Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 18(4): e1010458, 2022 04.
Article in English | MEDLINE | ID: mdl-35395062

ABSTRACT

Two-component regulatory systems (TCS) are among the most widespread mechanisms that bacteria use to sense and respond to environmental changes. In the human pathogen Streptococcus pneumoniae, a total of 13 TCS have been identified and many of them have been linked to pathogenicity. Notably, TCS01 strongly contributes to pneumococcal virulence in several infection models. However, it remains one of the least studied TCS in pneumococci and its functional role is still unclear. In this study, we demonstrate that TCS01 cooperates with a BceAB-type ABC transporter to sense and induce resistance to structurally-unrelated antimicrobial peptides of bacterial origin that all target undecaprenyl-pyrophosphate or lipid II, which are essential precursors of cell wall biosynthesis. Even though tcs01 and bceAB genes do not locate in the same gene cluster, disruption of either of them equally sensitized the bacterium to the same set of antimicrobial peptides. We show that the key function of TCS01 is to upregulate the expression of the transporter, while the latter appears the main actor in resistance. Electrophoretic mobility shift assays further demonstrated that the response regulator of TCS01 binds to the promoter region of the bceAB genes, implying a direct control of these genes. The BceAB transporter was overexpressed and purified from E. coli. After reconstitution in liposomes, it displayed substantial ATPase and GTPase activities that were stimulated by antimicrobial peptides to which it confers resistance to, revealing new functional features of a BceAB-type transporter. Altogether, this inducible defense mechanism likely contributes to the survival of the opportunistic microorganism in the human host, in which competition among commensal microorganisms is a key determinant for effective host colonization and invasive path.


Subject(s)
Antimicrobial Peptides , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial , Streptococcus pneumoniae , Antimicrobial Peptides/pharmacology , Bacteria/metabolism , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Escherichia coli/metabolism , Humans , Membrane Transport Proteins/metabolism , Peptides/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism
2.
Nucleic Acids Res ; 49(20): 11476-11490, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34718721

ABSTRACT

Pseudomonas aeruginosa possesses one of the most complex bacterial regulatory networks, which largely contributes to its success as a pathogen. However, most of its transcription factors (TFs) are still uncharacterized and the potential intra-species variability in regulatory networks has been mostly ignored so far. Here, we used DAP-seq to map the genome-wide binding sites of all 55 DNA-binding two-component systems (TCSs) response regulators (RRs) across the three major P. aeruginosa lineages. The resulting networks encompass about 40% of all genes in each strain and contain numerous new regulatory interactions across most major physiological processes. Strikingly, about half of the detected targets are specific to only one or two strains, revealing a previously unknown large functional diversity of TFs within a single species. Three main mechanisms were found to drive this diversity, including differences in accessory genome content, as exemplified by the strain-specific plasmid in IHMA87 outlier strain which harbors numerous binding sites of conserved chromosomally-encoded RRs. Additionally, most RRs display potential auto-regulation or RR-RR cross-regulation, bringing to light the vast complexity of this network. Overall, we provide the first complete delineation of the TCSs regulatory network in P. aeruginosa that will represent an important resource for future studies on this pathogen.


Subject(s)
Bacterial Proteins/genetics , Gene Regulatory Networks , Pseudomonas aeruginosa/genetics , Transcription Factors/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Pseudomonas aeruginosa/metabolism , Transcription Factors/metabolism
3.
Int J Mol Sci ; 21(12)2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32575393

ABSTRACT

Synergism between enzymes is of crucial importance in cell metabolism. This synergism occurs often through a spatial organisation favouring proximity and substrate channelling. In this context, we developed a strategy for evaluating the impact of the geometry between two enzymes involved in nature in the recycling of the carbon derived from plant cell wall polymers. By using an innovative covalent association process using two protein fragments, Jo and In, we produced two bi-modular chimeric complexes connecting a xylanase and a xylosidase, involved in the deconstruction of xylose-based plant cell wall polymer. We first show that the intrinsic activity of the individual enzymes was preserved. Small Angle X-rays Scattering (SAXS) analysis of the complexes highlighted two different spatial organisations in solution, affecting both the distance between the enzymes (53 Å and 28 Å) and the distance between the catalytic pockets (94 Å and 75 Å). Reducing sugar and HPAEC-PAD analysis revealed different behaviour regarding the hydrolysis of Beechwood xylan. After 24 h of hydrolysis, one complex was able to release a higher amount of reducing sugar compare to the free enzymes (i.e., 15,640 and 14,549 µM of equivalent xylose, respectively). However, more interestingly, the two complexes were able to release variable percentages of xylooligosaccharides compared to the free enzymes. The structure of the complexes revealed some putative steric hindrance, which impacted both enzymatic efficiency and the product profile. This report shows that controlling the spatial geometry between two enzymes would help to better investigate synergism effect within complex multi-enzymatic machinery and control the final product.


Subject(s)
Glycoside Hydrolases/chemistry , Plants/enzymology , Recombinant Fusion Proteins/metabolism , Xylose/chemistry , Biomass , Carbon Cycle , Glycoside Hydrolases/metabolism , Hydrolysis , Oligosaccharides/chemistry , Plant Proteins/chemistry , Plant Proteins/metabolism , Protein Domains , Protein Engineering , Scattering, Small Angle , X-Ray Diffraction , Xylosidases/chemistry , Xylosidases/metabolism
4.
J Biol Chem ; 292(6): 2217-2225, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28011643

ABSTRACT

Bacterial pathogens recruit circulating proteins to their own surfaces, co-opting the host protein functions as a mechanism of virulence. Particular attention has focused on the binding of plasminogen (Plg) to bacterial surfaces, as it has been shown that this interaction contributes to bacterial adhesion to host cells, invasion of host tissues, and evasion of the immune system. Several bacterial proteins are known to serve as receptors for Plg including glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a cytoplasmic enzyme that appears on the cell surface in this moonlighting role. Although Plg typically binds to these receptors via several lysine-binding domains, the specific interactions that occur have not been documented in all cases. However, identification of the relevant residues could help define strategies for mitigating the virulence of important human pathogens, such as Streptococcus pneumoniae (Sp). To shed light on this question, we have described a combination of peptide-spot array screening, competition and SPR assays, high-resolution crystallography, and mutational analyses to characterize the interaction between SpGAPDH and Plg. We identified three SpGAPDH lysine residues that were instrumental in defining the kinetic and thermodynamic parameters of the interaction. Altogether, the integration of the data presented in this work allows us to propose a structural model for the molecular interaction of the SpGAPDH-Plg complex.


Subject(s)
Plasminogen/metabolism , Streptococcus pneumoniae/pathogenicity , Amino Acid Sequence , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Kinetics , Protein Binding , Protein Conformation , Surface Plasmon Resonance , Thermodynamics
5.
Mol Microbiol ; 106(5): 832-846, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28960579

ABSTRACT

The peptidoglycan is a rigid matrix required to resist turgor pressure and to maintain the cellular shape. It is formed by linear glycan chains composed of N-acetylmuramic acid-(ß-1,4)-N-acetylglucosamine (MurNAc-GlcNAc) disaccharides associated through cross-linked peptide stems. The peptidoglycan is continually remodelled by synthetic and hydrolytic enzymes and by chemical modifications, including O-acetylation of MurNAc residues that occurs in most Gram-positive and Gram-negative bacteria. This modification is a powerful strategy developed by pathogens to resist to lysozyme degradation and thus to escape from the host innate immune system but little is known about its physiological function. In this study, we have investigated to what extend peptidoglycan O-acetylation is involved in cell wall biosynthesis and cell division of Streptococcus pneumoniae. We show that O-acetylation driven by Adr protects the peptidoglycan of dividing cells from cleavage by the major autolysin LytA and occurs at the septal site. Our results support a function for Adr in the formation of robust and mature MurNAc O-acetylated peptidoglycan and infer its role in the division of the pneumococcus.


Subject(s)
Cell Wall/metabolism , Peptidoglycan/metabolism , Streptococcus pneumoniae/metabolism , Acetylation , Acetylglucosamine/metabolism , Cell Division , Gram-Negative Bacteria/metabolism , Muramic Acids/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism
6.
BMC Microbiol ; 16(1): 239, 2016 Oct 12.
Article in English | MEDLINE | ID: mdl-27729019

ABSTRACT

BACKGROUND: Proteins from the LytR-CpsA-Psr family are found in almost all Gram-positive bacteria. Although LCP proteins have been studied in other pathogens, their functions in enterococci remain uncharacterized. The Psr protein from Enterococcus hirae, here renamed LcpA, previously associated with the regulation of the expression of the low-affinity PBP5 and ß-lactam resistance, has been characterized. RESULTS: LcpA protein of E. hirae ATCC 9790 has been produced and purified with and without its transmembrane helix. LcpA appears, through different methods, to be localized in the membrane, in agreement with in silico predictions. The interaction of LcpA with E. hirae cell wall indicates that LcpA binds enterococcal peptidoglycan, regardless of the presence of secondary cell wall polymers. Immunolocalization experiments showed that LcpA and PBP5 are localized at the division site of E. hirae. CONCLUSIONS: LcpA belongs to the LytR-CpsA-Psr family. Its topology, localization and binding to peptidoglycan support, together with previous observations on defective mutants, that LcpA plays a role related to the cell wall metabolism, probably acting as a phosphotransferase catalyzing the attachment of cell wall polymers to the peptidoglycan.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Enterococcus hirae/metabolism , Peptidoglycan/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cell Membrane/metabolism , Cell Wall/metabolism , Cloning, Molecular , DNA, Bacterial , Enterococcus hirae/cytology , Enterococcus hirae/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Genes, Bacterial , Phosphotransferases/metabolism , Protein Interaction Maps , Recombinant Proteins , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , beta-Lactam Resistance
7.
Amino Acids ; 48(9): 2205-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27209197

ABSTRACT

Snake venom metalloproteinases (SVMPs) participate in snakebite pathology such as hemorrhage, inflammation, and necrosis. They are synthesized as latent multi-domain precursors whose processing generates either catalytically active enzymes or free non-enzymatic domains. Recombinant expression of the precursor of P-III class SVMPs has failed due to the instability of the multi-domain polypeptide structure. Conversely, functional recombinant non-catalytic domains were obtained by prokaryotic expression systems. Here, we show for the first time the recombinant expression of the precursor of HF3, a highly hemorrhagic SVMP from Bothrops jararaca, and its non-catalytic domains, using an E. coli-based cell-free synthesis system. The precursor of HF3, composed of pro-, metalloproteinase-, disintegrin-like-, and cysteine-rich domains, and containing 38 Cys residues, was successfully expressed and purified. A protein composed of the disintegrin-like and cysteine-rich domains (DC protein) and the cysteine-rich domain alone (C protein) were expressed in vitro individually and purified. Both proteins were shown to be functional in assays monitoring the interaction with matrix proteins and in modulating the cleavage of fibrinogen by HF3. These data indicate that recombinant expression using prokaryotic-based cell-free synthesis emerges as an attractive alternative for the study of the structure and function of multi-domain proteins with a high content of Cys residues.


Subject(s)
Bothrops , Crotalid Venoms/biosynthesis , Crotalid Venoms/chemistry , Gene Expression , Metalloproteases/biosynthesis , Metalloproteases/chemistry , Animals , Cell-Free System/chemistry , Cell-Free System/metabolism , Crotalid Venoms/genetics , Metalloproteases/genetics , Protein Domains , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
8.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 6): 1373-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26057677

ABSTRACT

LytA is responsible for the autolysis of many Streptococcus species, including pathogens such as S. pneumoniae, S. pseudopneumoniae and S. mitis. However, how this major autolysin achieves full activity remains unknown. Here, the full-length structure of the S. pneumoniae LytA dimer is reported at 2.1 Å resolution. Each subunit has an N-terminal amidase domain and a C-terminal choline-binding domain consisting of six choline-binding repeats, which form five canonical and one single-layered choline-binding sites. Site-directed mutageneses combined with enzymatic activity assays indicate that dimerization and binding to choline are two independent requirements for the autolytic activity of LytA in vivo. Altogether, it is suggested that dimerization and full occupancy of all choline-binding sites through binding to choline-containing TA chains enable LytA to adopt a fully active conformation which allows the amidase domain to cleave two lactyl-amide bonds located about 103 Å apart on the peptidoglycan.


Subject(s)
Bacterial Proteins/chemistry , N-Acetylmuramoyl-L-alanine Amidase/chemistry , Streptococcus/chemistry , Protein Conformation
9.
Antimicrob Agents Chemother ; 59(1): 609-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385114

ABSTRACT

The human pathogen Streptococcus pneumoniae has been treated for decades with ß-lactam antibiotics. Its resistance is now widespread, mediated by the expression of mosaic variants of the target enzymes, the penicillin-binding proteins (PBPs). Understanding the mode of action of ß-lactams, not only in molecular detail but also in their physiological consequences, will be crucial to improving these drugs and any counterresistances. In this work, we investigate the piperacillin paradox, by which this ß-lactam selects primarily variants of PBP2b, whereas its most reactive target is PBP2x. These PBPs are both essential monofunctional transpeptidases involved in peptidoglycan assembly. PBP2x participates in septal synthesis, while PBP2b functions in peripheral elongation. The formation of the "lemon"-shaped cells induced by piperacillin treatment is consistent with the inhibition of PBP2x. Following the examination of treated and untreated cells by electron microscopy, the localization of the PBPs by epifluorescence microscopy, and the determination of the inhibition time course of the different PBPs, we propose a model of peptidoglycan assembly that accounts for the piperacillin paradox.


Subject(s)
Penicillin-Binding Proteins/antagonists & inhibitors , Piperacillin/pharmacology , Streptococcus pneumoniae/drug effects , beta-Lactam Resistance , Aminoacyltransferases/antagonists & inhibitors , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cytoskeletal Proteins/metabolism , Microbial Sensitivity Tests , Molecular Targeted Therapy , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/physiology , beta-Lactam Resistance/drug effects
10.
EMBO J ; 30(8): 1425-32, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21386816

ABSTRACT

Bacterial cell growth necessitates synthesis of peptidoglycan. Assembly of this major constituent of the bacterial cell wall is a multistep process starting in the cytoplasm and ending in the exterior cell surface. The intracellular part of the pathway results in the production of the membrane-anchored cell wall precursor, Lipid II. After synthesis this lipid intermediate is translocated across the cell membrane. The translocation (flipping) step of Lipid II was demonstrated to require a specific protein (flippase). Here, we show that the integral membrane protein FtsW, an essential protein of the bacterial division machinery, is a transporter of the lipid-linked peptidoglycan precursors across the cytoplasmic membrane. Using Escherichia coli membrane vesicles we found that transport of Lipid II requires the presence of FtsW, and purified FtsW induced the transbilayer movement of Lipid II in model membranes. This study provides the first biochemical evidence for the involvement of an essential protein in the transport of lipid-linked cell wall precursors across biogenic membranes.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cell Wall/metabolism , Escherichia coli/metabolism , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Peptidoglycan/metabolism , Biological Transport , Recombinant Proteins/metabolism
11.
J Mol Recognit ; 28(10): 635-44, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25960426

ABSTRACT

Antibody selectivity represents a major issue in the development of efficient immuno-therapeutics and detection assays. Its description requires a comparison of the affinities of the antibody for a significant number of antigen variants. In the case of peptide antigens, this task can now be addressed to a significant level of details owing to improvements in spot peptide array technologies. They allow the high-throughput mutational analysis of peptides with, depending on assay design, an evaluation of binding stabilities. Here, we examine the cross-reactive capacity of an antibody fragment using the PEPperCHIP(®) technology platform (PEPperPRINT GmbH, Heidelberg, Germany; >8800 peptides per microarray) combined with the surface plasmon resonance characterization (Biacore(®) technology; GE-Healthcare Biacore, Uppsala, Sweden) of a subset of interactions. ScFv1F4 recognizes the N-terminal end of oncoprotein E6 of human papilloma virus 16. The spot permutation analysis (i.e. each position substituted by all amino acids except cysteine) of the wild type decapeptide (sequence (6)TAMFQDPQER(15)) and of 15 variants thereof defined the optimal epitope and provided a ranking for variant recognition. The SPR affinity measurements mostly validated the ranking of complex stabilities deduced from array data and defined the sensitivity of spot fluorescence intensities, bringing further insight into the conditions for cross-reactivity. Our data demonstrate the importance of throughput and quantification in the assessment of antibody selectivity.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibody Specificity , Peptides/chemistry , Antibodies, Monoclonal/immunology , Cross Reactions/immunology , Epitopes/chemistry , Humans , Microarray Analysis/methods , Peptides/immunology , Surface Plasmon Resonance/methods
12.
Mol Microbiol ; 89(4): 690-701, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23796240

ABSTRACT

Proteins with LytM (Peptidase_M23) domains are broadly distributed in bacteria and have been implicated in a variety of important processes, including cell division and cell-shape determination. Most LytM-like proteins that have been structurally and/or biochemically characterized are metallo-endopeptidases that cleave cross-links in the peptidoglycan (PG) cell wall matrix. Notable exceptions are the Escherichia coli cell division proteins EnvC and NlpD. These LytM factors are not hydrolases themselves, but instead serve as activators that stimulate PG cleavage by target enzymes called amidases to promote cell separation. Here we report the structure of the LytM domain from EnvC, the first structure of a LytM factor implicated in the regulation of PG hydrolysis. As expected, the fold is highly similar to that of other LytM proteins. However, consistent with its role as a regulator, the active-site region is degenerate and lacks a catalytic metal ion. Importantly, genetic analysis indicates that residues in and around this degenerate active site are critical for amidase activation in vivo and in vitro. Thus, in the regulatory LytM factors, the apparent substrate binding pocket conserved in active metallo-endopeptidases has been adapted to control PG hydrolysis by another set of enzymes.


Subject(s)
Endopeptidases/chemistry , Endopeptidases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Amidohydrolases/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , DNA Mutational Analysis , Endopeptidases/genetics , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
13.
ACS Appl Mater Interfaces ; 16(29): 37390-37400, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39007843

ABSTRACT

Staphylococcus aureus (S. aureus) is an opportunistic infectious pathogen, which causes a high mortality rate during bloodstream infections. The early detection of virulent strains in patients' blood samples is of medical interest for rapid diagnosis. The main virulent factors identified in patient isolates include leukocidins that bind to specific membrane receptors and lyse immune cells and erythrocytes. Duffy antigen receptor for chemokines (DARC) on the surface of specific cells is a main target of leukocidins such as gamma-hemolysin AB (HlgAB) and leukocidin ED (LukED). Among them, HlgAB is a conserved and critical leukocidin that binds to DARC and forms pores on the cell membranes, leading to cell lysis. Current methods are based on ELISA or bacterial culture, which takes hours to days. For detecting HlgAB with faster response and higher sensitivity, we developed a biosensor that combines single-walled carbon nanotube field effect transistors (swCNT-FETs) with immobilized DARC receptors as biosensing elements. DARC was purified from a bacterial expression system and successfully reconstituted into nanodiscs that preserve binding capability for HlgAB. Dynamic light scattering (DLS) and scanning electron microscopy (SEM) showed an increase of the DARC-containing nanodisc size in the presence of HlgAB, indicating the formation of HlgAB prepore or pore complexes. We demonstrate that this sensor can specifically detect the leukocidins HlgA and HlgAB in a quantitative manner within the dynamic range of 1 fM to 100 pM with an LOD of 0.122 fM and an LOQ of 0.441 fM. The sensor was challenged with human serum spiked with HlgAB as simulated clinical samples. After dilution for decreasing nonspecific binding, it selectively detected the toxin with a similar detection range and apparent dissociation constant as in the buffer. This biosensor was demonstrated with remarkable sensitivity to detect HlgAB rapidly and has the potential as a tool for fundamental research and clinical applications, although this sensor cannot differentiate between HlgAB and LukED as both have the same receptor.


Subject(s)
Biosensing Techniques , Duffy Blood-Group System , Leukocidins , Staphylococcus aureus , Biosensing Techniques/methods , Duffy Blood-Group System/chemistry , Duffy Blood-Group System/metabolism , Leukocidins/chemistry , Leukocidins/metabolism , Humans , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/chemistry , Nanotubes, Carbon/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism
14.
J Biol Chem ; 287(51): 42620-33, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23086952

ABSTRACT

C1q, a key component of the classical complement pathway, is a major player in the response to microbial infection and has been shown to detect noxious altered-self substances such as apoptotic cells. In this work, using complementary experimental approaches, we identified the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a C1q partner when exposed at the surface of human pathogenic bacteria Streptococcus pneumoniae and human apoptotic cells. The membrane-associated GAPDH on HeLa cells bound the globular regions of C1q as demonstrated by pulldown and cell surface co-localization experiments. Pneumococcal strains deficient in surface-exposed GAPDH harbored a decreased level of C1q recognition when compared with the wild-type strains. Both recombinant human and pneumococcal GAPDHs interacted avidly with C1q as measured by surface plasmon resonance experiments (K(D) = 0.34-2.17 nm). In addition, GAPDH-C1q complexes were observed by transmission electron microscopy after cross-linking. The purified pneumococcal GAPDH protein activated C1 in an in vitro assay unlike the human form. Deposition of C1q, C3b, and C4b from human serum at the surface of pneumococcal cells was dependent on the presence of surface-exposed GAPDH. This ability of C1q to sense both human and bacterial GAPDHs sheds new insights on the role of this important defense collagen molecule in modulating the immune response.


Subject(s)
Cell Membrane/enzymology , Complement C1q/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Streptococcus pneumoniae/enzymology , Apoptosis , Cell Membrane Structures/metabolism , Complement Activation , Complement C1q/chemistry , Complement C1q/ultrastructure , Glyceraldehyde-3-Phosphate Dehydrogenases/ultrastructure , HeLa Cells , Humans , Immobilized Proteins/metabolism , Kinetics , Ligands , Mutation/genetics , Plasminogen/metabolism , Protein Binding , Protein Transport , Solubility , Solutions , Surface Plasmon Resonance
15.
Biochem J ; 441(3): 833-41, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22013894

ABSTRACT

RrgB is the major pilin which forms the pneumococcal pilus backbone. We report the high-resolution crystal structure of the full-length form of RrgB containing the IPQTG sorting motif. The RrgB fold is organized into four distinct domains, D1-D4, each of which is stabilized by an isopeptide bond. Crystal packing revealed a head-to-tail organization involving the interaction of the IPQTG motif into the D1 domain of two successive RrgB monomers. This fibrillar assembly, which fits into the electron microscopy density map of the native pilus, probably induces the formation of the D1 isopeptide bond as observed for the first time in the present study, since neither in published structures nor in soluble RrgB produced in Escherichia coli or in Streptococcus pneumoniae is the D1 bond present. Experiments performed in live bacteria confirmed that the intermolecular bond linking the RrgB subunits takes place between the IPQTG motif of one RrgB subunit and the Lys183 pilin motif residue of an adjacent RrgB subunit. In addition, we present data indicating that the D1 isopeptide bond is involved in RrgB stabilization. In conclusion, the crystal RrgB fibre is a compelling model for deciphering the molecular details required to generate the pneumococcal pilus.


Subject(s)
Fimbriae Proteins/chemistry , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Protein Multimerization , Streptococcus pneumoniae , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Crystallization , Crystallography, X-Ray , Fimbriae Proteins/genetics , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/genetics , Hydrogen Bonding , Mineral Fibers , Models, Biological , Models, Molecular , Molecular Conformation , Mutagenesis, Site-Directed , Protein Multimerization/genetics , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism
16.
Biochemistry ; 51(39): 7755-65, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22950454

ABSTRACT

All bacterial multidrug ABC transporters have been shown to work as either homodimers or heterodimers. Two possibly linked genes, patA and patB from Streptococcus pneumococcus, that encode half-ABC transporters have been shown previously to be involved in fluoroquinolone resistance. We showed that the ΔpatA, ΔpatB, or ΔpatA/ΔpatB mutant strains were more sensitive to unstructurally related compounds, i.e., ethidium bromide or fluoroquinolones, than the wild-type R6 strain. Inside-out vesicles prepared from Escherichia coli expressing PatA and/or PatB transported Hoechst 33342, a classical substrate of multidrug transporters, only when both PatA and PatB were coexpressed. This transport was inhibited either by orthovanadate or by reserpine, and mutation of the conserved Walker A lysine residue of either PatA or PatB fully abrogated Hoechst 33342 transport. PatA, PatB, and the PatA/PatB heterodimer were purified from detergent-solubilized E. coli membrane preparations. Protein dimers were identified in all cases, albeit in different proportions. In contrast to the PatA/PatB heterodimers, homodimers of PatA or PatB failed to show a vanadate-sensitive ATPase activity. Thus, PatA and PatB need to interact together to make a functional drug efflux transporter, and they work only as heterodimers.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Fluoroquinolones/pharmacology , Streptococcus pneumoniae/enzymology , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Anti-Bacterial Agents/metabolism , Fluoroquinolones/metabolism , Gene Deletion , Humans , Mutation , Pneumococcal Infections/drug therapy , Protein Multimerization , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Vanadates/metabolism
17.
J Biol Chem ; 286(50): 43004-12, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22013074

ABSTRACT

The ß-N-acetylhexosaminidase (EC 3.2.1.52) from glycoside hydrolase family 20 (GH20) catalyzes the hydrolysis of the ß-N-acetylglucosamine (NAG) group from the nonreducing end of various glycoconjugates. The putative surface-exposed N-acetylhexosaminidase StrH/Spr0057 from Streptococcus pneumoniae R6 was proved to contribute to the virulence by removal of ß(1,2)-linked NAG on host defense molecules following the cleavage of sialic acid and galactose by neuraminidase and ß-galactosidase, respectively. StrH is the only reported GH20 enzyme that contains a tandem repeat of two 53% sequence-identical catalytic domains (designated as GH20-1 and GH20-2, respectively). Here, we present the 2.1 Å crystal structure of the N-terminal domain of StrH (residues Glu-175 to Lys-642) complexed with NAG. It adopts an overall structure similar to other GH20 enzymes: a (ß/α)(8) TIM barrel with the active site residing at the center of the ß-barrel convex side. The kinetic investigation using 4-nitrophenyl N-acetyl-ß-d-glucosaminide as the substrate demonstrated that GH20-1 had an enzymatic activity (k(cat)/K(m)) of one-fourth compared with GH20-2. The lower activity of GH20-1 could be attributed to the substitution of active site Cys-469 of GH20-1 to the counterpart Tyr-903 of GH20-2. A complex model of NAGß(1,2)Man at the active site of GH20-1 combined with activity assays of the corresponding site-directed mutants characterized two key residues Trp-443 and Tyr-482 at subsite +1 of GH20-1 (Trp-876 and Tyr-914 of GH20-2) that might determine the ß(1,2) substrate specificity. Taken together, these findings shed light on the mechanism of catalytic specificity toward the ß(1,2)-linked ß-N-acetylglucosides.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Streptococcus pneumoniae/enzymology , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/metabolism , Bacterial Proteins/genetics , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Protein Structure, Secondary , Structure-Activity Relationship , Substrate Specificity , beta-N-Acetylhexosaminidases/genetics
18.
J Biol Chem ; 286(41): 35906-35914, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21865160

ABSTRACT

Spr1479 from Streptococcus pneumoniae R6 is a 33-kDa hypothetical protein of unknown function. Here, we determined the crystal structures of its apo-form at 1.90 Å and complex forms with inorganic phosphate and AMP at 2.30 and 2.20 Å, respectively. The core structure of Spr1479 adopts a four-layer αßßα-sandwich fold, with Fe(3+) and Mn(2+) coordinated at the binuclear center of the active site (similar to metallophosphoesterases). Enzymatic assays showed that, in addition to phosphodiesterase activity for bis(p-nitrophenyl) phosphate, Spr1479 has hydrolase activity for diadenosine polyphosphate (Ap(n)A) and ATP. Residues that coordinate with the two metals are indispensable for both activities. By contrast, the streptococcus-specific residue Trp-67, which binds to phosphate in the two complex structures, is indispensable for the ATP/Ap(n)A hydrolase activity only. Moreover, the AMP-binding pocket is conserved exclusively in all streptococci. Therefore, we named the protein SapH for streptococcal ATP/Ap(n)A and phosphodiester hydrolase.


Subject(s)
Acid Anhydride Hydrolases/chemistry , Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , Protein Folding , Streptococcus pneumoniae/enzymology , Apoenzymes/chemistry , Binding Sites , Crystallography, X-Ray , Protein Structure, Secondary
19.
Biochim Biophys Acta ; 1808(8): 2059-66, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21550334

ABSTRACT

Eukaryotic membrane protein expression is still a major bottleneck for structural studies. Production in E. coli often leads to low expression level and/or aggregated proteins. In the last decade, strategies relying on new fusion protein expression revealed promising results. Fusion with the amphipatic Mistic protein has been described to favor expression in E. coli membranes. Although, this approach has already been reported for a few membrane proteins, little is known about the activity of the fused proteins. We used this strategy and obtained high expression levels of a chloroplast ATP/ADP transporter from A. thaliana (NTT1) and characterized its transport properties. NTT1 fused to Mistic has a very low transport activity which can be recovered after in vivo Mistic fusion cleavage. Moreover, detailed molecular characterization of purified NTT1 mature form, NTT1 fused to Mistic or NTT1 cleaved-off from this fusion highlights the correct fold of the latter one. Therefore, considering the higher quantity of purified NTT1 mature form obtained via the Mistic fusion approach, this is a valuable strategy for obtaining quantities of pure and active proteins that are adequate for structural studies.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amphibian Proteins/biosynthesis , Arabidopsis Proteins/biosynthesis , Arabidopsis/metabolism , Cell Membrane/metabolism , Chloroplasts/metabolism , Cloning, Molecular/methods , Escherichia coli/metabolism , Nucleotide Transport Proteins/biosynthesis , Amphibian Proteins/genetics , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Biological Transport , Escherichia coli/genetics , Kinetics , Nucleotide Transport Proteins/chemistry , Nucleotide Transport Proteins/genetics , Peptide Hydrolases/metabolism , Protein Folding , Protein Structure, Quaternary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Structure-Activity Relationship
20.
Mol Microbiol ; 82(4): 904-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22023106

ABSTRACT

Zinc is an essential trace metal for living cells. The ABC transporter AdcABC was previously shown to be required for zinc uptake by Streptococcus pneumoniae. As we have recently described AdcAII as another zinc-binding lipoprotein, we have investigated the role of both AdcA and AdcAII in S. pneumoniae zinc metabolism. Deletion of either adcA or adcAII but not phtD reduced S. pneumoniae zinc uptake, with dual mutation of both adcA and adcAII further decreasing zinc import. For the Δ(adcA/adcAII) mutant, growth and intracellular concentrations of zinc were both greatly reduced in low zinc concentration. When grown in zinc-deficient medium, the Δ(adcA/adcAII) mutant displayed morphological defects related to aberrant septation. Growth and morphology of the Δ(adcA/adcAII) mutant recovered after supplementation with zinc. Dual deletion of adcA and adcAII strongly impaired growth of the pneumococcus in bronchoalveolar lavage fluid and human serum, and prevented S. pneumoniae establishing infection in mouse models of nasopharyngeal colonization, pneumonia and sepsis without altering the capsule. Taken together, our results show that AdcA and AdcAII play an essential and redundant role in specifically importing zinc into the pneumococcus, and that both zinc transporters are required for proper cell division and for S. pneumoniae survival during infection.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/pathogenicity , Virulence Factors/metabolism , Zinc/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Bacterial Proteins/genetics , Bronchoalveolar Lavage Fluid/microbiology , Cell Division , Culture Media/chemistry , Disease Models, Animal , Humans , Membrane Transport Proteins/genetics , Mice , Microbial Viability , Nasopharynx/microbiology , Pneumococcal Infections/microbiology , Sepsis/microbiology , Serum/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Virulence , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL