Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.433
Filtrar
1.
Sci Rep ; 14(1): 10882, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740792

RESUMEN

The aim of this study was to evaluate the antimicrobial efficacy of an air gas soft jet CAP for its potential use in removing oral biofilms, given that plasma-based technologies have emerged as promising methods in periodontology. Two types of biofilms were developed, one by Streptococcus mutans UA 159 bacterial strain and the other by a complex mixture of saliva microorganisms isolated from a patient with periodontitis. This latter biofilm was characterized via Next Generation Sequencing to determine the main bacterial phyla. The CAP source was applied at a distance of 6 mm for different time points. A statistically significant reduction of both CFU count and XTT was already detected after 60 s of CAP treatment. CLSM analysis supported CAP effectiveness in killing the microorganisms inside the biofilm and in reducing the thickness of the biofilm matrix. Cytotoxicity tests demonstrated the possible use of CAP without important side effects towards human gingival fibroblasts cell line. The current study showed that CAP treatment was able to significantly reduce preformed biofilms developed by both S. mutans and microorganisms isolated by a saliva sample. Further studies should be conducted on biofilms developed by additional saliva donors to support the potential of this innovative strategy to counteract oral pathogens responsible for periodontal diseases.


Asunto(s)
Biopelículas , Gases em Plasma , Saliva , Streptococcus mutans , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Humanos , Gases em Plasma/farmacología , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/fisiología , Saliva/microbiología , Fibroblastos/microbiología , Fibroblastos/efectos de los fármacos , Periodontitis/microbiología , Periodontitis/terapia , Línea Celular , Boca/microbiología
2.
mBio ; 15(5): e0018424, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38624207

RESUMEN

Microorganisms resist fluoride toxicity using fluoride export proteins from one of several different molecular families. Cariogenic species Streptococcus mutans and Candida albicans extrude intracellular fluoride using a CLCF F-/H+ antiporter and FEX fluoride channel, respectively, whereas oral commensal eubacteria, such as Streptococcus gordonii, export fluoride using a Fluc fluoride channel. In this work, we examine how genetic knockout of fluoride export impacts pathogen fitness in single-species and three-species dental biofilm models. For biofilms generated using S. mutans with the genetic knockout of the CLCF transporter, exposure to low fluoride concentrations decreased S. mutans counts, synergistically reduced the populations of C. albicans, increased the relative proportion of oral commensal S. gordonii, and reduced properties associated with biofilm pathogenicity, including acid production and hydroxyapatite dissolution. Biofilms prepared with C. albicans with genetic knockout of the FEX channel also exhibited reduced fitness in the presence of fluoride but to a lesser degree. Imaging studies indicate that S. mutans is highly sensitive to fluoride, with the knockout strain undergoing complete lysis when exposed to low fluoride for a moderate amount of time. Biochemical purification of the S. mutans CLCF transporter and functional reconstitution establishes that the functional protein is a dimer encoded by a single gene. Together, these findings suggest that fluoride export by oral pathogens can be targeted by specific inhibitors to restore biofilm symbiosis in dental biofilms and that S. mutans is especially susceptible to fluoride toxicity. IMPORTANCE: Dental caries is a globally prevalent condition that occurs when pathogenic species, including Streptococcus mutans and Candida albicans, outcompete beneficial species, such as Streptococcus gordonii, in the dental biofilm. Fluoride is routinely used in oral hygiene to prevent dental caries. Fluoride also has antimicrobial properties, although most microbes possess fluoride exporters to resist its toxicity. This work shows that sensitization of cariogenic species S. mutans and C. albicans to fluoride by genetic knockout of fluoride exporters alters the microbial composition and pathogenic properties of dental biofilms. These results suggest that the development of drugs that inhibit fluoride exporters could potentiate the anticaries effect of fluoride in over-the-counter products like toothpaste and mouth rinses. This is a novel strategy to treat dental caries.


Asunto(s)
Biopelículas , Candida albicans , Fluoruros , Streptococcus gordonii , Streptococcus mutans , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/fisiología , Candida albicans/metabolismo , Streptococcus mutans/genética , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/metabolismo , Streptococcus mutans/fisiología , Fluoruros/farmacología , Fluoruros/metabolismo , Streptococcus gordonii/efectos de los fármacos , Streptococcus gordonii/genética , Streptococcus gordonii/fisiología , Streptococcus gordonii/metabolismo , Técnicas de Inactivación de Genes , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Caries Dental/microbiología
3.
J Appl Oral Sci ; 32: e20230326, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38656049

RESUMEN

OBJECTIVE: This study evaluated the surface roughness, wettability and adhesion of multispecies biofilms (Candida albicans, Staphylococcus aureus and Streptococcus mutans) on 3D-printed resins for complete denture bases and teeth compared to conventional resins (heat-polymerized acrylic resin; artificial pre-fabricated teeth). METHODOLOGY: Circular specimens (n=39; 6.0 mm Ø × 2.0 mm) of each group were subjected to roughness (n=30), wettability (n=30) and biofilm adhesion (n=9) tests. Three roughness measurements were taken by laser confocal microscopy and a mean value was calculated. Wettability was evaluated by the contact angle of sessile drop method, considering the mean of the three evaluations per specimen. In parallel, microorganism adhesion to resin surfaces was evaluated using a multispecies biofilm model. Microbial load was evaluated by determining the number of Colony Forming Units (CFU/mL) and by scanning electron microscopy (SEM). Data were subjected to the Wald test in a generalized linear model with multiple comparisons and Bonferroni adjustment, as well as two-way ANOVA (α=5%). RESULTS: The roughness of the conventional base resin (0.01±0.04) was lower than that of the conventional tooth (0.14±0.04) (p=0.023) and 3D-printed base (0.18±0.08) (p<0.001). For wettability, conventional resin (84.20±5.57) showed a higher contact angle than the 3D-printed resin (60.58±6.18) (p<0.001). Higher microbial loads of S. mutans (p=0.023) and S. aureus (p=0.010) were observed on the surface of the conventional resin (S. mutans: 5.48±1.55; S. aureus: 7.01±0.57) compared to the 3D-printed resin (S. mutans: 4.11±1.96; S. aureus: 6.42±0.78). The adhesion of C. albicans was not affected by surface characteristics. The conventional base resin showed less roughness than the conventional dental resin and the printed base resin. CONCLUSION: The 3D-printed resins for base and tooth showed less hydrophobicity and less adhesion of S. mutans and S. aureus than conventional resins.


Asunto(s)
Resinas Acrílicas , Adhesión Bacteriana , Biopelículas , Candida albicans , Bases para Dentadura , Ensayo de Materiales , Microscopía Confocal , Microscopía Electrónica de Rastreo , Impresión Tridimensional , Staphylococcus aureus , Streptococcus mutans , Propiedades de Superficie , Humectabilidad , Streptococcus mutans/fisiología , Staphylococcus aureus/fisiología , Candida albicans/fisiología , Bases para Dentadura/microbiología , Resinas Acrílicas/química , Análisis de Varianza , Reproducibilidad de los Resultados , Dentadura Completa/microbiología , Valores de Referencia , Recuento de Colonia Microbiana , Modelos Lineales
4.
Methods Enzymol ; 696: 155-174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38658078

RESUMEN

The interactions between communities of microorganisms inhabiting the dental biofilm is a major determinant of oral health. These biofilms are periodically exposed to high concentrations of fluoride, which is present in almost all oral healthcare products. The microbes resist fluoride through the action of membrane export proteins. This chapter describes the culture, growth and harvest conditions of model three-species dental biofilm comprised of cariogenic pathogens Streptococcus mutans and Candida albicans and the commensal bacterium Streptococcus gordonii. In order to examine the role of fluoride export by S. mutans in model biofilms, procedures for generating a strain of S. mutans with a genetic knockout of the fluoride exporter are described. We present a case study examining the effects of this mutant strain on the biofilm mass, acid production and mineral dissolution under exposure to low levels of fluoride. These general approaches can be applied to study the effects of any gene of interest in physiologically realistic multispecies oral biofilms.


Asunto(s)
Biopelículas , Candida albicans , Fluoruros , Streptococcus gordonii , Streptococcus mutans , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/genética , Streptococcus mutans/fisiología , Streptococcus mutans/metabolismo , Streptococcus mutans/crecimiento & desarrollo , Fluoruros/farmacología , Fluoruros/metabolismo , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/metabolismo , Candida albicans/fisiología , Streptococcus gordonii/efectos de los fármacos , Streptococcus gordonii/genética , Streptococcus gordonii/fisiología , Humanos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana/genética , Caries Dental/microbiología
5.
Braz J Microbiol ; 55(1): 365-374, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38040990

RESUMEN

Candida albicans causes a variety of clinical manifestations through multiple virulence factors that act simultaneously to overcome the immune system and invade the host tissues. Owing to the limited number of antifungal agents available, new candidiasis therapeutic strategies are required. Previous studies have demonstrated that the metabolites produced by Streptococcus mutans lead to a decrease in the number of Candida cells. Here, for the first time, we evaluated whether the C. albicans cells that survived the pretreatment with S. mutans supernatant can modify their virulence factors and their capability to infect Galleria mellonella larvae. Streptococcus mutans supernatant (SM-S) was obtained by filtering the culture supernatant of this bacterium. Then, C. albicans cells were pretreated with SM-S for 24 h, and the surviving cells were evaluated using in vitro and in vivo assays. The C. albicans pretreated with SM-S showed a significant inhibition of hyphal growth, an altered adhesion pattern, and an impaired capability to form biofilms; however, its proteolytic activity was not affected. In the in vivo assays, C. albicans cells previously exposed to SM-S exhibited a reduced ability to infect G. mellonella and a higher amount of circulating hemocytes. Thus, SM-S could inhibit important virulence factors of C. albicans, which may contribute to the development of new candidiasis therapeutic strategies.


Asunto(s)
Candida albicans , Candidiasis , Animales , Virulencia , Streptococcus mutans/fisiología , Candidiasis/microbiología , Factores de Virulencia , Biopelículas
6.
Small ; 20(19): e2309230, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38112271

RESUMEN

Bone infection poses a major clinical challenge that can hinder patient recovery and exacerbate postoperative complications. This study has developed a bioactive composite scaffold through the co-assembly and intrafibrillar mineralization of collagen fibrils and zinc oxide (ZnO) nanowires (IMC/ZnO). The IMC/ZnO exhibits bone-like hierarchical structures and enhances capabilities for osteogenesis, antibacterial activity, and bacteria-infected bone healing. During co-cultivation with human bone marrow mesenchymal stem cells (BMMSCs), the IMC/ZnO improves BMMSC adhesion, proliferation, and osteogenic differentiation even under inflammatory conditions. Moreover, it suppresses the activity of Gram-negative Porphyromonas gingivalis and Gram-positive Streptococcus mutans by releasing zinc ions within the acidic infectious microenvironment. In vivo, the IMC/ZnO enables near-complete healing of infected bone defects within the intricate oral bacterial milieu, which is attributed to IMC/ZnO orchestrating M2 macrophage polarization, and fostering an osteogenic and anti-inflammatory microenvironment. Overall, these findings demonstrate the promise of the bioactive scaffold IMC/ZnO for treating bacteria-infected bone defects.


Asunto(s)
Regeneración Ósea , Colágeno , Células Madre Mesenquimatosas , Nanocables , Osteogénesis , Andamios del Tejido , Óxido de Zinc , Óxido de Zinc/química , Óxido de Zinc/farmacología , Nanocables/química , Regeneración Ósea/efectos de los fármacos , Andamios del Tejido/química , Humanos , Colágeno/química , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Animales , Porphyromonas gingivalis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Streptococcus mutans/fisiología , Streptococcus mutans/efectos de los fármacos , Proliferación Celular/efectos de los fármacos
7.
J Dent ; 141: 104805, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38101504

RESUMEN

INTRODUCTION: Childhood caries, a prevalent chronic disease, affects 60-90 % of children in industrialized regions, leading to lesions in both primary and permanent teeth. This condition precipitates hospital admissions, emergency room visits, elevated treatment costs, and missed school days, thereby impeding the child's academic engagement and increasing the likelihood of caries into adulthood. Despite multiple identified risk factors, significant interpersonal variability remains unexplained. The immune system generates a unique antibody repertoire, essential for maintaining a balanced and healthy oral microbiome. Streptococcus mutans is a primary contributor to the development of caries. METHODS: Employing mass spectrometry, we investigated the S. mutans proteins targeted by antibodies in children both with and without caries, delineating a fundamental suite of proteins discernible by the immune systems of a majority of individuals. Notably, this suite was enriched with proteins pivotal for bacterial adhesion. To ascertain the physiological implications of these discoveries, we evaluated the efficacy of saliva in thwarting S. mutans adherence to dental surfaces. RESULTS: Antibodies in most children recognized a core set of ten S. mutans proteins, with additional proteins identified in some individuals. There was no significant difference in the proteins identified by children with or without caries, but there was variation in antibody binding intensity to some proteins. Functionally, saliva from caries-free individuals, but not children with caries, was found to hinder the binding of S. mutans to teeth. These findings delineate the S. mutans proteome targeted by the immune system and suggest that the inhibition of bacterial adherence to teeth is a primary mechanism employed by the immune system to maintain oral balance and prevent caries formation. CONCLUSIONS: These findings enhance our knowledge of the immune system's function in oral health maintenance and caries prevention, shedding light on how immunoglobulins interact with S. mutans proteins. CLINICAL SIGNIFICANCE: Targeting S. mutans proteins implicated in bacterial adhesion could be a promising strategy for preventing childhood caries.


Asunto(s)
Caries Dental , Diente , Niño , Humanos , Streptococcus mutans/fisiología , Susceptibilidad a Caries Dentarias , Caries Dental/prevención & control , Caries Dental/microbiología , Adhesión Bacteriana , Saliva/química
8.
Braz Dent J ; 34(3): 73-81, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37466528

RESUMEN

Experimental models that consider host-pathogen interactions are relevant for improving knowledge about oral candidiasis. The aim of this study was to assess the epithelial immune responses, Candida penetration of cell monolayers, and virulence during mixed species culture infections. Single species cultures of Candida albicans and mixed cultures (C. albicans, Streptococcus mutans, and Streptococcus sanguinis) were used to infect monolayers of HaCaT and FaDu ATCC HTB-43 cells for 12 h. After infection, IL-18 and IL-34 gene expression was measured to assess epithelial cell immune responses, and lactate dehydrogenase (LDH) activity was measured as an indicator of cell damage. Microscopy determined C. albicans morphology and penetration of fungal cells through the keratinocyte monolayer. Monolayers devoid of infection served as controls. Data were analyzed by an ANOVA one-way test followed by Tukey's post-hoc test (α = 0.05). The results found that IL-18 and IL-34 gene expression and LDH activity were significantly (p < 0.05) upregulated for both cell lines exposed to mixed species cultures compared with C. albicans alone. Candida albicans yeast and hyphae were evident in C. albicans only infections. In contrast, monolayers infected by C. albicans, S. mutans, and S. sanguinis exhibited higher microbial invasion with several hyphal aggregates detected. The presence of streptococci in C. albicans infection enhances the virulence and pathogenicity of the fungus with associated increased immune responses and tissue damage. Extrapolation of these findings to oral infection would indicate the added potential benefit of managing bacterial components of biofilms during treatment.


Asunto(s)
Candida albicans , Interleucina-18 , Virulencia , Interleucina-18/metabolismo , Streptococcus , Streptococcus mutans/fisiología , Biopelículas
9.
Front Cell Infect Microbiol ; 13: 1053230, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37187472

RESUMEN

Objective: Various studies have indicated the application of Coenzyme Q10 and probiotic bacteria such as Ligilactobacillus salivarius (L. salivarius) and Lactiplantibacillus plantarum (L. plantarum) in combating periodontal disease. Considering the positive effect of these two on oral health, and the destructive effect of S. mutans, in this study, we investigate the outcomes of the administration of probiotics and Q10 on infected HEp-2 cell viability and S. mutans adhesion in different settings. Methods: A 3-week-old human epidermoid laryngeal (HEp-2) cell line was cultured and exposed to two different probiotics and 3 different doses of Q10 doses. Samples were contaminated by S. mutans immediately (therapeutic setting) and after 3 hours (preventive setting). Eventually, the viability of HEp-2 cells was investigated by MTT. Also, the number of adhered S. mutans was explored by direct and indirect adhesion assays. Results: L. plantarum and L. salivarius protect epithelial cells against S. mutans in both therapeutic and preventive settings, albeit not fully. In contrast, Q10 completely preserves the viability of infected Her HEp-2 cells at all concentrations. The effects of the coexistence of Q10 and probiotics were not quite equal, among which L. salivarius and 5 µg of Q10 form the best results. The microscopic adherence assay of S. mutans revealed that samples containing Q10 had significantly lower adhesion of probiotics and S. mutans to HEp-2 cells. Similarly, plates containing L. salivarius with 5µg or L. plantarum with 1µg Q10 or sole presence of L. salivarius had the lowest S. mutans adherence among others. Also, L. salivarius with 5µg Q10 had one of the highest probiotic adherences. Conclusion: In conclusion, co-administration of Q10 and probiotics especially in presence of L. salivarius with 5µg Q10 could have remarkable effects on HEp-2 cell viability, S. mutans, and probiotic adherence. Nevertheless, our study, for the first time, showed that Q10 might have an anti-bacterial activity by suppressing the adhesion of tested bacteria to HEp-2 cells. This hypothesis, if correct, suggests that due to their different mechanisms, co-prescription of Q10 and probiotics may lead to better clinical responses, especially in the mentiond dose.


Asunto(s)
Ligilactobacillus salivarius , Enfermedades Periodontales , Probióticos , Humanos , Femenino , Streptococcus mutans/fisiología , Supervivencia Celular , Probióticos/uso terapéutico
10.
Int J Mol Sci ; 24(8)2023 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-37108414

RESUMEN

A disturbed balance within the dental biofilm can result in the dominance of cariogenic and periodontopathogenic species and disease development. Due to the failure of pharmacological treatment of biofilm infection, a preventive approach to promoting healthy oral microbiota is necessary. This study analyzed the influence of Streptococcus salivarius K12 on the development of a multispecies biofilm composed of Streptococcus mutans, S. oralis and Aggregatibacter actinomycetemcomitans. Four different materials were used: hydroxyapatite, dentin and two dense polytetrafluoroethylene (d-PTFE) membranes. Total bacteria, individual species and their proportions in the mixed biofilm were quantified. A qualitative analysis of the mixed biofilm was performed using scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). The results showed that in the presence of S. salivarius K 12 in the initial stage of biofilm development, the proportion of S. mutans was reduced, which resulted in the inhibition of microcolony development and the complex three-dimensional structure of the biofilm. In the mature biofilm, a significantly lower proportion of the periodontopathogenic species A. actinomycetemcomitans was found in the salivarius biofilm. Our results show that S. salivarius K 12 can inhibit the growth of pathogens in the dental biofilm and help maintain the physiological balance in the oral microbiome.


Asunto(s)
Streptococcus mutans , Streptococcus salivarius , Streptococcus mutans/fisiología , Aggregatibacter actinomycetemcomitans , Biopelículas , Homeostasis
11.
Front Cell Infect Microbiol ; 13: 1116021, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36968105

RESUMEN

Objective: The aim of this study was to design and optimize a cold atmospheric plasma (CAP) device that could be applied in an oral environment and to study its effects on plaque biofilm metabolism and regrowth, as well as microbial flora composition and enamel demineralization. Method: CAP was obtained through a dielectric barrier discharge device; the optical properties were analyzed using emission spectroscopy. The electrochemical analysis of plasma devices includes voltametric characteristic curves and Lissajous. The Streptococcus mutans (UA159) and saliva biofilms were treated in vitro, and the effects of CAP on biofilm metabolism were investigated using MTT and lactate dehydrogenase assays. The duration of antibacterial activity on biofilms was examined, scanning electron microscopy was used to observe the morphology of biofilms, and 16S rRNA sequencing was used to explore the influence of CAP on the microbial flora composition of saliva biofilms. An in vitro model of biofilm-enamel demineralization was designed, and the effect of CAP on enamel demineralization was evaluated by micro surface hardness and micro-CT analysis. Results: CAP had antibacterial proliferative ability toward Streptococcus mutans biofilms and saliva biofilms and was stronger than ultraviolet under the same tested conditions. After 24 h, the antibacterial effect disappeared, which proved the short-term timeliness of its bactericidal ability. CAP can inhibit the acid production of biofilms, and its inhibitory effect on saliva biofilms can be extended to 24 h. CAP had a strong ability to regulate the composition of plaque biofilms, especially for Lactococcus proliferation, a major acid-producing bacterium in microcosm biofilms. The CAP-treated enamels were more acid-tolerant than non-treated controls. Conclusion: CAP had an explicit bactericidal effect on caries-related biofilms, which is a short-term antibacterial effect. It can inhibit the acid production of biofilms and has a downregulation effect on Lactococcus in saliva biofilms. CAP can help reduce demineralization of enamel.


Asunto(s)
Caries Dental , Desmineralización Dental , Humanos , Argón/farmacología , ARN Ribosómico 16S/genética , Desmineralización Dental/microbiología , Streptococcus mutans/fisiología , Biopelículas , Antibacterianos/farmacología
12.
Front Cell Infect Microbiol ; 13: 1130506, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36949812

RESUMEN

Dental biofilms are highly assembled microbial communities surrounded by an extracellular matrix, which protects the resident microbes. The microbes, including commensal bacteria and opportunistic pathogens, coexist with each other to maintain relative balance under healthy conditions. However, under hostile conditions such as sugar intake and poor oral care, biofilms can generate excessive acids. Prolonged low pH in biofilm increases proportions of acidogenic and aciduric microbes, which breaks the ecological equilibrium and finally causes dental caries. Given the complexity of oral microenvironment, controlling the acidic biofilms using antimicrobials that are activated at low pH could be a desirable approach to control dental caries. Therefore, recent researches have focused on designing novel kinds of pH-activated strategies, including pH-responsive antimicrobial agents and pH-sensitive drug delivery systems. These agents exert antibacterial properties only under low pH conditions, so they are able to disrupt acidic biofilms without breaking the neutral microenvironment and biodiversity in the mouth. The mechanisms of low pH activation are mainly based on protonation and deprotonation reactions, acids labile linkages, and H+-triggered reactive oxygen species production. This review summarized pH-activated antibiofilm strategies to control dental caries, concentrating on their effect, mechanisms of action, and biocompatibility, as well as the limitation of current research and the prospects for future study.


Asunto(s)
Antiinfecciosos , Caries Dental , Humanos , Caries Dental/prevención & control , Streptococcus sanguis , Streptococcus mutans/fisiología , Biopelículas , Antiinfecciosos/farmacología , Concentración de Iones de Hidrógeno
13.
Mar Drugs ; 21(2)2023 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-36827164

RESUMEN

The polymicrobial proliferation and development of complex biofilm morphologies by bacterial and fungal pathogens in the host are some of the key factors contributing to the failure of antimicrobial treatments. The polymicrobial interaction of Candida albicans and some bacterial species has been extensively studied in both in vitro and in vivo model systems. Alternative strategies for disrupting polymicrobial interaction and biofilm formation are constantly needed. Among several alternative strategies, the use of nanoparticles synthesized using a natural product in the treatment of microbial infection has been considered a promising approach. The current study aimed to synthesize gold nanoparticles (AuNPs) using a natural product, fucoidan, and to test their efficacy against mono and duo combinations of fungal (Candida albicans) and bacterial (Staphylococcus aureus/Streptococcus mutans) biofilms. Several methods were used to characterize and study Fu-AuNPs, including UV-vis absorption spectroscopy, FTIR, FE-TEM, EDS, DLS, zeta potential, and XRD. The concentration-dependent inhibition of early-stage biofilms and the eradication of mature biofilms of single species of C. albicans, S. aureus, and S. mutans have been observed. Early biofilms of a dual-species combination of C. albicans and S. aureus/S. mutans were also suppressed at an increasing concentration of Fu-AuNPs. Furthermore, Fu-AuNPs significantly eradicated the established mature biofilm of mixed species. The treatment method proposed in this study, which involves the use of marine-bioinspired nanoparticles, is a promising and biocompatible agent for preventing the growth of polymicrobial biofilms of bacterial and fungal pathogens.


Asunto(s)
Candida albicans , Nanopartículas del Metal , Oro , Staphylococcus aureus , Streptococcus mutans/fisiología , Biopelículas
14.
Food Funct ; 13(24): 12755-12765, 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36416047

RESUMEN

Postbiotics are functional biological compounds, such as bacterial lysates (BLs) released from probiotic bacteria. Although postbiotics exert various bioactivities, the anti-inflammatory and antibiofilm activities of BLs against oral pathogenic bacteria have not been investigated. In the present study, pretreatment with BLs extracted from Lactobacillus plantarum and L. rhamnosus GG suppressed the mRNA and protein expression levels of inflammatory mediators induced by the lipopolysaccharide (LPS) of Porphyromonas gingivalis in RAW 264.7 cells. Both BLs attenuated P. gingivalis LPS-induced phosphorylation of mitogen-activated protein kinases (MAPKs) and activation of nuclear factor-κB (NF-κB), suggesting that BLs inhibit periodontal inflammatory responses by regulating the MAPK and NF-κB signaling pathways. Moreover, both BLs interfered with biofilm formation by Streptococcus mutans; however, they did not eradicate the established S. mutans biofilm. Furthermore, both BLs downregulated gtfB, gtfC, and gtfD responsible for biofilm formation by S. mutans, suggesting that BLs reduce the synthesis of extracellular polysaccharide and thereby reduce S. mutans biofilm. Taken together, these results suggest that BLs of L. plantarum and L. rhamnosus GG can attenuate periodontal inflammation and dental caries and thus contribute to the improvement of oral health.


Asunto(s)
Antiinflamatorios , Biopelículas , Extractos Celulares , Caries Dental , Porphyromonas gingivalis , Humanos , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Caries Dental/microbiología , Caries Dental/prevención & control , Lipopolisacáridos , FN-kappa B/metabolismo , Células RAW 264.7 , Streptococcus mutans/fisiología , Probióticos , Extractos Celulares/farmacología , Extractos Celulares/uso terapéutico
15.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(5): 922-928, 2022 Sep.
Artículo en Chino | MEDLINE | ID: mdl-36224698

RESUMEN

Dental caries is a disease in which chronic progressive destruction of the hard dental tissues occurs under the influence of multiple factors, among which, bacterial infection being the most important one. Dental plaque biofilm is a key factor in the pathogenesis of dental caries. Under normal circumstances, microorganisms within the biofilm maintain a dynamic balance through coordination, competition, and antagonism. However, when the environment changes, the balance in the biofilm will be disrupted, and the number of cariogenic bacteria, especially Streptococcus mutans ( S. mutans), will increase significantly, thereby causing the production of large amounts of organic acids on the tooth surface, tooth demineralization, and the formation of dental caries. Therefore, finding ways to restore the dynamic balance of oral microorganisms through selective inhibition of S. mutans is key to the prevention and treatment of dental caries. Herein, we reviewed the research progress of recent years in the development of materials with selective antibacterial effect, intending to provide references for the further development of drugs for the prevention and treatment of dental caries. Future studies should focus on the following aspects, mechanism, clinical efficacy, chemical modification, and safety, to supplement and make improvements on the existing relevant research, and to promote progress in research and development of drugs for the prevention and treatment of dental caries.


Asunto(s)
Caries Dental , Streptococcus mutans , Antibacterianos/farmacología , Biopelículas , Caries Dental/prevención & control , Humanos , Streptococcus mutans/fisiología
16.
Mol Oral Microbiol ; 37(6): 244-255, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36156446

RESUMEN

Dental caries (tooth-decay) is caused by biofilms harboring polymicrobial communities on teeth that leads to the onset of localized areas of enamel demineralization. Streptococcus mutans has been clinically associated with severe caries in childhood. Although commensal bacteria can combat S. mutans using self-generated antimicrobials such as hydrogen peroxide (H2 O2 ), constant sugar-rich diet consumption disrupts microbial homeostasis shifting toward cariogenic community. Recently, Streptococcus oralis subsp. tigurinus strain J22, an oral isolate, was identified as a uniquely potent H2 O2 producer. Here, we assess whether a high H2 O2 -producing commensal streptococcus can modulate the spatial organization and virulence of S. mutans within biofilms. Using an experimental biofilm model, we find that the presence of S. oralis J22 can effectively inhibit the clustering, accumulation, and spatial organization of S. mutans on ex vivo human tooth surface, resulting in significant reduction of enamel demineralization. Notably, the generation of H2 O2 via pyruvate oxidase (SpxB) from S. oralis J22 is not repressed by sugars (a common repressor in other mitis group streptococci), resulting in enhanced inhibition of S. mutans growth (vs. Streptococcus gordonii). We further investigate its impact on biofilm virulence using an in vivo rodent caries model under sugar-rich diet. Coinfection of S. mutans with S. oralis results in reduced caries development compared to either species infected alone, whereas coinfection with S. gordonii has negligible effects, suggesting that the presence of an efficient, high H2 O2 -producer can disrupt S. mutans virulence. This work demonstrates that oral isolates with unusual high H2 O2 production may be capable of modulating biofilm cariogenicity in vivo. The findings also highlight the importance of bacterial antagonistic interactions within polymicrobial communities in health and in disease-causing state.


Asunto(s)
Coinfección , Caries Dental , Humanos , Streptococcus mutans/fisiología , Caries Dental/microbiología , Susceptibilidad a Caries Dentarias , Streptococcus gordonii/fisiología , Biopelículas , Azúcares/farmacología
17.
Nutrients ; 14(16)2022 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-36014799

RESUMEN

Streptococcus mutans (S. mutans) is a common cariogenic bacterium that secretes glucosyltransferases (GTFs) to synthesize extracellular polysaccharides (EPSs) and plays an important role in plaque formation. Propolis essential oil (PEO) is one of the main components of propolis, and its antibacterial activity has been proven. However, little is known about the potential effects of PEO against S. mutans. We found that PEO has antibacterial effects against S. mutans by decreasing bacterial viability within the biofilm, as demonstrated by the XTT assay, live/dead staining assay, LDH activity assay, and leakage of calcium ions. Furthermore, PEO also suppresses the total of biofilm biomasses and damages the biofilm structure. The underlying mechanisms involved may be related to inhibiting bacterial adhesion and GTFs activity, resulting in decreased production of EPSs. In addition, a CCK8 assay suggests that PEO has no cytotoxicity on normal oral epithelial cells. Overall, PEO has great potential for preventing and treating oral bacterial infections caused by S. mutans.


Asunto(s)
Antibacterianos , Biopelículas , Caries Dental , Aceites Volátiles , Própolis , Streptococcus mutans , Antibacterianos/química , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , China , Caries Dental/microbiología , Caries Dental/prevención & control , Glucosiltransferasas/farmacología , Humanos , Aceites Volátiles/farmacología , Polisacáridos/farmacología , Própolis/farmacología , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/fisiología
18.
Cancer Sci ; 113(11): 3980-3994, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35997541

RESUMEN

Recent studies have demonstrated a relationship between oral bacteria and systemic inflammation. Endothelial cells (ECs), which line blood vessels, control the opening and closing of the vascular barrier and contribute to hematogenous metastasis; however, the role of oral bacteria-induced vascular inflammation in tumor metastasis remains unclear. In this study, we examined the phenotypic changes in vascular ECs following Streptococcus mutans (S. mutans) stimulation in vitro and in vivo. The expression of molecules associated with vascular inflammation and barrier-associated adhesion was analyzed. Tumor metastasis was evaluated after intravenous injection of S. mutans in murine breast cancer hematogenous metastasis model. The results indicated that S. mutans invaded the ECs accompanied by inflammation and NF-κB activation. S. mutans exposure potentially disrupts endothelial integrity by decreasing vascular endothelial (VE)-cadherin expression. The migration and adhesion of tumor cells were enhanced in S. mutans-stimulated ECs. Furthermore, S. mutans-induced lung vascular inflammation promoted breast cancer cell metastasis to the lungs in vivo. The results indicate that oral bacteria promote tumor metastasis through vascular inflammation and the disruption of vascular barrier function. Improving oral hygiene in patients with cancer is of great significance in preventing postoperative pneumonia and tumor metastasis.


Asunto(s)
Neoplasias de la Mama , Streptococcus mutans , Humanos , Ratones , Animales , Femenino , Streptococcus mutans/fisiología , Células Endoteliales/metabolismo , Transducción de Señal , Inflamación/metabolismo , Neoplasias de la Mama/metabolismo
19.
Microbiol Spectr ; 10(4): e0112222, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35863019

RESUMEN

The oral cavity contains a number of microbes. They interact with each other and play an important role in human health. Among oral cariogenic microbes, Streptococcus mutans is recognized a major etiological bacteria of dental caries. Lactobacilli strains have been promoted as possible probiotic agents against S. mutans. However, their inhibitory mechanism has not been well elucidated yet. In the present study, two new compounds with strong antibiofilm activities were purified from the culture supernatant of Lactobacillus paragasseri MJM60645, which was isolated from the human oral cavity. These compounds showed strong inhibitory activities against S. mutans biofilm formation, with IC50 (concentration at which 50% biofilm was inhibited) of 30.4 µM for compound 1 and 18.9 µM for compound 2. However, these compounds did not show bactericidal activities against S. mutans. Structure elucidation by nuclear magnetic resonance (NMR) and mass spectrometry showed that compound 1 was composed of two arabinofuranose iminosugars jointed with one glycerol and oleic acid, and compound 2 was composed of two arabinofuranose iminosugars jointed with one glycerol and nervonic acid. To the best of our knowledge, these structures were discovered for the first time in this study. Treatment of S. mutans with compound 1 strongly downregulated expression levels of genes related to biofilm formation, including gtfB, gtfC, gtfD, gbpB, brpA, spaP, ftf, and smu0630 without affecting the expression of comDE or relA. This study provides new insights into novel molecules produced by Lactobacillus to regulate the pathogenesis of S. mutans, facilitating a better understanding of the mechanism for interactions between Lactobacillus and S. mutans. IMPORTANCE In this study, we isolated lactic acid bacteria that inhibit streptococcal biofilm from the oral cavity of infants and identified two novel compounds from the supernatant of their culture broth. The two compounds are structurally similar, and both consist of iminosugars, glycerol, and unsaturated fatty acid. A search of the SciFinder database revealed that these structures are novel and were discovered for the first time in this study. Mechanism studies have shown that these compounds can inhibit the expression of biofilm synthesis-related genes. This is the first report that lactic acid bacteria inhibit streptococcal biofilms by small molecules with new chemical structures. This study not only expands the understanding of natural products derived from lactic acid bacteria but also provides a new paradigm for the understanding of the interaction of bacteria in the oral microbiota.


Asunto(s)
Caries Dental , Streptococcus mutans , Biopelículas , Glicerol , Humanos , Lactobacillus , Streptococcus , Streptococcus mutans/fisiología
20.
Microbiol Spectr ; 10(4): e0119922, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35880891

RESUMEN

Streptococcus mutans is known as an important oral pathogen causing dental caries, a widespread oral infectious disease. S. mutans synthesize exopolysaccharide (EPS) using glucosyltransferases (Gtfs), resulting in biofilm formation on the tooth surface. Bacterial cells in the biofilms become strongly resistant to a harsh environment, such as antibiotics and host defense mechanisms, making biofilm-based infections difficult to eliminate. Discovering novel antibiofilm agents, especially from natural products, helps to develop effective strategies against this kind of diseases. The present study investigated the inhibitory effect of shikimic acid (SA), one abundant compound derived from Illicium verum extract, on the biofilm formation of S. mutans. We found SA can reduce the EPS synthesized by this oral pathogen and modulate the transcription of biofilm formation related genes, leading to fewer bacterial cells in its biofilm. SA also interacted with cell membrane and membrane proteins, causing damage to bacterial cells. Ex vivo testing of biofilm formation on bovine teeth showed SA strongly decreased the number of S. mutans cells and the number of EPS accumulated on dental enamel surfaces. Moreover, SA exhibits almost no toxicity to human oral cells evaluated by in vitro biocompatibility assay. In conclusion, shikimic acid exhibits remarkable antibiofilm activity against S. mutans and has the potential to be further developed as a novel anticaries agent. IMPORTANCE Natural products are an important and cost-effective source for screening antimicrobial agents. Here, we identified one compound, shikimic acid, from Illicium verum extract, exhibiting antimicrobial activity against S. mutans proliferation. It also inhibits biofilm formation of this bacteria through decreasing Gtf expression and EPS synthesis. Furthermore, this compound exhibits no significant cytotoxicity at its MIC against S. mutans, providing evidence for its clinical application.


Asunto(s)
Productos Biológicos , Caries Dental , Animales , Biopelículas , Bovinos , Humanos , Extractos Vegetales/farmacología , Ácido Shikímico/farmacología , Streptococcus mutans/fisiología , Factores de Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...