolysosomal pathways. Elevated intracellular amino acids, enhanced ATP production, and eventually prolonged cell survival were observed in the presence of βγ-CAT and extracellular proteins. Liposome assays indicated that high concentration of ATP negatively regulated the opening of βγ-CAT channels. Collectively, these results uncovered that βγ-CAT is an essential element in cell nutrient scavenging under cell nutrient deficiency by driving vesicular uptake of extracellular proteins, providing a new paradigm for PFPs in cell nutrient acquisition and metabolic flexibility.
Collapse
Affiliation(s)
- Ling-Zhen Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Long Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Human Aging Research Institute (HARI) and School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Zhi-Hong Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xian-Ling Bian
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- School of Life Science, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zi-Ru Si
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- School of Life Science, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qi-Quan Wang
- Human Aging Research Institute (HARI) and School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yang Xiang
- Human Aging Research Institute (HARI) and School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
- Corresponding author
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Corresponding author
| |
Collapse
|
19
|
Pacheco S, Gómez I, Peláez-Aguilar AE, Verduzco-Rosas LA, García-Suárez R, do Nascimento NA, Rivera-Nájera LY, Cantón PE, Soberón M, Bravo A. Structural changes upon membrane insertion of the insecticidal pore-forming toxins produced by Bacillus thuringiensis. FRONTIERS IN INSECT SCIENCE 2023; 3:1188891. [PMID: 38469496 PMCID: PMC10926538 DOI: 10.3389/finsc.2023.1188891] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 04/04/2023] [Indexed: 03/13/2024]
Abstract
Different Bacillus thuringiensis (Bt) strains produce a broad variety of pore-forming toxins (PFTs) that show toxicity against insects and other invertebrates. Some of these insecticidal PFT proteins have been used successfully worldwide to control diverse insect crop pests. There are several studies focused on describing the mechanism of action of these toxins that have helped to improve their performance and to cope with the resistance evolved by different insects against some of these proteins. However, crucial information that is still missing is the structure of pores formed by some of these PFTs, such as the three-domain crystal (Cry) proteins, which are the most commercially used Bt toxins in the biological control of insect pests. In recent years, progress has been made on the identification of the structural changes that certain Bt insecticidal PFT proteins undergo upon membrane insertion. In this review, we describe the models that have been proposed for the membrane insertion of Cry toxins. We also review the recently published structures of the vegetative insecticidal proteins (Vips; e.g. Vip3) and the insecticidal toxin complex (Tc) in the membrane-inserted state. Although different Bt PFTs show different primary sequences, there are some similarities in the three-dimensional structures of Vips and Cry proteins. In addition, all PFTs described here must undergo major structural rearrangements to pass from a soluble form to a membrane-inserted state. It is proposed that, despite their structural differences, all PFTs undergo major structural rearrangements producing an extended α-helix, which plays a fundamental role in perforating their target membrane, resulting in the formation of the membrane pore required for their insecticidal activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alejandra Bravo
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
20
|
Bian X, Si Z, Wang Q, Liu L, Shi Z, Tian C, Lee W, Zhang Y. IgG Fc-binding protein positively regulates the assembly of pore-forming protein complex βγ-CAT evolved to drive cell vesicular delivery and transport. J Biol Chem 2023; 299:104717. [PMID: 37068610 DOI: 10.1016/j.jbc.2023.104717] [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: 09/15/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023] Open
Abstract
Cell membranes form barriers for molecule exchange between the cytosol and the extracellular environments. βγ-CAT, a complex of pore-forming protein (PFP) BmALP1 (two βγ-crystallin domains with an aerolysin pore-forming domain) and the trefoil factor BmTFF3, has been identified in toad Bombina maxima. It plays pivotal roles, via inducing channel formation in various intra- or extra- cellular vesicles, as well as in nutrient acquisition, maintaining water balance, and antigen presentation. Thus, such a protein machine should be tightly regulated. Indeed, BmALP3 (a paralog of BmALP1) oxidizes BmALP1 to form a water-soluble polymer, leading to dissociation of the βγ-CAT complex and loss of biological activity. Here, we found that the B. maxima IgG Fc-binding protein (FCGBP), a well-conserved vertebrate mucin-like protein with unknown functions, acted as a positive regulator for βγ-CAT complex assembly. The interactions among FCGBP, BmALP1, and BmTFF3 were revealed by co-immunoprecipitation assays. Interestingly, FCGBP reversed the inhibitory effect of BmALP3 on the βγ-CAT complex. Furthermore, FCGBP reduced BmALP1 polymers and facilitated the assembly of βγ-CAT with the biological pore-forming activity in the presence of BmTFF3. Our findings define the role of FCGBP in mediating the assembly of a PFP machine evolved to drive cell vesicular delivery and transport.
Collapse
Affiliation(s)
- Xianling Bian
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ziru Si
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Qiquan Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Human Aging Research Institute (HARI) and School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Lingzhen Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Zhihong Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Changlin Tian
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Wenhui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
21
|
Pacheco S, Gómez I, Soberón M, Bravo A. A major conformational change of N-terminal helices of Bacillus thuringiensis Cry1Ab insecticidal protein is necessary for membrane insertion and toxicity. FEBS J 2022; 290:2692-2705. [PMID: 36560841 DOI: 10.1111/febs.16710] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/01/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Pore forming toxins rely on oligomerization for membrane insertion to kill their targets. Bacillus thuringiensis produces insecticidal Cry-proteins composed of three domains that form pores that kill the insect larvae. Domain I is involved in oligomerization and membrane insertion, whereas Domains II and III participate in receptor binding and specificity. However, the structural changes involved in membrane insertion of these proteins remain unsolved. The most widely accepted model for membrane insertion, the 'umbrella model', proposed that the α-4/α-5 hairpin of Domain I swings away and is inserted into the membrane. To determine the topology of Cry1Ab in the membrane, disulfide bonds linking α-helices of Domain I were introduced to restrict their movement. Disulfide bonds between helices α-2/α-3 or α-3/α-4 lost oligomerization and toxicity, indicating that movement of these helices is needed for insecticidal activity. By contrast, disulfide bonds linking helices α-5/α-6 did not affect toxicity, which contradicts the 'umbrella model'. Additionally, Föster resonance energy transfer closest approach analyses measuring distances of different points in the toxin to the membrane plane and collisional quenching assays analysing the protection of specific fluorescent-labeled residues to the soluble potassium iodide quencher in the membrane inserted state were performed. Overall, the data show that Domain I from Cry1Ab may undergo a major conformational change during its membrane insertion, where the N-terminal region (helices α-1 to α-4) participates in oligomerization and toxicity, probably forming an extended helix. These data break a paradigm, showing a new 'folding white-cane model', which better explains the structural changes of Cry toxins during insertion into the membrane.
Collapse
Affiliation(s)
- Sabino Pacheco
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| | - Isabel Gómez
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| | - Mario Soberón
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| | - Alejandra Bravo
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| |
Collapse
|
22
|
The mechanisms of chromogranin B-regulated Cl- homeostasis. Biochem Soc Trans 2022; 50:1659-1672. [PMID: 36511243 DOI: 10.1042/bst20220435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022]
Abstract
Chloride is the most abundant inorganic anions in almost all cells and in human circulation systems. Its homeostasis is therefore important for systems physiology and normal cellular activities. This topic has been extensively studied with chloride loaders and extruders expressed in both cell surfaces and intracellular membranes. With the newly discovered, large-conductance, highly selective Cl- channel formed by membrane-bound chromogranin B (CHGB), which differs from all other known anion channels of conventional transmembrane topology, and is distributed in plasma membranes, endomembrane systems, endosomal, and endolysosomal compartments in cells expressing it, we will discuss the potential physiological importance of the CHGB channels to Cl- homeostasis, cellular excitability and volume control, and cation uptake or release at the cellular and subcellular levels. These considerations and CHGB's association with human diseases make the CHGB channel a possible druggable target for future molecular therapeutics.
Collapse
|
23
|
Delgado A, Benedict C, Macrander J, Daly M. Never, Ever Make an Enemy… Out of an Anemone: Transcriptomic Comparison of Clownfish Hosting Sea Anemone Venoms. Mar Drugs 2022; 20:730. [PMID: 36547877 PMCID: PMC9782873 DOI: 10.3390/md20120730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Sea anemones are predatory marine invertebrates and have diverse venom arsenals. Venom is integral to their biology, and is used in competition, defense, and feeding. Three lineages of sea anemones are known to have independently evolved symbiotic relationships with clownfish, however the evolutionary impact of this relationship on the venom composition of the host is still unknown. Here, we investigate the potential of this symbiotic relationship to shape the venom profiles of the sea anemones that host clownfish. We use transcriptomic data to identify differences and similarities in venom profiles of six sea anemone species, representing the three known clades of clownfish-hosting sea anemones. We recovered 1121 transcripts matching verified toxins across all species, and show that hemolytic and hemorrhagic toxins are consistently the most dominant and diverse toxins across all species examined. These results are consistent with the known biology of sea anemones, provide foundational data on venom diversity of these species, and allow for a review of existing hierarchical structures in venomic studies.
Collapse
Affiliation(s)
- Alonso Delgado
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Charlotte Benedict
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Jason Macrander
- Department of Biology, Florida Southern College, Lakeland, FL 33815, USA
| | - Marymegan Daly
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Palacios-Ortega J, Amigot-Sánchez R, García-Montoya C, Gorše A, Heras-Márquez D, García-Linares S, Martínez-del-Pozo Á, Slotte JP. Determination of the boundary lipids of sticholysins using tryptophan quenching. Sci Rep 2022; 12:17328. [PMID: 36243747 PMCID: PMC9569322 DOI: 10.1038/s41598-022-21750-y] [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: 06/28/2022] [Accepted: 09/30/2022] [Indexed: 01/10/2023] Open
Abstract
Sticholysins are α-pore-forming toxins produced by the sea-anemone Stichodactyla helianthus. These toxins exert their activity by forming pores on sphingomyelin-containing membranes. Recognition of sphingomyelin by sticholysins is required to start the process of pore formation. Sphingomyelin recognition is coupled with membrane binding and followed by membrane penetration and oligomerization. Many features of these processes are known. However, the extent of contact with each of the different kinds of lipids present in the membrane has received little attention. To delve into this question, we have used a phosphatidylcholine analogue labeled at one of its acyl chains with a doxyl moiety, a known quencher of tryptophan emission. Here we present evidence for the contact of sticholysins with phosphatidylcholine lipids in the sticholysin oligomer, and for how each sticholysin isotoxin is affected differently by the inclusion of cholesterol in the membrane. Furthermore, using phosphatidylcholine analogs that were labeled at different positions of their structure (acyl chains and headgroup) in combination with a variety of sticholysin mutants, we also investigated the depth of the tryptophan residues of sticholysins in the bilayer. Our results indicate that the position of the tryptophan residues relative to the membrane normal is deeper when cholesterol is absent from the membrane.
Collapse
Affiliation(s)
- Juan Palacios-Ortega
- grid.13797.3b0000 0001 2235 8415Biochemistry Department, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland ,grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Rafael Amigot-Sánchez
- grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Carmen García-Montoya
- grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Ana Gorše
- grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Diego Heras-Márquez
- grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Sara García-Linares
- grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - Álvaro Martínez-del-Pozo
- grid.4795.f0000 0001 2157 7667Departamento de Bioquímica y Biología Molecular, Universidad Complutense, Madrid, Spain
| | - J. Peter Slotte
- grid.13797.3b0000 0001 2235 8415Biochemistry Department, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| |
Collapse
|
25
|
Four Cholesterol-Recognition Motifs in the Pore-Forming and Translocation Domains of Adenylate Cyclase Toxin Are Essential for Invasion of Eukaryotic Cells and Lysis of Erythrocytes. Int J Mol Sci 2022; 23:ijms23158703. [PMID: 35955837 PMCID: PMC9369406 DOI: 10.3390/ijms23158703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/21/2022] [Accepted: 08/02/2022] [Indexed: 12/05/2022] Open
Abstract
Adenylate Cyclase Toxin (ACT or CyaA) is one of the important virulence factors secreted by Bordetella pertussis, the bacterium causative of whooping cough. ACT debilitates host defenses by production of unregulated levels of cAMP into the cell cytosol upon delivery of its N-terminal domain with adenylate cyclase activity (AC domain) and by forming pores in the plasma membrane of macrophages. Binding of soluble toxin monomers to the plasma membrane of target cells and conversion into membrane-integrated proteins are the first and last step for these toxin activities; however, the molecular determinants in the protein or the target membrane that govern this conversion to an active toxin form are fully unknown. It was previously reported that cytotoxic and cytolytic activities of ACT depend on membrane cholesterol. Here we show that ACT specifically interacts with membrane cholesterol, and find in two membrane-interacting ACT domains, four cholesterol-binding motifs that are essential for AC domain translocation and lytic activities. We hypothesize that direct ACT interaction with membrane cholesterol through those four cholesterol-binding motifs drives insertion and stabilizes the transmembrane topology of several helical elements that ultimately build the ACT structure for AC delivery and pore-formation, thereby explaining the cholesterol-dependence of the ACT activities. The requirement for lipid-mediated stabilization of transmembrane helices appears to be a unifying mechanism to modulate toxicity in pore-forming toxins.
Collapse
|
26
|
Gupta T, Mondal AK, Pani I, Chattopadhyay K, Pal SK. Elucidating liquid crystal-aqueous interface for the study of cholesterol-mediated action of a β-barrel pore forming toxin. SOFT MATTER 2022; 18:5293-5301. [PMID: 35790122 DOI: 10.1039/d2sm00447j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pore-forming toxins (PFTs) produced by pathogenic bacteria serve as prominent virulence factors with potent cell-killing activity. Most of the β-barrel PFTs form transmembrane oligomeric pores in the membrane lipid bilayer in the presence of cholesterol. The pore-formation mechanisms of the PFTs highlight well-orchestrated regulated events in the membrane environment, which involve dramatic changes in the protein structure and organization. Also, concerted crosstalk between protein and membrane lipid components appears to play crucial roles in the process. Membrane-damaging lesions formed by the pore assembly of the PFTs would also be expected to impose drastic alterations in the membrane organization, details of which remain obscure in most of the cases. Prior reports have established that aqueous interfaces of liquid crystals (LCs) offer promise as responsive interfaces for biomolecular events (at physiologically relevant concentrations), which can be visualized as optical signals. Inspired by this, herein, we sought to understand the lipid membrane interactions of a β-barrel PFT i.e., Vibrio cholerae cytolysin (VCC), using LC-aqueous interfaces. Our results show the formation of dendritic patterns upon the addition of VCC to the lipid embedded with cholesterol over the LC film. In contrast, we did not observe any LC reorientation upon the addition of VCC to the lipid-laden LC-aqueous interface in the absence of cholesterol. An array of techniques such as polarizing optical microscopy (POM), atomic force microscopy (AFM), and fluorescence measurements were utilized to decipher the LC response to the lipid interactions of VCC occurring at these interfaces. Altogether, the results obtained from our study provide a novel platform to explore the mechanistic aspects of the protein-membrane interactions, in the process of membrane pore-formation by the membrane-damaging PFTs.
Collapse
Affiliation(s)
- Tarang Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, India.
| | - Anish Kumar Mondal
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, India.
| | - Ipsita Pani
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, India.
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, India.
| | - Santanu Kumar Pal
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, India.
| |
Collapse
|
27
|
Huang Y, Fuller G, Chandran Suja V. Physicochemical characteristics of droplet interface bilayers. Adv Colloid Interface Sci 2022; 304:102666. [PMID: 35429720 DOI: 10.1016/j.cis.2022.102666] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/01/2022]
Abstract
Droplet interface bilayer (DIB) is a lipid bilayer formed when two lipid monolayer-coated aqueous droplets are brought in contact within an oil phase. DIBs, especially post functionalization, are a facile model system to study the biophysics of the cell membrane. Continued advances in enhancing and functionalizing DIBs to be a faithful cell membrane mimetic requires a deep understanding of the physicochemical characteristics of droplet interface bilayers. In this review, we provide a comprehensive overview of the current scientific understanding of DIB characteristics starting with the key experimental frameworks for DIB generation, visualization and functionalization. Subsequently we report experimentally measured physical, electrical and transport characteristics of DIBs across physiologically relevant lipids. Advances in simulations and mathematical modelling of DIBs are also discussed, with an emphasis on revealing principles governing the key physicochemical characteristics. Finally, we conclude the review with important outstanding questions in the field.
Collapse
|
28
|
Wang H, Chen G, Li H. Templated folding of the RTX domain of the bacterial toxin adenylate cyclase revealed by single molecule force spectroscopy. Nat Commun 2022; 13:2784. [PMID: 35589788 PMCID: PMC9120197 DOI: 10.1038/s41467-022-30448-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
The RTX (repeats-in-toxin) domain of the bacterial toxin adenylate cyclase (CyaA) contains five RTX blocks (RTX-i to RTX-v) and its folding is essential for CyaA’s functions. It was shown that the C-terminal capping structure of RTX-v is critical for the whole RTX to fold. However, it is unknown how the folding signal transmits within the RTX domain. Here we use optical tweezers to investigate the interplay between the folding of RTX-iv and RTX-v. Our results show that RTX-iv alone is disordered, but folds into a Ca2+-loaded-β-roll structure in the presence of a folded RTX-v. Folding trajectories of RTX-iv-v reveal that the folding of RTX-iv is strictly conditional upon the folding of RTX-v, suggesting that the folding of RTX-iv is templated by RTX-v. This templating effect allows RTX-iv to fold rapidly, and provides significant mutual stabilization. Our study reveals a possible mechanism for transmitting the folding signal within the RTX domain. The authors use optical tweezers to show that the folding of repeats-in-toxin (RTX) block-iv in adenylate cyclase is templated by the folded RTX block-v. The findings suggest a possible mechanism for transmitting the folding signal in the RTX domain.
Collapse
Affiliation(s)
- Han Wang
- Department of Chemistry, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada.,State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, 300072, Tianjin, P. R. China
| | - Guojun Chen
- Department of Chemistry, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada
| | - Hongbin Li
- Department of Chemistry, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada.
| |
Collapse
|
29
|
Lata K, Singh M, Chatterjee S, Chattopadhyay K. Membrane Dynamics and Remodelling in Response to the Action of the Membrane-Damaging Pore-Forming Toxins. J Membr Biol 2022; 255:161-173. [PMID: 35305136 DOI: 10.1007/s00232-022-00227-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022]
Abstract
Pore-forming protein toxins (PFTs) represent a diverse class of membrane-damaging proteins that are produced by a wide variety of organisms. PFT-mediated membrane perforation is largely governed by the chemical composition and the physical properties of the plasma membranes. The interaction between the PFTs with the target membranes is critical for the initiation of the pore-formation process, and can lead to discrete membrane reorganization events that further aids in the process of pore-formation. Punching holes on the plasma membranes by the PFTs interferes with the cellular homeostasis by disrupting the ion-balance inside the cells that in turn can turn on multiple signalling cascades required to restore membrane integrity and cellular homeostasis. In this review, we discuss the physicochemical attributes of the plasma membranes associated with the pore-formation processes by the PFTs, and the subsequent membrane remodelling events that may start off the membrane-repair mechanisms.
Collapse
Affiliation(s)
- Kusum Lata
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Mahendra Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Shamaita Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India.
| |
Collapse
|
30
|
Yang F, Ye R, Ma C, Wang Y, Wang Y, Chen J, Yang J, Höfer J, Zhu Y, Xiao L, Zhang J, Xu Y. Toxicity evaluation, toxin screening and its intervention of the jellyfish Phacellophora camtschatica based on a combined transcriptome-proteome analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 233:113315. [PMID: 35189521 DOI: 10.1016/j.ecoenv.2022.113315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The application of multi-omics technologies provides a new perspective to solve three main problems including species identification, toxin screening and effective antagonist conformation in the studies of marine toxic jellyfish. METHODS A series of transcriptome-proteome based analysis accompanied with toxicity evaluations were performed for the ornamental jellyfish Phacellophora camtschatica. RESULTS Through combined morphological observation and Cytochrome c oxidase subunit Ⅰ (CO1) molecular alignment, the sample jellyfish was identified as P. camtschatica. A total of 25,747 unigenes and 3058 proteins were obtained from the successfully constructed transcriptome and proteome, in which 6869 (26.68%) and 6618 (25.70%) unigenes, as well as 2536 (82.93%) and 2844 (93.00%) proteins were annotated against the databases of Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), respectively. The jellyfish displayed obvious in vivo lethal effects with significant increases of multi-organ functional indexes as well as in vitro activities. Total of 62 toxins from 120 toxin-related unigenes were screened including 16 metalloproteases, 11 phospholipases and others. Moreover, 11 toxins were further screened by using the erythrocyte model, where the zinc metalloproteinase nas-15-like (1) was the most abundant. Finally, Diltiazem greatly improved the survival rate while EDTA slightly prolonged the survival time in ICR mice. CONCLUSION P. camtschatica is a poisonous jellyfish with diversified toxic components, in which metalloproteinase probably plays an important role in toxicities, and excessive Ca2+ entry may be the main mechanism of systemic lethal toxicity.
Collapse
Affiliation(s)
- Fengling Yang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Ruiwei Ye
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Chaoqun Ma
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Rd, Shanghai 200433, China.
| | - Yichao Wang
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Department of Clinical Laboratory, Taizhou Central Hospital, Taizhou 318000, China.
| | - Yi Wang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jianmei Chen
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jishun Yang
- Medical Insurance Center, Navy Medical Center, Navy Medical Center of PLA, Shanghai 200050, China.
| | - Juan Höfer
- Escuela de Ciencias del Mar, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| | - Yina Zhu
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Liang Xiao
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jing Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China.
| | - Yinghe Xu
- Department of Intensive Care Unit, Taizhou Central Hospital, Taizhou 318000, China.
| |
Collapse
|
31
|
Ulhuq FR, Mariano G. Bacterial pore-forming toxins. MICROBIOLOGY (READING, ENGLAND) 2022; 168:001154. [PMID: 35333704 PMCID: PMC9558359 DOI: 10.1099/mic.0.001154] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 12/11/2022]
Abstract
Pore-forming toxins (PFTs) are widely distributed in both Gram-negative and Gram-positive bacteria. PFTs can act as virulence factors that bacteria utilise in dissemination and host colonisation or, alternatively, they can be employed to compete with rival microbes in polymicrobial niches. PFTs transition from a soluble form to become membrane-embedded by undergoing large conformational changes. Once inserted, they perforate the membrane, causing uncontrolled efflux of ions and/or nutrients and dissipating the protonmotive force (PMF). In some instances, target cells intoxicated by PFTs display additional effects as part of the cellular response to pore formation. Significant progress has been made in the mechanistic description of pore formation for the different PFTs families, but in several cases a complete understanding of pore structure remains lacking. PFTs have evolved recognition mechanisms to bind specific receptors that define their host tropism, although this can be remarkably diverse even within the same family. Here we summarise the salient features of PFTs and highlight where additional research is necessary to fully understand the mechanism of pore formation by members of this diverse group of protein toxins.
Collapse
Affiliation(s)
- Fatima R. Ulhuq
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Giuseppina Mariano
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
A deep learning model to detect novel pore-forming proteins. Sci Rep 2022; 12:2013. [PMID: 35132124 PMCID: PMC8821639 DOI: 10.1038/s41598-022-05970-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Many pore-forming proteins originating from pathogenic bacteria are toxic against agricultural pests. They are the key ingredients in several pesticidal products for agricultural use, including transgenic crops. There is an urgent need to identify novel pore-forming proteins to combat development of resistance in pests to existing products, and to develop products that are effective against a broader range of pests. Existing computational methodologies to search for these proteins rely on sequence homology-based approaches. These approaches are based on similarities between protein sequences, and thus are limited in their usefulness for discovering novel proteins. In this paper, we outline a novel deep learning model trained on pore-forming proteins from the public domain. We compare different ways of encoding protein information during training, and contrast it with traditional approaches. We show that our model is capable of identifying known pore formers with no sequence similarity to the proteins used to train the model, and therefore holds promise for identifying novel pore formers.
Collapse
|
33
|
Cytolysin A (ClyA): A Bacterial Virulence Factor with Potential Applications in Nanopore Technology, Vaccine Development, and Tumor Therapy. Toxins (Basel) 2022; 14:toxins14020078. [PMID: 35202106 PMCID: PMC8880466 DOI: 10.3390/toxins14020078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/26/2022] Open
Abstract
Cytolysin A (ClyA) is a pore-forming toxin that is produced by some bacteria from the Enterobacteriaceae family. This review provides an overview of the current state of knowledge regarding ClyA, including the prevalence of the encoding gene and its transcriptional regulation, the secretion pathway used by the protein, and the mechanism of protein assembly, and highlights potential applications of ClyA in biotechnology. ClyA expression is regulated at the transcriptional level, primarily in response to environmental stressors, and ClyA can exist stably both as a soluble monomer and as an oligomeric membrane complex. At high concentrations, ClyA induces cytolysis, whereas at low concentrations ClyA can affect intracellular signaling. ClyA is secreted in outer membrane vesicles (OMVs), which has important implications for biotechnology applications. For example, the native pore-forming ability of ClyA suggests that it could be used as a component of nanopore-based technologies, such as sequencing platforms. ClyA has also been exploited in vaccine development owing to its ability to present antigens on the OMV surface and provoke a robust immune response. In addition, ClyA alone or OMVs carrying ClyA fusion proteins have been investigated for their potential use as anti-tumor agents.
Collapse
|
34
|
Zhou J, Duan M, Huang D, Shao H, Zhou Y, Fan Y. Label-free visible colorimetric biosensor for detection of multiple pathogenic bacteria based on engineered polydiacetylene liposomes. J Colloid Interface Sci 2022; 606:1684-1694. [PMID: 34500167 DOI: 10.1016/j.jcis.2021.07.155] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/09/2021] [Accepted: 07/30/2021] [Indexed: 11/19/2022]
Abstract
Bacterial infections are considered as a critical healthcare concern worldwide. Timely infection detection is crucial to effective antibiotic administration which can reduce the severity of infection and the occurrence of antibiotic resistance. We have developed label-free polydiacetylene (PDA) liposome-based colorimetric biosensor to detect and identify bacterial cultures at the genus and species level with naked eyes by simple color change. We found that among the various liposomal systems, moderate concentration of PDA, phospholipids and cholesterol in liposome assemblies can greatly influence the sensitivity to different bacteria, exhibiting unique chromatic properties of each bacterial strain. The strikingly different chromatic color change was due to the various mechanisms of interactions between bacterial toxins and biomimetic lipid bilayers. Furthermore, increase of cholesterol in liposome assemblies greatly enhanced the sensitivity of bacterial strains related to membrane destruction mediated by pore-formation mechanism such as S. aureus and E.coli, whereas the detection of the two bacterial strains was believed to rely on the specific recognition elements coupled with PDA moiety. As a proof of concept, a colorimetric finger-print array for distinguishing 6 bacterial species was studied. Particularly, the proposed bacterial detection platform is achieved through the interaction between bacterially secreted toxins and liposome bilayers instead of specific recognition of receptors-ligands. The results of both response time and sensitivity of label-free-liposome-based system show superior to previous reports on chromatic bacterial detection assays. By combing these results, the label-free-liposome-based colorimetric sensing platform shows great importance as a bacterial-sensing and discrimination platform.
Collapse
Affiliation(s)
- Jin Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 102402, China.
| | - Menglong Duan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Diwen Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Hui Shao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yue Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 102402, China.
| |
Collapse
|
35
|
Besançon H, Larpin Y, Babiychuk VS, Köffel R, Babiychuk EB. Engineered Liposomes Protect Immortalized Immune Cells from Cytolysins Secreted by Group A and Group G Streptococci. Cells 2022; 11:cells11010166. [PMID: 35011729 PMCID: PMC8749993 DOI: 10.3390/cells11010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/30/2021] [Accepted: 01/02/2022] [Indexed: 12/10/2022] Open
Abstract
The increasing antibiotic resistance of bacterial pathogens fosters the development of alternative, non-antibiotic treatments. Antivirulence therapy, which is neither bacteriostatic nor bactericidal, acts by depriving bacterial pathogens of their virulence factors. To establish a successful infection, many bacterial pathogens secrete exotoxins/cytolysins that perforate the host cell plasma membrane. Recently developed liposomal nanotraps, mimicking the outer layer of the targeted cell membranes, serve as decoys for exotoxins, thus diverting them from attacking host cells. In this study, we develop a liposomal nanotrap formulation that is capable of protecting immortalized immune cells from the whole palette of cytolysins secreted by Streptococcus pyogenes and Streptococcus dysgalactiae subsp. equisimilis—important human pathogens that can cause life-threatening bacteremia. We show that the mixture of cholesterol-containing liposomes with liposomes composed exclusively of phospholipids is protective against the combined action of all streptococcal exotoxins. Our findings pave the way for further development of liposomal antivirulence therapy in order to provide more efficient treatment of bacterial infections, including those caused by antibiotic resistant pathogens.
Collapse
|
36
|
Dai H, Meng XW, Ye K, Jia J, Kaufmann SH. Therapeutics targeting BCL2 family proteins. MECHANISMS OF CELL DEATH AND OPPORTUNITIES FOR THERAPEUTIC DEVELOPMENT 2022:197-260. [DOI: 10.1016/b978-0-12-814208-0.00007-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Sengupta N, Mondal AK, Mishra S, Chattopadhyay K, Dutta S. Single-particle cryo-EM reveals conformational variability of the oligomeric VCC β-barrel pore in a lipid bilayer. J Cell Biol 2021; 220:212683. [PMID: 34617964 PMCID: PMC8504180 DOI: 10.1083/jcb.202102035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/25/2021] [Accepted: 09/19/2021] [Indexed: 11/22/2022] Open
Abstract
Vibrio cholerae cytolysin (VCC) is a water-soluble, membrane-damaging, pore-forming toxin (PFT) secreted by pathogenic V. cholerae, which causes eukaryotic cell death by altering the plasma membrane permeability. VCC self-assembles on the cell surface and undergoes a dramatic conformational change from prepore to heptameric pore structure. Over the past few years, several high-resolution structures of detergent-solubilized PFTs have been characterized. However, high-resolution structural characterization of small β-PFTs in a lipid environment is still rare. Therefore, we used single-particle cryo-EM to characterize the structure of the VCC oligomer in large unilamellar vesicles, which is the first atomic-resolution cryo-EM structure of VCC. From our study, we were able to provide the first documented visualization of the rim domain amino acid residues of VCC interacting with lipid membrane. Furthermore, cryo-EM characterization of lipid bilayer–embedded VCC suggests interesting conformational variabilities, especially in the transmembrane channel, which could have a potential impact on the pore architecture and assist us in understanding the pore formation mechanism.
Collapse
Affiliation(s)
- Nayanika Sengupta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Anish Kumar Mondal
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India
| | - Suman Mishra
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Kausik Chattopadhyay
- Centre for Protein Science, Design and Engineering, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India
| | - Somnath Dutta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| |
Collapse
|
38
|
Alves RDC, Ferreira CGM, Ferreira de Melo IM, Baptista MGP, Lima de Albuquerque YM, do Nascimento BJ, dos Santos YB, Wanderley-Teixeira V, Teixeira ÁAC. Renal and hepatic changes in the offspring of rats that received biological insecticides during pregnancy and lactation. Acta Histochem 2021; 123:151799. [PMID: 34656827 DOI: 10.1016/j.acthis.2021.151799] [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: 07/29/2020] [Revised: 09/27/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022]
Abstract
Bacillus thuringiensis insecticides have been considered safe, being an alternative to the use of synthetic insecticides. However, studies have shown the effects of Bt Cry toxins on various organs, compromising their functions. The objective of this work was to test whether the administration of biological insecticides based on B. thuringiensis in pregnant rats will cause histopathological changes in the liver and kidneys, as well as in the levels of toxicity biomarkers, of their puppies in adulthood. Twenty rats, 90 days old, were used, divided into four groups: GC - Pregnant rats, GX - Pregnant rats that received XenTari®, GDi - Pregnant rats that received Dipel® and GDe - Pregnant rats that received deltamethrin. Insecticides were administered by gavage at a dosage of 1 mg/100 g/day (GX and GDi), and 2 mg/Kg/day (GDe) during pregnancy and lactation. In the animals of the groups whose matrices received the insecticides, there was a reduction in the levels of the biomarkers of toxicity alanine aminotransferase (ALT), aspartate aminotransferase (AST), urea and creatinine, about the control group. The biological insecticides promoted histopathological changes in the liver, with the presence of portal vein, centrilobular and sinusoidal capillaries congestion, and in the kidney, presence of cortical congestion and reduction of the subcapsular space. Histochemical evaluation in the liver demonstrated a significant reduction in glycogen in the groups that received insecticides when compared to the control group, whereas for collagen fibers in both the liver and the kidneys, no differences were observed between the experimental groups. The morphometry of the liver revealed a significant reduction in the lobular parenchyma and an increase in the non-lobular parenchyma, and in the kidney, there was a reduction in the diameter and volume of the glomerulus and Bowman's capsule of the animals whose matrices received both biological and synthetic insecticides. Thus, it is concluded that the biological insecticides XenTari® and Dipel® in sublethal doses in pregnant rats promote changes in the liver and kidney of the offspring similar to the insecticide deltamethrin, which extend into adulthood.
Collapse
|
39
|
Sannigrahi A, Chattopadhyay K. Pore formation by pore forming membrane proteins towards infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:79-111. [PMID: 35034727 DOI: 10.1016/bs.apcsb.2021.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the last 25 years, the biology of membrane proteins, including the PFPs-membranes interactions is seeking attention for the development of successful drug molecules against a number of infectious diseases. Pore forming toxins (PFTs), the largest family of PFPs are considered as a group of virulence factors produced in a large number of pathogenic systems which include streptococcus, pneumonia, Staphylococcus aureus, E. coli, Mycobacterium tuberculosis, group A and B streptococci, Corynebacterium diphtheria and many more. PFTs are generally utilized by the disease causing pathogens to disrupt the host first line of defense i.e. host cell membranes through pore formation strategy. Although, pore formation is the principal mode of action of the PFTs but they can have additional adverse effects on the hosts including immune evasion. Recently, structural investigation of different PFTs have imparted the molecular mechanistic insights into how PFTs get transformed from its inactive state to active toxic state. On the basis of their structural entity, PFTs have been classified in different types and their mode of actions alters in terms of pore formation and corresponding cellular toxicity. Although pathogen genome analysis can identify the probable PFTs depending upon their structural diversity, there are so many PFTs which utilize the local environmental conditions to generate their pore forming ability using a novel strategy which is known as "conformational switch" of a protein. This conformational switch is considered as characteristics of the phase shifting proteins which were often utilized by many pathogenic systems to protect them from the invaders through allosteric communication between distant regions of the protein. In this chapter, we discuss the structure function relationships of PFTs and how activity of PFTs varies with the change in the environmental conditions has been explored. Finally, we demonstrate these structural insights to develop therapeutic potential to treat the infections caused by multidrug resistant pathogens.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, Karnataka, India.
| | - Krishnananda Chattopadhyay
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.
| |
Collapse
|
40
|
Zhang Y, Zhang A, Li M, He K, Guo S. Nanoparticle-loaded microcapsules providing effective UV protection for Cry1Ac. J Microencapsul 2021; 38:522-532. [PMID: 34615422 DOI: 10.1080/02652048.2021.1990424] [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: 10/20/2022]
Abstract
AIM To prepare several novel microcapsules using chitosan (Cs) and Alginate (Alg) as coating materials, and nano-ZnO, nano-SiO2, nano-TiO2 as UV protective agents for improving UV resistance of Cry1Ac. METHODS Microcapsules were prepared by the layer-by-layer (LbL) self-assembly technique and electrostatic adsorption. The morphologies were observed by scanning electron microscopy (SEM), and the stability under UV radiation was studied by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and bioassay. RESULTS SEM showed that nano-ZnO and nano-TiO2 could be adsorbed on the negatively charged MC with the outermost layer being Alg, while nano-SiO2 could be adsorbed on the positively charged MC with Cs as the outermost layer. SDS-PAGE and bioassay showed that nano-ZnO and nano-SiO2 could provide effective UV protection after 8 h UV irradiation (p > 0.05), and nano-TiO2 could provide effective UV protection after 4 h UV irradiation (p > 0.05). CONCLUSION The microcapsules loaded with nanoparticles provided excellent UV resistance for Cry1Ac.
Collapse
Affiliation(s)
- Yongjing Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Aijing Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Mengyuan Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Kanglai He
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shuyuan Guo
- School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
41
|
Daskalov A, Glass NL. Gasdermin and Gasdermin-Like Pore-Forming Proteins in Invertebrates, Fungi and Bacteria. J Mol Biol 2021; 434:167273. [PMID: 34599942 DOI: 10.1016/j.jmb.2021.167273] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
The gasdermin family of pore-forming proteins (PFPs) has recently emerged as key molecular players controlling immune-related cell death in mammals. Characterized mammalian gasdermins are activated through proteolytic cleavage by caspases or serine proteases, which remove an inhibitory carboxy-terminal domain, allowing the pore-formation process. Processed gasdermins form transmembrane pores permeabilizing the plasma membrane, which often results in lytic and inflammatory cell death. While the gasdermin-dependent cell death (pyroptosis) has been predominantly characterized in mammals, it now has become clear that gasdermins also control cell death in early vertebrates (teleost fish) and invertebrate animals such as corals (Cnidaria). Moreover, gasdermins and gasdermin-like proteins have been identified and characterized in taxa outside of animals, notably Fungi and Bacteria. Fungal and bacterial gasdermins share many features with mammalian gasdermins including their mode of activation through proteolysis. It has been shown that in some cases the proteolytic activation is executed by evolutionarily related proteases acting downstream of proteins resembling immune receptors controlling pyroptosis in mammals. Overall, these findings establish gasdermins and gasdermin-regulated cell death as an extremely ancient mechanism of cellular suicide and build towards an understanding of the evolution of regulated cell death in the context of immunology. Here, we review the broader gasdermin family, focusing on recent discoveries in invertebrates, fungi and bacteria.
Collapse
Affiliation(s)
- Asen Daskalov
- Institut de Biochimie et Génétique Cellulaires, University of Bordeaux, France.
| | - N Louise Glass
- The Plant and Microbial Biology Department, The University of California, Berkeley, CA 94720-3102, United States
| |
Collapse
|
42
|
Outram MA, Sung YC, Yu D, Dagvadorj B, Rima SA, Jones DA, Ericsson DJ, Sperschneider J, Solomon PS, Kobe B, Williams SJ. The crystal structure of SnTox3 from the necrotrophic fungus Parastagonospora nodorum reveals a unique effector fold and provides insight into Snn3 recognition and pro-domain protease processing of fungal effectors. THE NEW PHYTOLOGIST 2021; 231:2282-2296. [PMID: 34053091 DOI: 10.1111/nph.17516] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/20/2021] [Indexed: 05/22/2023]
Abstract
Plant pathogens cause disease through secreted effector proteins, which act to promote infection. Typically, the sequences of effectors provide little functional information and further targeted experimentation is required. Here, we utilized a structure/function approach to study SnTox3, an effector from the necrotrophic fungal pathogen Parastagonospora nodorum, which causes cell death in wheat-lines carrying the sensitivity gene Snn3. We developed a workflow for the production of SnTox3 in a heterologous host that enabled crystal structure determination and functional studies. We show this approach can be successfully applied to study effectors from other pathogenic fungi. The β-barrel fold of SnTox3 is a novel fold among fungal effectors. Structure-guided mutagenesis enabled the identification of residues required for Snn3 recognition. SnTox3 is a pre-pro-protein, and the pro-domain of SnTox3 can be cleaved in vitro by the protease Kex2. Complementing this, an in silico study uncovered the prevalence of a conserved motif (LxxR) in an expanded set of putative pro-domain-containing fungal effectors, some of which can be cleaved by Kex2 in vitro. Our in vitro and in silico study suggests that Kex2-processed pro-domain (designated here as K2PP) effectors are common in fungi and this may have broad implications for the approaches used to study their functions.
Collapse
Affiliation(s)
- Megan A Outram
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yi-Chang Sung
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Daniel Yu
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Bayantes Dagvadorj
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Sharmin A Rima
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - David A Jones
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Daniel J Ericsson
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Synchrotron, Macromolecular Crystallography, Clayton, VIC, 3168, Australia
| | - Jana Sperschneider
- Biological Data Science Institute, The Australian National University, Canberra, ACT, 2601, Australia
| | - Peter S Solomon
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Simon J Williams
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
43
|
Outram MA, Sung YC, Yu D, Dagvadorj B, Rima SA, Jones DA, Ericsson DJ, Sperschneider J, Solomon PS, Kobe B, Williams SJ. The crystal structure of SnTox3 from the necrotrophic fungus Parastagonospora nodorum reveals a unique effector fold and provides insight into Snn3 recognition and pro-domain protease processing of fungal effectors. THE NEW PHYTOLOGIST 2021; 231:2282-2296. [PMID: 34053091 DOI: 10.1101/2020.05.27.120113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/20/2021] [Indexed: 05/25/2023]
Abstract
Plant pathogens cause disease through secreted effector proteins, which act to promote infection. Typically, the sequences of effectors provide little functional information and further targeted experimentation is required. Here, we utilized a structure/function approach to study SnTox3, an effector from the necrotrophic fungal pathogen Parastagonospora nodorum, which causes cell death in wheat-lines carrying the sensitivity gene Snn3. We developed a workflow for the production of SnTox3 in a heterologous host that enabled crystal structure determination and functional studies. We show this approach can be successfully applied to study effectors from other pathogenic fungi. The β-barrel fold of SnTox3 is a novel fold among fungal effectors. Structure-guided mutagenesis enabled the identification of residues required for Snn3 recognition. SnTox3 is a pre-pro-protein, and the pro-domain of SnTox3 can be cleaved in vitro by the protease Kex2. Complementing this, an in silico study uncovered the prevalence of a conserved motif (LxxR) in an expanded set of putative pro-domain-containing fungal effectors, some of which can be cleaved by Kex2 in vitro. Our in vitro and in silico study suggests that Kex2-processed pro-domain (designated here as K2PP) effectors are common in fungi and this may have broad implications for the approaches used to study their functions.
Collapse
Affiliation(s)
- Megan A Outram
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yi-Chang Sung
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Daniel Yu
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Bayantes Dagvadorj
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Sharmin A Rima
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - David A Jones
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Daniel J Ericsson
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Synchrotron, Macromolecular Crystallography, Clayton, VIC, 3168, Australia
| | - Jana Sperschneider
- Biological Data Science Institute, The Australian National University, Canberra, ACT, 2601, Australia
| | - Peter S Solomon
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Simon J Williams
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
44
|
Park J, Choi J, Kim DD, Lee S, Lee B, Lee Y, Kim S, Kwon S, Noh M, Lee MO, Le QV, Oh YK. Bioactive Lipids and Their Derivatives in Biomedical Applications. Biomol Ther (Seoul) 2021; 29:465-482. [PMID: 34462378 PMCID: PMC8411027 DOI: 10.4062/biomolther.2021.107] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022] Open
Abstract
Lipids, which along with carbohydrates and proteins are among the most important nutrients for the living organism, have a variety of biological functions that can be applied widely in biomedicine. A fatty acid, the most fundamental biological lipid, may be classified by length of its aliphatic chain, and the short-, medium-, and long-chain fatty acids and each have distinct biological activities with therapeutic relevance. For example, short-chain fatty acids have immune regulatory activities and could be useful against autoimmune disease; medium-chain fatty acids generate ketogenic metabolites and may be used to control seizure; and some metabolites oxidized from long-chain fatty acids could be used to treat metabolic disorders. Glycerolipids play important roles in pathological environments, such as those of cancers or metabolic disorders, and thus are regarded as a potential therapeutic target. Phospholipids represent the main building unit of the plasma membrane of cells, and play key roles in cellular signaling. Due to their physical properties, glycerophospholipids are frequently used as pharmaceutical ingredients, in addition to being potential novel drug targets for treating disease. Sphingolipids, which comprise another component of the plasma membrane, have their own distinct biological functions and have been investigated in nanotechnological applications such as drug delivery systems. Saccharolipids, which are derived from bacteria, have endotoxin effects that stimulate the immune system. Chemically modified saccharolipids might be useful for cancer immunotherapy or as vaccine adjuvants. This review will address the important biological function of several key lipids and offer critical insights into their potential therapeutic applications.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Bongjin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yunhee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sanghee Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungwon Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Quoc-Viet Le
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
45
|
Palacios-Ortega J, García-Linares S, Rivera-de-Torre E, Heras-Márquez D, Gavilanes JG, Slotte JP, Martínez-Del-Pozo Á. Structural foundations of sticholysin functionality. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140696. [PMID: 34246789 DOI: 10.1016/j.bbapap.2021.140696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023]
Abstract
Actinoporins constitute a family of α pore-forming toxins produced by sea anemones. The soluble fold of these proteins consists of a β-sandwich flanked by two α-helices. Actinoporins exert their activity by specifically recognizing sphingomyelin at their target membranes. Once there, they penetrate the membrane with their N-terminal α-helices, a process that leads to the formation of cation-selective pores. These pores kill the target cells by provoking an osmotic shock on them. In this review, we examine the role and relevance of the structural features of actinoporins, down to the residue level. We look at the specific amino acids that play significant roles in the function of actinoporins and their fold. Particular emphasis is given to those residues that display a high degree of conservation across the actinoporin sequences known to date. In light of the latest findings in the field, the membrane requirements for pore formation, the effect of lipid composition, and the process of pore formation are also discussed.
Collapse
Affiliation(s)
- Juan Palacios-Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain; Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
| | - Sara García-Linares
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - Esperanza Rivera-de-Torre
- Department of Biochemistry and Biotechnology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Diego Heras-Márquez
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - José G Gavilanes
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | - J Peter Slotte
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Álvaro Martínez-Del-Pozo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| |
Collapse
|
46
|
Qamar Z, Nasir IA, Abouhaidar MG, Hefferon KL, Rao AQ, Latif A, Ali Q, Anwar S, Rashid B, Shahid AA. Novel approaches to circumvent the devastating effects of pests on sugarcane. Sci Rep 2021; 11:12428. [PMID: 34127751 PMCID: PMC8203629 DOI: 10.1038/s41598-021-91985-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023] Open
Abstract
Sugarcane (Saccharum officinarum L.) is a cash crop grown commercially for its higher amounts of sucrose, stored within the mature internodes of the stem. Numerous studies have been done for the resistance development against biotic and abiotic stresses to save the sucrose yields. Quality and yield of sugarcane production is always threatened by the damages of cane borers and weeds. In current study two problems were better addressed through the genetic modification of sugarcane for provision of resistance against insects and weedicide via the expression of two modified cane borer resistant CEMB-Cry1Ac (1.8 kb), CEMB-Cry2A (1.9 kb) and one glyphosate tolerant CEMB-GTGene (1.4 kb) genes, driven by maize Ubiquitin Promoter and nos terminator. Insect Bio-toxicity assays were carried out for the assessment of Cry proteins through mortality percent of shoot borer Chilo infuscatellus at 2nd instar larvae stage. During V0, V1 and V2 generations young leaves from the transgenic sugarcane plants were collected at plant age of 20, 40, 60, 80 days and fed to the Chilo infuscatellus larvae. Up to 100% mortality of Chilo infuscatellus from 80 days old transgenic plants of V2 generation indicated that these transgenic plants were highly resistant against shoot borer and the gene expression level is sufficient to provide complete resistance against target pests. Glyphosate spray assay was carried out for complete removal of weeds. In V1-generation, 70-76% transgenic sugarcane plants were found tolerant against glyphosate spray (3000 mL/ha) under field conditions. While in V2-generation, the replicates of five selected lines 4L/2, 5L/5, 6L/5, L8/4, and L9/6 were found 100% tolerant against 3000 mL/ha glyphosate spray. It is evident from current study that CEMB-GTGene, CEMB-Cry1Ac and CEMB-Cry2A genes expression in sugarcane variety CPF-246 showed an efficient resistance against cane borers (Chilo infuscatellus) and was also highly tolerant against glyphosate spray. The selected transgenic sugarcane lines showed sustainable resistance against cane borer and glyphosate spray can be further exploited at farmer's field level after fulfilling the biosafety requirements to boost the sugarcane production in the country.
Collapse
Affiliation(s)
- Zahida Qamar
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan.
| | - Idrees Ahmad Nasir
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Mounir G Abouhaidar
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | | | - Abdul Qayyum Rao
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Ayesha Latif
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Qurban Ali
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Saima Anwar
- Pakistan Biomedical Engineering Centre University of Engineering and Technology, New Campus, Lahore, Pakistan
| | - Bushra Rashid
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Ahmad Ali Shahid
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
47
|
Xie J, Mei H, Jin S, Bu L, Wang X, Wang C, Zhao Q, Ma R, Zhou S. Outbreak of vibriosis associated with Vibrio parahaemolyticus in the mud crab Scylla paramamosain cultured in China. DISEASES OF AQUATIC ORGANISMS 2021; 144:187-196. [PMID: 34042066 DOI: 10.3354/dao03587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In this study, a Gram-negative bacterium was isolated from diseased Scylla paramamosain and tentatively named strain QX17. The bacterial isolate was identified as Vibrio parahaemolyticus based on morphological and biochemical characteristics and molecular identification with the 16S rRNA and HSP60 genes. In the challenge experiment, S. paramamosain injected intramuscularly with the V. parahaemolyticus isolate developed pathological signs similar to the naturally diseased mud crabs. The infection experiment also showed that the median lethal dosage (LD50) for QX17 was 4.79 × 102 CFU g-1 (crab weight). Histopathological analysis of the diseased mud crabs infected with V. parahaemolyticus showed deformation and basement membrane rupture of hepatopancreatic tubules in the hepatopancreas, and disordered and broken muscle fiber in the muscle. Antimicrobial susceptibility tests revealed that QX17 was highly sensitive to most of the tested aminoglycosides, tetracyclines, and quinolones. To the best of our knowledge, this is the first study reporting isolation and antibiotic sensitivities of V. parahaemolyticus from cultured mud crabs. The discovery of V. parahaemolyticus in cultured mud crabs not only adds to the growing list of emerging pathogens in crab aquaculture in China, but also highlights the necessity of developing early detection strategies and appropriate interventions to reduce the damage caused by V. parahaemolyticus in mud crab aquaculture.
Collapse
Affiliation(s)
- Jiasong Xie
- School of Marine Sciences, Ningbo University, Ningbo, Zhejiang Province 315832, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kaplan AR, Olson R, Alexandrescu AT. Protein yoga: Conformational versatility of the Hemolysin II C-terminal domain detailed by NMR structures for multiple states. Protein Sci 2021; 30:990-1005. [PMID: 33733504 DOI: 10.1002/pro.4066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 11/05/2022]
Abstract
The C-terminal domain of Bacillus cereus hemolysin II (HlyIIC), stabilizes the trans-membrane-pore formed by the HlyII toxin and may aid in target cell recognition. Initial efforts to determine the NMR structure of HlyIIC were hampered by cis/trans isomerization about the single proline at position 405 that leads to doubling of NMR resonances. We used the mutant P405M-HlyIIC that eliminates the cis proline to determine the NMR structure of the domain, which revealed a novel fold. Here, we extend earlier studies to the NMR structure determination of the cis and trans states of WT-HlyIIC that exist simultaneously in solution. The primary structural differences between the cis and trans states are in the loop that contains P405, and structurally adjacent loops. Thermodynamic linkage analysis shows that at 25 C the cis proline, which already has a large fraction of 20% in the unfolded protein, increases to 50% in the folded state due to coupling with the global stability of the domain. The P405M or P405A substitutions eliminate heterogeneity due to proline isomerization but lead to the formation of a new dimeric species. The NMR structure of the dimer shows that it is formed through domain-swapping of strand β5, the last segment of secondary structure following P405. The presence of P405 in WT-HlyIIC strongly disfavors the dimer compared to the P405M-HlyIIC or P405A-HlyIIC mutants. The WT proline may thus act as a "gatekeeper," warding off aggregative misfolding.
Collapse
Affiliation(s)
- Anne R Kaplan
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Rich Olson
- Department of Molecular Biology and Biochemistry, Molecular Biophysics Program, Wesleyan University, Middletown, Connecticut, USA
| | - Andrei T Alexandrescu
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
49
|
Žerovnik E. Viroporins vs. Other Pore-Forming Proteins: What Lessons Can We Take? Front Chem 2021; 9:626059. [PMID: 33681145 PMCID: PMC7930612 DOI: 10.3389/fchem.2021.626059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/07/2021] [Indexed: 11/13/2022] Open
Abstract
Pore-forming proteins (PFPs) exist in virtually all domains of life, and by disrupting cellular membranes, depending on the pore size, they cause ion dis-balance, small substances, or even protein efflux/influx, influencing cell’s signaling routes and fate. Such pore-forming proteins exist from bacteria to viruses and also shape host defense systems, including innate immunity. There is strong evidence that amyloid toxicity is also caused by prefibrillar oligomers making “amyloid pores” into cellular membranes. For most of the PFPs, a 2-step mechanism of protein-membrane interaction takes place on the “lipid rafts,” membrane microdomains rich in gangliosides and cholesterol. In this mini-review paper, common traits of different PFPs are looked at. Possible ways for therapy of channelopathies and/or modulating immunity relevant to the new threat of SARS-CoV-2 infections could be learnt from such comparisons.
Collapse
Affiliation(s)
- Eva Žerovnik
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
50
|
Besançon H, Babiychuk V, Larpin Y, Köffel R, Schittny D, Brockhus L, Hathaway LJ, Sendi P, Draeger A, Babiychuk E. Tailored liposomal nanotraps for the treatment of Streptococcal infections. J Nanobiotechnology 2021; 19:46. [PMID: 33588835 PMCID: PMC7885208 DOI: 10.1186/s12951-021-00775-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/11/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Streptococcal infections are associated with life-threatening pneumonia and sepsis. The rise in antibiotic resistance calls for novel approaches to treat bacterial diseases. Anti-virulence strategies promote a natural way of pathogen clearance by eliminating the advantage provided to bacteria by their virulence factors. In contrast to antibiotics, anti-virulence agents are less likely to exert selective evolutionary pressure, which is a prerequisite for the development of drug resistance. As part of their virulence mechanism, many bacterial pathogens secrete cytolytic exotoxins (hemolysins) that destroy the host cell by destabilizing their plasma membrane. Liposomal nanotraps, mimicking plasmalemmal structures of host cells that are specifically targeted by bacterial toxins are being developed in order to neutralize-by competitive sequestration-numerous exotoxins. RESULTS In this study, the liposomal nanotrap technology is further developed to simultaneously neutralize the whole palette of cytolysins produced by Streptococcus pneumoniae, Streptococcus pyogenes and Streptococcus dysgalactiae subspecies equisimilis-pathogens that can cause life-threatening streptococcal toxic shock syndrome. We show that the mixture of liposomes containing high amounts of cholesterol and liposomes composed exclusively of choline-containing phospholipids is fully protective against the combined action of exotoxins secreted by these pathogens. CONCLUSIONS Unravelling the universal mechanisms that define targeting of host cells by streptococcal cytolysins paves the way for a broad-spectrum anti-toxin therapy that can be applied without a diagnostic delay for the treatment of bacterial infections including those caused by antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Hervé Besançon
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | | | - Yu Larpin
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - René Köffel
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Dominik Schittny
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Lara Brockhus
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Lucy J Hathaway
- Institute for Infectious Diseases, University of Bern, 3001, Bern, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, 3001, Bern, Switzerland
| | - Annette Draeger
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland
| | - Eduard Babiychuk
- Institute of Anatomy, University of Bern, 3012, Bern, Switzerland.
| |
Collapse
|