Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 457
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39000324

RESUMEN

The objective of this review is to identify the microbiological alterations caused by various therapy modalities by critically analyzing the current findings. We limited our search to English-language papers published between 1 January 2004 and 7 May 2024 in PubMed, Scopus, and Web of Science that were relevant to our topic. In the search approach, the Boolean keywords "microbio*" AND "periodontitis" were used. A total of 5152 papers were obtained from the databases Web of Science (2205), PubMed (1793), and Scopus (1154). This resulted in 3266 articles after eliminating duplicates (1886), and 1411 entries were eliminated after their titles and abstracts were examined. The qualitative analysis of the 22 final articles is included in this study. Research on periodontal disease shows that periodontitis alters the oral microbiome and increases antibiotic resistance. Treatments like scaling and root planing (SRP), especially when combined with minocycline, improve clinical outcomes by reducing harmful bacteria. Comprehensive mechanical debridement with antibiotics, probiotics, EMD with bone grafts, and other adjunctive therapies enhances periodontal health. Personalized treatment strategies and advanced microbial analyses are crucial for effective periodontal management and antibiotic resistance control.


Asunto(s)
Enfermedades Periodontales , Humanos , Enfermedades Periodontales/terapia , Enfermedades Periodontales/microbiología , Enfermedades Periodontales/genética , Microbiota , Antibacterianos/uso terapéutico , Probióticos/uso terapéutico
2.
BMC Immunol ; 25(1): 39, 2024 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-38943064

RESUMEN

BACKGROUND: Previous observational studies have shown a bidirectional association between immune-mediated inflammatory disorders (IMID) and periodontal disease. However, evidence regarding the causal role of IMID and periodontal disease is still lacking. Therefore, we conducted a bidirectional two-sample Mendelian randomization (MR) study to uncover the potential genetic causal effects between IMID and periodontal disease. METHODS: Bidirectional two-sample MR analysis was employed. Data for ten IMIDs were sourced from genome-wide association studies (GWAS) conducted by the FinnGen Consortium (range from 1023 to 36321 cases) and UK Biobank (UKB) (range from 150 to 17574 cases). Furthermore, GWAS data for periodontal disease were obtained from the FinnGen Consortium (87497 cases), UKB (458 cases), and Gene Lifestyle Interactions in Dental Endpoints (GLIDE) consortium (17,353 periodontitis cases). Subsequently, the causal relationships were analyzed by random effects inverse variance weighting, weighted median, and MR-Egger. Sensitivity analyses were performed using the Cochrane Q test, funnel plot, and Mr-Egger intercept test to ensure robustness. Eventually, replication analysis and meta-analysis across different databases were carried out. RESULTS: Systemic lupus erythematosus (SLE) [IVW: OR = 1.079 (95% CI: 1.032-1.128) and P < 0.001], Sjogren syndrome [IVW: OR = 1.082 (95% CI: 1.012-1.157) and P = 0.022] and hypothyroidism [IVW: OR = 1.52 (95% CI: 1.13-2.04) and P = 0.005] may increase the risk of periodontal disease. In addition, periodontal disease may reduce the risk of SLE [IVW: OR = 0.8079 (95% CI: 0.6764-0.9650) and P = 0.019] and hyperthyroidism [IVW: OR = 5.59*10-9 (95% CI: 1.43*10-15-2.18*10-2) and P = 0.014]. Meta-analysis indicated a causal correlation between SLE and an increased risk of periodontal disease: [OR = 1.08 (95% CI: 1.03-1.13), P = 0.0009]. No significant evidence suggests bilateral causal relationships between other IMIDs and periodontal disease. No significant estimation of heterogeneity or pleiotropy is detected. CONCLUSIONS: Our study has confirmed a genetic causal relationship between IMIDs and periodontal disease, thereby unveiling novel potential mechanisms underlying IMIDs and periodontal disease. This discovery is promising in fostering interdisciplinary collaboration between clinicians and stomatologists to facilitate appropriate and precise screening, prevention, and early treatment of IMIDs and periodontal disease.


Asunto(s)
Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Enfermedades Periodontales , Humanos , Enfermedades Periodontales/genética , Enfermedades Periodontales/epidemiología , Enfermedades Periodontales/inmunología , Polimorfismo de Nucleótido Simple , Inflamación/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología
3.
Medicine (Baltimore) ; 103(24): e38490, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38875422

RESUMEN

BACKGROUND: Previous observational studies have suggested a possible association between periodontal disease and gastric cancer (GC); however, a causal relationship has not yet been established. This study aimed to explore the causal relationship between the 2 through a 2-sample bidirectional Mendelian randomization (MR) study. METHODS: Genome-wide association studies (GWAS) summary statistics were obtained from publicly available GWAS and relevant databases. Two-sample bidirectional MR analysis was conducted to investigate the causal relationship between periodontal disease and GC using the inverse-variance weighted (IVW) method selected as the primary analytical approach. Cochran Q test, MR-PRESSO, MR-pleiotropy, and leave-one-out analyses were performed to assess heterogeneity, pleiotropy, and sensitivity. RESULTS: In European ancestry, IVW analysis revealed no causal relationship between periodontal disease and GC (OR = 1.873; 95% CI [4.788e-10, 7.323e + 09]; P = .956), or between loose teeth and GC (OR = 1.064; 95% CI [0.708, 1.598]; P = .765). In East Asian ancestry, there was no causal relationship between periodontitis and GC according to IVW (OR = 0.948; 95% CI [0.886, 1.015]; P = .126). Conversely, according to the results of the IVW analysis, there was no causal relationship between GC and periodontal disease, regardless of European or East Asian ancestry. Furthermore, there was no heterogeneity or pleiotropy in the causal relationships between these variables (all P > .05), suggesting a certain level of reliability in our results. CONCLUSION: Within the limitations of this MR study, we found no mutual causal relationship between periodontal disease and GC. This finding can prevent overtreatment by clinical physicians and alleviate the psychological burden on patients.


Asunto(s)
Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Enfermedades Periodontales , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Enfermedades Periodontales/genética , Enfermedades Periodontales/epidemiología , Pueblo Asiatico/genética , Población Blanca/genética , Población Blanca/estadística & datos numéricos
4.
J Med Life ; 17(2): 195-200, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38813354

RESUMEN

Numerous studies have established a link between gene variants within the inflammasome complex and the incidence of periodontitis and cardiovascular illness across various ethnic groups. This study investigated the association between PYCARD gene polymorphism and susceptibility to periodontal disease and coronary heart disease (CHD) and their correlation with clinical periodontal indices. A total of 120 participants were enrolled, categorized into four groups: 30 healthy controls (C), 30 patients with generalized periodontitis (P), 30 patients with atherosclerotic CHD but clinically healthy periodontium (AS-C), and 30 patients with both atherosclerotic CHD and generalized periodontitis (AS-P). We recorded demographic data, collected blood samples, and measured periodontal indices, including plaque index, clinical attachment loss, bleeding on probing, and pocket depth. The genomic variant of the PYCARD gene was analyzed using a conventional polymerase reaction. A significant prevalence of T and G allele mutations and a higher distribution of CT and TT genotypes in PYCARD C/T (rs8056505) and the AG genotype in PYCARD A/G (rs372507365) were observed in groups P, AS-P, and AS-C. These single nucleotide polymorphisms (SNPs) were also positively correlated with the severity of clinical periodontitis indices. Our findings suggest that the increased frequency of T and G alleles and the distribution of CT, TT, and AG genotypes in PYCARD SNPs are significantly associated with an elevated risk for periodontal disease and CHD. These SNPs may participate in the pathogenesis of these conditions. The study reinforces the potential role of these genetic markers as risk factors for both diseases in the Iraqi population.


Asunto(s)
Enfermedad Coronaria , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Alelos , Proteínas Adaptadoras de Señalización CARD/genética , Estudios de Casos y Controles , Enfermedad Coronaria/genética , Genotipo , Enfermedades Periodontales/genética , Periodontitis/genética , Polimorfismo de Nucleótido Simple/genética
5.
PLoS One ; 19(5): e0304117, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38820296

RESUMEN

OBJECTIVE: This study aimed to explore the bidirectional causal relationship between periodontal disease-related phenotype (PDRP) and knee osteoarthritis (KOA) in a European population using a two-sample Mendelian Randomization (MR) approach. METHODS: We leveraged publicly available GWAS summary statistics for PDRP (n = 975) and KOA (n = 403,124), assessing their roles as both exposures and outcomes. Our comprehensive MR analysis employed various methods, including inverse variance weighting (IVW), weighted median, Egger regression, simple mode, and weighted mode, to enhance the robustness of our findings. To ensure the reliability of our instrumental variables, we implemented a rigorous screening process based on p-values and F-values, utilized Phenoscanner to investigate potential confounders, and conducted sensitivity analyses. RESULTS: Our analysis identified five SNPs associated with PDRP and three SNPs with KOA, all surpassing the genome-wide significance threshold, as instrumental variables. The IVW method demonstrated a significant causal relationship from PDRP to KOA (beta = 0.013, SE = 0.007, P = 0.035), without evidence of directional pleiotropy (MR-Egger regression intercept = 0.021, P = 0.706). No support was found for reverse causality from KOA to PDRP, as further MR analyses yielded non-significant P-values. Additionally, funnel plots and Cochran's Q test detected no significant heterogeneity or directional pleiotropy, confirming the robustness of our results. In multivariate analysis, when considering smoking, alcohol consumption, BMI collectively no direct causal relationship between KOA and PDRP. Conversely, smoking and higher BMI were independently associated with an increased risk of KOA. CONCLUSION: In conclusion, our analysis revealed no direct causal relationship from KOA to PDRP. However, a causal relationship from PDRP to KOA was observed. Notably, when adjusting for potential confounders like smoking, alcohol intake, and BMI, both the causal connection from PDRP to KOA and the inverse relationship were not substantiated.


Asunto(s)
Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Osteoartritis de la Rodilla , Enfermedades Periodontales , Fenotipo , Polimorfismo de Nucleótido Simple , Humanos , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/etiología , Enfermedades Periodontales/genética , Enfermedades Periodontales/complicaciones , Masculino , Femenino , Predisposición Genética a la Enfermedad , Factores de Riesgo
6.
Adv Clin Chem ; 120: 145-168, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38762240

RESUMEN

Due to technologic advancements, periodontology has witnessed a boost in biomarker research over the past three decades. Indeed, with the aid of omics, our understanding of the healthy periodontium, pathogenesis of periodontal diseases, and healing after periodontal treatment has improved significantly. Yet, the traditional methods, periodontal probing and radiographies, remain the most common methods to diagnose periodontal disease and monitor treatment. Although these approaches can produce reliable diagnostic outcomes, they generally detect disease only after significant tissue degradation thus making treatment outcome highly uncertain. Accordingly, laboratories worldwide have collaborated with clinicians to design accurate, rapid and cost-effective biomarkers for periodontal disease diagnosis. Despite these efforts, biomarkers that can be widely used in early disease diagnosis and for treatment outcome prediction are far from daily use. The aim of this chapter is to give a general overview on periodontal health and diseases, and review recent advancements in periodontal biomarker research. A second aim will discuss the strengths and limitations of translating periodontal biomarker research to clinical practice. Genetic biomarkers of periodontitis are not discussed as the available confirmatory data is scarce.


Asunto(s)
Biomarcadores , Enfermedades Periodontales , Humanos , Biomarcadores/análisis , Biomarcadores/metabolismo , Enfermedades Periodontales/diagnóstico , Enfermedades Periodontales/metabolismo , Enfermedades Periodontales/genética
7.
BMC Pulm Med ; 24(1): 220, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702679

RESUMEN

BACKGROUND: Recent research suggests that periodontitis can increase the risk of chronic obstructive pulmonary disease (COPD). In this study, we performed two-sample Mendelian randomization (MR) and investigated the causal effect of periodontitis (PD) on the genetic prediction of COPD. The study aimed to estimate how exposures affected outcomes. METHODS: Published data from the Gene-Lifestyle Interaction in the Dental Endpoints (GLIDE) Consortium's genome-wide association studies (GWAS) for periodontitis (17,353 cases and 28,210 controls) and COPD (16,488 cases and 169,688 controls) from European ancestry were utilized. This study employed a two-sample MR analysis approach and applied several complementary methods, including weighted median, inverse variance weighted (IVW), and MR-Egger regression. Multivariable Mendelian randomization (MVMR) analysis was further conducted to mitigate the influence of smoking on COPD. RESULTS: We chose five single-nucleotide polymorphisms (SNPs) as instrumental variables for periodontitis. A strong genetically predicted causal link between periodontitis and COPD, that is, periodontitis as an independent risk factor for COPD was detected. PD (OR = 1.102951, 95% CI: 1.005-1.211, p = 0.039) MR-Egger regression and weighted median analysis results were coincident with those of the IVW method. According to the sensitivity analysis, horizontal pleiotropy's effect on causal estimations seemed unlikely. However, reverse MR analysis revealed no significant genetic causal association between COPD and periodontitis. IVW (OR = 1.048 > 1, 95%CI: 0.973-1.128, p = 0.2082) MR Egger (OR = 0.826, 95%CI:0.658-1.037, p = 0.1104) and weighted median (OR = 1.043, 95%CI: 0.941-1.156, p = 0.4239). The results of multivariable Mendelian randomization (MVMR) analysis, after adjusting for the confounding effect of smoking, suggest a potential causal relationship between periodontitis and COPD (P = 0.035). CONCLUSION: In this study, periodontitis was found to be independent of COPD and a significant risk factor, providing new insights into periodontitis-mediated mechanisms underlying COPD development.


Asunto(s)
Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple , Enfermedad Pulmonar Obstructiva Crónica , Fumar , Humanos , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Factores de Riesgo , Fumar/epidemiología , Fumar/efectos adversos , Periodontitis/genética , Periodontitis/epidemiología , Índice de Severidad de la Enfermedad , Predisposición Genética a la Enfermedad , Enfermedades Periodontales/genética , Enfermedades Periodontales/epidemiología
8.
Brain Behav ; 14(4): e3486, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38648391

RESUMEN

BACKGROUND: Evidence from observational studies and clinical trials suggests an association between periodontal disease and Alzheimer's disease (AD). However, the causal relationship between periodontal disease and AD remains to be determined. METHODS: We obtained periodontal disease data from the FinnGen database and two sets of AD data from the IEU consortium and PGC databases. Subsequently, we conducted a two-sample Mendelian randomization (MR) analysis to investigate the causal relationship between periodontal disease and AD. RESULTS: The results of the random-effects IVW analysis revealed no evidence of a genetic causal relationship between periodontal disease and AD, regardless of whether the AD data from the IEU consortium or the AD data from the PGC database were utilized. No heterogeneity, multiple effects of levels, or outliers were observed in this study. CONCLUSIONS: Our findings indicate that there is no causal relationship between periodontal disease and AD at the genetic level.


Asunto(s)
Enfermedad de Alzheimer , Análisis de la Aleatorización Mendeliana , Enfermedades Periodontales , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/epidemiología , Humanos , Enfermedades Periodontales/genética , Enfermedades Periodontales/epidemiología , Polimorfismo de Nucleótido Simple , Predisposición Genética a la Enfermedad
9.
Sci Rep ; 14(1): 5360, 2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38438471

RESUMEN

In dogs, Porphyromonas gulae is a major periodontal pathogen with 41-kDa proteins polymerizing to form a filamentous structure called fimbriae or pili, termed FimA. FimA is classified into three genotypes: A, B, and C, and there are combinations of types A, B, C, A/B, A/C, B/C, and A/B/C. Periodontal disease is the most common oral disease in small dogs, but the periodontal disease status and P. gulae colonization at each dog age and breed remain unclear. In this study, we stratified 665 small dogs and analyzed the periodontal status and distribution of P. gulae with each FimA genotype. Dogs with periodontal disease and FimA genotype tended to increase with age. The dogs with at least one FimA genotype had significantly more severe periodontal disease compared with P. gulae-negative dogs (P < 0.01). Additionally, periodontal status was significantly associated with specific FimA genotype distribution in Toy Poodles and Chihuahuas (P < 0.05), whereas there was no such association in Dachshunds. These results suggest that the onset of periodontal disease and P. gulae colonization are related and progress with age. The relationship between periodontal disease and FimA genotype may differ depending on the dog breeds.


Asunto(s)
Enfermedades Periodontales , Perros , Animales , Enfermedades Periodontales/genética , Enfermedades Periodontales/veterinaria , Porphyromonas/genética , Citoesqueleto , Genotipo
10.
J Clin Periodontol ; 51(4): 464-473, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38185798

RESUMEN

AIM: WHIM (warts, hypogammaglobulinaemia, infections and myelokathexis) syndrome is a rare combined primary immunodeficiency disease caused by gain-of-function (GOF) mutations in the chemokine receptor CXCR4 and includes severe neutropenia as a common feature. Neutropenia is a known risk factor for periodontitis; however, a detailed periodontal evaluation of a WHIM syndrome cohort is lacking. This study aimed to establish the evidence base for the periodontal status of patients with WHIM syndrome. MATERIALS AND METHODS: Twenty-two adult WHIM syndrome patients and 22 age- and gender-matched healthy volunteers (HVs) were evaluated through a comprehensive medical and periodontal examination. A mouse model of WHIM syndrome was assessed for susceptibility to naturally progressing or inducible periodontitis. RESULTS: Fourteen patients with WHIM syndrome (63.6%) and one HV (4.5%) were diagnosed with Stage III/IV periodontitis. No WHIM patient presented with the early onset, dramatic clinical phenotypes typically associated with genetic forms of neutropenia. Age, but not the specific CXCR4 mutation or absolute neutrophil count, was associated with periodontitis severity in the WHIM cohort. Mice with a Cxcr4 GOF mutation did not exhibit increased alveolar bone loss in spontaneous or ligature-induced periodontitis. CONCLUSIONS: Overall, WHIM syndrome patients presented with an increased severity of periodontitis despite past and ongoing neutrophil mobilization treatments. GOF mutations in CXCR4 may be a risk factor for periodontitis in humans.


Asunto(s)
Síndromes de Inmunodeficiencia , Neutropenia , Enfermedades Periodontales , Periodontitis , Enfermedades de Inmunodeficiencia Primaria , Verrugas , Adulto , Humanos , Animales , Ratones , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/genética , Verrugas/genética , Verrugas/terapia , Neutropenia/complicaciones , Neutropenia/genética , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/genética , Periodontitis/complicaciones , Periodontitis/genética
11.
BMC Oral Health ; 24(1): 103, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233799

RESUMEN

Gene sequencing (GS) has numerous applications in combatting oral-cavity related disorders, including identifying genetic risk factors for diseases, developing targeted therapies, and improving diagnostic methods. It can help identify specific genetic mutations or variations that increase the risk of developing oral-cavity related disorders, such as oral cancer, periodontal disease, and cleft lip and palate. By the means of the following investigation, our primary objective was to assess the impact of GS technique in diagnosing and potentially treating diseases of the oral cavity by the means of a systematic review and meta-analysis. We commenced by defining the terms "gene sequencing," "oral cavity," and "disorders" as the important elements in our investigation's subject. Next, relevant databases like PubMed, Scopus, Embase, Web of Science, and Google Scholar were searched using keywords and synonyms for each concept, such as "genomic sequencing," "DNA sequencing," "oral health," "oral diseases," "dental caries," "periodontal disease," "oral cancer," and "salivary gland disorders." We combined several search terms, such as "gene sequencing AND oral disorders AND periodontal disease" or "oral cancer OR genomic sequencing," to further hone your search results using Boolean operators like "AND" and "OR." The oral cavity analysis obtained by CS in the selected articles revealed that most of the disorders were, in fact, a direct causal event influenced by the oral microbiome. Moreover, each sampled oral cavity evidenced a different microbial community, which predicted the precipitation of benign as well as malignant conditions, though not on a definitive basis. In the last ten years, genomic sequencing had advanced remarkably as majority of our selected studies observed, making it possible to diagnose and treat a variety of oral and maxillofacial disorders, including cancer. It was also used to ascertain a person's genetic make-up as well as to spot numerous genetic abnormalities that can predispose individuals to diseases. Understanding the different sequencing techniques and the resulting genetic anomalies may help with their clinical application and lead to an improvement in illness diagnosis and prognosis as a whole in the field of dentistry.


Asunto(s)
Labio Leporino , Fisura del Paladar , Caries Dental , Enfermedades de la Boca , Neoplasias de la Boca , Enfermedades Periodontales , Humanos , Enfermedades de la Boca/genética , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/genética , Enfermedades Periodontales/genética
13.
J Dent ; 141: 104804, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38122885

RESUMEN

OBJECTIVES: The present systematic review and meta-analysis aimed to assess the association between salivary protein polymorphisms and the risk of periodontal diseases (PD). DATA: The review incorporated cross-sectional, case-control, retrospective/prospective cohort, and randomized controlled trials assessing the influence of salivary protein polymorphisms on the risk of PD development were included in this review. SOURCES: A thorough literature search was conducted across electronic databases, namely PubMed, Scopus, Embase, and Web of Science, without any restrictions on publication language and year. STUDY SELECTION: A total of 168 studies were identified, of which 19 were eligible for inclusion. The risk of bias (RoB) assessment of the included studies was conducted at the methodological level. RESULTS: A total of 16 studies were included. Polymorphism in the gene encoding TNF-α was found to be protective against gingivitis, while those encoding IL-1α and IL-1ß were associated with developing gingivitis. Of the 42 proteins investigated, various gene polymorphisms were identified as protective or risk factors for periodontitis. Protective genes include CFH, DNMT1, OPRM1, and TLR9. Conversely, certain salivary protein genes (e.g., CRP, ERN1, FAM5C, IDH2, LTA, TET2, MPA, NLRP3, TLR4) were associated with periodontitis risk. Notably, IL6, MMP9, and MUC7 genes showed no association with PD, while MMP13 was linked to early implant loss. Overall, the meta-analysis found a statistically significant association between salivary proteins' polymorphisms and risk of PD. CONCLUSIONS: Salivary protein polymorphisms significantly influence PD, revealing protective and risk-associated genotypes. Despite limitations, findings suggest therapeutic targets, emphasizing the complex genetics-periodontal health interplay. CLINICAL SIGNIFICANCE: This study unveils salivary protein polymorphisms as pivotal factors in PD. Protective genes including CFH and TLR9, and risk-associated genes including CRP and TLR4, indicate a genetic basis for PD susceptibility.


Asunto(s)
Gingivitis , Enfermedades Periodontales , Periodontitis , Humanos , Estudios Retrospectivos , Estudios Transversales , Estudios Prospectivos , Receptor Toll-Like 4/genética , Receptor Toll-Like 9/genética , Enfermedades Periodontales/genética , Polimorfismo Genético/genética , Proteínas y Péptidos Salivales/genética
14.
Trop Biomed ; 40(3): 307-312, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37897163

RESUMEN

Trichomonas tenax, an oral flagellated protozoon found in humans, potentially associated with the inflammation of periodontal tissues and decreased immunity that causes the tissue damage and tooth loss from chronic infection. Currently, there is a lack of data regarding the prevalence of T. tenax infection in Thailand. Therefore, this study aimed to measure prevalence of T. tenax in periodontal disease patients by using polymerase chain reaction (PCR) to amplify the 18S ribosomal RNA (18S rRNA) gene and to determine the factors associated with the presence of this protozoan. A cross-sectional descriptive study was conducted among 230 patients with periodontal disease, who visited the oral health center of Suranaree University of Technology Hospital, Thailand from 2021 to 2022. Dental plaque specimens were collected and examined to identify the presence of T. tenax using the PCR-based 18S rRNA gene. The occurrence of factors associated with T. tenax infection was analyzed by the chi-square test and binary logistic regression. The prevalence of T. tenax infection was 13.48% (31/230), in patients, including 96.77% (30/31) and 3.23% (1/31) in periodontitis and gingivitis patients, respectively. The presence of T. tenax was associated with periodontal disease (p<0.001) and the Periodontal Screening and Record (PSR) index (p=0.001). The significant risk factors for T. tenax infection were periodontitis (ORadj=239.89, 95% CI=23.801-2417.746), no-underlying disease (ORadj=0.31, 95% CI=0.099-0.942), and male sex (ORadj=0.25, 95% CI=0.062-0.981). Dentists should be concerned about this oral protozoan in periodontitis patients. Furthermore, epidemiologic studies of T. tenax are still needed to investigate the mechanism of pathogenesis from T. tenax infection.


Asunto(s)
Enfermedades Periodontales , Periodontitis , Tricomoniasis , Trichomonas , Humanos , Masculino , Trichomonas/genética , ARN Ribosómico 18S/genética , Tricomoniasis/diagnóstico , Tricomoniasis/epidemiología , Tricomoniasis/genética , Estudios Transversales , Genes de ARNr , Enfermedades Periodontales/diagnóstico , Enfermedades Periodontales/epidemiología , Enfermedades Periodontales/genética , Periodontitis/epidemiología , Periodontitis/genética , Reacción en Cadena de la Polimerasa
15.
Int J Mol Sci ; 24(15)2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37569480

RESUMEN

miRNAs are major regulators of eukaryotic gene expression and host immunity, and play an important role in the inflammation-mediated pathways in periodontal disease (PD) pathogenesis. Expanding our previous observation with the global miRNA profiling using partial human mouth microbes, and lack of in vivo studies involving oral spirochete Treponema denticola-induced miRNAs, this study was designed to delineate the global miRNA expression kinetics during progression of periodontitis in mice infected with T. denticola by using NanoString nCounter® miRNA panels. All of the T. denticola-infected male and female mice at 8 and 16 weeks demonstrated bacterial colonization (100%) on the gingival surface, and an increase in alveolar bone resorption (p < 0.0001). A total of 70 miRNAs with at least 1.0-fold differential expression/regulation (DE) (26 upregulated and 44 downregulated) were identified. nCounter miRNA expression profiling identified 13 upregulated miRNAs (e.g., miR-133a, miR-378) and 25 downregulated miRNAs (e.g., miR-375, miR-34b-5p) in T. denticola-infected mouse mandibles during 8 weeks of infection, whereas 13 upregulated miRNAs (e.g., miR-486, miR-126-5p) and 19 downregulated miRNAs (miR-2135, miR-142-3p) were observed during 16 weeks of infection. One miRNA (miR-126-5p) showed significant difference between 8 and 16 weeks of infection. Interestingly, miR-126-5p has been presented as a potential biomarker in patients with periodontitis and coronary artery disease. Among the upregulated miRNAs, miR-486, miR-126-3p, miR-126-5p, miR-378a-3p, miR-22-3p, miR-151a-3p, miR-423-5p, and miR-221 were reported in human gingival plaques and saliva samples from periodontitis and with diabetes. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed various functional pathways of DE miRNAs, such as bacterial invasion of epithelial cells, Ras signaling, Fc gamma R-mediated phagocytosis, osteoclast differentiation, adherens signaling, and ubiquitin mediated proteolysis. This is the first study of DE miRNAs in mouse mandibles at different time-points of T. denticola infection; the combination of three specific miRNAs, miR-486, miR-126-3p, and miR-126-5p, may serve as an invasive biomarker of T. denticola in PD. These miRNAs may have a significant role in PD pathogenesis, and this research establishes a link between miRNA, periodontitis, and systemic diseases.


Asunto(s)
Enfermedades Transmisibles , MicroARNs , Enfermedades Periodontales , Periodontitis , Humanos , Masculino , Femenino , Animales , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Treponema denticola/genética , Spirochaetales/genética , Treponema/genética , Treponema/metabolismo , Cinética , Perfilación de la Expresión Génica , Periodontitis/genética , Enfermedades Periodontales/genética , Biomarcadores
16.
mSystems ; 8(4): e0119322, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37436062

RESUMEN

Periodontal disease is a chronic inflammatory disease in which the oral pathogen Porphyromonas gingivalis plays an important role. Porphyromonas gingivalis expresses virulence determinants in response to higher hemin concentrations, but the underlying regulatory processes remain unclear. Bacterial DNA methylation has the potential to fulfil this mechanistic role. We characterized the methylome of P. gingivalis, and compared its variation to transcriptome changes in response to hemin availability. Porphyromonas gingivalis W50 was grown in chemostat continuous culture with excess or limited hemin, prior to whole-methylome and transcriptome profiling using Nanopore and Illumina RNA-Seq. DNA methylation was quantified for Dam/Dcm motifs and all-context N6-methyladenine (6mA) and 5-methylcytosine (5mC). Of all 1,992 genes analyzed, 161 and 268 were respectively over- and under-expressed with excess hemin. Notably, we detected differential DNA methylation signatures for the Dam "GATC" motif and both all-context 6mA and 5mC in response to hemin availability. Joint analyses identified a subset of coordinated changes in gene expression, 6mA, and 5mC methylation that target genes involved in lactate utilization and ABC transporters. The results identify altered methylation and expression responses to hemin availability in P. gingivalis, with insights into mechanisms regulating its virulence in periodontal disease. IMPORTANCE DNA methylation has important roles in bacteria, including in the regulation of transcription. Porphyromonas gingivalis, an oral pathogen in periodontitis, exhibits well-established gene expression changes in response to hemin availability. However, the regulatory processes underlying these effects remain unknown. We profiled the novel P. gingivalis epigenome, and assessed epigenetic and transcriptome variation under limited and excess hemin conditions. As expected, multiple gene expression changes were detected in response to limited and excess hemin that reflect health and disease, respectively. Notably, we also detected differential DNA methylation signatures for the Dam "GATC" motif and both all-context 6mA and 5mC in response to hemin. Joint analyses identified coordinated changes in gene expression, 6mA, and 5mC methylation that target genes involved in lactate utilization and ABC transporters. The results identify novel regulatory processes underlying the mechanism of hemin regulated gene expression in P. gingivalis, with phenotypic impacts on its virulence in periodontal disease.


Asunto(s)
Hemina , Enfermedades Periodontales , Humanos , Hemina/farmacología , Porphyromonas gingivalis/genética , Metilación de ADN/genética , Enfermedades Periodontales/genética , Transportadoras de Casetes de Unión a ATP/genética , Expresión Génica
17.
J Clin Periodontol ; 50(9): 1140-1153, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37464577

RESUMEN

AIM: To investigate individual susceptibility to periodontitis by conducting an epigenome-wide association study using peripheral blood. MATERIALS AND METHODS: We included 1077 African American and 457 European American participants of the Atherosclerosis Risk in Communities (ARIC) study who had completed a dental examination or reported being edentulous at Visit 4 and had available data on DNA methylation from Visit 2 or 3. DNA methylation levels were compared by periodontal disease severity and edentulism through discovery analyses and subsequent testing of individual CpGs. RESULTS: Our discovery analysis replicated findings from a previous study reporting a region in gene ZFP57 (6p22.1) that was significantly hypomethylated in severe periodontal disease compared with no/mild periodontal disease in European American participants. Higher methylation levels in a separate region in an unknown gene (located in Chr10: 743,992-744,958) was associated with significantly higher odds of edentulism compared with no/mild periodontal disease in African American participants. In subsequent CpG testing, four CpGs in a region previously associated with periodontitis located within HOXA4 were significantly hypermethylated in severe periodontal disease compared with no/mild periodontal disease in African American participants (odds ratio per 1 SD increase in methylation level: cg11015251: 1.28 (1.02, 1.61); cg14359292: 1.24 (1.01, 1.54); cg07317062: 1.30 (1.05, 1.61); cg08657492: 1.25 (1.01, 1.55)). CONCLUSIONS: Our study highlights epigenetic variations in ZPF57 and HOXA4 that are significantly and reproducibly associated with periodontitis. Future studies should evaluate gene regulatory mechanisms in the candidate regions of these loci.


Asunto(s)
Aterosclerosis , Enfermedades Periodontales , Periodontitis , Humanos , Epigenoma , Estudio de Asociación del Genoma Completo , Enfermedades Periodontales/genética , Aterosclerosis/genética , Periodontitis/genética , Leucocitos , Genómica
18.
Genes (Basel) ; 14(6)2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37372382

RESUMEN

Periodontitis is a chronic inflammatory disease that affects the supporting structures of teeth. In the literature, the association between the pathogenicity of bacteria and environmental factors in this regard have been extensively examined. In the present study, we will shed light on the potential role that epigenetic change can play on different facets of its process, more particularly the modifications concerning the genes involved in inflammation, defense, and immune systems. Since the 1960s, the role of genetic variants in the onset and severity of periodontal disease has been widely demonstrated. These make some people more susceptible to developing it than others. It has been documented that the wide variation in its frequency for various racial and ethnic populations is due primarily to the complex interplay among genetic factors with those affecting the environment and the demography. In molecular biology, epigenetic modifications are defined as any change in the promoter for the CpG islands, in the structure of the histone protein, as well as post-translational regulation by microRNAs (miRNAs), being known to contribute to the alteration in gene expression for complex multifactorial diseases such as periodontitis. The key role of epigenetic modification is to understand the mechanism involved in the gene-environment interaction, and the development of periodontitis is now the subject of more and more studies that attempt to identify which factors are stimulating it, but also affect the reduced response to therapy.


Asunto(s)
Enfermedades Periodontales , Periodontitis , Humanos , Epigénesis Genética , Periodontitis/genética , Periodontitis/metabolismo , Histonas/genética , Enfermedades Periodontales/genética , Inflamación/genética
19.
Int J Mol Sci ; 24(9)2023 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-37175429

RESUMEN

Down syndrome patients show success rates in dental implants much lower than those observed in the general population. This retrospective case-control study aimed to identify possible genes that are related to the regulation of inflammatory responses and bone metabolism related to periimplantitis and implant loss, as well as genes related to bone quality. This process involved using the functional analysis of the gene expression software Transcriptome Analysis Console (TAC version 4.0 Applied BiosystemsTM, Thermo Fisher Scientific, Waltham, MA, USA) and a search for possible candidate genes involved. The focus was placed on the 93 genes related to periodontitis, periimplantitis, bone loss, implant loss, and genes related to bone quality and regulators underlying the establishment and maintenance of osseointegration. Five genes showed statistically significant results (p < 0.05) in our comparison. Four of them, IL1B (p = 0.023), IL1RN (p = 0.048), BGLAP (p = 0.0372) and PTK2 (p = 0.0075) were down-regulated in the periodontal disease and implant rejection group, and only one was overexpressed: FOXO1A (p = 0.0552). The genes with statistically significant alterations described in this article determine that the group of Down syndrome patients with periodontal disease and implant failure is a group of patients genetically susceptible to suffering from both conditions together.


Asunto(s)
Pérdida de Hueso Alveolar , Implantes Dentales , Síndrome de Down , Periimplantitis , Enfermedades Periodontales , Humanos , Estudios Retrospectivos , Estudios de Casos y Controles , Periimplantitis/metabolismo , Síndrome de Down/complicaciones , Síndrome de Down/genética , Enfermedades Periodontales/genética
20.
CRISPR J ; 6(2): 90-98, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36939849

RESUMEN

Periodontal disease (PD) is an immune-inflammatory disease affecting the supporting structures of the teeth, which results in progressive destruction of the hard and soft tissues surrounding teeth, ultimately resulting in tooth loss. The primary etiological factor for this disease is the presence of pathogenic microorganisms. Pathogenic bacteria face antagonistic conditions and foreign DNA components during the infection stage and depend on defense mechanisms such as clustered regularly interspaced short palindromic repeats (CRISPR)-Cas to counter them. Virulence genes regulated by the CRISPR-Cas system are often expressed by bacteria as part of the stress response to the presence of stress conditions and foreign elements. There is ever-growing evidence regarding the role of CRISPR-Cas in virulence of periodontal pathogens. The same CRISPR-Cas system may also be targeted to reduce bacterial virulence and it may also be utilized to develop diagnostic and therapeutic strategies for prevention and control of PD progression. This review article describes the CRISPR-Cas systems in the periodontal dysbiotic microbial communities, their role in the virulence of periodontal pathogens, and their potential role in understanding the pathogenesis of periodontitis and treatment of PD.


Asunto(s)
Sistemas CRISPR-Cas , Enfermedades Periodontales , Humanos , Sistemas CRISPR-Cas/genética , Edición Génica , Bacterias/genética , ADN , Enfermedades Periodontales/genética , Enfermedades Periodontales/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA