Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 13(29): 33821-33829, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34254515

ABSTRACT

Safe and effective antimicrobials are needed to combat emerging antibiotic-resistant bacteria. Structurally nanoengineered antimicrobial peptide polymers (termed SNAPPs) interact with bacterial cell membranes to potently kill bacteria but may also interact at some level with human cell membranes. We studied the association of four different SNAPPs with six different white blood cells within fresh whole human blood by flow cytometry. In whole human blood, SNAPPs had detectable association with phagocytic cells and B cells, but not natural killer and T cells. However, without plasma proteins and therefore no protein corona on the SNAPPs, a greater marked association of SNAPPs with all white blood cell types was detected, resulting in cytotoxicity against most blood cell components. Thus, the formation of a protein corona around the SNAPPs reduced the association and prevented human blood cell cytotoxicity of the SNAPPs. Understanding the bio-nano interactions of these SNAPPs will be crucial to ensuring that the design of next-generation SNAPPs and other promising antimicrobial nanomaterials continues to display high efficacy toward antibiotic-resistant bacteria while maintaining a low toxicity to primary human cells.


Subject(s)
Anti-Infective Agents/toxicity , Dendrimers/toxicity , Leukocytes/drug effects , Polyamines/toxicity , Pore Forming Cytotoxic Proteins/toxicity , Protein Corona/metabolism , Anti-Infective Agents/metabolism , Blood Proteins/metabolism , Dendrimers/metabolism , Humans , Polyamines/metabolism , Pore Forming Cytotoxic Proteins/metabolism
2.
ACS Appl Mater Interfaces ; 13(24): 28764-28773, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34110763

ABSTRACT

Titanium is widely utilized for manufacturing medical implants due to its inherent mechanical strength and biocompatibility. Recent studies have focused on developing coatings to impart unique properties to Ti implants, such as antimicrobial behavior, enhanced cell adhesion, and osteointegration. Ca- and Si-based ceramic (CS) coatings can enhance bone integration through the release of Ca and Si ions. However, high degradation rates of CS ceramics create a basic environment that reduces cell viability. Polymeric or protein-based coatings may be employed to modulate CS degradation. However, it is challenging to ensure coating stability over extended periods of time without compromising biocompatibility. In this study, we employed a fluorous-cured collagen shell as a drug-loadable scaffold around CS nanorod coatings on Ti implants. Fluorous-cured collagen coatings have enhanced mechanical and enzymatic stability and are able to regulate the release of Ca and Si ions. Furthermore, the collagen scaffold was loaded with antimicrobial peptides to impart antimicrobial activity while promoting cell adhesion. These multifunctional collagen coatings simultaneously regulate the degradation of CS ceramics and enhance antimicrobial activity, while maintaining biocompatibility.


Subject(s)
Anti-Bacterial Agents/pharmacology , Nanotubes/chemistry , Pore Forming Cytotoxic Proteins/pharmacology , Silicates/chemistry , Titanium/chemistry , Wound Healing/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/toxicity , Collagen/chemistry , Collagen/toxicity , Humans , Microbial Sensitivity Tests , Nanotubes/toxicity , Osteoblasts/drug effects , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/toxicity , Silicates/toxicity , Staphylococcus aureus/drug effects , Titanium/toxicity , Wettability
3.
Biomolecules ; 11(4)2021 04 09.
Article in English | MEDLINE | ID: mdl-33918933

ABSTRACT

Fire blight is a major pome fruit trees disease that is caused by the quarantine phytopathogenic Erwinia amylovora, leading to major losses, namely, in pear and apple productions. Nevertheless, no effective sustainable control treatments and measures have yet been disclosed. In that regard, antimicrobial peptides (AMPs) have been proposed as an alternative biomolecule against pathogens but some of those AMPs have yet to be tested against E. amylovora. In this study, the potential of five AMPs (RW-BP100, CA-M, 3.1, D4E1, and Dhvar-5) together with BP100, were assessed to control E. amylovora. Antibiograms, minimal inhibitory, and bactericidal concentrations (minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC), growth and IC50 were determined and membrane permeabilization capacity was evaluated by flow cytometry analysis and colony-forming units (CFUs) plate counting. For the tested AMPs, the higher inhibitory and bactericidal capacity was observed for RW-BP100 and CA-M (5 and 5-8 µM, respectively for both MIC and MBC), whilst for IC50 RW-BP100 presented higher efficiency (2.8 to 3.5 µM). Growth curves for the first concentrations bellow MIC showed that these AMPs delayed E. amylovora growth. Flow cytometry disclosed faster membrane permeabilization for CA-M. These results highlight the potential of RW-BP100 and CA-M AMPs as sustainable control measures against E. amylovora.


Subject(s)
Erwinia amylovora/drug effects , Pore Forming Cytotoxic Proteins/toxicity , Cell Membrane/drug effects , Cell Membrane/metabolism , Inhibitory Concentration 50 , Pore Forming Cytotoxic Proteins/chemical synthesis , Pore Forming Cytotoxic Proteins/pharmacology
4.
Toxins (Basel) ; 13(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33572185

ABSTRACT

Pore-forming toxins (PFTs) form multimeric trans-membrane pores in cell membranes that differ in pore channel diameter (PCD). Cellular resistance to large PFTs (>20 nm PCD) was shown to rely on Ca2+ influx activated membrane repair mechanisms. Small PFTs (<2 nm PCD) were shown to exhibit a high cytotoxic activity, but host cell response and membrane repair mechanisms are less well studied. We used monocytic immune cell lines to investigate the cellular resistance and host membrane repair mechanisms to small PFTs lysenin (Eisenia fetida) and aerolysin (Aeromonas hydrophila). Lysenin, but not aerolysin, is shown to induce Ca2+ influx from the extracellular space and to activate Ca2+ dependent membrane repair mechanisms. Moreover, lysenin binds to U937 cells with higher efficiency as compared to THP-1 cells, which is in line with a high sensitivity of U937 cells to lysenin. In contrast, aerolysin equally binds to U937 or THP-1 cells, but in different plasma membrane areas. Increased aerolysin induced cell death of U937 cells, as compared to THP-1 cells, is suggested to be a consequence of cap-like aerolysin binding. We conclude that host cell resistance to small PFTs attack comprises binding efficiency, pore localization, and capability to induce Ca2+ dependent membrane repair mechanisms.


Subject(s)
Bacterial Toxins/toxicity , Calcium Signaling/drug effects , Calcium/metabolism , Cell Membrane Permeability/drug effects , Cell Membrane/drug effects , Monocytes/drug effects , Pore Forming Cytotoxic Proteins/toxicity , Toxins, Biological/toxicity , Cell Death/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Drug Resistance , Genes, Reporter , Humans , Monocytes/metabolism , Monocytes/pathology , THP-1 Cells , U937 Cells
5.
Nucleic Acids Res ; 49(5): 2642-2654, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33591308

ABSTRACT

The CRISPR/Cas9 system is a technology for genome engineering, which has been applied to indel mutations in genes as well as targeted gene deletion and replacement. Here, we describe paired gRNA deletions along the PIGA locus on the human X chromosome ranging from 17 kb to 2 Mb. We found no compelling linear correlation between deletion size and the deletion efficiency, and there is no substantial impact of topologically associating domains on deletion frequency. Using this precise deletion technique, we have engineered a series of designer deletion cell lines, including one with deletions of two X-chromosomal counterselectable (negative selection) markers, PIGA and HPRT1, and additional cell lines bearing each individual deletion. PIGA encodes a component of the glycosylphosphatidylinositol (GPI) anchor biosynthetic apparatus. The PIGA gene counterselectable marker has unique features, including existing single cell level assays for both function and loss of function of PIGA and the existence of a potent counterselectable agent, proaerolysin, which we use routinely for selection against cells expressing PIGA. These designer cell lines may serve as a general platform with multiple selection markers and may be particularly useful for large scale genome engineering projects such as Genome Project-Write (GP-write).


Subject(s)
CRISPR-Cas Systems , Cell Engineering , Membrane Proteins/genetics , Sequence Deletion , Bacterial Toxins/toxicity , Cell Line , Chromosomes, Human, X , Genetic Markers , Heterozygote , Humans , Mutation , N-Acetylglucosaminyltransferases/genetics , Pore Forming Cytotoxic Proteins/toxicity , RNA/genetics
6.
Sci Rep ; 10(1): 16581, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024236

ABSTRACT

Reducing hurdles to clinical trials without compromising the therapeutic promises of peptide candidates becomes an essential step in peptide-based drug design. Machine-learning models are cost-effective and time-saving strategies used to predict biological activities from primary sequences. Their limitations lie in the diversity of peptide sequences and biological information within these models. Additional outlier detection methods are needed to set the boundaries for reliable predictions; the applicability domain. Antimicrobial peptides (AMPs) constitute an extensive library of peptides offering promising avenues against antibiotic-resistant infections. Most AMPs present in clinical trials are administrated topically due to their hemolytic toxicity. Here we developed machine learning models and outlier detection methods that ensure robust predictions for the discovery of AMPs and the design of novel peptides with reduced hemolytic activity. Our best models, gradient boosting classifiers, predicted the hemolytic nature from any peptide sequence with 95-97% accuracy. Nearly 70% of AMPs were predicted as hemolytic peptides. Applying multivariate outlier detection models, we found that 273 AMPs (~ 9%) could not be predicted reliably. Our combined approach led to the discovery of 34 high-confidence non-hemolytic natural AMPs, the de novo design of 507 non-hemolytic peptides, and the guidelines for non-hemolytic peptide design.


Subject(s)
Drug Design , Machine Learning , Pore Forming Cytotoxic Proteins/chemistry , Amino Acid Sequence , Cost-Benefit Analysis , Hemolysis/drug effects , Machine Learning/economics , Pore Forming Cytotoxic Proteins/toxicity
7.
Proc Natl Acad Sci U S A ; 117(43): 26936-26945, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33046640

ABSTRACT

Novel antibiotics are urgently needed to combat multidrug-resistant pathogens. Venoms represent previously untapped sources of novel drugs. Here we repurposed mastoparan-L, the toxic active principle derived from the venom of the wasp Vespula lewisii, into synthetic antimicrobials. We engineered within its N terminus a motif conserved among natural peptides with potent immunomodulatory and antimicrobial activities. The resulting peptide, mast-MO, adopted an α-helical structure as determined by NMR, exhibited increased antibacterial properties comparable to standard-of-care antibiotics both in vitro and in vivo, and potentiated the activity of different classes of antibiotics. Mechanism-of-action studies revealed that mast-MO targets bacteria by rapidly permeabilizing their outer membrane. In animal models, the peptide displayed direct antimicrobial activity, led to enhanced ability to attract leukocytes to the infection site, and was able to control inflammation. Permutation studies depleted the remaining toxicity of mast-MO toward human cells, yielding derivatives with antiinfective activity in animals. We demonstrate a rational design strategy for repurposing venoms into promising antimicrobials.


Subject(s)
Bacteremia/drug therapy , Pore Forming Cytotoxic Proteins/chemistry , Wasp Venoms/chemistry , Animals , Drug Design , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Mice , Microbial Sensitivity Tests , Pore Forming Cytotoxic Proteins/therapeutic use , Pore Forming Cytotoxic Proteins/toxicity , Wasp Venoms/therapeutic use , Wasp Venoms/toxicity
8.
Sci Rep ; 10(1): 13206, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32764602

ABSTRACT

The use of non-standard toxicity models is a hurdle in the early development of antimicrobial peptides towards clinical applications. Herein we report an extensive in vitro and in vivo toxicity study of a library of 24 peptide-based antimicrobials with narrow spectrum activity towards veterinary pathogens. The haemolytic activity of the compounds was evaluated against four different species and the relative sensitivity against the compounds was highest for canine erythrocytes, intermediate for rat and human cells and lowest for bovine cells. Selected peptides were additionally evaluated against HeLa, HaCaT and HepG2 cells which showed increased stability towards the peptides. Therapeutic indexes of 50-500 suggest significant cellular selectivity in comparison to bacterial cells. Three peptides were administered to rats in intravenous acute dose toxicity studies up to 2-8 × MIC. None of the injected compounds induced any systemic toxic effects in vivo at the concentrations employed illustrating that the correlation between the different assays is not obvious. This work sheds light on the in vitro and in vivo toxicity of this class of promising compounds and provides insights into the relationship between the different toxicity models often employed in different manners to evaluate the toxicity of novel bioactive compounds in general.


Subject(s)
Hemolysis/drug effects , Pore Forming Cytotoxic Proteins/toxicity , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Erythrocytes/cytology , Erythrocytes/drug effects , Humans , Pore Forming Cytotoxic Proteins/chemistry , Rats
9.
Biomolecules ; 10(5)2020 05 07.
Article in English | MEDLINE | ID: mdl-32392805

ABSTRACT

Trichoderma species are widely used as biofungicides for the control of fungal plant pathogens. Several studies have been performed to identify the main genes and compounds involved in Trichoderma-plant-microbial pathogen cross-talks. However, there is not much information about the exact mechanism of this profitable interaction. Peptaibols secreted mainly by Trichoderma species are linear, 5-20 amino acid residue long, non-ribosomally synthesized peptides rich in α-amino isobutyric acid, which seem to be effective in Trichoderma-plant pathogenic fungus interactions. In the present study, reversed phase (RP) high-performance liquid chromatography (HPLC) coupled with electrospray ionization (ESI) mass spectrometry (MS) was used to detect peptaibol profiles of Trichoderma strains during interactions with fungal plant pathogens. MS investigations of the crude extracts deriving from in vitro confrontations of Trichodermaasperellum and T.longibrachiatum with different plant pathogenic fungi (Fusariummoniliforme, F.culmorum, F.graminearum, F.oxysporum species complex, Alternariasolani and Rhizoctoniasolani) were performed to get a better insight into the role of these non-ribosomal antimicrobial peptides. The results revealed an increase in the total amount of peptaibols produced during the interactions, as well as some differences in the peptaibol profiles between the confrontational and control tests. Detection of the expression level of the peptaibol synthetase tex1 by qRT-PCR showed a significant increase in T.asperellum/R.solani interaction in comparison to the control. In conclusion, the interaction with plant pathogens highly influenced the peptaibol production of the examined Trichoderma strains.


Subject(s)
Antibiosis , Peptaibols/metabolism , Trichoderma/metabolism , Alternaria/drug effects , Alternaria/physiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fusarium/drug effects , Fusarium/physiology , Peptaibols/chemistry , Peptaibols/toxicity , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/toxicity , Rhizoctonia/drug effects , Rhizoctonia/physiology , Trichoderma/physiology
10.
Toxins (Basel) ; 12(5)2020 05 22.
Article in English | MEDLINE | ID: mdl-32456013

ABSTRACT

Pore-forming toxins are alluring tools for delivering biologically-active, impermeable cargoes to intracellular environments by introducing large conductance pathways into cell membranes. However, the lack of regulation often leads to the dissipation of electrical and chemical gradients, which might significantly affect the viability of cells under scrutiny. To mitigate these problems, we explored the use of lysenin channels to reversibly control the barrier function of natural and artificial lipid membrane systems by controlling the lysenin's transport properties. We employed artificial membranes and electrophysiology measurements in order to identify the influence of labels and media on the lysenin channel's conductance. Two cell culture models: Jurkat cells in suspension and adherent ATDC5 cells were utilized to demonstrate that lysenin channels may provide temporary cytosol access to membrane non-permeant propidium iodide and phalloidin. Permeability and cell viability were assessed by fluorescence spectroscopy and microscopy. Membrane resealing by chitosan or specific media addition proved to be an effective way of maintaining cellular viability. In addition, we loaded non-permeant dyes into liposomes via lysenin channels by controlling their conducting state with multivalent metal cations. The improved control over membrane permeability might prove fruitful for a large variety of biological or biomedical applications that require only temporary, non-destructive access to the inner environment enclosed by natural and artificial membranes.


Subject(s)
Cell Membrane Permeability/drug effects , Lipid Bilayers , Membranes/drug effects , Pore Forming Cytotoxic Proteins/pharmacology , Toxins, Biological/pharmacology , Cell Survival/drug effects , Chitosan/pharmacology , Humans , Jurkat Cells , Membrane Potentials , Membranes/metabolism , Membranes/pathology , Phalloidine/metabolism , Pore Forming Cytotoxic Proteins/toxicity , Propidium/metabolism , Toxins, Biological/toxicity
11.
J Invest Dermatol ; 140(8): 1619-1628.e2, 2020 08.
Article in English | MEDLINE | ID: mdl-31981578

ABSTRACT

The microbiome represents a vast resource for drug discovery, as its members engage in constant conflict to outcompete one another by deploying diverse strategies for survival. Cutibacterium acnes is one of the most common bacterial species on human skin and can promote the common disease acne vulgaris. By employing a combined strategy of functional screening, genetics, and proteomics we discovered a strain of Staphylococcus capitis (S. capitis E12) that selectively inhibited growth of C. acnes with potency greater than antibiotics commonly used in the treatment of acne. Antimicrobial peptides secreted from S. capitis E12 were identified as four distinct phenol-soluble modulins acting synergistically. These peptides were not toxic to human keratinocytes and the S. capitis extract did not kill other commensal skin bacteria but was effective against C. acnes on pig skin and on mice. Overall, these data show how a member of the human skin microbiome can be useful as a biotherapy for acne vulgaris.


Subject(s)
Acne Vulgaris/therapy , Biological Therapy/methods , Skin/microbiology , Staphylococcus capitis/immunology , Symbiosis/immunology , Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Adult , Animals , Female , Humans , Keratinocytes/immunology , Male , Mice , Microbial Sensitivity Tests , Pore Forming Cytotoxic Proteins/isolation & purification , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/toxicity , Primary Cell Culture , Propionibacterium acnes/immunology , Propionibacterium acnes/pathogenicity , Skin/immunology , Staphylococcus capitis/isolation & purification , Staphylococcus capitis/metabolism , Swine , Toxicity Tests , Young Adult
12.
Toxins (Basel) ; 11(7)2019 07 10.
Article in English | MEDLINE | ID: mdl-31295915

ABSTRACT

Random mutations and selective pressure drive protein adaptation to the changing demands of the environment. As a consequence, nature favors the evolution of protein diversity. A group of proteins subject to exceptional environmental stress and known for their widespread diversity are the pore-forming hemolytic proteins from sea anemones, known as actinoporins. In this study, we identified and isolated new isoforms of actinoporins from the sea anemone Actinia fragacea (fragaceatoxins). We characterized their hemolytic activity, examined their stability and structure, and performed a comparative analysis of their primary sequence. Sequence alignment reveals that most of the variability among actinoporins is associated with non-functional residues. The differences in the thermal behavior among fragaceatoxins suggest that these variability sites contribute to changes in protein stability. In addition, the protein-protein interaction region showed a very high degree of identity (92%) within fragaceatoxins, but only 25% among all actinoporins examined, suggesting some degree of specificity at the species level. Our findings support the mechanism of evolutionary adaptation in actinoporins and reflect common pathways conducive to protein variability.


Subject(s)
Cnidarian Venoms/isolation & purification , Pore Forming Cytotoxic Proteins/isolation & purification , Sea Anemones , Animals , Cnidarian Venoms/chemistry , Cnidarian Venoms/toxicity , Erythrocytes/drug effects , Hemolysis , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/toxicity , Protein Conformation , Sheep
13.
Toxins (Basel) ; 11(6)2019 06 25.
Article in English | MEDLINE | ID: mdl-31242582

ABSTRACT

Animal venoms are complex mixtures of highly specialized toxic molecules. Cnidarians and arachnids produce pore-forming proteins (PFPs) directed against the plasma membrane of their target cells. Among PFPs from cnidarians, actinoporins stand out for their small size and molecular simplicity. While native actinoporins require only sphingomyelin for membrane binding, engineered chimeras containing a recognition antibody-derived domain fused to an actinoporin isoform can nonetheless serve as highly specific immunotoxins. Examples of such constructs targeted against malignant cells have been already reported. However, PFPs from arachnid venoms are less well-studied from a structural and functional point of view. Spiders from the Latrodectus genus are professional insect hunters that, as part of their toxic arsenal, produce large PFPs known as latrotoxins. Interestingly, some latrotoxins have been identified as potent and highly-specific insecticides. Given the proteinaceous nature of these toxins, their promising future use as efficient bioinsecticides is discussed throughout this Perspective. Protein engineering and large-scale recombinant production are critical steps for the use of these PFPs as tools to control agriculturally important insect pests. In summary, both families of PFPs, from Cnidaria and Arachnida, appear to be molecules with promising biotechnological applications.


Subject(s)
Cnidarian Venoms , Pore Forming Cytotoxic Proteins , Spider Venoms , Animals , Arachnida , Biotechnology , Cnidaria , Cnidarian Venoms/chemistry , Cnidarian Venoms/toxicity , Genomics , Humans , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/toxicity , Spider Venoms/chemistry , Spider Venoms/toxicity
14.
Toxins (Basel) ; 11(6)2019 06 18.
Article in English | MEDLINE | ID: mdl-31216745

ABSTRACT

Pore-forming toxins (PFTs) form nanoscale pores across target membranes causing cell death. The pore-forming cytolysins of the RTX (repeats in toxin) family belong to a steadily increasing family of proteins characterized by having in their primary sequences a number of glycine- and aspartate-rich nonapeptide repeats. They are secreted by a variety of Gram-negative bacteria and form ion-permeable pores in several cell types, such as immune cells, epithelial cells, or erythrocytes. Pore-formation by RTX-toxins leads to the dissipation of ionic gradients and membrane potential across the cytoplasmic membrane of target cells, which results in cell death. The pores formed in lipid bilayers by the RTX-toxins share some common properties such as cation selectivity and voltage-dependence. Hemolytic and cytolytic RTX-toxins are important virulence factors in the pathogenesis of the producing bacteria. And hence, understanding the function of these proteins at the molecular level is critical to elucidating their role in disease processes. In this review we summarize the current state of knowledge on pore-formation by RTX toxins, and include recent results from our own laboratory regarding the pore-forming activity of adenylate cyclase toxin (ACT or CyaA), a large protein toxin secreted by Bordetella pertussis, the bacterium causative of whooping cough.


Subject(s)
Bacterial Toxins/toxicity , Cell Membrane Permeability/drug effects , Pore Forming Cytotoxic Proteins/toxicity , Animals , Bacterial Toxins/chemistry , Humans , Pore Forming Cytotoxic Proteins/chemistry
15.
Biomolecules ; 9(5)2019 05 10.
Article in English | MEDLINE | ID: mdl-31083482

ABSTRACT

RTX (Repeats in ToXin) pore-forming toxins constitute an expanding family of exoproteins secreted by many Gram-negative bacteria and involved in infectious diseases caused by said pathogens. Despite the relevance in the host/pathogen interactions, the structure and characteristics of the lesions formed by these toxins remain enigmatic. Here, we capture the first direct nanoscale pictures of lytic pores formed by an RTX toxin, the Adenylate cyclase (ACT), secreted by the whooping cough bacterium Bordetella pertussis. We reveal that ACT associates into growing-size oligomers of variable stoichiometry and heterogeneous architecture (lines, arcs, and rings) that pierce the membrane, and that, depending on the incubation time and the toxin concentration, evolve into large enough "holes" so as to allow the flux of large molecular mass solutes, while vesicle integrity is preserved. We also resolve ACT assemblies of similar variable stoichiometry in the cell membrane of permeabilized target macrophages, proving that our model system recapitulates the process of ACT permeabilization in natural membranes. Based on our data we propose a non-concerted monomer insertion and sequential mechanism of toroidal pore formation by ACT. A size-tunable pore adds a new regulatory element to ACT-mediated cytotoxicity, with different pore sizes being putatively involved in different physiological scenarios or cell types.


Subject(s)
Adenylate Cyclase Toxin/toxicity , Bordetella pertussis/pathogenicity , Cell Membrane/metabolism , Pore Forming Cytotoxic Proteins/toxicity , Adenylate Cyclase Toxin/chemistry , Adenylate Cyclase Toxin/metabolism , Animals , Bordetella pertussis/enzymology , Cell Line , Cell Membrane Permeability , Macrophages/microbiology , Mice , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/metabolism , Protein Binding , Protein Multimerization
16.
Toxins (Basel) ; 10(10)2018 09 25.
Article in English | MEDLINE | ID: mdl-30257425

ABSTRACT

Bacterial pore-forming toxins induce a rapid and massive increase in cytosolic Ca2+ concentration due to the formation of pores in the plasma membrane and/or activation of Ca2+-channels. As Ca2+ is an essential messenger in cellular signaling, a sustained increase in Ca2+ concentration has dramatic consequences on cellular behavior, eventually leading to cell death. However, host cells have adapted mechanisms to protect against Ca2+ intoxication, such as Ca2+ efflux and membrane repair. The final outcome depends upon the nature and concentration of the toxin and on the cell type. This review highlights the repercussions of Ca2+ overload on the induction of cell death, repair mechanisms, cellular adhesive properties, and the inflammatory response.


Subject(s)
Bacterial Toxins/toxicity , Calcium/metabolism , Pore Forming Cytotoxic Proteins/toxicity , Animals , Cell Death/drug effects , Humans , Intercellular Junctions/drug effects
17.
Toxins (Basel) ; 10(9)2018 08 23.
Article in English | MEDLINE | ID: mdl-30142951

ABSTRACT

Recent technological advances have seen increasing numbers of complex structures from diverse pore-forming toxins (PFT). The ClyA family of α-PFTs comprises a broad variety of assemblies including single-, two- and three-component toxin systems. With crystal structures available for soluble subunits of all major groups in this extended protein family, efforts now focus on obtaining molecular insights into physiological pore formation. This review provides an up-to-date discussion on common and divergent structural and functional traits that distinguish the various ClyA family PFTs. Open questions of this research topic are outlined and discussed.


Subject(s)
Pore Forming Cytotoxic Proteins , Cell Membrane/physiology , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/toxicity , Protein Conformation
18.
Environ Microbiol ; 20(9): 3442-3456, 2018 09.
Article in English | MEDLINE | ID: mdl-30136361

ABSTRACT

Aeromonas species are ubiquitous inhabitants of freshwater environments, and are responsible for fish motile aeromonad septicemia (MAS). A. hydrophila is implicated as the primary etiologic agent of MAS. Here, we analysed MAS epidemiological data for cyprinid fish in southern China, and found that A. veronii infections dominated. Consistent with this observation, A. veronii isolates were generally more virulent than A. hydrophila isolates when infecting germ-free zebrafish larvae via continuous immersion challenge. Through in vivo screening of the transposon library of the A. veronii strain Hm091, aerolysin was identified as the key virulence factor. Further results indicated that A. veronii Hm091 aerolysin disrupts the intestinal barrier of zebrafish, enabling systematic invasion by not only A. veronii Hm091 in a mono-infection, but also A. hydrophila NJ-1 in a mixed infection. Moreover, the differences in aerolysin expression and activity were the major contributor to the observed differences between the A. veronii and A. hydrophila strains regarding invasion efficacy via intestine. Together, our results provide new insights into the aetiology and pathogenesis of Aeromonas infections, and highlight the importance of A. veronii-targeted treatments in future efforts against MAS.


Subject(s)
Aeromonas veronii/metabolism , Aeromonas veronii/pathogenicity , Bacterial Toxins/metabolism , Fish Diseases/microbiology , Gram-Negative Bacterial Infections/veterinary , Pore Forming Cytotoxic Proteins/metabolism , Sepsis/veterinary , Aeromonas/isolation & purification , Aeromonas veronii/genetics , Animals , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , China , Gram-Negative Bacterial Infections/microbiology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/toxicity , Sepsis/microbiology , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Virulence Factors/toxicity , Zebrafish/microbiology
19.
J Appl Toxicol ; 38(3): 408-417, 2018 03.
Article in English | MEDLINE | ID: mdl-29134676

ABSTRACT

Plague has led to millions of deaths in history and outbreaks continue to the present day. The efficacy limitations and safety concerns of the existing killed whole cell and live-attenuated vaccines call for the development of new vaccines. In this study, we evaluated the immunogenicity and safety of a novel subunit plague vaccine, comprising native F1 antigen and recombinant V antigen. The cynomolgus macaques in low- and high-dose vaccine groups were vaccinated at weeks 0, 2, 4 and 6, at dose levels of 15 µg F1 + 15 µg rV and 30 µg F1 + 30 µg rV respectively. Specific antibodies and interferon-γ and interleukin-2 expression in lymphocytes were measured. For safety, except for the general toxicity and local irritation, we made a systematic immunotoxicity study on the vaccine including immunostimulation, autoimmunity and anaphylactic reaction. The vaccine induced high levels of serum anti-F1 and anti-rV antibodies, and caused small increases of interferon-γ and interleukin-2 in monkeys. The vaccination led to a reversible increase in the number of peripheral blood eosinophils, the increases in serum IgE level in a few animals and histopathological change of granulomas at injection sites. The vaccine had no impact on general conditions, most clinical pathology parameters, percentages of T-cell subsets, organ weights and gross pathology of treated monkeys and had passable local tolerance. The F1 + rV subunit plague vaccine can induce very strong humoral immunity and low level of cellular immunity in cynomolgus macaques and has a good safety profile.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Immunity, Humoral/drug effects , Immunogenicity, Vaccine , Plague Vaccine/immunology , Pore Forming Cytotoxic Proteins/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/toxicity , Bacterial Proteins/administration & dosage , Bacterial Proteins/toxicity , Eosinophils/drug effects , Eosinophils/immunology , Female , Granuloma/chemically induced , Granuloma/immunology , Granuloma/pathology , Immunity, Cellular/drug effects , Immunoglobulin E/blood , Injection Site Reaction/immunology , Injection Site Reaction/pathology , Injections, Intramuscular , Interferon-gamma/blood , Interleukin-2/blood , Macaca fascicularis , Male , Plague Vaccine/administration & dosage , Plague Vaccine/toxicity , Pore Forming Cytotoxic Proteins/administration & dosage , Pore Forming Cytotoxic Proteins/toxicity , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
20.
Toxins (Basel) ; 10(1)2017 12 27.
Article in English | MEDLINE | ID: mdl-29280959

ABSTRACT

Venoms are produced by a wide variety of species including spiders, scorpions, reptiles, cnidarians, and fish for the purpose of harming or incapacitating predators or prey. While some venoms are of relatively simple composition, many contain hundreds to thousands of individual components with distinct pharmacological activity. Pain-inducing or "algesic" venom compounds have proven invaluable to our understanding of how physiological nociceptive neural networks operate. In this review, we present an overview of some of the diverse nociceptive pathways that can be modulated by specific venom components to evoke pain.


Subject(s)
Pain/chemically induced , Peptides/toxicity , Venoms/toxicity , Animals , Humans , Ion Channels/physiology , Pain/physiopathology , Phospholipases A2/toxicity , Pore Forming Cytotoxic Proteins/toxicity , Sensory Receptor Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...