Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 509(2): 335-340, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30579592

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Liases de Carbono-Enxofre/genética , Endopeptidase Clp/genética , Proteínas de Fímbrias/genética , Regulação Bacteriana da Expressão Gênica , Porphyromonas gingivalis/genética , Proteínas de Bactérias/metabolismo , Liases de Carbono-Enxofre/metabolismo , Endopeptidase Clp/deficiência , Proteínas de Fímbrias/metabolismo , Deleção de Genes , Violeta Genciana , Recombinação Homóloga , Microscopia Eletrônica de Varredura , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/ultraestrutura
2.
Anaerobe ; 55: 107-111, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30423437

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Fusobacterium nucleatum/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Propriedades de Superfície/efeitos dos fármacos , Treponema denticola/efeitos dos fármacos , Bactérias Anaeróbias/química , Bactérias Anaeróbias/efeitos dos fármacos , Bactérias Anaeróbias/ultraestrutura , Fusobacterium nucleatum/química , Fusobacterium nucleatum/ultraestrutura , Humanos , Interações Hidrofóbicas e Hidrofílicas , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Porphyromonas gingivalis/química , Porphyromonas gingivalis/ultraestrutura , Proteoma/análise , Treponema denticola/química , Treponema denticola/ultraestrutura
3.
J Proteome Res ; 17(7): 2377-2389, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29766714

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células/química , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Heme/farmacologia , Porphyromonas gingivalis/química , Proteoma/metabolismo , Proteínas da Membrana Bacteriana Externa , Micropartículas Derivadas de Células/efeitos dos fármacos , Heme/análise , Porphyromonas gingivalis/citologia , Porphyromonas gingivalis/ultraestrutura , Proteoma/efeitos dos fármacos
4.
PLoS Pathog ; 12(8): e1005820, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27509186

RESUMO

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.


Assuntos
Proteínas de Bactérias/ultraestrutura , Sistemas de Secreção Bacterianos/ultraestrutura , Porphyromonas gingivalis/ultraestrutura , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Eletroforese em Gel de Poliacrilamida , Immunoblotting , Imunoprecipitação , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Porphyromonas gingivalis/metabolismo
5.
Microb Pathog ; 116: 26-32, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29325862

RESUMO

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.


Assuntos
Acroleína/análogos & derivados , Antibacterianos/farmacologia , Cinnamomum zeylanicum/química , Óleos Voláteis/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Acroleína/isolamento & purificação , Acroleína/farmacologia , Antibacterianos/isolamento & purificação , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Óleos Voláteis/isolamento & purificação , Permeabilidade/efeitos dos fármacos , Casca de Planta/química , Porphyromonas gingivalis/ultraestrutura
6.
Artigo em Inglês | MEDLINE | ID: mdl-27993858

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos Hormonais/farmacologia , Biofilmes/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Streptococcus mutans/efeitos dos fármacos , Toremifeno/farmacologia , Biofilmes/crescimento & desenvolvimento , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Permeabilidade da Membrana Celular/efeitos dos fármacos , Placa Dentária/tratamento farmacológico , Placa Dentária/microbiologia , Reposicionamento de Medicamentos , Farmacorresistência Bacteriana Múltipla/fisiologia , Humanos , Testes de Sensibilidade Microbiana , Periodontite/tratamento farmacológico , Periodontite/microbiologia , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/ultraestrutura , Streptococcus mutans/metabolismo , Streptococcus mutans/ultraestrutura , Titânio/análise
7.
J Immunol ; 190(3): 1148-57, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264656

RESUMO

Porphyromonas gingivalis is a primary etiological agent of chronic periodontal disease, an infection-driven chronic inflammatory disease that leads to the resorption of tooth-supporting alveolar bone. We previously reported that TLR2 is required for P. gingivalis-induced alveolar bone loss in vivo, and our in vitro work implicated TNF as a key downstream mediator. In this study, we show that TNF-deficient (Tnf(-/-)) mice are resistant to alveolar bone loss following oral infection with P. gingivalis, and thus establish a central role for TNF in experimental periodontal disease. Using bone marrow-derived macrophages (BMDM) from wild-type and gene-specific knockout mice, we demonstrate that the initial inflammatory response to P. gingivalis in naive macrophages is MyD88 dependent and requires cooperative signaling of TLR2 and TLR4. The ability of P. gingivalis to activate cells via TLR2 or TLR4 was confirmed in TLR2- or TLR4-transformed human embryonic kidney cells. Additional studies using bacterial mutants demonstrated a role for fimbriae in the modulation of TLR-mediated activation of NF-κB. Whereas both TLR2 and TLR4 contributed to TNF production in naive macrophages, P. gingivalis preferentially exploited TLR2 in endotoxin-tolerant BMDM to trigger excessive TNF production. We found that TNF induced surface TLR2 expression and augmented TLR-induced cytokine production in P. gingivalis-stimulated BMDM, establishing a previously unidentified TNF-dependent feedback loop. Adoptive transfer of TLR2-expressing macrophages to TLR2-deficient mice restored the ability of P. gingivalis to induce alveolar bone loss in vivo. Collectively, our results identify a TLR2- and TNF-dependent macrophage-specific mechanism underlying pathogen-induced inflammatory bone loss in vivo.


Assuntos
Perda do Osso Alveolar/etiologia , Infecções por Bacteroidaceae/imunologia , Gengivite/fisiopatologia , Macrófagos/fisiologia , Porphyromonas gingivalis/patogenicidade , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Transferência Adotiva , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/fisiopatologia , Animais , Antibioticoprofilaxia , Infecções por Bacteroidaceae/microbiologia , Fímbrias Bacterianas/fisiologia , Regulação da Expressão Gênica/imunologia , Gengivite/complicações , Gengivite/imunologia , Células HEK293 , Humanos , Teste de Cultura Mista de Linfócitos , Ativação de Macrófagos , Macrófagos/transplante , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/ultraestrutura , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/deficiência
8.
Am J Dent ; 28(2): 81-4, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26087572

RESUMO

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.


Assuntos
Biofilmes , Porphyromonas gingivalis/fisiologia , Semicondutores , Energia Solar , Titânio/química , Escovação Dentária/instrumentação , Carga Bacteriana , Técnicas Bacteriológicas , Corantes , Citoplasma/ultraestrutura , Placa Dentária/microbiologia , Equipamentos e Provisões Elétricas , Violeta Genciana , Humanos , Teste de Materiais , Microscopia Eletrônica de Varredura , Porphyromonas gingivalis/ultraestrutura , Fatores de Tempo , Raios Ultravioleta
9.
Klin Lab Diagn ; 60(12): 59-64, 2015 Dec.
Artigo em Russo | MEDLINE | ID: mdl-27032256

RESUMO

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).


Assuntos
Aggregatibacter actinomycetemcomitans/ultraestrutura , Biofilmes/crescimento & desenvolvimento , Candida albicans/ultraestrutura , Gengivite/diagnóstico , Periodontite/diagnóstico , Porphyromonas gingivalis/ultraestrutura , Prevotella intermedia/ultraestrutura , Adolescente , Adulto , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Biofilmes/classificação , Candida albicans/genética , Candida albicans/isolamento & purificação , Estudos de Casos e Controles , Doença Crônica , Diagnóstico Diferencial , Feminino , Gengiva/microbiologia , Gengiva/ultraestrutura , Gengivite/microbiologia , Gengivite/patologia , Humanos , Masculino , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Periodontite/microbiologia , Periodontite/patologia , Reação em Cadeia da Polimerase , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/isolamento & purificação , Prevotella intermedia/genética , Prevotella intermedia/isolamento & purificação , Kit de Reagentes para Diagnóstico , Índice de Gravidade de Doença
10.
J Proteome Res ; 13(5): 2420-32, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24620993

RESUMO

Porphyromonas gingivalis, a keystone pathogen associated with chronic periodontitis, produces outer membrane vesicles (OMVs) that carry a cargo of virulence factors. In this study, the proteome of OMVs was determined by LC-MS/MS analyses of SDS-PAGE fractions, and a total of 151 OMV proteins were identified, with all but one likely to have originated from either the outer membrane or periplasm. Of these, 30 exhibited a C-terminal secretion signal known as the CTD that localizes them to the cell/vesicle surface, 79 and 27 were localized to the vesicle membrane and lumen respectively while 15 were of uncertain location. All of the CTD proteins along with other virulence factors were found to be considerably enriched in the OMVs, while proteins exhibiting the OmpA peptidoglycan-binding motif and TonB-dependent receptors were preferentially retained on the outer membrane of the cell. Cryo-transmission electron microscopy analysis revealed that an electron dense surface layer known to comprise CTD proteins accounted for a large proportion of the OMVs' volume providing an explanation for the enrichment of CTD proteins. Together the results show that P. gingivalis is able to specifically concentrate and release a large number of its virulence factors into the environment in the form of OMVs.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas Periplásmicas/metabolismo , Porphyromonas gingivalis/metabolismo , Fatores de Virulência/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Cromatografia Líquida , Microscopia Crioeletrônica , Eletroforese em Gel de Poliacrilamida , Microscopia Eletrônica de Transmissão , Periplasma/metabolismo , Periplasma/ultraestrutura , Porphyromonas gingivalis/patogenicidade , Porphyromonas gingivalis/ultraestrutura , Proteoma/metabolismo , Proteômica/métodos , Transdução de Sinais , Espectrometria de Massas em Tandem , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestrutura , Virulência
11.
Microb Pathog ; 77: 100-4, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25448131

RESUMO

The periodontopathogen Porphyromonas gingivalis is represented by a spectrum of phenotypes ranging from commensals to pathogenic lineages. Capsule and fimbriae are considered key virulence factors in this specie, involved in colonization and host defenses evasion. Since these virulence traits may not be expressed by certain strains, we aimed to test the hypothesis that certain clusters or genotypes of P. gingivalis correlate with the production of capsule and fimbriae. Sixteen P. gingivalis isolates were evaluated. Capsule (K) was detected by optical microscopy of negatively stained cells. The presence of fimbriae (F) was determined by TEM. Genotypes were determined by NotI macrorestriction fragments analysis through Pulsed-Field Gel Electrophoresis (PFGE) and Multi-locus sequence typing (MLST) based on seven house-keeping genes. The phenotypes included F(+)K(+) (n = 4), F(-)K(+) (n = 5), F(+)K(-) (n = 5) and F(-)K(-) (n = 2). The analysis of whole genome macrorestriction fragments revealed 14 different clusters. MLST data also revealed extensive genetic diversity; however, PFGE and MLST profiles showed evident differences. There was no association between P. gingivalis clusters and encapsulated and/or fimbriated phenotypes. Genotyping methods were not able to discriminate isolates according to the production of virulence factors such as capsule and major fimbriae, indicating that recombination played a key role in the expression of capsule and fimbriae in P. gingivalis.


Assuntos
Cápsulas Bacterianas , Fímbrias Bacterianas/ultraestrutura , Variação Genética , Porphyromonas gingivalis/genética , Propriedades de Superfície , DNA Bacteriano/química , DNA Bacteriano/genética , Eletroforese em Gel de Campo Pulsado , Genótipo , Microscopia , Tipagem de Sequências Multilocus , Fenótipo , Porphyromonas gingivalis/química , Porphyromonas gingivalis/classificação , Porphyromonas gingivalis/ultraestrutura
12.
J Periodontal Res ; 49(1): 62-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23550921

RESUMO

BACKGROUND AND OBJECTIVE: Porphyromonas gingivalis is a major etiological agent in the development and progression of periodontal diseases. In this study, we isolated a cell growth inhibitor against P. gingivalis species from rice protein extract. MATERIAL AND METHODS: The cell growth inhibitor active against P. gingivalis was purified from polished rice extract using a six-step column chromatography process. Its antimicrobial properties were investigated through microscope analysis, spectrum of activity and general structure. RESULTS: The inhibitor was identified as AmyI-1, an α-amylase, and showed significant cell growth inhibitory activity against P. gingivalis species. Scanning electron microscopy micrograph analysis and bactericidal assay indicated an intriguing possibility that the inhibitor compromises the cell membrane structure of the bacterial cells and leads to cell death. Moreover, α-amylases from human saliva and porcine pancreas showed inhibitory activity similar to that of AmyI-1. CONCLUSIONS: This is the first study to report that α-amylases cause cell death of periodontal pathogenic bacteria. This finding highlights the potential importance and therapeutic potential of α-amylases in treating periodontal diseases.


Assuntos
Antibacterianos/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , alfa-Amilases/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Oryza/enzimologia , alfa-Amilases Pancreáticas/farmacologia , Doenças Periodontais/microbiologia , Extratos Vegetais/farmacologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/ultraestrutura , Saliva/enzimologia , Proteínas e Peptídeos Salivares/farmacologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Suínos , alfa-Amilases/isolamento & purificação
13.
J Appl Microbiol ; 116(5): 1164-71, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24471579

RESUMO

AIMS: The aim of this study was to investigate the effects of tea catechin epigallocatechin gallate (EGCg) on established biofilms and biofilm formation by Porphyromonas gingivalis, a major pathogen of periodontal disease. METHODS AND RESULTS: Biofilm cell survival was measured using adenosine triphosphate (ATP) bioluminescence. In the presence of EGCg, the ATP level in cells of established biofilms was significantly decreased compared to the controls (P < 0·0001). Transmission electron microscopy revealed that EGCg damaged the cell membrane and cell wall of P. gingivalis. Confocal laser-scanning microscopy revealed that the proportion of dead cells was higher in biofilms treated with EGCg. Moreover, the effects of subminimal inhibitory concentrations (MICs) of EGCg on P. gingivalis biofilm formation were dose-dependent (P < 0·0001). CONCLUSION: Our results suggest that EGCg destroys established P. gingivalis biofilms and inhibits biofilm formation. SIGNIFICANCE AND IMPACT OF THE STUDY: Development of chemical control agents against oral biofilms is necessary, because oral biofilms can be only removed using mechanical debridement. This article indicates that EGCg may represent a novel antibiofilm agent that prevents infections involving bacterial biofilms such as periodontitis.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Catequina/análogos & derivados , Porphyromonas gingivalis/efeitos dos fármacos , Catequina/farmacologia , Porphyromonas gingivalis/fisiologia , Porphyromonas gingivalis/ultraestrutura
14.
Anaerobe ; 21: 34-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23583539

RESUMO

Laminaria japonica is a brown alga, which is consumed widely in Korea, Japan, and China. This study investigated the antimicrobial activity of ethanol extracts of L. japonica against oral microbial species to assess the possible application of L. japonica extracts in dental care products. The minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) were determined in culture medium using a microdilution method. The MICs of ethanol extracts of L. japonica with oral streptococci were 62.5-500 µg/ml and the MBCs were 125-1000 µg/ml. The MICs of Actinomyces naeslundii and Actinomyces odontolyticus were 250 and 62.5 µg/ml, respectively. The MBCs of A. naeslundii and A. odontolyticus were 500 and 250 µg/ml, respectively. The MICs were 250 and 62.5 µg/ml for Fusobacterium nucleatum and Porphyromonas gingivalis, respectively. The killing of Streptococcus mutans and P. gingivalis was dependent on the incubation time. The killing of S. mutans, A. odontolyticus, and P. gingivalis was significantly dependent on the extract concentration. Bacterial treatment with L. japonica extracts changed the cell surface texture of S. mutans, A. odontolyticus, and P. gingivalis. The results of this study suggest that L. japonica extracts may be useful for the development of antimicrobial agents to combat oral pathogens.


Assuntos
Anti-Infecciosos/farmacologia , Cárie Dentária/prevenção & controle , Placa Dentária/tratamento farmacológico , Laminaria/química , Boca/microbiologia , Actinomyces/efeitos dos fármacos , Actinomyces/ultraestrutura , Cariostáticos/química , Cariostáticos/farmacologia , Cárie Dentária/microbiologia , Placa Dentária/microbiologia , Etanol , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/ultraestrutura , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/ultraestrutura , Proteobactérias/efeitos dos fármacos , Proteobactérias/ultraestrutura , Streptococcus/efeitos dos fármacos , Streptococcus/ultraestrutura , Fatores de Tempo
15.
Infect Immun ; 80(1): 3-13, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22025518

RESUMO

The oral bacterium Porphyromonas gingivalis is a key etiological agent of human periodontitis, a prevalent chronic disease that affects up to 80% of the adult population worldwide. P. gingivalis exhibits neuraminidase activity. However, the enzyme responsible for this activity, its biochemical features, and its role in the physiology and virulence of P. gingivalis remain elusive. In this report, we found that P. gingivalis encodes a neuraminidase, PG0352 (SiaPg). Transcriptional analysis showed that PG0352 is monocistronic and is regulated by a sigma70-like promoter. Biochemical analyses demonstrated that SiaPg is an exo-α-neuraminidase that cleaves glycosidic-linked sialic acids. Cryoelectron microscopy and tomography analyses revealed that the PG0352 deletion mutant (ΔPG352) failed to produce an intact capsule layer. Compared to the wild type, in vitro studies showed that ΔPG352 formed less biofilm and was less resistant to killing by the host complement. In vivo studies showed that while the wild type caused a spreading type of infection that affected multiple organs and all infected mice were killed, ΔPG352 only caused localized infection and all animals survived. Taken together, these results demonstrate that SiaPg is an important virulence factor that contributes to the biofilm formation, capsule biosynthesis, and pathogenicity of P. gingivalis, and it can potentially serve as a new target for developing therapeutic agents against P. gingivalis infection.


Assuntos
Cápsulas Bacterianas/biossíntese , Biofilmes/crescimento & desenvolvimento , Neuraminidase/metabolismo , Porphyromonas gingivalis/enzimologia , Porphyromonas gingivalis/fisiologia , Sequência de Aminoácidos , Animais , Cápsulas Bacterianas/ultraestrutura , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/mortalidade , Infecções por Bacteroidaceae/patologia , Microscopia Crioeletrônica , Modelos Animais de Doenças , Tomografia com Microscopia Eletrônica , Deleção de Genes , Histocitoquímica , Experimentação Humana , Fígado/patologia , Pulmão/patologia , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/genética , Porphyromonas gingivalis/ultraestrutura , Regiões Promotoras Genéticas , Alinhamento de Sequência , Análise de Sobrevida , Transcrição Gênica , Virulência
16.
J Appl Microbiol ; 112(2): 404-11, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22093286

RESUMO

AIMS: To investigate the effects of the combined application of an N-acyl homoserine lactone (HSL) analog and antibiotics on biofilms of Porphyromonas gingivalis, a major pathogen of periodontal disease. METHODS AND RESULTS: Antibiotics used were cefuroxime, ofloxacin and minocycline. A flow-cell model was used for biofilm formation. Samples were divided into four groups: control, analog-treated, antibiotic-treated and combined application groups. Biofilm cell survival was determined using adenosine triphosphate (ATP) bioluminescence and confocal laser microscopy (CLSM). In the combined application group, the ATP count in biofilm cells was significantly decreased compared with the antibiotic-treated group (Games-Howell test, P < 0·05). A combination of cefuroxime and the analog was most effective against the P. gingivalis biofilm. CLSM observations revealed that the proportion of dead cells was highest in the combined application group. CONCLUSIONS: The combined application of the N-acyl HSL analog and antibiotics was effective at reducing the viability of P. gingivalis cells in biofilms. SIGNIFICANCE AND IMPACT OF THE STUDY: The combined application of the N-acyl HSL analog and antibiotics may be successful for eradicating infections involving bacterial biofilms, such as periodontitis.


Assuntos
Acil-Butirolactonas/farmacologia , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Doenças Periodontais/tratamento farmacológico , Porphyromonas gingivalis/efeitos dos fármacos , Sinergismo Farmacológico , Viabilidade Microbiana/efeitos dos fármacos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Doenças Periodontais/microbiologia , Porphyromonas gingivalis/ultraestrutura
17.
Antimicrob Agents Chemother ; 55(2): 806-12, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21098243

RESUMO

Polyphosphate [poly(P)] has antibacterial activity against various Gram-positive bacteria. In contrast, Gram-negative bacteria are generally resistant to poly(P). Here, we describe the antibacterial characterization of poly(P) against a Gram-negative periodontopathogen, Porphyromonas gingivalis. The MICs of pyrophosphate (Na(4)P(2)O(7)) and all poly(P) (Na(n + 2)P(n)O(3n + 1); n = 3 to 75) tested for the bacterium by the agar dilution method were 0.24% and 0.06%, respectively. Orthophosphate (Na(2)HPO(4)) failed to inhibit bacterial growth. Poly-P75 was chosen for further study. In liquid medium, 0.03% poly-P75 was bactericidal against P. gingivalis irrespective of the growth phase and inoculum size, ranging from 10(5) to 10(9) cells/ml. UV-visible spectra of the pigments from P. gingivalis grown on blood agar with or without poly-P75 showed that poly-P75 reduced the formation of µ-oxo bisheme by the bacterium. Poly-P75 increased hemin accumulation on the P. gingivalis surface and decreased energy-driven uptake of hemin by the bacterium. The expression of the genes encoding hemagglutinins, gingipains, hemin uptake loci, chromosome replication, and energy production was downregulated, while that of the genes related to iron storage and oxidative stress was upregulated by poly-P75. The transmission electron microscope showed morphologically atypical cells with electron-dense granules and condensed nucleoid in the cytoplasm. Collectively, poly(P) is bactericidal against P. gingivalis, in which hemin/heme utilization is disturbed and oxidative stress is increased by poly(P).


Assuntos
Antibacterianos/farmacologia , Polifosfatos/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Meios de Cultura , Regulação Bacteriana da Expressão Gênica , Heme/metabolismo , Hemina/metabolismo , Humanos , Testes de Sensibilidade Microbiana/métodos , Estresse Oxidativo , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/ultraestrutura
18.
Antimicrob Agents Chemother ; 55(12): 5887-92, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21911560

RESUMO

Antibiotic resistance of biofilm-grown bacteria contributes to chronic infections, such as marginal and periapical periodontitis, which are strongly associated with Porphyromonas gingivalis. Concurrent azithromycin (AZM) administration and mechanical debridement improve the clinical parameters of periodontal tissue in situ. We examined the in vitro efficacy of AZM against P. gingivalis biofilms. The susceptibilities of adherent P. gingivalis strains 381, HW24D1, 6/26, and W83 to AZM, erythromycin (ERY), ampicillin (AMP), ofloxacin (OFX), and gentamicin (GEN) were investigated using a static model. The optical densities of adherent P. gingivalis cells were significantly decreased by using AZM and ERY at sub-MIC levels compared with those of the controls in all the strains tested, except for the effect of ERY on strain W83. AMP and OFX inhibited P. gingivalis adherent cells at levels over their MICs, and GEN showed no inhibition in the static model. The effects of AZM and ERY against biofilm cells were investigated using a flow cell model. The ATP levels of P. gingivalis biofilms were significantly decreased by AZM at concentrations below the sub-MICs; however, ERY was not effective for inhibition of P. gingivalis biofilm cells at their sub-MICs. Furthermore, decreased density of P. gingivalis biofilms was observed three-dimensionally with sub-MIC AZM, using confocal laser scanning microscopy. These findings suggest that AZM is effective against P. gingivalis biofilms at sub-MIC levels and could have future clinical application for oral biofilm infections, such as chronic marginal and periapical periodontitis.


Assuntos
Antibacterianos/farmacologia , Azitromicina/farmacologia , Biofilmes/efeitos dos fármacos , Eritromicina/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Humanos , Testes de Sensibilidade Microbiana/normas , Microscopia Confocal , Porphyromonas gingivalis/fisiologia , Porphyromonas gingivalis/ultraestrutura
19.
Lasers Med Sci ; 26(2): 193-204, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20309597

RESUMO

The erbium:yttrium-aluminum-garnet (Er:YAG) laser is now increasingly used in periodontal therapy. The purpose of this study was to investigate the effect of Er:YAG laser irradiation on the morphology of periodontopathic bacteria and to compare the bacterial elimination effect of the laser and the ultrasonic scaler on diseased root surfaces in vitro. Colonies of Porphyromonas gingivalis were exposed to a single-pulse Er:YAG laser at 40 mJ and were examined by scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Also, 20 pairs of periodontally diseased root surfaces with subgingival calculi of freshly extracted teeth were treated by Er:YAG laser scaling at 40 mJ/pulse (14.2 J/cm(2) per pulse) and 10 Hz with water spray or ultrasonic scaling, or were not treated. The efficiency of each treatment was determined as the area treated per second, and the treated surfaces were examined by SEM. The material scraped from the treated root surfaces was cultured in aerobic and anaerobic conditions, and the numbers of colony forming units (CFUs) were compared. SEM and TEM showed that the Er:YAG laser had easily ablated the bacterial colony, leaving an ablation spot with a crater and the surrounding affected area showing melted branch-like structures. The laser irradiation was as equally effective and efficient as the ultrasonic scaler in performing root surface debridement. The CFUs after laser treatment were significantly fewer than those after ultrasonic scaling in aerobic and anaerobic culture conditions. Er:YAG laser ablates periodontopathic bacteria with thermal vaporization, and its bacterial elimination effect on the diseased root surfaces appears to be superior to that of the ultrasonic scaler.


Assuntos
Terapia a Laser , Desbridamento Periodontal/instrumentação , Periodontite/terapia , Porphyromonas gingivalis/ultraestrutura , Raiz Dentária/microbiologia , Terapia por Ultrassom , Infecções por Bacteroidaceae , Descontaminação , Raspagem Dentária/instrumentação , Humanos , Lasers de Estado Sólido , Microscopia Eletrônica , Periodontite/microbiologia , Doenças Dentárias , Raiz Dentária/diagnóstico por imagem , Raiz Dentária/efeitos da radiação , Ultrassonografia
20.
Sci Rep ; 11(1): 23954, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34907255

RESUMO

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.


Assuntos
Biofilmes/crescimento & desenvolvimento , Desinfecção , Fibroblastos/metabolismo , Gengiva/metabolismo , Lipopolissacarídeos/química , Porphyromonas gingivalis/fisiologia , Titânio/química , Fibroblastos/ultraestrutura , Humanos , Lasers de Estado Sólido , Microscopia Eletrônica de Varredura , Porphyromonas gingivalis/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA