Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
Add more filters

Publication year range
1.
Biochem Biophys Res Commun ; 716: 149954, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38704887

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.


Subject(s)
Cell Membrane , Cholesterol , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , Vero Cells , Chlorocebus aethiops , Cholesterol/metabolism , Animals , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Cell Membrane/metabolism , Cell Membrane/virology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Humans , Carrier Proteins/metabolism , COVID-19/virology , COVID-19/metabolism , Protein Binding
2.
Fish Shellfish Immunol ; 151: 109698, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38871141

ABSTRACT

In the course of searching for genes controlling the immune system in caenogastropod mollusks, we characterized and phylogenetically placed five new actinoporin-like cytolysins expressed in periwinkles of the genus Littorina. These newly discovered proteins, named littoporins (LitP), contain a central cytolysin/lectin domain and exhibit a predicted protein fold that is almost identical to the three-dimensional structures of actinoporins. Two of these proteins, LitP-1 and LitP-2, were found to be upregulated in L. littorea kidney tissues and immune cells in response to natural and experimental infection with the trematode Himasthla elongata, suggesting their potential role as perforins in the systemic anti-trematode immune response. The primary sequence divergence of littoporins is hypothesized to be attributed to the taxonomic range of cell membranes they can recognize and permeabilize.


Subject(s)
Amino Acid Sequence , Phylogeny , Animals , Sequence Alignment/veterinary , Trematoda/physiology , Perforin/genetics , Perforin/immunology , Perforin/chemistry , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Snails/immunology , Snails/genetics , Gene Expression Profiling/veterinary
3.
Nucleic Acids Res ; 50(11): 6562-6574, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35670660

ABSTRACT

DNA transcription is regulated by a range of diverse mechanisms and primarily by transcription factors that recruit the RNA polymerase complex to the promoter region on the DNA. Protein binding to DNA at nearby or distant sites can synergistically affect this process in a variety of ways, but mainly through direct interactions between DNA-binding proteins. Here we show that a Transcription Activator-Like Effector (TALE), which lacks an activation domain, can enhance transcription in mammalian cells when it binds in the vicinity of and without direct interaction with several different dimeric or monomeric transcription factors. This effect was observed for several TALEs regardless of the recognition sequences and their DNA-bound orientation. TALEs can exert an effect over the distance of tens of nucleotides and it also potentiated KRAB-mediated repression. The augmentation of transcriptional regulation of another transcription factor is characteristic of TALEs, as it was not observed for dCas9/gRNA, zinc finger, or Gal4 DNA-binding domains. We propose that this mechanism involves an allosteric effect exerted on DNA structure or dynamics. This mechanism could be used to modulate transcription but may also play a role in the natural context of TALEs.


Subject(s)
Transcription Activator-Like Effectors , Transcription Factors , Transcription, Genetic , Animals , Binding Sites , DNA/genetics , Gene Expression Regulation , Mammals/genetics , Transcription Activator-Like Effectors/genetics , Transcription Factors/metabolism
4.
J Biol Chem ; 298(10): 102455, 2022 10.
Article in English | MEDLINE | ID: mdl-36063994

ABSTRACT

Pore-forming proteins perforate lipid membranes and consequently affect their integrity and cell fitness. Therefore, it is not surprising that many of these proteins from bacteria, fungi, or certain animals act as toxins. While pore-forming proteins have also been found in plants, there is little information about their molecular structure and mode of action. Bryoporin is a protein from the moss Physcomitrium patens, and its corresponding gene was found to be upregulated by various abiotic stresses, especially dehydration, as well as upon fungal infection. Based on the amino acid sequence, it was suggested that bryoporin was related to the actinoporin family of pore-forming proteins, originally discovered in sea anemones. Here, we provide the first detailed structural and functional analysis of this plant cytolysin. The crystal structure of monomeric bryoporin is highly similar to those of actinoporins. Our cryo-EM analysis of its pores showed an actinoporin-like octameric structure, thereby revealing a close kinship of proteins from evolutionarily distant organisms. This was further confirmed by our observation of bryoporin's preferential binding to and formation of pores in membranes containing animal sphingolipids, such as sphingomyelin and ceramide phosphoethanolamine; however, its binding affinity was weaker than that of actinoporin equinatoxin II. We determined bryoporin did not bind to major sphingolipids found in fungi or plants, and its membrane-binding and pore-forming activity was enhanced by various sterols. Our results suggest that bryoporin could represent a part of the moss defense arsenal, acting as a pore-forming toxin against membranes of potential animal pathogens, parasites, or predators.


Subject(s)
Bryopsida , Porins , Animals , Amino Acid Sequence , Bryopsida/genetics , Bryopsida/metabolism , Cnidarian Venoms/chemistry , Cytotoxins , Porins/genetics , Porins/metabolism , Sea Anemones/chemistry
5.
Biochem Biophys Res Commun ; 674: 44-52, 2023 Sep 24.
Article in English | MEDLINE | ID: mdl-37393643

ABSTRACT

The bacterium Bacillus thuringiensis (Bt) produces insecticidal proteins during the sporulation phase. These proteins are located in parasporal crystals consisting of two delta-endotoxin classes, crystal (Cry) and cytolytic (Cyt) toxins. In vitro, Cyt toxins show cytolytic activity against bacterial and a variety of insect and mammalian cells. They bind to cell membranes with unsaturated phospholipids and sphingomyelin. Although Bt and its parasporal crystals containing both Cry and Cyt toxins have been successfully used as bioinsecticides, the molecular mechanism of action of Cyt toxins is not yet fully understood. To address this, we exposed Cyt2Aa to lipid membranes and visualized membrane disruption process using cryo-electron microscopy. We observed two types of Cyt2Aa oligomers. First, Cyt2Aa forms smaller curved oligomers on the membrane surface that become linear over time, and detach when the membrane ruptures. Similar linear filamentous oligomers were also formed by Cyt2Aa in the presence of detergents without prior exposure to lipid membranes, which exhibited attenuated cytolytic activity. Furthermore, our data suggest that Cyt2Aa adopts different conformations between its monomeric and oligomeric forms. Overall, our results provide new evidence for a detergent-like mechanism of action of Cyt2Aa rather than the pore-forming model of target membrane disruption of this important class of insecticidal proteins.

6.
PLoS Pathog ; 17(4): e1009477, 2021 04.
Article in English | MEDLINE | ID: mdl-33857257

ABSTRACT

The lack of efficient methods to control the major diseases of crops most important to agriculture leads to huge economic losses and seriously threatens global food security. Many of the most important microbial plant pathogens, including bacteria, fungi, and oomycetes, secrete necrosis- and ethylene-inducing peptide 1 (Nep1)-like proteins (NLPs), which critically contribute to the virulence and spread of the disease. NLPs are cytotoxic to eudicot plants, as they disturb the plant plasma membrane by binding to specific plant membrane sphingolipid receptors. Their pivotal role in plant infection and broad taxonomic distribution makes NLPs a promising target for the development of novel phytopharmaceutical compounds. To identify compounds that bind to NLPs from the oomycetes Pythium aphanidermatum and Phytophthora parasitica, a library of 587 small molecules, most of which are commercially unavailable, was screened by surface plasmon resonance. Importantly, compounds that exhibited the highest affinity to NLPs were also found to inhibit NLP-mediated necrosis in tobacco leaves and Phytophthora infestans growth on potato leaves. Saturation transfer difference-nuclear magnetic resonance and molecular modelling of the most promising compound, anthranilic acid derivative, confirmed stable binding to the NLP protein, which resulted in decreased necrotic activity and reduced ion leakage from tobacco leaves. We, therefore, confirmed that NLPs are an appealing target for the development of novel phytopharmaceutical agents and strategies, which aim to directly interfere with the function of these major microbial virulence factors. The compounds identified in this study represent lead structures for further optimization and antimicrobial product development.


Subject(s)
Phytophthora/pathogenicity , Plant Diseases/prevention & control , Pythium/pathogenicity , Solanum tuberosum/genetics , Molecular Dynamics Simulation , Necrosis , Phytophthora/genetics , Plant Diseases/parasitology , Plant Leaves/genetics , Plant Leaves/parasitology , Pythium/genetics , Solanum tuberosum/parasitology , Surface Plasmon Resonance , Nicotiana/genetics , Nicotiana/parasitology
7.
New Phytol ; 237(3): 746-750, 2023 02.
Article in English | MEDLINE | ID: mdl-36210522

ABSTRACT

Lipid membrane destruction by microbial pore-forming toxins (PFTs) is a ubiquitous mechanism of damage to animal cells, but is less prominent in plants. Nep1-like proteins (NLPs) secreted by phytopathogens that cause devastating crop diseases, such as potato late blight, represent the only family of microbial PFTs that effectively damage plant cells by disrupting the integrity of the plant plasma membrane. Recent research has elucidated the molecular mechanism of NLP-mediated membrane damage, which is unique among microbial PFTs and highly adapted to the plant membrane environment. In this review, we cover recent insight into how NLP cytolysins damage plant membranes and cause cell death.


Subject(s)
Plants , Proteins , Animals , Cell Membrane , Cell Death
8.
Cell Mol Life Sci ; 78(17-18): 6229-6249, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34387717

ABSTRACT

Pore-forming proteins (PFPs) are a heterogeneous group of proteins that are expressed and secreted by a wide range of organisms. PFPs are produced as soluble monomers that bind to a receptor molecule in the host cell membrane. They then assemble into oligomers that are incorporated into the lipid membrane to form transmembrane pores. Such pore formation alters the permeability of the plasma membrane and is one of the most common mechanisms used by PFPs to destroy target cells. Interestingly, PFPs can also indirectly manipulate diverse cellular functions. In recent years, increasing evidence indicates that the interaction of PFPs with lipid membranes is not only limited to pore-induced membrane permeabilization but is also strongly associated with extensive plasma membrane reorganization. This includes lateral rearrangement and deformation of the lipid membrane, which can lead to the disruption of target cell function and finally death. Conversely, these modifications also constitute an essential component of the membrane repair system that protects cells from the lethal consequences of pore formation. Here, we provide an overview of the current knowledge on the changes in lipid membrane organization caused by PFPs from different organisms.


Subject(s)
Cell Membrane/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane Permeability , Complement Membrane Attack Complex/metabolism , Endocytosis , Humans , Membrane Lipids/metabolism , Porins/metabolism
9.
PLoS Pathog ; 15(9): e1007951, 2019 09.
Article in English | MEDLINE | ID: mdl-31479498

ABSTRACT

Necrosis and ethylene-inducing peptide 1 (Nep1)-like proteins (NLPs) are secreted by several phytopathogenic microorganisms. They trigger necrosis in various eudicot plants upon binding to plant sphingolipid glycosylinositol phosphorylceramides (GIPC). Interestingly, HaNLP3 from the obligate biotroph oomycete Hyaloperonospora arabidopsidis does not induce necrosis. We determined the crystal structure of HaNLP3 and showed that it adopts the NLP fold. However, the conformations of the loops surrounding the GIPC headgroup-binding cavity differ from those of cytotoxic Pythium aphanidermatum NLPPya. Essential dynamics extracted from µs-long molecular dynamics (MD) simulations reveals a limited conformational plasticity of the GIPC-binding cavity in HaNLP3 relative to toxic NLPs. This likely precludes HaNLP3 binding to GIPCs, which is the underlying reason for the lack of toxicity. This study reveals that mutations at key protein regions cause a switch between non-toxic and toxic phenotypes within the same protein scaffold. Altogether, these data provide evidence that protein flexibility is a distinguishing trait of toxic NLPs and highlight structural determinants for a potential functional diversification of non-toxic NLPs utilized by biotrophic plant pathogens.


Subject(s)
Oomycetes/genetics , Oomycetes/metabolism , Plant Diseases/parasitology , Amino Acid Sequence , Ethylenes/metabolism , Necrosis/metabolism , Peptides/metabolism , Peronospora/genetics , Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
FASEB J ; 34(8): 11068-11086, 2020 08.
Article in English | MEDLINE | ID: mdl-32648626

ABSTRACT

NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is a multiprotein complex which forms within cells in response to various microbial and self-derived triggers. Mutations in the gene encoding NLRP3 cause rare cryopyrin-associated periodic syndromes (CAPS) and growing evidence links NLRP3 inflammasome to common diseases such as Alzheimer´s disease. In order to modulate different stages of NLRP3 inflammasome assembly nine peptides whose sequences correspond to segments of inflammasome components NLRP3 and apoptosis-associated speck-like protein containing a CARD (ASC) were selected. Five peptides inhibited IL-1ß release, caspase-1 activation and ASC oligomerization in response to soluble and particulate NLRP3 triggers. Modulatory peptides also attenuated IL-1ß maturation induced by constitutive CAPS-associated NLRP3 mutants. Peptide corresponding to H2-H3 segment of ASC pyrin domain selectively inhibited NLRP3 inflammasome by binding to NLRP3 pyrin domain in the micromolar range. The peptide had no effect on AIM2 and NLRC4 inflammasomes as well as NF-κB pathway. The peptide effectively dampened neutrophil infiltration in the silica-induced peritonitis and when equipped with Antennapedia or Angiopep-2 motifs crossed the blood-brain barrier in a mouse model. Our study demonstrates that peptides represent an important tool for targeting multiprotein inflammatory complexes and can serve as the basis for the development of novel anti-inflammatory strategies for neurodegeneration.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peptides/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Blood-Brain Barrier/metabolism , Caspase 1/metabolism , Cells, Cultured , Cryopyrin-Associated Periodic Syndromes/metabolism , DNA-Binding Proteins/metabolism , Female , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Neutrophil Infiltration/physiology , Peritonitis/metabolism
11.
Int J Mol Sci ; 22(7)2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33805017

ABSTRACT

Identification of novel agents for bladder cancer treatment is highly desirable due to the high incidence of tumor recurrence and the risk of progression to muscle-invasive disease. The key feature of the cholesterol-dependent toxin listeriolysin O mutant (LLO Y406A) is its preferential activity at pH 5.7, which could be exploited either directly for selective targeting of cancer cells or the release of accumulated therapeutics from acidic endosomes. Therefore, our goal was to compare the cytotoxic effect of LLO Y406A on cancer cells (RT4) and normal urothelial cells (NPU), and to identify which cell membranes are the primary target of LLO Y406A by viability assays, life-cell imaging, fluorescence, and electron microscopy. LLO Y406A decreased viability, altered cell morphology, provoked membrane blebbing, and induced apoptosis in RT4 cells, while it did not affect NPU cells. LLO Y406A did not cause endosomal escape in RT4 cells, while the plasma membrane of RT4 cells was revealed as the primary target of LLO Y406A. It has been concluded that LLO Y406A has the ability to selectively eliminate cancer urothelial cells through pore-forming activity at the plasma membrane, without cytotoxic effects on normal urothelial cells. This promising selective activity merits further testing as an anti-cancer agent.


Subject(s)
Antineoplastic Agents/toxicity , Bacterial Toxins/toxicity , Cell Membrane/drug effects , Heat-Shock Proteins/toxicity , Hemolysin Proteins/toxicity , Urinary Bladder Neoplasms/metabolism , Urothelium/drug effects , Animals , Bacterial Toxins/genetics , Calcium/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Heat-Shock Proteins/genetics , Hemolysin Proteins/genetics , Humans , Mutation , Swine , Urothelium/metabolism
12.
J Infect Dis ; 222(9): 1505-1516, 2020 10 01.
Article in English | MEDLINE | ID: mdl-31970394

ABSTRACT

BACKGROUND: Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol. METHODS: Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO. RESULTS: Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin. CONCLUSIONS: Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Heat-Shock Proteins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Listeria monocytogenes/drug effects , Phosphorylcholine/pharmacology , Apoptosis , Calcium/metabolism , Caspase 3/metabolism , HeLa Cells , Humans , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
13.
Mol Microbiol ; 112(4): 1253-1269, 2019 10.
Article in English | MEDLINE | ID: mdl-31376198

ABSTRACT

Proteins of the aegerolysin family have a high abundance in Fungi. Due to their specific binding to membrane lipids, and their membrane-permeabilization potential in concert with protein partner(s) belonging to a membrane-attack-complex/perforin (MACPF) superfamily, they were proposed as useful tools in different biotechnological and biomedical applications. In this work, we performed functional studies on expression of the genes encoding aegerolysin and MACPF-like proteins in Aspergillus niger. Our results suggest the sporulation process being crucial for strong induction of the expression of all these genes. However, deletion of either of the aegerolysin genes did not influence the growth, development, sporulation efficiency and phenotype of the mutants, indicating that aegerolysins are not key factors in the sporulation process. In all our expression studies we noticed a strong correlation in the expression of one aegerolysin and MACPF-like gene. Aegerolysins were confirmed to be secreted from the fungus. We also showed the specific interaction of a recombinant A. niger aegerolysin with an invertebrate-specific membrane sphingolipid. Moreover, using this protein labelled with mCherry we successfully stained insect cells membranes containing this particular sphingolipid. Our combined results suggest, that aegerolysins in this species, and probably also in other aspergilli, could be involved in defence against predators.


Subject(s)
Complement Membrane Attack Complex/metabolism , Fungal Proteins/metabolism , Hemolysin Proteins/metabolism , Perforin/metabolism , Aspergillus niger/genetics , Aspergillus niger/metabolism , Complement Membrane Attack Complex/genetics , Fungal Proteins/physiology , Gene Expression Regulation, Fungal/genetics , Hemolysin Proteins/physiology , Membrane Proteins/metabolism , Perforin/genetics , Sphingolipids/metabolism , Spores, Fungal/genetics , Spores, Fungal/metabolism
14.
Anal Biochem ; 608: 113899, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32763307

ABSTRACT

A major drawback of the IgG capture step is the high cost of the protein A resin. For a better utilization of the resin, a continuous multi-column operation was recently proposed. In this method, accurate detection of leaking IgG is crucial to divert the breakthrough fluid from the waste to the next column and prolong the loading step without product loss. The detection of a breakthrough point as a change in UV absorption is based on a relatively small signal addition of IgGs to the bulk signal of host cell proteins. To achieve specificity, we used a quartz crystal microbalance and immobilized protein A as specific ligand on the sensor surface. We integrated the quartz crystal microbalance sensor in-line after the protein A column for real-time detection of IgGs in the breakthrough fluid. We show that this specific IgG detection in the breakthrough fluid can be more sensitive than with the UV detector. The use of the same product-specific ligand in the affinity column and in the sensor allows simultaneous in-line regeneration of column and sensor in a single step. Such a sensor could support cost-efficient load control during the entire continuous multi-column capture step in downstream processing.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Biosensing Techniques/methods , Chromatography, Affinity/methods , Quartz Crystal Microbalance Techniques/methods , Staphylococcal Protein A/chemistry , Biosensing Techniques/instrumentation , Chromatography, Affinity/instrumentation , Gold/chemistry , Quartz Crystal Microbalance Techniques/instrumentation , Spectrophotometry, Ultraviolet/methods
15.
Semin Cell Dev Biol ; 72: 133-141, 2017 12.
Article in English | MEDLINE | ID: mdl-28751252

ABSTRACT

The ancient phylum of Cnidaria contains many aquatic species with peculiar lifestyle. In order to survive, these organisms have evolved attack and defense mechanisms that are enabled by specialized cells and highly developed venoms. Pore-forming toxins are an important part of their venomous arsenal. Along some other types, the most representative are examples of four protein families that are commonly found in other kingdoms of life: actinoporins, Cry-like proteins, aerolysin-like toxins and MACPF/CDC toxins. Some of the homologues of pore-forming toxins may serve other functions, such as in food digestion, development and response against pathogenic organisms. Due to their interesting physico-chemical properties, the cnidarian pore-forming toxins may also serve as tools in medical research and nanobiotechnological applications.


Subject(s)
Cnidaria/metabolism , Cnidarian Venoms/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Toxins, Biological/metabolism , Animals , Cell Membrane/metabolism , Cnidaria/genetics , Cnidarian Venoms/chemistry , Cnidarian Venoms/genetics , Models, Molecular , Phylogeny , Pore Forming Cytotoxic Proteins/classification , Pore Forming Cytotoxic Proteins/genetics , Protein Conformation , Toxins, Biological/chemistry , Toxins, Biological/genetics
16.
J Nanobiotechnology ; 17(1): 108, 2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31623647

ABSTRACT

BACKGROUND: A major bottleneck in drug delivery is the breakdown and degradation of the delivery system through the endosomal/lysosomal network of the host cell, hampering the correct delivery of the drug of interest. In nature, the bacterial pathogen Listeria monocytogenes has developed a strategy to secrete Listeriolysin O (LLO) toxin as a tool to escape the eukaryotic lysosomal system upon infection, allowing it to grow and proliferate unharmed inside the host cell. RESULTS: As a "proof of concept", we present here the use of purified His-LLO H311A mutant protein and its conjugation on the surface of gold nanoparticles to promote the lysosomal escape of 40 nm-sized nanoparticles in mouse embryonic fibroblasts. Surface immobilization of LLO was achieved after specific functionalization of the nanoparticles with nitrile acetic acid, enabling the specific binding of histidine-tagged proteins. CONCLUSIONS: Endosomal acidification leads to release of the LLO protein from the nanoparticle surface and its self-assembly into a 300 Å pore that perforates the endosomal/lysosomal membrane, enabling the escape of nanoparticles.


Subject(s)
Bacterial Toxins/metabolism , Drug Carriers/metabolism , Endosomes/metabolism , Gold/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Nanoparticles/metabolism , Animals , Cell Line , Fibroblasts/metabolism , Hydrogen-Ion Concentration , Listeria monocytogenes/metabolism , Lysosomes/metabolism , Mice , Models, Molecular
17.
Int J Mol Sci ; 20(3)2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30736336

ABSTRACT

Diarrhea caused by enterotoxigenic Escherichia coli (ETEC) is one of the leading causes of mortality in children under five years of age and is a great burden on developing countries. The major virulence factor of the bacterium is the heat-labile enterotoxin (LT), a close homologue of the cholera toxin. The toxins bind to carbohydrate receptors in the gastrointestinal tract, leading to toxin uptake and, ultimately, to severe diarrhea. Previously, LT from human- and porcine-infecting ETEC (hLT and pLT, respectively) were shown to have different carbohydrate-binding specificities, in particular with respect to N-acetyllactosamine-terminating glycosphingolipids. Here, we probed 11 single-residue variants of the heat-labile enterotoxin with surface plasmon resonance spectroscopy and compared the data to the parent toxins. In addition we present a 1.45 Å crystal structure of pLTB in complex with branched lacto-N-neohexaose (Galß4GlcNAcß6[Galß4GlcNAcß3]Galß4Glc). The largest difference in binding specificity is caused by mutation of residue 94, which links the primary and secondary binding sites of the toxins. Residue 95 (and to a smaller extent also residues 7 and 18) also contribute, whereas residue 4 shows no effect on monovalent binding of the ligand and may rather be important for multivalent binding and avidity.


Subject(s)
Enterotoxigenic Escherichia coli/genetics , Enterotoxins/chemistry , Enterotoxins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Binding Sites , Carbohydrates/chemistry , Crystallography, X-Ray , Humans , Molecular Conformation , Protein Binding , Structure-Activity Relationship , Surface Plasmon Resonance
18.
Trends Biochem Sci ; 39(11): 510-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25440714

ABSTRACT

Pore-forming proteins (PFPs) interact with lipid bilayers to compromise membrane integrity. Many PFPs function by inserting a ring of oligomerized subunits into the bilayer to form a protein-lined hydrophilic channel. However, mounting evidence suggests that PFPs can also generate 'proteolipidic' pores by contributing to the fusion of inner and outer bilayer leaflets to form a toroidal structure. We discuss here toroidal pore formation by peptides including melittin, protegrin, and Alzheimer's Aß1-41, as well as by PFPs from several evolutionarily unrelated families: the colicin/Bcl-2 grouping including the pro-apoptotic protein Bax, actinoporins derived from sea anemones, and the membrane attack complex-perforin/cholesterol dependent cytolysin (MACPF/CDC) set of proteins. We also explore how the structure and biological role of toroidal pores might be investigated further.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Pore Forming Cytotoxic Proteins/chemistry , Cell Membrane/metabolism , Colicins/chemistry , Colicins/metabolism , Lipid Bilayers/metabolism , Melitten/chemistry , Melitten/metabolism , Membrane Lipids/metabolism , Models, Molecular , Pore Forming Cytotoxic Proteins/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
19.
J Membr Biol ; 251(3): 491-505, 2018 06.
Article in English | MEDLINE | ID: mdl-29476261

ABSTRACT

Archaeosomes are vesicles made of lipids from archaea. They possess many unique features in comparison to other lipid systems, with their high stability being the most prominent one, making them a promising system for biotechnological applications. Here, we report a preparation protocol of large unilamellar vesicles, giant unilamellar vesicles (GUVs), and nanodiscs from archaeal lipids with incorporated cholesterol. Incorporation of cholesterol led to additional increase in thermal stability of vesicles. Surface plasmon resonance, sedimentation assays, intrinsic tryptophan fluorescence measurements, calcein release experiments, and GUVs experiments showed that members of cholesterol-dependent cytolysins, listeriolysin O (LLO), and perfringolysin O (PFO), bind to cholesterol-rich archaeosomes and thereby retain their pore-forming activity. Interestingly, we observed specific binding of LLO, but not PFO, to archaeosomes even in the absence of cholesterol. This suggests a new capacity of LLO to bind to carbohydrate headgroups of archaeal lipids. Furthermore, we were able to express LLO inside GUVs by cell-free expression. GUVs made from archaeal lipids were highly stable, which could be beneficial for synthetic biology applications. In summary, our results describe novel model membrane systems for studying membrane interactions of proteins and their potential use in biotechnology.


Subject(s)
Archaea/metabolism , Cholesterol/chemistry , Cytotoxins/chemistry , Unilamellar Liposomes/chemistry , Cholesterol/metabolism , Cytotoxins/metabolism , Unilamellar Liposomes/metabolism
20.
PLoS Pathog ; 12(4): e1005597, 2016 04.
Article in English | MEDLINE | ID: mdl-27104344

ABSTRACT

Listeriolysin-O (LLO) plays a crucial role during infection by Listeria monocytogenes. It enables escape of bacteria from phagocytic vacuole, which is the basis for its spread to other cells and tissues. It is not clear how LLO acts at phagosomal membranes to allow bacterial escape. The mechanism of action of LLO remains poorly understood, probably due to unavailability of suitable experimental tools that could monitor LLO membrane disruptive activity in real time. Here, we used high-speed atomic force microscopy (HS-AFM) featuring high spatio-temporal resolution on model membranes and optical microscopy on giant unilamellar vesicles (GUVs) to investigate LLO activity. We analyze the assembly kinetics of toxin oligomers, the prepore-to-pore transition dynamics and the membrane disruption in real time. We reveal that LLO toxin efficiency and mode of action as a membrane-disrupting agent varies strongly depending on the membrane cholesterol concentration and the environmental pH. We discovered that LLO is able to form arc pores as well as damage lipid membranes as a lineactant, and this leads to large-scale membrane defects. These results altogether provide a mechanistic basis of how large-scale membrane disruption leads to release of Listeria from the phagocytic vacuole in the cellular context.


Subject(s)
Bacterial Toxins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Phagosomes/microbiology , Animals , Cell Line , Cholesterol/metabolism , Flow Cytometry , Microscopy, Atomic Force , Microscopy, Confocal
SELECTION OF CITATIONS
SEARCH DETAIL