Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 468
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34992142

RESUMO

Bacterial behavior and virulence during human infection is difficult to study and largely unknown, as our vast knowledge of infection microbiology is primarily derived from studies using in vitro and animal models. Here, we characterize the physiology of Porphyromonas gingivalis, a periodontal pathogen, in its native environment using 93 published metatranscriptomic datasets from periodontally healthy and diseased individuals. P. gingivalis transcripts were more abundant in samples from periodontally diseased patients but only above 0.1% relative abundance in one-third of diseased samples. During human infection, P. gingivalis highly expressed genes encoding virulence factors such as fimbriae and gingipains (proteases) and genes involved in growth and metabolism, indicating that P. gingivalis is actively growing during disease. A quantitative framework for assessing the accuracy of model systems showed that 96% of P. gingivalis genes were expressed similarly in periodontitis and in vitro midlogarithmic growth, while significantly fewer genes were expressed similarly in periodontitis and in vitro stationary phase cultures (72%) or in a murine abscess infection model (85%). This high conservation in gene expression between periodontitis and logarithmic laboratory growth is driven by overall low variance in P. gingivalis gene expression, relative to other pathogens including Pseudomonas aeruginosa and Staphylococcus aureus Together, this study presents strong evidence for the use of simple test tube growth as the gold standard model for studying P. gingivalis biology, providing biological relevance for the thousands of laboratory experiments performed with logarithmic phase P. gingivalis Furthermore, this work highlights the need to quantitatively assess the accuracy of model systems.


Assuntos
Infecções por Bacteroidaceae/microbiologia , Periodontite/microbiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/metabolismo , Animais , Fímbrias Bacterianas/metabolismo , Cisteína Endopeptidases Gingipaínas , Humanos , Laboratórios , Camundongos , Porphyromonas gingivalis/patogenicidade , Transcriptoma , Virulência/genética , Fatores de Virulência
2.
J Microbiol ; 60(1): 70-78, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34826101

RESUMO

Low electric current can inhibit certain microbial biofilms and enhance the efficacy of antimicrobials against them. This study investigated the electricidal and bioelectric effects of direct current (DC) against Porphyromonas gingivalis biofilms as well as the underlying mechanisms. Here, we firstly showed that DC significantly suppressed biofilm formation of P. gingivalis in time- and intensity-dependent manners, and markedly inhibited preformed P. gingivalis biofilms. Moreover, DC enhanced the killing efficacy of metronidazole (MTZ) and amoxicillin with clavulanate potassium (AMC) against the biofilms. Notably, DC-treated biofilms displayed upregulated intracellular ROS and expression of ROS related genes (sod, feoB, and oxyR) as well as porin gene. Interestingly, DC-induced killing of biofilms was partially reversed by ROS scavenger N-dimethylthiourea (DMTU), and the synergistic effect of DC with MTZ/AMC was weakened by small interfering RNA of porin gene (si-Porin). In conclusion, DC can exert electricidal and bioelectric effects against P. gingivalis biofilms partially via promotion of oxidative stress and antibiotic transport, which offers a promising approach for effective management of periodontitis.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Porphyromonas gingivalis/química , Porphyromonas gingivalis/efeitos dos fármacos , Amoxicilina/farmacologia , Eletricidade , Humanos , Metronidazol/farmacologia , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Periodontite/microbiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/fisiologia , Espécies Reativas de Oxigênio/metabolismo
3.
Microbiol Spectr ; 9(3): e0121221, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34756068

RESUMO

In the periodontal pocket, there is a direct correlation between environmental conditions, the dynamic oral microbial flora, and disease. The relative abundance of several newly recognized microbial species in the oral microenvironment has raised questions on their impact on disease development. One such organism, Filifactor alocis, is significant to the pathogenic biofilm structure. Moreover, its pathogenic characteristics are highlighted by its ability to survive in the oxidative-stress microenvironment of the periodontal pocket and alter the microbial community dynamics. There is a gap in our understanding of its mechanism(s) of oxidative stress resistance and impact on pathogenicity. Several proteins, including HMPRFF0389-00519 (FA519), were observed in high abundance in F. alocis during coinfection of epithelial cells with Porphyromonas gingivalis W83. Bioinformatics analysis shows that FA519 contains a "Cys-X-X-Cys zinc ribbon domain" which could be involved in DNA binding and oxidative stress resistance. We have characterized FA519 to elucidate its roles in the oxidative stress resistance and virulence of F. alocis. Compared to the wild-type strain, the F. alocis isogenic gene deletion mutant, FLL1013 (ΔFA519::ermF), showed significantly reduced sensitivity to hydrogen peroxide and nitric oxide-induced stress. The ability to form biofilm and adhere to and invade gingival epithelial cells was also reduced in the isogenic mutant. The recombinant FA519 protein was shown to protect DNA from Fenton-mediated damage with an intrinsic ability to reduce hydrogen peroxide and disulfide bonds. Collectively, these results suggest that FA519 is involved in oxidative stress resistance and can modulate important virulence attributes in F. alocis. IMPORTANCE Filifactor alocis is an emerging member of the periodontal community and is now proposed to be a diagnostic indicator of periodontal disease. However, due to the lack of genetic tools available to study this organism, not much is known about its virulence attributes. The mechanism(s) of oxidative stress resistance in F. alocis is unknown. Therefore, identifying the adaptive mechanisms utilized by F. alocis to survive in the oxidative stress environment of the periodontal pocket would lead to understanding its virulence regulation, which could help develop novel therapeutic treatments to combat the effects of periodontal disease. This study is focused on the characterization of FA519, a hypothetical protein in F. alocis, as a multifunctional protein that plays an important role in the reactive oxygen species-detoxification pathway. Collectively, our results suggest that FA519 is involved in oxidative stress resistance and can modulate important virulence attributes in F. alocis.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridiales/metabolismo , Inativação Metabólica/fisiologia , Estresse Oxidativo/fisiologia , Bolsa Periodontal/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/metabolismo , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Clostridiales/genética , Clostridiales/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Inativação Metabólica/genética , Microbiota/fisiologia , Oxirredutases/genética , Oxirredutases/metabolismo , Doenças Periodontais/microbiologia , Doenças Periodontais/patologia , Peroxidase/metabolismo , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/metabolismo , Tiorredoxinas/metabolismo , Fatores de Virulência/genética
5.
Dis Mon ; 67(9): 101166, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33663798

RESUMO

OBJECTIVES: The effect of antiretroviral therapy (ART) on the oral pathogenic microbes in human immunodeficiency virus-1 seropositive patients remains relatively unexplored. Thus, the present study assessed the effect of ART on the sub-gingival levels of 3 pathogenic microbes. MATERIALS AND METHODS: The study groups consisted of 60 human immunodeficiency virus-1 seropositive patients divided into 3 groups of 20 each. Group 1 had periodontitis and did not start with the ART. Group 2 had periodontitis and started with ART (Tenofovir Disoproxil Fumarate 300 mg + Lamivudine 300 mg + Efavirenz 600 mg) at least 6 months before the study. Group 3 with normal periodontium, and have not started ART. The sub-gingival loads of Cytomegalovirus, Epstein-Barr virus, and the Porphyromonas gingivalis levels were assessed, along with the CD4 counts. RESULTS: The cytomegalovirus load was highest in group 1, followed by groups 2, and 3 (p-value of 0.271). The Epstein-Barr load was highest for group 2, followed by group 3, and 1 (p-value of 0.022). The P.gingivalis load was highest in group 2, followed by groups 1 and 3, (p-value of 0.028). The Epstein-Barr and Cytomegalovirus counts were significantly higher (p-value < 0.02) when the CD4 counts were less than 500 cells/cu3. CONCLUSION: ART did not cause any significant reduction in the sub-gingival levels of any of the 3 examined microbes. Given the lack of any significant effect on the sub-gingival microbial loads by the ART, human immunodeficiency virus patients may require additional anti-microbial agents and regular mechanical plaque removal to maintain their periodontal status.


Assuntos
Terapia Antirretroviral de Alta Atividade , Citomegalovirus/crescimento & desenvolvimento , Infecções por HIV , HIV-1/crescimento & desenvolvimento , Herpesvirus Humano 4/crescimento & desenvolvimento , Periodontite , Porphyromonas gingivalis/crescimento & desenvolvimento , Adulto , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Infecções por Bacteroidaceae/complicações , Infecções por Bacteroidaceae/microbiologia , Contagem de Linfócito CD4 , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/virologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/virologia , Feminino , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Humanos , Masculino , Periodontite/complicações , Periodontite/microbiologia , Periodontite/virologia , Periodonto/efeitos dos fármacos , Periodonto/microbiologia , Periodonto/patologia , Periodonto/virologia
6.
PLoS One ; 16(1): e0246194, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33497417

RESUMO

Periodontal diseases, including gingivitis and periodontitis, are a global oral health problem. Porphyromonas gingivalis, a key pathogen involved in the onset of periodontitis, is able to colonize the subgingival epithelium and invade the underlying connective tissue due to the contribution of cysteine proteases known as gingipains. In this study, we investigated the effects of a phenolic extract prepared from tart cherry (Prunus cerasus L.) juice on the growth, adherence, and protease activity of P. gingivalis. We also assessed the protective effect of the tart cherry extract on the disruption of the oral epithelial barrier induced by P. gingivalis. The tart cherry extract that contains procyanidins and quercetin and its derivatives (rutinoside, glucoside) as the most important phenolic compounds attenuated P. gingivalis growth, reduced adherence to an experimental basement membrane matrix model, and decreased the protease activities of P. gingivalis. The tart cherry extract also exerted a protective effect on the integrity of the oral epithelial barrier in an in vitro model infected with P. gingivalis. More specifically, the extract prevented a decrease in transepithelial electrical resistance as well as the destruction of tight junction proteins (zonula occludens-1 and occludin). These results suggest that the tart cherry phenolic extract may be a promising natural product for the treatment of periodontitis through its ability to attenuate the virulence properties of P. gingivalis and curtail the ability of this pathogen to impair the oral epithelial barrier.


Assuntos
Células Epiteliais , Mucosa Bucal , Extratos Vegetais/farmacologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Prunus/química , Junções Íntimas/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Humanos , Mucosa Bucal/metabolismo , Mucosa Bucal/microbiologia , Mucosa Bucal/patologia , Periodontite/tratamento farmacológico , Periodontite/metabolismo , Periodontite/microbiologia , Extratos Vegetais/química
7.
J Tissue Eng Regen Med ; 15(3): 232-243, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33434402

RESUMO

Infectious bone defects remain a significant challenge in orthopedics and dentistry. Calcium phosphate cement (CPC) have attracted significant interest in use as local drug delivery system, which with great potential to control release of antibiotics for the treatment of infectious bone defects. Within the current study, a novel antibacterial scaffold of chitosan-reinforced calcium phosphate cement delivering doxycycline hyclate (CPCC + DOX) was developed. Furthermore, the capacity of CPCC + DOX scaffolds for bone regeneration was enhanced by the human periodontal ligament stem cells (hPDLSCs) encapsulated in alginate beads. CPCC + DOX scaffolds were fabricated to contain different concentrations of DOX. Flexural strength of CPCC + DOX ranged from 5.56 ± 0.70 to 6.2 ± 0.72 MPa, which exceeded the reported strength of cancellous bone. Scaffolds exhibited continual DOX release, reaching 80% at 21 days. Scaffold with 5 mg/ml DOX (CPCC + DOX5mg) had a strong antibacterial effect, with a 4-log colony forming unit reduction against S. aureus and P. gingivalis. The proliferation and osteogenic differentiation of hPDLSCs encapsulated in alginate hydrogel microbeads were investigated in culture with CPCC + DOX scaffolds. CPCC + DOX5mg had no negative effect on proliferation of hPDLSCs. Alkaline phosphatase activity, mineral synthesis, and osteogenic gene expressions for CPCC + DOX5mg group were much higher than control group. DOX did not compromise the osteogenic induction. In summary, the novel CPCC + DOX scaffold exhibited excellent mechanical properties and strong antibacterial activity, while supporting the proliferation and osteogenic differentiation of hPDLSCs. The CPCC + DOX + hPDLSCs construct is promising to enhance bone regeneration and combat bone infections in dental, craniofacial, and orthopedic applications.


Assuntos
Antibacterianos , Infecções por Bacteroidaceae , Cimentos Ósseos , Regeneração Óssea/efeitos dos fármacos , Microesferas , Osteogênese , Ligamento Periodontal , Porphyromonas gingivalis/crescimento & desenvolvimento , Infecções Estafilocócicas , Staphylococcus aureus/crescimento & desenvolvimento , Células-Tronco , Antibacterianos/química , Antibacterianos/farmacologia , Infecções por Bacteroidaceae/tratamento farmacológico , Infecções por Bacteroidaceae/microbiologia , Cimentos Ósseos/química , Cimentos Ósseos/farmacologia , Fosfatos de Cálcio , Humanos , Ligamento Periodontal/metabolismo , Ligamento Periodontal/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/metabolismo , Células-Tronco/metabolismo , Células-Tronco/microbiologia
8.
Biochim Biophys Acta Mol Cell Res ; 1868(3): 118923, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33285176

RESUMO

The keystone pathogen Porphyromonas gingivalis (P. gingivalis) elicits inflammation and autophagy in periodontal tissues. Transcription factor CXXC-type zinc finger protein 5 (CXXC5) and various signals are sensitive to P. gingivalis invasion. Herein, we investigated the P. gingivalis-elicited autophagy activity, the contribution of CXXC5, and the involvement of signals in cementoblasts, tooth root surface cells crucial in periodontal and periapical regions. After coculture with P. gingivalis, cementoblasts exhibited inflammatory cytokine increase, light chain 3(LC3)-I/II conversion, autophagosome activation, and CXXC5 reduction. Cementoblasts with loss and gain of CXXC5 were developed. CXXC5 silencing suppressed autophagy and inflammation, thereby partially compensating for the effects of P. gingivalis, and vice versa. We then screened potential signals and verified the positive participation of Stat3/Akt/Erk networks through specific inhibitor employment. P. gingivalis and CXXC5 induced autophagy through Beclin1 and Atg5 activation. Intriguingly, Annexin V/PI assay and EdU detection revealed that P. gingivalis promoted apoptosis and repressed cell proliferation. In sum, coculture with P. gingivalis enhanced autophagy activity in cementoblasts, which was partially suppressed by CXXC5 downregulation and mediated by Jak/Stat3, PI3K-Akt, and Erk1/2 signaling. This process probably influenced cell apoptosis and proliferation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Cemento Dentário/citologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/metabolismo , Animais , Autofagossomos/metabolismo , Autofagia , Técnicas Bacteriológicas , Técnicas de Cultura de Células , Linhagem Celular , Cemento Dentário/metabolismo , Cemento Dentário/microbiologia , Regulação para Baixo , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Porphyromonas gingivalis/patogenicidade
9.
FEBS J ; 288(5): 1479-1495, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32681704

RESUMO

Several studies have shown a clear association between periodontal disease and increased risk of cardiovascular disease. Porphyromonas gingivalis (Pg), a key oral pathogen, and its cell surface-expressed gingipains, induce oedema in a zebrafish larvae infection model although the mechanism of these vascular effects is unknown. Here, we aimed to determine whether Pg-induced vascular damage is mediated by gingipains. In vitro, human endothelial cells from different vascular beds were invaded by wild-type (W83) but not gingipain-deficient (ΔK/R-ab) Pg. W83 infection resulted in increased endothelial permeability as well as decreased cell surface abundance of endothelial adhesion molecules PECAM-1 and VE-cadherin compared to infection with ΔK/R-ab. In agreement, when transgenic zebrafish larvae expressing fluorescently labelled PECAM-1 or VE-cadherin were systemically infected with W83 or ΔK/R-ab, a significant reduction in adhesion molecule fluorescence was observed specifically in endothelium proximal to W83 bacteria through a gingipain-dependent mechanism. Furthermore, this was associated with increased vascular permeability in vivo when assessed by dextran leakage microangiography. These data are the first to show that Pg directly mediates vascular damage in vivo by degrading PECAM-1 and VE-cadherin. Our data provide a molecular mechanism by which Pg might contribute to cardiovascular disease.


Assuntos
Infecções por Bacteroidaceae/etiologia , Cardiomegalia/etiologia , Edema/etiologia , Células Endoteliais/efeitos dos fármacos , Cisteína Endopeptidases Gingipaínas/toxicidade , Porphyromonas gingivalis/patogenicidade , Animais , Animais Geneticamente Modificados , Antígenos CD/genética , Antígenos CD/metabolismo , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroidaceae/patologia , Caderinas/genética , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Edema/genética , Edema/metabolismo , Edema/patologia , Embrião não Mamífero , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Angiofluoresceinografia , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Cisteína Endopeptidases Gingipaínas/biossíntese , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Larva/efeitos dos fármacos , Larva/microbiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/metabolismo , Cultura Primária de Células , Transdução de Sinais , Peixe-Zebra
10.
Arch Oral Biol ; 121: 104956, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33157493

RESUMO

OBJECTIVE: Rice peptide has antibacterial properties that have been tested in planktonic bacterial culture. However, bacteria form biofilm at disease sites and are resistant to antibacterial agents. The aim of this study was to clarify the mechanisms of action of rice peptide and its amino acid substitution against periodontopathic bacteria and their antibiofilm effects. DESIGN: Porphyromonas gingivalis and Fusobacterium nucleatum were treated with AmyI-1-18 rice peptide or its arginine-substituted analog, G12R, under anaerobic conditions. The amount of biofilm was evaluated by crystal violet staining. The integrity of the bacteria cytoplasmic membrane was studied in a propidium iodide (PI) stain assay and transmission electron microscopy (TEM). RESULTS: Both AmyI-1-18 and G12R inhibited biofilm formation of P. gingivalis and F. nucleatum; in particular, G12R inhibited F. nucleatum at lower concentrations. However, neither peptide eradicated established biofilms significantly. According to the minimum inhibitory concentration and minimum bactericidal concentration against P. gingivalis, AmyI-1-18 has bacteriostatic properties and G12R has bactericidal activity, and both peptides showed bactericidal activity against F. nucleatum. PI staining and TEM analysis indicated that membrane disruption by G12R was enhanced, which suggests that the replacement amino acid reinforced the electostatic interaction between the peptide and bacteria by increase of cationic charge and α-helix content. CONCLUSIONS: Rice peptide inhibited biofilm formation of P. gingivalis and F. nucleatum, and bactericidal activity via membrane destruction was enhanced by amino acid substitution.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Fusobacterium nucleatum/efeitos dos fármacos , Oryza/química , Peptídeos/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Substituição de Aminoácidos , Fusobacterium nucleatum/crescimento & desenvolvimento , Proteínas de Plantas/farmacologia , Porphyromonas gingivalis/crescimento & desenvolvimento
11.
Sci Rep ; 10(1): 14745, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32901057

RESUMO

The control of inflammation and infection is crucial for periodontal wound healing and regeneration. M101, an oxygen carrier derived from Arenicola marina, was tested for its anti-inflammatory and anti-infectious potential based on its anti-oxidative and tissue oxygenation properties. In vitro, no cytotoxicity was observed in oral epithelial cells (EC) treated with M101. M101 (1 g/L) reduced significantly the gene expression of pro-inflammatory markers such as TNF-α, NF-κΒ and RANKL in P. gingivalis-LPS stimulated and P. gingivalis-infected EC. The proteome array revealed significant down-regulation of pro-inflammatory cytokines (IL-1ß and IL-8) and chemokine ligands (RANTES and IP-10), and upregulation of pro-healing mediators (PDGF-BB, TGF-ß1, IL-10, IL-2, IL-4, IL-11 and IL-15) and, extracellular and immune modulators (TIMP-2, M-CSF and ICAM-1). M101 significantly increased the gene expression of Resolvin-E1 receptor. Furthermore, M101 treatment reduced P. gingivalis biofilm growth over glass surface, observed with live/dead analysis and by decreased P. gingivalis 16 s rRNA expression (51.7%) (p < 0.05). In mice, M101 reduced the clinical abscess size (50.2%) in P. gingivalis-induced calvarial lesion concomitant with a decreased inflammatory score evaluated through histomorphometric analysis, thus, improving soft tissue and bone healing response. Therefore, M101 may be a novel therapeutic agent that could be beneficial in the management of P. gingivalis associated diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Infecções por Bacteroidaceae/complicações , Abscesso Encefálico/tratamento farmacológico , Inflamação/tratamento farmacológico , Oxigênio/farmacologia , Poliquetos/química , Crânio/efeitos dos fármacos , Animais , Infecções por Bacteroidaceae/tratamento farmacológico , Infecções por Bacteroidaceae/microbiologia , Abscesso Encefálico/microbiologia , Abscesso Encefálico/patologia , Gengiva/química , Gengiva/microbiologia , Humanos , Inflamação/microbiologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/isolamento & purificação , Transdução de Sinais , Crânio/microbiologia , Crânio/patologia
12.
J Biosci Bioeng ; 130(6): 644-649, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32847740

RESUMO

Porphyromonas gingivalis, a major pathogen associated with chronic periodontitis, produces several virulence agents in the outer cell membrane, including gingipains and hemagglutinins. These virulence factors enable the bacteria to adhere to periodontal tissue and degrade host proteins to obtain the nutrients needed for dental plaque formation. P. gingivalis TDC60 was recently identified as the most aggressive P. gingivalis strain to dates. In this study, we isolated a known pregnane glycoside, argeloside I, from the aqueous extract of Solenostemma argel leaves. Argeloside I completely hindered the growth of P. gingivalis TDC60 and inhibited the production of hemagglutinins as well as Arg- and Lys-specific gingipains. Our results demonstrate a new function of pregnane glycosides. Argeloside I may be a candidate for reducing the risk associated with P. gingivalis TDC60 and its adhesion factors.


Assuntos
Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/patogenicidade , Cisteína Endopeptidases/metabolismo , Hemaglutininas/biossíntese , Humanos , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/metabolismo , Virulência/efeitos dos fármacos
13.
Biocontrol Sci ; 25(2): 45-53, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32507790

RESUMO

Povidone-iodine (PVP-I) is used for infection control and preoperative sterilization of the oral and pharyngeal regions. Marketed preparations containing cetylpyridinium chloride (CPC) are used to inhibit growth of oral bacteria. We conducted an in vitro study of the sterilizing effects of these microbicides on 10 oral bacterial strains and fungi related to pneumonia and periodontal disease, after dilution with phosphate-buffered saline (PBS), saliva, and components in saliva. The CPC solution was evaluated at 50 mg/100 mL, which is the concentration used in products. CPC sterilized all strains within 1 minute. Prolongation of the sterilization time associated with dilution was more gradual in comparison to PVP-I solution. CPC sterilized 7 of 10 microbial strains within 3 minutes at 3 mg/100 mL. At 500 mg/100 mL, which is near the upper limit of the concentration that is actually used, PVP-I solution sterilized 7 microbial strains within 3 minutes. However, PVP-I had no sterilization effect when diluted to 100 mg/100 mL or lower. With addition of saliva, PVP-I sterilized 2 microbial strains within 3 minutes at 500 mg/100 mL, whereas CPC solution sterilized 9 microbial strains within 1 minute at 50 mg/100 mL. Our results show that in use influenced by dilution with saliva, CPC is likely to maintain a strong sterilization effect, whereas PVP-I may have a reduced effect.


Assuntos
Anti-Infecciosos Locais/farmacologia , Cetilpiridínio/farmacologia , Povidona-Iodo/farmacologia , Esterilização/métodos , Candida albicans/efeitos dos fármacos , Candida albicans/crescimento & desenvolvimento , Clostridiales/efeitos dos fármacos , Clostridiales/crescimento & desenvolvimento , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/crescimento & desenvolvimento , Humanos , Testes de Sensibilidade Microbiana , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/crescimento & desenvolvimento , Saliva/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Streptococcus constellatus/efeitos dos fármacos , Streptococcus constellatus/crescimento & desenvolvimento , Streptococcus intermedius/efeitos dos fármacos , Streptococcus intermedius/crescimento & desenvolvimento , Streptococcus mutans/efeitos dos fármacos , Streptococcus mutans/crescimento & desenvolvimento , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/crescimento & desenvolvimento
14.
Nat Microbiol ; 5(8): 1016-1025, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32393857

RESUMO

Porphyromonas gingivalis, an asaccharolytic member of the Bacteroidetes, is a keystone pathogen in human periodontitis that may also contribute to the development of other chronic inflammatory diseases. P. gingivalis utilizes protease-generated peptides derived from extracellular proteins for growth, but how these peptides enter the cell is not clear. Here, we identify RagAB as the outer-membrane importer for these peptides. X-ray crystal structures show that the transporter forms a dimeric RagA2B2 complex, with the RagB substrate-binding surface-anchored lipoprotein forming a closed lid on the RagA TonB-dependent transporter. Cryo-electron microscopy structures reveal the opening of the RagB lid and thus provide direct evidence for a 'pedal bin' mechanism of nutrient uptake. Together with mutagenesis, peptide-binding studies and RagAB peptidomics, our work identifies RagAB as a dynamic, selective outer-membrane oligopeptide-acquisition machine that is essential for the efficient utilization of proteinaceous nutrients by P. gingivalis.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Oligopeptídeos/metabolismo , Porphyromonas gingivalis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Microscopia Crioeletrônica , Cristalografia por Raios X , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Simulação de Dinâmica Molecular , Periodontite/microbiologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/crescimento & desenvolvimento , Conformação Proteica
15.
Carbohydr Polym ; 237: 116163, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32241426

RESUMO

A polyelectrolyte complex nanoparticle comprising chitosan (CS) and carboxymethyl chitosan (CMCS) was prepared (CS/CMCS-NPs) by ionic gelation, which was then used as a doxycycline carrier (Dox:CS/CMCS-NPs). The obtained CS/CMCS-NPs and Dox:CS/CMCS-NPs were characterized for various parameters and bacteriostatic ability against Porphyromonas gingivalis. The regulation of related genes and proteins of NLRP3 inflammasome and IL-1ß in human gingival fibroblasts (HGFs) was characterized by qRT-PCR, western blotting and ELISA. The results showed that Dox:CS/CMCS-NPs had an orderly morphology and an excellent cytocompatibility. P. gingivalis was strongly inhibited by Dox:CS/CMCS-NPs contrasted with control group. Dox:CS/CMCS-NPs effectively down-regulated both gene and protein levels of NLRP3 inflammasome and IL-1ß in HGFs. This study provides a new method for rational application of Dox in the clinical treatment of periodontal disease and a new direction for explaining the mechanism of action of Dox:CS/CMCS-NPs and more drug-carrying nanoparticles.


Assuntos
Antibacterianos/administração & dosagem , Quitosana/análogos & derivados , Doxiciclina/administração & dosagem , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nanopartículas/administração & dosagem , Doenças Periodontais/metabolismo , Adolescente , Antibacterianos/química , Células Cultivadas , Quitosana/administração & dosagem , Quitosana/química , Doxiciclina/química , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Inflamassomos/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Nanopartículas/química , Doenças Periodontais/genética , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento
16.
Lasers Med Sci ; 35(6): 1403-1410, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32193820

RESUMO

Bactericidal and detoxification effects of diode laser (DL) have been reported in periodontal treatment. The objective of this study was investigating the additional effect of DL with nonsurgical periodontal treatment on the red complex bacteria in type 2 diabetes mellitus (DM) patients with chronic periodontitis (CP). Sixty type 2 DM patients with chronic periodontitis (CP) were randomly assigned in two parallel groups to receive scaling root planning (SRP, n = 30) or SRP followed by DL periodontal pocket irradiation (SRP + DL, n = 30). Recording of clinical parameters and subgingival plaque sampling were performed at baseline, and post therapy (1 and 3 months after treatment). Amounts of Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia were evaluated with quantitative RT-PCR. Significant reductions for numbers of all three bacterial species were observed at 1 and 3 months compared with baseline for both treatments (p < 0.001), but no significant differences were found between two groups regarding bacterial reductions at these follow-up time points. No additional benefit of DL as an adjunct to nonsurgical periodontal therapy was recognized in the reduction of P. gingivalis, T. denticola, and T. forsythia for type 2 DM patients with CP. Further studies are required to clarify the effects of diode laser on the other periodontopathogens.


Assuntos
Periodontite Crônica/microbiologia , Periodontite Crônica/cirurgia , Diabetes Mellitus Tipo 2/complicações , Lasers Semicondutores/uso terapêutico , Adulto , Periodontite Crônica/tratamento farmacológico , Contagem de Colônia Microbiana , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/efeitos da radiação , Aplainamento Radicular
17.
Curr Pharm Biotechnol ; 21(9): 862-871, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32081107

RESUMO

BACKGROUND AND OBJECTIVE: Dental peri-implantitis, which can be caused by several different microbial factors, is characterized by inflammatory lesions of the surrounding hard and soft tissues of an oral implant. Matrix Metalloproteinase 9 (MMP9) is thought to be involved in the pathogenesis of peri-implantitis. However, the regulatory mechanism of MMP9 in peri-implantitis has not been fully elucidated. In this study, we tried to evaluate the regulatory mechanism of MMP9 in peri-implantitis. METHODS: We collected Peri-Implant Crevicular Fluid (PICF) from ten healthy implants and ten periimplantitis patients and compared their expression level of MMP9. We also cultured macrophages from the peripheral blood of healthy volunteers infected by Porphyromonas gingivalis to reveal the regulatory mechanism of MMP9 in peri-implantitis. Western blot, immunofluorescence staining and quantitative Polymerase Chain Reaction (RT-PCR) were used to better characterize the mechanism of MMP9. RESULTS: The expression of MMP9 was up-regulated in peri-implantitis patient PICF and P. gingivalis infected human macrophages. LOX-1, not dectin-1, was found to mediate MMP9 expression in human macrophages with P. gingivalis infection. Expression of Erk1/2 was responsible for infection-induced MMP9 expression. Finally, use of a broad-spectrum metalloproteinase inhibitor impaired LOX-1 expression in infected macrophages. CONCLUSION: Our results demonstrate that MMP9 is involved in dental peri-implantitis and is regulated by LOX-1 and Erk1/2. This LOX-1/MMP9 signaling pathway may represent a potential drug target for peri-implantitis.


Assuntos
Implantes Dentários , Sistema de Sinalização das MAP Quinases/genética , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/genética , Peri-Implantite/metabolismo , Receptores Depuradores Classe E/metabolismo , Humanos , Macrófagos/microbiologia , Masculino , Peri-Implantite/etiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Regulação para Cima
18.
J Oleo Sci ; 69(2): 115-122, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32023578

RESUMO

A new difunctional Zn(II) coordination polymer (CP) with the chemical formula of [Zn(TBTA) (L)1.5]n (1) has been synthesized hydrothermally from tetrabromoterephthalic acid (H2TBTA) and 4,4'-bis(imidazole-1-yl)-biphenyl (L) ligands. Furthermore, due to its strong intense emission and open N donor sites, complex 1 could be used as a light-emitting sensor to determine 2,4,6-trinitrophenol (TNP) which has high selectivity and sensitivity. Furthermore, the anti-bacterial effect of the compound against P. gingivalis in vitro was evaluated by measuring the P. gingivalis growth curves after compound treatment. And the RT-PCR assay was performed to detect the relative expression of ragA and ragB, which are important for the P. gingivalis growth. The potential anti-infectious mechanism was further studied by using molecular docking technique.


Assuntos
Doenças Periodontais/tratamento farmacológico , Porphyromonas gingivalis/crescimento & desenvolvimento , Trinitrobenzenos/química , Trinitrobenzenos/uso terapêutico , Compostos de Zinco/química , Compostos de Zinco/uso terapêutico , Depressão Química , Humanos , Ligantes , Doenças Periodontais/microbiologia , Polímeros , Trinitrobenzenos/farmacologia , Compostos de Zinco/farmacologia
19.
Clin Oral Investig ; 24(4): 1465-1477, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31940064

RESUMO

OBJECTIVES: This study presents a surface modification method to treat the zirconia implant abutment materials using a helium cold atmospheric plasma (CAP) jet in order to evaluate its efficacy on oral bacteria adhesion and growth. MATERIALS AND METHODS: Yttrium-Stabilized Zirconia disks were subjected to helium CAP treatment; after the treatment, zirconia surface was evaluated using scanning electron microscopy, a contact angle measuring device, X-ray photoelectron spectroscopy for surface characteristics. The response of Streptococcus mutans and Porphyromonas gingivalis on treated surface was evaluated by a scanning electron microscopy, MTT assay, and LIVE/DEAD staining. The biofilm formation was analyzed using a crystal violet assay. RESULTS: After the helium CAP jet treatment, the zirconia surface chemistry has been changed while the surface topography remains unchanged, the bacterial growth was inhibited, and the biofilm forming decreased. As the treatment time increases, the zirconia abutment showed a better bacterial inhibition efficacy. CONCLUSIONS: The helium CAP jet surface modification approach can eliminate bacterial growth on zirconia surface with surface chemistry change, while surface topography remained. CLINICAL RELEVANCE: Soft tissue seal around dental implant abutment plays a crucial role in maintaining long-term success. However, it is weaker than periodontal barriers and vulnerable to bacterial invasion. CAP has a potential prospect for improving soft tissue seal around the zirconia abutment, therefore providing better esthetics and most of all, prevent peri-implant lesions from happening.


Assuntos
Aderência Bacteriana , Materiais Dentários , Hélio , Gases em Plasma , Porphyromonas gingivalis/crescimento & desenvolvimento , Streptococcus mutans/crescimento & desenvolvimento , Zircônio , Temperatura Baixa , Dente Suporte , Estética Dentária , Teste de Materiais , Microscopia Eletrônica de Varredura , Propriedades de Superfície
20.
FEMS Microbiol Lett ; 366(16)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31578552

RESUMO

Chronic periodontitis is caused by dysbiosis of human oral commensals and especially by increase in Porphyromonas gingivalis. Inhibitors of P. gingivalis growth are expected to serve as effective drugs for the periodontal therapy. In the present study, we isolated new growth inhibitors of P. gingivalis using minimal media for P. gingivalis. The minimal media included the previously reported Globulin-Albumin (GA) and the newly developed Lactalbumin-Ferric chloride (LF) and Globulin-Calcium chloride (GC); all supported growth of the wild-type strain of P. gingivalis but did not support the growth of a mutant defective for a type IX secretion system. GC contains CaCl2, indicating that P. gingivalis requires a calcium ion for growth. Using LF and GA, we screened about 100 000 compounds and identified 73 that strongly inhibited the growth of P. gingivalis. More than half of these candidates would not have been obtained if these minimal media had not been used in our screen. One of our candidate inhibitors was diphenyleneiodonium chloride (DPIC), which showed strong bactericidal activity against P. gingivalis. Excess amounts of flavin adenine dinucleotide or flavin mononucleotide suppressed the inhibitory activity of DPIC, suggesting that DPIC would be a novel potent growth inhibitor.


Assuntos
Antibacterianos/metabolismo , Meios de Cultura/química , Dinitrocresóis/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos/métodos , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana/métodos , Viabilidade Microbiana/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...