Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nature ; 610(7931): 349-355, 2022 10.
Article in English | MEDLINE | ID: mdl-36171290

ABSTRACT

Entomopathogenic nematodes are widely used as biopesticides1,2. Their insecticidal activity depends on symbiotic bacteria such as Photorhabdus luminescens, which produces toxin complex (Tc) toxins as major virulence factors3-6. No protein receptors are known for any Tc toxins, which limits our understanding of their specificity and pathogenesis. Here we use genome-wide CRISPR-Cas9-mediated knockout screening in Drosophila melanogaster S2R+ cells and identify Visgun (Vsg) as a receptor for an archetypal P. luminescens Tc toxin (pTc). The toxin recognizes the extracellular O-glycosylated mucin-like domain of Vsg that contains high-density repeats of proline, threonine and serine (HD-PTS). Vsg orthologues in mosquitoes and beetles contain HD-PTS and can function as pTc receptors, whereas orthologues without HD-PTS, such as moth and human versions, are not pTc receptors. Vsg is expressed in immune cells, including haemocytes and fat body cells. Haemocytes from Vsg knockout Drosophila are resistant to pTc and maintain phagocytosis in the presence of pTc, and their sensitivity to pTc is restored through the transgenic expression of mosquito Vsg. Last, Vsg knockout Drosophila show reduced bacterial loads and lethality from P. luminescens infection. Our findings identify a proteinaceous Tc toxin receptor, reveal how Tc toxins contribute to P. luminescens pathogenesis, and establish a genome-wide CRISPR screening approach for investigating insecticidal toxins and pathogens.


Subject(s)
Bacterial Toxins , CRISPR-Cas Systems , Drosophila Proteins , Drosophila melanogaster , Gene Editing , Virulence Factors , Animals , Bacterial Toxins/metabolism , Biological Control Agents , Culicidae , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/microbiology , Fat Body/cytology , Gene Knockdown Techniques , Hemocytes , Humans , Moths , Mucins , Pest Control, Biological , Phagocytosis , Photorhabdus/metabolism , Repetitive Sequences, Amino Acid , Transgenes , Virulence Factors/metabolism
2.
Annu Rev Microbiol ; 73: 247-265, 2019 09 08.
Article in English | MEDLINE | ID: mdl-31140906

ABSTRACT

Tc toxin complexes are virulence factors of many bacteria, including insect and human pathogens. Tc toxins are composed of three subunits that act together to perforate the host membrane, similar to a syringe, and translocate toxic enzymes into the host cell. The reactions of the toxic enzymes lead to deterioration and ultimately death of the cell. We review recent high-resolution structural and functional data that explain the mechanism of action of this type of bacterial toxin at an unprecedented level of molecular detail. We focus on the steps that are necessary for toxin activation and membrane permeation. This is where the largest conformational transitions appear. Furthermore, we compare the architecture and function of Tc toxins with those of anthrax toxin and vertebrate teneurin.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Animals , Humans , Insecta , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Protein Transport
3.
Nature ; 563(7730): 209-213, 2018 11.
Article in English | MEDLINE | ID: mdl-30232455

ABSTRACT

Tc toxins secrete toxic enzymes into host cells using a unique syringe-like injection mechanism. They are composed of three subunits, TcA, TcB and TcC. TcA forms the translocation channel and the TcB-TcC heterodimer functions as a cocoon that shields the toxic enzyme. Binding of the cocoon to the channel triggers opening of the cocoon and translocation of the toxic enzyme into the channel. Here we show in atomic detail how the assembly of the three components activates the toxin. We find that part of the cocoon completely unfolds and refolds into an alternative conformation upon binding. The presence of the toxic enzyme inside the cocoon is essential for its subnanomolar binding affinity for the TcA subunit. The enzyme passes through a narrow negatively charged constriction site inside the cocoon, probably acting as an extruder that releases the unfolded protein with its C terminus first into the translocation channel.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cryoelectron Microscopy , Multiprotein Complexes/ultrastructure , Photorhabdus/ultrastructure , Protein Refolding , Protein Unfolding , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/ultrastructure , Bacterial Toxins/biosynthesis , Cytotoxins/biosynthesis , Cytotoxins/chemistry , Cytotoxins/metabolism , Models, Biological , Models, Molecular , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Photorhabdus/chemistry , Protein Conformation , Protein Transport
4.
Biol Cell ; 114(6): 160-176, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35304921

ABSTRACT

BACKGROUND INFORMATION: Like most other cell surface proteins, α5 ß1 integrin is glycosylated, which is required for its various activities in ways that mostly remain to be determined. RESULTS: Here, we have established the first comprehensive site-specific glycan map of α5 ß1 integrin that was purified from a natural source, that is, rat liver. This analysis revealed striking site selective variations in glycan composition. Complex bi, tri, or tetraantennary N-glycans were predominant at various proportions at most potential N-glycosylation sites. A few of these sites were nonglycosylated or contained high mannose or hybrid glycans, indicating that early N-glycan processing was hindered. Almost all complex N-glycans had fully galactosylated and sialylated antennae. Moderate levels of core fucosylation and high levels of O-acetylation of NeuAc residues were observed at certain sites. An O-linked HexNAc was found in an EGF-like domain of ß1 integrin. The extensive glycan information that results from our study was projected onto a map of α5 ß1 integrin that was obtained by homology modeling. We have used this model for the discussion of how glycosylation might be used in the functional cycle of α5 ß1 integrin. A striking example concerns the involvement of glycan-binding galectins in the regulation of the molecular homeostasis of glycoproteins at the cell surface through the formation of lattices or endocytic pits according to the glycolipid-lectin (GL-Lect) hypothesis. CONCLUSION: We expect that the glycoproteomics data of the current study will serve as a resource for the exploration of structural mechanisms by which glycans control α5 ß1 integrin activity and endocytic trafficking. SIGNIFICANCE: Glycosylation of α5 ß1 integrin has been implicated in multiple aspects of integrin function and structure. Yet, detailed knowledge of its glycosylation, notably the specific sites of glycosylation, is lacking. Furthermore, the α5 ß1 integrin preparation that was analyzed here is from a natural source, which is of importance as there is not a lot of literature in the field about the glycosylation of "native" glycoproteins.


Subject(s)
Integrin alpha5 , Integrin beta1 , Polysaccharides , Animals , Glycoproteins/chemistry , Glycosylation , Integrin alpha5/chemistry , Integrin beta1/chemistry , Liver/metabolism , Polysaccharides/chemistry , Rats
5.
Proc Natl Acad Sci U S A ; 116(46): 23083-23090, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31666324

ABSTRACT

Tc toxins are modular toxin systems of insect and human pathogenic bacteria. They are composed of a 1.4-MDa pentameric membrane translocator (TcA) and a 250-kDa cocoon (TcB and TcC) encapsulating the 30-kDa toxic enzyme (C terminus of TcC). Binding of Tc toxins to target cells and a pH shift trigger the conformational transition from the soluble prepore state to the membrane-embedded pore. Subsequently, the toxic enzyme is translocated and released into the cytoplasm. A high-resolution structure of a holotoxin embedded in membranes is missing, leaving open the question of whether TcB-TcC has an influence on the conformational transition of TcA. Here we show in atomic detail a fully assembled 1.7-MDa Tc holotoxin complex from Photorhabdus luminescens in the membrane. We find that the 5 TcA protomers conformationally adapt to fit around the cocoon during the prepore-to-pore transition. The architecture of the Tc toxin complex allows TcB-TcC to bind to an already membrane-embedded TcA pore to form a holotoxin. Importantly, assembly of the holotoxin at the membrane results in spontaneous translocation of the toxic enzyme, indicating that this process is not driven by a proton gradient or other energy source. Mammalian lipids with zwitterionic head groups are preferred over other lipids for the integration of Tc toxins. In a nontoxic Tc toxin variant, we can visualize part of the translocating toxic enzyme, which transiently interacts with alternating negative charges and hydrophobic stretches of the translocation channel, providing insights into the mechanism of action of Tc toxins.


Subject(s)
Bacterial Toxins/chemistry , Photorhabdus/chemistry , ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Cryoelectron Microscopy , Photorhabdus/metabolism , Protein Structure, Quaternary
6.
J Biol Chem ; 291(11): 5652-5663, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26757820

ABSTRACT

The α-pore-forming toxin Cytolysin A (ClyA) is responsible for the hemolytic activity of various Escherichia coli and Salmonella enterica strains. Soluble ClyA monomers spontaneously assemble into annular dodecameric pore complexes upon contact with membranes or detergent. At ClyA monomer concentrations above ∼100 nm, the rate-limiting step in detergent- or membrane- induced pore assembly is the unimolecular reaction from the monomer to the assembly-competent protomer, which then oligomerizes rapidly to active pore complexes. In the absence of detergent, ClyA slowly forms soluble oligomers. Here we show that soluble ClyA oligomers cannot form dodecameric pore complexes after the addition of detergent and are hemolytically inactive. In addition, we demonstrate that the natural cysteine pair Cys-87/Cys-285 of ClyA forms a disulfide bond under oxidizing conditions and that both the oxidized and reduced ClyA monomers assemble to active pores via the same pathway in the presence of detergent, in which an unstructured, monomeric intermediate is transiently populated. The results show that the oxidized ClyA monomer assembles to pore complexes about one order of magnitude faster than the reduced monomer because the unstructured intermediate of oxidized ClyA is less stable and dissolves more rapidly than the reduced intermediate. Moreover, we show that oxidized ClyA forms soluble, inactive oligomers in the absence of detergent much faster than the reduced monomer, providing an explanation for several contradictory reports in which oxidized ClyA had been described as inactive.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Hemolysin Proteins/metabolism , Animals , Detergents/metabolism , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/ultrastructure , Hemolysin Proteins/chemistry , Hemolysin Proteins/ultrastructure , Hemolysis , Horses , Models, Molecular , Oxidation-Reduction , Protein Conformation , Protein Multimerization , Solubility
7.
Chembiochem ; 16(15): 2162-6, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26382254

ABSTRACT

The incorporation of the non-natural amino acids (4R)- and (4S)-fluoroproline (Flp) has been successfully used to improve protein stability, but little is known about their effect on protein folding kinetics. Here we analyzed the influence of (4R)- and (4S)-Flp on the rate-limiting trans-to-cis isomerization of the Ile75-Pro76 peptide bond in the folding of Escherichia coli thioredoxin (Trx). While (4R)-Flp at position 76 had essentially no effect on the isomerization rate in the context of the intact tertiary structure, (4S)-Flp accelerated the folding reaction ninefold. Similarly, tenfold faster trans-to-cis isomerization of Ile75-(4S)-Flp76 relative to Ile75-Pro76 was observed in the unfolded state of Trx. Our results show that the replacement of cis prolines by non-natural proline analogues can be used for modulating the folding rates of proteins with cis prolyl-peptide bonds in the native state.


Subject(s)
Proline/analogs & derivatives , Proline/chemistry , Protein Folding , Thioredoxins/chemistry , Escherichia coli/chemistry , Models, Molecular , Proline/metabolism , Stereoisomerism
8.
Biochemistry ; 53(40): 6357-69, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25222267

ABSTRACT

The α-pore-forming toxin Cytolysin A (ClyA) is responsible for the hemolytic phenotype of several Escherichia coli and Salmonella enterica strains. ClyA is a soluble, 34 kDa monomer that assembles into a dodecameric pore complex in the presence of membranes or detergent. The comparison of the X-ray structures of monomeric ClyA and the ClyA protomer in the pore complex revealed one of the largest conformational transitions observed so far in proteins, involving the structural rearrangement of more than half of all residues, which is consistent with the finding that conversion from the monomer to the assembly competent protomer is rate-limiting for pore assembly. Here, we introduced artificial disulfide bonds at two distinct sites into the ClyA monomer that both prevent a specific structural rearrangement required for protomer formation. Using electron microscopy and hemolytic activity assays, we show that the engineered disulfides indeed trap these ClyA variants in an assembly incompetent state. Assembly of the variants into functional pore complexes can be completely recovered by disulfide reduction. The assembly kinetics of the ClyA variants recorded with circular dichroism and fluorescence spectroscopy revealed the same mechanism of protomer formation that was observed for wild-type ClyA, proceeding via an intermediate with decreased secondary structure content.


Subject(s)
Escherichia coli Proteins/chemistry , Hemolysin Proteins/chemistry , Hemolytic Agents/chemistry , Amino Acid Substitution , Animals , Crystallography, X-Ray , Cystine/chemistry , Erythrocytes/drug effects , Erythrocytes/physiology , Escherichia coli , Escherichia coli Proteins/genetics , Escherichia coli Proteins/pharmacology , Fluorescence Resonance Energy Transfer , Hemolysin Proteins/genetics , Hemolysin Proteins/pharmacology , Hemolysis , Hemolytic Agents/pharmacology , Horses , Kinetics , Models, Molecular , Oxidation-Reduction , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary
9.
Nat Commun ; 13(1): 4202, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35858890

ABSTRACT

Tc toxins deliver toxic enzymes into host cells by a unique injection mechanism. One of these enzymes is the actin ADP-ribosyltransferase TccC3, whose activity leads to the clustering of the cellular cytoskeleton and ultimately cell death. Here, we show in atomic detail how TccC3 modifies actin. We find that the ADP-ribosyltransferase does not bind to G-actin but interacts with two consecutive actin subunits of F-actin. The binding of TccC3 to F-actin occurs via an induced-fit mechanism that facilitates access of NAD+ to the nucleotide binding pocket. The following nucleophilic substitution reaction results in the transfer of ADP-ribose to threonine-148 of F-actin. We demonstrate that this site-specific modification of F-actin prevents its interaction with depolymerization factors, such as cofilin, which impairs actin network turnover and leads to steady actin polymerization. Our findings reveal in atomic detail a mechanism of action of a bacterial toxin through specific targeting and modification of F-actin.


Subject(s)
Actins , Threonine , ADP Ribose Transferases/metabolism , ADP-Ribosylation , Actin Cytoskeleton/metabolism , Actins/metabolism , Threonine/metabolism
10.
Nat Commun ; 11(1): 2694, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483155

ABSTRACT

Toxin complex (Tc) toxins are virulence factors of pathogenic bacteria. Tcs are composed of three subunits: TcA, TcB and TcC. TcA facilitates receptor-toxin interaction and membrane permeation, TcB and TcC form a toxin-encapsulating cocoon. While the mechanisms of holotoxin assembly and pore formation have been described, little is known about receptor binding of TcAs. Here, we identify heparins/heparan sulfates and Lewis antigens as receptors for different TcAs from insect and human pathogens. Glycan array screening reveals that all tested TcAs bind negatively charged heparins. Cryo-EM structures of Morganella morganii TcdA4 and Xenorhabdus nematophila XptA1 reveal that heparins/heparan sulfates unexpectedly bind to different regions of the shell domain, including receptor-binding domains. In addition, Photorhabdus luminescens TcdA1 binds to Lewis antigens with micromolar affinity. Here, the glycan interacts with the receptor-binding domain D of the toxin. Our results suggest a glycan dependent association mechanism of Tc toxins on the host cell surface.


Subject(s)
Bacterial Toxins/toxicity , Cell Adhesion/drug effects , Cell Adhesion/physiology , Polysaccharides/metabolism , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/pharmacokinetics , Binding Sites , Cell Membrane/drug effects , Cell Membrane/metabolism , HEK293 Cells , Heparin/chemistry , Heparin/metabolism , Humans , Insecta/microbiology , Lewis X Antigen/chemistry , Lewis X Antigen/metabolism , Models, Molecular , Molecular Docking Simulation , Morganella morganii/pathogenicity , Photorhabdus/pathogenicity , Polysaccharides/chemistry , Xenorhabdus/pathogenicity
11.
Nat Commun ; 10(1): 5263, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748551

ABSTRACT

Tc toxins are bacterial protein complexes that inject cytotoxic enzymes into target cells using a syringe-like mechanism. Tc toxins are composed of a membrane translocator and a cocoon that encapsulates a toxic enzyme. The toxic enzyme varies between Tc toxins from different species and is not conserved. Here, we investigate whether the toxic enzyme can be replaced by other small proteins of different origin and properties, namely Cdc42, herpes simplex virus ICP47, Arabidopsis thaliana iLOV, Escherichia coli DHFR, Ras-binding domain of CRAF kinase, and TEV protease. Using a combination of electron microscopy, X-ray crystallography and in vitro translocation assays, we demonstrate that it is possible to turn Tc toxins into customizable molecular syringes for delivering proteins of interest across membranes. We also infer the guidelines that protein cargos must obey in terms of size, charge, and fold in order to apply Tc toxins as a universal protein translocation system.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Protein Translocation Systems/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Crystallography, X-Ray , Endopeptidases/chemistry , Endopeptidases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/metabolism , Microscopy, Electron , Models, Molecular , Photorhabdus/chemistry , Photorhabdus/metabolism , Protein Translocation Systems/chemistry , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism
12.
Commun Biol ; 2: 218, 2019.
Article in English | MEDLINE | ID: mdl-31240256

ABSTRACT

Selecting particles from digital micrographs is an essential step in single-particle electron cryomicroscopy (cryo-EM). As manual selection of complete datasets-typically comprising thousands of particles-is a tedious and time-consuming process, numerous automatic particle pickers have been developed. However, non-ideal datasets pose a challenge to particle picking. Here we present the particle picking software crYOLO which is based on the deep-learning object detection system You Only Look Once (YOLO). After training the network with 200-2500 particles per dataset it automatically recognizes particles with high recall and precision while reaching a speed of up to five micrographs per second. Further, we present a general crYOLO network able to pick from previously unseen datasets, allowing for completely automated on-the-fly cryo-EM data preprocessing during data acquisition. crYOLO is available as a standalone program under http://sphire.mpg.de/ and is distributed as part of the image processing workflow in SPHIRE.


Subject(s)
Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/methods , Software , Datasets as Topic , Deep Learning , Neural Networks, Computer
13.
Philos Trans R Soc Lond B Biol Sci ; 372(1726)2017 Aug 05.
Article in English | MEDLINE | ID: mdl-28630151

ABSTRACT

The cytolytic toxin cytolysin A (ClyA) from Escherichia coli is probably one of the best-characterized examples of bacterial, α-pore-forming toxins (α-PFTs). Like other PFTs, ClyA exists in a soluble, monomeric form that assembles to an annular, homo-oligomeric pore complex upon contact with detergent or target membranes. Comparison of the three-dimensional structures of the 34 kDa monomer and the protomer in the context of the dodecameric pore complex revealed that ClyA undergoes one of the largest conformational transitions described for proteins so far, in which 55% of the residues change their position and 16% of the residues adopt a different secondary structure in the protomer. Studies on the assembly of ClyA revealed a unique mechanism that differs from the assembly mechanism of other PFTs. The rate-liming step of pore formation proved to be the unimolecular conversion of the monomer to an assembly-competent protomer, during which a molten globule-like off-pathway intermediate accumulates. The oligomerization of protomers to pore complexes is fast and follows a kinetic scheme in which mixtures of linear oligomers of different size are formed first, followed by very rapid and specific association of pairs of oligomers that can directly perform ring closure to the dodecameric pore complex.This article is part of the themed issue 'Membrane pores: from structure and assembly, to medicine and technology'.


Subject(s)
Bacterial Toxins/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Hemolysin Proteins/chemistry , Cell Membrane/metabolism , Kinetics , Molecular Conformation
14.
Nat Struct Mol Biol ; 23(10): 884-890, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27571177

ABSTRACT

Tc toxins from pathogenic bacteria use a special syringe-like mechanism to perforate the host cell membrane and inject a deadly enzyme into the host cytosol. The molecular mechanism of this unusual injection system is poorly understood. Using electron cryomicroscopy, we determined the structure of TcdA1 from Photorhabdus luminescens embedded in lipid nanodiscs. In our structure, compared with the previous structure of TcdA1 in the prepore state, the transmembrane helices rearrange in the membrane and open the initially closed pore. However, the helices do not span the complete membrane; instead, the loops connecting the helices form the rim of the funnel. Lipid head groups reach into the space between the loops and consequently stabilize the pore conformation. The linker domain is folded and packed into a pocket formed by the other domains of the toxin, thereby considerably contributing to stabilization of the pore state.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Membrane Lipids/chemistry , Photorhabdus/chemistry , Pore Forming Cytotoxic Proteins/chemistry , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Thermodynamics
15.
Sci Rep ; 5: 11840, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26121966

ABSTRACT

Cis prolyl peptide bonds are conserved structural elements in numerous protein families, although their formation is energetically unfavorable, intrinsically slow and often rate-limiting for folding. Here we investigate the reasons underlying the conservation of the cis proline that is diagnostic for the fold of thioredoxin-like thiol-disulfide oxidoreductases. We show that replacement of the conserved cis proline in thioredoxin by alanine can accelerate spontaneous folding to the native, thermodynamically most stable state by more than four orders of magnitude. However, the resulting trans alanine bond leads to small structural rearrangements around the active site that impair the function of thioredoxin as catalyst of electron transfer reactions by more than 100-fold. Our data provide evidence for the absence of a strong evolutionary pressure to achieve intrinsically fast folding rates, which is most likely a consequence of proline isomerases and molecular chaperones that guarantee high in vivo folding rates and yields.


Subject(s)
Escherichia coli Proteins/chemistry , Protein Folding , Thioredoxins/chemistry , Amino Acid Substitution , Catalytic Domain , Circular Dichroism , Conserved Sequence , Crystallography, X-Ray , Escherichia coli , Escherichia coli Proteins/genetics , Isomerism , Kinetics , Models, Molecular , Oxidation-Reduction , Proline/chemistry , Protein Structure, Secondary , Thermodynamics , Thioredoxins/genetics
16.
Nat Commun ; 6: 6198, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25652783

ABSTRACT

Pore-forming toxins are protein assemblies used by many organisms to disrupt the membranes of target cells. They are expressed as soluble monomers that assemble spontaneously into multimeric pores. However, owing to their complexity, the assembly processes have not been resolved in detail for any pore-forming toxin. To determine the assembly mechanism for the ring-shaped, homododecameric pore of the bacterial cytolytic toxin ClyA, we collected a diverse set of kinetic data using single-molecule spectroscopy and complementary techniques on timescales from milliseconds to hours, and from picomolar to micromolar ClyA concentrations. The entire range of experimental results can be explained quantitatively by a surprisingly simple mechanism. First, addition of the detergent n-dodecyl-ß-D-maltopyranoside to the soluble monomers triggers the formation of assembly-competent toxin subunits, accompanied by the transient formation of a molten-globule-like intermediate. Then, all sterically compatible oligomers contribute to assembly, which greatly enhances the efficiency of pore formation compared with simple monomer addition.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Hemolysin Proteins/chemistry , Protein Multimerization , Protein Subunits/chemistry , Animals , Cells, Cultured , Erythrocytes/cytology , Erythrocytes/drug effects , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Escherichia coli Proteins/toxicity , Fluorescence Resonance Energy Transfer , Gene Expression , Hemolysin Proteins/biosynthesis , Hemolysin Proteins/genetics , Hemolysin Proteins/toxicity , Hemolysis/drug effects , Horses , Kinetics , Maltose/analogs & derivatives , Maltose/chemistry , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/biosynthesis , Protein Subunits/genetics , Protein Subunits/toxicity , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL