Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 300(5): 107282, 2024 May.
Article in English | MEDLINE | ID: mdl-38604564

ABSTRACT

The major human pathogen Streptococcus pneumoniae encounters the immune-derived oxidant hypothiocyanous acid (HOSCN) at sites of colonization and infection. We recently identified the pneumococcal hypothiocyanous acid reductase (Har), a member of the flavoprotein disulfide reductase enzyme family, and showed that it contributes to the HOSCN tolerance of S. pneumoniae in vitro. Here, we demonstrate in mouse models of pneumococcal infection that Har is critical for colonization and invasion. In a colonization model, bacterial load was attenuated dramatically in the nasopharynx when har was deleted in S. pneumoniae. The Δhar strain was also less virulent compared to wild type in an invasion model as reflected by a significant reduction in bacteria in the lungs and no dissemination to the blood and brain. Kinetic measurements with recombinant Har demonstrated that this enzyme reduced HOSCN with near diffusion-limited catalytic efficiency, using either NADH (kcat/KM = 1.2 × 108 M-1s-1) or NADPH (kcat/KM = 2.5 × 107 M-1s-1) as electron donors. We determined the X-ray crystal structure of Har in complex with the FAD cofactor to 1.50 Å resolution, highlighting the active site architecture characteristic for this class of enzymes. Collectively, our results demonstrate that pneumococcal Har is a highly efficient HOSCN reductase, enabling survival against oxidative host immune defenses. In addition, we provide structural insights that may aid the design of Har inhibitors.


Subject(s)
Bacterial Proteins , Pneumococcal Infections , Streptococcus pneumoniae , Streptococcus pneumoniae/enzymology , Animals , Mice , Pneumococcal Infections/microbiology , Pneumococcal Infections/enzymology , Pneumococcal Infections/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Humans , Female , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Thiocyanates
2.
J Biol Chem ; 297(2): 101000, 2021 08.
Article in English | MEDLINE | ID: mdl-34303706

ABSTRACT

DNA gyrase is a type II topoisomerase that is responsible for maintaining the topological state of bacterial and some archaeal genomes. It uses an ATP-dependent two-gate strand-passage mechanism that is shared among all type II topoisomerases. During this process, DNA gyrase creates a transient break in the DNA, the G-segment, to form a cleavage complex. This allows a second DNA duplex, known as the T-segment, to pass through the broken G-segment. After the broken strand is religated, the T-segment is able to exit out of the enzyme through a gate called the C-gate. Although many steps of the type II topoisomerase mechanism have been studied extensively, many questions remain about how the T-segment ultimately exits out of the C-gate. A recent cryo-EM structure of Streptococcus pneumoniae GyrA shows a putative T-segment in close proximity to the C-gate, suggesting that residues in this region may be important for coordinating DNA exit from the enzyme. Here, we show through site-directed mutagenesis and biochemical characterization that three conserved basic residues in the C-gate of DNA gyrase are important for DNA supercoiling activity, but not for ATPase or cleavage activity. Together with the structural information previously published, our data suggest a model in which these residues cluster to form a positively charged region that facilitates T-segment passage into the cavity formed between the DNA gate and C-gate.


Subject(s)
Catalytic Domain , DNA Gyrase/metabolism , DNA, Bacterial/chemistry , DNA, Superhelical , Pneumococcal Infections/enzymology , Protein Structural Elements , Streptococcus pneumoniae/enzymology , DNA Gyrase/chemistry , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Models, Molecular , Mutagenesis, Site-Directed/methods , Pneumococcal Infections/microbiology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/pathogenicity
3.
Nat Microbiol ; 6(2): 257-269, 2021 02.
Article in English | MEDLINE | ID: mdl-33349663

ABSTRACT

Streptococcus pneumoniae is a natural colonizer of the human respiratory tract and an opportunistic pathogen. Although epithelial cells are among the first to encounter pneumococci, the cellular processes and contribution of epithelial cells to the host response are poorly understood. Here, we show that a S. pneumoniae serotype 6B ST90 strain, which does not cause disease in a murine infection model, induces a unique NF-κB signature response distinct from an invasive-disease-causing isolate of serotype 4 (TIGR4). This signature is characterized by activation of p65 and requires a histone demethylase KDM6B. We show, molecularly, that the interaction of the 6B strain with epithelial cells leads to chromatin remodelling within the IL-11 promoter in a KDM6B-dependent manner, where KDM6B specifically demethylates histone H3 lysine 27 dimethyl. Remodelling of the IL-11 locus facilitates p65 access to three NF-κB sites that are otherwise inaccessible when stimulated by IL-1ß or TIGR4. Finally, we demonstrate through chemical inhibition of KDM6B with GSK-J4 inhibitor and through exogenous addition of IL-11 that the host responses to the 6B ST90 and TIGR4 strains can be interchanged both in vitro and in a murine model of infection in vivo. Our studies therefore reveal how a chromatin modifier governs cellular responses during infection.


Subject(s)
Chromatin Assembly and Disassembly , Host-Pathogen Interactions/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/pathogenicity , A549 Cells , Alveolar Epithelial Cells , Animals , Benzazepines/pharmacology , Disease Models, Animal , Enzyme Inhibitors , Epithelial Cells/microbiology , Gene Expression Regulation , Humans , Interleukin-11/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NF-kappa B/pharmacology , Pneumococcal Infections/enzymology , Pneumococcal Infections/genetics , Promoter Regions, Genetic , Pyrimidines/pharmacology
4.
Cell Rep ; 30(12): 4016-4026.e4, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32209465

ABSTRACT

Pathogenic bacteria can alter host gene expression through post-translational modifications of histones. We show that a natural colonizer, Streptococcus pneumoniae, induces specific histone modifications, including robust dephosphorylation of histone H3 on serine 10 (H3S10), during infection of respiratory epithelial cells. The bacterial pore-forming toxin pneumolysin (PLY), along with the pyruvate oxidase SpxB responsible for H2O2 production, play important roles in the induction of this modification. The combined effects of PLY and H2O2 trigger host signaling that culminates in H3S10 dephosphorylation, which is mediated by the host cell phosphatase PP1. Strikingly, S. pneumoniae infection induces dephosphorylation and subsequent activation of PP1 catalytic activity. Colonization of PP1 catalytically deficient cells results in impaired intracellular S. pneumoniae survival and infection. Interestingly, PP1 activation and H3S10 dephosphorylation are not restricted to S. pneumoniae and appear to be general epigenomic mechanisms favoring intracellular survival of pathogenic bacteria.


Subject(s)
Histones/metabolism , Host-Pathogen Interactions , Phosphoprotein Phosphatases/metabolism , Pneumococcal Infections/enzymology , Streptococcus pneumoniae/physiology , Animals , Bacterial Proteins/metabolism , Cell Line , Female , Gene Expression Regulation, Bacterial , Humans , Hydrogen Peroxide/metabolism , Inflammation/genetics , Mice, Inbred C57BL , Phosphorylation , Phosphoserine/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/genetics , Streptolysins/metabolism , Transcription, Genetic
5.
J Recept Signal Transduct Res ; 37(5): 522-534, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28768454

ABSTRACT

The discovery of clinically relevant inhibitors against MurF enzyme has proven to be a challenging task. In order to get further insight into the structural features required for the MurF inhibitory activity, we performed pharmacophore and atom-based three-dimensional quantitative structure-activity relationship studies for novel thiophene-3-carbonitriles based MurF inhibitors. The five-feature pharmacophore model was generated using 48 inhibitors having IC50 values ranging from 0.18 to 663 µm. The best-fitted model showed a higher coefficient of determination (R2 = 0.978), cross-validation coefficient (Q2 = 0.8835) and Pearson coefficient (0.9406) at four component partial least-squares factor. The model was validated with external data set and enrichment study. The effectiveness of the docking protocol was validated by docking the co-crystallized ligand into the catalytic pocket of MurF enzyme. Further, binding free energy calculated by the molecular mechanics generalized Born surface area approach showed that van der Waals and non-polar solvation energy terms are the main contributors to ligand binding in the active site of MurF enzyme. A 10-ns molecular dynamic simulation was performed to confirm the stability of the 3ZM6-ligand complex. Four new molecules are also designed as potent MurF inhibitors. These results provide insights regarding the development of novel MurF inhibitors with better binding affinity.


Subject(s)
Peptide Synthases/chemistry , Pneumococcal Infections/drug therapy , Streptococcus pneumoniae/enzymology , Thiophenes/chemistry , Binding Sites/drug effects , Catalytic Domain/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding/drug effects , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/genetics , Pneumococcal Infections/enzymology , Pneumococcal Infections/microbiology , Protein Binding , Quantitative Structure-Activity Relationship , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity , Thiophenes/metabolism , Thiophenes/pharmacology
6.
mBio ; 6(1)2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25604786

ABSTRACT

UNLABELLED: Streptococcus pneumoniae is a leading pathogen with an extracellular lifestyle; however, it is detected by cytosolic surveillance systems of macrophages. The innate immune response that follows cytosolic sensing of cell wall components results in recruitment of additional macrophages, which subsequently clear colonizing organisms from host airways. In this study, we monitored cytosolic access by following the transit of the abundant bacterial surface component capsular polysaccharide, which is linked to the cell wall. Confocal and electron microscopy visually characterized the location of cell wall components in murine macrophages outside membrane-bound organelles. Quantification of capsular polysaccharide through cellular fractionation demonstrated that cytosolic access of bacterial cell wall components is dependent on phagocytosis, bacterial sensitivity to the host's degradative enzyme lysozyme, and release of the pore-forming toxin pneumolysin. Activation of p38 mitogen-activated protein kinase (MAPK) signaling is important for limiting access to the cytosol; however, ultimately, these are catastrophic events for both the bacteria and the macrophage, which undergoes cell death. Our results show how expression of a pore-forming toxin ensures the death of phagocytes that take up the organism, although cytosolic sensing results in innate immune detection that eventually allows for successful host defense. These findings provide an example of how cytosolic access applies to an extracellular microbe and contributes to its pathogenesis. IMPORTANCE: Streptococcus pneumoniae (the pneumococcus) is a bacterial pathogen that is a leading cause of pneumonia. Pneumococcal disease is preceded by colonization of the nasopharynx, which lasts several weeks before being cleared by the host's immune system. Although S. pneumoniae is an extracellular microbe, intracellular detection of pneumococcal components is critical for bacterial clearance. In this study, we show that following bacterial uptake and degradation by phagocytes, pneumococcal products access the host cell cytosol via its pore-forming toxin. This phenomenon of cytosolic access results in phagocyte death and may serve to combat the host cells responsible for clearing the organism. Our results provide an example of how intracellular access and subsequent immune detection occurs during infection with an extracellular pathogen.


Subject(s)
Bacterial Toxins/metabolism , Cytosol/microbiology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/metabolism , Streptolysins/metabolism , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Cytosol/immunology , Humans , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Pneumococcal Infections/enzymology , Pneumococcal Infections/immunology , Streptolysins/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism
7.
PLoS One ; 8(7): e68647, 2013.
Article in English | MEDLINE | ID: mdl-23874703

ABSTRACT

Pathogenic bacteria produce a wide variety of virulence factors that are considered to be potential antibiotic targets. In this study, we report the crystal structure of a novel S. pneumoniae virulence factor, GHIP, which is a streptococcus-specific glycosyl hydrolase. This novel structure exhibits an α/ß-barrel fold that slightly differs from other characterized hydrolases. The GHIP active site, located at the negatively charged groove in the barrel, is very similar to the active site in known peptidoglycan hydrolases. Functionally, GHIP exhibited weak enzymatic activity to hydrolyze the PNP-(GlcNAc)5 peptidoglycan by the general acid/base catalytic mechanism. Animal experiments demonstrated a marked attenuation of S. pneumoniae-mediated virulence in mice infected by ΔGHIP-deficient strains, suggesting that GHIP functions as a novel S. pneumoniae virulence factor. Furthermore, GHIP participates in allowing S. pneumoniae to colonize the nasopharynx and invade host epithelial cells. Taken together, these findings suggest that GHIP can potentially serve as an antibiotic target to effectively treat streptococcus-mediated infection.


Subject(s)
Bacterial Proteins/chemistry , Hydrolases/chemistry , Pneumococcal Infections/enzymology , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/pathogenicity , Virulence Factors/chemistry , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cell Line, Tumor , Crystallography, X-Ray , Female , Humans , Hydrolases/genetics , Hydrolases/metabolism , Mice , Streptococcus pneumoniae/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
8.
J Microbiol ; 51(2): 194-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23625220

ABSTRACT

Antimicrobial peptides act as important innate immune defense mediators against invading microbes such as Streptococcus pneumoniae. Among a number of antimicrobial peptides, ß-defensin 2 (BD2) has strong antimicrobial activity against S. pneumoniae. However, little is known about the molecular signaling mechanisms leading to the BD2 expression. Here, we report that BD2 is strongly induced by S. pneumoniae in human airway cells including human middle-ear cells. Among diverse pneumococcal virulence factors, pneumolysin is required for inducing BD2 whose expression is under the control of p38 mitogen-activated protein kinase (MAPK). Pneumolysin also selectively regulates the expression of MAPK phosphatase 1 (MKP1), which inhibits the p38 signaling pathway, thereby leading to upregulation of BD2 to mount an effective defense against S. pneumoniae infection. These results provide novel insights into the molecular mechanisms underlying the coordinative regulation of BD2 expression via p38-MKP1 in the pathogenesis of airway infectious diseases.


Subject(s)
Pneumococcal Infections/enzymology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/metabolism , Streptolysins/metabolism , Up-Regulation , Virulence Factors/metabolism , beta-Defensins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Bacterial Proteins/metabolism , Cell Line , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Pneumococcal Infections/genetics , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/genetics , beta-Defensins/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
9.
J Immunol ; 190(1): 307-16, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23209321

ABSTRACT

IRAK4 is critical for MyD88-dependent TLR signaling, and patients with Irak4 mutations are extremely susceptible to recurrent bacterial infections. In these studies, mice homozygous for a mutant IRAK4 that lacks kinase activity (IRAK4(KDKI)) were used to address the role of IRAK4 in response to TLR agonists or bacterial infection. IRAK4(KDKI) macrophages exhibited diminished responsiveness to the TLR4 agonist LPS and little to no response to the TLR2 agonist Pam3Cys compared with wild-type macrophages as measured by cytokine mRNA, cytokine protein expression, and MAPK activation. Importantly, we identified two kinases downstream of the MAPKs, MNK1 and MSK1, whose phosphorylation is deficient in IRAK4(KDKI) macrophages stimulated through either TLR2 or TLR4, suggesting that IRAK4 contributes to TLR signaling beyond the initial phosphorylation of MAPKs. Additionally, IRAK4(KDKI) macrophages produced minimal cytokine mRNA expression in response to the Gram-positive bacteria Streptococcus pneumoniae and Staphylococcus aureus compared with WT cells, and IRAK4(KDKI) mice exhibited increased susceptibility and decreased cytokine production in vivo upon S. pneumoniae infection. Treatment of infected mice with a complex of polyinosinic-polycytidylic acid with poly-L-lysine and carboxymethyl cellulose (Hiltonol), a potent TLR3 agonist, significantly improved survival of both WT and IRAK4(KDKI) mice, thereby providing a potential treatment strategy in both normal and immunocompromised patients.


Subject(s)
Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Interleukin-1 Receptor-Associated Kinases/metabolism , Pneumococcal Infections/immunology , Signal Transduction/immunology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Animals , Cells, Cultured , Cytokines/genetics , Down-Regulation/immunology , Genetic Predisposition to Disease , Humans , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/enzymology , Pneumococcal Infections/genetics , Signal Transduction/genetics
10.
Mol Oral Microbiol ; 27(4): 270-83, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22759312

ABSTRACT

Streptococcus pneumoniae colonizes the upper respiratory tract from where the organisms may disseminate systemically to cause life threatening infections. The mechanisms by which pneumococci colonize epithelia are not understood, but neuraminidase A (NanA) has a major role in promoting growth and survival in the upper respiratory tract. In this article we show that mutants of S. pneumoniae D39 deficient in NanA or neuraminidase B (NanB) are abrogated in adherence to three epithelial cell lines, and to primary nasopharyngeal cells. Adherence levels were partly restored by nanA complementation in trans. Enzymic activity of NanA was shown to be necessary for pneumococcal adherence to epithelial cells, and adherence of the nanA mutant was restored to wild-type level by pre-incubation of epithelial cells with Lactococcus lactis cells expressing NanA. Pneumococcal nanA or nanB mutants were deficient in biofilm formation, while expression of NanA on L. lactis or Streptococcus gordonii promoted biofilm formation by these heterologous host organisms. The results suggest that NanA is an enzymic factor mediating adherence to epithelial cells by decrypting receptors for adhesion, and functions at least in part as an adhesin in biofilm formation. Neuraminidase A thus appears to play multiple temporal roles in pneumococcal infection, from adherence to host tissues, colonization, and community development, to systemic spread and crossing of the blood-brain barrier.


Subject(s)
Bacterial Proteins/physiology , Neuraminidase/physiology , Pneumococcal Infections/enzymology , Streptococcus pneumoniae/enzymology , Virulence Factors , Bacterial Adhesion/physiology , Bacterial Proteins/genetics , Biofilms/growth & development , Cells, Cultured , Epithelial Cells/microbiology , Humans , Mutation , Nasopharynx/cytology , Neuraminidase/genetics , Protein Binding , Receptors, Cell Surface/metabolism , Respiratory System/cytology , Streptococcus pneumoniae/genetics
11.
Nat Commun ; 3: 771, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22491319

ABSTRACT

Lung injury, whether induced by infection or caustic chemicals, initiates a series of complex wound-healing responses. If uncontrolled, these responses may lead to fibrotic lung diseases and loss of function. Thus, resolution of lung injury must be tightly regulated. The key regulatory proteins required for tightly controlling the resolution of lung injury have yet to be identified. Here we show that loss of deubiquitinase CYLD led to the development of lung fibrosis in mice after infection with Streptococcus pneumoniae. CYLD inhibited transforming growth factor-ß-signalling and prevented lung fibrosis by decreasing the stability of Smad3 in an E3 ligase carboxy terminus of Hsc70-interacting protein-dependent manner. Moreover, CYLD decreases Smad3 stability by deubiquitinating K63-polyubiquitinated Akt. Together, our results unveil a role for CYLD in tightly regulating the resolution of lung injury and preventing fibrosis by deubiquitinating Akt. These studies may help develop new therapeutic strategies for preventing lung fibrosis.


Subject(s)
Cysteine Endopeptidases/metabolism , Down-Regulation , Pneumococcal Infections/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Humans , Lung/enzymology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/enzymology , Pneumococcal Infections/genetics , Proto-Oncogene Proteins c-akt/genetics , Streptococcus pneumoniae/physiology , Transforming Growth Factor beta1/genetics , Tumor Suppressor Proteins/genetics , Ubiquitination
12.
Eur Respir J ; 40(6): 1458-67, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22441740

ABSTRACT

The majority of cases of community-acquired pneumonia are caused by Streptococcus pneumoniae and most studies on pneumococcal host interaction are based on cell culture or animal experiments. Thus, little is known about infections in human lung tissue. Cyclooxygenase-2 and its metabolites play an important regulatory role in lung inflammation. Therefore, we established a pneumococcal infection model on human lung tissue demonstrating mitogen-activated protein kinase (MAPK)-dependent induction of cyclooxygenase-2 and its related metabolites. In addition to alveolar macrophages and the vascular endothelium, cyclooxygenase-2 was upregulated in alveolar type II but not type I epithelial cells, which was confirmed in lungs of patients suffering from acute pneumonia. Moreover, we demonstrated the expression profile of all four E prostanoid receptors at the mRNA level and showed functionality of the E prostanoid(4) receptor by cyclic adenosine monophosphate production. Additionally, in comparison to previous studies, cyclooxygenase-2/prostaglandin E(2) related pro- and anti-inflammatory mediator regulation was partly confirmed in human lung tissue after pneumococcal infection. Overall, cell type-specific and MAPK-dependent cyclooxygenase-2 expression and prostaglandin E(2) formation in human lung tissue may play an important role in the early phase of pneumococcal infections.


Subject(s)
Cyclooxygenase 2/metabolism , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Lung/enzymology , Lung/microbiology , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/metabolism , Colony-Forming Units Assay , Dinoprostone/metabolism , Epithelial Cells/microbiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Humans , Immunohistochemistry/methods , Inflammation , MAP Kinase Signaling System , Microscopy, Fluorescence/methods , Pneumococcal Infections/enzymology , Prostaglandins/metabolism , Pulmonary Alveoli/microbiology
13.
J Biol Chem ; 287(13): 10472-10481, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22262863

ABSTRACT

Streptococcus pneumoniae is the most common causative agent of community-acquired pneumonia throughout the world, with high morbidity and mortality rates. A major feature of pneumococcal pneumonia is abundant neutrophil infiltration. In this study, we identified S. pneumoniae α-enolase as a neutrophil binding protein in ligand blot assay and mass spectrometry findings. Scanning electron microscopic and fluorescence microscopic analyses also revealed that S. pneumoniae α-enolase induces formation of neutrophil extracellular traps, which have been reported to bind and kill microbes. In addition, cytotoxic assay results showed that α-enolase dose-dependently increased the release of extracellular lactate dehydrogenase from human neutrophils as compared with untreated neutrophils. Furthermore, an in vitro cell migration assay using Chemotaxicell culture chambers demonstrated that α-enolase possesses neutrophil migrating activity. Interestingly, bactericidal assay findings showed that α-enolase increased neutrophil extracellular trap-dependent killing of S. pneumoniae in human blood. Moreover, pulldown assay and mass spectrometry results identified myoblast antigen 24.1D5 as an α-enolase-binding protein on human neutrophils, whereas flow cytometric analysis revealed that 24.1D5 was expressed on human neutrophils, but not on human monocytes or T cells. Together, our results indicate that α-enolase from S. pneumoniae increases neutrophil migrating activity and induces cell death of human neutrophils by releasing neutrophil extracellular traps. Furthermore, we found that myoblast antigen 24.1D5, which expressed on the surface of neutrophils, bound to α-enolase of S. pneumoniae.


Subject(s)
Neutrophils/metabolism , Phosphopyruvate Hydratase/metabolism , Pneumococcal Infections/enzymology , Streptococcus pneumoniae/enzymology , Cell Line, Tumor , Cell Movement/immunology , Humans , L-Lactate Dehydrogenase/immunology , L-Lactate Dehydrogenase/metabolism , Neutrophil Infiltration/immunology , Neutrophils/immunology , Phosphopyruvate Hydratase/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology
14.
J Immunol ; 187(9): 4890-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21957143

ABSTRACT

Streptococcus pneumoniae is a Gram-positive, extracellular bacterium that is responsible for significant mortality and morbidity worldwide. Pneumolysin (PLY), a cytolysin produced by all clinical isolates of the pneumococcus, is one of the most important virulence factors of this pathogen. We have previously reported that PLY is an essential factor for activation of caspase-1 and consequent secretion of IL-1ß and IL-18 in macrophages infected with S. pneumoniae. However, the host molecular factors involved in caspase-1 activation are still unclear. To further elucidate the mechanism of caspase-1 activation in macrophages infected with S. pneumoniae, we examined the involvement of inflammasomes in inducing this cellular response. Our study revealed that apoptosis-associated specklike protein containing a caspase recruitment domain (ASC), an adaptor protein for inflammasome receptors such as nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2), is essentially required for the induction of caspase-1 activation by S. pneumoniae. Caspase-1 activation was partially impaired in NLRP3(-/-) macrophages, whereas knockdown and knockout of AIM2 resulted in a clear decrease in caspase-1 activation in response to S. pneumoniae. These results suggest that ASC inflammasomes, including AIM2 and NLRP3, are critical for caspase-1 activation induced by S. pneumoniae. Furthermore, ASC(-/-) mice were more susceptible than wild-type mice to S. pneumoniae, with impaired secretion of IL-1ß and IL-18 into the bronchoalveolar lavage after intranasal infection, suggesting that ASC inflammasomes contribute to the protection of host from infection with PLY-producing S. pneumoniae.


Subject(s)
Caspase 1/metabolism , Cytoskeletal Proteins/physiology , Immunity, Innate , Inflammasomes/physiology , Pneumococcal Infections/immunology , Pneumococcal Infections/metabolism , Animals , Apoptosis Regulatory Proteins , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/biosynthesis , CARD Signaling Adaptor Proteins , Carrier Proteins/physiology , Caspase 1/deficiency , Caspase 1/genetics , Cell Line , Cell Line, Transformed , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , DNA-Binding Proteins , Disease Resistance/immunology , Enzyme Activation/immunology , Female , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/physiology , Pneumococcal Infections/enzymology , Streptolysins/antagonists & inhibitors , Streptolysins/biosynthesis
15.
Curr Eye Res ; 35(9): 787-98, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20795860

ABSTRACT

PURPOSE: The pneumococcal capsule is required for pathogenesis in systemic infections, yet reports show most conjunctivitis outbreaks are caused by nonencapsulated pneumococci, while keratitis infections are caused by encapsulated strains. This study aims to determine the effect of capsule in pneumococcal keratitis and conjunctivitis in rabbit models of infection. METHODS: A capsule-deficient isogenic mutant was created using homologous transformation. Parent and mutant strains were injected within the upper bulbar conjunctiva (conjunctivitis) or into the corneal stroma (keratitis) of New Zealand white rabbits. Clinical examinations were performed 24 and 48 hr post-infection at which time corneas or conjunctivae were removed, homogenized, and plated to determine the recovered bacterial load. Whole eyes were removed for histological examination. The neuraminidase activity was determined following in vitro and in vivo growth. RESULTS: There were no significant differences in clinical scores between the eyes infected with the parent or mutant for either infection, nor was there a difference in the amount of bacteria recovered from the cornea. In the conjunctivae, however, the mutant strain was cleared by the host faster than the parent strain. Histological examination showed slightly more infiltrating polymorphonuclear leukocytes (PMN) and macrophages in the conjunctivae infected with the parent strain. The neuraminidase activity of both strains was not significantly different when the strains were grown in vitro. However, the neuraminidase activity of the parent was significantly less than that of the mutant at 3 and 12 hr post conjunctival infection. CONCLUSIONS: Although more outbreaks of pneumococcal conjunctivitis are tied to nonencapsulated S. pneumoniae strains, this study showed that an encapsulated strain was capable of establishing conjunctivitis in a rabbit injection model and survive attack by the host immune system longer than its nonencapsulated isogenic mutant. Nonetheless, the nonencapsulated pneumococci had an increased neuraminidase activity level in vivo when compared to the parent strain.


Subject(s)
Bacterial Capsules/physiology , Conjunctivitis, Bacterial/microbiology , Keratitis/microbiology , Neuraminidase/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Animals , Colony Count, Microbial , Conjunctivitis, Bacterial/enzymology , Conjunctivitis, Bacterial/pathology , DNA Primers , Disease Models, Animal , Granulocytes/physiology , Keratitis/enzymology , Keratitis/pathology , Macrophages/physiology , Neutrophils/physiology , Oligonucleotides/chemistry , Pneumococcal Infections/enzymology , Pneumococcal Infections/pathology , Rabbits
16.
Am J Physiol Lung Cell Mol Physiol ; 298(1): L67-78, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19783640

ABSTRACT

The immunological explanation for the "hygiene hypothesis" has been proposed to be induction of T helper 1 (Th1) responses by microbial products. However, the protective results of hygiene hypothesis-linked microbial exposures are currently shown to be unlikely to result from a Th1-skewed response. Until now, effect of microbial exposure early in life on airway innate resistance remained unclear. We examined the role of early life exposure to microbes in airway innate resistance to a respiratory pathogen. Specific pathogen-free weanling mice were nasally exposed to the mixture of microbial extracts or PBS (control) every other day for 28 days and intratracheally infected with Streptococcus pneumoniae 10 days after the last exposure. Exposure to microbial extracts facilitated colonization of aerobic gram-positive bacteria, anaerobic microorganisms, and Lactobacillus in the airway, compared with control exposure. In pneumococcal pneumonia, the exposure prolonged mouse survival days by suppressing bacterial growth and by retarding pneumococcal blood invasion, despite significantly low levels of leukocyte recruitment in the lung. Enhancement of airway resistance was associated with a significant decrease in production of leukocyte chemokine (KC) and TNFalpha, and suppression of matrix metalloproteinase (MMP-9) expression/activation with enhancement of tissue inhibitor of MMP (TIMP-3) activation. The exposure increased production of IFN-gamma, IL-4, and monocyte chemoattractant-1 following infection. Furthermore, expression of Toll-like receptor 2, 4, and 9 was promoted by the exposure but no longer upregulated upon pneumococcal infection. Thus, we suggest that hygiene hypothesis is more important in regulating the PMN-dominant inflammatory response than in inducing a Th1-dominant response.


Subject(s)
Immunity, Innate/immunology , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumonia/immunology , Pneumonia/microbiology , Streptococcus pneumoniae/physiology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Movement , Chemokines/metabolism , Colony Count, Microbial , Enzyme Activation , Immunoglobulin A/immunology , Lung/enzymology , Lung/microbiology , Lung/pathology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/microbiology , Pneumococcal Infections/complications , Pneumococcal Infections/enzymology , Pneumonia/complications , Pneumonia/enzymology , Streptococcus pneumoniae/growth & development , Tissue Inhibitor of Metalloproteinase-3/metabolism , Toll-Like Receptors/metabolism
17.
J Endocrinol ; 203(2): 263-70, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19656972

ABSTRACT

We have previously shown that skeletal muscle deiodinase type 2 (D2) mRNA (listed as Dio2 in MGI Database) is upregulated in an animal model of acute illness. However, human studies on the expression of muscle D2 during illness report conflicting data. Therefore, we evaluated the expression of skeletal muscle D2 and D2-regulating factors in two mouse models of illness that differ in timing and severity of illness: 1) turpentine-induced inflammation, and 2) Streptococcus pneumoniae infection. During turpentine-induced inflammation, D2 mRNA and activity increased compared to pair-fed controls, most prominently at day 1 and 2, whereas after S. pneumoniae infection D2 mRNA decreased. We evaluated the association of D2 expression with serum thyroid hormones, (de-)ubiquitinating enzymes ubiquitin-specific peptidase 33 and WD repeat and SOCS box-containing 1 (Wsb1), cytokine expression and activation of inflammatory pathways and cAMP pathway. During chronic inflammation the increased muscle D2 expression is associated with the activation of the cAMP pathway. The normalization of D2 5 days after turpentine injection coincides with increased Wsb1 and tumor necrosis factor alpha expression. Muscle interleukin-1beta (Il1b) expression correlated with decreased D2 mRNA expression after S. pneumoniae infection. In conclusion, muscle D2 expression is differentially regulated during illness, probably related to differences in the inflammatory response and type of pathology. D2 mRNA and activity increases in skeletal muscle during the acute phase of chronic inflammation compared to pair-fed controls probably due to activation of the cAMP pathway. In contrast, muscle D2 mRNA decreases 48 h after a severe bacterial infection, which is associated with local Il1b mRNA expression and might also be due to diminished food-intake.


Subject(s)
Gene Expression Regulation, Enzymologic , Iodide Peroxidase/genetics , Muscle, Skeletal/enzymology , Pneumococcal Infections/enzymology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Inflammation/chemically induced , Inflammation/enzymology , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Messenger/analysis , Signal Transduction , Thyroid Hormones/blood , Turpentine , Ubiquitin-Protein Ligases/genetics , Iodothyronine Deiodinase Type II
18.
J Biol Chem ; 284(38): 26161-73, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19608744

ABSTRACT

The presence of a fucose utilization operon in the Streptococcus pneumoniae genome and its established importance in virulence indicates a reliance of this bacterium on the harvesting of host fucose-containing glycans. The identities of these glycans, however, and how they are harvested is presently unknown. The biochemical and high resolution x-ray crystallographic analysis of two family 98 glycoside hydrolases (GH98s) from distinctive forms of the fucose utilization operon that originate from different S. pneumoniae strains reveal that one enzyme, the predominant type among pneumococcal isolates, has a unique endo-beta-galactosidase activity on the LewisY antigen. Altered active site topography in the other species of GH98 enzyme tune its endo-beta-galactosidase activity to the blood group A and B antigens. Despite their different specificities, these enzymes, and by extension all family 98 glycoside hydrolases, use an inverting catalytic mechanism. Many bacterial and viral pathogens exploit host carbohydrate antigens for adherence as a precursor to colonization or infection. However, this is the first evidence of bacterial endoglycosidase enzymes that are known to play a role in virulence and are specific for distinct host carbohydrate antigens. The strain-specific distribution of two distinct types of GH98 enzymes further suggests that S. pneumoniae strains may specialize to exploit host-specific antigens that vary from host to host, a factor that may feature in whether a strain is capable of colonizing a host or establishing an invasive infection.


Subject(s)
Bacterial Proteins/chemistry , Glycoside Hydrolases/chemistry , Lewis Blood Group Antigens/chemistry , Streptococcus pneumoniae/enzymology , Bacterial Proteins/metabolism , Glycoside Hydrolases/metabolism , Humans , Lewis Blood Group Antigens/metabolism , Operon , Pneumococcal Infections/enzymology , Species Specificity , Streptococcus pneumoniae/pathogenicity , Substrate Specificity/physiology
19.
J Cell Sci ; 122(Pt 2): 256-67, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19118218

ABSTRACT

By interacting with components of the human host, including extracellular matrix (ECM) proteins, Streptococcus pneumoniae has evolved various strategies for colonization. Here, we characterized the interaction of pneumococci with the adhesive glycoprotein vitronectin and the contribution of this protein to pneumococcal uptake by host cells in an integrin-dependent manner. Specific interaction of S. pneumoniae with the heparin-binding sites of purified multimeric vitronectin was demonstrated by flow cytometry analysis. Host-cell-bound vitronectin promoted pneumococcal adherence to and invasion into human epithelial and endothelial cells. Pneumococci were trapped by microspike-like structures, which were induced upon contact of pneumococci with host-cell-bound vitronectin. Alphavbeta3 integrin was identified as the major cellular receptor for vitronectin-mediated adherence and uptake of pneumococci. Ingestion of pneumococci by host cells via vitronectin required a dynamic actin cytoskeleton and was dependent on integrin-linked kinase (ILK), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt), as demonstrated by gene silencing or in inhibition experiments. In conclusion, pneumococci exploit the vitronectin-alphavbeta3-integrin complex as a cellular receptor for invasion and this integrin-mediated internalization requires the cooperation between the host signalling molecules ILK, PI3K and Akt.


Subject(s)
Endothelial Cells/metabolism , Epithelial Cells/metabolism , Pneumococcal Infections/enzymology , Protein Serine-Threonine Kinases/metabolism , Streptococcus pneumoniae/metabolism , Vitronectin/metabolism , Actins/metabolism , Cell Line , Cytoskeleton/metabolism , Cytoskeleton/microbiology , Endothelial Cells/cytology , Endothelial Cells/microbiology , Epithelial Cells/cytology , Epithelial Cells/microbiology , Heparin/metabolism , Humans , Integrin alphaVbeta3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pneumococcal Infections/microbiology , Proto-Oncogene Proteins c-akt/metabolism
20.
PLoS Pathog ; 4(12): e1000241, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19079576

ABSTRACT

The abundance of lysozyme on mucosal surfaces suggests that successful colonizers must be able to evade its antimicrobial effects. Lysozyme has a muramidase activity that hydrolyzes bacterial peptidoglycan and a non-muramidase activity attributable to its function as a cationic antimicrobial peptide. Two enzymes (PgdA, a N-acetylglucosamine deacetylase, and Adr, an O-acetyl transferase) that modify different sites on the peptidoglycan of Streptococcus pneumoniae have been implicated in its resistance to lysozyme in vitro. Here we show that the antimicrobial effect of human lysozyme is due to its muramidase activity and that both peptidoglycan modifications are required for full resistance by pneumococci. To examine the contribution of lysozyme and peptidoglycan modifications during colonization of the upper respiratory tract, competition experiments were performed with wild-type and pgdAadr mutant pneumococci in lysozyme M-sufficient (LysM(+/+)) and -deficient (LysM(-/-)) mice. The wild-type strain out-competed the double mutant in LysM(+/+), but not LysM(-/-) mice, indicating the importance of resistance to the muramidase activity of lysozyme during mucosal colonization. In contrast, strains containing single mutations in either pgdA or adr prevailed over the wild-type strain in both LysM(+/+) and LysM(-/-) mice. Our findings demonstrate that individual peptidoglycan modifications diminish fitness during colonization. The competitive advantage of wild-type pneumococci in LysM(+/+) but not LysM(-/-) mice suggests that the combination of peptidoglycan modifications reduces overall fitness, but that this is outweighed by the benefits of resistance to the peptidoglycan degrading activity of lysozyme.


Subject(s)
Muramidase/metabolism , Peptidoglycan/metabolism , Pneumococcal Infections/microbiology , Respiratory Mucosa/enzymology , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , Acetyltransferases/genetics , Acetyltransferases/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Bacterial Capsules/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Wall/metabolism , Female , Humans , Mice , Mutation , Nasopharynx/enzymology , Nasopharynx/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Peptidoglycan/chemistry , Pneumococcal Infections/enzymology , Pneumococcal Infections/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Tract Infections/enzymology , Respiratory Tract Infections/immunology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL