Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
1.
Microbiol Res ; 282: 127655, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38402726

RESUMEN

Quorum sensing (QS), an integral component of bacterial communication, is essential in coordinating the collective response of diverse bacterial pathogens. Central carbon metabolism (CCM), serving as the primary metabolic hub for substances such as sugars, lipids, and amino acids, plays a crucial role in the life cycle of bacteria. Pathogenic bacteria often utilize CCM to regulate population metabolism and enhance the synthesis of specific cellular structures, thereby facilitating in adaptation to the host microecological environment and expediting infection. Research has demonstrated that QS can both directly or indirectly affect the CCM of numerous pathogenic bacteria, thus altering their virulence and pathogenicity. This article reviews the interplay between QS and CCM in Gram-positive pathogenic bacteria, details the molecular mechanisms by which QS modulates CCM, and lays the groundwork for investigating bacterial pathogenicity and developing innovative infection treatment drugs.


Asunto(s)
Bacterias Gramnegativas , Percepción de Quorum , Percepción de Quorum/fisiología , Bacterias Gramnegativas/fisiología , Bacterias , Bacterias Grampositivas/fisiología , Virulencia
2.
Int J Mol Sci ; 24(12)2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37373533

RESUMEN

Porphyromonas gingivalis is a periodontopathogenic bacterium that can adhere to and colonize periodontal tissues, leading to an inflammatory process, and, consequently, tissue destruction. New therapies using flavonoids, such as hesperidin, are being studied, and their promising properties have been highlighted. The aim of this study was to evaluate the effect of hesperidin on the epithelial barrier function, reactive oxygen species (ROS) production, and on the inflammatory response caused by P. gingivalis in in vitro models. The integrity of the epithelial tight junctions challenged by P. gingivalis was determined by monitoring the transepithelial electrical resistance (TER). P. gingivalis adherence to a gingival keratinocyte monolayer and a basement membrane model were evaluated by a fluorescence assay. A fluorometric assay was used to determine the ROS production in gingival keratinocytes. The level of pro-inflammatory cytokines and matrix metalloproteinases (MMPs) secretion was evaluated by ELISA; to assess NF-κB activation, the U937-3xjB-LUC monocyte cell line transfected with a luciferase reporter gene was used. Hesperidin protected against gingival epithelial barrier dysfunction caused by P. gingivalis and reduced the adherence of P. gingivalis to the basement membrane model. Hesperidin dose-dependently inhibited P. gingivalis-mediated ROS production by oral epithelial cells as well as the secretion of IL-1ß, TNF-α, IL-8, MMP-2, and MMP-9 by macrophages challenged with P. gingivalis. Additionally, it was able to attenuate NF-κB activation in macrophages stimulated with P. gingivalis. These findings suggest that hesperidin has a protective effect on the epithelial barrier function, in addition to reducing ROS production and attenuating the inflammatory response associated with periodontal disease.


Asunto(s)
Hesperidina , Porphyromonas gingivalis , Porphyromonas gingivalis/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hesperidina/farmacología , FN-kappa B/metabolismo , Células Epiteliales/metabolismo , Macrófagos/metabolismo , Encía
3.
Front Microbiol ; 13: 999839, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36406439

RESUMEN

Dental caries and periodontal diseases remain a challenge for oral health, especially given the lack of effective and safe treatment options that are currently available. Against the backdrop of an ongoing antimicrobial resistance crisis, a renewed interest in traditional medicinal plants as a potential source of new bioactive compounds has surfaced. In this context, we systematically screened the antimicrobial and anti-biofilm activities of both ethanolic and aqueous extracts of nine Algerian medicinal plants (Artemisia herba alba, Centaurium erythraea, Juglans regia, Laurus nobilis, Matricaria recutita, Mentha pulegium, Mentha piperita, Origanum vulgare and Taraxacum officinale). To evaluate the activity spectrum of the extracts, the screening was carried out against an extensive collection of Streptococcus, Enterococcus and Lacticaseibacillus isolates recovered from dental plaques of Algerian patients. Broad-spectrum antimicrobial and anti-biofilm properties were observed, especially among ethanolic extracts, which marks them as a promising source for bioactive compounds to control oral biofilms. The ethanolic extract of O. vulgare, which showed the most promising effects in the initial screening, was further characterized. We first verified the biocompatibility of this extract using human oral keratinocytes and selected a range of non-cytotoxic concentrations (0.195-0.781 mg/ml) to further validate its anti-biofilm and anti-virulence potential. At these concentrations, the extract not only prevented biofilm formation (10.04 ± 0.75-87.91 ± 9.08% of reduction) of most dental plaque isolates on a polystyrene surface, but also significantly reduced their adherence to hydroxyapatite (34.58 ± 9.09-62.77 ± 0.95%). Moreover, the extract showed curative potential against mature biofilms grown under conditions mimicking the oral niche. In addition to its anti-biofilm properties, we observed an inhibition of glucosyltransferase activity, a reduction in acidogenesis and a downregulation in the expression of multiple virulence-associated genes for extract-treated samples. Since anti-virulence properties are more robust to the development of resistance, they provide an attractive complementation to the antimicrobial activities of the extract. Thymol was identified as an important active compound of the extract using GC-MS analysis, but synergy with other compounds was also detected, suggesting a potential advantage of using the whole extract over purified thymol. Further research into the bioactive compounds of the O. vulgare ethanolic extract could yield novel products to fight dental caries.

4.
J Antimicrob Chemother ; 77(12): 3275-3282, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36173390

RESUMEN

BACKGROUND: The increased resistance of bacterial pathogens to fluoroquinolones (FQs), such as norfloxacin and ciprofloxacin, supports the need to develop new antibacterial drugs and combination therapies using conventional antibiotics. The LuxS/AI-2 quorum sensing (QS) system can regulate the complex group behaviour of Streptococcus suis and impact its susceptibility to FQs. OBJECTIVES: We investigated the combination of paeoniflorin and norfloxacin as a novel and effective strategy against FQ-resistant S. suis. METHODS: FIC, AI-2 activity assay, real-time RT-PCR and biofilm inhibition assays were performed to investigate the in vitro effect of paeoniflorin combined with norfloxacin. Mouse protection and mouse anti-infection assays were performed to investigate the in vivo effect of paeoniflorin combined with norfloxacin. RESULTS: FIC results showed that paeoniflorin and norfloxacin exert a synergistic bactericidal effect. Evidence was brought that paeoniflorin reduces the S. suis AI-2 activity and significantly down-regulates the transcription of the FQ efflux pump gene. In addition, paeoniflorin can inhibit biofilm formation, thereby promoting the ability of norfloxacin to kill S. suis. Finally, we showed in a mouse model that paeoniflorin in association with norfloxacin is effective to treat S. suis infections. CONCLUSIONS: This study highlighted the inhibitory potential of paeoniflorin on the LuxS/AI-2 QS system of S. suis, and provided evidence that it can inhibit the FQ efflux pump and prevent biofilm formation to cooperate with norfloxacin in the treatment of resistant S. suis-related infections.


Asunto(s)
Antibacterianos , Monoterpenos , Norfloxacino , Infecciones Estreptocócicas , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Biopelículas , Fluoroquinolonas/farmacología , Glucósidos/farmacología , Monoterpenos/farmacología , Monoterpenos/uso terapéutico , Norfloxacino/farmacología , Norfloxacino/uso terapéutico , Streptococcus suis , Infecciones Estreptocócicas/tratamiento farmacológico , Farmacorresistencia Bacteriana
5.
Front Cell Infect Microbiol ; 12: 898412, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35992166

RESUMEN

Respiratory infections seriously affect the swine industry worldwide. Co-infections of two vital pathogenic bacteria Streptococcus suis (S. suis) and Actinobacillus pleuropneumoniae (A. pleuropneumoniae), colonizing the respiratory tract often occurs in veterinary clinical practice. Moreover, our previous research found that S. suis and A. pleuropneumoniae can form biofilm in vitro. The formation of a mixed biofilm not only causes persistent infections, but also increases the multiple drug resistance of bacteria, which brings difficulties to disease prevention and control. However, the methods for detecting S. suis and A. pleuropneumoniae in co-infection and biofilm are immature. Therefore, in this study, primers and probes were designed based on the conservative sequence of S. suis gdh gene and A. pleuropneumoniae apxIVA gene. Then, a TaqMan duplex real-time PCR method for simultaneous detection of S. suis and A. pleuropneumoniae was successfully established via optimizing the reaction system and conditions. The specificity analysis results showed that this TaqMan real-time PCR method had strong specificity and high reliability. The sensitivity test results showed that the minimum detection concentration of S. suis and A. pleuropneumoniae recombinant plasmid was 10 copies/µL, which is 100 times more sensitive than conventional PCR methods. The amplification efficiencies of S. suis and A. pleuropneumoniae were 95.9% and 104.4% with R2 value greater than 0.995, respectively. The slopes of the calibration curves of absolute cell abundance of S. suis and A. pleuropneumoniae were 1.02 and 1.09, respectively. The assays were applied to cultivated mixed biofilms and approximately 108 CFUs per biofilm were quantified when 108 CFUs planktonic bacteria of either S. suis or A. pleuropneumoniae were added to biofilms. In summary, this study developed a TaqMan real-time PCR assay for specific, accurate quantification of S. suis or A. pleuropneumoniae in mixed biofilms, which may help for the detection, prevention and control of diseases caused by a bacterial mixed infection involving S. suis and A. pleuropneumoniae.


Asunto(s)
Infecciones por Actinobacillus , Actinobacillus pleuropneumoniae , Coinfección , Streptococcus suis , Enfermedades de los Porcinos , Infecciones por Actinobacillus/diagnóstico , Infecciones por Actinobacillus/microbiología , Infecciones por Actinobacillus/veterinaria , Actinobacillus pleuropneumoniae/genética , Animales , Biopelículas , Coinfección/diagnóstico , Coinfección/veterinaria , Reproducibilidad de los Resultados , Streptococcus suis/genética , Porcinos , Enfermedades de los Porcinos/diagnóstico , Enfermedades de los Porcinos/microbiología
6.
Front Oral Health ; 3: 923663, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784661

RESUMEN

Porphyromonas gingivalis expresses a broad array of virulence factors that enable it to play a central role in the etiopathogenesis of periodontitis. The objective of the present study was to assess the effects of a berry polyphenolic fraction (Orophenol®) composed of extracts from cranberry, wild blueberry, and strawberry on the main pathogenic determinants of P. gingivalis. Orophenol® attenuated the growth of P. gingivalis and decreased its hemolytic activity, its adherence to a basement membrane matrix model, and its proteinase activities. The berry polyphenolic fraction also impaired the production of reactive oxygen species (ROS) by oral keratinocytes stimulated with P. gingivalis. Lastly, using an in vitro model of oral keratinocyte barrier, the fraction exerted a protective effect against the damages mediated by P. gingivalis. In conclusion, the berry polyphenolic fraction investigated in the present study attenuated several pathogenic properties of P. gingivalis. Although future clinical investigations are required, our study provided evidence that the polyphenols contained in this fraction may represent bioactive molecules of high interest for the prevention and/or treatment of periodontal disease.

7.
Front Oral Health ; 3: 867793, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35392377

RESUMEN

Periodontitis, an inflammatory disease that affects tooth-supporting tissues, is the result of a polymicrobial infection involving mainly Gram negative anaerobic bacteria. The aim of the present study was to investigate the effects of a phenolic-rich extract of cocoa (Theobroma cacao L.) beans on the pathogenic properties of Porphyromonas gingivalis, which is well-known as a keystone pathogen in the development of periodontitis. The effect of the cocoa extract on P. gingivalis-induced activation of the nuclear factor kappa B (NF-κB) transcription factor in a monocyte model was also assessed. The cocoa extract, whose major phenolic compound was epicatechin, inhibited the growth, hemolytic activity, proteolytic activities, and adherence properties (basement membrane matrix, erythrocytes) of P. gingivalis in a dose-dependent manner. It also protected the barrier function of a keratinocyte model against the deleterious effects mediated by P. gingivalis, and attenuated reactive oxygen species (ROS) production by oral keratinocytes treated with P. gingivalis. Lastly, the cocoa extract showed an anti-inflammatory property by preventing P. gingivalis-induced NF-κB activation in monocytes. In conclusion, this in vitro study highlighted the potential value of an epicatechin-rich extract of cocoa beans for preventing and/or treating periodontal diseases.

8.
Biotechnol Adv ; 59: 107965, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35487393

RESUMEN

Quorum sensing (QS), which is a key part of cell/cell communication, is widely distributed in microorganisms, especially in bacteria. Bacteria can produce and detect the presence of QS signal molecule, perceive the composition and density of microorganisms in their complex habitat, and then dynamically regulate their own gene expression to adapt to their environment. Among the many traits controlled by QS in pathogenic bacteria is the expression of virulence factors and antibiotic resistance. Many pathogenic bacteria rely on QS to govern the production of virulence factors and express drug-resistance, especially in zoonotic bacteria. The threat of antibiotic resistant zoonotic bacteria has called for alternative antimicrobial strategies that would mitigate the increase of classical resistance mechanism. Targeting QS has proven to be a promising alternative to conventional antibiotic for controlling infections. Here we review the QS systems in common zoonotic pathogenic bacteria and outline how QS may control the virulence and antibiotic resistance of zoonotic bacteria.


Asunto(s)
Percepción de Quorum , Factores de Virulencia , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias/metabolismo , Farmacorresistencia Microbiana/genética , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Factores de Virulencia/farmacología
9.
Front Oral Health ; 3: 847914, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35295880

RESUMEN

Porphyromonas gingivalis is a key pathogen of periodontitis, an inflammatory disease that affects the tooth-supporting tissues. The aim of the present study was to investigate the effects of the flavanone eriodictyol on P. gingivalis-induced reactive oxygen species (ROS) production by gingival keratinocytes and the inflammatory response of macrophages. Porphyromonas gingivalis and H2O2 acted synergistically to induce ROS production by keratinocytes. The presence of eriodictyol significantly attenuated ROS production in a dose-dependent manner. We used a macrophage model to show that eriodictyol decreases the secretion of IL-1ß, IL-6, IL-8, and TNF-α induced by P. gingivalis. Evidence has been brought that this anti-inflammatory property of eriodictyol may be related to its ability to prevent the activation of the NF-κB signaling pathway by P. gingivalis. This periodontal pathogen was also found to be a potent inducer of matrix metalloproteinase (MMP) production by macrophages, including MMP-2, MMP-8, and MMP-9. Eriodictyol dose-dependently inhibited the production of all three MMPs. Lastly, eriodictyol inhibited the catalytic activity of both MMP-9 and P. gingivalis collagenase. In conclusion, eriodictyol may be a potential therapeutic agent for preventing and/or treating periodontal disease due to its antioxidant, anti-inflammatory, and anti-proteinase properties.

10.
J Agric Food Chem ; 70(2): 429-445, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-34989570

RESUMEN

Quorum sensing (QS) is a signaling mechanism for cell-to-cell communication between bacteria, fungi, and even eukaryotic hosts such as plant and animal cells. Bacteria in real life do not exist as isolated organisms but are found in complex, dynamic, and microecological environments. The study of interspecies QS and interkingdom QS is a valuable approach for exploring bacteria-bacteria interactions and bacteria-host interaction mechanisms and has received considerable attention from researchers. The correct combination of QS signals and receptors is key to initiating the QS process. Compared with intraspecies QS, the signal regulation mechanism of interspecies QS and interkingdom QS is often more complicated, and the distribution of receptors is relatively wide. The present review focuses on the latest progress with respect to the distribution, structure, and signal transduction of interspecies and interkingdom QS receptors and provides a guide for the investigation of new QS receptors in the future.


Asunto(s)
Bacterias , Percepción de Quorum , Bacterias/genética , Plantas , Transducción de Señal
11.
Probiotics Antimicrob Proteins ; 14(1): 204-215, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35067836

RESUMEN

In addition to be an important zoonotic agent, Streptococcus suis serotype 2 causes severe infections in pigs. In this study, we characterized a new bacteriocin produced by Streptococcus pluranimalium 2N12 isolated from a pig nasal sample. The bacteriocin, termed pluranimalicin 2N12, was a two-peptide class IIb bacteriocin active against S. suis. The gene cluster responsible for the biosynthesis of pluranimalicin 2N12 by S. pluranimalium contained seven open reading frames, including putative genes for peptides (pluα, pluß), export (pluA, pluB), and regulation (pluC, pluD, pluE). The deduced amino acid sequences of the peptides Pluα (33 amino acids) and Pluß (29 amino acids) showed 73% and 69% identity in amino acid residues, respectively, with the peptides SthA and SthB of the streptocin produced by Streptococcus gordonii. The antibacterial activity of pluranimalicin 2N12 against S. suis was dependent on the presence of the two peptides Pluα and Pluß that exhibited a membrane permeabilization effect. No activity was found against the other swine pathogens tested. Depending on the concentrations used, Pluα and Pluß displayed no or low toxicity towards swine tracheal epithelial cells. The pluranimalicin peptides Pluα and Pluß, either individually or in combination, exhibited anti-inflammatory activity since they attenuated IL-6 and TNF-α production by macrophages challenged with lipopolysaccharide. Given its dual action (antibacterial and anti-inflammatory), pluranimalicin 2N12 holds promise as a potential therapeutic agent for controlling S. suis infections.


Asunto(s)
Bacteriocinas , Cavidad Nasal , Streptococcus suis , Animales , Cavidad Nasal/microbiología , Péptidos/metabolismo , Péptidos/farmacología , Streptococcus , Streptococcus suis/genética , Streptococcus suis/metabolismo , Porcinos
12.
Sci Rep ; 11(1): 23724, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34887426

RESUMEN

The mouth environment comprises the second most significant microbiome in the body, and its equilibrium is critical in oral health. Secretory calcium-binding phosphoprotein proline-glutamine rich 1 (SCPPPQ1), a protein normally produced by the gingival epithelium to mediate its attachment to teeth, was suggested to be bactericidal. Our aim was to further explore the antibacterial potential of human SCPPPQ1 by characterizing its mode of action and identifying its active portions. In silico analysis showed that it has molecular parallels with antimicrobial peptides. Incubation of Porphyromonas gingivalis, a major periodontopathogen, with the full-length protein resulted in decrease in bacterial number, formation of aggregates and membrane disruptions. Analysis of SCPPPQ1-derived peptides indicated that these effects are sustained by specific regions of the molecule. Altogether, these data suggest that human SCPPPQ1 exhibits antibacterial capacity and provide new insight into its mechanism of action.


Asunto(s)
Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/farmacología , Fosfoproteínas/química , Fosfoproteínas/farmacología , Porphyromonas gingivalis/efectos de los fármacos , Secuencia de Aminoácidos , Péptidos Antimicrobianos/biosíntesis , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/farmacología , Proteínas de Unión al Calcio/metabolismo , Resistencia a la Enfermedad , Interacciones Huésped-Patógeno , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Fosfoproteínas/metabolismo , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
13.
Front Vet Sci ; 8: 787241, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34957284

RESUMEN

Actinobacillus pleuropneumoniae is the causal agent of porcine pleuropneumonia, a highly contagious and often deadly respiratory disease that causes major economic losses in the swine industry worldwide. The aim of the present study was to investigate the hydrogen peroxide (H2O2)-dependent antagonistic activity of Streptococcus pluranimalium 2N12 (pig nasal isolate) against A. pleuropneumoniae. A fluorimetric assay showed that S. pluranimalium produces H2O2 dose- and time-dependently. The production of H2O2 increased in the presence of exogenous lactate, suggesting the involvement of lactate oxidase. All 20 strains of A. pleuropneumoniae tested, belonging to 18 different serovars, were susceptible to H2O2, with minimal inhibitory concentrations and minimal bactericidal concentrations ranging from 0.57 to 2.3 mM. H2O2, as well as a culture supernatant of S. pluranimalium, killed planktonic cells of A. pleuropneumoniae. Treating the culture supernatant with catalase abolished its bactericidal property. H2O2 was also active against a pre-formed biofilm-like structure of A. pleuropneumoniae albeit to a lesser extent. A checkerboard assay was used to show that there were antibacterial synergistic interactions between H2O2 and conventional antibiotics, more particularly ceftiofur. Based on our results and within the limitations of this in vitro study, the production of H2O2 by S. pluranimalium could be regarded as a potential protective mechanism of the upper respiratory tract against H2O2-sensitive pathogens such as A. pleuropneumoniae.

14.
Virulence ; 12(1): 3062-3073, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34923916

RESUMEN

Streptococcus suis (S. suis), more specifically serotype 2, is a bacterial pathogen that threatens the lives of pigs and humans. Like many other pathogens, S. suis exhibits quorum sensing (QS) system-controlled virulence factors, such as biofilm formation that complicates treatment. Therefore, impairing the QS involving LuxS/AI-2 cycle in S. suis, may be a promising alternative strategy for overcoming S. suis infections. In this study, we investigated paeoniflorin (PF), a monoterpenoid glycoside compound extracted from peony, as an inhibitor of S. suis LuxS/AI-2 system. At a sub-minimal inhibitory concentration (MIC) (1/16 MIC; 25 µg/ml), PF significantly reduced biofilm formation by S. suis through inhibition of extracellular polysaccharide (EPS) production, without affecting bacterial growth. Moreover, evidence was brought that PF reduces AI-2 activity in S. suis biofilm. Molecular docking indicated that LuxS may be the target of PF. Monitoring LuxS enzymatic activity confirmed that PF had a partial inhibitory effect. Finally, we showed that the use of PF in a mouse model can relieve S. suis infections. This study highlighted the anti-biofilm potential of PF against S. suis, and brought evidence that it may as an inhibitor of the LuxS/AI-2 system to prevent S. suis biofilm-related infections. PF can thus be used as a new type of natural biofilm inhibitor for clinical application.


Asunto(s)
Streptococcus suis , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Biopelículas , Liasas de Carbono-Azufre/genética , Liasas de Carbono-Azufre/farmacología , Glucósidos , Homoserina , Lactonas/farmacología , Ratones , Simulación del Acoplamiento Molecular , Monoterpenos/farmacología , Percepción de Quorum , Porcinos , Virulencia
15.
ACS Omega ; 6(42): 27791-27797, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34722979

RESUMEN

Quebecol is a polyphenolic compound initially isolated from Canadian maple syrup in 2011. Recently, our group demonstrated in a macrophage model that quebecol inhibits the secretion of pro-inflammatory cytokines and reduces the activation of the NF-κB transcription factor. In this study, we further explored the therapeutic potential of quebecol against periodontal disease, an inflammatory disorder of bacterial origin affecting tooth-supporting tissues. More specifically, the effects of this natural compound on matrix metalloproteinase (MMP) activity and macrophage secretion, as well as on the mineralization activity of osteoblasts (bone-forming cells), were investigated. Results showed that exposing lipopolysaccharide (LPS)-treated macrophages to quebecol led to a significant decrease in the secretion of MMP-8 and MMP-9. In addition, quebecol dose dependently inhibited the catalytic activity of MMP-9. Quebecol also enhanced the mineralization activity of osteoblasts. This study brought forward additional evidence to support the potential of quebecol as a nutraceutical agent against periodontitis.

16.
Food Funct ; 12(21): 10470-10483, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34554173

RESUMEN

Cranberries are widely recognized as a functional food that can promote oral health. However, the high concentration of organic acids in cranberry juice can cause tooth enamel erosion. Electrodialysis with bipolar membrane (EDBM) is a process used for the deacidification of cranberry juice. The present study investigated whether the removal of organic acids (0%, 19%, 42%, 60%, and 79%) from cranberry juice by EDBM affects its antibacterial activity against major periodontopathogens as well as its anti-inflammatory properties in an oral epithelial cell model. A deacidification rate ≥60% attenuated the bactericidal effect against planktonic and biofilm-embedded Aggregatibacter actinomycetemcomitans but had no impact on Porphyromonas gingivalis and Fusobacterium nucleatum. Cranberry juice increased the adherence of A. actinomycetemcomitans and P. gingivalis to oral epithelial cells, but reduced the adherence of F. nucleatum by half regardless of the deacidification rate. F. nucleatum produced more hydrogen sulfide when it was exposed to deacidified cranberry juice with a deacidification rate ≥42% compared to the raw beverage. Interestingly, the removal of organic acids from cranberry juice lowered the cytotoxicity of the beverage for oral epithelial cells. Deacidification attenuated the anti-inflammatory effect of cranberry juice in an in vitro oral epithelial cell model. The secretion of IL-6 by lipopolysaccharide (LPS)-stimulated oral epithelial cells exposed to cranberry juice increased proportionally with the deacidification rate. No such effect was observed with respect to the production of IL-8. This study provided evidence that organic acids, just like phenolic compounds, might contribute to the health benefits of cranberry juice against periodontitis.


Asunto(s)
Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Antibacterianos/farmacología , Antiinflamatorios/farmacología , Fusobacterium nucleatum/efectos de los fármacos , Extractos Vegetales/farmacología , Porphyromonas gingivalis/efectos de los fármacos , Vaccinium macrocarpon/química , Ácidos/química , Antibacterianos/química , Antiinflamatorios/química , Biopelículas , Células Cultivadas , Técnicas Electroquímicas/métodos , Células Epiteliales/efectos de los fármacos , Jugos de Frutas y Vegetales , Extractos Vegetales/química
17.
Foods ; 10(7)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34359504

RESUMEN

Cranberry (Vaccinium macrocarpon) may be a potent natural adjuvant for the prevention of oral diseases due to its anti-adherence, anti-cariogenic, and anti-inflammatory properties. However, the high titrable acidity of cranberry juice (CJ) has been reported to cause gastrointestinal discomfort, leading consumers to restrict their intake of this beverage. Electrodialysis with a bipolar membrane (EDBM) can reduce the organic acid content of CJ while retaining the flavonoids associated with potential health benefits. This study aimed to assess how the deacidification of CJ by EDBM impacts the antibacterial properties of the beverage against cariogenic (Streptococcus mutans, Streptococcus sobrinus) and commensal (Streptococcus gordonii, Streptococcus oralis, Streptococcus salivarius) streptococci, and how it affects oral epithelial barrier function and inflammatory response in an in vitro model. The removal of organic acids from CJ (deacidification rate ≥42%) reduced the bactericidal activity of the beverage against planktonic S. mutans and S. gordonii after a 15-min exposure, whereas only the viability of S. gordonii was significantly impacted by CJ deacidification rate when the bacteria were embedded in a biofilm. Moreover, conditioning saliva-coated hydroxyapatite with undiluted CJ samples significantly lowered the adherence of S. mutans, S. sobrinus, and S. oralis. With respect to epithelial barrier function, exposure to CJ deacidified at a rate of ≥19% maintained the integrity of a keratinocyte monolayer over the course of 24 h compared to raw CJ, as assessed by the determination of transepithelial electrical resistance (TER) and fluorescein isothiocyanate-conjugated dextran paracellular transport. These results can be in part attributed to the inability of the deacidified CJ to disrupt two tight junction proteins, zonula occludens-1 and occludin, following exposure, unlike raw CJ. Deacidification of CJ impacted the secretion of IL-6, but not of IL-8, by oral epithelial cells. In conclusion, deacidification of CJ appears to provide benefits with respect to the maintenance of oral health.

18.
PLoS One ; 16(5): e0252029, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34029354

RESUMEN

Fusobacterium nucleatum is associated with many conditions and diseases, including periodontal diseases that affect tooth-supporting tissues. The aim of the present study was to investigate the effects of a cocoa extract (Theobroma cacao L.) on F. nucleatum with respect to growth, biofilm formation, adherence, and hydrogen sulfide (H2S) production. The anti-inflammatory properties and the effect on epithelial barrier function of the cocoa extract were also assessed. The cocoa extract, whose major phenolic compound is epicatechin, dose-dependently inhibited the growth, biofilm formation, adherence properties (basement membrane matrix, oral epithelial cells), and H2S production of F. nucleatum. It also decreased IL-6 and IL-8 production by F. nucleatum-stimulated oral epithelial cells and inhibited F. nucleatum-induced NF-κB activation in monocytes. Lastly, the cocoa extract enhanced the barrier function of an oral epithelial model by increasing the transepithelial electrical resistance. We provide evidence that the beneficial properties of an epicatechin-rich cocoa extract may be useful for preventing and/or treating periodontal diseases.


Asunto(s)
Cacao/química , Fusobacterium nucleatum/efectos de los fármacos , Enfermedades Periodontales/tratamiento farmacológico , Fenoles/farmacología , Biopelículas/efectos de los fármacos , Catequina/química , Catequina/farmacología , Adhesión Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Fusobacterium nucleatum/patogenicidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Sulfuro de Hidrógeno/metabolismo , Interleucina-6/genética , Interleucina-8/genética , Monocitos/efectos de los fármacos , Enfermedades Periodontales/microbiología , Fenoles/química , Extractos Vegetales/química , Extractos Vegetales/farmacología
19.
Arch Oral Biol ; 126: 105135, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33930649

RESUMEN

OBJECTIVE: To investigate the ability of a green tea extract and epigallocatechin-3-gallate (EGCG) to protect oral epithelial cells against the deleterious effects of the chemotherapeutic agent irinotecan, with respect to cytotoxicity; reactive oxygen species (ROS) generation; cytokine and matrix metalloproteinase (MMP) production; and cell proliferation and migration. METHODS: The B11 oral keratinocyte and GMSM-K oral epithelial cell lines were used in this study. Cell viability was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay. A fluorometric assay was used to quantify ROS production. Cell proliferation was assessed using a fluorescent cell tracker dye, while a migration assay kit was used to monitor cell migration. Cytokine and MMP secretion was quantified by an enzyme-linked immunosorbent assay. RESULTS: The green tea extract and EGCG reduced the cytotoxicity of irinotecan toward oral keratinocyte and epithelial cell lines. Irinotecan-induced intracellular ROS generation by oral keratinocytes was reduced by the green tea extract and EGCG. Irinotecan negatively affected the proliferation and migration of oral keratinocytes in a dose-dependent manner. However, these effects were not neutralized by the green tea extract, while EGCG showed a trend to attenuate the irinotecan-induced decrease in cell migration. The green tea extract and EGCG also had a dose-dependent inhibitory effect on irinotecan-induced secretion of interleukin-6 and interleukin-8 by oral epithelial cells. Lastly, the irinotecan-induced decrease in the secretion of MMP-2 and MMP-9 by oral epithelial cells was partially restored by the green tea extract and EGCG. CONCLUSIONS: The green tea extract and EGCG, through anti-cytotoxic, anti-oxidative, and anti-inflammatory properties, may protect the oral mucosa against the deleterious effects of the chemotherapeutic agent irinotecan and may be of interest for treating oral mucositis.


Asunto(s)
Catequina , , Catequina/análogos & derivados , Catequina/farmacología , Células Epiteliales , Irinotecán/toxicidad , Extractos Vegetales/farmacología
20.
Arch Oral Biol ; 126: 105126, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33872861

RESUMEN

OBJECTIVE: To investigate the effects of Dual Zinc plus Arginine formulations (aqueous solution and dentifrice) on tumor necrosis factor-alpha (TNF-α)-induced barrier dysfunction as well as on cell proliferation and migration in an in vitro gingival keratinocyte model. DESIGN: Gingival keratinocytes were seeded onto the membrane of a double-chamber system in the absence and presence of recombinant TNF-α and the formulations under investigation. The barrier function was assessed by determination of transepithelial electrical resistance (TER) and paracellular transport of fluorescein isothiocyanate (FITC)-dextran. The distribution of zonula occludens-1 (ZO-1) and occludin was visualized by immunofluorescence microscopy. The effects of the formulations on keratinocyte cell proliferation were determined using a fluorescent cell tracker dye, while a migration assay kit was used to investigate their effects on cell migration. RESULTS: Under conditions where TNF-α induces loss of keratinocyte barrier integrity, the Dual Zinc plus Arginine formulations (aqueous solution and dentifrice) protected the keratinocyte tight junction against the damages since they prevented the TNF-α-induced drop in TER and increase in FITC-dextran paracellular flux in the in vitro model. The treatment of keratinocytes with the formulations markedly mitigated the altered distribution of ZO-1 and occludin. Both formulations increased the proliferation of keratinocytes and alleviated the negative impact caused by TNF-α. Lastly, the formulations increased the migration capacity of keratinocytes. CONCLUSIONS: The ability of the Dual Zinc plus Arginine formulations to protect the barrier integrity of gingival keratinocytes from TNF-α-induced damage and to promote their proliferation and migration suggests that they may offer benefits for oral health.


Asunto(s)
Arginina , Factor de Necrosis Tumoral alfa , Arginina/farmacología , Proliferación Celular , Mucosa Intestinal , Queratinocitos , Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...