Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Extracell Vesicles ; 13(1): e12407, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38251423

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is associated with nonalcoholic fatty liver disease (NAFLD) by lipid accumulation in the liver. In this study, we showed that extracellular vesicles (EVs) from the periodontal pathogens Filifactor alocis and Porphyromonas gingivalis induced steatosis by inducing PAI-1 in the liver and serum of mice fed a low-fat diet. PAI-1 induction was not observed in TLR2-/- mice. When tested using HEK-Blue hTLR2 cells, human TLR2 reporter cells, the TLR2-activating ability of serum from NAFLD patients (n = 100) was significantly higher than that of serum from healthy subjects (n = 100). Correlation analysis confirmed that PAI-1 levels were positively correlated with the TLR2-activating ability of serum from NAFLD patients and healthy subjects. Amphiphilic molecules in EVs were involved in PAI-1 induction. Our data demonstrate that the TLR2/PAI-1 axis is important for hepatic steatosis by EVs of periodontal pathogens.


Assuntos
Vesículas Extracelulares , Hepatopatia Gordurosa não Alcoólica , Inibidor 1 de Ativador de Plasminogênio , Receptor 2 Toll-Like , Animais , Humanos , Camundongos
2.
J Extracell Vesicles ; 12(12): e12381, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38014595

RESUMO

Periodontitis is a chronic inflammatory disease caused by periodontal pathogens in subgingival plaque and is associated with systemic inflammatory diseases. Extracellular vesicles (EVs) released from host cells and pathogens carry a variety of biological molecules and are of interest for their role in disease progression and as diagnostic markers. In the present study, we analysed the proteome and inflammatory response of EVs derived from macrophages infected with Tannerella forsythia, a periodontal pathogen. The EVs isolated from the cell conditioned medium of T. forsythia-infected macrophages were divided into two distinct vesicles, macrophage-derived EVs and T. forsythia-derived OMVs, by size exclusion chromatography combined with density gradient ultracentrifugation. Proteome analysis showed that in T. forsythia infection, macrophage-derived EVs were enriched with pro-inflammatory cytokines and inflammatory mediators associated with periodontitis progression. T. forsythia-derived OMVs harboured several known virulence factors, including BspA, sialidase, GroEL and various bacterial lipoproteins. T. forsythia-derived OMVs induced pro-inflammatory responses via TLR2 activation. In addition, we demonstrated that T. forsythia actively released OMVs when T. forsythia encountered macrophage-derived soluble molecules. Taken together, our results provide insight into the characterisation of EVs derived from cells infected with a periodontal pathogen.


Assuntos
Vesículas Extracelulares , Periodontite , Humanos , Tannerella forsythia , Proteoma , Periodontite/microbiologia , Macrófagos , Imunidade
3.
Mol Oral Microbiol ; 38(1): 71-81, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35866308

RESUMO

Outer membrane vesicles (OMVs) of bacteria harbor physiologically active molecules, and quorum sensing inhibitors (QSIs) are expected to regulate bacterial virulence. In this study, we analyzed the proinflammatory activity of OMVs of the periodontal pathogen Tannerella forsythia treated with d-arabinose and d-galactose as QSIs, which inhibit the biofilm formation of periodontal pathogens and autoinducer 2 activity. Compared to OMVs of nontreated T. forsythia (TF OMVs), OMVs released from QSI-treated T. forsythia, designated TF ara-OMVs and TF gal-OMVs, showed reduced production of TNF-α, IL-1ß, IL-6, and IL-8 in THP-1 monocytes through decreased activation of NF-κB/MAPKs. Using a human NF-κB reporter cell line and bone marrow-derived macrophages from TLR2-/- mice, TF ara-OMVs and TF gal-OMVs showed less activation of TLR2 than TF OMVs. These results demonstrated that QSIs provide a dual advantage against bacterial infection by inhibiting bacterial biofilm formation and generating OMVs with reduced proinflammatory activity.


Assuntos
NF-kappa B , Tannerella forsythia , Humanos , Animais , Camundongos , NF-kappa B/metabolismo , Receptor 2 Toll-Like/metabolismo , Percepção de Quorum , Macrófagos/metabolismo
4.
J Oral Microbiol ; 14(1): 2123550, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36312320

RESUMO

Outer membrane vesicles (OMVs) released from gram-negative bacteria harbor diverse molecules to communicate with host cells. In this study, we evaluated the OMVs of periodontal pathogens for their effects on the activation of dendritic cells and CD4+ T cell differentiation. OMVs of Porphyromonas gingivalis ATCC 33277, Treponema denticola ATCC 33521, and Tannerella forsythia ATCC 43037 ('red complex' pathogens) were isolated by density gradient ultracentrifugation. Mouse bone marrow-derived dendritic cells (BMDCs) were treated with OMVs, and OMV-primed BMDCs were cocultured with naïve CD4+ T cells to analyze the polarization of effector helper T cells. The OMVs upregulated maturation markers, including MHC class II, CD80, CD86, and CD40, on BMDCs. OMVs of P. gingivalis and T. forsythia induced the expression of the proinflammatory cytokines IL-1ß, IL-6, IL-23, and IL-12p70 in BMDCs. In T. denticola OMV-primed BMDCs, proinflammatory cytokines were poorly detected, which may be attributed to posttranslational degradation due to the highly proteolytic nature of OMVs. In cocultures of naïve CD4+ T cells with OMV-primed BMDCs, OMVs of P. gingivalis and T. denticola induced the differentiation of Th17 cells, whereas T. forsythia OMVs induced Th1 cell differentiation. These results demonstrate that OMVs derived from the 'red complex' periodontal pathogens induce maturation of BMDCs and differentiation of naïve CD4+ T cells to Th1 or Th17 cells.

5.
Sci Rep ; 12(1): 14239, 2022 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-35987920

RESUMO

Dysbiosis of the oral microbiota plays an important role in the progression of periodontitis, which is characterized by chronic inflammation and alveolar bone loss, and associated with systemic diseases. Bacterial extracellular vesicles (EVs) contain various bioactive molecules and show diverse effects on host environments depending on the bacterial species. Recently, we reported that EVs derived from Filifactor alocis, a Gram-positive periodontal pathogen, had osteoclastogenic activity. In the present study, we analysed the osteoclastogenic potency and immunostimulatory activity of EVs derived from the Gram-negative periodontal pathogens Porphyromonas gingivalis and Tannerella forsythia, the oral commensal bacterium Streptococcus oralis, and the gut probiotic strain Lactobacillus reuteri. Bacterial EVs were purified by density gradient ultracentrifugation using OptiPrep (iodixanol) reagent. EVs from P. gingivalis, T. forsythia, and S. oralis increased osteoclast differentiation and osteoclstogenic cytokine expression in osteoclast precursors, whereas EVs from L. reuteri did not. EVs from P. gingivalis, T. forsythia, and S. oralis preferentially activated Toll-like receptor 2 (TLR2) rather than TLR4 or TLR9, and induced osteoclastogenesis mainly through TLR2. The osteoclastogenic effects of EVs from P. gingivalis and T. forsythia were reduced by both lipoprotein lipase and polymyxin B, an inhibitor of lipopolysaccharide (LPS), while the osteoclastogenic effects of EVs from S. oralis were reduced by lipoprotein lipase alone. These results demonstrate that EVs from periodontal pathogens and oral commensal have osteoclastogenic activity through TLR2 activation by lipoproteins and/or LPS.


Assuntos
Vesículas Extracelulares , Boca , Osteoclastos , Diferenciação Celular , Vesículas Extracelulares/metabolismo , Lipopolissacarídeos , Lipase Lipoproteica , Microbiota , Boca/microbiologia , Osteoclastos/metabolismo , Porphyromonas gingivalis/fisiologia , Receptor 2 Toll-Like , Receptor 4 Toll-Like
6.
Anaerobe ; 75: 102533, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35143955

RESUMO

OBJECTIVES: Biofilm formation on dental implant surfaces can cause peri-implant mucositis and peri-implantitis. Lectins are involved in interactions between bacteria or between bacteria and their hosts. Disrupting these interactions via specific sugars can result in reduced adhesion and biofilm formation. The purpose of this study was to identify sugars that function as antiadhesion or antibiofilm agents on titanium discs. METHODS: Of the sugars tested, the sugars that did not affect the planktonic growth of Streptococcus oralis, Fusobacterium nucleatum, and Porphyromonas gingivalis were selected. The selected sugars were assessed for their ability to inhibit biofilm formation of bacteria in single and consortium species by crystal violet staining, confocal laser scanning microscopy after live/dead staining, and scanning electron microscopy. The sugars were evaluated for their ability to inhibit activity of the quorum sensing molecule autoinducer 2 (AI-2) by bioluminescence assay. RESULTS: Biofilm formation of single bacteria or consortia of S. oralis, F. nucleatum, and P. gingivalis on titanium discs was significantly inhibited in the presence of d-arabinose. Pretreating titanium discs with d-arabinose for 3 min inhibited biofilm formation at a level comparable to that observed when d-arabinose was present over the entire period, suggesting that d-arabinose had initial anti-adhesive activity. In addition, d-arabinose inhibited the activity of AI-2. CONCLUSIONS: d-Arabinose may be a good candidate for application as an antibiofilm agent and AI-2 inhibitor.


Assuntos
Peri-Implantite , Titânio , Arabinose/farmacologia , Biofilmes , Fusobacterium nucleatum , Humanos , Porphyromonas gingivalis , Titânio/farmacologia
7.
J Extracell Vesicles ; 10(12): e12157, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34648247

RESUMO

Periodontitis is an inflammatory disease induced by local infection in tooth-supporting tissue. Periodontitis is associated with systemic bone diseases, but little is known about the mechanism of the causal effect of periodontitis on systemic bone resorption. Bacteria-derived extracellular vesicles (EVs) act as natural carriers of virulence factors that are responsible for systemic inflammation. In this study, we investigated the role of EVs derived from Filifactor alocis, a Gram-positive, anaerobic periodontal pathogen, in systemic bone loss and osteoclast differentiation. F. alocis EVs accumulated in the long bones of mice after intraperitoneal administration. These EVs induced proinflammatory cytokines, osteoclastogenesis, and bone resorption via Toll-like receptor 2 (TLR2). The phase separation of F. alocis EVs showed that amphiphilic molecules were responsible for the induced bone resorption and osteoclastogenesis. The osteoclastogenic effects of F. alocis EVs were reduced by lipoprotein lipase. Proteomic analysis of the amphiphilic molecules identified seven lipoproteins. Our results indicate that lipoprotein-like molecules in F. alocis EVs may contribute to systemic bone loss via TLR2.


Assuntos
Doenças Ósseas/microbiologia , Vesículas Extracelulares/metabolismo , Periodontite/microbiologia , Receptor 2 Toll-Like/metabolismo , Animais , Clostridiales , Humanos , Camundongos
8.
Mol Oral Microbiol ; 35(5): 202-210, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33044804

RESUMO

Filifactor alocis, an asaccharolytic anaerobic Gram-positive rod (AAGPR), is an emerging marker of periodontitis. Severe periodontitis causes destruction of the alveolar bone that supports teeth and can even lead to tooth loss. Based on our previous report that F. alocis-derived extracellular vesicles (FA EVs) contain various effector molecules and have immunostimulatory activity, we investigated the effect of FA EVs on osteogenesis using mouse bone-derived mesenchymal stromal cells (BMSCs). FA EVs dramatically inhibited bone mineralization similar to whole bacteria and reduced the expression levels of osteogenic marker genes. The osteogenic differentiation of TLR2-deficient BMSCs was not inhibited by FA EVs, suggesting that their inhibitory effect on osteogenesis is dependent on TLR2 signaling. FA EVs effectively activated TLR2 downstream signaling of the MAPK and NF-κB pathways. In addition, FA EVs regulated RANKL and OPG gene expression, increasing the RANKL/OPG ratio in BMSCs in a TLR2-dependent manner. Our study suggests that F. alocis-derived EVs interfere with bone metabolism via TLR2 activation, providing insight into the pathogenesis of bone loss associated with periodontitis.


Assuntos
Clostridiales , Vesículas Extracelulares , Células-Tronco Mesenquimais/citologia , Osteogênese , Receptor 2 Toll-Like/metabolismo , Animais , Diferenciação Celular , Camundongos , Transdução de Sinais
9.
Mol Oral Microbiol ; 35(5): 222-230, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32794617

RESUMO

Uric acid is a potential metabolite that serves as a danger-associated molecular pattern (DAMP) and induces inflammatory responses in sterile environments. Porphyromonas gingivalis is a keystone periodontopathogen, and its gingipain proteases play a critical role in the pathogenesis of periodontitis. In this study, we demonstrate that P. gingivalis gingipains play a role in THP-1 macrophage uric acid production by increasing the expression and activity of xanthine oxidoreductase (XOR). Uric acid sodium salt induces caspase-1 activation, cell death, and the expression of proinflammatory cytokines, including IL-1α, IL-6, and IL-8, in the human keratinocyte HOK-16B cell line. Our results suggest that gingipain-induced uric acid can mediate inflammation in periodontal tissue cells.


Assuntos
Cisteína Endopeptidases Gingipaínas/metabolismo , Porphyromonas gingivalis/enzimologia , Ácido Úrico/metabolismo , Linhagem Celular , Citocinas/metabolismo , Humanos , Inflamação , Queratinócitos , Porphyromonas gingivalis/patogenicidade , Células THP-1 , Xantina Desidrogenase/metabolismo
10.
Arch Oral Biol ; 111: 104666, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31955046

RESUMO

In the oral microbial community, commensals can compete with pathogens and reduce their colonization in the oral cavity. A substance that can inhibit harmful bacteria and enrich beneficial bacteria is required to maintain oral health. The purpose of this study was to examine the effect of d-galactose on the biofilm formation of the cariogenic bacteria Streptococcus mutans and oral commensal streptococci and to evaluate their use in solution and in paste form. Biofilms of S. mutans, Streptococcus oralis, and Streptococcus mitis were formed on saliva-coated glass slips in the absence or presence of d-galactose and evaluated by staining with 1 % crystal violet. d-Galactose significantly inhibited the biofilm formation of S. mutans at concentrations ranging from 2 µM to 200 mM but increased the biofilm formation of S. oralis and S. mitis at concentrations of 2-200 mM. d-Galactose significantly inhibited three glucosyltransferase genes, gtfB, gtfC, and gtfD. The effect of d-galactose in the form of solution and paste was evaluated using bovine teeth. Pretreatment with 100 mM d-galactose on bovine teeth resulted in significantly reduced S. mutans biofilm formation. Our results suggest that d-galactose can be a candidate substance for the development of oral hygiene products to prevent caries by inhibiting the biofilm formation of S. mutans and simultaneously increasing the biofilm formation of commensal oral streptococci.


Assuntos
Streptococcus , Animais , Biofilmes , Bovinos , Galactose
11.
Mol Oral Microbiol ; 35(1): 1-9, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31675472

RESUMO

Filifactor alocis, a gram-positive, obligate anaerobic rod, is an emerging periodontal pathogen that is frequently isolated from patients with periodontitis, peri-implantitis, and apical periodontitis. Recent studies have shown that extracellular vesicles (EVs) from gram-negative periodontal pathogens, so-called outer membrane vesicles (OMVs), harbor various effector molecules responsible for inducing host inflammatory responses. However, there are no reports of EVs from F. alocis. In this study, we purified and characterized the protein profiles of EVs from F. alocis and investigated their immunostimulatory activity on human monocytic THP-1 and human oral keratinocyte HOK-16B cell lines. Highly pure EVs were obtained from F. alocis using density gradient ultracentrifugation. Nanoparticle tracking analysis and transmission electron microscopy showed that F. alocis EVs were between 50 and 270 nm in diameter. Proteome analysis identified 28 proteins, including lipoproteins, autolysins, F. alocis complement inhibitor (FACIN), transporter-related proteins, metabolism-related proteins, and ribosomal proteins. Human cytokine array analysis showed that F. alocis EVs remarkably induced the expression of CCL1, CCL2, MIP-1, CCL5, CXCL1, CXCL10, ICAM-1, IL-1ß, IL-1ra, IL-6, IL-8, MIF, SerpinE, and TNF-α in THP-1 cells and CXCL1, G-CSF, GM-CSF, IL-6, and IL-8 in HOK-16B cells. The immunostimulatory activity of F. alocis EVs was similar to that of the whole bacterial cells. Our findings provide new insight into the role of EVs from gram-positive oral bacteria in periodontal diseases.


Assuntos
Vesículas Extracelulares , Periodontite , Clostridiales , Bactérias Gram-Positivas , Humanos , Proteoma
12.
Mol Oral Microbiol ; 34(5): 209-218, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31332969

RESUMO

Interleukin-24 is a pleiotropic immunoregulatory cytokine and a member of the IL-20R subfamily of the IL-10 family. The aim of this study was to investigate the regulation of IL-24 in the human oral keratinocyte cell line HOK-16B following infection with Tannerella forsythia, a major periodontal pathogen. T. forsythia induced the expression of IL-24 mRNA and the secretion of glycosylated IL-24 in HOK-16B cells. Glycosylation of IL-24 is linked to its solubility and bioavailability. T. forsythia-stimulated reactive oxygen species (ROS) induced the expression of IL-24, which was regulated by IL-6. The ROS inhibitor N-acetylcysteine and MAPK inhibitors significantly reduced the expression of IL-6 and IL-24 induced by T. forsythia. Recombinant human IL-24 significantly enhanced the expression of IL-1α, IL-8, CXCL10, and MCP-1 in HOK-16B cells. Together, these results indicate that ROS, MAPKs, and IL-6 comprise the axis of IL-24 expression in HOK-16B cells stimulated with T. forsythia. Thus, IL-24 may be involved in inflammation in oral keratinocytes.


Assuntos
Inflamação , Interleucinas , Queratinócitos , Tannerella forsythia , Humanos , Interleucina-6/fisiologia , Interleucinas/metabolismo , Queratinócitos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Ligação Proteica , Espécies Reativas de Oxigênio , Transdução de Sinais , Tannerella forsythia/patogenicidade
13.
Eur J Oral Sci ; 126(6): 449-457, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30230039

RESUMO

This study aimed to verify, in in vivo settings, whether quorum-sensing inhibition molecules could attenuate alveolar bone loss induced by Porphyromonas gingivalis/Fusobacterium nucleatum co-infection and reduce the bacterial colonization of periodontal tissues. In BALB/c mice, periodontitis was induced through oral inoculation with P. gingivalis and F. nucleatum six times during a 42-d period. Quorum sensing inhibitors (a furanone compound and D-ribose) were administered simultaneously with bacterial infection. Linear and volumetric modifications of interproximal alveolar bone levels were compared between groups using micro-computed tomography. Total bacteria, and P. gingivalis and F. nucleatum DNA in periodontal tissues, were quantified using real-time PCR. Radiographic linear measurements demonstrated a significant reduction of alveolar bone loss, of approximately 40%, in mice treated with quorum sensing inhibitors when compared with the co-infection group. This was confirmed by a significant increase of residual bone volume in the test group. While total bacterial genes in the treatment group significantly decreased by 93% in periodontal tissue samples when quorum sensing inhibitors were administered, no significant differences of P. gingivalis DNA were found. Quorum sensing inhibitors reduced periodontal breakdown and bacterial infection in periodontal tissues after co-infection with P. gingivalis and F. nucleatum.


Assuntos
Coinfecção , Periodontite , Percepção de Quorum/efeitos dos fármacos , Perda do Osso Alveolar , Animais , DNA Bacteriano/análise , Modelos Animais de Doenças , Furanos/administração & dosagem , Furanos/antagonistas & inibidores , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/patogenicidade , Expressão Gênica , Genes Bacterianos , Interações Hospedeiro-Patógeno , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Periodontite/diagnóstico por imagem , Periodontite/microbiologia , Periodontite/patologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidade , Ribose/administração & dosagem , Ribose/antagonistas & inibidores , Microtomografia por Raio-X
14.
Cell Death Differ ; 25(2): 380-391, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29077095

RESUMO

Caspase-4 is an inflammatory caspase; however, its mechanism of activation is poorly understood. In this study, we demonstrate that Td92, a surface protein of the periodontal pathogen Treponema denticola and a homolog of the Treponema pallidum surface protein Tp92, activates caspase-4 and induces pyroptosis in primary cultured human gingival fibroblasts (HGFs) via cathepsin G activation. Cathepsin G inhibition or siRNA knockdown of cathepsin G inhibited Td92-induced caspase-4 activation and cell death. Td92-induced cell death was significantly inhibited by siRNA knockdown of gasdermin D. Td92 treatment resulted in the binding of cathepsin G to caspase-4 and the coaggregation of these two molecules. In addition, Td92 induced IL-1α expression and secretion, and this was inhibited by caspase-4 knockdown. Cytochalasin D did not block Td92-induced caspase-4 activation, suggesting that Td92 internalization is not required for caspase-4 activation. Our results demonstrate that cathepsin G is directly engaged in caspase-4 activation by a bacterial ligand, which is responsible for cell death and IL-1α secretion in HGFs.


Assuntos
Proteínas de Bactérias/metabolismo , Caspases Iniciadoras/metabolismo , Catepsina G/metabolismo , Fibroblastos/metabolismo , Gengiva/metabolismo , Treponema denticola/química , Treponema pallidum/química , Células Cultivadas , Fibroblastos/citologia , Gengiva/citologia , Humanos , Células THP-1 , Treponema denticola/metabolismo , Treponema pallidum/metabolismo
15.
Arch Oral Biol ; 84: 133-138, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28987726

RESUMO

OBJECTIVE: The antimicrobial efficacy of zinc- (ZnCl2) and cetylpyridinium-chloride (CPC) and their inhibition capacity on volatile sulfur compound (VSC) production by oral bacterial strains were investigated. DESIGN: Minimum inhibitory concentrations (MIC) and growth curves were determined for ZnCl2, CPC, and CPC with ZnCl2 solutions against eight oral microorganisms (Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, Porphyromonas gingivalis, Prevotella intermedia, Treponema denticola, Tannerella forsythia, Staphylococcus aureus and Streptococcus mutans) known to be involved in the pathophysiology of both halitosis and periodontal disease. Gas chromatography was applied to measure VSCs (H2S, CH3SH, (CH3)2S) production levels of each strains following exposure to the solutions. RESULTS: ZnCl2 and CPC effectively inhibited growth of all eight strains. ZnCl2 was generally more effective than CPC in suppressing bacterial growth excluding A. actinomycetemcomitans, P. intermedia, and T. forsythia. Synergism between CPC and ZnCl2 was shown in A. actinomycetemcomitans. The MIC for CPC was significantly lower than ZnCl2. VSC production was detected in five bacterial strains (A. actinomycetemcomitans, F. nucleatum, P. gingivalis, T. denticola, and T. forsythia). Each bacterial strain showed unique VSCs production profiles. H2S was produced by F. nucleatum, P. gingivalis, and T. denticola, CH3SH by all five strains and (CH3)2S by A. actinomycetemcomitans, F. nucleatum, P. gingivalis, and T. denticola. Production of CH3SH, the most malodorous component among the three major VSCs from mouth air was evident in F. nucleatum and T. forsythia. CONCLUSION: Both ZnCl2 and CPC effectively inhibit bacterial growth causative of halitosis and periodontal disease, resulting in a direct decrease of bacterial VSCs production.


Assuntos
Anti-Infecciosos Locais/farmacologia , Cetilpiridínio/farmacologia , Cloretos/farmacologia , Gases/metabolismo , Halitose/microbiologia , Compostos de Enxofre/metabolismo , Compostos de Zinco/farmacologia , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Cromatografia Gasosa , Fusobacterium nucleatum/efeitos dos fármacos , Técnicas In Vitro , Porphyromonas gingivalis/efeitos dos fármacos , Prevotella intermedia/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Streptococcus mutans/efeitos dos fármacos , Tannerella forsythia/efeitos dos fármacos , Treponema denticola/efeitos dos fármacos
16.
Innate Immun ; 23(7): 569-577, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28766990

RESUMO

This study investigated the pathogenesis of periodontitis and the role of nucleotide-binding oligomerization domain-like receptor protein 10 (NLRP10). The human oral epithelial cell line HOK-16B was infected with two periodontal pathogens, Tannerella forsythia and Fusobacterium nucleatum, at various MOIs. RT-PCR and immunoblotting demonstrated that infection increased mRNA and protein expression of NLRP10, respectively. The siRNA-mediated NLRP10 knockdown significantly reduced IL-1α expression and secretion. Both bacteria induced phosphorylation of ERK, JNK and p38 MAP kinases in HOK-16B cells. NLRP10 knockdown impaired ERK phosphorylation only. ERK inhibition significantly decreased the expression of T. forsythia- and F. nucleatum-induced IL-1α. Our data suggest that NLRP10 is involved in activating the ERK signalling pathway in HOK-16B cells infected with T. forsythia and F. nucleatum. This pathway likely augments the pro-inflammatory cytokine IL-1α levels, which may play a critical role in periodontitis.


Assuntos
Proteínas de Transporte/metabolismo , Células Epiteliais/imunologia , Infecções por Fusobacterium/imunologia , Fusobacterium nucleatum/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Periodontite/imunologia , Tannerella forsythia/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Linhagem Celular , Células Epiteliais/microbiologia , Regulação da Expressão Gênica , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Sistema de Sinalização das MAP Quinases , Boca/citologia , RNA Interferente Pequeno/genética
17.
J Oral Microbiol ; 9(1): 1320193, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28748028

RESUMO

Invasion of periodontal pathogens into periodontal tissues is an important step that can cause tissue destruction in periodontal diseases. Porphyromonas gingivalis is a keystone pathogen and its gingipains are key virulence factors. Fusobacterium nucleatum is a bridge organism that mediates coadhesion of disease-causing late colonizers such as P. gingivalis and early colonizers during the development of dental biofilms. The aim of this study was to investigate how P. gingivalis, in particular its gingipains, influences the invasion of coinfecting F. nucleatum into gingival epithelial cells. When invasion of F. nucleatum was analyzed after 4 h of infection, invasion of F. nucleatum was suppressed in the presence of P. gingivalis compared with during monoinfection. However, coinfection with a gingipain-null mutant of P. gingivalis did not affect invasion of F. nucleatum. Inhibition of PI3K reduced invasion of F. nucleatum. P. gingivalis inactivated the PI3K/AKT pathway, which was also dependent on gingipains. Survival of intracellular F. nucleatum was promoted by P. gingivalis with Arg gingipain mutation. The results suggest that P. gingivalis, in particular its gingipains, can affect the invasion of coinfecting F. nucleatum through modulating intracellular signaling of the host cells.

18.
Eur J Med Chem ; 137: 76-87, 2017 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-28554093

RESUMO

Bacterial behaviors such as virulence factor secretion and biofilm formation are critical for survival, and are effectively regulated through quorum sensing, a mechanism of intra- and interspecies communication in response to changes in cell density and species complexity. Many bacterial species colonize host tissues and form a defensive structure called a biofilm, which can be the basis of inflammatory diseases. Periodontitis, a chronic inflammatory disease affecting the periodontium, is caused by subgingival biofilms related to periodontopathogens. In particular, Fusobacterium nucleatum is a major co-aggregation bridge organism in the formation and growth of subgingival biofilms, linking the early and late colonizers in periodontal biofilms. According to our previous study, the intergeneric quorum-sensing signal molecule autoinducer-2 (AI-2) of F. nucleatum plays a key role in intra- and interspecies interactions of periodontopathogens, and may be a good target for periodontal biofilm inhibition. Recently, brominated furanones produced by the macroalga Delisea pulchra were shown to inhibit biofilm formation via AI-2, and have been investigated toward the goal of increasing the inhibition effect. In this study, we describe the synthesis of new bromofuranone analogs, i.e., 3-(dibromomethylene)isobenzofuran-1(3H)-one derivatives, and demonstrate their inhibitory activities against biofilm formation by periodontopathogens, including F. nucleatum, Porphyromonas gingivalis, and Tannerella forsythia.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Furanos/farmacologia , Homosserina/análogos & derivados , Lactonas/antagonistas & inibidores , Antibacterianos/síntese química , Antibacterianos/química , Relação Dose-Resposta a Droga , Furanos/síntese química , Fusobacterium nucleatum/efeitos dos fármacos , Homosserina/antagonistas & inibidores , Testes de Sensibilidade Microbiana , Estrutura Molecular , Porphyromonas gingivalis/efeitos dos fármacos , Percepção de Quorum/efeitos dos fármacos , Relação Estrutura-Atividade , Tannerella forsythia/efeitos dos fármacos
20.
Arch Oral Biol ; 73: 72-78, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27697692

RESUMO

OBJECTIVE: The aim of this study was to analyze whether periodontopathogens induced inflammatory cell death and the release of diverse endogenous danger molecules in THP-1-derived macrophages. METHODS: The macrophages were treated with Treponema denticola, Porphyromonas gingivalis, and Tannerella forsythia. Activation of caspase-1 and caspase-4 was detected by Western blotting. Cell death of bacteria-stimulated macrophages was examined using a lactate dehydrogenase (LDH) assay and propidium iodide (PI)/annexin V (AV) staining. Levels of endogenous danger signals, including adenosine triphosphate (ATP), uric acid, heat shock protein 60 (HSP60), high-mobility group box protein 1 (HMGB1), and fibronectin in the culture supernatants were determined using an ATP bioluminescence assay kit, a uric acid assay kit, and Western blotting, respectively. RESULTS: T. denticola, P. gingivalis, and T. forsythia induced activation of caspase-1 and caspase-4. The LDH assay and PI/AV staining showed that all three pathogens induced pyroptotic cell death. All three bacteria induced release of ATP, which is an important ligand for inflammasome activation; the increase in ATP ultimately leads to caspase-1 activation. T. denticola induced release of HSP60 and fibronectin, while T. forsythia induced release of HMGB1 in addition to HSP60 and fibronectin. None of the endogenous molecules except for fibronectin were detected in P. gingivalis-infected cells, possibly due to degradation of these factors by the proteolytic activity of the bacteria. Interestingly, P. gingivalis induced uric acid release. CONCLUSION: Inflammatory cell death and endogenous danger molecules released from cells infected with periodontopathogens may play critical roles in the pathogenesis and progression of periodontitis by augmenting immune and inflammatory responses.


Assuntos
Morte Celular/fisiologia , Periodontite/microbiologia , Porphyromonas gingivalis/patogenicidade , Tannerella forsythia/patogenicidade , Treponema denticola/patogenicidade , Trifosfato de Adenosina/metabolismo , Western Blotting , Caspase 1/metabolismo , Caspases Iniciadoras/metabolismo , Células Cultivadas , Chaperonina 60/metabolismo , Fibronectinas/metabolismo , Citometria de Fluxo , Proteínas HMGB/metabolismo , Humanos , Macrófagos , Porphyromonas gingivalis/enzimologia , Transdução de Sinais , Tannerella forsythia/enzimologia , Treponema denticola/enzimologia , Ácido Úrico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...