Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Sci Rep ; 11(1): 23954, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907255

ABSTRACT

To investigate the effect of Er:YAG laser treatment on lipopolysaccharide (LPS) clearance and fibroblast adhesion on titanium disks. Grade IV titanium discs (n = 216) were used and allocated to 6 groups. Group 1 was the negative control without Porphyromonas gingivalis inoculation. Discs in Groups 2-6 were incubated with P. gingivalis to form a biofilm. Group 3 received 0.12% chlorhexidine irrigation and Group 4 received titanium curettage to remove the biofilm. Group 5 was treated with Er:YAG laser irradiation and Group 6 was treated with titanium curettage plus Er:YAG laser irradiation. The contact angle and surface roughness were measured after the various treatments. The surface microstructure and residual bacteria were examined using scanning electron microscopy and confocal laser scanning microscopy, respectively. Residual LPS was examined using a limulus amoebocyte lysate assay and human gingival fibroblast adhesion was quantified using fluorescent microscopy. Curettage plus Er:YAG laser irradiation was the most effective method for removing bacteria and LPS. No significant difference in the amount of fibroblast adhesion was found between the control and Group 6. Combined use of Er:YAG laser irradiation and curettage optimizes LPS clearance and fibroblast adhesion on titanium discs.


Subject(s)
Biofilms/growth & development , Disinfection , Fibroblasts/metabolism , Gingiva/metabolism , Lipopolysaccharides/chemistry , Porphyromonas gingivalis/physiology , Titanium/chemistry , Fibroblasts/ultrastructure , Humans , Lasers, Solid-State , Microscopy, Electron, Scanning , Porphyromonas gingivalis/ultrastructure
2.
Sci Rep ; 10(1): 18313, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110205

ABSTRACT

Porphyromonas gingivalis is a causative agent in the onset and progression of periodontal disease. This study aims to investigate the effects of quercetin, a natural plant product, on P. gingivalis virulence properties including gingipain, haemagglutinin and biofilm formation. Antimicrobial effects and morphological changes of quercetin on P. gingivalis were detected. The effects of quercetin on gingipains activities and hemolytic, hemagglutination activities were evaluated using chromogenic peptides and sheep erythrocytes. The biofilm biomass and metabolism with different concentrations of quercetin were assessed by the crystal violet and MTT assay. The structures and thickness of the biofilms were observed by confocal laser scanning microscopy. Bacterial cell surface properties including cell surface hydrophobicity and aggregation were also evaluated. The mRNA expression of virulence and iron/heme utilization was assessed using real time-PCR. Quercetin exhibited antimicrobial effects and damaged the cell structure. Quercetin can inhibit gingipains, hemolytic, hemagglutination activities and biofilm formation at sub-MIC concentrations. Molecular docking analysis further indicated that quercetin can interact with gingipains. The biofilm became sparser and thinner after quercetin treatment. Quercetin also modulate cell surface hydrophobicity and aggregation. Expression of the genes tested was down-regulated in the presence of quercetin. In conclusion, our study demonstrated that quercetin inhibited various virulence factors of P. gingivalis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Periodontal Diseases/microbiology , Porphyromonas gingivalis/drug effects , Quercetin/pharmacology , Biofilms/drug effects , Gingipain Cysteine Endopeptidases/metabolism , Hemagglutinins/metabolism , Humans , Microbial Sensitivity Tests , Microscopy, Confocal , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Porphyromonas gingivalis/ultrastructure , Virulence/drug effects
3.
Sci Rep ; 10(1): 7468, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366945

ABSTRACT

Recent epidemiological  studies link Periodontal disease(PD) to age-related macular degeneration (AMD). We documented earlier that Porphyromonas gingivalis(Pg), keystone oral-pathobiont, causative of PD, efficiently invades human gingival epithelial and blood-dendritic cells. Here, we investigated the ability of dysbiotic Pg-strains to invade human-retinal pigment epithelial cells(ARPE-19), their survival, intracellular localization, and the pathological effects, as dysfunction of RPEs leads to AMD. We show that live, but not heat-killed Pg-strains adhere to and invade ARPEs. This involves early adhesion to ARPE cell membrane, internalization and localization of Pg within single-membrane vacuoles or cytosol, with some nuclear localization apparent. No degradation of Pg or localization inside double-membrane autophagosomes was evident, with dividing Pg suggesting a metabolically active state during invasion. We found significant downregulation of autophagy-related genes particularly, autophagosome complex. Antibiotic protection-based recovery assay further confirmed distinct processes of adhesion, invasion and amplification of Pg within ARPE cells. This is the first study to demonstrate invasion of human-RPEs, begin to characterize intracellular localization and survival of Pg within these cells. Collectively, invasion of RPE by Pg and its prolonged survival by autophagy evasion within these cells suggest a strong rationale for studying the link between oral infection and AMD pathogenesis in individuals with periodontitis.


Subject(s)
Autophagosomes , Autophagy , Bacteroidaceae Infections , Cytosol , Porphyromonas gingivalis , Retinal Pigment Epithelium , Vacuoles , Autophagosomes/metabolism , Autophagosomes/microbiology , Autophagosomes/ultrastructure , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Cell Line , Cytosol/metabolism , Cytosol/microbiology , Cytosol/ultrastructure , Humans , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/ultrastructure , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/microbiology , Retinal Pigment Epithelium/ultrastructure , Vacuoles/microbiology , Vacuoles/pathology , Vacuoles/ultrastructure
4.
Biomed Res Int ; 2019: 3547858, 2019.
Article in English | MEDLINE | ID: mdl-31886204

ABSTRACT

Porphyromonas gingivalis is an important pathogenic anaerobic bacterium that causes aspiration pneumonia. This bacterium frequently forms biofilms in the oral cavity and in respiratory tract-associated medical devices. Bacterial colonization that occurs in association with this biofilm formation is the main reason for incurable aspiration pneumonia. The Lonicera caerulea var. emphyllocalyx (LCE) fruit has been used in folk medicine in Hokkaido, the northern part of Japan. The aim of this study was to elucidate one of the antimicrobial mechanisms of LCE methanol extract (LCEE)-the inhibitory effect of LCEE on biofilm formation by P. gingivalis. Our results show that LCEE significantly reduced biofilm formation by three different P. gingivalis isolates in a concentration- and time-dependent manner that were quantified by the adsorption of safranin red. When LCEE was added to biofilms already formed by P. gingivalis, LCEE did not degrade the biofilm. However, treatment with LCEE significantly promoted the removal of existing biofilm by vibration compared to that of control. We also confirmed biofilm formation in LCEE-treated P. gingivalis in tracheal tubes using scanning electron microscopic (SEM) analysis. Cyanidin 3-O-glucoside (C3G), one of the components of LCE, also inhibited the formation of biofilm by P. gingivalis in a concentration-dependent manner. Our results reveal that LCEE may be an effective antibacterial substance for P. gingivalis-induced aspiration pneumonia because of its role in the suppression of bacterial biofilm formation in the oral cavity.


Subject(s)
Anti-Bacterial Agents , Biofilms , Fruit/chemistry , Lonicera/chemistry , Plant Extracts , Porphyromonas gingivalis/physiology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Dose-Response Relationship, Drug , Plant Extracts/chemistry , Plant Extracts/pharmacology , Porphyromonas gingivalis/ultrastructure
5.
Sci Rep ; 9(1): 4935, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894633

ABSTRACT

Porphyromonas gingivalis is a member of the dysbiotic oral microbiome and a "keystone pathogen" that causes severe periodontal disease, which is among the most prevalent infectious diseases. Part of the virulence factors secreted by P. gingivalis are the essential cysteine peptidases gingipain K (Kgp) and R (RgpA and RgpB), which account for 85% of the extracellular proteolytic activity of the pathogen and are thus prime targets for inhibition. We report the high-resolution (1.20 Å) complex structure of Kgp with KYT-36, a peptide-derived, potent, bioavailable and highly selective inhibitor, which is widely used for studies in vitro, in cells and in vivo. Sub-nanomolar inhibition of Kgp is achieved by tight binding to the active-site cleft, which is covered for its sub-sites S3 through S1' under establishment of nine hydrophobic interactions, 14 hydrogen bonds and one salt bridge. In addition, an inhibitor carbonyl carbon that mimics the scissile carbonyl of substrates is pyramidalized and just 2.02 Å away from the catalytic nucleophile of Kgp, C477Sγ. Thus, the crystal structure emulates a reaction intermediate of the first nucleophilic attack during catalysis of cysteine peptidases. The present study sets the pace for the development of tailored next-generation drugs to tackle P. gingivalis.


Subject(s)
Bacteroidaceae Infections/drug therapy , Benzylamines/chemistry , Carbamates/chemistry , Gingipain Cysteine Endopeptidases/antagonists & inhibitors , Hydrazines/chemistry , Periodontitis/drug therapy , Porphyromonas gingivalis/ultrastructure , Protease Inhibitors/chemistry , Bacteroidaceae Infections/microbiology , Benzylamines/pharmacology , Benzylamines/therapeutic use , Carbamates/pharmacology , Carbamates/therapeutic use , Catalytic Domain/drug effects , Crystallography, X-Ray , Drug Development , Gingipain Cysteine Endopeptidases/metabolism , Gingipain Cysteine Endopeptidases/ultrastructure , Hydrazines/pharmacology , Hydrazines/therapeutic use , Hydrophobic and Hydrophilic Interactions , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Protein Domains , Structure-Activity Relationship , Virulence Factors/antagonists & inhibitors , Virulence Factors/metabolism
6.
Biochem Biophys Res Commun ; 509(2): 335-340, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30579592

ABSTRACT

Porphyromonas gingivalis is one of the most commonly detected pathogens in periodontal disease and root canal infections. Its viability and pathogenicity are greatly increased in plaque biofilms. Some caseinolytic proteases (Clp) reportedly regulate biofilm formation by various pathogenic bacteria, including P. gingivalis. However, the specific influence of ClpP and its mechanism of regulating biofilm formation by P. gingivalis remains unclear. Hence, in this study, a clpP deletion strain and complemented strain were constructed by homologous recombination, and an in vitro biofilm model was established. Biofilm architecture was observed by scanning electron microscopy. Bacterial cells within the biofilms were examined using confocal scanning laser microscopy. Crystal violet staining was used to determine the amount of formed biofilm. mRNA levels of related regulatory genes were assessed using real-time PCR. The clpP deletion and complemented strains of P. gingivalis were successfully constructed. The biofilm formation ability of the deletion strain was significantly reduced compared with that of the wild-type strain, while that of the complemented strain did not differ from that of the wild-type strain. The expression of fimA, mfa1, and luxS in the deletion strain was lower than in the wild-type and complemented strains at each timepoint. It can be concluded that ClpP increases the biofilm formation of P. gingivalis by regulating the expression levels of fimA, mfa1, and luxS.


Subject(s)
Bacterial Proteins/genetics , Biofilms/growth & development , Carbon-Sulfur Lyases/genetics , Endopeptidase Clp/genetics , Fimbriae Proteins/genetics , Gene Expression Regulation, Bacterial , Porphyromonas gingivalis/genetics , Bacterial Proteins/metabolism , Carbon-Sulfur Lyases/metabolism , Endopeptidase Clp/deficiency , Fimbriae Proteins/metabolism , Gene Deletion , Gentian Violet , Homologous Recombination , Microscopy, Electron, Scanning , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/ultrastructure
7.
Anaerobe ; 55: 107-111, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30423437

ABSTRACT

It has been reported that sub-minimal inhibitory concentrations (sub-MICs) of antibiotics are capable of altering bacterial surface properties and phenotype. In this study, the effects of sub-MICs of certain antibiotics on surface hydrophobicity, cell morphology, and protein profile were ascertained using Fusobacterium nucleatum, Porphyromonas gingivalis and Treponema denticola strains, which are pathogenic bacterial species in periodontal diseases. The MICs of antibiotics were determined by culturing bacteria in media supplemented with serially diluted antibiotic solutions, and sub-MIC of antibiotics was used. The effect of sub-MIC of antibiotics on cell morphology was determined by scanning electron microscopy. Microscopic observation of F. nucleatum and P. gingivalis grown at a sub-MIC of amoxicillin revealed cell enlargement. T. denticola grown at a sub-MIC of doxycycline also showed cell elongation. The relative surface hydrophobicity determined by measuring the ability of the bacteria to absorb n-hexadecane revealed an increase in surface hydrophobicity of F. nucleatum grown at sub-MIC of penicillin and amoxicillin, but a decrease with metronidazole; whereas increased hydrophobicity was observed in T. denticola grown at sub-MIC of doxycycline, metronidazole and tetracycline. The surface hydrophobicity of P. gingivalis increased only when grown in sub-MIC of metronidazole. The protein expression profile of the treated bacteria differed from their respective controls. These results confirmed that sub-MIC concentrations of antibiotics can affect the phenotype, surface properties and morphology of periodontal pathogenic anaerobic bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fusobacterium nucleatum/drug effects , Porphyromonas gingivalis/drug effects , Surface Properties/drug effects , Treponema denticola/drug effects , Bacteria, Anaerobic/chemistry , Bacteria, Anaerobic/drug effects , Bacteria, Anaerobic/ultrastructure , Fusobacterium nucleatum/chemistry , Fusobacterium nucleatum/ultrastructure , Humans , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/ultrastructure , Proteome/analysis , Treponema denticola/chemistry , Treponema denticola/ultrastructure
8.
Sci Rep ; 8(1): 14914, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297793

ABSTRACT

Porphyromonas gingivalis-induced inflammatory effects are mostly investigated in monolayer cultured cells. The aim of this study was to develop a 3D spheroid model of gingiva to take into account epithelio-fibroblastic interactions. Human gingival epithelial cells (ECs) and human oral fibroblasts (FBs) were cultured by hanging drop method to generate 3D microtissue (MT) whose structure was analyzed on histological sections and the cell-to-cell interactions were observed by scanning and transmission electron microscopy (SEM and TEM). MTs were infected by P. gingivalis and the impact on cell death (Apaf-1, caspase-3), inflammatory markers (TNF-α, IL-6, IL-8) and extracellular matrix components (Col-IV, E-cadherin, integrin ß1) was evaluated by immunohistochemistry and RT-qPCR. Results were compared to those observed in situ in experimental periodontitis and in human gingival biopsies. MTs exhibited a well-defined spatial organization where ECs were organized in an external cellular multilayer, while, FBs constituted the core. The infection of MT demonstrated the ability of P. gingivalis to bypass the epithelial barrier in order to reach the fibroblastic core and induce disorganization of the spheroid structure. An increased cell death was observed in fibroblastic core. The development of such 3D model may be useful to define the role of EC-FB interactions on periodontal host-immune response and to assess the efficacy of new therapeutics.


Subject(s)
Epithelial Cells/pathology , Fibroblasts/microbiology , Fibroblasts/pathology , Inflammation/microbiology , Inflammation/pathology , Models, Biological , Porphyromonas gingivalis/physiology , Spheroids, Cellular/pathology , Adult , Apoptosis/genetics , Epithelial Cells/ultrastructure , Female , Fibroblasts/ultrastructure , Gene Expression Regulation , Gingiva/pathology , Humans , Male , Middle Aged , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/ultrastructure
9.
J Proteome Res ; 17(7): 2377-2389, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29766714

ABSTRACT

Porphyromonas gingivalis is an anaerobic, Gram-negative oral pathogen associated with chronic periodontitis. P. gingivalis has an obligate requirement for heme, which it obtains from the host. Heme availability has been linked to disease initiation and progression. In this study we used continuous culture of the bacterium to determine the effect of heme limitation and excess on the P. gingivalis proteome. Four biological replicates of whole cell lysate (WCL) and outer membrane vesicle (OMV) samples were digested with trypsin and analyzed by tandem mass spectrometry and MaxQuant label-free quantification. In total, 1211 proteins were quantified, with 108 and 49 proteins significantly changing in abundance more than 1.5-fold ( p < 0.05) in the WCLs and OMVs, respectively. The proteins most upregulated in response to heme limitation were those involved in binding and transporting heme, whereas the four proteins most upregulated under the heme-excess condition constitute a putative heme efflux system. In general, the protein abundance ratios obtained for OMVs and WCLs agreed, indicating that changes to the OM protein composition are passed onto OMVs; however, 16 proteins were preferentially packaged into OMVs under one condition more than the other. In particular, moonlighting cytoplasmic proteins were preferentially associated with OMVs under heme excess.


Subject(s)
Cell-Derived Microparticles/chemistry , Gene Expression Regulation, Bacterial/drug effects , Heme/pharmacology , Porphyromonas gingivalis/chemistry , Proteome/metabolism , Bacterial Outer Membrane Proteins , Cell-Derived Microparticles/drug effects , Heme/analysis , Porphyromonas gingivalis/cytology , Porphyromonas gingivalis/ultrastructure , Proteome/drug effects
10.
Microb Pathog ; 116: 26-32, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29325862

ABSTRACT

The objective of this study was to investigate the antibacterial effects of cinnamon (Cinnamomum zeylanicum) bark essential oil (CBEO) and its principal constituent cinnamaldehyde against Porphyromonas gingivalis and to elucidate the antibacterial mechanism. GC-MS analysis showed that cinnamaldehyde was the major constituent in CBEO (57.97%). The minimum inhibition concentrations (MICs) of CBEO and cinnamaldehyde were 6.25 µg/mL and 2.5 µM for P. gingivalis, respectively. Nucleic acid and protein leakage was observed with increasing concentrations of CBEO and cinnamaldehyde. Additionally, propidium iodide uptake assays revealed CBEO and cinnamaldehyde at 1 × MIC impaired P. gingivalis membrane integrity by enhancing cell permeability. Morphological changes in P. gingivalis cells were observed by scanning electron microscopy, which indicated cell membrane destruction. To further determine the anti-biofilm effect, relative biofilm formation and established biofilms were examined, which demonstrated that both CBEO and cinnamaldehyde at sub-MIC levels inhibited P. gingivalis biofilm formation by 74.5% and 67.3% separately, but only CBEO slightly decreased established biofilms by 33.5% at 4 × MIC. These results suggest the potential of CBEO as a natural antimicrobial agent against periodontal disease. Furthermore, cinnamaldehyde was confirmed to be the antibacterial substance of CBEO with inhibitory action against P. gingivalis.


Subject(s)
Acrolein/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Cinnamomum zeylanicum/chemistry , Oils, Volatile/pharmacology , Porphyromonas gingivalis/drug effects , Acrolein/isolation & purification , Acrolein/pharmacology , Anti-Bacterial Agents/isolation & purification , Cell Membrane/drug effects , Cell Membrane/physiology , Gas Chromatography-Mass Spectrometry , Humans , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Oils, Volatile/isolation & purification , Permeability/drug effects , Plant Bark/chemistry , Porphyromonas gingivalis/ultrastructure
11.
Sci Rep ; 7(1): 1413, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28469253

ABSTRACT

Dental plaque is a complex multispecies biofilm, and is a direct precursor of periodontal disease. The virulence of periodontal pathogens, such as Porphyromonas gingivalis, is expressed in the context of this polymicrobial community. Previously, we reported an antagonistic relationship between Streptococcus cristatus and P. gingivalis, and identified arginine deiminase (ArcA) of S. cristatus as the signaling molecule to which P. gingivalis responds by repressing the expression and production of FimA protein. Here we demonstrate that direct interaction between P. gingivalis and S. cristatus is necessary for the cell-cell communication. Two surface proteins of P. gingivalis, PGN_0294 and PGN_0806, were found to interact with S. cristatus ArcA. Using a peptide array analysis, we identified several P. gingivalis-binding sites of ArcA, which led to the discovery of an 11-mer peptide with the native sequence of ArcA that repressed expression of fimbriae and of gingipains. These data indicate that a functional motif of ArcA is sufficient to selectively alter virulence gene expression in P. gingivalis, and PGN_0294 and PGN_0806 may serve as receptors for ArcA. Our findings provide a molecular basis for future rational design of agents that interfere with the initiation and formation of a P. gingivalis-induced pathogenic community.


Subject(s)
Gene Expression Regulation, Bacterial , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/pathogenicity , Streptococcus/genetics , Streptococcus/pathogenicity , Bacterial Proteins/isolation & purification , Cell Communication , Membrane Proteins/isolation & purification , Porphyromonas gingivalis/ultrastructure , Virulence
12.
Article in English | MEDLINE | ID: mdl-27993858

ABSTRACT

The spread of antibiotic resistance and the challenges associated with antiseptics such as chlorhexidine have necessitated a search for new antibacterial agents against oral bacterial pathogens. As a result of failing traditional approaches, drug repurposing has emerged as a novel paradigm to find new antibacterial agents. In this study, we examined the effects of the FDA-approved anticancer agent toremifene against the oral bacteria Porphyromonas gingivalis and Streptococcus mutans We found that the drug was able to inhibit the growth of both pathogens, as well as prevent biofilm formation, at concentrations ranging from 12.5 to 25 µM. Moreover, toremifene was shown to eradicate preformed biofilms at concentrations ranging from 25 to 50 µM. In addition, we found that toremifene prevents P. gingivalis and S. mutans biofilm formation on titanium surfaces. A time-kill study indicated that toremifene is bactericidal against S. mutans Macromolecular synthesis assays revealed that treatment with toremifene does not cause preferential inhibition of DNA, RNA, or protein synthesis pathways, indicating membrane-damaging activity. Biophysical studies using fluorescent probes and fluorescence microscopy further confirmed the membrane-damaging mode of action. Taken together, our results suggest that the anticancer agent toremifene is a suitable candidate for further investigation for the development of new treatment strategies for oral bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Biofilms/drug effects , Cell Membrane/drug effects , Porphyromonas gingivalis/drug effects , Streptococcus mutans/drug effects , Toremifene/pharmacology , Biofilms/growth & development , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Membrane Permeability/drug effects , Dental Plaque/drug therapy , Dental Plaque/microbiology , Drug Repositioning , Drug Resistance, Multiple, Bacterial/physiology , Humans , Microbial Sensitivity Tests , Periodontitis/drug therapy , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/ultrastructure , Streptococcus mutans/metabolism , Streptococcus mutans/ultrastructure , Titanium/analysis
13.
PLoS Pathog ; 12(8): e1005820, 2016 08.
Article in English | MEDLINE | ID: mdl-27509186

ABSTRACT

The type IX secretion system (T9SS) has been recently discovered and is specific to Bacteroidetes species. Porphyromonas gingivalis, a keystone pathogen for periodontitis, utilizes the T9SS to transport many proteins including the gingipain virulence factors across the outer membrane and attach them to the cell surface via a sortase-like mechanism. At least 11 proteins have been identified as components of the T9SS including PorK, PorL, PorM, PorN and PorP, however the precise roles of most of these proteins have not been elucidated and the structural organization of these components is unknown. In this study, we purified PorK and PorN complexes from P. gingivalis and using electron microscopy we have shown that PorN and the PorK lipoprotein interact to form a 50 nm diameter ring-shaped structure containing approximately 32-36 subunits of each protein. The formation of these rings was dependent on both PorK and PorN, but was independent of PorL, PorM and PorP. PorL and PorM were found to form a separate stable complex. PorK and PorN were protected from proteinase K cleavage when present in undisrupted cells, but were rapidly degraded when the cells were lysed, which together with bioinformatic analyses suggests that these proteins are exposed in the periplasm and anchored to the outer membrane via the PorK lipid. Chemical cross-linking and mass spectrometry analyses confirmed the interaction between PorK and PorN and further revealed that they interact with the PG0189 outer membrane protein. Furthermore, we established that PorN was required for the stable expression of PorK, PorL and PorM. Collectively, these results suggest that the ring-shaped PorK/N complex may form part of the secretion channel of the T9SS. This is the first report showing the structural organization of any T9SS component.


Subject(s)
Bacterial Proteins/ultrastructure , Bacterial Secretion Systems/ultrastructure , Porphyromonas gingivalis/ultrastructure , Amino Acid Sequence , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Electrophoresis, Polyacrylamide Gel , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Microscopy, Electron, Transmission , Models, Molecular , Porphyromonas gingivalis/metabolism
14.
Sci Rep ; 6: 29985, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27457788

ABSTRACT

Formation of bacterial biofilms on dental implant material surfaces (titanium) may lead to the development of peri-implant diseases influencing the long term success of dental implants. In this study, a novel Cu-bearing titanium alloy (Ti-Cu) was designed and fabricated in order to efficiently kill bacteria and discourage formation of biofilms, and then inhibit bacterial infection and prevent implant failure, in comparison with pure Ti. Results from biofilm based gene expression studies, biofilm growth observation, bacterial viability measurements and morphological examination of bacteria, revealed antimicrobial/antibiofilm activities of Ti-Cu alloy against the oral specific bacterial species, Streptococcus mutans and Porphyromonas gingivalis. Proliferation and adhesion assays with mesenchymal stem cells, and measurement of the mean daily amount of Cu ion release demonstrated Ti-Cu alloy to be biocompatible. In conclusion, Ti-Cu alloy is a promising dental implant material with antimicrobial/antibiofilm activities and acceptable biocompatibility.


Subject(s)
Alloys/pharmacology , Anti-Bacterial Agents/pharmacology , Copper/pharmacology , Porphyromonas gingivalis/drug effects , Streptococcus mutans/drug effects , Animals , Biofilms/drug effects , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Fluorescence , Gene Expression Regulation, Bacterial/drug effects , Imaging, Three-Dimensional , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Microbial Sensitivity Tests , Models, Biological , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/ultrastructure , Rats , Streptococcus mutans/genetics , Streptococcus mutans/ultrastructure
15.
Mol Oral Microbiol ; 31(5): 365-78, 2016 10.
Article in English | MEDLINE | ID: mdl-26466922

ABSTRACT

Outer membrane vesicles (OMVs) are asymmetrical single bilayer membranous nanostructures produced by Gram-negative bacteria important for bacterial interaction with the environment. Porphyromonas gingivalis, a keystone pathogen associated with chronic periodontitis, produces OMVs that act as a virulence factor secretion system contributing to its pathogenicity. Despite their biological importance, the mechanisms of OMV biogenesis have not been fully elucidated. The ~14 times more curvature of the OMV membrane than cell outer membrane (OM) indicates that OMV biogenesis requires energy expenditure for significant curvature of the OMV membrane. In P. gingivalis, we propose that this may be achieved by upregulating the production of certain inner or outer leaflet lipids, which causes localized outward curvature of the OM. This results in selection of anionic lipopolysaccharide (A-LPS) and associated C-terminal domain (CTD) -family proteins on the outer surface due to their ability to accommodate the curvature. Deacylation of A-LPS may further enable increased curvature leading to OMV formation. Porphyromonas gingivalis OMVs that are selectively enriched in CTD-family proteins, largely the gingipains, can support bacterial coaggregation, promote biofilm development and act as an intercessor for the transport of non-motile bacteria by motile bacteria. The P. gingivalis OMVs are also believed to contribute to host interaction and colonization, evasion of immune defense mechanisms, and destruction of periodontal tissues. They may be crucial for both micro- and macronutrient capture, especially heme and probably other assimilable compounds for its own benefit and that of the wider biofilm community.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/ultrastructure , Porphyromonas gingivalis/physiology , Porphyromonas gingivalis/ultrastructure , Adhesins, Bacterial , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Biofilms , Cell Membrane/chemistry , Cell Membrane/immunology , Cell Membrane/metabolism , Cysteine Endopeptidases , Gingipain Cysteine Endopeptidases , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/pathogenicity , Virulence Factors
16.
PLoS One ; 10(10): e0139454, 2015.
Article in English | MEDLINE | ID: mdl-26437277

ABSTRACT

Porphyromonas gingivalis, a gram-negative obligate anaerobic bacterium, is considered to be a key pathogen in periodontal disease. The bacterium expresses Mfa1 fimbriae, which are composed of polymers of Mfa1. The minor accessory components Mfa3, Mfa4, and Mfa5 are incorporated into these fimbriae. In this study, we characterized Mfa4 using genetically modified strains. Deficiency in the mfa4 gene decreased, but did not eliminate, expression of Mfa1 fimbriae. However, Mfa3 and Mfa5 were not incorporated because of defects in posttranslational processing and leakage into the culture supernatant, respectively. Furthermore, the mfa4-deficient mutant had an increased tendency to auto-aggregate and form biofilms, reminiscent of a mutant completely lacking Mfa1. Notably, complementation of mfa4 restored expression of structurally intact and functional Mfa1 fimbriae. Taken together, these results indicate that the accessory proteins Mfa3, Mfa4, and Mfa5 are necessary for assembly of Mfa1 fimbriae and regulation of auto-aggregation and biofilm formation of P. gingivalis. In addition, we found that Mfa3 and Mfa4 are processed to maturity by the same RgpA/B protease that processes Mfa1 subunits prior to polymerization.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Proteins/metabolism , Biofilms , Fimbriae Proteins/physiology , Fimbriae, Bacterial/physiology , Porphyromonas gingivalis/physiology , Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Fimbriae Proteins/deficiency , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Genes, Bacterial , Genetic Complementation Test , Gingipain Cysteine Endopeptidases , Multigene Family , Mutagenesis , Organelle Biogenesis , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/ultrastructure , Protein Processing, Post-Translational , Species Specificity
17.
Future Microbiol ; 10(9): 1517-27, 2015.
Article in English | MEDLINE | ID: mdl-26343879

ABSTRACT

Porphyromonas gingivalis is one of the keystone pathogens associated with chronic periodontitis. All P. gingivalis strains examined thus far produce outer membrane vesicles. Recent studies have found that vesicles possess some well-known virulence factors of P. gingivalis such as adhesins, toxins and proteolytic enzymes. Carrying most of the characteristic features of their parent P. gingivalis cells, vesicles communicate with host cells and other members of microbial biofilms, resulting in the transmission of virulence factors into these host cells and the formation of pathogenic bacteria-dominated microbial communities. An in-depth understanding of both the nature and role of vesicles in the pathogenicity of P. gingivalis is both important and timely, particularly when speaking of periodontitis and its related systemic effects.


Subject(s)
Periodontitis/microbiology , Porphyromonas gingivalis/cytology , Porphyromonas gingivalis/physiology , Transport Vesicles/physiology , Adhesins, Bacterial/metabolism , Biofilms , Humans , Microbial Consortia/physiology , Organelle Biogenesis , Porphyromonas gingivalis/pathogenicity , Porphyromonas gingivalis/ultrastructure , Virulence Factors/metabolism
18.
Am J Dent ; 28(2): 81-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26087572

ABSTRACT

PURPOSE: To determine the efficacy of a solar-powered TiO2 semiconductor electric toothbrush on Porphyromonas gingivalis biofilm. METHODS: P. gingivalis cells were cultivated on sterilized coverslips under anaerobic conditions and were used as a biofilm. To evaluate the efficacy of the solar-powered TiO2 electric toothbrush on the P. gingivalis biofilm, the bacterial cell biofilm coverslips were placed into sterilized phosphate buffered saline (PBS) and brushed for 1 minute. Following mechanical brushing, the coverslips were stained with 1% crystal violet (CV) for 10 seconds at room temperature. The efficacy of P. gingivalis biofilm removal by the solar-powered TiO2 electric toothbrush was measured through the absorbance of the CV-stained solution containing the removed biofilm at 595 nm. The antimicrobial effect of the solar-powered TiO2 semiconductor was evaluated by the P. gingivalis bacterial count in PBS by blacklight irradiation for 0 to 60 minutes at a distance of 7 cm. The electrical current though the solar-powered TiO2 semiconductor was measured by a digital multimeter. The biofilm removal by the solar-powered TiO2 semiconductor was also evaluated by scanning electron microscopy (SEM). RESULTS: The biofilm removal rate of the solar-powered TiO2 electric toothbrush was 90.1 ± 1.4%, which was 1.3-fold greater than that of non-solar-powered electric toothbrushes. The solar-powered TiO2 semiconductor significantly decreased P. gingivalis cells and biofilm microbial activity in a time-dependent manner (P< 0.01). The electrical current passing through the solar-powered TiO2 semiconductor was 70.5 ± 0.1 µA, which was a 27-fold higher intensity than the non-solar-powered brush. SEM analysis revealed that the solar-powered TiO2 semiconductor caused a biofilm disruption and that cytoplasmic contents were released from the microbial cells.


Subject(s)
Biofilms , Porphyromonas gingivalis/physiology , Semiconductors , Solar Energy , Titanium/chemistry , Toothbrushing/instrumentation , Bacterial Load , Bacteriological Techniques , Coloring Agents , Cytoplasm/ultrastructure , Dental Plaque/microbiology , Electrical Equipment and Supplies , Gentian Violet , Humans , Materials Testing , Microscopy, Electron, Scanning , Porphyromonas gingivalis/ultrastructure , Time Factors , Ultraviolet Rays
19.
Mol Oral Microbiol ; 30(5): 361-75, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25869817

ABSTRACT

Neutrophils are regarded as the sentinel cells of innate immunity and are found in abundance within the gingival crevice. Discovery of neutrophil extracellular traps (NETs) within the gingival pockets prompted us to probe the nature of the interactions of neutrophils with the prominent periopathogen Porphyromonas gingivalis. Some of the noted virulence factors of this Gram-negative anaerobe are gingipains: arginine gingipains (RgpA/B) and lysine gingipain (Kgp). The aim of this study was to evaluate the role of gingipains in phagocytosis, formation of reactive oxygen species, NETs and CXCL8 modulation by using wild-type strains and isogenic gingipain mutants. Confocal imaging showed that gingipain mutants K1A (Kgp) and E8 (RgpA/B) induced extracellular traps in neutrophils, whereas ATCC33277 and W50 were phagocytosed. The viability of both ATCC33277 and W50 dwindled as the result of phagocytosis and could be salvaged by cytochalasin D, and the bacteria released high levels of lipopolysaccharide in the culture supernatant. Porphyromonas gingivalis induced reactive oxygen species and CXCL8 with the most prominent effect being that of the wild-type strain ATCC33277, whereas the other wild-type strain W50 was less effective. Quantitative real-time polymerase chain reaction revealed a significant CXCL8 expression by E8. All the tested P. gingivalis strains increased cytosolic free calcium. In conclusion, phagocytosis is the primary neutrophil response to P. gingivalis, although NETs could play an accessory role in infection control. Although gingipains do not seem to directly regulate phagocytosis, NETs or oxidative burst in neutrophils, their proteolytic properties could modulate the subsequent outcomes such as nutrition acquisition and survival by the bacteria.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Extracellular Traps/microbiology , Neutrophils/immunology , Phagocytosis , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/pathogenicity , Respiratory Burst , Cells, Cultured , Extracellular Traps/immunology , Gingipain Cysteine Endopeptidases , Host-Pathogen Interactions , Humans , Interleukin-8/genetics , Interleukin-8/immunology , Microbial Viability , Mutation , Neutrophils/ultrastructure , Porphyromonas gingivalis/physiology , Porphyromonas gingivalis/ultrastructure , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
20.
Klin Lab Diagn ; 60(12): 59-64, 2015 Dec.
Article in Russian | MEDLINE | ID: mdl-27032256

ABSTRACT

The study was carried out to analyze morphology of biofilm of periodontium and to develop electronic microscopic criteria of differentiated diagnostic of inflammatory diseases of gums. The scanning electronic microscopy was applied to analyze samples of bioflm of periodont from 70 patients. Including ten patients with every nosologic form of groups with chronic catarrhal periodontitis. of light, mean and severe degree, chronic catarrhal gingivitis, Candida-associated paroperiodontitis and 20 healthy persons with intact periodontium. The analysis was implemented using dual-beam scanning electronic microscope Quanta 200 3D (FEI company, USA) and walk-through electronic micJEM 100B (JEOL, Japan). To detect marker DNA of periodont pathogenic bacteria in analyzed samples the kit of reagentsfor polymerase chain reaction "MultiDent-5" ("GenLab", Russia). The scanning electronic microscopy in combination with transmission electronic microscopy and polymerase chain reaction permits analyzing structure, composition and degree of development of biofilm of periodontium and to apply differentiated diagnostic of different nosologic forms of inflammatory diseases of periodontium, including light form of chronic periodontitis and gingivitis. The electronic microscopical indications of diseases ofperiodontium of inflammatory character are established: catarrhal gingivitis, (coccal morphological alternate), chronic periodontitis (bacillary morphological alternate), Candida-associated periodontitis (Candida morphological alternate of biofilm ofperiodontium).


Subject(s)
Aggregatibacter actinomycetemcomitans/ultrastructure , Biofilms/growth & development , Candida albicans/ultrastructure , Gingivitis/diagnosis , Periodontitis/diagnosis , Porphyromonas gingivalis/ultrastructure , Prevotella intermedia/ultrastructure , Adolescent , Adult , Aggregatibacter actinomycetemcomitans/genetics , Aggregatibacter actinomycetemcomitans/isolation & purification , Biofilms/classification , Candida albicans/genetics , Candida albicans/isolation & purification , Case-Control Studies , Chronic Disease , Diagnosis, Differential , Female , Gingiva/microbiology , Gingiva/ultrastructure , Gingivitis/microbiology , Gingivitis/pathology , Humans , Male , Microscopy, Electron, Scanning , Middle Aged , Periodontitis/microbiology , Periodontitis/pathology , Polymerase Chain Reaction , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/isolation & purification , Prevotella intermedia/genetics , Prevotella intermedia/isolation & purification , Reagent Kits, Diagnostic , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL