1
|
Wu Y, Liu Q, Tian R, Zhao S, Ding B, Jiang Q. Predesigned DNA Origami Nanodrills Mediate Controlled Pore Formation on a Plasma Membrane and Cell Death. NANO LETTERS 2025; 25:8220-8226. [PMID: 40343847 DOI: 10.1021/acs.nanolett.5c01290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
We presented an engineered DNA origami nanodrill that is capable of controlled attachment and penetration of the plasma membrane, thereby inducing cell damage and lytic death. The cap-and-stem-like DNA origami nanodrills were constructed, equipping them with addressable cholesterol tags on their caps for adherence to lipid bilayers. The subsequent insertion of the origami stems was initiated, enabling the generation of distinct transmembrane channels in various artificial and biological membranes. These nanodrills with plasma-membrane-disrupting functions resulted in mechanical damage to living cell membranes, leading to increased cytosolic calcium levels, enhanced intracellular protein release, and potent cytotoxicity against tumor cells. The further stimuli-responsive design enabled acid-triggered attachment of DNA nanodrills to the plasma membrane and subsequent cell damage in a pH-controlled fashion. This programmable origami nanodrill could serve as an artificial mediator of plasma-membrane rupture and cell death, thus potentially leading to the development of new therapeutic strategies to combat cancer.
Collapse
Affiliation(s)
- Yushuai Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Run Tian
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Zhao
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Qiao Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
2
|
Policarpo M, Salzburger W, Maumus F, Gilbert C. Multiple Horizontal Transfers of Immune Genes Between Distantly Related Teleost Fishes. Mol Biol Evol 2025; 42:msaf107. [PMID: 40378191 PMCID: PMC12107551 DOI: 10.1093/molbev/msaf107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
Horizontal gene transfer (HGT) is less frequent in eukaryotes than in prokaryotes, yet can have strong functional implications and was proposed as a causal factor for major adaptations in several eukaryotic lineages. Most cases of eukaryote HGT reported to date are inter-domain transfers, and few studies have investigated eukaryote-to-eukaryote HGTs. Here, we performed a large-scale survey of HGT among 242 species of ray-finned fishes. We found multiple lines of evidence supporting 19 teleost-to-teleost HGT events that involve 17 different genes in 11 teleost fish orders. The genes involved in these transfers show lower synonymous divergence than expected under vertical transmission, their phylogeny is inconsistent with that of teleost fishes, and they occur at non-syntenic positions in donor and recipient lineages. The distribution of HGT events in the teleost tree is heterogenous, with 8 of the 19 transfers occurring between the same two orders (Osmeriformes and Clupeiformes). Though we favor a scenario involving multiple HGT events, future work should evaluate whether hybridization between species belonging to different teleost orders may generate HGT-like patterns. Besides the previously reported transfer of an antifreeze protein, most transferred genes play roles in immunity or are pore-forming proteins, suggesting that such genes may be more likely than others to confer a strong selective advantage to the recipient species. Overall, our work shows that teleost-to-teleost HGT has occurred on multiple occasions, and it will be worth further quantifying these transfers and evaluating their impact on teleost evolution as more genomes are sequenced.
Collapse
Affiliation(s)
- Maxime Policarpo
- Zoological Institute, Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Walter Salzburger
- Zoological Institute, Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Florian Maumus
- URGI, INRAE, Université Paris-Saclay, Versailles 78026, France
| | - Clément Gilbert
- Université Paris-Saclay, CNRS, IRD, UMR Évolution, Génomes, Comportement et Écologie, Gif-sur-Yvette 91198, France
| |
Collapse
|
3
|
Lortal L, Lyon CM, Sprague JL, Sonnberger J, Paulin OKA, Wickramasinghe DN, Richardson JP, Hube B, Naglik JR. Candidalysin biology and activation of host cells. mBio 2025:e0060324. [PMID: 40293285 DOI: 10.1128/mbio.00603-24] [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: 04/30/2025] Open
Abstract
Candida albicans is an opportunistic fungal pathogen that can cause life-threatening systemic infections and distressing mucosal infections. A major breakthrough in understanding C. albicans pathogenicity was the discovery of candidalysin, the first cytolytic peptide toxin identified in a human pathogenic fungus. Secreted by C. albicans hyphae and encoded by the ECE1 gene, this 31-amino acid peptide integrates into and permeabilizes host cell membranes, causing damage across diverse cell types. Beyond its cytolytic activity, candidalysin can trigger potent innate immune responses in epithelial cells, macrophages, and neutrophils. Additionally, candidalysin plays a key role in nutrient acquisition during infection. This review explores the biology of candidalysin, its role in host cell activation, and extends the discussion to non-candidalysin Ece1p peptides, shedding light on their emerging significance.
Collapse
Affiliation(s)
- Léa Lortal
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Claire M Lyon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Johannes Sonnberger
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Olivia K A Paulin
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Don N Wickramasinghe
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
4
|
Nguyen CTG, Meng F. Unleashing the power of nucleic acid therapeutics through efficient cytosolic delivery. J Control Release 2025; 383:113774. [PMID: 40280238 DOI: 10.1016/j.jconrel.2025.113774] [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: 12/11/2024] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The approval of siRNA-based therapy for liver disease in 2018 and the subsequent success of mRNA-based SARS-CoV-2 vaccines have inaugurated a new era in nucleic acid-based therapeutics. These breakthroughs underscore the transformative potential of nucleic acid-based therapeutics, which modulate gene function, correct genetic defects, or disrupt pathological molecular processes. Such advances represent a paradigm shift in modern medicine. Despite their immense promise, the clinical realization of nucleic acid-based therapies is fundamentally constrained by endosomal entrapment, a critical barrier that significantly limits therapeutic efficacy. Overcoming this obstacle is imperative to fully unlock the potential of these therapies. Designing effective strategies to facilitate the escape of nucleic acids from endosomes-or bypassing endosomal pathways altogether-remains a central challenge in the field. In this review, we provide a comprehensive and critical analysis of current approaches aimed at enhancing endosomal escape or circumventing endosomal entrapment. By highlighting both the successes and limitations of these strategies, we aim to offer valuable insights to inform the development of more efficient and clinically viable nucleic acid delivery systems, advancing the future of molecular medicine.
Collapse
Affiliation(s)
- Cao Thuy Giang Nguyen
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, MA 01854, USA
| | - Fanfei Meng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, MA 01854, USA.
| |
Collapse
|
5
|
Shekhar A, Di Lucrezia R, Jerye K, Korotkov VS, Harmrolfs K, Rox K, Weich HA, Ghai I, Delhommel F, Becher I, Degenhart C, Fansa E, Unger A, Habenberger P, Klebl B, Lukat P, Schmelz S, Henke S, Borgert S, Lang JC, Sasse F, Diestel R, Richter C, Schneider-Daum N, Hinkelmann B, Niemz J, Lehr CM, Jänsch L, Huehn J, Alm R, Savitski M, Welte T, Hesterkamp T, Sattler M, Winterhalter M, Blankenfeldt W, Medina E, Bilitewski U, Dinkel K, Brönstrup M. Highly potent quinoxalinediones inhibit α-hemolysin and ameliorate Staphylococcus aureus lung infections. Cell Host Microbe 2025; 33:560-572.e21. [PMID: 40168998 DOI: 10.1016/j.chom.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/27/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
Hospital-acquired pneumonia caused by Staphylococcus aureus is associated with patient morbidity and mortality, despite adequate antibiotic therapy. This illustrates the need for treatments beyond antibiotics. The pore-forming heptameric toxin α-hemolysin (Hla) is a major pathogenicity factor of S. aureus and a clinically validated target. We identify quinoxalinediones (QDS) as highly potent Hla inhibitors, conferring protection against the hallmarks of Hla-induced pathogenicity such as Ca2+ influx, cytotoxicity, hemolysis, and monolayer destruction. The effects were exerted across major Hla subtypes in all relevant cell types. QDS prevented the formation of functional pores by interacting with Hla near the phospholipid-binding site. The QDS analog, H052, was active in mouse models of S. aureus lung infections, when administered prophylactically or therapeutically, either as monotherapy or when given in combination with the antibiotic linezolid. The study provides evidence that complex bacterial toxins can be targeted in vivo by drug-like small molecules.
Collapse
Affiliation(s)
- Aditya Shekhar
- Compound Profiling and Screening, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | | | - Karoline Jerye
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Vadim S Korotkov
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kirsten Harmrolfs
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Katharina Rox
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig, Germany
| | - Herbert A Weich
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Ishan Ghai
- Life Sciences and Chemistry, Constructor University, 28759 Bremen, Germany
| | - Florent Delhommel
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85747 Garching, Germany; Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Isabelle Becher
- Proteomics Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | - Eyad Fansa
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Anke Unger
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | | | - Bert Klebl
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Peer Lukat
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Stefan Schmelz
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Steffi Henke
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Sebastian Borgert
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Julia C Lang
- Infection Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Florenz Sasse
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Randi Diestel
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Clémentine Richter
- Department of Drug Delivery, Helmholtz-Institute for Pharmaceutical Research (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery, Helmholtz-Institute for Pharmaceutical Research (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Bettina Hinkelmann
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jana Niemz
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz-Institute for Pharmaceutical Research (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | | | - Mikhail Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine and Infectious Disease, Hannover Medical School, Carl-Neuberg-Strasse 1, Hannover 30625, Germany
| | - Thomas Hesterkamp
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig, Germany
| | - Michael Sattler
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85747 Garching, Germany; Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | | | - Wulf Blankenfeldt
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Eva Medina
- Infection Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Ursula Bilitewski
- Compound Profiling and Screening, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Klaus Dinkel
- Lead Discovery Center, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Mark Brönstrup
- Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany; German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig, Germany; Center of Biomolecular Drug Research (BMWZ), Leibniz Universität, 30167 Hannover, Germany.
| |
Collapse
|
6
|
Wang Y, Li M. QDS pathoblockers target and lock α-hemolysin. Cell Host Microbe 2025; 33:464-466. [PMID: 40209674 DOI: 10.1016/j.chom.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/12/2025]
Abstract
Staphylococcus aureus is a major pathogen with rising antibiotic resistance. In this issue of Cell Host & Microbe, Shekhar et al. find that quinoxalinediones reduce S. aureus lung infections through α-hemolysin inhibition, offering a therapeutic strategy for S. aureus pneumonia.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Nursing, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
7
|
Vázquez RF, Daza Millone MA, Giglio ML, Brola TR, Maté SM, Heras H. Insights into the Protein-Lipid Interaction of Perivitellin-2, an Unusual Snail Pore-Forming Toxin. Toxins (Basel) 2025; 17:183. [PMID: 40278681 PMCID: PMC12031178 DOI: 10.3390/toxins17040183] [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: 03/02/2025] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
The perivitellin-2 (PV2) from snails is an unusual neuro and enterotoxin comprising a pore-forming domain of the Membrane Attack Complex and Perforin Family (MACPF) linked to a lectin. While both domains have membrane binding capabilities, PV2's mechanism of action remains unclear. We studied the apple snail Pomacea maculata PV2's (PmPV2's) interaction with lipid membranes using various biophysical and cell biology approaches. In vitro studies showed that PmPV2 toxicity decreased when cholesterol (Chol) was diminished from enterocyte cell membranes. Chol enhanced PmPV2 association with phosphatidylcholine membranes but did not induce pore formation. In contrast, using rat brain lipid models, rich in glycolipids, PmPV2 exhibited high affinity and induced vesicle permeabilization. Negative stain electron microscopy and atomic force microscopy confirmed the formation of pore-like structures in brain lipid vesicles. Our findings suggest that Chol is a necessary lipid component and point to PmPV2-glycolipid interactions as potential activators critical to triggering PmPV2's pore-forming activity, providing insights into this novel toxin's mechanism.
Collapse
Affiliation(s)
- Romina F. Vázquez
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, INIBIOLP, CCT-La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, La Plata 1900, Argentina; (R.F.V.); (M.L.G.)
| | - M. Antonieta Daza Millone
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas, INIFTA, CCT-La Plata, CONICET, Universidad Nacional de La Plata, Diagonal 113 y 64, La Plata 1900, Argentina;
| | - Matías L. Giglio
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, INIBIOLP, CCT-La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, La Plata 1900, Argentina; (R.F.V.); (M.L.G.)
- Department of Biology, University of Utah, Salt Lake City, UT 84115, USA
| | - Tabata R. Brola
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, INIBIOLP, CCT-La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, La Plata 1900, Argentina; (R.F.V.); (M.L.G.)
| | - Sabina M. Maté
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, INIBIOLP, CCT-La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, La Plata 1900, Argentina; (R.F.V.); (M.L.G.)
| | - Horacio Heras
- Instituto de Investigaciones Bioquímicas de La Plata “Prof. Dr. Rodolfo R. Brenner”, INIBIOLP, CCT-La Plata, CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, La Plata 1900, Argentina; (R.F.V.); (M.L.G.)
- Cátedra de Química Biológica, Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata, 122 y 60, La Plata 1900, Argentina
| |
Collapse
|
8
|
Chow SH, Jeon Y, Deo P, Yeung ATY, Hale C, Sridhar S, Abraham G, Nickson J, Olivier FAB, Jiang JH, Ding Y, Han ML, Le Brun AP, Anderson D, Creek D, Tong J, Gabriel K, Li J, Traven A, Dougan G, Shen HH, Naderer T. Staphylococcal toxin PVL ruptures model membranes under acidic conditions through interactions with cardiolipin and phosphatidic acid. PLoS Biol 2025; 23:e3003080. [PMID: 40233125 PMCID: PMC12052211 DOI: 10.1371/journal.pbio.3003080] [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: 06/10/2024] [Revised: 05/05/2025] [Accepted: 02/24/2025] [Indexed: 04/17/2025] Open
Abstract
Panton-Valentine leukocidin (PVL) is a pore-forming toxin secreted by Staphylococcus aureus strains that cause severe infections. Bicomponent PVL kills phagocytes depending on cell surface receptors, such as complement 5a receptor 1 (C5aR1). How the PVL-receptor interaction enables assembly of the leukocidin complex, targeting of membranes, and insertion of a pore channel remains incompletely understood. Here, we demonstrate that PVL binds the anionic phospholipids, phosphatidic acid, and cardiolipin, under acidic conditions and targets lipid bilayers that mimic lysosomal and mitochondrial membranes, but not the plasma membrane. The PVL-lipid interaction was sufficient to enable leukocidin complex formation as determined by neutron reflectometry and the rupture of model membranes, independent of protein receptors. In phagocytes, PVL and its C5aR1 receptor were internalized depending on sphingomyelin and cholesterol, which were dispensable for the interaction of the toxin with the plasma membrane. Internalized PVL compromised the integrity of lysosomes and mitochondria before plasma membrane rupture. Preventing the acidification of organelles or the genetic loss of PVL impaired the escape of intracellular S. aureus from macrophages. Together, the findings advance our understanding of how an S. aureus toxin kills host cells and provide key insights into how leukocidins target membranes.
Collapse
Affiliation(s)
- Seong H. Chow
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Yusun Jeon
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Pankaj Deo
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Amy T. Y. Yeung
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Christine Hale
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Sushmita Sridhar
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Gilu Abraham
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Joshua Nickson
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Françios A. B. Olivier
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Yue Ding
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Australia
| | - Mei-Ling Han
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Anton P. Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Kirrawee DC, Australia
| | - Dovile Anderson
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Darren Creek
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Janette Tong
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Kip Gabriel
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Jian Li
- Centre to Impact AMR, Monash University, Clayton, Australia
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Gordon Dougan
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Hsin-Hui Shen
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Australia
| | - Thomas Naderer
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| |
Collapse
|
9
|
Chatterjee A, Naskar P, Mishra S, Dutta S. Pore Formation by Pore Forming Proteins in Lipid Membranes: Structural Insights Through Cryo-EM. J Membr Biol 2025:10.1007/s00232-025-00344-5. [PMID: 40155553 DOI: 10.1007/s00232-025-00344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Many pathogenic bacteria utilize their complicated appalling arsenal, bacterial virulence factors, to attack host cells by damaging the host cell membrane and neutralizing host defense mechanisms. Bacterial pore-forming proteins (PFPs) are one of them, they include a distinct class of secreted soluble toxin monomers, which binds to the specific cell surface receptors and /or lipids, oligomerizes as an amphipathic transmembrane pore complex on host cell membranes, and deforms the integrity of the plasma membrane. Researchers have focused on characterizing the structure and function of different Pore Forming Toxins (PFTs) from various organisms, where most of the structural studies employed X-ray crystallography, single-particle cryo-EM, and cryo-electron tomography. However, historically, most of these previous studies focused on using detergent to solubilize and oligomerize the PFTs. Additionally, previous studies have also shown that lipid membranes and lipid components, including cell surface receptors, play a critical role in pore formation and oligomerization. However, there are limited studies available that aim to resolve the structure and function of PFTs in liposomes. In this review article, we majorly focused on structural and functional studies of pore-forming toxins in the presence of detergents, lipid nanodiscs, and liposomes. We will also discuss the challenges and benefits of using liposomes to study pore-forming proteins in more biologically relevant membrane environments.
Collapse
Affiliation(s)
- Arnab Chatterjee
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Prasenjit Naskar
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Suman Mishra
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Somnath Dutta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, 560012, India.
| |
Collapse
|
10
|
Xie J, Xiang J, Shen Y, Shao S. Mechanistic Insights into the Tools for Intracellular Protein Delivery. CHEM & BIO ENGINEERING 2025; 2:132-155. [PMID: 40171130 PMCID: PMC11955855 DOI: 10.1021/cbe.4c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 04/03/2025]
Abstract
Proteins are an important therapeutic modality in modern medicine. However, their inherent inability to traverse cell membranes essentially limits their application to extracellular targets. Recent advances in intracellular protein delivery have enabled access to traditionally "undruggable" intracellular targets and paved the way to precisely modulate cellular functions. This Review provides a comprehensive examination of the key mechanisms and emerging technologies that facilitate the transport of functional proteins across cellular membranes. Delivery methods are categorized into physical, chemical, and biological approaches, each with distinct advantages and limitations. Physical methods enable direct protein entry but often pose challenges related to invasiveness and technical complexity. Chemical strategies offer customizable solutions with enhanced control over cellular targeting and uptake, yet may face issues with cytotoxicity and scalability. Biological approaches leverage naturally occurring processes to achieve efficient intracellular transport, though regulatory and production consistency remain hurdles. By highlighting recent advancements, challenges, and opportunities within each approach, this review underscores the potential of intracellular protein delivery technologies to unlock new therapeutic pathways and transform drug development paradigms.
Collapse
Affiliation(s)
- Jingwen Xie
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Jiajia Xiang
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Youqing Shen
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Shiqun Shao
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| |
Collapse
|
11
|
Liu R, Buttaci DR, Sokol CL. Neurogenic inflammation and itch in barrier tissues. Semin Immunol 2025; 77:101928. [PMID: 39798211 PMCID: PMC11893243 DOI: 10.1016/j.smim.2024.101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
Once regarded as distinct systems, the nervous system and the immune system are now recognized for their complex interactions within the barrier tissues. The neuroimmune circuitry comprises a dual-network system that detects external and internal disturbances, providing critical information to tailor a context-specific response to various threats to tissue integrity, such as wounding or exposure to noxious and harmful stimuli like pathogens, toxins, or allergens. Using the skin as an example of a barrier tissue with the polarized sensory neuronal responses of itch and pain, we explore the molecular pathways driving neuronal activation and the effects of this activation on the immune response. We then apply these findings to other barrier tissues, to find common pathways controlling neuroimmune responses in the barriers.
Collapse
Affiliation(s)
- Rebecca Liu
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dean R Buttaci
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy & Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
12
|
Astacio JD, Rodríguez-Pires S, Melgarejo P, De Cal A, Espeso EA. Differences in Behavior During Early Nectarine Infection Among Main Monilinia spp. Causing Brown Rot. PHYTOPATHOLOGY 2025; 115:269-280. [PMID: 39746049 DOI: 10.1094/phyto-05-24-0159-r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Brown rot is a disease that affects stone and pome fruit crops worldwide. It is caused by fungal members of the genus Monilinia, mainly M. fructicola, M. laxa, and M. fructigena. This study presents evidence that, despite having a very similar battery of cell wall-degrading enzymes, the three species behave differently during the early stages of infection, suggesting differences at the regulatory level, which could also explain the differences in host preference among the three species. We have shown that M. fructicola infection is accelerated by red light, and the first symptoms appear much earlier than in darkness or in the other two species. The overexpression of genes encoding for CAZymes, such as pme3, pme2, pg1, cel1, pnl1, and pnl2, as well as the necrosis factor nep2, can be associated with the etiology of Monilinia spp. In addition, we found that nep2 in M. fructigena lacks binding sites in its promoter sequence for the white-collar complex, which is the major transcription factor responsible for regulating photoreception processes in fungi. Finally, we found that AlphaFold models of the NEP1-like proteins present on the three Monilinia species predict proteins with a very high degree of similarity.
Collapse
Affiliation(s)
- Juan Diego Astacio
- Grupo de Hongos Fitopatógenos, Departamento de Protección Vegetal, Centro Nacional INIA-CSIC, 28040 Madrid, Spain
- Programa Biotecnología y Recursos Genéticos de Plantas y Microorganismos Asociados, ETSIA, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Silvia Rodríguez-Pires
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Paloma Melgarejo
- Grupo de Hongos Fitopatógenos, Departamento de Protección Vegetal, Centro Nacional INIA-CSIC, 28040 Madrid, Spain
| | - Antonieta De Cal
- Grupo de Hongos Fitopatógenos, Departamento de Protección Vegetal, Centro Nacional INIA-CSIC, 28040 Madrid, Spain
| | - Eduardo Antonio Espeso
- Laboratorio de Biología Celular de Aspergillus, Departamento de Biociencias Celulares y Moleculares, Centro Investigaciones Biológicas Margarita Salas, CSIC (CIB-CSIC), 28040 Madrid, Spain
| |
Collapse
|
13
|
Yabrag A, Ullah N, Baryalai P, Ahmad I, Zlatkov N, Toh E, Lindbäck T, Uhlin BE, Wai SN, Nadeem A. A new understanding of Acanthamoeba castellanii: dispelling the role of bacterial pore-forming toxins in cyst formation and amoebicidal actions. Cell Death Discov 2025; 11:66. [PMID: 39971918 PMCID: PMC11839945 DOI: 10.1038/s41420-025-02345-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/24/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
Pore-forming toxins (PFTs) are recognized as major virulence factors produced by both Gram-positive and Gram-negative bacteria. While the effects of PFTs have been extensively investigated using mammalian cells as a model system, their interactions with the environmental host, Acanthamoeba castellanii remains less understood. This study employed high-throughput image screening (HTI), advanced microscopy, western blot analysis, and cytotoxicity assays to evaluate the impact of PFT-producing bacterial species on their virulence against A. castellanii. Our unbiased HTI data analysis reveals that the cyst induction of A. castellanii in response to various bacterial species does not correlate with the presence of PFT-producing bacteria. Moreover, A. castellanii demonstrates resistance to PFT-mediated cytotoxicity, in contrast to mammalian macrophages. Notably, Vibrio anguillarum and Ralstonia eutropha triggered a high frequency of cyst formation and cytotoxicity in infected A. castellanii. In summary, our findings reveal that A. castellanii exhibits a unique resistance to PFTs, unlike mammalian cells, suggesting its potential ecological role as a reservoir for diverse pathogenic species and its influence on their persistence and proliferation in the environment.
Collapse
Affiliation(s)
- Abdelbasset Yabrag
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
| | - Naeem Ullah
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
| | - Palwasha Baryalai
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden
| | - Irfan Ahmad
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, SE-75123, Uppsala, Sweden
| | - Nikola Zlatkov
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Eric Toh
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden
| | - Toril Lindbäck
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Bernt Eric Uhlin
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
| | - Sun Nyunt Wai
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden
| | - Aftab Nadeem
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187, Umeå, Sweden.
| |
Collapse
|
14
|
Li JG, Ying YL, Long YT. Aerolysin Nanopore Electrochemistry. Acc Chem Res 2025; 58:517-528. [PMID: 39874057 DOI: 10.1021/acs.accounts.4c00630] [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: 01/30/2025]
Abstract
Ions are the crucial signaling components for living organisms. In cells, their transportation across pore-forming membrane proteins is vital for regulating physiological functions, such as generating ionic current signals in response to target molecule recognition. This ion transport is affected by confined interactions and local environments within the protein pore. Therefore, the pore-forming protein can efficiently transduce the characteristics of each target molecule into ion-transport-mediated signals with high sensitivity. Inspired by nature, various protein pores have been developed into high-throughput and label-free nanopore sensors for single-molecule detection, enabling rapid and accurate readouts. In particular, aerolysin, a key virulence factor of Aeromonas hydrophila, exhibits a high sensitivity in generating ionic current fingerprints for detecting subtle differences in the sequence, conformation, and structure of DNA, proteins, polypeptides, oligosaccharides, and other molecules. Aerolysin features a cap that is approximately 14 nm wide on the cis side and a central pore that is about 10 nm long with a minimum diameter of around 1 nm. Its long lumen, with 11 charged rings at two entrances and neutral amino acids in between, facilitates the dwelling of the single analyte within the pore. This characteristic enables rich interactions between the well-defined residues within the pore and the analyte. As a result, the ionic current signal offers a unique molecular fingerprint, extending beyond the traditional volume exclusion model in nanopore sensing. In 2006, aerolysin was first reported to discriminate conformational differences of single peptides, opening the door for a rapidly growing field of aerolysin nanopore electrochemistry. Over the years, various mutant aerolysin nanopores have emerged, associated with advanced instrumentation and data analysis algorithms, enabling the simultaneous identification of over 30 targets with the number still increasing. Aerolysin nanopore electrochemistry in particular allows time-resolved qualitative and quantitative analysis ranging from DNA sequencing, proteomics, enzyme kinetics, and single-molecule reactions to potential clinical diagnostics. Especially, the feasibility of aerolysin nanopore electrochemistry in dynamic quantitative analysis would revolutionize omics studies at the single-molecule level, paving the way for the promising field of single-molecule temporal omics. Despite the success of this approach so far, it remains challenging to understand how confined interactions correlate to the distinguishable ionic signatures. Recent attempts have added correction terms to the volume exclusion model to account for variations in ion mobility within the nanopore caused by the confined interactions between the aerolysin and the analyte. Therefore, in this Account, we revisit the origin of the current blockade induced by target molecules inside the aerolysin nanopore. We highlight the contributions of the confined noncovalent interactions to the sensing ability of the aerolysin nanopore through the corrected conductance model. This Account then describes the design of interaction networks within the aerolysin nanopore, including electrostatic, hydrophobic, hydrogen-bonding, cation-π, and ion-charged amino acid interactions, for ultrasensitive biomolecular identification and quantification. Finally, we provide an outlook on further understanding the noncovalent interaction network inside the aerolysin nanopore, improving the manipulating and fine-tuning of confined electrochemistry toward a broad range of practical applications.
Collapse
Affiliation(s)
- Jun-Ge Li
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yi-Lun Ying
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Yi-Tao Long
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
15
|
Anton JS, Iacovache I, Bada Juarez JF, Abriata LA, Perrin LW, Cao C, Marcaida MJ, Zuber B, Dal Peraro M. Aerolysin Nanopore Structures Revealed at High Resolution in a Lipid Environment. J Am Chem Soc 2025; 147:4984-4992. [PMID: 39900531 PMCID: PMC11826888 DOI: 10.1021/jacs.4c14288] [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: 10/15/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
Aerolysin is a β-pore-forming toxin produced by most Aeromonas bacteria, which has attracted large attention in the field of nanopore sensing due to its narrow and charged pore lumen. Structurally similar proteins, belonging to the aerolysin-like family, are present throughout all kingdoms of life, but very few of them have been structurally characterized in a lipid environment. Here, we present the first high-resolution atomic cryo-EM structures of aerolysin prepore and pore in a membrane-like environment. These structures allow the identification of key interactions, which are relevant for understanding the pore formation mechanism and for correctly positioning the pore β-barrel and its anchoring β-turn motif in the membrane. Moreover, we elucidate at high resolution the architecture of key pore mutations and precisely identify four constriction rings in the pore lumen that are highly relevant for nanopore sensing experiments.
Collapse
Affiliation(s)
- Jana S. Anton
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ioan Iacovache
- Institute
of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| | - Juan F. Bada Juarez
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Luciano A. Abriata
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Louis W. Perrin
- Department
of Inorganic and Analytical Chemistry, Chemistry and Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Chan Cao
- Department
of Inorganic and Analytical Chemistry, Chemistry and Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Maria J. Marcaida
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Benoît Zuber
- Institute
of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| | - Matteo Dal Peraro
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
16
|
Lan X, Yang M, Wang J, Huang C, Wu A, Cui L, Guo Y, Zeng L, Guo X, Zhang Y, Xiang Y, Wang Q. Pore-Forming Protein LIN-24 Enhances Starvation Resilience in Caenorhabditis elegans by Modulating Lipid Metabolism and Mitochondrial Dynamics. Toxins (Basel) 2025; 17:72. [PMID: 39998089 PMCID: PMC11860826 DOI: 10.3390/toxins17020072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
The ability to survive starvation is a critical evolutionary adaptation, yet the molecular mechanisms underlying this capability remain incompletely understood. Pore-forming proteins (PFPs) are typically associated with immune defense, where they disturb the membranes of target cells. However, the role of PFPs in non-immune functions, particularly in metabolic and structural adaptations to starvation, is less explored. Here, we investigate the aerolysin-like PFP LIN-24 in Caenorhabditis elegans and uncover its novel function in enhancing starvation resistance. We found that LIN-24 expression is upregulated during starvation, leading to increased expression of the lipase-encoding gene lipl-3. This upregulation accelerates the mobilization and degradation of lipid stores, thereby sustaining energy levels. Additionally, LIN-24 overexpression significantly preserves muscle integrity, as evidenced by the maintenance of muscle structure compared to wild-type worms. Furthermore, we demonstrate that LIN-24 induces the formation of donut-shaped mitochondria, a structural change likely aimed at reducing ATP production to conserve energy during prolonged nutrient deprivation. This mitochondrial remodeling depends on genes involved in mitochondrial dynamics, including mff-1, mff-2, drp-1, and clk-1. Collectively, these findings expand our understanding of PFPs, demonstrating their multifaceted role in stress resistance beyond immune defense. LIN-24's involvement in regulating metabolism, preserving muscle structure, and remodeling mitochondria highlights its crucial role in the adaptive response to starvation, offering novel insights into the evolution of stress resistance mechanisms and potential therapeutic targets for conditions related to muscle preservation and metabolic regulation.
Collapse
Affiliation(s)
- Xinqiang Lan
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Mengqi Yang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Jiali Wang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Chunping Huang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Andong Wu
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Leilei Cui
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Yingqi Guo
- Institutional Center for Shared Technologies and Facilities of the Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650204, China; (Y.G.); (L.Z.)
| | - Lin Zeng
- Institutional Center for Shared Technologies and Facilities of the Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650204, China; (Y.G.); (L.Z.)
| | - Xiaolong Guo
- School of Physical Education, Yunnan Normal University, Kunming 650500, China;
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650204, China;
| | - Yang Xiang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| | - Qiquan Wang
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Nanchang University, Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China; (X.L.); (M.Y.); (J.W.); (C.H.); (A.W.); (L.C.)
| |
Collapse
|
17
|
Malter KE, Dunbar TL, Westin C, Darin E, Alfaro JR, Shikuma NJ. A bacterial membrane-disrupting protein stimulates animal metamorphosis. mBio 2025; 16:e0357324. [PMID: 39727418 PMCID: PMC11796346 DOI: 10.1128/mbio.03573-24] [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: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Diverse marine animals undergo a metamorphic larval-to-juvenile transition in response to surface-bound bacteria. Although this host-microbe interaction is critical to establishing and maintaining marine animal populations, the functional activity of bacterial products and how they activate the host's metamorphosis program has not yet been defined for any animal. The marine bacterium Pseudoalteromonas luteoviolacea stimulates the metamorphosis of a tubeworm called Hydroides elegans by producing a molecular syringe called metamorphosis-associated contractile structures (MACs). MACs stimulate metamorphosis by injecting a protein effector termed metamorphosis-inducing factor 1 (Mif1) into tubeworm larvae. Here, we show that MACs bind to tubeworm cilia and form visible pores on the cilia membrane surface, which are smaller and less numerous in the absence of Mif1. In vitro, Mif1 associates with eukaryotic lipid membranes and possesses phospholipase activity. MACs can also deliver Mif1 to human cell lines and cause parallel phenotypes, including cell surface binding, membrane disruption, calcium flux, and mitogen-activated protein kinase activation. Finally, MACs can also stimulate metamorphosis by delivering two unrelated membrane-disrupting proteins, MLKL and RegIIIɑ. Our findings demonstrate that membrane disruption by MACs and Mif1 is necessary for Hydroides metamorphosis, connecting the activity of a bacterial protein effector to the developmental transition of a marine animal. IMPORTANCE This research describes a mechanism wherein a bacterium prompts the metamorphic development of an animal from larva to juvenile form by injecting a protein that disrupts membranes in the larval cilia. Specifically, results show that a bacterial contractile injection system and the protein effector it injects form pores in larval cilia, influencing critical signaling pathways like mitogen-activated protein kinase and calcium flux, ultimately driving animal metamorphosis. This discovery sheds light on how a bacterial protein effector exerts its activity through membrane disruption, a phenomenon observed in various bacterial toxins affecting cellular functions, and elicits a developmental response. This work reveals a potential strategy used by marine organisms to respond to microbial cues, which could inform efforts in coral reef restoration and biofouling prevention. The study's insights into metamorphosis-associated contractile structures' delivery of protein effectors to specific anatomical locations highlight prospects for future biomedical and environmental applications.
Collapse
Affiliation(s)
- Kyle E. Malter
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Tiffany L. Dunbar
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Carl Westin
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Emily Darin
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Josefa Rivera Alfaro
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Nicholas J. Shikuma
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| |
Collapse
|
18
|
Piselli C. How to isolate channel-forming membrane proteins using the E. coli expression system. Nat Protoc 2025; 20:462-479. [PMID: 39367089 DOI: 10.1038/s41596-024-01055-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 07/08/2024] [Indexed: 10/06/2024]
Abstract
The recombinant expression, isolation and characterization of pore-forming proteins is one of the most commonly used strategies for understanding the permeability properties of the biological membrane into which they are embedded. This protocol describes how to quantify the expression of your protein of interest and use this information to optimize its production using the Escherichia coli strain BL21Gold(de3)ΔABCF. It explains with a step-by-step approach how to separate the bacterial compartments according to their solubility and how to extract your protein of interest in its native conformation using detergent solutions. Finally, it describes how to improve its purity via ion-exchange chromatography and insert the purified porins into outer membrane vesicles, from which they can be copurified. The protocol is simpler and less empirical than those described for most channel-forming membrane proteins and also provides a solid foundation for the isolation of soluble proteins. Several parameters can be optimized on a case-by-case basis: expression time and temperature, concentration of the inducer, nature and concentration of the detergent, incubation time and temperature, pH and ionic strength of the purification buffers. This protocol is effective with prokaryotic channel-forming membrane proteins and can be employed for the production of pore-forming proteins from chloroplasts, mitochondria or eukaryotes in general. With minor optimization, this protocol can be adapted for the isolation of receptors, carrier, pumps or any other membrane-active proteins.
Collapse
Affiliation(s)
- Claudio Piselli
- Department of Protein Evolution, Departmental Research Group Molecular Recognition and Catalysis, Max Planck Institute for Biology, Tubingen, Germany.
| |
Collapse
|
19
|
He X, Jiang X, Guo J, Sun H, Yang J. PANoptosis in Bacterial Infections: A Double-Edged Sword Balancing Host Immunity and Pathogenesis. Pathogens 2025; 14:43. [PMID: 39861004 PMCID: PMC11768250 DOI: 10.3390/pathogens14010043] [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: 12/10/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
PANoptosis is a newly identified programmed cell death pathway that integrates characteristics of apoptosis, pyroptosis, and necroptosis. It plays a dual role in the host immune response to bacterial infections. On one hand, PANoptosis acts as a protective mechanism by inducing the death of infected cells to eliminate pathogens and releasing pro-inflammatory cytokines to amplify the immune response. On the other hand, bacteria can exploit PANoptosis to evade host immune defenses. This dual nature underscores the potential of PANoptosis as a target for developing novel therapies against bacterial infections. This review summarizes the molecular mechanisms of PANoptosis, along with the crosstalk and integration of different cell death pathways in response to various bacterial pathogens. We also discuss the dual roles of PANoptosis in bacterial infectious diseases, including sepsis, pulmonary infections, and intestinal infections. Elucidating the molecular mechanisms underlying PANoptosis and how bacteria manipulate this pathway offers critical insights into host-pathogen interactions. These insights provide a foundation for designing targeted antibacterial strategies, modulating inflammation, and advancing precision medicine to improve clinical outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Jing Yang
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (X.H.); (X.J.); (J.G.); (H.S.)
| |
Collapse
|
20
|
Su Z, Chen T, Liu X, Kang X. Size-tunable transmembrane nanopores assembled from decomposable molecular templates. Biosens Bioelectron 2025; 267:116780. [PMID: 39277918 DOI: 10.1016/j.bios.2024.116780] [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: 05/18/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Transmembrane nanopores, as key elements in molecular transport and single-molecule sensors, are assembled naturally from multiple monomers in the presence of lipid bilayers. The nanopore size, especially the precise diameter of the inner space, determines its sensing targets and further biological application. In this paper, we introduce a template molecule-aided assembly strategy for constructing size-tunable transmembrane nanopores. Inspired by the barrel-like structure, similar to many transmembrane proteins, cyclodextrin molecules of different sizes are utilized as templates and modulators to assemble the α-helical barreled peptide of polysaccharide transporters (Wza). The functional nanopores assembled by this strategy possess high biological and chemical activity and can be inserted into lipid bilayers, forming stable single channels for single-molecule sensing. After enzyme digestion, the cyclodextrins on protein nanopores can be degraded, and the remaining nontemplate transmembrane protein nanopores can also preserve the integrity of their structure and function. The template molecule-aided assembly strategy employed a simple and convenient method for fully artificially synthesizing transmembrane protein nanopores; the pore size is completely dependent on the size of the template molecule and controllable, ranging from 1.1 to 1.8 nm. Furthermore, by chemically synthesized peptides and modifications, the pore function is easily modulated and does not involve the cumbersome genetic mutations of other biological techniques.
Collapse
Affiliation(s)
- Zhuoqun Su
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China; School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
| | - Tingting Chen
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China
| | - Xingtong Liu
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China
| | - Xiaofeng Kang
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
21
|
Abo-Zeid FS, Abdurabbah EM, Fares NH, Al-Karmalawy AA. Ultrastructural effects of Staphylococcus aureus toxicity on albino mice kidney. Microb Pathog 2025; 198:107110. [PMID: 39527986 DOI: 10.1016/j.micpath.2024.107110] [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: 04/18/2024] [Revised: 10/25/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Staphylococcus aureus (S. aureus) is a prominent infectious etiological agent in humans, dairy animals, and camels. Camel milk has all the nutrients which are nutritious and advantageous to the growth of S. aureus that dominates most bacterial species. So, the present work was designed to investigate the effect of certain toxin gene of S. aureus bacteria isolated from raw camel milk on the fine structure of mice kidneys. Two toxin genes of S. aureus were identified and described through PCR using 23s rRNA primer for 23s rRNA gene. In addition, the effect of the isolated S. aureus bacteria, especially those carrying the gene "tst" was studied on the ultrastructure of mice kidney. Twenty male albino mice were allocated into 2 groups, control group and infected group (orally administered with a single dosage of S. aureus aqueous solutions from camel milk at a concentration of 5 × 108 colony forming unit/0.1 mL for three days). Infection of mice with S. aureus resulted in vacuolation, necrosis, and degeneration of tubular epithelial cells, intertubular congestion and inflammation, as well as dilatation and congestion of glomeruli. Therefore, this current study most likely indicates that camel milk infected with S. aureus has a significant and clear impact in inducing symptoms of kidney failure in communities that use these dairy products without health supervision, especially nomadic and pastoral communities.
Collapse
Affiliation(s)
- Faten S Abo-Zeid
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Ebtisam M Abdurabbah
- Department of Zoology, Faculty of Science, Omar Al-Mukhtar University, Al-Bayda, Libya
| | - Nagui H Fares
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt.
| |
Collapse
|
22
|
Snoj T, Lukan T, Gruden K, Anderluh G. Interaction of an Oomycete Nep1-like Cytolysin with Natural and Plant Cell-Mimicking Membranes. J Membr Biol 2024:10.1007/s00232-024-00330-3. [PMID: 39692881 DOI: 10.1007/s00232-024-00330-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 11/30/2024] [Indexed: 12/19/2024]
Abstract
Plants are attacked by various pathogens that secrete a variety of effectors to damage host cells and facilitate infection. One of the largest and so far understudied microbial protein families of effectors is necrosis- and ethylene-inducing peptide-1-like proteins (NLPs), which are involved in important plant diseases. Many NLPs act as cytolytic toxins that cause cell death and tissue necrosis by disrupting the plant's plasma membrane. Their mechanism of action is unique and leads to the formation of small, transient membrane ruptures. Here, we capture the interaction of the cytotoxic model NLP from the oomycete Pythium aphanidermatum, NLPPya, with plant cell-mimicking membranes of giant unilamellar vesicles (GUVs) and tobacco protoplasts using confocal fluorescence microscopy. We show that the permeabilization of GUVs by NLPPya is concentration- and time-dependent, confirm the small size of the pores by observing the inability of NLPPya monomers to pass through them, image the morphological changes of GUVs at higher concentrations of NLPPya and confirm its oligomerization on the membrane of GUVs. In addition, NLPPya bound to plasma membranes of protoplasts, which showed varying responses. Our results provide new insights into the interaction of NLPPya with model lipid membranes containing plant-derived sphingolipids.
Collapse
Affiliation(s)
- Tina Snoj
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
- Graduate School of Biosciences, Biotehnical Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Tjaša Lukan
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia.
| |
Collapse
|
23
|
Zárate-Potes A, Schulenburg H, Dierking K. Unanticipated specificity in effector-triggered immunity. Trends Immunol 2024; 45:939-942. [PMID: 39550315 DOI: 10.1016/j.it.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/18/2024]
Abstract
Effector-triggered immunity (ETI) enables hosts to react to pathogens by monitoring few key cellular processes. ETI responses are assumed to be similar toward related pathogen effectors. However, recent evidence from the invertebrate model Caenorhabditis elegans and pore-forming toxins indicates a much more complex and specific ETI than previously anticipated.
Collapse
Affiliation(s)
- Alejandra Zárate-Potes
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Hinrich Schulenburg
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Am Botanischen Garten 1-9, 24118 Kiel, Germany; Max Planck Institute for Evolutionary Biology, 24306 Ploen, Germany.
| | - Katja Dierking
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Am Botanischen Garten 1-9, 24118 Kiel, Germany.
| |
Collapse
|
24
|
Meyer N, Torrent J, Balme S. Characterizing Prion-Like Protein Aggregation: Emerging Nanopore-Based Approaches. SMALL METHODS 2024; 8:e2400058. [PMID: 38644684 PMCID: PMC11672191 DOI: 10.1002/smtd.202400058] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/10/2024] [Indexed: 04/23/2024]
Abstract
Prion-like protein aggregation is characteristic of numerous neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. This process involves the formation of aggregates ranging from small and potentially neurotoxic oligomers to highly structured self-propagating amyloid fibrils. Various approaches are used to study protein aggregation, but they do not always provide continuous information on the polymorphic, transient, and heterogeneous species formed. This review provides an updated state-of-the-art approach to the detection and characterization of a wide range of protein aggregates using nanopore technology. For each type of nanopore, biological, solid-state polymer, and nanopipette, discuss the main achievements for the detection of protein aggregates as well as the significant contributions to the understanding of protein aggregation and diagnostics.
Collapse
Affiliation(s)
- Nathan Meyer
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonCedex 5Montpellier34095France
- INMUniversity of MontpellierINSERMMontpellier34095France
| | - Joan Torrent
- INMUniversity of MontpellierINSERMMontpellier34095France
| | - Sébastien Balme
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonCedex 5Montpellier34095France
| |
Collapse
|
25
|
Liu H, Zhang W, He Q, Aikemu R, Xu H, Guo Z, Wang L, Li W, Wang G, Wang X, Guo W. Re-localization of a repeat-containing fungal effector by apoplastic protein Chitinase-like 1 blocks its toxicity. Nat Commun 2024; 15:10122. [PMID: 39578470 PMCID: PMC11584738 DOI: 10.1038/s41467-024-54470-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
A fungal effector that is toxic to plant cells was identified in Verticillium dahliae. The effector contains a non-canonical Common in several Fungal Extracellular Membrane proteins (CFEM) domain, a tandem repeat region consisting of four 14-amino acid repeats rich in proline, and a C-terminal region, thus is designated V. dahliae tetrapeptide repeat protein (VdTRP). The membrane targeting of VdTRP is vital for its cell toxicity. CFEM mediates the membrane targeting and the tandem repeat region exerts the toxic function upon cell membrane. The chitinase-like 1 (CTL1), an essential apoplastic protein of cotton, can redirect VdTRP from cell membrane to apoplast. Transgenic cotton overexpressing CTL1 greatly enhances cotton resistance to V. dahliae without affecting cotton growth and development, implicating its potential application in breeding cotton with high wilt resistance. Our data demonstrates that genetic manipulation of effector target constitutes potential strategy for improving crop resistance to fungal pathogens.
Collapse
Affiliation(s)
- Hanqiao Liu
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wenshu Zhang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qinqfei He
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Reyila Aikemu
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Huijuan Xu
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhan Guo
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lu Wang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weixi Li
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guilin Wang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xinyu Wang
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China.
- College of Life Sciences, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Wangzhen Guo
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing, 210095, China.
- Engineering Research Center of Ministry of Education for Cotton Germplasm Enhancement and Application, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
26
|
Liu X, Ni Y, Ye L, Guo Z, Tan L, Li J, Yang M, Chen S, Li R. Nanopore strand-specific mismatch enables de novo detection of bacterial DNA modifications. Genome Res 2024; 34:2025-2038. [PMID: 39358016 PMCID: PMC11610603 DOI: 10.1101/gr.279012.124] [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: 01/30/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
DNA modifications in bacteria present diverse types and distributions, playing crucial functional roles. Current methods for detecting bacterial DNA modifications via nanopore sequencing typically involve comparing raw current signals to a methylation-free control. In this study, we found that bacterial DNA modification induces errors in nanopore reads. And these errors are found only in one strand but not the other, showing a strand-specific bias. Leveraging this discovery, we developed Hammerhead, a pioneering pipeline designed for de novo methylation discovery that circumvents the necessity of raw signal inference and a methylation-free control. The majority (14 out of 16) of the identified motifs can be validated by raw signal comparison methods or by identifying corresponding methyltransferases in bacteria. Additionally, we included a novel polishing strategy employing duplex reads to correct modification-induced errors in bacterial genome assemblies, achieving a reduction of over 85% in such errors. In summary, Hammerhead enables users to effectively locate bacterial DNA methylation sites from nanopore FASTQ/FASTA reads, thus holds promise as a routine pipeline for a wide range of nanopore sequencing applications, such as genome assembly, metagenomic binning, decontaminating eukaryotic genome assemblies, and functional analysis for DNA modifications.
Collapse
Affiliation(s)
- Xudong Liu
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
| | - Ying Ni
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518000, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong 999077, China
| | - Lianwei Ye
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
| | - Zhihao Guo
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
| | - Lu Tan
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
| | - Jun Li
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
| | - Mengsu Yang
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518000, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518000, China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Runsheng Li
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong 999077, China;
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518000, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
27
|
Roesel T, Cao C, Bada Juarez JF, Dal Peraro M, Roke S. Dissecting the Membrane Association Mechanism of Aerolysin Pores at Femtomolar Concentrations Using Water as a Probe. NANO LETTERS 2024; 24:13888-13894. [PMID: 39469905 PMCID: PMC11544699 DOI: 10.1021/acs.nanolett.4c00035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024]
Abstract
Aerolysin is a bacterial toxin that forms transmembrane pores at the host plasma membrane and has a narrow internal diameter and great stability. These assets make it a highly promising nanopore for detecting biopolymers such as nucleic acids and peptides. Although much is known about aerolysin from a microbiological and structural perspective, its membrane association and pore-formation mechanism are not yet fully understood. Here, we used angle-resolved second harmonic scattering (AR-SHS) and single-channel current measurements to investigate how wild-type (wt) aerolysin and its mutants interact with liposomes in aqueous solutions at femtomolar concentrations. Our AR-SHS experiments were sensitive enough to detect changes in the electrostatic properties of membrane-bound aerolysin, which were induced by variations in pH levels. We reported for the first time the membrane binding affinity of aerolysin at different stages of the pore formation mechanism: while wt aerolysin has a binding affinity as high as 20 fM, the quasi-pore and the prepore states show gradually decreasing membrane affinities, incomplete insertion, and a pore opening signature. Moreover, we quantitatively characterized the membrane affinity of mutants relevant for applications to nanopore sensing. Our study provides a label-free method for efficiently screening biological pores suitable for conducting molecular sensing and sequencing measurements as well as for probing pore-forming processes.
Collapse
Affiliation(s)
- Tereza Roesel
- Laboratory
for Fundamental BioPhotonics (LBP), Institute of Bioengineering (IBI),
and Institute of Materials Science (IMX), School of Engineering (STI),
and Lausanne Centre for Ultrafast Science (LACUS), École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Chan Cao
- Department
of Inorganic and Analytical Chemistry, School of Chemistry and Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Juan F. Bada Juarez
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Matteo Dal Peraro
- Institute
of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Sylvie Roke
- Laboratory
for Fundamental BioPhotonics (LBP), Institute of Bioengineering (IBI),
and Institute of Materials Science (IMX), School of Engineering (STI),
and Lausanne Centre for Ultrafast Science (LACUS), École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
28
|
Zhang J, Feng K, Shen WT, Gao W, Zhang L. Research Advances of Cellular Nanoparticles as Multiplex Countermeasures. ACS NANO 2024; 18:30211-30223. [PMID: 39441568 PMCID: PMC11544708 DOI: 10.1021/acsnano.4c09830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Cellular nanoparticles (CNPs), fabricated by coating natural cell membranes onto nanoparticle cores, have been widely used to replicate cellular functions for various therapeutic applications. Specifically, CNPs act as cell decoys, binding harmful molecules or infectious pathogens and neutralizing their bioactivity. This neutralization strategy leverages the target's functional properties rather than its structure, resulting in broad-spectrum efficacy. Since their inception, CNP platforms have undergone significant advancements to enhance their neutralizing capabilities and efficiency. This review traces the research advances of CNP technology as multiplex countermeasures across four categories with progressive functions: neutralization through cell membrane binding, simultaneous neutralization using both cell membrane and nanoparticle core, continuous neutralization via enzymatic degradation, and enhanced neutralization through membrane modification. The review highlights the structure-property relationship in CNP designs, showing the functional advances of each category of CNP. By providing an overview of CNPs in multiplex neutralization of a wide range of chemical and biological threat agents, this article aims to inspire the development of more advanced CNP nanoformulations and uncover innovative applications to address unresolved medical challenges.
Collapse
Affiliation(s)
- Jiayuan
Alex Zhang
- Aiiso Yufeng Li Family Department of
Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Kailin Feng
- Aiiso Yufeng Li Family Department of
Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Wei-Ting Shen
- Aiiso Yufeng Li Family Department of
Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Weiwei Gao
- Aiiso Yufeng Li Family Department of
Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| | - Liangfang Zhang
- Aiiso Yufeng Li Family Department of
Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell
Collaboratory, University of California
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
29
|
Caliot E, Firon A, Solgadi A, Trieu-Cuot P, Dramsi S. Lipid lysination by MprF contributes to hemolytic pigment retention in group B Streptococcus. Res Microbiol 2024; 175:104231. [PMID: 39197696 DOI: 10.1016/j.resmic.2024.104231] [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: 05/03/2024] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Group B Streptococcus (GBS) is the leading cause of neonatal sepsis and meningitis. A major virulence factor is a pigmented beta-haemolytic/cyto-lysin (β-h/c) toxin with an ornithine rhamnolipid structure. We initially observed that absence of MprF enzyme altered pigmentation and haemolytic activity in GBS. Next, we showed that MprF-dependent lipid lysination contributes to the retention of the ornithine rhamnolipid within GBS membrane. Furthermore, cationic lipidation by MprF altered membrane properties contributing to resistance to the cyclic lipopeptide daptomycin and to acidic pH. This study highlights the importance of cationic lipids in cell envelope homeostasis and in modulating β-h/c activity.
Collapse
Affiliation(s)
- Elise Caliot
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Biology of Gram-positive Pathogens Unit, F-75015 Paris, France
| | - Arnaud Firon
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Biology of Gram-positive Pathogens Unit, F-75015 Paris, France
| | - Audrey Solgadi
- UMS-IPSIT SAMM Facility, Université Paris-Saclay, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, F-91400 Orsay, France
| | - Patrick Trieu-Cuot
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Biology of Gram-positive Pathogens Unit, F-75015 Paris, France.
| | - Shaynoor Dramsi
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Biology of Gram-positive Pathogens Unit, F-75015 Paris, France.
| |
Collapse
|
30
|
Cases M, Dorca-Arévalo J, Blanch M, Rodil S, Terni B, Martín-Satué M, Llobet A, Blasi J, Solsona C. The epsilon toxin from Clostridium perfringens stimulates calcium-activated chloride channels, generating extracellular vesicles in Xenopus oocytes. Pharmacol Res Perspect 2024; 12:e70005. [PMID: 39320019 PMCID: PMC11423345 DOI: 10.1002/prp2.70005] [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: 02/23/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 09/26/2024] Open
Abstract
The epsilon toxin (Etx) from Clostridium perfringens has been identified as a potential trigger of multiple sclerosis, functioning as a pore-forming toxin that selectively targets cells expressing the plasma membrane (PM) myelin and lymphocyte protein (MAL). Previously, we observed that Etx induces the release of intracellular ATP in sensitive cell lines. Here, we aimed to re-examine the mechanism of action of the toxin and investigate the connection between pore formation and ATP release. We examined the impact of Etx on Xenopus laevis oocytes expressing human MAL. Extracellular ATP was assessed using the luciferin-luciferase reaction. Activation of calcium-activated chloride channels (CaCCs) and a decrease in the PM surface were recorded using the two-electrode voltage-clamp technique. To evaluate intracellular Ca2+ levels and scramblase activity, fluorescent dyes were employed. Extracellular vesicles were imaged using light and electron microscopy, while toxin oligomers were identified through western blots. Etx triggered intracellular Ca2+ mobilization in the Xenopus oocytes expressing hMAL, leading to the activation of CaCCs, ATP release, and a reduction in PM capacitance. The toxin induced the activation of scramblase and, thus, translocated phospholipids from the inner to the outer leaflet of the PM, exposing phosphatidylserine outside in Xenopus oocytes and in an Etx-sensitive cell line. Moreover, Etx caused the formation of extracellular vesicles, not derived from apoptotic bodies, through PM fission. These vesicles carried toxin heptamers and doughnut-like structures in the nanometer size range. In conclusion, ATP release was not directly attributed to the formation of pores in the PM, but to scramblase activity and the formation of extracellular vesicles.
Collapse
Affiliation(s)
- Mercè Cases
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Jonatan Dorca-Arévalo
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Marta Blanch
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Sergi Rodil
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
| | - Beatrice Terni
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Mireia Martín-Satué
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
| | - Artur Llobet
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Juan Blasi
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Carles Solsona
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences-Campus Bellvitge, University of Barcelona, Barcelona, Spain
- Laboratory of Molecular and Cellular Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Tang J, Song H, Li S, Lam SM, Ping J, Yang M, Li N, Chang T, Yu Z, Liu W, Lu Y, Zhu M, Tang Z, Liu Z, Guo YR, Shui G, Veillette A, Zeng Z, Wu N. TMEM16F Expressed in Kupffer Cells Regulates Liver Inflammation and Metabolism to Protect Against Listeria Monocytogenes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402693. [PMID: 39136057 PMCID: PMC11497084 DOI: 10.1002/advs.202402693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/20/2024] [Indexed: 10/25/2024]
Abstract
Infection by bacteria leads to tissue damage and inflammation, which need to be tightly controlled by host mechanisms to avoid deleterious consequences. It is previously reported that TMEM16F, a calcium-activated lipid scramblase expressed in various immune cell types including T cells and neutrophils, is critical for the control of infection by bacterium Listeria monocytogenes (Lm) in vivo. This function correlated with the capacity of TMEM16F to repair the plasma membrane (PM) damage induced in T cells in vitro, by the Lm toxin listeriolysin O (LLO). However, whether the protective effect of TMEM16F on Lm infection in vivo is mediated by an impact in T cells, or in other cell types, is not determined. Herein, the immune cell types and mechanisms implicated in the protective effect of TMEM16F against Lm in vivo are elucidated. Cellular protective effects of TMEM16F correlated with its capacity of lipid scrambling and augment PM fluidity. Using cell type-specific TMEM16F-deficient mice, the indication is obtained that TMEM16F expressed in liver Kupffer cells (KCs), but not in T cells or B cells, is key for protection against Listeria in vivo. In the absence of TMEM16F, Listeria induced PM rupture and fragmentation of KCs in vivo. KC death associated with greater liver damage, inflammatory changes, and dysregulated liver metabolism. Overall, the results uncovered that TMEM16F expressed in Kupffer cells is crucial to protect the host against Listeria infection. This influence is associated with the capacity of Kupffer cell-expressed TMEM16F to prevent excessive inflammation and abnormal liver metabolism.
Collapse
Affiliation(s)
- Jianlong Tang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Hua Song
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
| | - Shimin Li
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - Jieming Ping
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Mengyun Yang
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Na Li
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Teding Chang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ze Yu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Weixiang Liu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
| | - Yan Lu
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Min Zhu
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zhaohui Tang
- Department of Traumatic SurgeryTongji Trauma CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Zheng Liu
- Department of Otolaryngology‐Head and Neck SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyNo. 1095 Jiefang AvenueWuhan430030China
| | - Yusong R. Guo
- Department of biochemistry and molecular biologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100101China
| | - André Veillette
- Laboratory of Molecular OncologyInstitut de recherches cliniques de Montréal (IRCM)MontréalQuébecH2W1R7Canada
- Department of MedicineUniversity of MontréalMontréalQuébecH3T 1J4Canada
- Department of MedicineMcGill UniversityMontréalQuébecH3G 1Y6Canada
| | - Zhutian Zeng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Department of OncologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefei230001China
| | - Ning Wu
- Department of ImmunologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and Technology (HUST)Wuhan430030China
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical ImmunologyAnhui Medical UniversityHefei230032China
- Cell Architecture Research CenterTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
32
|
Martin CL, Hill JH, Aller SG. Host Tropism and Structural Biology of ABC Toxin Complexes. Toxins (Basel) 2024; 16:406. [PMID: 39330864 PMCID: PMC11435725 DOI: 10.3390/toxins16090406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
ABC toxin complexes are a class of protein toxin translocases comprised of a multimeric assembly of protein subunits. Each subunit displays a unique composition, contributing to the formation of a syringe-like nano-machine with natural cargo carrying, targeting, and translocation capabilities. Many of these toxins are insecticidal, drawing increasing interest in agriculture for use as biological pesticides. The A subunit (TcA) is the largest subunit of the complex and contains domains associated with membrane permeation and targeting. The B and C subunits, TcB and TcC, respectively, package into a cocoon-like structure that contains a toxic peptide and are coupled to TcA to form a continuous channel upon final assembly. In this review, we outline the current understanding and gaps in the knowledge pertaining to ABC toxins, highlighting seven published structures of TcAs and how these structures have led to a better understanding of the mechanism of host tropism and toxin translocation. We also highlight similarities and differences between homologues that contribute to variations in host specificity and conformational change. Lastly, we review the biotechnological potential of ABC toxins as both pesticides and cargo-carrying shuttles that enable the transport of peptides into cells.
Collapse
Affiliation(s)
- Cole L. Martin
- Graduate Biomedical Sciences Pathobiology, Physiology and Pharmacology Theme, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - John H. Hill
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Stephen G. Aller
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
33
|
Liguori F, Pellicciotta N, Milanetti E, Xi Windemuth S, Ruocco G, Di Leonardo R, Danino T. Dynamic Gene Expression Mitigates Mutational Escape in Lysis-Driven Bacteria Cancer Therapy. BIODESIGN RESEARCH 2024; 6:0049. [PMID: 39301524 PMCID: PMC11411163 DOI: 10.34133/bdr.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/09/2024] [Accepted: 08/25/2024] [Indexed: 09/22/2024] Open
Abstract
Engineered bacteria have the potential to deliver therapeutic payloads directly to tumors, with synthetic biology enabling precise control over therapeutic release in space and time. However, it remains unclear how to optimize therapeutic bacteria for durable colonization and sustained payload release. Here, we characterize nonpathogenic Escherichia coli expressing the bacterial toxin Perfringolysin O (PFO) and dynamic strategies that optimize therapeutic efficacy. While PFO is known for its potent cancer cell cytotoxicity, we present experimental evidence that expression of PFO causes lysis of bacteria in both batch culture and microfluidic systems, facilitating its efficient release. However, prolonged expression of PFO leads to the emergence of a mutant population that limits therapeutic-releasing bacteria in a PFO expression level-dependent manner. We present sequencing data revealing the mutant takeover and employ molecular dynamics to confirm that the observed mutations inhibit the lysis efficiency of PFO. To analyze this further, we developed a mathematical model describing the evolution of therapeutic-releasing and mutant bacteria populations revealing trade-offs between therapeutic load delivered and fraction of mutants that arise. We demonstrate that a dynamic strategy employing short and repeated inductions of the pfo gene better preserves the original population of therapeutic bacteria by mitigating the effects of mutational escape. Altogether, we demonstrate how dynamic modulation of gene expression can address mutant takeovers giving rise to limitations in engineered bacteria for therapeutic applications.
Collapse
Affiliation(s)
- Filippo Liguori
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Nicola Pellicciotta
- NANOTEC-CNR, Soft and Living Matter Laboratory, Institute of Nanotechnology, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Edoardo Milanetti
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sophia Xi Windemuth
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Giancarlo Ruocco
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Roberto Di Leonardo
- Department of Physics, Sapienza University of Rome, Rome, Italy
- NANOTEC-CNR, Soft and Living Matter Laboratory, Institute of Nanotechnology, Rome, Italy
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| |
Collapse
|
34
|
Artuyants A, Hong J, Dauros-Singorenko P, Phillips A, Simoes-Barbosa A. Lactobacillus gasseri and Gardnerella vaginalis produce extracellular vesicles that contribute to the function of the vaginal microbiome and modulate host-Trichomonas vaginalis interactions. Mol Microbiol 2024; 122:357-371. [PMID: 37485746 DOI: 10.1111/mmi.15130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/22/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
Trichomonas vaginalis is an extracellular protozoan parasite of the human urogenital tract, responsible for a prevalent sexually transmitted infection. Trichomoniasis is accompanied by a dysbiotic microbiome that is characterised by the depletion of host-protective commensals such as Lactobacillus gasseri, and the flourishing of a bacterial consortium that is comparable to the one seen for bacterial vaginosis, including the founder species Gardnerella vaginalis. These two vaginal bacteria are known to have opposite effects on T. vaginalis pathogenicity. Studies on extracellular vesicles (EVs) have been focused on the direction of a microbial producer (commensal or pathogen) to a host recipient, and largely in the context of the gut microbiome. Here, taking advantage of the simplicity of the human cervicovaginal microbiome, we determined the molecular cargo of EVs produced by L. gasseri and G. vaginalis and examined how these vesicles modulate the interaction of T. vaginalis and host cells. We show that these EVs carry a specific cargo of proteins, which functions can be attributed to the opposite roles that these bacteria play in the vaginal biome. Furthermore, these bacterial EVs are delivered to host and protozoan cells, modulating host-pathogen interactions in a way that mimics the opposite effects that these bacteria have on T. vaginalis pathogenicity. This is the first study to describe side-by-side the protein composition of EVs produced by two bacteria belonging to the opposite spectrum of a microbiome and to demonstrate that these vesicles modulate the pathogenicity of a protozoan parasite. Such as in trichomoniasis, infections and dysbiosis co-occur frequently resulting in significant co-morbidities. Therefore, studies like this provide the knowledge for the development of antimicrobial therapies that aim to clear the infection while restoring a healthy microbiome.
Collapse
Affiliation(s)
| | - Jiwon Hong
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, University of Auckland, Auckland, New Zealand
| | | | - Anthony Phillips
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Surgical and Translational Research Centre, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
35
|
Wu G, Chen J, Wang A, Yan F. Unveiling the viroporin arsenal in plant viruses: Implications for the future. PLoS Pathog 2024; 20:e1012473. [PMID: 39235994 PMCID: PMC11376509 DOI: 10.1371/journal.ppat.1012473] [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: 09/07/2024] Open
Abstract
Viroporins are small, hydrophobic viral proteins that modify cellular membranes to form tiny pores for influx of ions and small molecules. Previously, viroporins were identified exclusively in vertebrate viruses. Recent studies have shown that both plant-infecting positive-sense single-stranded (+ss) and negative-sense single-stranded (-ss) RNA viruses also encode functional viroporins. These seminal discoveries not only advance our understanding of the distribution and evolution of viroporins, but also open up a new field of plant virus research.
Collapse
Affiliation(s)
- Guanwei Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agroproducts, Institute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Provincial Key Laboratory of Green Plant Protection, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jianping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agroproducts, Institute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Provincial Key Laboratory of Green Plant Protection, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Aiming Wang
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, Ontario, Canada
| | - Fei Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agroproducts, Institute of Plant Virology, Ningbo University, Ningbo, China
- Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Provincial Key Laboratory of Green Plant Protection, Institute of Plant Virology, Ningbo University, Ningbo, China
| |
Collapse
|
36
|
Wickramasinghe DN, Lyon CM, Lee S, Hepworth OW, Priest EL, Maufrais C, Ryan AP, Permal E, Sullivan D, McManus BA, Hube B, Butler G, d'Enfert C, Naglik JR, Richardson JP. Variations in candidalysin amino acid sequence influence toxicity and host responses. mBio 2024; 15:e0335123. [PMID: 38953356 PMCID: PMC11323794 DOI: 10.1128/mbio.03351-23] [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: 12/09/2023] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
Candida albicans causes millions of mucosal infections in humans annually. Hyphal overgrowth on mucosal surfaces is frequently associated with tissue damage caused by candidalysin, a secreted peptide toxin that destabilizes the plasma membrane of host cells thereby promoting disease and immunopathology. Candidalysin was first identified in C. albicans strain SC5314, but recent investigations have revealed candidalysin "variants" of differing amino acid sequence in isolates of C. albicans, and the related species C. dubliniensis, and C tropicalis, suggesting that sequence variation among candidalysins may be widespread in natural populations of these Candida species. Here, we analyzed ECE1 gene sequences from 182 C. albicans isolates, 10 C. dubliniensis isolates, and 78 C. tropicalis isolates and identified 10, 3, and 2 candidalysin variants in these species, respectively. Application of candidalysin variants to epithelial cells revealed differences in the ability to cause cellular damage, changes in metabolic activity, calcium influx, MAPK signalling, and cytokine secretion, while biophysical analyses indicated that variants exhibited differences in their ability to interact with and permeabilize a membrane. This study identifies candidalysin variants with differences in biological activity that are present in medically relevant Candida species. IMPORTANCE Fungal infections are a significant burden to health. Candidalysin is a toxin produced by Candida albicans that damages host tissues, facilitating infection. Previously, we demonstrated that candidalysins exist in the related species C. dubliniensis and C. tropicalis, thereby identifying these molecules as a toxin family. Recent genomic analyses have highlighted the presence of a small number of candidalysin "variant" toxins, which have different amino acid sequences to those originally identified. Here, we screened genome sequences of isolates of C. albicans, C. dubliniensis, and C. tropicalis and identified candidalysin variants in all three species. When applied to epithelial cells, candidalysin variants differed in their ability to cause damage, activate intracellular signaling pathways, and induce innate immune responses, while biophysical analysis revealed differences in the ability of candidalysin variants to interact with lipid bilayers. These findings suggest that intraspecies variation in candidalysin amino acid sequence may influence fungal pathogenicity.
Collapse
Affiliation(s)
- Don N. Wickramasinghe
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Claire M. Lyon
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Sejeong Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Olivia W. Hepworth
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Emily L. Priest
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Corinne Maufrais
- Institut Pasteur, Université Paris Cité, INRAe USC 2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Adam P. Ryan
- School of Biomedical and Biomolecular Science and UCD Conway Institute of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Emmanuelle Permal
- Institut Pasteur, Université Paris Cité, INRAe USC 2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Derek Sullivan
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Brenda A. McManus
- Division of Oral Biosciences, Dublin Dental University Hospital, and School of Dental Science, Trinity College Dublin, Dublin, Ireland
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Geraldine Butler
- School of Biomedical and Biomolecular Science and UCD Conway Institute of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Christophe d'Enfert
- Institut Pasteur, Université Paris Cité, INRAe USC 2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Julian R. Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| | - Jonathan P. Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
37
|
Desai N, Pande S, Salave S, Singh TRR, Vora LK. Antitoxin nanoparticles: design considerations, functional mechanisms, and applications in toxin neutralization. Drug Discov Today 2024; 29:104060. [PMID: 38866357 DOI: 10.1016/j.drudis.2024.104060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
The application of nanotechnology has significantly advanced the development of novel platforms that enhance disease treatment and diagnosis. A key innovation in this field is the creation of antitoxin nanoparticles (ATNs), designed to address toxin exposure. These precision-engineered nanosystems have unique physicochemical properties and selective binding capabilities, allowing them to effectively capture and neutralize toxins from various biological, chemical, and environmental sources. In this review, we thoroughly examine their therapeutic and diagnostic potential for managing toxin-related challenges. We also explore recent advancements and offer critical insights into the design and clinical implementation of ATNs.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Kandi, Telangana, India
| | - Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Kandi, Telangana, India
| | - Sagar Salave
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat, India
| | | | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
38
|
Guo Z, Zhu AT, Wei X, Jiang Y, Yu Y, Noh I, Gao W, Fang RH, Zhang L. A genetically engineered neuronal membrane-based nanotoxoid elicits protective immunity against neurotoxins. Bioact Mater 2024; 38:321-330. [PMID: 38764446 PMCID: PMC11101676 DOI: 10.1016/j.bioactmat.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Given their dangerous effects on the nervous system, neurotoxins represent a significant threat to public health. Various therapeutic approaches, including chelating agents, receptor decoys, and toxin-neutralizing antibodies, have been explored. While prophylactic vaccines are desirable, it is oftentimes difficult to effectively balance their safety and efficacy given the highly dangerous nature of neurotoxins. To address this, we report here on a nanovaccine against neurotoxins that leverages the detoxifying properties of cell membrane-coated nanoparticles. A genetically modified cell line with constitutive overexpression of the α7 nicotinic acetylcholine receptor is developed as a membrane source to generate biomimetic nanoparticles that can effectively and irreversibly bind to α-bungarotoxin, a model neurotoxin. This abrogates the biological activity of the toxin, enabling the resulting nanotoxoid to be safely delivered into the body and processed by the immune system. When co-administered with an immunological adjuvant, a strong humoral response against α-bungarotoxin is generated that protects vaccinated mice against a lethal dose of the toxin. Overall, this work highlights the potential of using genetic modification strategies to develop nanotoxoid formulations against various biological threats.
Collapse
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Audrey T. Zhu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xiaoli Wei
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yiyan Yu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ilkoo Noh
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
39
|
Berhanu S, Majumder S, Müntener T, Whitehouse J, Berner C, Bera AK, Kang A, Liang B, Khan N, Sankaran B, Tamm LK, Brockwell DJ, Hiller S, Radford SE, Baker D, Vorobieva AA. Sculpting conducting nanopore size and shape through de novo protein design. Science 2024; 385:282-288. [PMID: 39024453 PMCID: PMC11549965 DOI: 10.1126/science.adn3796] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/03/2024] [Indexed: 07/20/2024]
Abstract
Transmembrane β-barrels have considerable potential for a broad range of sensing applications. Current engineering approaches for nanopore sensors are limited to naturally occurring channels, which provide suboptimal starting points. By contrast, de novo protein design can in principle create an unlimited number of new nanopores with any desired properties. Here we describe a general approach to designing transmembrane β-barrel pores with different diameters and pore geometries. Nuclear magnetic resonance and crystallographic characterization show that the designs are stably folded with structures resembling those of the design models. The designs have distinct conductances that correlate with their pore diameter, ranging from 110 picosiemens (~0.5 nanometer pore diameter) to 430 picosiemens (~1.1 nanometer pore diameter). Our approach opens the door to the custom design of transmembrane nanopores for sensing and sequencing applications.
Collapse
Affiliation(s)
- Samuel Berhanu
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Sagardip Majumder
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - James Whitehouse
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT
| | - Carolin Berner
- Structural Biology Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- VUB-VIB Center for Structural Biology, Brussels, Belgium
| | - Asim K. Bera
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Binyong Liang
- Department of Molecular Physiology and Biological Physics and Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Nasir Khan
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Lukas K. Tamm
- Department of Molecular Physiology and Biological Physics and Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - David J. Brockwell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT
| | | | - Sheena E. Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT
| | - David Baker
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Anastassia A. Vorobieva
- Structural Biology Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- VUB-VIB Center for Structural Biology, Brussels, Belgium
- VIB Center for AI and Computational Biology, Belgium
| |
Collapse
|
40
|
Kulma M, Šakanović A, Bedina-Zavec A, Caserman S, Omersa N, Šolinc G, Orehek S, Hafner-Bratkovič I, Kuhar U, Slavec B, Krapež U, Ocepek M, Kobayashi T, Kwiatkowska K, Jerala R, Podobnik M, Anderluh G. Sequestration of membrane cholesterol by cholesterol-binding proteins inhibits SARS-CoV-2 entry into Vero E6 cells. Biochem Biophys Res Commun 2024; 716:149954. [PMID: 38704887 DOI: 10.1016/j.bbrc.2024.149954] [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: 02/13/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
Membrane lipids and proteins form dynamic domains crucial for physiological and pathophysiological processes, including viral infection. Many plasma membrane proteins, residing within membrane domains enriched with cholesterol (CHOL) and sphingomyelin (SM), serve as receptors for attachment and entry of viruses into the host cell. Among these, human coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use proteins associated with membrane domains for initial binding and internalization. We hypothesized that the interaction of lipid-binding proteins with CHOL in plasma membrane could sequestrate lipids and thus affect the efficiency of virus entry into host cells, preventing the initial steps of viral infection. We have prepared CHOL-binding proteins with high affinities for lipids in the plasma membrane of mammalian cells. Binding of the perfringolysin O domain four (D4) and its variant D4E458L to membrane CHOL impaired the internalization of the receptor-binding domain of the SARS-CoV-2 spike protein and the pseudovirus complemented with the SARS-CoV-2 spike protein. SARS-CoV-2 replication in Vero E6 cells was also decreased. Overall, our results demonstrate that the integrity of CHOL-rich membrane domains and the accessibility of CHOL in the membrane play an essential role in SARS-CoV-2 cell entry.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Aleksandra Šakanović
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Apolonija Bedina-Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Simon Caserman
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Urška Kuhar
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Brigita Slavec
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Uroš Krapež
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Matjaž Ocepek
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan; UMR 7021 CNRS, Université de Strasbourg, F-67401, Illkirch, France
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Marjetka Podobnik
- 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
|
41
|
Li F, Zhu P, Zheng B, Lu Z, Fang C, Fu Y, Li X. A Customized Biohybrid Presenting Cascade Responses to Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404901. [PMID: 38723206 DOI: 10.1002/adma.202404901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Indexed: 05/16/2024]
Abstract
Intrinsic characteristics of microorganisms, including non-specific metabolism sites, toxic byproducts, and uncontrolled proliferation constrain their exploitation in medical applications such as tumor therapy. Here, the authors report an engineered biohybrid that can efficiently target cancerous sites through a pre-determined metabolic pathway to enable precise tumor ablation. In this system, DH5α Escherichia coli is engineered by the introduction of hypoxia-inducible promoters and lactate oxidase genes, and further surface-armored with iron-doped ZIF-8 nanoparticles. This bioengineered E. coli can produce and secrete lactate oxidase to reduce lactate concentration in response to hypoxic tumor microenvironment, as well as triggering immune activation. The peroxidase-like functionality of the nanoparticles extends the end product of the lactate metabolism, enabling the conversion of hydrogen peroxide (H2O2) into highly cytotoxic hydroxyl radicals. This, coupled with the transformation of tirapazamine loaded on nanoparticles to toxic benzotriazinyl, culminates in severe tumor cell ferroptosis. Intravenous injection of this biohybrid significantly inhibits tumor growth and metastasis.
Collapse
Affiliation(s)
- Feiyu Li
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Peipei Zhu
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Bingzhu Zheng
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Zijie Lu
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Chao Fang
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Yike Fu
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Xiang Li
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| |
Collapse
|
42
|
Ribeiro TP, Martins-de-Sa D, Macedo LLP, Lourenço-Tessutti IT, Ruffo GC, Sousa JPA, Rósario Santana JMD, Oliveira-Neto OB, Moura SM, Silva MCM, Morgante CV, Oliveira NG, Basso MF, Grossi-de-Sa MF. Cotton plants overexpressing the Bacillus thuringiensis Cry23Aa and Cry37Aa binary-like toxins exhibit high resistance to the cotton boll weevil (Anthonomus grandis). PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2024; 344:112079. [PMID: 38588981 DOI: 10.1016/j.plantsci.2024.112079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/10/2024]
Abstract
The cotton boll weevil (CBW, Anthonomus grandis) stands as one of the most significant threats to cotton crops (Gossypium hirsutum). Despite substantial efforts, the development of a commercially viable transgenic cotton event for effective open-field control of CBW has remained elusive. This study describes a detailed characterization of the insecticidal toxins Cry23Aa and Cry37Aa against CBW. Our findings reveal that CBW larvae fed on artificial diets supplemented exclusively with Cry23Aa decreased larval survival by roughly by 69%, while supplementation with Cry37Aa alone displayed no statistical difference compared to the control. However, the combined provision of both toxins in the artificial diet led to mortality rates approaching 100% among CBW larvae (LC50 equal to 0.26 PPM). Additionally, we engineered transgenic cotton plants by introducing cry23Aa and cry37Aa genes under control of the flower bud-specific pGhFS4 and pGhFS1 promoters, respectively. Seven transgenic cotton events expressing high levels of Cry23Aa and Cry37Aa toxins in flower buds were selected for greenhouse bioassays, and the mortality rate of CBW larvae feeding on their T0 and T1 generations ranged from 75% to 100%. Our in silico analyses unveiled that Cry23Aa displays all the hallmark characteristics of β-pore-forming toxins (β-PFTs) that bind to sugar moieties in glycoproteins. Intriguingly, we also discovered a distinctive zinc-binding site within Cry23Aa, which appears to be involved in protein-protein interactions. Finally, we discuss the major structural features of Cry23Aa that likely play a role in the toxin's mechanism of action. In view of the low LC50 for CBW larvae and the significant accumulation of these toxins in the flower buds of both T0 and T1 plants, we anticipate that through successive generations of these transgenic lines, cotton plants engineered to overexpress cry23Aa and cry37Aa hold promise for effectively managing CBW infestations in cotton crops.
Collapse
Affiliation(s)
- Thuanne Pires Ribeiro
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Diogo Martins-de-Sa
- Department of Cellular Biology, University of Brasília, Brasília, DF 70910-900, Brazil; Genesilico Biotech, Brasília, DF 71503-508, Brazil
| | - Leonardo Lima Pepino Macedo
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Isabela Tristan Lourenço-Tessutti
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Gustavo Caseca Ruffo
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil; Graduate Program in Genomic Science and Biotechnology, Catholic University of Brasília, Brasília, DF 71966-700, Brazil
| | - João Pedro Abreu Sousa
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil; Graduate Program in Genomic Science and Biotechnology, Catholic University of Brasília, Brasília, DF 71966-700, Brazil
| | - Julia Moura do Rósario Santana
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil; Graduate Program in Genomic Science and Biotechnology, Catholic University of Brasília, Brasília, DF 71966-700, Brazil
| | - Osmundo Brilhante Oliveira-Neto
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil; Euroamerican University Center, Unieuro, Brasília, DF 70790-160, Brazil
| | - Stéfanie Menezes Moura
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Maria Cristina Mattar Silva
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Carolina Vianna Morgante
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil; Embrapa Semi-Arid, Pretrolina, PE 56302-970, Brazil
| | - Nelson Geraldo Oliveira
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Marcos Fernando Basso
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil
| | - Maria Fatima Grossi-de-Sa
- Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil; National Institute of Science and Technology, INCT PlantStress Biotech, Embrapa, Brasília, DF 70770-917, Brazil; Graduate Program in Genomic Science and Biotechnology, Catholic University of Brasília, Brasília, DF 71966-700, Brazil; Graduate Program in Biotechnology, Catholic University Dom Bosco, Campo Grande, MS 79117-900, Brazil.
| |
Collapse
|
43
|
Goudarzi MH, Robinson SD, Cardoso FC, Mitchell ML, Cook LG, King GF, Walker AA. Phylogeny, envenomation syndrome, and membrane permeabilising venom produced by Australia's electric caterpillar Comana monomorpha. Sci Rep 2024; 14:14172. [PMID: 38898081 PMCID: PMC11187147 DOI: 10.1038/s41598-024-65078-1] [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: 04/17/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024] Open
Abstract
Zygaenoidea is a superfamily of lepidopterans containing many venomous species, including the Limacodidae (nettle caterpillars) and Megalopygidae (asp caterpillars). Venom proteomes have been recently documented for several species from each of these families, but further data are required to understand the evolution of venom in Zygaenoidea. In this study, we examined the 'electric' caterpillar from North-Eastern Australia, a limacodid caterpillar densely covered in venomous spines. We used DNA barcoding to identify this caterpillar as the larva of the moth Comana monomorpha (Turner, 1904). We report the clinical symptoms of C. monomorpha envenomation, which include acute pain, and erythema and oedema lasting for more than a week. Combining transcriptomics of venom spines with proteomics of venom harvested from the spine tips revealed a venom markedly different in composition from previously examined limacodid venoms that are rich in peptides. In contrast, the venom of C. monomorpha is rich in aerolysin-like proteins similar to those found in venoms of asp caterpillars (Megalopygidae). Consistent with this composition, the venom potently permeabilises sensory neurons and human neuroblastoma cells. This study highlights the diversity of venom composition in Limacodidae.
Collapse
Affiliation(s)
- Mohaddeseh H Goudarzi
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Protein and Peptide Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Samuel D Robinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Protein and Peptide Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michela L Mitchell
- Department of Toxinology, Women's and Children's Health Network, North Adelaide, SA, 5006, Australia
| | - Lyn G Cook
- School of the Environment, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
- Australian Research Council Centre of Excellence for Innovations in Protein and Peptide Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Andrew A Walker
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.
- Australian Research Council Centre of Excellence for Innovations in Protein and Peptide Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
44
|
Zhu X, Shi Z, Mao Y, Lächelt U, Huang R. Cell Membrane Perforation: Patterns, Mechanisms and Functions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310605. [PMID: 38344881 DOI: 10.1002/smll.202310605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/21/2023] [Indexed: 02/21/2024]
Abstract
Cell membrane is crucial for the cellular activities, and any disruption to it may affect the cells. It is demonstrated that cell membrane perforation is associated with some biological processes like programmed cell death (PCD) and infection of pathogens. Specific developments make it a promising technique to perforate the cell membrane controllably and precisely. The pores on the cell membrane provide direct pathways for the entry and exit of substances, and can also cause cell death, which means reasonable utilization of cell membrane perforation is able to assist intracellular delivery, eliminate diseased or cancerous cells, and bring about other benefits. This review classifies the patterns of cell membrane perforation based on the mechanisms into 1) physical patterns, 2) biological patterns, and 3) chemical patterns, introduces the characterization methods and then summarizes the functions according to the characteristics of reversible and irreversible pores, with the aim of providing a comprehensive summary of the knowledge related to cell membrane perforation and enlightening broad applications in biomedical science.
Collapse
Affiliation(s)
- Xinran Zhu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Ulrich Lächelt
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria
| | - Rongqin Huang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Huashan Hospital, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
45
|
Zhao J, Wang Z, Yang M, Guo J, Gao Z, Song P, Song YY. Pore-Forming Toxin-Driven Recovery of Peroxidase-Mimicking Activity in Biomass Channels for Label-Free Electrochemical Bacteria Sensing. Anal Chem 2024; 96:7661-7668. [PMID: 38687969 DOI: 10.1021/acs.analchem.4c00589] [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: 05/02/2024]
Abstract
The development of sensitive, selective, and rapid methods to detect bacteria in complex media is essential to ensuring human health. Virulence factors, particularly pore-forming toxins (PFTs) secreted by pathogenic bacteria, play a crucial role in bacterial diseases and serve as indicators of disease severity. In this study, a nanochannel-based label-free electrochemical sensing platform was developed for the detection of specific pathogenic bacteria based on their secreted PFTs. In this design, wood substrate channels were functionalized with a Fe-based metal-organic framework (FeMOF) and then protected with a layer of phosphatidylcholine (PC)-based phospholipid membrane (PM) that serves as a peroxidase mimetic and a channel gatekeeper, respectively. Using Staphylococcus aureus (S. aureus) as the model bacteria, the PC-specific PFTs secreted by S. aureus perforate the PM layer. Now exposed to the FeMOF, uncharged 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonate) (ABTS) molecules in the electrolyte undergo oxidation to cationic products (ABTS•+). The measured transmembrane ionic current indicates the presence of S. aureus and methicillin-resistant S. aureus (MRSA) with a low detection limit of 3 cfu mL-1. Besides excellent specificity, this sensing approach exhibits satisfactory performance for the detection of target bacteria in the complex media of food.
Collapse
Affiliation(s)
- Junjian Zhao
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Zirui Wang
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Mei Yang
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Junli Guo
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
- Foshan Graduate School of Innovation, Northeastern University, Foshan 528311, China
| | - Zhida Gao
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| | - Pei Song
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Yan-Yan Song
- Department of Chemistry, College of Science, Northeastern University, Shenyang 110819, China
| |
Collapse
|
46
|
Zheng Y, Yang Y, Liu X, Liu P, Li X, Zhang M, Zhou E, Zhao Z, Wang X, Zhang Y, Zheng B, Yan Y, Liu Y, Xu D, Cao L. Accelerated corrosion of 316L stainless steel in a simulated oral environment via extracellular electron transfer and acid metabolites of subgingival microbiota. Bioact Mater 2024; 35:56-66. [PMID: 38283387 PMCID: PMC10810744 DOI: 10.1016/j.bioactmat.2024.01.007] [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: 10/16/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/30/2024] Open
Abstract
316L stainless steel (SS) is widely applied as microimplant anchorage (MIA) due to its excellent mechanical properties. However, the risk that the oral microorganisms can corrode 316L SS is fully neglected. Microbiologically influenced corrosion (MIC) of 316L SS is essential to the health and safety of all patients because the accelerated corrosion caused by the oral microbiota can trigger the release of Cr and Ni ions. This study investigated the corrosion behavior and mechanism of subgingival microbiota on 316L SS by 16S rRNA and metagenome sequencing, electrochemical measurements, and surface characterization techniques. Multispecies biofilms were formed by the oral subgingival microbiota in the simulated oral anaerobic environment on 316L SS surfaces, significantly accelerating the corrosion in the form of pitting. The microbiota samples collected from the subjects differed in biofilm compositions, corrosion behaviors, and mechanisms. The oral subgingival microbiota contributed to the accelerated corrosion of 316L SS via acidic metabolites and extracellular electron transfer. Our findings provide a new insight into the underlying mechanisms of oral microbial corrosion and guide the design of oral microbial corrosion-resistant materials.
Collapse
Affiliation(s)
- Ying Zheng
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yi Yang
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- State Key Laboratory of Rolling and Automation, Northeastern University, Shenyang, China
| | - Xianbo Liu
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Pan Liu
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- State Key Laboratory of Rolling and Automation, Northeastern University, Shenyang, China
| | - Xiangyu Li
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- State Key Laboratory of Rolling and Automation, Northeastern University, Shenyang, China
| | - Mingxing Zhang
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- State Key Laboratory of Rolling and Automation, Northeastern University, Shenyang, China
| | - Enze Zhou
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- State Key Laboratory of Rolling and Automation, Northeastern University, Shenyang, China
| | - Zhenjin Zhao
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xue Wang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yuanyuan Zhang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Bowen Zheng
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yuwen Yan
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yi Liu
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Dake Xu
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
- State Key Laboratory of Rolling and Automation, Northeastern University, Shenyang, China
- Electrobiomaterials Institute, Key Laboratory for Anisotropy and Texture of Materials (Ministry of Education), Northeastern University, Shenyang, China
| | - Liu Cao
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
- Institute of Health Sciences, China Medical University, Shenyang, China
| |
Collapse
|
47
|
Schmidt G. Some Examples of Bacterial Toxins as Tools. Toxins (Basel) 2024; 16:202. [PMID: 38787054 PMCID: PMC11125981 DOI: 10.3390/toxins16050202] [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: 03/05/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/25/2024] Open
Abstract
Pathogenic bacteria produce diverse protein toxins to disturb the host's defenses. This includes the opening of epithelial barriers to establish bacterial growth in deeper tissues of the host and to modulate immune cell functions. To achieve this, many toxins share the ability to enter mammalian cells, where they catalyze the modification of cellular proteins. The enzymatic activity is diverse and ranges from ribosyl- or glycosyl-transferase activity, the deamidation of proteins, and adenylate-cyclase activity to proteolytic cleavage. Protein toxins are highly active enzymes often with tight specificity for an intracellular protein or a protein family coupled with the intrinsic capability of entering mammalian cells. A broad understanding of their molecular mechanisms established bacterial toxins as powerful tools for cell biology. Both the enzymatic part and the pore-forming/protein transport capacity are currently used as tools engineered to study signaling pathways or to transport cargo like labeled compounds, nucleic acids, peptides, or proteins directly into the cytosol. Using several representative examples, this review is intended to provide a short overview of the state of the art in the use of bacterial toxins or parts thereof as tools.
Collapse
Affiliation(s)
- Gudula Schmidt
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| |
Collapse
|
48
|
Gigon L, Müller P, Haenni B, Iacovache I, Barbo M, Gosheva G, Yousefi S, Soragni A, von Ballmoos C, Zuber B, Simon HU. Membrane damage by MBP-1 is mediated by pore formation and amplified by mtDNA. Cell Rep 2024; 43:114084. [PMID: 38583154 DOI: 10.1016/j.celrep.2024.114084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/28/2023] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Eosinophils play a crucial role in host defense while also contributing to immunopathology through the release of inflammatory mediators. Characterized by distinctive cytoplasmic granules, eosinophils securely store and rapidly release various proteins exhibiting high toxicity upon extracellular release. Among these, major basic protein 1 (MBP-1) emerges as an important mediator in eosinophil function against pathogens and in eosinophil-associated diseases. While MBP-1 targets both microorganisms and host cells, its precise mechanism remains elusive. We demonstrate that formation of small pores by MBP-1 in lipid bilayers induces membrane permeabilization and disrupts potassium balance. Additionally, we reveal that mitochondrial DNA (mtDNA) present in eosinophil extracellular traps (EETs) amplifies MBP-1 toxic effects, underscoring the pivotal role of mtDNA in EETs. Furthermore, we present evidence indicating that absence of CpG methylation in mtDNA contributes to the regulation of MBP-1-mediated toxicity. Taken together, our data suggest that the mtDNA scaffold within extracellular traps promotes MBP-1 toxicity.
Collapse
Affiliation(s)
- Lea Gigon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Philipp Müller
- Department of Chemistry, Biochemistry, and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Beat Haenni
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Ioan Iacovache
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Maruša Barbo
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Gordana Gosheva
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Alice Soragni
- Department of Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christoph von Ballmoos
- Department of Chemistry, Biochemistry, and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Benoît Zuber
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany.
| |
Collapse
|
49
|
Beltrán JF, Herrera-Belén L, Parraguez-Contreras F, Farías JG, Machuca-Sepúlveda J, Short S. MultiToxPred 1.0: a novel comprehensive tool for predicting 27 classes of protein toxins using an ensemble machine learning approach. BMC Bioinformatics 2024; 25:148. [PMID: 38609877 PMCID: PMC11010298 DOI: 10.1186/s12859-024-05748-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
Protein toxins are defense mechanisms and adaptations found in various organisms and microorganisms, and their use in scientific research as therapeutic candidates is gaining relevance due to their effectiveness and specificity against cellular targets. However, discovering these toxins is time-consuming and expensive. In silico tools, particularly those based on machine learning and deep learning, have emerged as valuable resources to address this challenge. Existing tools primarily focus on binary classification, determining whether a protein is a toxin or not, and occasionally identifying specific types of toxins. For the first time, we propose a novel approach capable of classifying protein toxins into 27 distinct categories based on their mode of action within cells. To accomplish this, we assessed multiple machine learning techniques and found that an ensemble model incorporating the Light Gradient Boosting Machine and Quadratic Discriminant Analysis algorithms exhibited the best performance. During the tenfold cross-validation on the training dataset, our model exhibited notable metrics: 0.840 accuracy, 0.827 F1 score, 0.836 precision, 0.840 sensitivity, and 0.989 AUC. In the testing stage, using an independent dataset, the model achieved 0.846 accuracy, 0.838 F1 score, 0.847 precision, 0.849 sensitivity, and 0.991 AUC. These results present a powerful next-generation tool called MultiToxPred 1.0, accessible through a web application. We believe that MultiToxPred 1.0 has the potential to become an indispensable resource for researchers, facilitating the efficient identification of protein toxins. By leveraging this tool, scientists can accelerate their search for these toxins and advance their understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Jorge F Beltrán
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile.
| | - Lisandra Herrera-Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Temuco, Chile
| | - Fernanda Parraguez-Contreras
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile
| | - Jorge G Farías
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile
| | - Jorge Machuca-Sepúlveda
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile
| | - Stefania Short
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar, 01145, Temuco, Chile
| |
Collapse
|
50
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|