Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.922
Filtrar
1.
Biofouling ; 40(2): 99-113, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38425046

RESUMO

This scoping review focused on exploring the efficacy of flavonoids against bacteria associated with dental caries and periodontal diseases. Inclusion criteria comprise studies investigating the antibacterial effects of flavonoids against bacteria linked to caries or periodontal diseases, both pure or diluted in vehicle forms. The search, conducted in August 2023, in databases including PubMed/MEDLINE, Scopus, Web of Science, Embase, LILACS, and Gray Literature. Out of the initial 1125 studies, 79 met the inclusion criteria, majority in vitro studies. Prominent flavonoids tested included epigallocatechin-gallate, apigenin, quercetin, and myricetin. Predominant findings consistently pointed to bacteriostatic, bactericidal, and antibiofilm activities. The study primarily investigated bacteria associated with dental caries, followed by periodontopathogens. A higher number of publications presented positive antibacterial results against Streptococcus mutans in comparison to Porphyromonas gingivalis. These encouraging findings underline the potential applicability of commercially available flavonoids in materials or therapies, underscoring the need for further exploration in this field.


Assuntos
Cárie Dentária , Doenças Periodontais , Humanos , Cárie Dentária/prevenção & controle , Biofilmes , Doenças Periodontais/tratamento farmacológico , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Flavonoides/farmacologia , Antibacterianos/farmacologia , Streptococcus mutans
2.
NPJ Biofilms Microbiomes ; 10(1): 3, 2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233485

RESUMO

Oral microbiome dysbiosis mediates chronic periodontal disease, gut microbial dysbiosis, and mucosal barrier disfunction that leads to steatohepatitis via the enterohepatic circulation. Improving this dysbiosis towards health may improve liver disease. Treatment with antibiotics and probiotics have been used to modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. The aim of the present investigation was to evaluate the potential for nisin, an antimicrobial peptide produced by Lactococcus lactis, to counteract the periodontitis-associated gut dysbiosis and to modulate the glycolipid-metabolism and inflammation in the liver. Periodontal pathogens, namely Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Fusobacterium nucleatum, were administrated topically onto the oral cavity to establish polymicrobial periodontal disease in mice. In the context of disease, nisin treatment significantly shifted the microbiome towards a new composition, commensurate with health while preventing the harmful inflammation in the small intestine concomitant with decreased villi structural integrity, and heightened hepatic exposure to bacteria and lipid and malondialdehyde accumulation in the liver. Validation with RNA Seq analyses, confirmed the significant infection-related alteration of several genes involved in mitochondrial dysregulation, oxidative phosphorylation, and metal/iron binding and their restitution following nisin treatment. In support of these in vivo findings indicating that periodontopathogens induce gastrointestinal and liver distant organ lesions, human autopsy specimens demonstrated a correlation between tooth loss and severity of liver disease. Nisin's ability to shift the gut and liver microbiome towards a new state commensurate with health while mitigating enteritis, represents a novel approach to treating NAFLD-steatohepatitis-associated periodontal disease.


Assuntos
Bacteriocinas , Nisina , Hepatopatia Gordurosa não Alcoólica , Doenças Periodontais , Camundongos , Humanos , Animais , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/metabolismo , Nisina/farmacologia , Nisina/metabolismo , Disbiose , Doenças Periodontais/microbiologia , Porphyromonas gingivalis/metabolismo , Inflamação/complicações , Estresse Oxidativo
3.
Crit Rev Microbiol ; 50(2): 212-223, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36883683

RESUMO

The oral microbiome, populated by a diverse range of species, plays a critical role in the initiation and progression of periodontal disease. The most dominant yet little-discussed players in the microbiome, the bacteriophages, influence the health and disease of the host in various ways. They, not only contribute to periodontal health by preventing the colonization of pathogens and disrupting biofilms but also play a role in periodontal disease by upregulating the virulence of periodontal pathogens through the transfer of antibiotic resistance and virulence factors. Since bacteriophages selectively infect only bacterial cells, they have an enormous scope to be used as a therapeutic strategy; recently, phage therapy has been successfully used to treat antibiotic-resistant systemic infections. Their ability to disrupt biofilms widens the scope against periodontal pathogens and dental plaque biofilms in periodontitis. Future research focussing on the oral phageome and phage therapy's effectiveness and safety could pave way for new avenues in periodontal therapy. This review explores our current understanding of bacteriophages, their interactions in the oral microbiome, and their therapeutic potential in periodontal disease.


Assuntos
Bacteriófagos , Doenças Periodontais , Periodontite , Humanos , Bacteriófagos/genética , Doenças Periodontais/terapia , Doenças Periodontais/microbiologia , Periodontite/terapia , Periodontite/microbiologia , Biofilmes , Virulência
4.
Front Cell Infect Microbiol ; 13: 1159822, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37124035

RESUMO

Microbiome is a keystone polymicrobial community that coexist with human body in a beneficial relationship. These microorganisms enable the human body to maintain homeostasis and take part in mechanisms of defense against infection and in the absorption of nutrients. Even though microbiome is involved in physiologic processes that are beneficial to host health, it may also cause serious detrimental issues. Additionally, it has been proven that bacteria can migrate to other human body compartments and colonize them even although significant structural differences with the area of origin exist. Such migrations have been clearly observed when the causes of genesis and progression of colorectal cancer (CRC) have been investigated. It has been demonstrated that the oral microbiome is capable of penetrating into the large intestine and cause impairments leading to dysbiosis and stimulation of cancerogenic processes. The main actors of such events seem to be oral pathogenic bacteria belonging to the red and orange complex (regarding classification of bacteria in the context of periodontal diseases), such as Porphyromonas gingivalis and Fusobacterium nucleatum respectively, which are characterized by significant amount of cancerogenic virulence factors. Further examination of oral microbiome and its impact on CRC may be crucial on early detection of this disease and would allow its use as a precise non-invasive biomarker.


Assuntos
Neoplasias Colorretais , Microbiota , Doenças Periodontais , Humanos , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Fatores de Virulência , Fusobacterium nucleatum
5.
Microbiol Spectr ; 11(3): e0432722, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37191548

RESUMO

The purposes of this study were to examine the compositional changes in the salivary microbiota according to the severity of periodontal disease and to verify whether the distribution of specific bacterial species in saliva can distinguish the severity of disease. Saliva samples were collected from 8 periodontally healthy controls, 16 patients with gingivitis, 19 patients with moderate periodontitis, and 29 patients with severe periodontitis. The V3 and V4 regions of the 16S rRNA gene in the samples were sequenced, and the levels of 9 bacterial species showing significant differences among the groups by sequencing analysis were identified using quantitative real-time PCR (qPCR). The predictive performance of each bacterial species in distinguishing the severity of disease was evaluated using a receiver operating characteristic curve. Twenty-nine species, including Porphyromonas gingivalis, increased as the severity of disease increased, whereas 6 species, including Rothia denticola, decreased. The relative abundances of P. gingivalis, Tannerella forsythia, Filifactor alocis, and Prevotella intermedia determined by qPCR were significantly different among the groups. The three bacterial species P. gingivalis, T. forsythia, and F. alocis were positively correlated with the sum of the full-mouth probing depth and were moderately accurate at distinguishing the severity of periodontal disease. In conclusion, the salivary microbiota showed gradual compositional changes according to the severity of periodontitis, and the levels of P. gingivalis, T. forsythia, and F. alocis in mouth rinse saliva had the ability to distinguish the severity of periodontal disease. IMPORTANCE Periodontal disease is one of the most widespread medical conditions and the leading cause of tooth loss, imposing high economic costs and an increasing burden worldwide as life expectancy increases. Changes in the subgingival bacterial community during the progression of periodontal disease can affect the entire oral ecosystem, and bacteria in saliva can reflect the degree of bacterial imbalance in the oral cavity. This study explored whether the specific bacterial species in saliva can distinguish the severity of periodontal disease by analyzing the salivary microbiota and suggested P. gingivalis, T. forsythia, and F. alocis as biomarkers for distinguishing the severity of periodontal disease in saliva.


Assuntos
Microbiota , Doenças Periodontais , Periodontite , Humanos , Bacteroides , RNA Ribossômico 16S/genética , Doenças Periodontais/diagnóstico , Doenças Periodontais/microbiologia , Porphyromonas gingivalis/genética , Periodontite/diagnóstico , Periodontite/microbiologia
6.
Int. j. morphol ; 41(2): 431-436, abr. 2023. ilus, tab
Artigo em Espanhol | LILACS | ID: biblio-1440308

RESUMO

La enfermedad periodontal es una de las principales causas de pérdida dentaria. Clínicamente, esta patología, mediada por la desregulación del sistema inmune producto de una disbiosis ocurrida en el surco gingival, inicia con la inflamación de la encía y evoluciona con el daño irreversible de los tejidos que rodean el diente. El hueso alveolar es uno de los tejidos afectados esta patología, esto debido a la activación de osteoclastos por la sobreexpresión de la proteína RANKL en el huésped. El propósito de este trabajo es determinar el nivel de sobreexpresión de RANKL, en un modelo de células tumorales U2OS, frente a la infección con Porphyromonas gingivalis y Prevotella intermedia. Para identificar el nivel de RANKL, se definieron cuatro grupos: Un grupo control, no tratado; Grupo PG, tratado con P. gingivalis; Grupo PI, tratado con P. Intermedia; y un grupo PG+PI, tratado con ambas bacterias. El nivel relativo de la proteína RANKL fue determinado en el sobrenadante y en los extractos celulares de manera independiente, mediante la técnica Western blot. En sobrenadantes, el grupo PG mostró mayores niveles de RANKL comparados con PI (p < 0,05). En extractos celulares los niveles fueron mayores en el grupo PG+PI (p < 0,05). El grupo PI mostró los niveles más bajos de RANKL. La infección polimicrobiana resulta en una mayor expresión de RANKL en células tumorales U2OS, mientras que frente a la infección P. gingivalis, se observó mayor cantidad de RANKL soluble.


SUMMARY: Periodontal disease is one of the main causes of tooth loss. Clinically, this pathology, mediated by the deregulation of the immune system due to a dysbiosis occurred in the gingival sulcus, begins with the inflammation of the gum and evolves with the irreversible damage of the tissues that surround the tooth. Alveolar bone is one of the most affected tissues by this disease, due to the activation of osteoclasts by the upregulation of RANKL in the host. The aim of this study is to determine the increase of RANKL, in a U2OS tumor cells model, inoculated with Porphyromonas gingivalis and Prevotella intermedia. To identify the level of RANKL, four groups were defined: A control group, not treated; PG group, treated with P.gingivalis; PI group, treated with P. intermedia; and a PG+PI group, treated with both bacteria. The relative level of RANKL was determined in the supernatant and cell extracts independently, using the Western blot technique. In supernatants, the PG group showed higher RANKL levels compared to PI (p < 0.05). In cell extracts the levels were higher in the PG+PI group (p < 0.05.). The PI group showed the lowest levels of RANKL.Polymicrobial infection results in a greater expression of of soluble RANKL was observed.


Assuntos
Doenças Periodontais/microbiologia , Bactérias Anaeróbias/fisiologia , Reabsorção Óssea/microbiologia , Ligante RANK/metabolismo , Células Cultivadas , Western Blotting , Porphyromonas gingivalis/fisiologia , Prevotella intermedia/fisiologia , Linhagem Celular Tumoral , Eletroforese , Ligante RANK/análise
7.
J Periodontal Res ; 58(3): 621-633, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36919705

RESUMO

OBJECTIVES: Periodontal disease occurs frequently in patients with limited cutaneous systemic sclerosis (lcSSc) while data about underlying pathways contributing to periodontal changes are scarce. The aim of this study was to evaluate periodontal disease and to investigate its association with endothelial dysfunction and clinical changes in patients with lcSSc. METHODS: In 38 lcSSc patients and 38 controls, periodontal status was evaluated by disease-specific questionnaire, dental examination including bleeding on probing (BOP), pocket depth, and plaque index, and dental panoramic radiograph. Periodontopathogen bacteria were collected subgingivally using paper points and interleukin-1 (IL-1) gene polymorphisms were evaluated using buccal swabs. Endothelial dysfunction was measured by flow-mediated dilatation, pulse-wave velocity and biochemical analysis, including arginine metabolites and endothelial microparticles. Additionally, lcSSc-specific clinical changes and parameters were recorded. RESULTS: Periodontitis was present in 31 patients with lcSSc (81.6%) and in 27 controls (71.1%) (p = .280). LcSSc patients had a lower teeth number (p = .039) and Eikenella corrodens was to a higher degree detectable in patients with lcSSc (p = .041) while the remaining periodontal parameters revealed no differences between both cohorts. Significant correlations between parameters of arterial stiffness, EUSTAR index, number of teeth and BOP were observed (all p < .05). Detection of Prevotella intermedia was associated with selected IL-1 gene polymorphisms (p = .032) and Porphyromonas gingivalis was associated with severe periodontitis (p = .041). CONCLUSION: Periodontal disease may occur frequently in patients with lcSSc and may be associated with arterial stiffness and with SSc activity.


Assuntos
Doenças Periodontais , Periodontite , Escleroderma Sistêmico , Humanos , Estudos de Casos e Controles , Índice Periodontal , Doenças Periodontais/complicações , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Periodontite/complicações , Prevotella intermedia , Interleucina-1 , Escleroderma Sistêmico/complicações , Aggregatibacter actinomycetemcomitans , Perda da Inserção Periodontal/complicações
8.
BMC Oral Health ; 23(1): 187, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36998066

RESUMO

BACKGROUND: There is increasing evidence that diagnostic salivary tests measuring inflammatory biomarkers are being developed to assess inflammatory status for early detection, prevention, and progression of periodontal disease. Therefore, the aim of the present study was to investigate and identify the salivary biomarker that can predict the inflammatory status of periodontal disease. METHODS: A total of 36 patients (28 women and 8 men) with an average age of 57 years were investigated. Unstimulated saliva was collected from the recruited subjects and analyzed using SillHa, a saliva-testing device that measures bacteria count, saliva buffer capacity, acidity, leukocyte esterase, protein, and ammonia. Periodontal parameters were then obtained by clinical examination and initial periodontal therapy was performed. Data obtained with SillHa were compared with clinical periodontal parameters at baseline, re-examination (three months from baseline), and final examination (six months from re-examination). RESULTS: Leukocyte esterase activity in saliva measured by SillHa; BOP and PCR measured by clinical examination showed a significant difference between baseline and final examination and between re-examination and final examination. Patients in the lower median group (group 1) had a significant difference in leukocyte esterase activity between baseline and final examination and re-examination and final examination. In addition, patients in Group 1 had significantly lower BOP between baseline and final examination. While patients in the higher median group (group 2) showed a modest decrease in leukocyte esterase activity, which was significant only between baseline and final examination, no significant changes were observed concerning BOP. Furthermore, the associated systemic disease was observed in 30% and 81.2% of group 1 and 2 patients, respectively. CONCLUSION: The results suggest that leukocyte esterase activity in saliva measured by SillHa could serve as a reliable diagnostic marker for monitoring inflammatory status in periodontal disease.


Assuntos
Doenças Periodontais , Masculino , Humanos , Feminino , Pessoa de Meia-Idade , Doenças Periodontais/diagnóstico , Doenças Periodontais/microbiologia , Hidrolases de Éster Carboxílico , Biomarcadores/análise , Saliva/química
9.
Nutrients ; 15(4)2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36839184

RESUMO

Obesity causes gut dysbiosis; nevertheless, little is known about the oral microbiome. We aimed to identify differences in the subgingival microbiota influenced by body weight and periodontal status. Patients (n = 75) recruited at the University Dental Hospital Sharjah, United Arab Emirates, were distributed into three equal groups (healthy weight, overweight, and obese) sub-divided into having either no-mild (NM) or moderate-severe (MS) periodontitis. Subgingival plaques were collected. Microbiota were identified by 16S rRNA sequencing using nanopore technology. Linear discriminant analysis demonstrated significant bacterial biomarkers for body weight and periodontal health. Unique microbiota signatures were identified, with enrichment of periopathogens in patients with MS periodontitis (Aggregatibacter actinomycetemcomitans in obese, Tannerella forsythia and Treponema denticola in overweight, Porphyromonas gingivalis and Fusobacterium nucleatum in healthy weight), thus reflecting differences in the microbiota affected by body weight. Other pathogenic bacteria, such as Salmonella enterica and Klebsiella pneumoniae, were enriched in overweight subjects with NM periodontitis, suggesting an increase in the relative abundance of pathogens even in patients with good periodontal health if they were overweight. Alpha and beta diversities were significantly different among the groups. Dysbiosis of the subgingival microbiota in obese and overweight individuals was associated with increased prevalence and severity of periodontal disease, which was correlated with the body mass index. This study highlights the immense importance of the oral microbiome and the need for lifestyle and dental interventions to resolve oral dysbiosis and restore normal homeostasis.


Assuntos
Microbiota , Doenças Periodontais , Periodontite , Humanos , Sobrepeso , Disbiose , RNA Ribossômico 16S/genética , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Periodontite/microbiologia , Obesidade
10.
Acta Biomater ; 158: 759-768, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36638945

RESUMO

Biofilm of oral pathogenic microorganisms induced by their multiplication and coaggregation would lead to periodontitis. In biofilms, the extracellular polymeric substances (EPS) as a protective shield encapsulates the individual bacteria, protecting them against attack. To alleviate periodontal disease, disrupting the EPS of pathogenic bacteria is crucial and challenging. Based on the sufficient capacity of disorganizing EPS of our designed cationic dextrans, we hypothesized that these polymers could be competent in relieving periodontitis. We validated that cationic dextrans could induce the phase transition of EPS in biofilms, especially the Porphyromonas gingivalis (P. gingivalis), a keystone periodontal pathogen, thus effectively destroying biofilm in vitro. More importantly, satisfactory in vivo treatment was achieved in a rat periodontal disease model. In summary, the study exploited a practical and effective strategy to treat periodontitis with cationic dextrans' powerful biofilm-controlling potential. STATEMENT OF SIGNIFICANCE: Periodontal disease is closely related to dental plaque biofilms on the tooth surface. The biofilm forms gel structures and shields the bacteria underneath, thus protecting oral pathogens from traditional anti-bacterial reagents. Due to limited penetration into gel, the efficacy of these reagents in biofilm elimination is restricted. Our designed cationic dextran could wipe out the coverage of gel-like EPS to disperse encapsulated bacteria. Such superior capacity endowed them with satisfactory effect in disrupting biofilm. Notably, in a rat periodontitis model, cationic dextrans dramatically suppressed alveolar bone loss and alleviated periodontal inflammation by controlling dental plaque. Given the increasing global concerns about periodontal disease, it's worth expanding the application of cationic dextrans both scientifically and clinically.


Assuntos
Placa Dentária , Doenças Periodontais , Periodontite , Animais , Ratos , Dextranos/farmacologia , Doenças Periodontais/microbiologia , Biofilmes , Periodontite/tratamento farmacológico , Periodontite/microbiologia , Porphyromonas gingivalis
11.
Quintessence Int ; 54(5): 372-383, 2023 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-36622053

RESUMO

OBJECTIVES: Gingivitis refers to inflammation of the gingiva and its connective tissues. Research has revealed a higher prevalence of gingivitis in individuals with intellectual disability than in healthy individuals. Milk fermented with Lacticaseibacillus rhamnosus L8020 (L8020 yogurt) inhibits the accumulation of periodontal disease-related pathogens in vitro and alleviates the symptoms of periodontal disease. The aim of this study was to investigate the influence of L8020 yogurt on oral microbiota and the abundance of four periodontal pathogens (Tannerella forsythia, Porphyromonas gingivalis, Prevotella intermedia, and Treponema denticola) and on the microbiota in individuals with intellectual disability and gingivitis. METHOD AND MATERIALS: Forty-one outpatients with intellectual disability participated in this study. To examine the effects of daily consumption of L8020 yogurt, the patients were randomly divided into L8020 (test group, n = 21) and placebo (n = 20) yogurt groups. All patients consumed 80 g of yogurt for 12 weeks. Oral examination was performed before the first intake of yogurt and dental plaque was collected before and after the intake of yogurt. DNA was extracted from dental plaque and subjected to next-generation sequencing. RESULTS: The relative abundance of T forsythia was significantly lower in the test group than in the placebo group. Additionally, the relative abundance of the four pathogens reduced after 84 days of consuming L8020 yogurt compared with that after consuming placebo yogurt. CONCLUSION: Mixing L rhamnosus L8020 with probiotic products that are consumed daily would be effective in suppressing the increase in periodontal disease-causing bacteria and beneficial for individuals with intellectual disability.


Assuntos
Placa Dentária , Gengivite , Deficiência Intelectual , Lacticaseibacillus rhamnosus , Doenças Periodontais , Humanos , Lacticaseibacillus , Placa Dentária/microbiologia , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Prevotella intermedia , Treponema denticola , Aggregatibacter actinomycetemcomitans
12.
Sci Rep ; 13(1): 544, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36631604

RESUMO

Periodontal disease can induce dysbiosis, a compositional and functional alteration in the microbiota. Dysbiosis induced by periodontal disease is known to cause systemic inflammation and may affect transplant immunity. Here, we examined the effects of periodontal disease-related intestinal dysbiosis on transplant immunity using a mouse model of allogenic skin graft in which the mice were orally administered the periodontal pathogen Porphyromonas gingivalis (Pg). For 6 weeks, the Pg group orally received Pg while the control group orally received phosphate-buffered saline solution. After that, both groups received allogenic skin grafts. 16 s rRNA analysis of feces revealed that oral administration of Pg significantly increased three short chain fatty acids (SCFAs) producing genera. SCFA (acetate and propionate) levels were significantly higher in the Pg group (p = 0.040 and p = 0.005). The ratio of regulatory T cells, which are positively correlated with SCFAs, to total CD4+ T cells in the peripheral blood and spleen was significantly greater (p = 0.002 and p < 0.001) in the Pg group by flowcytometry. Finally, oral administration of Pg significantly prolonged skin graft survival (p < 0.001) and reduced pathological inflammation in transplanted skin grafts. In conclusion, periodontal pathogen-induced intestinal dysbiosis may affect transplant immunity through increased levels of SCFAs and regulatory T cells. (198 words).


Assuntos
Disbiose , Microbioma Gastrointestinal , Rejeição de Enxerto , Doenças Periodontais , Transplante de Pele , Disbiose/complicações , Disbiose/microbiologia , Ácidos Graxos Voláteis , Inflamação/patologia , Doenças Periodontais/complicações , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/microbiologia , Animais
13.
Biotechnol Genet Eng Rev ; 39(1): 85-117, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35861776

RESUMO

Oral microbial ecosystems are vital in maintaining the health of the oral cavity and the entire body. Oral microbiota is associated with the progression of oral diseases such as dental caries, periodontal diseases, head and neck cancer, and several systemic diseases such as cardiovascular disease, rheumatoid arthritis, adverse pregnancy outcomes, diabetes, lung infection, colorectal cancer, and pancreatic cancer. Buccal mucosa, tongue dorsum, hard palate, saliva, palatine tonsils, throat, keratinized gingiva, supra-gingival plaque, subgingival plaque, dentures, and lips are microbial habitats of the oral cavity. Porphyromonas gingivalis may have a role in the development of periodontal diseases, oral cancer, diabetes, and atherosclerotic disease. Fusobacterium nucleatum showed a higher abundance in periodontal diseases, oral and colon cancer, adverse pregnancy outcomes, diabetes, and rheumatoid arthritis. The higher abundance of Prevotella intermedia is typical in periodontal diseases, rheumatoid arthritis, and adverse pregnancy outcome. S. salivarius displayed higher abundance in both dental caries and OSCC. Oral bacteria may influence systemic diseases through inflammation by releasing pro inflammatory cytokines. Identification of oral bacteria using culture-dependent approaches and next-generation sequencing-based metagenomic approaches is believed to significantly identify the therapeutic targets and non-invasive diagnostic indicators in different human diseases. Oral bacteria in saliva could be exploited as a non-invasive diagnostic indicator for the early detection of oral and systemic disorders. Other therapeutic approaches such as the use of probiotics, green tea polyphenol, cold atmospheric plasma (CAP) therapy, antimicrobial photodynamic therapy, and antimicrobial peptides are used to inhibit the growth of biofilm formation by oral bacteria.


Porphyromonas gingivalis may have a role in the development of periodontal diseases, oral cancer, diabetes, and atherosclerotic diseaseFusobacterium nucleatum showed a higher abundance in periodontal diseases, oral and colon cancer, adverse pregnancy outcomes, diabetes, and rheumatoid arthritisOral bacteria may influence systemic diseases through inflammation by releasing pro inflammatory cytokines.Identification of oral bacteria in saliva may be used as a non-invasive diagnostic indicator for the early detection of oral and systemic disorders.


Assuntos
Artrite Reumatoide , Cárie Dentária , Microbiota , Doenças Periodontais , Feminino , Humanos , Gravidez , Doenças Periodontais/microbiologia , Porphyromonas gingivalis
14.
J Complement Integr Med ; 20(1): 129-136, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36398568

RESUMO

OBJECTIVES: Lycium barbarum, commonly known as goji berry, Himalayan berry, or Tibetian berry, is emerging as a popular "superfood" with anti-inflammatory and antioxidant properties. However, its use for the management of oral inflammatory diseases has not been explored. The present study aims to evaluate the antimicrobial, anti-adhesion, anti-biofilm, and cytotoxic properties of an ethanolic extract of L. barbarum (LBE) against common oral and periodontal pathogens. METHODS: The antimicrobial properties of LBE against five potential periodontal pathogens (Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, Prevotella intermedia, Tanerella forsythia) were tested and compared to chlorhexidine and doxycycline using serial dilution and disc diffusion assay. The MTT Assay was performed for evaluating the cytotoxicity and cell viability of the LBE on the gingival fibroblast and modified keratinocyte cell lines. The anti-adhesion and anti-biofilm properties of LBE against P. gingivalis at its minimal bactericidal value were also assessed. RESULTS: LBE inhibited the growth of periodontal pathogens as compared to control, however, the zone of inhibition of LBE was less when compared to doxycycline and chlorhexidine. The de novo extract showed a maximum zone of inhibition against Tf and Aa. The LBE extract was also compatible to gingival fibroblast tissues and oral keratinocytes at 1 mg/mL. CONCLUSIONS: L. barbarum is a promising alternative to Chlorhexidine for the management of oral and periodontal infections.


Assuntos
Anti-Infecciosos , Lycium , Doenças Periodontais , Clorexidina/farmacologia , Doxiciclina , Doenças Periodontais/tratamento farmacológico , Doenças Periodontais/microbiologia , Porphyromonas gingivalis
15.
J Oral Biosci ; 65(1): 72-79, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36473619

RESUMO

OBJECTIVES: Periodontal disease is triggered by oral microbiome dysbiosis. Thus, to prevent its onset, it is important to maintain relative abundance of periodontal pathogenic bacteria in the oral microbiome at a low level. While Phellodendron bark extract (PBE) and its active ingredient, berberine, exert antibacterial effects on periodontal pathogenic bacteria, such as Porphyromonas gingivalis, their effects on the oral microbiome as a whole remain unknown. Therefore, we aimed to clarify the potential of PBE and berberine chloride (BC) in regulating the relative abundance of periodontal pathogenic bacteria in the oral microbiome. METHODS: Saliva was collected from 20 participants. Each participant's saliva was combined separately with P. gingivalis suspension and either PBE or BC in a modified basal medium. The samples were then incubated under anaerobic conditions for 24 h. After cultivation, we determined the total bacterial concentration using quantitative polymerase chain reaction analysis and the bacterial composition using 16 S ribosomal RNA gene sequencing. RESULTS: The total bacterial concentration was reduced because of treatment with PBE and BC. Bacterial 16 S ribosomal RNA gene sequencing confirmed that treatment with PBE and BC significantly reduced the relative abundance of periodontal pathogenic bacteria, including red and orange complex bacteria. CONCLUSIONS: Our findings suggest that PBE and BC reduce the relative abundance of periodontal pathogenic bacteria in the oral microbiome. Thus, PBE and BC can aid in preventing periodontal disease, given their ability to regulate the oral microbiome composition and their anti-inflammatory effects.


Assuntos
Berberina , Microbiota , Doenças Periodontais , Phellodendron , Humanos , Cloretos , Casca de Planta , Doenças Periodontais/microbiologia , Porphyromonas gingivalis , Microbiota/genética
16.
Braz. j. oral sci ; 22: e231137, Jan.-Dec. 2023. ilus
Artigo em Inglês | LILACS, BBO - Odontologia | ID: biblio-1523140

RESUMO

The purpose of this in vitro study was to analyze the influence of nicotine on the extracellular polysaccharides in Fusobacterium nucleatum biofilm. Methods: F. nucleatum (ATCC 10953) biofilms supplemented with different concentrations of nicotine (0, 0.5, 1, 2, 4, and 8 mg/mL) were grown in two different BHI broth conditions [no sucrose and 1% sucrose]. Extracellular polysaccharides assay, pH measurements, and a spectrophotometric assay were performed. Data were submitted for ANOVA and Tukey honestly significant difference analyses (HSD) tests (α =.05). Results: Extracellular polysaccharides synthesis was influenced by an interaction between nicotine concentrations and growth medium solution containing sucrose (P<.05). The pH values declined in the sucrose-exposed biofilm were greater than in the group exposed only to nicotine (P<.05). The biofilm exposed to sucrose and nicotine had a higher total biofilm growth (P<.05) than the nicotine-treated biofilm without sucrose. Conclusions: Regardless of sucrose exposure, biofilms exposed to different nicotine concentrations influenced the amount of extracellular polysaccharides


Assuntos
Humanos , Polissacarídeos Bacterianos/síntese química , Fusobacterium nucleatum/crescimento & desenvolvimento , Biofilmes/crescimento & desenvolvimento , Nicotina/farmacologia , Doenças Periodontais/microbiologia , Espectrofotometria , Sacarose/administração & dosagem , Meios de Cultura , Cárie Dentária/microbiologia , Nicotina/administração & dosagem
17.
Front Cell Infect Microbiol ; 12: 1035324, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36579339

RESUMO

Dental calculus has long been considered as a vital contributing factor of periodontal diseases. Our review focuses on the role of dental calculus as a repository and discusses the bioinformation recently reported to be concealed in dental calculus from three perspectives: time-varying oral condition, systemic diseases, and anthropology at various times. Molecular information representing an individual's contemporary oral health status could be detected in dental calculus. Additionally, pathogenic factors of systemic diseases were found in dental calculus, including bacteria, viruses and toxic heavy metals. Thus, dental calculus has been proposed to play a role as biological data storage for detection of molecular markers of latent health concerns. Through the study of environmental debris in dental calculus, an overview of an individual's historical dietary habits and information about the environment, individual behaviors and social culture changes can be unveiled. This review summarizes a new role of dental calculus as a repository of bioinformation, with potential use in the prediction of oral diseases, systemic diseases, and even anthropology.


Assuntos
Microbiota , Doenças Periodontais , Humanos , Cálculos Dentários , Doenças Periodontais/microbiologia , Bactérias/genética
18.
Bol. malariol. salud ambient ; 62(6): 1205-1211, dic. 2022. tab., ilus.
Artigo em Espanhol | LILACS, LIVECS | ID: biblio-1427327

RESUMO

Entamoeba gingivalis y Trichomonas tenaxson protozoos que generalmente se encuentran en la cavidad oral de los humanos. E. gingivalis vive en la superficie de los dientes y las encías, bolsas gingivales cerca de la base de los dientes y rara vez en las criptas de las amígdalas. Por lo general, se transmiten por contacto directo de una persona a otra al besarse, rociarse con gotas o compartir utensilios para comer. Se sabe que hasta el 95% de las personas con poca higiene bucal pueden estar infectadas con esta ameba. Por otro lado, Trichomonas tenax es una pequeña tricomona que generalmente se encuentra en la cavidad oral del 5 al 10% de los humanos. Aunque se consideran comensales anaeróbicos no patógenos, el refugio de este protozoo es más común entre personas con mala higiene bucal o enfermedades. Los informes epidemiológicos demostraron variables tasas de prevalencia de la infección por E. gingivalis entre los niños, y su ocurrencia puede variar de acuerdo a la edad, presencia de gingivitis, periodontitis y condiciones inmunosupresoras. Los niños y adolescentes en edad escolar son extremadamente susceptibles a una amplia gama de infecciones gingivales como bacterias y protozoos. El objetivo de este trabajo es determinar la prevalencia de los protozoarios E. gingivalis y T. tenax en adolescentes con edades comprendidas entre 10 y 19 años en una población de 185 pacientes de la ciudad peruana de Huancayo, Perú. La prevalencia fue mayor en la población femenina (14,05%) que en la población masculina (11,35%), con un mayor porcentaje de prevalencia entre las edades de 17 a 19 años (12,43%), pH salival menor a 6,7 y con enfermedades periodentales (16,22%). Los resultados estadísticos demostraron que la edad (p=0,001), pH salival (p=0,024) y diagnóstico de otras patologías periodentales (p<0,001) fueron estadísticamente significativas(AU)


Entamoeba gingivalis and Trichomonas tenax are protozoa that are usually found in the oral cavity of humans. E. gingivalis lives on the surface of the teeth and gums, gum pockets near the base of the teeth, and rarely in the crypts of the tonsils. They are usually spread by direct person-to-person contact through kissing, dousing, or sharing eating utensils. It is known that up to 95% of people with poor oral hygiene may be infected with this amoeba. On the other hand, Trichomonas tenax is a small trichomona that is usually found in the oral cavity of 5-10% of humans. Although they are considered non-pathogenic anaerobic commensals, the harborage of this protozoan is more common among people with poor oral hygiene or disease. Epidemiological reports have shown variable prevalence rates of E. gingivalis infection among children, and its occurrence may vary according to age, presence of gingivitis, periodontitis, and immunosuppressive conditions. School-age children and adolescents are extremely susceptible to a wide range of gingival infections including bacteria and protozoa. The objective of this work is to determine the prevalence of the protozoa E. gingivalis and T. tenax in adolescents between the ages of 10 and 19 in a population of 185 patients from the Peruvian city of Huancayo, Peru. The prevalence was higher in the female population (14.05%) than in the male population (11.35%), with a higher percentage of prevalence between the ages of 17 to 19 years (12.43%), lower salivary pH to 6.7 and with periodontal diseases (16.22%). The statistical results showed that age (p=0.001), salivary pH (p=0.024) and diagnosis of other periodontal pathologies (p<0.001) were statistically significant(AU)


Assuntos
Humanos , Masculino , Feminino , Criança , Adolescente , Adulto Jovem , Doenças Periodontais/microbiologia , Doenças Periodontais/epidemiologia , Infecções por Protozoários/epidemiologia , Peru/epidemiologia , Prevalência , Estudos Transversais
19.
Br Dent J ; 233(10): 855-858, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36434225

RESUMO

Necrotising periodontal diseases can present with significant morbidity and, whilst unusual, they are not entirely uncommon in primary care. With this in mind, members of the dental team should be aware of relatively recent changes to the classification and management of these diseases to optimise patient outcomes. Similarly, understanding the bacteriology, patterns of tissue breakdown, and management and maintenance of these conditions will allow clinicians to manage these diseases should they present. The progression of necrotising diseases in Sub-Saharan Africa to present as noma is also discussed. The objective is to help the reader understand the classification and management of necrotising periodontal diseases.


Assuntos
Doenças Periodontais , Humanos , Doenças Periodontais/diagnóstico , Doenças Periodontais/terapia , Doenças Periodontais/microbiologia
20.
J Periodontal Res ; 57(6): 1227-1237, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36261869

RESUMO

OBJECTIVE: This investigation explored oral-gut microbial signatures with potential to distinguish among periodontal conditions. BACKGROUND DATA: The interplay between the oral and gut microbiomes may be a critical pathway linking periodontal diseases and systemic inflammatory disorders. The mechanisms by which oral microorganisms translocate to the gut and cause microbial dysbiosis, favoring an inflammatory state, are still unknown. As a first approach, characterization of oral-gut microbial profiles associated with periodontal health and diseases can provide insights on such mechanisms of etiology and pathogenesis. METHODS: Fecal and saliva samples from individuals with periodontal health (PH, 8), gingivitis (GG, 17), and periodontitis (PD, 24) were analyzed for their microbial composition by 16S rRNA gene sequencing. Microbial taxa were compared and correlated to periodontal parameters. Multivariate discriminant analysis (MDA) was carried out to identify profiles related to health and disease. RESULTS: Few significant differences in oral-gut taxa were detected among clinical groups, although increase in fecal Fusobacterium nucleatum ss vincentii and salivary Aggregatibacter actinomycetemcomitans, Parvimonas micra, and Fretibacterium sp. HMT358 were strongly correlated with deep pockets and inflammation (p < .01). Over 50% of the fecal microbiota comprised microorganisms shared between oral and gut sites, whereas oral taxa were detected in approximately 9%, particularly enriched in GG fecal samples (p = .04). Trends for lower fecal richness and higher salivary diversity in PD compared to PH were observed. MDA was able to classify correctly 82% of the patients into the clinical groups. Main classifiers of periodontitis were high BMI, older age, and enrichment of oral-fecal Leptotrichia sp. HMT4, Peptostreptococcus stomatis, Dialister invisus, and a novel Lautropia sp. HMTC89-like organism. CONCLUSION: Within the limitations of an exploratory investigation, specific profiles of oral-gut taxa, including known and potential novel organisms, combined with social-demographic features were able to discriminate individuals with periodontal diseases in this study population.


Assuntos
Microbioma Gastrointestinal , Microbiota , Doenças Periodontais , Periodontite , Humanos , RNA Ribossômico 16S/genética , Microbiota/genética , Periodontite/diagnóstico , Periodontite/microbiologia , Aggregatibacter actinomycetemcomitans/genética , Doenças Periodontais/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...