Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Sci Rep ; 11(1): 13038, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158520

ABSTRACT

Translocation is essential to the anthrax toxin mechanism. Protective antigen (PA), the binding component of this AB toxin, forms an oligomeric pore that translocates lethal factor (LF) or edema factor, the active components of the toxin, into the cell. Structural details of the translocation process have remained elusive despite their biological importance. To overcome the technical challenges of studying translocation intermediates, we developed a method to immobilize, transition, and stabilize anthrax toxin to mimic important physiological steps in the intoxication process. Here, we report a cryoEM snapshot of PApore translocating the N-terminal domain of LF (LFN). The resulting 3.3 Å structure of the complex shows density of partially unfolded LFN near the canonical PApore binding site. Interestingly, we also observe density consistent with an α helix emerging from the 100 Å ß barrel channel suggesting LF secondary structural elements begin to refold in the pore channel. We conclude the anthrax toxin ß barrel aids in efficient folding of its enzymatic payload prior to channel exit. Our hypothesized refolding mechanism has broader implications for pore length of other protein translocating toxins.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Protein Refolding , Protein Unfolding , Antigens, Bacterial/ultrastructure , Models, Molecular , Nanoparticles/chemistry
2.
Biochim Biophys Acta Gen Subj ; 1864(3): 129499, 2020 03.
Article in English | MEDLINE | ID: mdl-31785327

ABSTRACT

Lyme disease is a tick-borne infection caused by Borrelia burgdorferi sensu lato complex spirochetes. The spirochete is located in the gut of the tick; as the infected tick starts the blood meal, the spirochete must travel through the hemolymph to the salivary glands, where it can spread to and infect the new host organism. In this study, we determined the crystal structures of the key outer surface protein BBE31 from B. burgdorferi and its orthologous protein BSE31 (BSPA14S_RS05060 gene product) from B. spielmanii. BBE31 is known to be important for the transfer of B. burgdorferi from the gut to the hemolymph in the tick after a tick bite. While BBE31 exerts its function by interacting with the Ixodes scapularis tick gut protein TRE31, structural and mass spectrometry data revealed that BBE31 has a glutathione (GSH) covalently attached to Cys142 suggesting that the protein may have acquired some additional functions in contrast to its orthologous protein BSE31, which lacks any interactions with GSH. In the current study, in addition to analyzing the potential reasons for GSH binding, the three-dimensional structure of BBE31 provides new insights into the molecular details of the transmission process as the protein plays an important role in the initial phase before the spirochete is physically transferred to the new host. This knowledge will be potentially used for the development of new strategies to fight against Lyme disease.


Subject(s)
Antigens, Bacterial/ultrastructure , Bacterial Outer Membrane Proteins/ultrastructure , Borrelia burgdorferi/metabolism , Lyme Disease/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/pathogenicity , Glutathione/metabolism , Humans , Ixodes/metabolism , Lyme Disease/transmission , Spirochaetales , Spirochaetales Infections/metabolism
3.
Toxins (Basel) ; 9(10)2017 09 22.
Article in English | MEDLINE | ID: mdl-28937604

ABSTRACT

The anthrax lethal toxin consists of protective antigen (PA) and lethal factor (LF). Understanding both the PA pore formation and LF translocation through the PA pore is crucial to mitigating and perhaps preventing anthrax disease. To better understand the interactions of the LF-PA engagement complex, the structure of the LFN-bound PA pore solubilized by a lipid nanodisc was examined using cryo-EM. CryoSPARC was used to rapidly sort particle populations of a heterogeneous sample preparation without imposing symmetry, resulting in a refined 17 Å PA pore structure with 3 LFN bound. At pH 7.5, the contributions from the three unstructured LFN lysine-rich tail regions do not occlude the Phe clamp opening. The open Phe clamp suggests that, in this translocation-compromised pH environment, the lysine-rich tails remain flexible and do not interact with the pore lumen region.


Subject(s)
Antigens, Bacterial/ultrastructure , Anthrax , Bacterial Toxins , Cryoelectron Microscopy , Molecular Dynamics Simulation , Protein Structure, Tertiary
4.
Micron ; 93: 1-8, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27883989

ABSTRACT

Negatively stained transmission electron microscope images are presented that depict the aggregation of recombinant anthrax protective antigen (rPA83 monomer and the PA63 prepore oligomer) under varying in vitro biochemical conditions. Heat treatment (50°C) of rPA83 produced clumped fibrils, but following heating the PA63 prepore formed disordered aggregates. Freeze-thaw treatment of the PA63 prepore generated linear flexuous aggregates of the heptameric oligomers. Aqueous suspensions of cholesterol microcrystals were shown to bind small rPA83 aggregates at the edges of the planar bilayers. With PA63 a more discrete binding of the prepores to the crystalline cholesterol bilayer edges occurs. Sodium deoxycholate (NaDOC) treatment of rPA83 produced quasi helical fibrillar aggregate, similar but not identical to that produced by heat treatment. Remarkably, NaDOC treatment of the PA63 prepores induced transformation into pores, with a characteristic extended ß-barrel. The PA63 pores aggregated as dimers, that aggregated further as angular chains and closed structures in higher NaDOC concentrations. The significance of the sterol interaction is discussed in relation to its likely importance for PA action in vivo.


Subject(s)
Antigens, Bacterial/ultrastructure , Protein Aggregates/physiology , Sterols/pharmacology , Antigens, Bacterial/drug effects , Antigens, Bacterial/genetics , Bacterial Toxins/genetics , Cholesterol/chemistry , Cholesterol/pharmacology , Crystallization , Deoxycholic Acid/administration & dosage , Deoxycholic Acid/pharmacology , Freezing , Hot Temperature , Hydrogen-Ion Concentration , Microscopy, Electron, Transmission/methods , Models, Molecular , Negative Staining , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/ultrastructure
5.
Mol Microbiol ; 102(4): 593-610, 2016 11.
Article in English | MEDLINE | ID: mdl-27507539

ABSTRACT

Three pathogenic species of the genus Yersinia assemble adhesive fimbriae via the FGL-chaperone/usher pathway. Closely related Y. pestis and Y. pseudotuberculosis elaborate the pH6 antigen (Psa), which mediates bacterial attachment to alveolar cells of the lung. Y. enterocolitica, instead, assembles the homologous fimbriae Myf of unknown function. Here, we discovered that Myf, like Psa, specifically recognizes ß1-3- or ß1-4-linked galactose in glycosphingolipids, but completely lacks affinity for phosphatidylcholine, the main receptor for Psa in alveolar cells. The crystal structure of a subunit of Psa (PsaA) complexed with choline together with mutagenesis experiments revealed that PsaA has four phosphatidylcholine binding pockets that enable super-high-avidity binding of Psa-fibres to cell membranes. The pockets are arranged as six tyrosine residues, which are all missing in the MyfA subunit of Myf. Conversely, the crystal structure of the MyfA-galactose complex revealed that the galactose-binding site is more extended in MyfA, enabling tighter binding to lactosyl moieties. Our results suggest that during evolution, Psa has acquired a tyrosine-rich surface that enables it to bind to phosphatidylcholine and mediate adhesion of Y. pestis/pseudotuberculosis to alveolar cells, whereas Myf has specialized as a carbohydrate-binding adhesin, facilitating the attachment of Y. enterocolitica to intestinal cells.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Fimbriae, Bacterial/metabolism , Yersinia/metabolism , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Antigens, Bacterial/genetics , Antigens, Bacterial/ultrastructure , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/ultrastructure , Binding Sites , Fimbriae Proteins/metabolism , Molecular Chaperones/metabolism , Tropism/genetics , Virulence/genetics , Yersinia enterocolitica/metabolism , Yersinia pestis/metabolism , Yersinia pseudotuberculosis/metabolism
6.
J Mol Biol ; 427(20): 3340-3349, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26344833

ABSTRACT

Membrane-embedded molecular machines are utilized to move water-soluble proteins across these barriers. Anthrax toxin forms one such machine through the self-assembly of its three component proteins--protective antigen (PA), lethal factor, and edema factor. Upon endocytosis into host cells, acidification of the endosome induces PA to form a membrane-inserted channel, which unfolds lethal factor and edema factor and translocates them into the host cytosol. Translocation is driven by the proton motive force, composed of the chemical potential, the proton gradient (ΔpH), and the membrane potential (Δψ). A crystal structure of the lethal toxin core complex revealed an "α clamp" structure that binds to substrate helices nonspecifically. Here, we test the hypothesis that, through the recognition of unfolding helical structure, the α clamp can accelerate the rate of translocation. We produced a synthetic PA mutant in which an α helix was crosslinked into the α clamp to block its function. This synthetic construct impairs translocation by raising a yet uncharacterized translocation barrier shown to be much less force dependent than the known unfolding barrier. We also report that the α clamp more stably binds substrates that can form helices than those, such as polyproline, that cannot. Hence, the α clamp recognizes substrates by a general shape-complementarity mechanism. Substrates that are incapable of forming compact secondary structure (due to the introduction of a polyproline track) are severely deficient for translocation. Therefore, the α clamp and its recognition of helical structure in the translocating substrate play key roles in the molecular mechanism of protein translocation.


Subject(s)
Antigens, Bacterial/metabolism , Antigens, Bacterial/ultrastructure , Bacterial Toxins/metabolism , Membrane Potentials , Protein Transport/physiology , Proton-Motive Force , Bacillus anthracis/metabolism , Binding Sites , Cell Membrane/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Endocytosis
7.
Nature ; 521(7553): 545-9, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25778700

ABSTRACT

Anthrax toxin, comprising protective antigen, lethal factor, and oedema factor, is the major virulence factor of Bacillus anthracis, an agent that causes high mortality in humans and animals. Protective antigen forms oligomeric prepores that undergo conversion to membrane-spanning pores by endosomal acidification, and these pores translocate the enzymes lethal factor and oedema factor into the cytosol of target cells. Protective antigen is not only a vaccine component and therapeutic target for anthrax infections but also an excellent model system for understanding the mechanism of protein translocation. On the basis of biochemical and electrophysiological results, researchers have proposed that a phi (Φ)-clamp composed of phenylalanine (Phe)427 residues of protective antigen catalyses protein translocation via a charge-state-dependent Brownian ratchet. Although atomic structures of protective antigen prepores are available, how protective antigen senses low pH, converts to active pore, and translocates lethal factor and oedema factor are not well defined without an atomic model of its pore. Here, by cryo-electron microscopy with direct electron counting, we determine the protective antigen pore structure at 2.9-Å resolution. The structure reveals the long-sought-after catalytic Φ-clamp and the membrane-spanning translocation channel, and supports the Brownian ratchet model for protein translocation. Comparisons of four structures reveal conformational changes in prepore to pore conversion that support a multi-step mechanism by which low pH is sensed and the membrane-spanning channel is formed.


Subject(s)
Antigens, Bacterial/metabolism , Antigens, Bacterial/ultrastructure , Bacillus anthracis/chemistry , Bacillus anthracis/ultrastructure , Bacterial Toxins/metabolism , Cryoelectron Microscopy , Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Biocatalysis , Hydrogen-Ion Concentration , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/ultrastructure , Models, Molecular , Phenylalanine/metabolism , Protein Conformation , Protein Transport , Structure-Activity Relationship
8.
J Membr Biol ; 248(3): 595-607, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25578459

ABSTRACT

Bacterial toxin or viral entry into the cell often requires cell surface binding and endocytosis. The endosomal acidification induces a limited unfolding/refolding and membrane insertion reaction of the soluble toxins or viral proteins into their translocation competent or membrane inserted states. At the molecular level, the specific orientation and immobilization of the pre-transitioned toxin on the cell surface is often an important prerequisite prior to cell entry. We propose that structures of some toxin membrane insertion complexes may be observed through procedures where one rationally immobilizes the soluble toxin so that potential unfolding â†” refolding transitions that occur prior to membrane insertion orientate away from the immobilization surface in the presence of lipid micelle pre-nanodisc structures. As a specific example, the immobilized prepore form of the anthrax toxin pore translocon or protective antigen can be transitioned, inserted into a model lipid membrane (nanodiscs), and released from the immobilized support in its membrane solubilized form. This particular strategy, although unconventional, is a useful procedure for generating pure membrane-inserted toxins in nanodiscs for electron microscopy structural analysis. In addition, generating a similar immobilized platform on label-free biosensor surfaces allows one to observe the kinetics of these acid-induced membrane insertion transitions. These platforms can facilitate the rational design of inhibitors that specifically target the toxin membrane insertion transitions that occur during endosomal acidification. This approach may lead to a new class of direct anti-toxin inhibitors.


Subject(s)
Lipid Bilayers/chemistry , Antigens, Bacterial/ultrastructure , Bacterial Toxins , Biosensing Techniques , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Chaperonin 60/ultrastructure , Cryoelectron Microscopy , Endosomes/chemistry , Models, Molecular , Nanostructures/chemistry , Nanostructures/ultrastructure , Protein Structure, Tertiary
9.
Biochem Biophys Res Commun ; 450(1): 335-40, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24944022

ABSTRACT

NMR-based structural biology urgently needs cost- and time-effective methods to assist both in the process of acquiring high-resolution NMR spectra and their subsequent analysis. Especially for bigger proteins (>20 kDa) selective labeling is a frequently used means of sequence-specific assignment. In this work we present the successful overexpression of a polypeptide of 233 residues, corresponding to the structured part of the N-terminal domain of Anthrax Lethal Factor, using Escherichia coli expression system. The polypeptide was subsequently isolated in pure, soluble form and analyzed structurally by solution NMR spectroscopy. Due to the non-satisfying quality and resolution of the spectra of this 27 kDa protein, an almost complete backbone assignment became feasible only by the combination of uniform and novel amino acid-selective labeling schemes. Moreover, amino acid-type selective triple-resonance NMR experiments proved to be very helpful.


Subject(s)
Amino Acids/chemistry , Antigens, Bacterial/chemistry , Antigens, Bacterial/ultrastructure , Bacterial Toxins/chemistry , Magnetic Resonance Spectroscopy/methods , Protein Structure, Secondary , Protein Structure, Tertiary , Staining and Labeling/methods
10.
Int J Nanomedicine ; 9: 1367-77, 2014.
Article in English | MEDLINE | ID: mdl-24648734

ABSTRACT

Development of lipid-based adjuvant formulations to enhance the immunogenicity of recombinant vaccine antigens is a focus of modern vaccine research. Characterizing interactions between vaccine antigens and formulation excipients is important for establishing compatibility between the different components and optimizing vaccine stability and potency. Cryogenic transmission electron microscopy (TEM) is a highly informative analytical technique that may elucidate various aspects of protein- and lipid-based structures, including morphology, size, shape, and phase structure, while avoiding artifacts associated with staining-based TEM. In this work, cryogenic TEM is employed to characterize a recombinant tuberculosis vaccine antigen, an anionic liposome formulation, and antigen-liposome interactions. By performing three-dimensional tomographic reconstruction analysis, the formation of a population of protein-containing flattened liposomes, not present in the control samples, was detected. It is shown that cryogenic TEM provides unique information regarding antigen-liposome interactions not detectable by light-scattering-based methods. Employing a suite of complementary analytical techniques is important to fully characterize interactions between vaccine components.


Subject(s)
Antigens, Bacterial/chemistry , Tuberculosis Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Bacterial/ultrastructure , Cryoelectron Microscopy , Humans , Imaging, Three-Dimensional , Liposomes/administration & dosage , Liposomes/chemistry , Nanomedicine , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Particle Size , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/chemistry , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
11.
J Immunol ; 191(10): 5268-77, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24108697

ABSTRACT

MR1-restricted mucosal-associated invariant T (MAIT) cells represent a subpopulation of αß T cells with innate-like properties and limited TCR diversity. MAIT cells are of interest because of their reactivity against bacterial and yeast species, suggesting that they play a role in defense against pathogenic microbes. Despite the advances in understanding MAIT cell biology, the molecular and structural basis behind their ability to detect MR1-Ag complexes is unclear. In this study, we present our structural and biochemical characterization of MAIT TCR engagement of MR1 presenting an Escherichia coli-derived stimulatory ligand, rRL-6-CH2OH, previously found in Salmonella typhimurium. We show a clear enhancement of MAIT TCR binding to MR1 due to the presentation of this ligand. Our structure of a MAIT TCR/MR1/rRL-6-CH2OH complex shows an evolutionarily conserved binding orientation, with a clear role for both the CDR3α and CDR3ß loops in recognizing the rRL-6-CH2OH stimulatory ligand. We also present two additional xenoreactive MAIT TCR/MR1 complexes that recapitulate the docking orientation documented previously, despite having variation in the CDR2ß and CDR3ß loop sequences. Our data support a model by which MAIT TCRs engage MR1 in a conserved fashion, with their binding affinities modulated by the nature of the MR1-presented Ag or diversity introduced by alternate Vß usage or CDR3ß sequences.


Subject(s)
Antigens, Bacterial/immunology , Histocompatibility Antigens Class I/metabolism , Multiprotein Complexes/ultrastructure , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/immunology , Antigen Presentation/immunology , Antigens, Bacterial/ultrastructure , Crystallography, X-Ray , Escherichia coli/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/ultrastructure , Humans , Lymphocyte Activation/immunology , Minor Histocompatibility Antigens , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/ultrastructure , Salmonella typhimurium/immunology , T-Lymphocyte Subsets/metabolism
12.
Biochemistry ; 52(37): 6335-47, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23964683

ABSTRACT

Domain 2 of the anthrax protective antigen (PA) prepore heptamer unfolds and refolds during endosome acidification to generate an extended 100 Å ß barrel pore that inserts into the endosomal membrane. The PA pore facilitates the pH-dependent unfolding and translocation of bound toxin enzymic components, lethal factor (LF) and/or edema factor, from the endosome to the cytoplasm. We constructed immobilized complexes of the prepore with the PA-binding domain of LF (LFN) to monitor the real-time prepore to pore kinetic transition using surface plasmon resonance and biolayer interferometry (BLI). The kinetics of this transition increased as the solution pH was decreased from 7.5 to 5.0, mirroring acidification of the endosome. Once it had undergone the transition, the LFN-PA pore complex was removed from the BLI biosensor tip and deposited onto electron microscopy grids, where PA pore formation was confirmed by negative stain electron microscopy. When the soluble receptor domain (ANTRX2/CMG2) binds the immobilized PA prepore, the transition to the pore state was observed only after the pH was lowered to early (pH 5.5) or late (pH 5.0) endosomal pH conditions. Once the pore formed, the soluble receptor readily dissociated from the PA pore. Separate binding experiments with immobilized PA pores and the soluble receptor indicate that the receptor has a weakened propensity to bind to the transitioned pore. This immobilized anthrax toxin platform can be used to identify or validate potential antimicrobial lead compounds capable of regulating and/or inhibiting anthrax toxin complex formation or pore transitions.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Endosomes/metabolism , Immobilized Proteins/chemistry , Intracellular Membranes/metabolism , Antigens, Bacterial/ultrastructure , Biosensing Techniques , Humans , Hydrogen-Ion Concentration , Interferometry , Kinetics , Micelles , Microscopy, Electron , Protein Folding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Peptide/metabolism , Surface Plasmon Resonance
13.
Protein Sci ; 22(5): 586-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23494942

ABSTRACT

We have visualized by cryo-electron microscopy (cryo-EM) the complex of the anthrax protective antigen (PA) translocon and the N-terminal domain of anthrax lethal factor (LF(N) inserted into a nanodisc model lipid bilayer. We have determined the structure of this complex at a nominal resolution of 16 Å by single-particle analysis and three-dimensional reconstruction. Consistent with our previous analysis of negatively stained unliganded PA, the translocon comprises a globular structure (cap) separated from the nanodisc bilayer by a narrow stalk that terminates in a transmembrane channel (incompletely distinguished in this reconstruction). The globular cap is larger than the unliganded PA pore, probably due to distortions introduced in the previous negatively stained structures. The cap exhibits larger, more distinct radial protrusions, previously identified with PA domain three, fitted by elements of the NMFF PA prepore crystal structure. The presence of LF(N), though not distinguished due to the seven-fold averaging used in the reconstruction, contributes to the distinct protrusions on the cap rim volume distal to the membrane. Furthermore, the lumen of the cap region is less resolved than the unliganded negatively stained PA, due to the low contrast obtained in our images of this specimen. Presence of the LF(N) extended helix and N terminal unstructured regions may also contribute to this additional internal density within the interior of the cap. Initial NMFF fitting of the cryoEM-defined PA pore cap region positions the Phe clamp region of the PA pore translocon directly above an internal vestibule, consistent with its role in toxin translocation.


Subject(s)
Anthrax/microbiology , Antigens, Bacterial/chemistry , Antigens, Bacterial/ultrastructure , Bacillus anthracis/chemistry , Bacterial Toxins/chemistry , Bacillus anthracis/ultrastructure , Cryoelectron Microscopy , Lipid Bilayers/chemistry
14.
Protein Sci ; 22(4): 492-501, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23389868

ABSTRACT

We have devised a procedure to incorporate the anthrax protective antigen (PA) pore complexed with the N-terminal domain of anthrax lethal factor (LFN ) into lipid nanodiscs and analyzed the resulting complexes by negative-stain electron microscopy. Insertion into nanodiscs was performed without relying on primary and secondary detergent screens. The preparations were relatively pure, and the percentage of PA pore inserted into nanodiscs on EM grids was high (∼43%). Three-dimensional analysis of negatively stained single particles revealed the LFN -PA nanodisc complex mirroring the previous unliganded PA pore nanodisc structure, but with additional protein density consistent with multiple bound LFN molecules on the PA cap region. The assembly procedure will facilitate collection of higher resolution cryo-EM LFN -PA nanodisc structures and use of advanced automated particle selection methods.


Subject(s)
Antigens, Bacterial/ultrastructure , Lipids/chemistry , Nanostructures/ultrastructure , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Cryoelectron Microscopy , Models, Molecular , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
15.
J Mol Biol ; 425(7): 1119-26, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23353825

ABSTRACT

An outer membrane protein BP26/OMP28 of Brucella, BP26, is identified as a major immunodominant antigen and widely used as a diagnostic marker and for vaccination against Brucellosis. BP26 belongs to the family of proteins that contains a SIMPL (signaling molecule that associates with the mouse pelle-like kinase) domain, whose structure and function have been unknown. Here, we present the crystal structure of BP26 revealing that 16 BP26 molecules form a novel channel-like assembly as also shown by electron microscopy analysis. Eight BP26 molecules forming a ring structure contain a hole at the center of the octamer, and another octamer interacts with each other to form a channel having a large internal cavity. BP26 is found to be structurally similar to a bacteriophage protein involved in infection, implicating that BP26 might function during Brucella infection. In addition, the BP26 structure suggests that the protein functions as a multimeric channel-like form and provides a canonical model for the SIMPL domains.


Subject(s)
Bacterial Proteins/chemistry , Brucella/metabolism , Ion Channels/chemistry , Membrane Proteins/chemistry , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/ultrastructure , Bacterial Proteins/genetics , Bacterial Proteins/ultrastructure , Brucella/genetics , Brucella/immunology , Crystallography, X-Ray , Ion Channels/genetics , Ion Channels/ultrastructure , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
16.
J Mol Biol ; 420(1-2): 29-39, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22480614

ABSTRACT

Shigella flexneri is a Gram-negative enteric pathogen that is the predominant cause of bacillary dysentery. Shigella uses a type III secretion system to deliver effector proteins that alter normal target cell functions to promote pathogen invasion. The type III secretion apparatus (T3SA) consists of a basal body, an extracellular needle, and a tip complex that is responsible for delivering effectors into the host cell cytoplasm. IpaD [Ipa (invasion plasmid antigen)] is the first protein to localize to the T3SA needle tip, where it prevents premature effector secretion and serves as an environmental sensor for triggering recruitment of the translocator protein IpaB to the needle tip. Thus, IpaD would be expected to form a stable structure whose overall architecture supports its functions. It is not immediately obvious from the published IpaD crystal structure (Protein Data Bank ID 2j0o) how a multimer of IpaD would be incorporated at the tip of the first static T3SA intermediate, nor what its functional role would be in building a mature T3SA. Here, we produce three-dimensional reconstructions from transmission electron microscopy images of IpaD localized at the Shigella T3SA needle tip for comparison to needle tips from a Shigella ipaD-null mutant. The results demonstrate that IpaD resides as a homopentamer at the needle tip of the T3SA. Furthermore, comparison to tips assembled from the distal domain IpaD(Δ192-267) mutation shows that IpaD adopts an elongated conformation that facilitates its ability to control type III secretion and stepwise assembly of the T3SA needle tip complex.


Subject(s)
Antigens, Bacterial/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Bacterial Secretion Systems , Shigella flexneri/chemistry , Shigella flexneri/ultrastructure , Antigens, Bacterial/chemistry , Imaging, Three-Dimensional , Microscopy, Electron, Scanning , Models, Molecular , Protein Structure, Quaternary , Shigella flexneri/pathogenicity
17.
Vaccine ; 28(35): 5746-54, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20600492

ABSTRACT

Caf1 of the plague bacterium, Yersinia pestis is a polymeric virulence factor and vaccine component, formed from monomers by a donor strand exchange (DSE) mechanism. Here, EM images of Caf1 reveal flexible polymers up to 1.5 microm long (4MDa). The bead-like structures along the polymer are 5.8 + or - 1 nm long and correspond to single Caf1 proteins. Short polymers often form circles, presumably by DSE. We also provide the first images of proteins bound to alhydrogel adjuvant. Caf1, hemocyanin and anthrax PA are all resolved clearly and Caf1 exhibits adjuvant bound stretches with long intervening loops draped from the edges.


Subject(s)
Adjuvants, Immunologic , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Polymers/chemistry , Virulence Factors/chemistry , Antigens, Bacterial/immunology , Antigens, Bacterial/ultrastructure , Bacterial Capsules/chemistry , Bacterial Capsules/immunology , Bacterial Proteins/immunology , Bacterial Proteins/ultrastructure , Microscopy, Electron, Transmission , Models, Molecular , Plague/prevention & control , Plague Vaccine/immunology , Protein Folding , Protein Structure, Quaternary , Virulence Factors/immunology , Yersinia pestis/chemistry , Yersinia pestis/immunology
18.
J Mol Biol ; 399(5): 741-58, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20433851

ABSTRACT

Anthrax is caused by strains of Bacillus anthracis that produce two key virulence factors, anthrax toxin (Atx) and a poly-gamma-D-glutamic acid capsule. Atx is comprised of three proteins: protective antigen (PA) and two enzymes, lethal factor (LF) and edema factor (EF). To disrupt cell function, these components must assemble into holotoxin complexes, which contain either a ring-shaped homooctameric or homoheptameric PA oligomer bound to multiple copies of LF and/or EF, producing lethal toxin (LT), edema toxin, or mixtures thereof. Once a host cell endocytoses these complexes, PA converts into a membrane-inserted channel that translocates LF and EF into the cytosol. LT can assemble on host cell surfaces or extracellularly in plasma. We show that, under physiological conditions in bovine plasma, LT complexes containing heptameric PA aggregate and inactivate more readily than LT complexes containing octameric PA. LT complexes containing octameric PA possess enhanced stability, channel-forming activity, and macrophage cytotoxicity relative to those containing heptameric PA. Under physiological conditions, multiple biophysical probes reveal that heptameric PA can prematurely adopt the channel conformation, but octameric PA complexes remain in their soluble prechannel configuration, which allows them to resist aggregation and inactivation. We conclude that PA may form an octameric oligomeric state as a means to produce a more stable and active LT complex that could circulate freely in the blood.


Subject(s)
Antigens, Bacterial/blood , Antigens, Bacterial/chemistry , Bacterial Toxins/blood , Bacterial Toxins/chemistry , Animals , Antigens, Bacterial/ultrastructure , Bacillus anthracis/metabolism , Binding Sites , Cattle , Circular Dichroism , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Protein Multimerization
19.
Clin Nephrol ; 71(6): 703-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19473640

ABSTRACT

We herein report the case of a 12-year-old boy with dense deposit disease (DDD) evoked by streptococcal infection. He had been diagnosed to have asymptomatic hematuria syndrome at the age of 6 during school screening. At 12 years of age, he was found to have macrohematuria and overt proteinuria with hypocomplementemia 2 months after streptococcal pharyngitis. Renal biopsy showed endocapillary proliferative glomerulonephritis with double contours of the glomerular basement membrane. Hypocomplementemia and proteinuria were sustained for over 8 weeks. He was suspected to have dense deposit disease due to intramembranous deposits in the first and the second biopsies. 1 month after treatment with methylprednisolone pulse therapy, proteinuria decreased to a normal level. Microscopic hematuria disappeared 2 years later, but mild hypocomplementemia persisted for more than 7 years. Nephritis-associated plasmin receptor (NAPlr), a nephritic antigen for acute poststreptococcal glomerulonephritis, was found to be positive in the glomeruli for more than 8 weeks. DDD is suggested to be caused by dysgeneration of the alternative pathway due to C3NeF and impaired Factor H activity. A persistent deposition of NAPlr might be one of the factors which lead to complement dysgeneration. A close relationship was suggested to exist between the streptococcal infection and dense deposit disease in this case.


Subject(s)
Glomerulonephritis, Membranoproliferative/microbiology , Streptococcal Infections/complications , Antigens, Bacterial/ultrastructure , Child , Glomerulonephritis, Membranoproliferative/diagnosis , Glomerulonephritis, Membranoproliferative/drug therapy , Glomerulonephritis, Membranoproliferative/immunology , Glucocorticoids/administration & dosage , Hematuria/drug therapy , Hematuria/microbiology , Humans , Kidney/immunology , Kidney/pathology , Kidney/ultrastructure , Male , Methylprednisolone/administration & dosage , Proteinuria/drug therapy , Proteinuria/microbiology , Pulse Therapy, Drug , Receptors, Cell Surface/ultrastructure , Severity of Illness Index , Streptococcal Infections/diagnosis , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology
20.
Biophys J ; 95(3): 1157-64, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18645196

ABSTRACT

Nonelectrolyte polymers of poly(ethylene glycol) (PEG) were used to estimate the diameter of the ion channel formed by the Bacillus anthracis protective antigen 63 (PA(63)). Based on the ability of different molecular weight PEGs to partition into the pore and reduce channel conductance, the pore appears to be narrower than the one formed by Staphylococcus aureus alpha-hemolysin. Numerical integration of the PEG sample mass spectra and the channel conductance data were used to refine the estimate of the pore's PEG molecular mass cutoff (approximately 1400 g/mol). The results suggest that the limiting diameter of the PA(63) pore is <2 nm, which is consistent with an all-atom model of the PA(63) channel and previous experiments using large ions.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/ultrastructure , Bacillus anthracis/chemistry , Bacterial Toxins/chemistry , Models, Chemical , Models, Molecular , Polyethylene Glycols/chemistry , Computer Simulation , Electrolytes/chemistry , Porosity , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...