Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(23): e2122386119, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35648835

ABSTRACT

Pneumococcal conjugate vaccines (PCVs) used in childhood vaccination programs have resulted in replacement of vaccine-type with nonvaccine-type pneumococci in carriage and invasive pneumococcal disease (IPD). A vaccine based on highly conserved and protective pneumococcal antigens is urgently needed. Here, we performed intranasal immunization of mice with pneumococcal membrane particles (MPs) to mimic natural nasopharyngeal immunization. MP immunization gave excellent serotype-independent protection against IPD that was antibody dependent but independent of the cytotoxin pneumolysin. Using Western blotting, immunoprecipitation, mass spectrometry, and different bacterial mutants, we identified the conserved lipoproteins MalX and PrsA as the main antigens responsible for cross-protection. Additionally, we found that omitting the variable surface protein and vaccine candidate PspA from MPs enhanced protective immune responses to the conserved proteins. Our findings suggest that MPs containing MalX and PrsA could serve as a platform for pneumococcal vaccine development targeting the elderly and immunocompromised.


Subject(s)
Bacterial Proteins , Lipoproteins , Membrane Proteins , Membrane Transport Proteins , Pneumococcal Infections , Pneumococcal Vaccines , Administration, Intranasal , Animals , Bacterial Proteins/immunology , Cell Membrane/immunology , Conserved Sequence , Cross Reactions , Humans , Immunization/methods , Lipoproteins/immunology , Membrane Proteins/immunology , Membrane Transport Proteins/immunology , Mice , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/immunology , Serogroup , Streptococcus pneumoniae/immunology
2.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34785593

ABSTRACT

Emerging antibiotic resistance demands identification of novel antibacterial compound classes. A bacterial whole-cell screen based on pneumococcal autolysin-mediated lysis induction was developed to identify potential bacterial cell wall synthesis inhibitors. A hit class comprising a 1-amino substituted tetrahydrocarbazole (THCz) scaffold, containing two essential amine groups, displayed bactericidal activity against a broad range of gram-positive and selected gram-negative pathogens in the low micromolar range. Mode of action studies revealed that THCz inhibit cell envelope synthesis by targeting undecaprenyl pyrophosphate-containing lipid intermediates and thus simultaneously inhibit peptidoglycan, teichoic acid, and polysaccharide capsule biosynthesis. Resistance did not readily develop in vitro, and the ease of synthesizing and modifying these small molecules, as compared to natural lipid II-binding antibiotics, makes THCz promising scaffolds for development of cell wall-targeting antimicrobials.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Cell Wall/chemistry , Cell Wall/drug effects , Lipids/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial/drug effects , Microbial Sensitivity Tests , N-Acetylmuramoyl-L-alanine Amidase , Peptidoglycan/biosynthesis , Polyisoprenyl Phosphates , Streptococcus pneumoniae/drug effects , Teichoic Acids/chemistry , Uridine Diphosphate N-Acetylmuramic Acid/analogs & derivatives
3.
mBio ; 11(6)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323510

ABSTRACT

Streptococcus pneumoniae, a major cause of pneumonia, sepsis, and meningitis worldwide, has the nasopharynges of small children as its main ecological niche. Depletion of pneumococci from this niche would reduce the disease burden and could be achieved using small molecules with narrow-spectrum antibacterial activity. We identified the alkylated dicyclohexyl carboxylic acid 2CCA-1 as a potent inducer of autolysin-mediated lysis of S. pneumoniae, while having low activity against Staphylococcus aureus 2CCA-1-resistant strains were found to have inactivating mutations in fakB3, known to be required for uptake of host polyunsaturated fatty acids, as well as through inactivation of the transcriptional regulator gene fabT, vital for endogenous, de novo fatty acid synthesis regulation. Structure activity relationship exploration revealed that, besides the central dicyclohexyl group, the fatty acid-like structural features of 2CCA-1 were essential for its activity. The lysis-inducing activity of 2CCA-1 was considerably more potent than that of free fatty acids and required growing bacteria, suggesting that 2CCA-1 needs to be metabolized to exert its antimicrobial activity. Total lipid analysis of 2CCA-1 treated bacteria identified unique masses that were modeled to 2CCA-1 containing lysophosphatidic and phosphatidic acid in wild-type but not in fakB3 mutant bacteria. This suggests that 2CCA-1 is metabolized as a fatty acid via FakB3 and utilized as a phospholipid building block, leading to accumulation of toxic phospholipid species. Analysis of FabT-mediated fakB3 expression elucidates how the pneumococcus could ensure membrane homeostasis and concurrent economic use of host-derived fatty acids.IMPORTANCE Fatty acid biosynthesis is an attractive antibiotic target, as it affects the supply of membrane phospholipid building blocks. In Streptococcus pneumoniae, it is not sufficient to target only the endogenous fatty acid synthesis machinery, as uptake of host fatty acids may bypass this inhibition. Here, we describe a small-molecule compound, 2CCA-1, with potent bactericidal activity that upon interactions with the fatty acid binding protein FakB3, which is present in a limited number of Gram-positive species, becomes metabolized and incorporated as a toxic phospholipid species. Resistance to 2CCA-1 developed specifically in fakB3 and the regulatory gene fabT These mutants reveal a regulatory connection between the extracellular polyunsaturated fatty acid metabolism and endogenous fatty acid synthesis in S. pneumoniae, which could ensure balance between efficient scavenging of host polyunsaturated fatty acids and membrane homeostasis. The data might be useful in the identification of narrow-spectrum treatment strategies to selectively target members of the Lactobacillales such as S. pneumoniae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carboxylic Acids/pharmacology , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids/pharmacology , Streptococcus pneumoniae/drug effects , Anti-Bacterial Agents/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriolysis/drug effects , Biosynthetic Pathways/drug effects , Carboxylic Acids/chemistry , Drug Resistance, Bacterial , Fatty Acids/chemistry , Gene Expression Regulation, Bacterial , Mutation , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism
4.
Sci Rep ; 10(1): 3670, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111864

ABSTRACT

Handling of submicron-sized objects is important in many biochemical and biomedical applications, but few methods today can precisely manipulate this range of particles. We present gradient acoustic focusing that enables flow-through particle separation of submicron particles and cells and we apply it for separation of bacteria from blood lysate to facilitate their detection in whole blood for improved diagnostics. To control suspended objects below the classical 2µm size limit for acoustic focusing, we introduce a co-flowing acoustic impedance gradient to generate a stabilizing acoustic volume force that supresses acoustic streaming. The method is validated theoretically and experimentally using polystyrene particles, Staphylococcus aureus, Streptococcus pneumoniae and Escherichia coli. The applicability of the method is demonstrated by the separation of bacteria from selectively chemically lysed blood. Combined with downstream operations, this new approach opens up for novel methods for sepsis diagnostics.


Subject(s)
Escherichia coli/cytology , Microfluidic Analytical Techniques , Polystyrenes/chemistry , Staphylococcus aureus/cytology , Streptococcus pneumoniae/cytology , Particle Size , Sound
5.
Elife ; 72018 01 13.
Article in English | MEDLINE | ID: mdl-29331015

ABSTRACT

Blood is arguably the most important bodily fluid and its analysis provides crucial health status information. A first routine measure to narrow down diagnosis in clinical practice is the differential blood count, determining the frequency of all major blood cells. What is lacking to advance initial blood diagnostics is an unbiased and quick functional assessment of blood that can narrow down the diagnosis and generate specific hypotheses. To address this need, we introduce the continuous, cell-by-cell morpho-rheological (MORE) analysis of diluted whole blood, without labeling, enrichment or separation, at rates of 1000 cells/sec. In a drop of blood we can identify all major blood cells and characterize their pathological changes in several disease conditions in vitro and in patient samples. This approach takes previous results of mechanical studies on specifically isolated blood cells to the level of application directly in blood and adds a functional dimension to conventional blood analysis.


Subject(s)
Blood Cells/cytology , Blood Cells/physiology , Cytological Techniques/methods , Diagnostic Tests, Routine/methods , Single-Cell Analysis/methods , Humans
6.
Cell Rep ; 20(1): 251-263, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28683318

ABSTRACT

Genome delivery to the proper cellular compartment for transcription and replication is a primary goal of viruses. However, methods for analyzing viral genome localization and differentiating genomes with high identity are lacking, making it difficult to investigate entry-related processes and co-examine heterogeneous RNA viral populations. Here, we present an RNA labeling approach for single-cell analysis of RNA viral replication and co-infection dynamics in situ, which uses the versatility of padlock probes. We applied this method to identify influenza A virus (IAV) infections in cells and lung tissue with single-nucleotide specificity and to classify entry and replication stages by gene segment localization. Extending the classification strategy to co-infections of IAVs with single-nucleotide variations, we found that the dependence on intracellular trafficking places a time restriction on secondary co-infections necessary for genome reassortment. Altogether, these data demonstrate how RNA viral genome labeling can help dissect entry and co-infections.


Subject(s)
Genome, Viral , Influenza A virus/physiology , RNA, Viral/metabolism , Single-Cell Analysis/methods , Staining and Labeling/methods , Virus Replication , Animals , Dogs , Epithelial Cells/virology , HEK293 Cells , Humans , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Proteins/metabolism
7.
J Exp Med ; 214(6): 1619-1630, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28515075

ABSTRACT

Streptococcus pneumoniae is the main cause of bacterial meningitis, a life-threating disease with a high case fatality rate despite treatment with antibiotics. Pneumococci cause meningitis by invading the blood and penetrating the blood-brain barrier (BBB). Using stimulated emission depletion (STED) super-resolution microscopy of brain biopsies from patients who died of pneumococcal meningitis, we observe that pneumococci colocalize with the two BBB endothelial receptors: polymeric immunoglobulin receptor (pIgR) and platelet endothelial cell adhesion molecule (PECAM-1). We show that the major adhesin of the pneumococcal pilus-1, RrgA, binds both receptors, whereas the choline binding protein PspC binds, but to a lower extent, only pIgR. Using a bacteremia-derived meningitis model and mutant mice, as well as antibodies against the two receptors, we prevent pneumococcal entry into the brain and meningitis development. By adding antibodies to antibiotic (ceftriaxone)-treated mice, we further reduce the bacterial burden in the brain. Our data suggest that inhibition of pIgR and PECAM-1 has the potential to prevent pneumococcal meningitis.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Brain/microbiology , Fimbriae Proteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Polymeric Immunoglobulin/metabolism , Streptococcus pneumoniae/pathogenicity , Virulence Factors/metabolism , Animals , Antibodies, Bacterial/pharmacology , Antibodies, Bacterial/therapeutic use , Biopsy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/pathology , Brain/drug effects , Brain/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Humans , Meningitis, Bacterial/drug therapy , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Mice, Inbred C57BL , Microscopy , Protein Binding/drug effects , Streptococcus pneumoniae/isolation & purification
8.
mBio ; 7(5)2016 10 25.
Article in English | MEDLINE | ID: mdl-27795406

ABSTRACT

Understanding the interplay between antibiotic resistance and bacterial fitness and virulence is essential to guide individual treatments and improve global antibiotic policies. A paradigmatic example of a resistance mechanism is the intrinsic inducible chromosomal ß-lactamase AmpC from multiple Gram-negative bacteria, including Pseudomonas aeruginosa, a major nosocomial pathogen. The regulation of ampC expression is intimately linked to peptidoglycan recycling, and AmpC-mediated ß-lactam resistance is frequently mediated by inactivating mutations in ampD, encoding an N-acetyl-anhydromuramyl-l-alanine amidase, affecting the levels of ampC-activating muropeptides. Here we dissect the impact of the multiple pathways causing AmpC hyperproduction on P. aeruginosa fitness and virulence. Through a detailed analysis, we demonstrate that the lack of all three P. aeruginosa AmpD amidases causes a dramatic effect in fitness and pathogenicity, severely compromising growth rates, motility, and cytotoxicity; the latter effect is likely achieved by repressing key virulence factors, such as protease LasA, phospholipase C, or type III secretion system components. We also show that ampC overexpression is required but not sufficient to confer the growth-motility-cytotoxicity impaired phenotype and that alternative pathways leading to similar levels of ampC hyperexpression and resistance, such as those involving PBP4, had no fitness-virulence cost. Further analysis indicated that fitness-virulence impairment is caused by overexpressing ampC in the absence of cell wall recycling, as reproduced by expressing ampC from a plasmid in an AmpG (muropeptide permease)-deficient background. Thus, our findings represent a major step in the understanding of ß-lactam resistance biology and its interplay with fitness and pathogenesis. IMPORTANCE: Understanding the impact of antibiotic resistance mechanisms on bacterial pathogenesis is critical to curb the spread of antibiotic resistance. A particularly noteworthy antibiotic resistance mechanism is the ß-lactamase AmpC, produced by Pseudomonas aeruginosa, a major pathogen causing hospital-acquired infections. The regulation of AmpC is linked to the cell wall recycling pathways, and frequently, resistance to ß-lactams is caused by mutation of several of the components of the cell wall recycling pathways such as AmpD. Here we dissect the impact of the pathways for AmpC hyperproduction on virulence, showing that the lack of all three P. aeruginosa AmpD amidases causes a major effect in fitness and pathogenicity, compromising growth, motility, and cytotoxicity. Further analysis indicated that fitness-virulence impairment is specifically caused by the hyperproduction of AmpC in the absence of cell wall recycling. Our work provides valuable information for delineating future strategies for combating P. aeruginosa infections by simultaneously targeting virulence and antibiotic resistance.


Subject(s)
Bacterial Proteins/biosynthesis , Cell Wall/metabolism , Peptidoglycan/metabolism , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/physiology , beta-Lactamases/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Deletion , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Pseudomonas aeruginosa/growth & development , Virulence , beta-Lactamases/genetics
9.
Mol Microbiol ; 101(6): 954-67, 2016 09.
Article in English | MEDLINE | ID: mdl-27273793

ABSTRACT

The pneumococcal autolysin LytA is a key virulence factor involved in several important functions including DNA competence, immune evasion and biofilm formation. Here, we present the 1.05 Å crystal structure of the catalytic domain of LytA in complex with a synthetic cell-wall-based peptidoglycan (PG) ligand that occupies the entire Y-shaped substrate-binding crevice. As many as twenty-one amino-acid residues are engaged in ligand interactions with a majority of these interactions directed towards the glycan strand. All saccharides are intimately bound through hydrogen bond, van der Waals and CH-π interactions. Importantly, the structure of LytA is not altered upon ligand binding, whereas the bound ligand assumes a different conformation compared to the unbound NMR-based solution structure of the same PG-fragment. Mutational study reveals that several non-catalytic glycan-interacting residues, structurally conserved in other amidases from Gram-positive Firmicutes, are pivotal for enzymatic activity. The three-dimensional structure of the LytA/PG complex provides a novel structural basis for ligand restriction by the pneumococcal autolysin, revealing for the first time an importance of the multivalent binding to PG saccharides.


Subject(s)
N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Cell Wall/enzymology , Cell Wall/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Structure, Tertiary , Protein Subunits , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/metabolism , Structure-Activity Relationship
10.
mBio ; 5(1): e01120-13, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24520066

ABSTRACT

UNLABELLED: The cytosolic N-acetylmuramoyl-l-alanine amidase LytA protein of Streptococcus pneumoniae, which is released by bacterial lysis, associates with the cell wall via its choline-binding motif. During exponential growth, LytA accesses its peptidoglycan substrate to cause lysis only when nascent peptidoglycan synthesis is stalled by nutrient starvation or ß-lactam antibiotics. Here we present three-dimensional structures of LytA and establish the requirements for substrate binding and catalytic activity. The solution structure of the full-length LytA dimer reveals a peculiar fold, with the choline-binding domains forming a rigid V-shaped scaffold and the relatively more flexible amidase domains attached in a trans position. The 1.05-Å crystal structure of the amidase domain reveals a prominent Y-shaped binding crevice composed of three contiguous subregions, with a zinc-containing active site localized at the bottom of the branch point. Site-directed mutagenesis was employed to identify catalytic residues and to investigate the relative impact of potential substrate-interacting residues lining the binding crevice for the lytic activity of LytA. In vitro activity assays using defined muropeptide substrates reveal that LytA utilizes a large substrate recognition interface and requires large muropeptide substrates with several connected saccharides that interact with all subregions of the binding crevice for catalysis. We hypothesize that the substrate requirements restrict LytA to the sites on the cell wall where nascent peptidoglycan synthesis occurs. IMPORTANCE: Streptococcus pneumoniae is a human respiratory tract pathogen responsible for millions of deaths annually. Its major pneumococcal autolysin, LytA, is required for autolysis and fratricidal lysis and functions as a virulence factor that facilitates the spread of toxins and factors involved in immune evasion. LytA is also activated by penicillin and vancomycin and is responsible for the lysis induced by these antibiotics. The factors that regulate the lytic activity of LytA are unclear, but it was recently demonstrated that control is at the level of substrate recognition and that LytA required access to the nascent peptidoglycan. The present study was undertaken to structurally and functionally investigate LytA and its substrate-interacting interface and to determine the requirements for substrate recognition and catalysis. Our results reveal that the amidase domain comprises a complex substrate-binding crevice and needs to interact with a large-motif epitope of peptidoglycan for catalysis.


Subject(s)
N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptidoglycan/metabolism , Streptococcus pneumoniae/enzymology , Catalytic Domain , Crystallography, X-Ray , DNA Mutational Analysis , Magnetic Resonance Spectroscopy , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , N-Acetylmuramoyl-L-alanine Amidase/genetics , Protein Binding , Protein Conformation
11.
J Biol Chem ; 287(14): 11018-29, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334685

ABSTRACT

The pneumococcal autolysin LytA is a virulence factor involved in autolysis as well as in fratricidal- and penicillin-induced lysis. In this study, we used biochemical and molecular biological approaches to elucidate which factors control the cytoplasmic translocation and lytic activation of LytA. We show that LytA is mainly localized intracellularly, as only a small fraction was found attached to the extracellular cell wall. By manipulating the extracellular concentration of LytA, we found that the cells were protected from lysis during exponential growth, but not in the stationary phase, and that a defined threshold concentration of extracellular LytA dictates the onset of autolysis. Stalling growth through nutrient depletion, or the specific arrest of cell wall synthesis, sensitized cells for LytA-mediated lysis. Inhibition of cell wall association via the choline binding domain of an exogenously added enzymatically inactive form of LytA revealed a potential substrate for the amidase domain within the cell wall where the formation of nascent peptidoglycan occurs.


Subject(s)
Bacteriolysis , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptidoglycan/biosynthesis , Peptidoglycan/metabolism , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/enzymology , Anti-Bacterial Agents/pharmacology , Bacteriolysis/drug effects , Cell Division/drug effects , Cell Proliferation/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Deoxycholic Acid/pharmacology , Detergents/pharmacology , Extracellular Space/drug effects , Extracellular Space/metabolism , N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/pharmacology , Protein Sorting Signals , Protein Transport/drug effects , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/metabolism
12.
Infect Immun ; 79(10): 4210-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21788380

ABSTRACT

Streptococcus pneumoniae is a leading cause of bacterial pneumonia, meningitis, and sepsis in children. Human immunity to pneumococcal infections has been assumed to depend on anticapsular antibodies. However, recent findings from murine models suggest that alternative mechanisms, dependent on T helper cells, are also involved. Although the immunological events in which T helper cells contribute to acquired immunity have been studied in mice, little is known about how these responses are generated in humans. Therefore, we examined bacterial and host factors involved in the induction of Th1 and Th17 responses, using a coculture model of human monocytes and CD4(+) T cells. We show that monocytes promote effector cytokine production by memory T helper cells, leading to a mixed Th1/Th17 (gamma interferon [IFN-γ]/interleukin-17 [IL-17]) profile. Both T helper cytokines were triggered by purified pneumococcal peptidoglycan; however, the balance between the two immune effector arms depended on bacterial viability. Accordingly, live pneumococci triggered a Th1-biased response via monocyte production of IL-12p40, whereas heat-killed pneumococci triggered a Th17 response through TLR2 signaling. An increased understanding of human T helper responses is essential for the development of novel pneumococcal vaccines designed to elicit cell-mediated immunity.


Subject(s)
Monocytes/immunology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/immunology , Th1 Cells/immunology , Th17 Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Coculture Techniques , Cytokines/metabolism , Humans , Interleukin-12 Subunit p40/metabolism , Interleukin-17/immunology , Peptidoglycan/immunology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Toll-Like Receptor 2/metabolism
13.
J Biol Chem ; 285(5): 3300-9, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-19940132

ABSTRACT

Most secretion pathways in bacteria and eukaryotic cells are challenged by the requirement for their substrate proteins to mature after they traverse a membrane barrier and enter a reactive oxidizing environment. For Gram-positive bacteria, the mechanisms that protect their exported proteins from misoxidation during their post-translocation maturation are poorly understood. To address this, we separated numerous bacterial species according to their tolerance for oxygen and divided their proteomes based on the predicted subcellular localization of their proteins. We then applied a previously established computational approach that utilizes cysteine incorporation patterns in proteins as an indicator of enzymatic systems that may exist in each species. The Sec-dependent exported proteins from aerobic Gram-positive Actinobacteria were found to encode cysteines in an even-biased pattern indicative of a functional disulfide bond formation system. In contrast, aerobic Gram-positive Firmicutes favor the exclusion of cysteines from both their cytoplasmic proteins and their substantially longer exported proteins. Supporting these findings, we show that Firmicutes, but not Actinobacteria, tolerate growth in reductant. We further demonstrate that the actinobacterium Corynebacterium glutamicum possesses disulfide-bonded proteins and two dimeric Dsb-like enzymes that can efficiently catalyze the formation of disulfide bonds. Our results suggest that cysteine exclusion is an important adaptive strategy against the challenges presented by oxidative environments.


Subject(s)
Cysteine/chemistry , Gram-Positive Bacteria/metabolism , Actinobacteria/metabolism , Bacterial Proteins/chemistry , Corynebacterium glutamicum/metabolism , Cytoplasm/metabolism , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Bacterial , Oxidation-Reduction , Oxygen/chemistry , Protein Folding , Protein Transport , Proteome
14.
Dev Comp Immunol ; 31(8): 790-804, 2007.
Article in English | MEDLINE | ID: mdl-17240448

ABSTRACT

Despite the ecological and evolutionary importance of echinoderms, very little is known about the immune mechanisms in this group especially regarding humoral immunity. In this paper, we screened for proteins putatively involved in immunity in the common European seastar Asterias rubens using a mass spectrometry-based proteomic approach. Two proteins showed striking sequence similarities with peptidoglycan recognition proteins (PGRPs). The two seastar proteins were identified as a single protein, termed PGRP-S1a, occurring in two forms in the coelomic plasma, one of 20kDa and another of 22kDa. We also cloned and sequenced a second member of the PGRP family, termed PGRP-S2a. It has a calculated molecular mass of 21.3kDa and is expressed in circulating phagocytes. Both the S1a-cDNA from coelomic epithelium RNA and the S2a-cDNA from phagocytes code for the amino acid residues necessary for peptidoglycan degradation. PGRP-S1a did not affect the phagocytic activity of seastar immune cells towards Micrococcus luteus but inhibited their production of reactive oxygen species (ROS). A recombinant, His-tagged, PGRP-S2a degrades peptidoglycan and increases the phagocytosis of M. luteus cells by seastar phagocytes.


Subject(s)
Amidohydrolases/genetics , Asterias/enzymology , Asterias/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amino Acid Sequence , Animals , Asterias/genetics , Base Sequence , Carrier Proteins/chemistry , Cloning, Molecular , Immunity, Innate , Micrococcus luteus , Molecular Sequence Data , Peptidoglycan/metabolism , Phagocytosis , Protein Binding , Reactive Oxygen Species/metabolism , Sequence Homology, Amino Acid
15.
Biochem Biophys Res Commun ; 350(4): 994-9, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17046713

ABSTRACT

The peptidoglycan recognition protein (PGRP) family is conserved from insects to mammals and is involved in immune regulation and bacterial clearance. They form at least three functional classes; receptors required for immune gene expression; amidases that degrade peptidoglycan and scavenge the tissues from immune-stimulating peptidoglycan; and as proteins with antibacterial activity. We here report that PGRP-SB1 is an N-acetylmuramoyl l-alanine amidase, which (in contrast to the previously described PGRP-amidases) shows antibacterial activity. PGRP-SB1 is highly active against peptidoglycans that have a diaminopimelic acid (DAP) residue in the cross-linking peptide, but lack activity to most lysine-containing peptidoglycans. The antibacterial activity is pronounced against Bacillus megaterium with an LD(50) of 1.5microg ml(-1). The bactericidal effect of PGRP-SB1 is dependent on its enzymatic activity, as the zinc co-factor is essential. The bactericidal mode of action is thus different from non-enzymatic vertebrate PGRPs that have been reported to be antibacterial.


Subject(s)
Bacillus megaterium/cytology , Bacillus megaterium/drug effects , Carrier Proteins/administration & dosage , Drosophila Proteins/administration & dosage , Anti-Bacterial Agents/administration & dosage , Cell Survival/drug effects , Dose-Response Relationship, Drug
16.
Proc Natl Acad Sci U S A ; 102(18): 6455-60, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15843462

ABSTRACT

Drosophila knockout mutants have placed peptidoglycan recognition proteins (PGRPs) in the two major pathways controlling immune gene expression. We now examine PGRP affinities for peptidoglycan. PGRP-SA and PGRP-LCx are bona fide pattern recognition receptors, and PGRP-SA, the peptidoglycan receptor of the Toll/Dif pathway, has selective affinity for different peptidoglycans. PGRP-LCx, the default peptidoglycan receptor of the Imd/Relish pathway, has strong affinity for all polymeric peptidoglycans tested and for monomeric peptidoglycan. PGRP-LCa does not have affinity for polymeric or monomeric peptidoglycan. Instead, PGRP-LCa can form heterodimers with LCx when the latter is bound to monomeric peptidoglycan. Hence, PGRP-LCa can be said to function as an adaptor, thus adding a new function to a member of the PGRP family.


Subject(s)
Bacteria/metabolism , Carrier Proteins/metabolism , Drosophila/immunology , Peptidoglycan/metabolism , Signal Transduction/immunology , Animals , Bacteria/immunology , Cell Line , Cell Wall/metabolism , Dimerization , Drosophila/metabolism , Drosophila/microbiology , Genetic Vectors/genetics , Immunohistochemistry , Ligands , Models, Biological , Organophosphorus Compounds , Sequence Analysis, Protein
17.
Immunity ; 20(5): 637-49, 2004 May.
Article in English | MEDLINE | ID: mdl-15142531

ABSTRACT

Insects depend solely upon innate immune responses to survive infection. These responses include the activation of extracellular protease cascades, leading to melanization and clotting, and intracellular signal transduction pathways inducing antimicrobial peptide gene expression. In Drosophila, the IMD pathway is required for antimicrobial gene expression in response to gram-negative bacteria. The exact molecular component(s) from these bacteria that activate the IMD pathway remain controversial. We found that highly purified LPS did not stimulate the IMD pathway. However, lipid A, the active portion of LPS in mammals, activated melanization in the silkworm Bombyx morii. On the other hand, the IMD pathway was remarkably sensitive to polymeric and monomeric gram-negative peptidoglycan. Recognition of peptidoglycan required the stem-peptide sequence specific to gram-negative peptidoglycan and the receptor PGRP-LC. Recognition of monomeric and polymeric peptidoglycan required different PGRP-LC splice isoforms, while lipid A recognition required an unidentified soluble factor in the hemolymph of Bombyx morii.


Subject(s)
Drosophila Proteins/immunology , Drosophila/immunology , Lipopolysaccharides/immunology , Peptidoglycan/immunology , Signal Transduction/immunology , Animals , Blotting, Northern , Bombyx/immunology , Carrier Proteins/immunology , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/immunology , Lipid A/immunology , Peptidoglycan/chemistry , Polymerase Chain Reaction , Protein Isoforms/immunology
18.
J Biol Chem ; 278(29): 26319-22, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12777387

ABSTRACT

The peptidoglycan recognition protein PGRP-LC is a major activator of the imd/Relish pathway in the Drosophila immune response. Three transcripts are generated by alternative splicing of the complex PGRP-LC gene. The encoded transmembrane proteins share an identical intracellular part, but each has a separate extracellular PGRP-domain: x, y, or a. Here we show that two of these isoforms play unique roles in the response to different microorganisms. Using RNA interference in Drosophila mbn-2 cells, we found that PGRP-LCx is the only isoform required to mediate signals from Gram-positive bacteria and purified bacterial peptidoglycan. By contrast, the recognition of Gram-negative bacteria and bacterial lipopolysaccharide requires both PGRP-LCa and LCx. The third isoform, LCy, is expressed at lower levels and may be partially redundant. Two additional PGRP domains in the gene cluster, z and w, are both included in a single transcript of a separate gene, PGRP-LF. Suppression of this transcript does not block the response to any of the microorganisms tested.


Subject(s)
Carrier Proteins/genetics , Drosophila Proteins/genetics , Drosophila/genetics , Genes, Insect , Multigene Family , Alternative Splicing , Animals , Cell Line , Drosophila/drug effects , Drosophila/immunology , Drosophila/microbiology , Genes, Insect/drug effects , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/pathogenicity , Lipopolysaccharides/pharmacology , Multigene Family/drug effects , Peptidoglycan/pharmacology , RNA Interference
19.
J Biol Chem ; 278(9): 7059-64, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12496260

ABSTRACT

Recent studies of peptidoglycan recognition protein (PGRP) have shown that 2 of the 13 Drosophila PGRP genes encode proteins that function as receptors mediating immune responses to bacteria. We show here that another member, PGRP-SC1B, has a totally different function because it has enzymatic activity and thereby can degrade peptidoglycan. A mass spectrometric analysis of the cleavage products demonstrates that the enzyme hydrolyzes the lactylamide bond between the glycan strand and the cross-linking peptides. This result assigns the protein as an N-acetylmuramoyl-l-alanine amidase (EC ), and the corresponding gene is thus the first of this class to be described from a eukaryotic organism. Mutant forms of PGRP-SC1B lacking a potential zinc ligand are enzymatically inactive but retain their peptidoglycan affinity. The immunostimulatory properties of PGRP-SC1B-degraded peptidoglycan are much reduced. This is in striking contrast to lysozyme-digested peptidoglycan, which retains most of its elicitor activity. This points toward a scavenger function for PGRP-SC1B. Furthermore, a sequence homology comparison with phage T7 lysozyme, also an N-acetylmuramoyl-l-alanine amidase, shows that as many as six of the Drosophila PGRPs could belong to this class of proteins.


Subject(s)
Carrier Proteins/physiology , Drosophila/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Binding Sites , Blotting, Northern , Carrier Proteins/metabolism , Cell Line , Chromatography, High Pressure Liquid , Cross-Linking Reagents/pharmacology , DNA, Complementary/metabolism , Hydrolysis , Insecta , Kinetics , Ligands , Mass Spectrometry , Muramidase/metabolism , Mutation , N-Acetylmuramoyl-L-alanine Amidase/chemistry , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptides/chemistry , Peptidoglycan/metabolism , Protein Binding , RNA/metabolism , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...