Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Microlife ; 5: uqae008, 2024.
Article in English | MEDLINE | ID: mdl-38665235

ABSTRACT

On September 20-22 September 2023, the international conference 'Microbiology 2023: from single cell to microbiome and host' convened microbiologists from across the globe for a very successful symposium, showcasing cutting-edge research in the field. Invited lecturers delivered exceptional presentations covering a wide range of topics, with a major emphasis on phages and microbiomes, on the relevant bacteria within these ecosystems, and their multifaceted roles in diverse environments. Discussions also spanned the intricate analysis of fundamental bacterial processes, such as cell division, stress resistance, and interactions with phages. Organized by four renowned Academies, the German Leopoldina, the French Académie des sciences, the Royal Society UK, and the Royal Swedish Academy of Sciences, the symposium provided a dynamic platform for experts to share insights and discoveries, leaving participants inspired and eager to integrate new knowledge into their respective projects. The success of Microbiology 2023 prompted the decision to host the next quadrennial academic meeting in Sweden. This choice underscores the commitment to fostering international collaboration and advancing the frontiers of microbiological knowledge. The transition to Sweden promises to be an exciting step in the ongoing global dialogue and specific collaborations on microbiology, a field where researchers will continue to push the boundaries of knowledge, understanding, and innovation not only in health and disease but also in ecology.

2.
J Immunol ; 212(1): 69-80, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37982695

ABSTRACT

Staphylococcus aureus is a significant cause of morbidity and mortality in pulmonary infections. Patients with autosomal-dominant hyper-IgE syndrome due to STAT3 deficiency are particularly susceptible to acquiring staphylococcal pneumonia associated with lung tissue destruction. Because macrophages are involved in both pathogen defense and inflammation, we investigated the impact of murine myeloid STAT3 deficiency on the macrophage phenotype in vitro and on pathogen clearance and inflammation during murine staphylococcal pneumonia. Murine bone marrow-derived macrophages (BMDM) from STAT3 LysMCre+ knockout or Cre- wild-type littermate controls were challenged with S. aureus, LPS, IL-4, or vehicle control in vitro. Pro- and anti-inflammatory responses as well as polarization and activation markers were analyzed. Mice were infected intratracheally with S. aureus, bronchoalveolar lavage and lungs were harvested, and immunohistofluorescence was performed on lung sections. S. aureus infection of STAT3-deficient BMDM led to an increased proinflammatory cytokine release and to enhanced upregulation of costimulatory MHC class II and CD86. Murine myeloid STAT3 deficiency did not affect pathogen clearance in vitro or in vivo. Matrix metalloproteinase 9 was upregulated in Staphylococcus-treated STAT3-deficient BMDM and in lung tissues of STAT3 knockout mice infected with S. aureus. Moreover, the expression of miR-155 was increased. The enhanced inflammatory responses and upregulation of matrix metalloproteinase 9 and miR-155 expression in murine STAT3-deficient as compared with wild-type macrophages during S. aureus infections may contribute to tissue damage as observed in STAT3-deficient patients during staphylococcal pneumonia.


Subject(s)
Job Syndrome , MicroRNAs , Pneumonia, Staphylococcal , Staphylococcal Infections , Humans , Mice , Animals , Staphylococcus aureus/metabolism , Macrophage Activation , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Inflammation/genetics , Mice, Knockout , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism
3.
Nat Commun ; 14(1): 3170, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37264013

ABSTRACT

The bacterial cell envelope consists of multiple layers, including the peptidoglycan cell wall, one or two membranes, and often an external layer composed of capsular polysaccharides (CPS) or other components. How the synthesis of all these layers is precisely coordinated remains unclear. Here, we identify a mechanism that coordinates the synthesis of CPS and peptidoglycan in Streptococcus pneumoniae. We show that CPS synthesis initiates from the division septum and propagates along the long axis of the cell, organized by the tyrosine kinase system CpsCD. CpsC and the rest of the CPS synthesis complex are recruited to the septum by proteins associated with the divisome (a complex involved in septal peptidoglycan synthesis) but not the elongasome (involved in peripheral peptidoglycan synthesis). Assembly of the CPS complex starts with CpsCD, then CpsA and CpsH, the glycosyltransferases, and finally CpsJ. Remarkably, targeting CpsC to the cell pole is sufficient to reposition CPS synthesis, leading to diplococci that lack CPS at the septum. We propose that septal CPS synthesis is important for chain formation and complement evasion, thereby promoting bacterial survival inside the host.


Subject(s)
Peptidoglycan , Streptococcus pneumoniae , Streptococcus pneumoniae/metabolism , Peptidoglycan/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Polysaccharides/metabolism , Cell Membrane/metabolism , Bacterial Capsules/metabolism , Polysaccharides, Bacterial/metabolism
4.
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
5.
Oncogene ; 41(15): 2173-2186, 2022 04.
Article in English | MEDLINE | ID: mdl-35197571

ABSTRACT

Increasing evidence highlights the role of bacteria in the physiopathology of cancer. However, the underlying molecular mechanisms remains poorly understood. Several cancer-associated bacteria have been shown to produce toxins which interfere with the host defense against tumorigenesis. Here, we show that lipopolysaccharides from Klebsiella pneumoniae and other Enterobacteria strongly inhibit the host tumor suppressor p53 pathway through a novel mechanism of p53 regulation. We found that lipopolysaccharides destabilize TP53 mRNA through a TLR4-NF-κB-mediated inhibition of the RNA-binding factor Wig-1. Importantly, we show that K. pneumoniae disables two major tumor barriers, oncogene-induced DNA damage signaling and senescence, by impairing p53 transcriptional activity upon DNA damage and oncogenic stress. Furthermore, we found an inverse correlation between the levels of TLR4 and p53 mutation in colorectal tumors. Hence, our data suggest that the repression of p53 by Enterobacteria via TLR4 alleviates the selection pressure for p53 oncogenic mutations and shapes the genomic evolution of cancer.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Enterobacteriaceae/genetics , Enterobacteriaceae/metabolism , Humans , Lipopolysaccharides/pharmacology , RNA, Messenger/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Suppressor Protein p53/metabolism
6.
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
7.
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
9.
Proc Natl Acad Sci U S A ; 117(49): 31386-31397, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229573

ABSTRACT

Influenza A virus (IAV)-related mortality is often due to secondary bacterial infections, primarily by pneumococci. Here, we study how IAV-modulated changes in the lungs affect bacterial replication in the lower respiratory tract (LRT). Bronchoalveolar lavages (BALs) from coinfected mice showed rapid bacterial proliferation 4 to 6 h after pneumococcal challenge. Metabolomic and quantitative proteomic analyses demonstrated capillary leakage with efflux of nutrients and antioxidants into the alveolar space. Pneumococcal adaptation to IAV-induced inflammation and redox imbalance increased the expression of the pneumococcal chaperone/protease HtrA. Presence of HtrA resulted in bacterial growth advantage in the IAV-infected LRT and protection from complement-mediated opsonophagocytosis due to capsular production. Absence of HtrA led to growth arrest in vitro that was partially restored by antioxidants. Pneumococcal ability to grow in the IAV-infected LRT depends on the nutrient-rich milieu with increased levels of antioxidants such as ascorbic acid and its ability to adapt to and cope with oxidative damage and immune clearance.


Subject(s)
Antioxidants/metabolism , Capillaries/pathology , Influenza, Human/microbiology , Pneumococcal Infections/microbiology , Respiratory System/microbiology , Respiratory System/virology , Streptococcus pneumoniae/growth & development , Animals , Bacterial Proteins/metabolism , Glucose/metabolism , Humans , Inflammation/complications , Inflammation/pathology , Mice, Inbred C57BL , Models, Biological , Molecular Chaperones/metabolism , Orthomyxoviridae Infections/microbiology , Oxidation-Reduction , Oxidative Stress , Phagocytosis , Respiratory System/pathology
10.
mBio ; 11(5)2020 09 29.
Article in English | MEDLINE | ID: mdl-32994320

ABSTRACT

Estrogen, a major female sex steroid hormone, has been shown to promote the selection of mucoid Pseudomonas aeruginosa in the airways of patients with chronic respiratory diseases, including cystic fibrosis. This results in long-term persistence, poorer clinical outcomes, and limited therapeutic options. In this study, we demonstrate that at physiological concentrations, sex steroids, including testosterone and estriol, induce membrane stress responses in P. aeruginosa This is characterized by increased virulence and consequent inflammation and release of proinflammatory outer membrane vesicles promoting in vivo persistence of the bacteria. The steroid-induced P. aeruginosa response correlates with the molecular polarity of the hormones and membrane fluidic properties of the bacteria. This novel mechanism of interaction between sex steroids and P. aeruginosa explicates the reported increased disease severity observed in females with cystic fibrosis and provides evidence for the therapeutic potential of the modulation of sex steroids to achieve better clinical outcomes in patients with hormone-responsive strains.IMPORTANCE Molecular mechanisms by which sex steroids interact with P. aeruginosa to modulate its virulence have yet to be reported. Our work provides the first characterization of a steroid-induced membrane stress mechanism promoting P. aeruginosa virulence, which includes the release of proinflammatory outer membrane vesicles, resulting in inflammation, host tissue damage, and reduced bacterial clearance. We further demonstrate that at nanomolar (physiological) concentrations, male and female sex steroids promote virulence in clinical strains of P. aeruginosa based on their dynamic membrane fluidic properties. This work provides, for the first-time, mechanistic insight to better understand and predict the P. aeruginosa related response to sex steroids and explain the interindividual patient variability observed in respiratory diseases such as cystic fibrosis that are complicated by gender differences and chronic P. aeruginosa infection.


Subject(s)
Bacterial Outer Membrane/drug effects , Cystic Fibrosis/complications , Gonadal Steroid Hormones/metabolism , Pseudomonas aeruginosa/pathogenicity , Stress, Physiological/drug effects , Alginates/metabolism , Animals , Biofilms/drug effects , Biofilms/growth & development , Cystic Fibrosis/microbiology , Estradiol/chemistry , Estradiol/pharmacology , Female , Gonadal Steroid Hormones/pharmacology , Humans , Inflammation , Male , Mice , Mice, Inbred BALB C , Pseudomonas aeruginosa/genetics , Sex Factors , Testosterone/chemistry , Testosterone/pharmacology , Virulence
11.
Mol Microbiol ; 113(3): 650-658, 2020 03.
Article in English | MEDLINE | ID: mdl-32185835

ABSTRACT

The Gram-positive bacterium Streptococcus pneumoniae, the pneumococcus, is an important commensal resident of the human nasopharynx. Carriage is usually asymptomatic, however, S. pneumoniae can become invasive and spread from the upper respiratory tract to the lungs causing pneumonia, and to other organs to cause severe diseases such as bacteremia and meningitis. Several pneumococcal proteins important for its disease-causing capability have been described and many are expressed on the bacterial surface. The surface located pneumococcal type-1 pilus has been associated with virulence and the inflammatory response, and it is present in 20%-30% of clinical isolates. Its tip protein RrgA has been shown to be a major adhesin to human cells and to promote invasion through the blood-brain barrier. In this review we discuss recent findings of the impact of RrgA on bacterial colonization of the upper respiratory tract and on pneumococcal virulence, and use epidemiological data and genome-mining to suggest trade-off mechanisms potentially explaining the rather low prevalence of pilus-1 expressing pneumococci in humans.


Subject(s)
Fimbriae Proteins/metabolism , Streptococcus pneumoniae/metabolism , Virulence Factors/metabolism , Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Fimbriae Proteins/physiology , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/physiology , Protein Binding , Streptococcus pneumoniae/pathogenicity , Virulence/genetics , Virulence Factors/physiology
12.
13.
Cell Microbiol ; 21(11): e13077, 2019 11.
Article in English | MEDLINE | ID: mdl-31251447

ABSTRACT

Streptococcus pneumoniae (the pneumococcus) is a human respiratory tract pathogen and a major cause of morbidity and mortality globally. Although the pneumococcus is a commensal bacterium that colonizes the nasopharynx, it also causes lethal diseases such as meningitis, sepsis, and pneumonia, especially in immunocompromised patients, in the elderly, and in young children. Due to the acquisition of antibiotic resistance and the emergence of nonvaccine serotypes, the pneumococcus has been classified as one of the priority pathogens for which new antibacterials are urgently required by the World Health Organization, 2017. Understanding molecular mechanisms behind the pathogenesis of pneumococcal infections and bacterial interactions within the host is crucial to developing novel therapeutics. Previously considered to be an extracellular pathogen, it is becoming evident that pneumococci may also occasionally establish intracellular niches within the body to escape immune surveillance and spread within the host. Intracellular survival within host cells also enables pneumococci to resist many antibiotics. Within the host cell, the bacteria exist in unique vacuoles, thereby avoiding degradation by the acidic lysosomes, and modulate the expression of its virulence genes to adapt to the intracellular environment. To invade and survive intracellularly, the pneumococcus utilizes a combination of virulence factors such as pneumolysin (PLY), pneumococcal surface protein A (PspA), pneumococcal adhesion and virulence protein B (PavB), the pilus-1 adhesin RrgA, pyruvate oxidase (SpxB), and metalloprotease (ZmpB). In this review, we discuss recent findings showing the intracellular persistence of Streptococcus pneumoniae and its underlying mechanisms.


Subject(s)
Pneumococcal Infections/microbiology , Streptococcus pneumoniae/pathogenicity , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Dendritic Cells/immunology , Drug Resistance, Microbial , Heart/microbiology , Heart/physiopathology , Humans , Lung/immunology , Lung/microbiology , Macrophages/immunology , Myocardium/metabolism , Myocardium/pathology , Nasopharynx/microbiology , Respiratory System/immunology , Respiratory System/microbiology , Spleen/cytology , Spleen/microbiology , Spleen/pathology , Streptococcus pneumoniae/immunology , Virulence Factors/metabolism
14.
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
15.
Cell Host Microbe ; 18(4): 390-1, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468741

ABSTRACT

Virulent group A streptococci have become a serious threat, with the emergence of the hypervirulent lineage M1T1. In this issue of Cell Host & Microbe, LaRock et al. (2015), uncover a role for the streptococcal M1 protein in neutralizing a key human antimicrobial peptide, cathelicidin.


Subject(s)
Antigens, Bacterial/metabolism , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Immune Evasion , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Animals , Humans
16.
Bioessays ; 37(4): 426-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640084

ABSTRACT

Transport of DNA across bacterial membranes involves complex DNA uptake systems. In Gram-positive bacteria, the DNA uptake machinery shares fundamental similarities with type IV pili and type II secretion systems. Although dedicated pilus structures, such as type IV pili in Gram-negative bacteria, are necessary for efficient DNA uptake, the role of similar structures in Gram-positive bacteria is just beginning to emerge. Recently two essentially very different pilus structures composed of the same major pilin protein ComGC were proposed to be involved in transformation of the Gram-positive bacterium Streptococcus pneumoniae - one is a long, thin, type IV pilus-like fiber with DNA binding capacity and the other one is a pilus structure that was thicker, much shorter and not able to bind DNA. Here we discuss how competence induced pili, either by pilus retraction or by a transient pilus-related opening in the cell wall, may mediate DNA uptake in S. pneumoniae.


Subject(s)
Fimbriae, Bacterial/metabolism , Streptococcus pneumoniae/metabolism , Transformation, Bacterial , Fimbriae, Bacterial/genetics , Streptococcus pneumoniae/genetics
17.
NPJ Biofilms Microbiomes ; 1: 15004, 2015.
Article in English | MEDLINE | ID: mdl-28721228
18.
Ups J Med Sci ; 119(2): 205-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24694025

ABSTRACT

Spread of antibiotic resistance is mediated by clonal lineages of bacteria that besides being resistant also possess other properties promoting their success. Some vaccines already in use, such as the pneumococcal conjugate vaccines, have had an effect on these successful clones, but at the same time have allowed for the expansion and resistance evolution of previously minor clones not covered by the vaccine. Since resistance frequently is horizontally transferred it will be difficult to generate a vaccine that covers all possible genetic lineages prone to develop resistance unless the vaccine target(s) is absolutely necessary for spread and/or disease development. Targeting the resistance mechanism itself by a vaccine is an interesting but hitherto unexplored approach.


Subject(s)
Bacterial Vaccines/administration & dosage , Drug Resistance, Microbial
19.
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
20.
Proc Natl Acad Sci U S A ; 111(7): E758-65, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550320

ABSTRACT

Streptococcus pneumoniae is a major human pathogen that successfully adapts to the host environment via an efficient uptake system for free DNA liberated from other organisms in the upper respiratory tract, facilitating immune evasion and drug resistance. Although the initial signaling events leading to pneumococcal competence for DNA transformation and the fate of DNA when it has been taken up have been extensively studied, the actual mechanism by which DNA in the environment may traverse the thick capsular and cell wall layers remains unknown. Here we visualize that induction of competence results in the formation of a native morphologically distinct pilus structure on the bacterial surface. This plaited pilus is encoded by the competence (com)G locus, and, after assembly, it is rapidly released into the surrounding medium. Heterologous pneumococcal pilus expression in Escherichia coli was obtained by replacing the pulE-K putative pilin genes of the Klebsiella oxytoca type II secretion system with the complete comG locus. In the pneumococcus, the coordinated secretion of pili from the cells correlates to DNA transformation. A model for DNA transformation is proposed whereby pilus assembly "drills" a channel across the thick cell wall that becomes transiently open by secretion of the pilus, providing the entry port for exogenous DNA to gain access to DNA receptors associated with the cytoplasmic membrane.


Subject(s)
Bacterial Secretion Systems/physiology , DNA Transformation Competence/genetics , DNA/metabolism , Fimbriae, Bacterial/metabolism , Streptococcus pneumoniae/metabolism , Transformation, Bacterial/physiology , Electrophoresis, Polyacrylamide Gel , Fimbriae, Bacterial/ultrastructure , Microscopy, Electron, Transmission , Transformation, Bacterial/genetics , Trichloroacetic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...