Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Oral Biosci ; 65(1): 119-125, 2023 03.
Article in English | MEDLINE | ID: mdl-36464149

ABSTRACT

OBJECTIVES: Commensal bacteria in the host body play a fundamental role in the differentiation and maintenance of the immune system. Studies on intestinal immunity have revealed that, under steady-state conditions, microflora have an important role in the maintenance of health. However, the role of oral commensal bacteria on the oral immune system is still unclear. Here, we clarify the interactions between commensal bacteria and the oral mucosal immune system under steady-state conditions. METHODS: We used germ-free mice that had never been exposed to bacteria and conventional mice grown with normal bacterial flora. Oral cells were isolated from the oral mucosa, stained with specific antibodies, and analyzed by flow cytometry. For the detection of myeloperoxidase and intracellular cytokines, oral cells were stimulated with N-formyl-methionine-leucyl-phenylalanine and phorbol 12-myristate 13-acetate/ionomycin, respectively. RESULTS: We found that the oral mucosa harbored more neutrophils in germ-free mice than in conventional mice. However, the majority of neutrophils in the germ-free oral mucosa exhibited an immature phenotype. Other immune cells, including macrophages, T cells, and B cells, in the oral mucosa of germ-free mice showed similar differentiation to those in conventional mice. These results indicate that in the steady-state oral mucosa, the normal commensal flora promote the peripheral differentiation of neutrophils. CONCLUSIONS: The presence of commensal flora is critical for the development of adequate immune system in the oral mucosa.


Subject(s)
Mouth Mucosa , Neutrophils , Animals , Mice , Cytokines , Bacteria , Cell Differentiation
2.
PLoS One ; 17(11): e0276293, 2022.
Article in English | MEDLINE | ID: mdl-36350830

ABSTRACT

Members of the mitis group streptococci are the most abundant inhabitants of the oral cavity and dental plaque. Influenza A virus (IAV), the causative agent of influenza, infects the upper respiratory tract, and co-infection with Streptococcus pneumoniae is a major cause of morbidity during influenza epidemics. S. pneumoniae is a member of mitis group streptococci and shares many features with oral mitis group streptococci. In this study, we investigated the effect of viable Streptococcus oralis, a representative member of oral mitis group, on the infectivity of H1N1 IAV. The infectivity of IAV was measured by a plaque assay using Madin-Darby canine kidney cells. When IAV was incubated in growing culture of S. oralis, the IAV titer decreased in a time- and dose-dependent manner and became less than 100-fold, whereas heat-inactivated S. oralis had no effect. Other oral streptococci such as Streptococcus mutans and Streptococcus salivarius also reduced the viral infectivity to a lesser extent compared to S. oralis and Streptococcus gordonii, another member of the oral mitis group. S. oralis produces hydrogen peroxide (H2O2) at a concentration of 1-2 mM, and its mutant deficient in H2O2 production showed a weaker effect on the inactivation of IAV, suggesting that H2O2 contributes to viral inactivation. The contribution of H2O2 was confirmed by an inhibition assay using catalase, an H2O2-decomposing enzyme. These oral streptococci produce short chain fatty acids (SCFA) such as acetic acid as a by-product of sugar metabolism, and we also found that the inactivation of IAV was dependent on the mildly acidic pH (around pH 5.0) of these streptococcal cultures. Although inactivation of IAV in buffers of pH 5.0 was limited, incubation in the same buffer containing 2 mM H2O2 resulted in marked inactivation of IAV, which was similar to the effect of growing S. oralis culture. Taken together, these results reveal that viable S. oralis can inactivate IAV via the production of SCFAs and H2O2. This finding also suggests that the combination of mildly acidic pH and H2O2 at low concentrations could be an effective method to inactivate IAV.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Humans , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Influenza A virus/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Streptococcus mitis , Streptococcus oralis , Viridans Streptococci/metabolism , Streptococcus gordonii/metabolism , Acids/metabolism , Hydrogen-Ion Concentration
3.
Microbiol Immunol ; 66(12): 539-551, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36114681

ABSTRACT

Members of the oral mitis group streptococci including Streptococcus oralis, Streptococcus sanguinis, and Streptococcus gordonii are the most abundant inhabitants of human oral cavity and dental plaque, and have been implicated in infectious complications such as bacteremia and infective endocarditis. Oral mitis group streptococci are genetically close to Streptococcus pneumoniae; however, they do not produce cytolysin (pneumolysin), which is a key virulence factor of S. pneumoniae. Similar to S. pneumoniae, oral mitis group streptococci possess several cell surface proteins that bind to the cell surface components of host mammalian cells. S. sanguinis expresses long filamentous pili that bind to the matrix proteins of host cells. The cell wall-anchored nuclease of S. sanguinis contributes to the evasion of the neutrophil extracellular trap by digesting its web-like extracellular DNA. Oral mitis group streptococci produce glucosyltransferases, which synthesize glucan (glucose polymer) from sucrose of dietary origin. Neuraminidase (NA) is a virulent factor in oral mitis group streptococci. Influenza type A virus (IAV) relies on viral NA activity to release progeny viruses from infected cells and spread the infection, and NA-producing oral streptococci elevate the risk of IAV infection. Moreover, oral mitis group streptococci produce hydrogen peroxide (H2 O2 ) as a by-product of sugar metabolism. Although the concentrations of streptococcal H2 O2 are low (1-2 mM), they play important roles in bacterial competition in the oral cavity and evasion of phagocytosis by host macrophages and neutrophils. In this review, we intended to describe the diverse pathogenicity of oral mitis group streptococci.


Subject(s)
Mouth , Humans
4.
Microbiol Immunol ; 66(6): 253-263, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35088451

ABSTRACT

Secondary bacterial infection following influenza type A virus (IAV) infection is a major cause of morbidity and mortality during influenza epidemics. Streptococcus pneumoniae has been identified as a predominant pathogen in secondary pneumonia cases that develop following influenza. Although IAV has been shown to enhance susceptibility to the secondary bacterial infection, the underlying mechanism of the viral-bacterial synergy leading to disease progression is complex and remains elusive. In this review, cooperative interactions of viruses and streptococci during co- or secondary infection with IAV are described. IAV infects the upper respiratory tract, therefore, streptococci that inhabit or infect the respiratory tract are of special interest. As many excellent reviews on the co-infection of IAV and S. pneumoniae have already been published, this review is intended to describe the unique interactions between other streptococci and IAV. Both streptococcal and IAV infections modulate the host epithelial barrier of the respiratory tract in various ways. IAV infection directly disrupts epithelial barriers, though at the same time the virus modifies the properties of infected cells to enhance streptococcal adherence and invasion. Mitis group streptococci produce neuraminidases, which promote IAV infection in a unique manner. The studies reviewed here have revealed intriguing mechanisms underlying secondary streptococcal infection following influenza.


Subject(s)
Coinfection , Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Streptococcal Infections , Coinfection/complications , Humans , Influenza, Human/complications , Streptococcal Infections/microbiology , Streptococcus pneumoniae
5.
PLoS One ; 15(4): e0231101, 2020.
Article in English | MEDLINE | ID: mdl-32302339

ABSTRACT

Mast cells and basophils are central players in allergic reactions triggered by immunoglobulin E (IgE). They have intracellular granules containing allergic mediators (e.g., histamine, serotonin, inflammatory cytokines, proteases and ß-hexosaminidase), and stimulation by IgE-allergen complex leads to the release of such allergic mediators from the granules, that is, degranulation. Mast cells are residents of mucosal surfaces, including those of nasal and oral cavities, and play an important role in the innate defense system. Members of the mitis group streptococci such as Streptococcus oralis, are primary colonizers of the human oral cavity. They produce hydrogen peroxide (H2O2) as a by-product of sugar metabolism. In this study, we investigated the effects of streptococcal infection on RBL-2H3 mast cell/basophil cell line. Infection by oral streptococci did not induce degranulation of the cells. Stimulation of the RBL-2H3 cells with anti-dinitrophenol (DNP) IgE and DNP-conjugated human serum albumin triggers degranulation with the release of ß-hexosaminidase. We found that S. oralis and other mitis group streptococci inhibited the IgE-triggered degranulation of RBL-2H3 cells. Since mitis group streptococci produce H2O2, we examined the effect of S. oralis mutant strain deficient in producing H2O2, and found that they lost the ability to suppress the degranulation. Moreover, H2O2 alone inhibited the IgE-induced degranulation. Subsequent analysis suggested that the inhibition of degranulation was related to the cytotoxicity of streptococcal H2O2. Activated RBL-2H3 cells produce interleukin-4 (IL-4); however, IL-4 production was not induced by streptococcal H2O2. Furthermore, an in vivo study using the murine pollen-induced allergic rhinitis model suggested that the streptococcal H2O2 reduces nasal allergic reaction. These findings reveal that H2O2 produced by oral mitis group streptococci inhibits IgE-stimulated degranulation by inducing cell death. Consequently, streptococcal H2O2 can be considered to modulate the allergic reaction in mucosal surfaces.


Subject(s)
Allergens/metabolism , Hypersensitivity/immunology , Immunoglobulin E/immunology , Streptococcal Infections/drug therapy , Allergens/immunology , Animals , Basophils/immunology , Basophils/microbiology , Basophils/pathology , Cell Degranulation/immunology , Cell Survival/immunology , Dinitrophenols/pharmacology , Humans , Hydrogen Peroxide/metabolism , Hypersensitivity/drug therapy , Hypersensitivity/pathology , Immunoglobulin E/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Mast Cells/immunology , Mast Cells/microbiology , Mast Cells/pathology , Mice , Plant Extracts/chemistry , Plant Extracts/pharmacology , Serum Albumin, Human/immunology , Serum Albumin, Human/metabolism , Streptococcal Infections/immunology , Streptococcus oralis/immunology , Streptococcus oralis/pathogenicity , Sugars/metabolism
6.
Infect Immun ; 88(1)2019 12 17.
Article in English | MEDLINE | ID: mdl-31611274

ABSTRACT

Food allergy is a life-threatening response to specific foods, and microbiota imbalance (dysbiosis) in gut is considered a cause of this disease. Meanwhile, the host immune response also plays an important role in the disease. Notably, interleukin 33 (IL-33) released from damaged or necrotic intestinal epithelial cells facilitates IL-2-producing CD4 helper T (Th2) responses. However, causal relationships between the gut and oral dysbiosis and food allergy remain unknown. In this study, we analyzed effects of gut and oral dysbiosis on development of food allergy. A murine model of food allergy was established via ovalbumin (OVA) injection in BALB/c mice. Viable fecal bacteria were identified using matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS). il33 expression in colon-26 mouse colon cells stimulated by isolated fecal bacteria was quantified by real-time PCR. Intestinal T cells from the mice were analyzed by flow cytometry. Salivary IgA levels were quantified by enzyme-linked immunosorbent assay (ELISA), and IgA-bound oral bacteria were detected by flow cytometry. Among fecal bacteria, the abundance of Citrobacter sp. increased in the feces of allergic mice and induced il33 expression in colon-26 cells. Orally administered Citrobacter koseri JCM1658 exacerbated systemic allergic symptoms and reduced intestinal Th17 cells. Salivary IgA and IgA-bound oral bacteria increased in the allergic mice. Based on the results described above, food allergy induced both gut and oral dysbiosis. Citrobacter sp. aggravated allergy symptoms by inducing IL-33 release from intestinal epithelial cells.


Subject(s)
Dysbiosis , Food Hypersensitivity/complications , Gastrointestinal Tract/microbiology , Immunoglobulin A/metabolism , Immunologic Factors/metabolism , Microbiota/drug effects , Mouth/microbiology , Animals , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Mice, Inbred BALB C
7.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29263111

ABSTRACT

Small noncoding RNAs (sRNAs) have been identified as important regulators of gene expression in various cellular processes. cia-dependent small RNAs (csRNAs), a group of sRNAs that are controlled by the two-component regulatory system CiaRH, are widely conserved in streptococci, but their targets have been identified only in Streptococcus pneumoniaeStreptococcus sanguinis, a pioneer colonizer of teeth and one of the most predominant bacteria in the early oral biofilm, has been shown to have six csRNAs. Using computational target prediction and the luciferase reporter assay, we identified pilT, a constituent of the type IV pilus operon, as a negative regulatory target for one of the csRNAs, namely, csRNA1-1, in S. sanguinis RNA-RNA electrophoretic mobility shift assay using a nucleotide exchange mutant of csRNA1-1 revealed that csRNA1-1 binds directly to pilT mRNA. In addition, csRNA1-1 and csRNA1-2, a putative gene duplication product of csRNA1-1 that is tandemly located in the S. sanguinis genome, negatively regulated S. sanguinis biofilm formation. These results suggest the involvement of csRNAs in the colonization step of S. sanguinis.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Biofilms , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Small Untranslated/metabolism , Streptococcal Infections/microbiology , Streptococcus sanguis/genetics , Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , Down-Regulation , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , RNA, Bacterial/metabolism , RNA, Small Untranslated/genetics , Streptococcus sanguis/physiology
8.
PLoS One ; 12(2): e0172223, 2017.
Article in English | MEDLINE | ID: mdl-28222125

ABSTRACT

Streptococcus is the dominant bacterial genus in the human oral cavity and a leading cause of infective endocarditis. Streptococcus sanguinis belongs to the mitis group of streptococci and produces hydrogen peroxide (H2O2) by the action of SpxB, a pyruvate oxidase. In this study, we investigated the involvement of SpxB in survival of S. sanguinis in human blood and whether bacterial H2O2 exhibits cytotoxicity against human neutrophils. Results of a bactericidal test with human whole blood revealed that the spxB mutation in S. sanguinis is detrimental to its survival in blood. When S. sanguinis strains were exposed to isolated neutrophils, the bacterial survival rate was significantly decreased by spxB deletion. Furthermore, human neutrophils exposed to the S. sanguinis wild-type strain, in contrast to those exposed to an spxB mutant strain, underwent cell death with chromatin de-condensation and release of web-like extracellular DNA, reflecting induction of neutrophil extracellular traps (NETs). Since reactive oxygen species-mediated NET induction requires citrullination of arginine residues in histone proteins and subsequent chromatin de-condensation, we examined citrullination levels of histone in infected neutrophils. It is important to note that the citrullinated histone H3 was readily detected in neutrophils infected with the wild-type strain, as compared to infection with the spxB mutant strain. Moreover, decomposition of streptococcal H2O2 with catalase reduced NET induction. These results suggest that H2O2 produced by S. sanguinis provokes cell death of neutrophils and NET formation, thus potentially affecting bacterial survival in the bloodstream.


Subject(s)
Bacterial Proteins/physiology , Hydrogen Peroxide/metabolism , Neutrophils/microbiology , Pyruvate Oxidase/physiology , Streptococcus sanguis/physiology , Adult , Bacterial Proteins/genetics , Blood Bactericidal Activity , Cell Death , Chromatin/ultrastructure , Citrulline/analysis , Extracellular Traps , Gene Deletion , Histones/blood , Humans , Neutrophils/physiology , Protein Processing, Post-Translational , Pyruvate Oxidase/deficiency , Pyruvate Oxidase/genetics , Reactive Oxygen Species , Streptococcus sanguis/genetics , Streptococcus sanguis/pathogenicity , Virulence
9.
Biochem Biophys Res Commun ; 485(2): 461-467, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202416

ABSTRACT

Oral streptococci including mitis group streptococci are commensal residents and are also the first to colonize the oral cavity. However, various species of these oral streptococci have the potential to invade the host and occasionally lead to severe infectious disease such as cardiovascular diseases. Oral streptococci have close interactions with the host immune system including macrophages at the oral mucosal surface. One notable common trait of oral streptococcus including Streptococcus oralis (S. oralis) is the production of hydrogen peroxide (H2O2). Using a comprehensive microarray approach, we sought to understand the innate immune response profiling affected by H2O2 production from oral streptococci. We compared the gene expression patterns of macrophages infected with S. oralis wild type (WT) and streptococcal pyruvate oxidase knockout (SpxB-KO), a strain that does not produce H2O2. We found that H2O2 from S. oralis suppressed proinflammatory gene expression such as TNF-α, that is induced in response to infection, and activated the cellular stress genes such as Egr-1 in response to oxidative stress. A comparative gene ontology analysis of S. oralis WT and SpxB-KO strains revealed that during infection, down regulated genes were closely related to the processes involved in the host defense reaction and up regulated genes were related with the cellular stress responses. Using qPCR analysis, we also confirmed the same pattern of expression changes such as TNF-α, IL-6 and Egr-1. Furthermore, supernatant from SpxB-KO could not suppress the expression of TNF-α in macrophages stimulated with LPS. These findings suggested that H2O2 production from S. oralis leads to the suppression of inflammatory responses and NF-κB signaling pathways in macrophages as well as the induction of the oxidative stress response. We concluded that streptococcal H2O2 production has the beneficial effects of modulating the innate immune response, thereby stabilizing streptococcal colonization at the mucosal surface and even in the bloodstream leading to cardiovascular disease after invasion, in addition to the commensal role to compete other bacterial species as initial colonizer at oral cavity.


Subject(s)
Gene Expression Profiling/methods , Hydrogen Peroxide/metabolism , Macrophages/metabolism , Oligonucleotide Array Sequence Analysis/methods , Streptococcus oralis/metabolism , 3T3 Cells , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blotting, Western , Cell Line , Cluster Analysis , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Ontology , Host-Pathogen Interactions , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mutation , Pyruvate Oxidase/genetics , Pyruvate Oxidase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Streptococcus oralis/genetics , Streptococcus oralis/physiology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Infect Immun ; 84(7): 2042-2050, 2016 07.
Article in English | MEDLINE | ID: mdl-27113357

ABSTRACT

Streptococcus oralis, an oral commensal, belongs to the mitis group of streptococci and occasionally causes opportunistic infections, such as bacterial endocarditis and bacteremia. Recently, we found that the hydrogen peroxide (H2O2) produced by S. oralis is sufficient to kill human monocytes and epithelial cells, implying that streptococcal H2O2 is a cytotoxin. In the present study, we investigated whether streptococcal H2O2 impacts lysosomes, organelles of the intracellular digestive system, in relation to cell death. S. oralis infection induced the death of RAW 264 macrophages in an H2O2-dependent manner, which was exemplified by the fact that exogenous H2O2 also induced cell death. Infection with either a mutant lacking spxB, which encodes pyruvate oxidase responsible for H2O2 production, or Streptococcus mutans, which does not produce H2O2, showed less cytotoxicity. Visualization of lysosomes with LysoTracker revealed lysosome deacidification after infection with S. oralis or exposure to H2O2, which was corroborated by acridine orange staining. Similarly, fluorescent labeling of lysosome-associated membrane protein-1 gradually disappeared during infection with S. oralis or exposure to H2O2 The deacidification and the following induction of cell death were inhibited by chelating iron in lysosomes. Moreover, fluorescent staining of cathepsin B indicated lysosomal destruction. However, treatment of infected cells with a specific inhibitor of cathepsin B had negligible effects on cell death; instead, it suppressed the detachment of dead cells from the culture plates. These results suggest that streptococcal H2O2 induces cell death with lysosomal destruction and then the released lysosomal cathepsins contribute to the detachment of the dead cells.


Subject(s)
Hydrogen Peroxide/metabolism , Lysosomes/metabolism , Macrophages/immunology , Macrophages/microbiology , Streptococcus oralis/physiology , Animals , Catalase/metabolism , Cathepsin B/metabolism , Cell Death/drug effects , Cell Death/immunology , Deferoxamine/pharmacology , Hydrogen-Ion Concentration , Lysosomal-Associated Membrane Protein 1/metabolism , Macrophages/metabolism , Mice , Models, Biological , Streptococcal Infections/immunology , Streptococcal Infections/microbiology
11.
PLoS One ; 9(8): e103125, 2014.
Article in English | MEDLINE | ID: mdl-25084357

ABSTRACT

Streptococcus sanguinis, a member of the commensal mitis group of streptococci, is a primary colonizer of the tooth surface, and has been implicated in infectious complications including bacteremia and infective endocarditis. During disease progression, S. sanguinis may utilize various cell surface molecules to evade the host immune system to survive in blood. In the present study, we discovered a novel cell surface nuclease with a cell-wall anchor domain, termed SWAN (streptococcal wall-anchored nuclease), and investigated its contribution to bacterial resistance against the bacteriocidal activity of neutrophil extracellular traps (NETs). Recombinant SWAN protein (rSWAN) digested multiple forms of DNA including NET DNA and human RNA, which required both Mg(2+) and Ca(2+) for optimum activity. Furthermore, DNase activity of S. sanguinis was detected around growing colonies on agar plates containing DNA. In-frame deletion of the swan gene mostly reduced that activity. These findings indicated that SWAN is a major nuclease displayed on the surface, which was further confirmed by immuno-detection of SWAN in the cell wall fraction. The sensitivity of S. sanguinis to NET killing was reduced by swan gene deletion. Moreover, heterologous expression of the swan gene rendered a Lactococcus lactis strain more resistant to NET killing. Our results suggest that the SWAN nuclease on the bacterial surface contributes to survival in the potential situation of S. sanguinis encountering NETs during the course of disease progression.


Subject(s)
Cell Wall/enzymology , Extracellular Traps/immunology , Extracellular Traps/microbiology , Micrococcal Nuclease/metabolism , Neutrophils/immunology , Streptococcus sanguis/physiology , DNA, Viral/metabolism , Enzyme Activation , Extracellular Space/metabolism , Gene Order , Genetic Loci , Humans , Hydrolysis , Protein Transport , RNA, Viral/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Streptococcus sanguis/pathogenicity
12.
PLoS One ; 9(1): e88136, 2014.
Article in English | MEDLINE | ID: mdl-24498253

ABSTRACT

Members of the mitis group of streptococci are normal inhabitants of the commensal flora of the oral cavity and upper respiratory tract of humans. Some mitis group species, such as Streptococcus oralis and Streptococcus sanguinis, are primary colonizers of the human oral cavity. Recently, we found that hydrogen peroxide (H2O2) produced by S. oralis is cytotoxic to human macrophages, suggesting that streptococcus-derived H2O2 may act as a cytotoxin. Since epithelial cells provide a physical barrier against pathogenic microbes, we investigated their susceptibility to infection by H2O2-producing streptococci in this study. Infection by S. oralis and S. sanguinis was found to stimulate cell death of Detroit 562, Calu-3 and HeLa epithelial cell lines at a multiplicity of infection greater than 100. Catalase, an enzyme that catalyzes the decomposition of H2O2, inhibited S. oralis cytotoxicity, and H2O2 alone was capable of eliciting epithelial cell death. Moreover, S. oralis mutants lacking the spxB gene encoding pyruvate oxidase, which are deficient in H2O2 production, exhibited reduced cytotoxicity toward Detroit 562 epithelial cells. In addition, enzyme-linked immunosorbent assays revealed that both S. oralis and H2O2 induced interleukin-6 production in Detroit 562 epithelial cells. These results suggest that streptococcal H2O2 is cytotoxic to epithelial cells, and promotes bacterial evasion of the host defense systems in the oral cavity and upper respiratory tracts.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/metabolism , Hydrogen Peroxide/metabolism , Pyruvate Oxidase/metabolism , Streptococcal Infections/enzymology , Streptococcus oralis/enzymology , Streptococcus sanguis/enzymology , Cell Death , Cell Line , Epithelial Cells/pathology , Humans , Streptococcal Infections/pathology
13.
PLoS One ; 8(5): e62563, 2013.
Article in English | MEDLINE | ID: mdl-23658745

ABSTRACT

Hydrogen peroxide (H2O2) produced by members of the mitis group of oral streptococci plays important roles in microbial communities such as oral biofilms. Although the cytotoxicity of H2O2 has been widely recognized, the effects of H2O2 produced by oral streptococci on host defense systems remain unknown. In the present study, we investigated the effect of H2O2 produced by Streptococcus oralis on human macrophage cell death. Infection by S. oralis was found to stimulate cell death of a THP-1 human macrophage cell line at multiplicities of infection greater than 100. Catalase, an enzyme that catalyzes the decomposition of H2O2, inhibited the cytotoxic effect of S. oralis. S. oralis deletion mutants lacking the spxB gene, which encodes pyruvate oxidase, and are therefore deficient in H2O2 production, showed reduced cytotoxicity toward THP-1 macrophages. Furthermore, H2O2 alone was capable of inducing cell death. The cytotoxic effect seemed to be independent of inflammatory responses, because H2O2 was not a potent stimulator of tumor necrosis factor-α production in macrophages. These results indicate that streptococcal H2O2 plays a role as a cytotoxin, and is implicated in the cell death of infected human macrophages.


Subject(s)
Cytotoxins/pharmacology , Hydrogen Peroxide/pharmacology , Macrophages/drug effects , Streptococcus oralis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalase/metabolism , Catalase/pharmacology , Cell Death/drug effects , Cell Line , Cytotoxins/biosynthesis , Humans , Hydrogen Peroxide/metabolism , Macrophages/microbiology , Macrophages/pathology , Pyruvate Oxidase/deficiency , Pyruvate Oxidase/genetics , Streptococcus oralis/drug effects , Streptococcus oralis/pathogenicity , Tumor Necrosis Factor-alpha/biosynthesis
14.
Endocrinology ; 154(2): 773-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23291450

ABSTRACT

Periodontitis, an inflammatory disease of periodontal tissues, is characterized by excessive alveolar bone resorption. An increase in the receptor activator of nuclear factor-κB ligand (RANKL) to osteoprotegerin (OPG) ratio is thought to reflect the severity of periodontitis. Here, we examined alveolar bone loss in OPG-deficient (OPG(-/-)) mice and RANKL-overexpressing transgenic (RANKL-Tg) mice. Alveolar bone loss in OPG(-/-) mice at 12 weeks was significantly higher than that in RANKL-Tg mice. OPG(-/-) but not RANKL-Tg mice exhibited severe bone resorption especially in cortical areas of the alveolar bone. An increased number of osteoclasts was observed in the cortical areas in OPG(-/-) but not in RANKL-Tg mice. Immunohistochemical analyses showed many OPG-positive signals in osteocytes but not osteoblasts. OPG-positive osteocytes in the cortical area of alveolar bones and long bones were abundant in both wild-type and RANKL-Tg mice. This suggests the resorption in cortical bone areas to be prevented by OPG produced locally. To test the usefulness of OPG(-/-) mice as an animal model for screening drugs to prevent alveolar bone loss, we administered an antimouse RANKL antibody or risedronate, a bisphosphonate, to OPG(-/-) mice. They suppressed alveolar bone resorption effectively. OPG(-/-) mice are useful for screening therapeutic agents against alveolar bone loss.


Subject(s)
Mandibular Diseases/physiopathology , Alveolar Bone Loss , Animals , Male , Mice , Mice, Transgenic , Osteoclasts/metabolism , Osteoprotegerin , Periodontitis , RANK Ligand
15.
FEMS Microbiol Lett ; 323(2): 164-70, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22092716

ABSTRACT

Streptococcus sanguinis, a normal inhabitant of the human oral cavity, is a common streptococcal species implicated in infective endocarditis. Herein, we investigated the effects of infection with S. sanguinis on foam cell formation and cell death of macrophages. Infection with S. sanguinis stimulated foam cell formation of THP-1, a human macrophage cell line. At a multiplicity of infection >100, S. sanguinis-induced cell death of the macrophages. Viable bacterial infection was required to trigger cell death because heat-inactivated S. sanguinis did not induce cell death. The production of cytokines interleukin-1ß and tumor necrosis factor-α from macrophages was also stimulated during bacterial infection. Inhibition of the production of reactive oxygen species (ROS) resulted in reduced cell death, suggesting an association of ROS with cell death. Furthermore, S. sanguinis-induced cell death appeared to be independent of activation of inflammasomes, because cleavage of procaspase-1 was not evident in infected macrophages.


Subject(s)
Cell Death , Foam Cells , Macrophages/immunology , Macrophages/microbiology , Reactive Oxygen Species/toxicity , Streptococcus/pathogenicity , Cell Line , Humans , Interleukin-1beta/metabolism , Reactive Oxygen Species/metabolism , Streptococcus/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
J Biol Chem ; 286(43): 37566-77, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21880740

ABSTRACT

The human pathogen Streptococcus pyogenes produces diverse pili depending on the serotype. We investigated the assembly mechanism of FCT type 1 pili in a serotype M6 strain. The pili were found to be assembled from two precursor proteins, the backbone protein T6 and ancillary protein FctX, and anchored to the cell wall in a manner that requires both a housekeeping sortase enzyme (SrtA) and pilus-associated sortase enzyme (SrtB). SrtB is primarily required for efficient formation of the T6 and FctX complex and subsequent polymerization of T6, whereas proper anchoring of the pili to the cell wall is mainly mediated by SrtA. Because motifs essential for polymerization of pilus backbone proteins in other Gram-positive bacteria are not present in T6, we sought to identify the functional residues involved in this process. Our results showed that T6 encompasses the novel VAKS pilin motif conserved in streptococcal T6 homologues and that the lysine residue (Lys-175) within the motif and cell wall sorting signal of T6 are prerequisites for isopeptide linkage of T6 molecules. Because Lys-175 and the cell wall sorting signal of FctX are indispensable for substantial incorporation of FctX into the T6 pilus shaft, FctX is suggested to be located at the pilus tip, which was also implied by immunogold electron microscopy findings. Thus, the elaborate assembly of FCT type 1 pili is potentially organized by sortase-mediated cross-linking between sorting signals and the amino group of Lys-175 positioned in the VAKS motif of T6, thereby displaying T6 and FctX in a temporospatial manner.


Subject(s)
Cell Wall/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Protein Multimerization/physiology , Streptococcus pyogenes/metabolism , Amino Acid Motifs , Base Sequence , Cell Wall/genetics , Cell Wall/ultrastructure , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Humans , Molecular Sequence Data , Streptococcus pyogenes/genetics , Streptococcus pyogenes/ultrastructure
17.
Clin Vaccine Immunol ; 18(9): 1552-61, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21795464

ABSTRACT

In the development of a component vaccine against caries, the catalytic region (CAT) and glucan-binding domain (GBD) of glucosyltransferase B (GtfB) from Streptococcus mutans have been employed as target antigens. These regions were adopted as primary targets because they theoretically include epitopes associated with enzyme function. However, their antigenicities have not been fully evaluated. Although there are many reports about successful vaccination using these components, the principle has not yet been put to practical use. For these reasons, we came to doubt the effectiveness of the epitopes in vaccine production and reevaluated the antigenic region of GtfB by using in silico analyses combined with in vitro and in vivo experiments. The results suggested that the ca. 360-amino-acid variable region (VR) in the N terminus of GtfB is more reactive than CAT and GBD. This region is S. mutans and/or GtfB specific, nonconserved among other streptococcal Gtfs, and of unknown function. Immunization using an adenovirus vector-borne DNA vaccine confirmed that VR is an epitope that shows promise for the development of a caries vaccine.


Subject(s)
Antigens, Bacterial , Epitopes , Glucosyltransferases , Streptococcus mutans/enzymology , Adenoviridae/genetics , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Catalytic Domain/immunology , Cell Line , Dental Caries/prevention & control , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Female , Genetic Vectors , Glucans/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/immunology , Humans , Immunization , Mice , Streptococcus mutans/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/chemistry , Vaccines, DNA/genetics , Vaccines, DNA/immunology
18.
Microb Pathog ; 50(3-4): 148-54, 2011.
Article in English | MEDLINE | ID: mdl-21238567

ABSTRACT

Streptococcus sanguinis is a member of oral streptococci and one of the most abundant species found in oral biofilm called dental plaque. Colonization of the oral streptococci on the tooth surface depends on the adhesion of bacteria to salivary components adsorbed to the tooth surface. Recently, we identified unique cell surface long filamentous structures named pili in this species. Herein, we investigated the role of S. sanguinis pili in biofilm formation. We found that pili-deficient mutant, in which the genes encoding the three pilus proteins PilA, PilB and PilC have been deleted, showed an impaired bacterial accumulation on saliva-coated surfaces. Confocal microscopic observations suggested that the mutant was incapable of producing typical three-dimensional layer of biofilm. Ligand blot analysis showed that the ancillary pilus proteins PilB and PilC bound to human whole saliva. Additional analysis demonstrated that PilC bound to multiple salivary components, and one of which was found to be salivary α-amylase. These results indicate that pilus proteins are members of saliva-binding proteins of oral S. sanguinis, and suggest the involvement of pili in its colonization on saliva-coated tooth surfaces and in the human oral cavity.


Subject(s)
Amylases/metabolism , Biofilms , Fimbriae, Bacterial/metabolism , Saliva/enzymology , Streptococcal Infections/enzymology , Streptococcus sanguis/physiology , Amylases/genetics , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/genetics , Humans , Mouth/enzymology , Mouth/microbiology , Protein Binding , Saliva/microbiology , Streptococcal Infections/microbiology , Streptococcus sanguis/genetics
19.
J Biol Chem ; 285(29): 22666-75, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20472552

ABSTRACT

Streptococcus pyogenes (group A streptococcus (GAS)) is a pathogen that invades non-phagocytic host cells, and causes a variety of acute infections such as pharyngitis. Our group previously reported that intracellular GAS is effectively degraded by the host-cell autophagic machinery, and that a cholesterol-dependent cytolysin, streptolysin O (SLO), is associated with bacterial escape from endosomes in epithelial cells. However, the details of both the intracellular behavior of GAS and the process leading to its autophagic degradation remain unknown. In this study, we found that two host small G proteins, Rab5 and Rab7, were associated with the pathway of autophagosome formation and the fate of intracellular GAS. Rab5 was involved in bacterial invasion and endosome fusion. Rab7 was clearly multifunctional, with roles in bacterial invasion, endosome maturation, and autophagosome formation. In addition, this study showed that the bacterial cytolysin SLO supported the escape of GAS into the cytoplasm from endosomes, and surprisingly, a SLO-deficient mutant of GAS was viable longer than the wild-type strain although it failed to escape the endosomes. This intracellular behavior of GAS is unique and distinct from that of other types of bacterial invaders. Our results provide a new picture of GAS infection and host-cell responses in epithelial cells.


Subject(s)
Autophagy , Intracellular Space/microbiology , Streptolysins/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Bacterial Proteins/metabolism , Biomarkers/metabolism , Endosomes/microbiology , Endosomes/ultrastructure , HeLa Cells , Humans , Intracellular Space/metabolism , Lysosomes/microbiology , Lysosomes/ultrastructure , Microbial Viability , Models, Biological , Mutant Proteins/metabolism , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/ultrastructure , Protein Transport , Streptococcus pyogenes/cytology , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/ultrastructure , Time Factors , rab7 GTP-Binding Proteins
20.
Biochem Biophys Res Commun ; 391(2): 1192-6, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-20004645

ABSTRACT

Streptococcus sanguinis is a predominant bacterium in the human oral cavity and occasionally causes infective endocarditis. We identified a unique cell surface polymeric structure named pili in this species and investigated its functions in regard to its potential virulence. Pili of S. sanguinis strain SK36 were shown to be composed of three distinctive pilus proteins (PilA, PilB, and PilC), and a pili-deficient mutant demonstrated reduced bacterial adherence to HeLa and human oral epithelial cells. PilC showed a binding ability to fibronectin, suggesting that pili are involved in colonization by this species. In addition, ATCC10556, a standard S. sanguinis strain, was unable to produce pili due to defective pilus genes, which indicates a diversity of pilus expression among various S. sanguinis strains.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/metabolism , Fibronectins/metabolism , Fimbriae, Bacterial/metabolism , Mouth/microbiology , Streptococcus sanguis/pathogenicity , Bacterial Proteins/genetics , Fibronectins/blood , Fimbriae, Bacterial/genetics , Humans , Streptococcus sanguis/genetics , Streptococcus sanguis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...