Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pathog Dis ; 76(8)2018 11 01.
Article in English | MEDLINE | ID: mdl-30452651

ABSTRACT

The adenylate cyclase toxin (CyaA) is a multi-domain protein secreted by Bordetella pertussis, the causative agent of whooping cough. CyaA is involved in the early stages of respiratory tract colonization by Bordetella pertussis. CyaA is produced and acylated in the bacteria, and secreted via a dedicated secretion system. The cell intoxication process involves a unique mechanism of transport of the CyaA toxin catalytic domain (ACD) across the plasma membrane of eukaryotic cells. Once translocated, ACD binds to and is activated by calmodulin and produces high amounts of cAMP, subverting the physiology of eukaryotic cells. Here, we review our work on the identification and characterization of a critical region of CyaA, the translocation region, required to deliver ACD into the cytosol of target cells. The translocation region contains a segment that exhibits membrane-active properties, i.e. is able to fold upon membrane interaction and permeabilize lipid bilayers. We proposed that this region is required to locally destabilize the membrane, decreasing the energy required for ACD translocation. To further study the translocation process, we developed a tethered bilayer lipid membrane (tBLM) design that recapitulate the ACD transport across a membrane separating two hermetic compartments. We showed that ACD translocation is critically dependent on calcium, membrane potential, CyaA acylation and on the presence of calmodulin in the trans compartment. Finally, we describe how calmodulin-binding triggers key conformational changes in ACD, leading to its activation and production of supraphysiological concentrations of cAMP.


Subject(s)
Adenylate Cyclase Toxin/metabolism , Bordetella pertussis/metabolism , Calmodulin/metabolism , Cyclic AMP/metabolism , Acylation , Adenylate Cyclase Toxin/chemistry , Calcium/metabolism , Cell Membrane/metabolism , Eukaryotic Cells , Humans , Membrane Potentials , Permeability , Protein Binding , Protein Conformation , Protein Folding , Protein Processing, Post-Translational , Protein Transport
2.
Toxicon ; 149: 37-44, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29337218

ABSTRACT

The adenylate cyclase toxin (CyaA) plays an essential role in the early stages of respiratory tract colonization by Bordetella pertussis, the causative agent of whooping cough. Once secreted, CyaA invades eukaryotic cells, leading to cell death. The cell intoxication process involves a unique mechanism of translocation of the CyaA catalytic domain directly across the plasma membrane of the target cell. Herein, we review our recent results describing how calcium is involved in several steps of this intoxication process. In conditions mimicking the low calcium environment of the crowded bacterial cytosol, we show that the C-terminal, calcium-binding Repeat-in-ToXin (RTX) domain of CyaA, RD, is an extended, intrinsically disordered polypeptide chain with a significant level of local, secondary structure elements, appropriately sized for transport through the narrow channel of the secretion system. Upon secretion, the high calcium concentration in the extracellular milieu induces the refolding of RD, which likely acts as a scaffold to favor the refolding of the upstream domains of the full-length protein. Due to the presence of hydrophobic regions, CyaA is prone to aggregate into multimeric forms in vitro, in the absence of a chaotropic agent. We have recently defined the experimental conditions required for CyaA folding, comprising both calcium binding and molecular confinement. These parameters are critical for CyaA folding into a stable, monomeric and functional form. The monomeric, calcium-loaded (holo) toxin exhibits efficient liposome permeabilization and hemolytic activities in vitro, even in a fully calcium-free environment. By contrast, the toxin requires sub-millimolar calcium concentrations in solution to translocate its catalytic domain across the plasma membrane, indicating that free calcium in solution is actively involved in the CyaA toxin translocation process. Overall, this data demonstrates the remarkable adaptation of bacterial RTX toxins to the diversity of calcium concentrations it is exposed to in the successive environments encountered in the course of the intoxication process.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Calcium/chemistry , Models, Biological , Whooping Cough/microbiology , Adenylate Cyclase Toxin/metabolism , Bordetella pertussis , Eukaryotic Cells/microbiology , Protein Domains , Protein Folding , Protein Translocation Systems , Protein Transport
3.
J Biol Chem ; 289(44): 30702-30716, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25231985

ABSTRACT

The adenylate cyclase (CyaA) toxin, a multidomain protein of 1706 amino acids, is one of the major virulence factors produced by Bordetella pertussis, the causative agent of whooping cough. CyaA is able to invade eukaryotic target cells in which it produces high levels of cAMP, thus altering the cellular physiology. Although CyaA has been extensively studied by various cellular and molecular approaches, the structural and functional states of the toxin remain poorly characterized. Indeed, CyaA is a large protein and exhibits a pronounced hydrophobic character, making it prone to aggregation into multimeric forms. As a result, CyaA has usually been extracted and stored in denaturing conditions. Here, we define the experimental conditions allowing CyaA folding into a monomeric and functional species. We found that CyaA forms mainly multimers when refolded by dialysis, dilution, or buffer exchange. However, a significant fraction of monomeric, folded protein could be obtained by exploiting molecular confinement on size exclusion chromatography. Folding of CyaA into a monomeric form was found to be critically dependent upon the presence of calcium and post-translational acylation of the protein. We further show that the monomeric preparation displayed hemolytic and cytotoxic activities suggesting that the monomer is the genuine, physiologically active form of the toxin. We hypothesize that the structural role of the post-translational acylation in CyaA folding may apply to other RTX toxins.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Bacterial Proteins/chemistry , Bordetella pertussis/enzymology , Calcium/chemistry , Acylation , Adenylate Cyclase Toxin/isolation & purification , Adenylate Cyclase Toxin/pharmacology , Animals , Bacterial Proteins/isolation & purification , Bacterial Proteins/pharmacology , Chromatography, Gel , Circular Dichroism , Erythrocytes/drug effects , Erythrocytes/physiology , Hemolysis , Protein Processing, Post-Translational , Protein Refolding , Protein Structure, Quaternary , Protein Structure, Secondary , Sheep , Urea/chemistry
4.
J Biol Chem ; 288(45): 32585-32598, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24064217

ABSTRACT

Bordetella pertussis, the pathogenic bacteria responsible for whooping cough, secretes several virulence factors, among which is the adenylate cyclase toxin (CyaA) that plays a crucial role in the early stages of human respiratory tract colonization. CyaA invades target cells by translocating its catalytic domain directly across the plasma membrane and overproduces cAMP, leading to cell death. The molecular process leading to the translocation of the catalytic domain remains largely unknown. We have previously shown that the catalytic domain per se, AC384, encompassing residues 1-384 of CyaA, did not interact with lipid bilayer, whereas a longer polypeptide, AC489, spanning residues 1-489, binds to membranes and permeabilizes vesicles. Moreover, deletion of residues 375-485 within CyaA abrogated the translocation of the catalytic domain into target cells. Here, we further identified within this region a peptidic segment that exhibits membrane interaction properties. A synthetic peptide, P454, corresponding to this sequence (residues 454-485 of CyaA) was characterized by various biophysical approaches. We found that P454 (i) binds to membranes containing anionic lipids, (ii) adopts an α-helical structure oriented in plane with respect to the lipid bilayer, and (iii) permeabilizes vesicles. We propose that the region encompassing the helix 454-485 of CyaA may insert into target cell membrane and induce a local destabilization of the lipid bilayer, thus favoring the translocation of the catalytic domain across the plasma membrane.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Bacterial Proteins/chemistry , Bordetella pertussis/chemistry , Lipid Bilayers/chemistry , Peptides/chemistry , Adenylate Cyclase Toxin/metabolism , Bacterial Proteins/metabolism , Bordetella pertussis/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Humans , Lipid Bilayers/metabolism , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Protein Transport
5.
Methods Mol Biol ; 896: 163-77, 2012.
Article in English | MEDLINE | ID: mdl-22821523

ABSTRACT

Size exclusion chromatography coupled online to a Tetra Detector Array in combination with analytical ultracentrifugation (or with quasi-elastic light scattering) is a useful methodology to characterize hydrodynamic properties of macromolecules, including intrinsically disordered proteins. The time-averaged apparent hydration and the shape factor of proteins can be estimated from the measured parameters (molecular mass, intrinsic viscosity, hydrodynamic radius) by these techniques. Here we describe in detail this methodology and its application to characterize hydrodynamic and conformational changes in proteins.


Subject(s)
Chemistry Techniques, Analytical/methods , Hydrodynamics , Proteins/chemistry , Chromatography, Gel , Light , Proteins/isolation & purification , Scattering, Radiation , Spectrophotometry , Time Factors , Ultracentrifugation , Water/chemistry
6.
Methods Mol Biol ; 896: 331-49, 2012.
Article in English | MEDLINE | ID: mdl-22821535

ABSTRACT

Under physiological conditions, intrinsically disordered proteins (IDPs) are unfolded, mainly because of their low hydrophobicity and the strong electrostatic repulsion between charged residues of the same sign within the protein. Softwares have been designed to facilitate the computation of the mean net charge of proteins (formally protein valence) from their amino acid sequences. Nevertheless, discrepancies between experimental and computed valence values for several proteins have been reported in the literature. Hence, experimental approaches are required to obtain accurate estimation of protein valence in solution. Moreover, ligand-induced disorder-to-order transition is involved in the folding of numerous IDPs. Some of the ligands are cations or anions, which, upon protein binding, decrease the mean net charge of the protein, favoring its folding via a charge reduction effect. An accurate determination of the mean net charge of protein in both its ligand-free intrinsically disordered state and in its folded, ligand-bound state allows one to estimate the number of ligands bound to the protein in the holo-state. Here, we describe an experimental protocol to determine the mean net charge of protein, from its electrophoretic mobility, its molecular mass and its hydrodynamic radius.


Subject(s)
Electrophoresis/methods , Motion , Proteins/chemistry , Calcium/pharmacology , Hydrodynamics , Molecular Weight , Protein Folding/drug effects
7.
J Biol Chem ; 287(12): 9200-12, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22241477

ABSTRACT

The adenylate cyclase (CyaA) toxin, one of the virulence factors secreted by Bordetella pertussis, the pathogenic bacteria responsible for whooping cough, plays a critical role in the early stages of respiratory tract colonization by this bacterium. The CyaA toxin is able to invade eukaryotic cells by translocating its N-terminal catalytic domain directly across the plasma membrane of the target cells, where, activated by endogenous calmodulin, it produces supraphysiological levels of cAMP. How the catalytic domain is transferred from the hydrophilic extracellular medium into the hydrophobic environment of the membrane and then to the cell cytoplasm remains an unsolved question. In this report, we have characterized the membrane-interacting properties of the CyaA catalytic domain. We showed that a protein covering the catalytic domain (AC384, encompassing residues 1-384 of CyaA) displayed no membrane association propensity. However, a longer polypeptide (AC489), encompassing residues 1-489 of CyaA, exhibited the intrinsic property to bind to membranes and to induce lipid bilayer destabilization. We further showed that deletion of residues 375-485 within CyaA totally abrogated the toxin's ability to increase intracellular cAMP in target cells. These results indicate that, whereas the calmodulin dependent enzymatic domain is restricted to the amino-terminal residues 1-384 of CyaA, the membrane-interacting, translocation-competent domain extends up to residue 489. This thus suggests an important role of the region adjacent to the catalytic domain of CyaA in promoting its interaction with and its translocation across the plasma membrane of target cells.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Adenylate Cyclase Toxin/metabolism , Bordetella pertussis/metabolism , Cell Membrane/microbiology , Whooping Cough/microbiology , Adenylate Cyclase Toxin/genetics , Bordetella pertussis/chemistry , Bordetella pertussis/genetics , Catalytic Domain , Cell Line , Humans , Protein Transport
8.
J Mol Biol ; 415(3): 584-99, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22119486

ABSTRACT

The accumulation of amyloid fibers due to protein misfolding is associated with numerous human diseases. For example, the formation of amyloid deposits in neurodegenerative pathologies is correlated with abnormal apoptosis. We report here the in vitro formation of various types of aggregates by Bcl-xL, a protein of the Bcl-2 family involved in the regulation of apoptosis. Bcl-xL forms aggregates in three states, micelles, native-like fibrils, and amyloid fibers, and their biophysical characterization has been performed in detail. Bcl-xL remains in its native state within micelles and native-like fibrils, and our results suggest that native-like fibrils are formed by the association of micelles. Formation of amyloid structures, that is, nonnative intermolecular ß-sheets, is favored by the proximity of proteins within fibrils at the expense of the Bcl-xL native structure. Finally, we provide evidence of a direct relationship between the amyloid character of the fibers and the tertiary-structure stability of the native Bcl-xL. The potential causality between the accumulation of Bcl-xL into amyloid deposits and abnormal apoptosis during neurodegenerative diseases is discussed.


Subject(s)
Amyloid/metabolism , bcl-X Protein/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Humans , Microscopy, Electron , Models, Molecular , Protein Conformation , Protein Denaturation , Protein Multimerization , Protein Stability , bcl-X Protein/chemistry
9.
Biophys J ; 99(11): 3744-53, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21112299

ABSTRACT

The adenylate cyclase toxin (CyaA) is one of the major virulence factors of Bordetella pertussis, the causative agent of whooping cough. Its C-terminal region, the receptor-binding domain (RD), contains ∼40 calcium-binding Repeat in ToXin (RTX) motifs, which are characteristic of many virulence factors of pathogenic bacteria. We previously showed that RD is intrinsically disordered in the absence of calcium and acquires its functional three-dimensional structure upon calcium binding. To gain further insight into the physicochemical properties of RD, we characterized its calcium-induced conformational and stability changes by combining spectroscopic approaches. We show that RD, in the absence of calcium, adopts premolten globule conformations, due in part to the strong internal electrostatic repulsions between the negative charges of the aspartate-rich polypeptide sequence. Accordingly, sodium is able to screen these electrostatic repulsions, allowing a partial compaction of the polypeptide, whereas calcium triggers a strong compaction as well as the acquisition of secondary and tertiary structures in a highly cooperative manner. The differential sensitivity of the calcium-loaded state to guanidinium- and urea-induced denaturations provides further evidence that electrostatic interactions play a critical role in the folding and stability of RD. These results provide new insights into the folding/function relationship of the RTX motifs.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Adenylate Cyclase Toxin/metabolism , Bordetella pertussis/metabolism , Calcium/pharmacology , Protein Folding/drug effects , Circular Dichroism , Fluorescence , Guanidine/pharmacology , Models, Molecular , Protein Stability/drug effects , Protein Structure, Tertiary , Sodium Chloride/pharmacology , Spectroscopy, Fourier Transform Infrared , Tryptophan/metabolism , Urea/pharmacology
10.
Proc Natl Acad Sci U S A ; 107(25): 11277-82, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20534570

ABSTRACT

Allostery plays a key role in the regulation of the activity and function of many biomolecules. And although many ligands act through allostery, no systematic use is made of it in drug design strategies. Here we describe a procedure for identifying the regions of a protein that can be used to control its activity through allostery. This procedure is based on the construction of a plausible conformational path, which describes protein transition between known active and inactive conformations. The path is calculated by using a framework approach that steers and markedly improves the conjugate peak refinement method. The evolution of conformations along this path was used to identify a putative allosteric site that could regulate activation of Bacillus anthracis adenylyl cyclase toxin (EF) by calmodulin. Conformations of the allosteric site at different steps along the path from the inactive (free) to the active (bound to calmodulin) forms of EF were used to perform virtual screenings and propose candidate EF inhibitors. Several candidates then proved to inhibit calmodulin-induced activation in an in vitro assay. The most potent compound fully inhibited EF at a concentration of 10 microM. The compounds also inhibited the related adenylyl cyclase toxin from Bordetella pertussis (CyaA). The specific homology between the putative allosteric sites in both toxins supports that these pockets are the actual binding sites of the selected inhibitors.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Calmodulin/chemistry , Allosteric Site , Bacterial Toxins/antagonists & inhibitors , Bordetella pertussis/metabolism , Chemistry, Pharmaceutical/methods , Computational Biology/methods , Databases, Protein , Drug Design , Humans , Models, Molecular , Protein Conformation , Protein Structure, Tertiary
11.
J Mol Biol ; 397(2): 534-49, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20096704

ABSTRACT

Repeat in toxin (RTX) motifs are nonapeptide sequences found among numerous virulence factors of Gram-negative bacteria. In the presence of calcium, these RTX motifs are able to fold into an idiosyncratic structure called the parallel beta-roll. The adenylate cyclase toxin (CyaA) produced by Bordetella pertussis, the causative agent of whooping cough, is one of the best-characterized RTX cytolysins. CyaA contains a C-terminal receptor domain (RD) that mediates toxin binding to the eukaryotic cell receptor. The receptor-binding domain is composed of about forty RTX motifs organized in five successive blocks (I to V). The RTX blocks are separated by non-RTX flanking regions of variable lengths. It has been shown that block V with its N- and C-terminal flanking regions constitutes an autonomous subdomain required for the toxicity of CyaA. Here, we investigated the calcium-induced biophysical changes of this subdomain to identify the respective contributions of the flanking regions to the folding process of the RTX motifs. We showed that the RTX polypeptides, in the absence of calcium, exhibited the hallmarks of intrinsically disordered proteins and that the C-terminal flanking region was critical for the calcium-dependent folding of the RTX polypeptides, while the N-terminal flanking region was not involved. Furthermore, the secondary and tertiary structures were acquired concomitantly upon cooperative binding of several calcium ions. This suggests that the RTX polypeptide folding is a two-state reaction, from a calcium-free unfolded state to a folded and compact conformation, in which the calcium-bound RTX motifs adopt a beta-roll structure. The relevance of these results to the toxin physiology, in particular to its secretion, is discussed.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Adenylate Cyclase Toxin/metabolism , Bordetella pertussis/enzymology , Calcium/metabolism , Amino Acid Motifs , Bordetella pertussis/chemistry , Circular Dichroism , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , Spectrum Analysis
12.
Biochemistry ; 49(2): 318-28, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20000337

ABSTRACT

Bordetella pertussis, the causative agent of whooping cough, secretes among various toxins an adenylate cyclase (CyaA) that displays a unique mechanism of cell invasion, which involves a direct translocation of its N-terminal catalytic domain (AC, 400 residues) across the plasma membrane of the eukaryotic targeted cells. Once into the cytosol, AC is activated by endogenous calmodulin and produces toxic amounts of cAMP. The structure of AC in complex with the C-terminal part of calmodulin has recently been determined. However, as the structure of the catalytic domain in the absence of calmodulin is still lacking, the molecular basis of AC activation by calmodulin remains largely unknown. To characterize this activation mechanism, we investigated here the biophysical properties of the isolated catalytic domain in solution with or without calmodulin. We found that calmodulin triggered only minor modifications of the protein secondary and tertiary structure but had a pronounced effect on the hydrodynamic properties of AC. Indeed, while the isolated catalytic domain was spherical and hydrated, it underwent a significant elongation as well as compaction and dehydration upon calmodulin interaction. On the basis of these data, we propose a model for the structural transition between the calmodulin-free and calmodulin-bound AC.


Subject(s)
Adenylate Cyclase Toxin/chemistry , Adenylate Cyclase Toxin/metabolism , Bordetella pertussis/enzymology , Calmodulin/pharmacology , Adenylate Cyclase Toxin/genetics , Adenylate Cyclase Toxin/isolation & purification , Bordetella pertussis/genetics , Catalytic Domain , Chromatography, Gel , Chromatography, Ion Exchange , Circular Dichroism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Protein Conformation , Spectrophotometry
13.
Microbiology (Reading) ; 155(Pt 3): 944-956, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19246765

ABSTRACT

Characterization of 'unknown' proteins is one of the challenges of the post-genomic era. Here, we report a study of Bacillus subtilis YdiB, which belongs to an uncharted class of bacterial P-loop ATPases. Precise deletion of the ydiB gene yielded a mutant with much reduced growth rate compared to the wild-type strain. In vitro, purified YdiB was in equilibrium among different forms, monomers, dimers and oligomers, and this equilibrium was strongly affected by salts; high concentrations of NaCl favoured the monomeric over the oligomeric form of the enzyme. Interestingly, the ATPase activity of the monomer was about three times higher than that of the oligomer, and the monomer showed a K(m) of about 60 microM for ATP and a V(max) of about 10 nmol min(-1) (mg protein)(-1) (k(cat) approximately 10 h(-1)). This low ATPase activity was shown to be specific to YdiB because mutation of an invariant lysine residue in the P-loop motif (K41A) strongly attenuated this rate. This mutant was unable to restore a normal growth phenotype when introduced into a conditional knockout strain for ydiB, showing that the ATPase activity of YdiB is required for the in vivo function of the protein. Oligomerization was also observed with the purified YjeE from Escherichia coli, a YdiB orthologue, suggesting that this property is shared by all members of this family of ATPases. Importantly, dimers of YdiB were also observed in a B. subtilis extract, or when stabilized by formaldehyde cross-linking for YjeE from E. coli, suggesting that oligomerization might regulate the function of this new class of proteins in vivo.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Protein Multimerization , Adenosine Triphosphatases/genetics , Bacillus subtilis/genetics , Bacillus subtilis/growth & development , Bacterial Proteins/genetics , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Gene Deletion , Gene Knockout Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...