Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 228, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37803465

RESUMEN

INTRODUCTION: Periodontitis-related oral microbial dysbiosis is thought to contribute to Alzheimer's disease (AD) neuroinflammation and brain amyloid production. Since probiotics can modulate periodontitis/oral dysbiosis, this study examined the effects of a probiotic/lantibiotic, nisin, in modulating brain pathology triggered by periodontitis. METHODS: A polymicrobial mouse model of periodontal disease was used to evaluate the effects of this disease on brain microbiome dysbiosis, neuroinflammation, Alzheimer's-related changes, and nisin's therapeutic potential in this context. RESULTS: 16S sequencing and real-time PCR data revealed that Nisin treatment mitigated the changes in the brain microbiome composition, diversity, and community structure, and reduced the levels of periodontal pathogen DNA in the brain induced by periodontal disease. Nisin treatment significantly decreased the mRNA expression of pro-inflammatory cytokines (Interleukin-1ß/IL-1 ß, Interleukin 6/IL-6, and Tumor Necrosis Factor α/TNF-α) in the brain that were elevated by periodontal infection. In addition, the concentrations of amyloid-ß 42 (Aß42), total Tau, and Tau (pS199) (445.69 ± 120.03, 1420.85 ± 331.40, 137.20 ± 36.01) were significantly higher in the infection group compared to the control group (193.01 ± 31.82, 384.27 ± 363.93, 6.09 ± 10.85), respectively. Nisin treatment markedly reduced the Aß42 (261.80 ± 52.50), total Tau (865.37 ± 304.93), and phosphorylated Tau (82.53 ± 15.77) deposition in the brain of the infection group. DISCUSSION: Nisin abrogation of brain microbiome dysbiosis induces beneficial effects on AD-like pathogenic changes and neuroinflammation, and thereby may serve as a potential therapeutic for periodontal-dysbiosis-related AD.


Asunto(s)
Enfermedad de Alzheimer , Bacteriocinas , Microbiota , Nisina , Periodontitis , Probióticos , Ratones , Animales , Enfermedad de Alzheimer/patología , Nisina/metabolismo , Bacteriocinas/metabolismo , Enfermedades Neuroinflamatorias , Disbiosis/tratamiento farmacológico , Disbiosis/metabolismo , Periodontitis/metabolismo , Encéfalo/metabolismo , Péptidos beta-Amiloides/metabolismo , Interleucina-6/metabolismo , Probióticos/uso terapéutico
2.
PLoS Pathog ; 17(7): e1009311, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34255809

RESUMEN

Periodontal disease is driven by dysbiosis in the oral microbiome, resulting in over-representation of species that induce the release of pro-inflammatory cytokines, chemokines, and tissue-remodeling matrix metalloproteinases (MMPs) in the periodontium. These chronic tissue-destructive inflammatory responses result in gradual loss of tooth-supporting alveolar bone. The oral spirochete Treponema denticola, is consistently found at significantly elevated levels in periodontal lesions. Host-expressed Toll-Like Receptor 2 (TLR2) senses a variety of bacterial ligands, including acylated lipopolysaccharides and lipoproteins. T. denticola dentilisin, a surface-expressed protease complex comprised of three lipoproteins has been implicated as a virulence factor in periodontal disease, primarily due to its proteolytic activity. While the role of acylated bacterial components in induction of inflammation is well-studied, little attention has been given to the potential role of the acylated nature of dentilisin. The purpose of this study was to test the hypothesis that T. denticola dentilisin activates a TLR2-dependent mechanism, leading to upregulation of tissue-destructive genes in periodontal tissue. RNA-sequencing of periodontal ligament cells challenged with T. denticola bacteria revealed significant upregulation of genes associated with extracellular matrix organization and degradation including potentially tissue-specific inducible MMPs that may play novel roles in modulating host immune responses that have yet to be characterized within the context of oral disease. The Gram-negative oral commensal, Veillonella parvula, failed to upregulate these same MMPs. Dentilisin-induced upregulation of MMPs was mediated via TLR2 and MyD88 activation, since knockdown of expression of either abrogated these effects. Challenge with purified dentilisin upregulated the same MMPs while a dentilisin-deficient T. denticola mutant had no effect. Finally, T. denticola-mediated activation of TLR2/MyD88 lead to the nuclear translocation of the transcription factor Sp1, which was shown to be a critical regulator of all T. denticola-dependent MMP expression. Taken together, these data suggest that T. denticola dentilisin stimulates tissue-destructive cellular processes in a TLR2/MyD88/Sp1-dependent fashion.


Asunto(s)
Proteínas Bacterianas/metabolismo , Péptido Hidrolasas/metabolismo , Enfermedades Periodontales , Infecciones por Treponema/metabolismo , Factores de Virulencia/metabolismo , Células Cultivadas , Humanos , Metaloproteinasas de la Matriz/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Enfermedades Periodontales/metabolismo , Enfermedades Periodontales/microbiología , Enfermedades Periodontales/patología , Ligamento Periodontal , Factor de Transcripción Sp1/metabolismo , Receptor Toll-Like 2/metabolismo , Treponema denticola , Infecciones por Treponema/patología , Regulación hacia Arriba
3.
Periodontol 2000 ; 2023 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-38073011

RESUMEN

The oral squamous cell carcinoma (OSCC) 5 year survival rate of 41% has marginally improved in the last few years, with less than a 1% improvement per year from 2005 to 2017, with higher survival rates when detected at early stages. Based on histopathological grading of oral dysplasia, it is estimated that severe dysplasia has a malignant transformation rate of 7%-50%. Despite these numbers, oral dysplasia grading does not reliably predict its clinical behavior. Thus, more accurate markers predicting oral dysplasia progression to cancer would enable better targeting of these lesions for closer follow-up, especially in the early stages of the disease. In this context, molecular biomarkers derived from genetics, proteins, and metabolites play key roles in clinical oncology. These molecular signatures can help predict the likelihood of OSCC development and/or progression and have the potential to detect the disease at an early stage and, support treatment decision-making and predict treatment responsiveness. Also, identifying reliable biomarkers for OSCC detection that can be obtained non-invasively would enhance management of OSCC. This review will discuss biomarkers for OSCC that have emerged from different biological areas, including genomics, transcriptomics, proteomics, metabolomics, immunomics, and microbiomics.

4.
Periodontol 2000 ; 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37277934

RESUMEN

Three years into the coronavirus disease 2019 (COVID-19) pandemic, there are still growing concerns with the emergence of different variants, unknown long- and short-term effects of the virus, and potential biological mechanisms underlying etiopathogenesis and increased risk for morbidity and mortality. The role of the microbiome in human physiology and the initiation and progression of several oral and systemic diseases have been actively studied in the past decade. With the proof of viral transmission, carriage, and a potential role in etiopathogenesis, saliva and the oral environment have been a focus of COVID-19 research beyond diagnostic purposes. The oral environment hosts diverse microbial communities and contributes to human oral and systemic health. Several investigations have identified disruptions in the oral microbiome in COVID-19 patients. However, all these studies are cross-sectional in nature and present heterogeneity in study design, techniques, and analysis. Therefore, in this undertaking, we (a) systematically reviewed the current literature associating COVID-19 with changes in the microbiome; (b) performed a re-analysis of publicly available data as a means to standardize the analysis, and (c) reported alterations in the microbial characteristics in COVID-19 patients compared to negative controls. Overall, we identified that COVID-19 is associated with oral microbial dysbiosis with significant reduction in diversity. However, alterations in specific bacterial members differed across the study. Re-analysis from our pipeline shed light on Neisseria as the potential key microbial member associated with COVID-19.

5.
Oral Dis ; 29(3): 1333-1340, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34637557

RESUMEN

OBJECTIVE: To determine the in vitro antiviral activity of oral care products containing stabilized chlorine dioxide toward infectious viruses that harbor in the oral cavity. Specfically, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), SARS-CoV, human coronavirus (HCoV) 229E, influenza A (H3N2), rhinovirus type 14, adenovirus type 5, and herpes simplex virus (HSV) type 1 and 2 were examined. METHODS: Validated in vitro suspension virucidal assays were used. Test product was mixed with the test virus for 30, 60, or 120 s, neutralized with sodium thiosulfate, serially diluted in dilution medium in a 96-well plate and incubated in a carbon dioxide incubator for 7 days. The 50% Tissue Culture Infectious Dose per milliliter was determined. RESULTS: Two rinses, one oral spray and one fluoride toothpaste showed log reduction of severe acute respiratory syndrome coronavirus-2 ranging from 1.81 to 2.98 and of influenza A from 2.58 to 4.13, respectively, within 30 s of contact time; similar results were obtained at 60 s. Further, the Ultra Sensitive rinse showed 0.19, 0.75, 1.58, 1.75, 2.66, and 3.24 log reduction of severe acute respiratory syndrome coronavirus, human coronavirus 229E, rhinovirus type 14, adenovirus type 5, and herpes simplex virus type 1 and type 2, respectively, within 30 s of contact time. CONCLUSION: Stabilized chlorine dioxide containing CloSYS® oral care products reduced the viral load of multiple viruses within 30 s. The results warrant further investigation for potential in vivo applications.


Asunto(s)
COVID-19 , Gripe Humana , Humanos , SARS-CoV-2 , Antivirales/farmacología , Subtipo H3N2 del Virus de la Influenza A
6.
J Evid Based Dent Pract ; 23(4): 101916, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38035893

RESUMEN

ARTICLE TITLE AND BIBLIOGRAPHIC INFORMATION: Hu D, Zhong T, Dai Q. Clinical efficacy of probiotics as an adjunctive therapy to scaling and root planning in the management of periodontitis: a systematic review and meta-analysis of randomized controlled trails. J Evid Based Dent Pract. 2021;21(2):101547. doi:10.1016/j.jebdp.2021.101547. SOURCE OF FUNDING: Self-funded. TYPE OF STUDY/DESIGN: Systematic review with meta-analysis of data.

7.
PLoS Pathog ; 16(10): e1008881, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33002094

RESUMEN

Epidemiological studies reveal significant associations between periodontitis and oral cancer. However, knowledge about the contribution of periodontal pathogens to oral cancer and potential regulatory mechanisms involved is limited. Previously, we showed that nisin, a bacteriocin and commonly used food preservative, reduced oral cancer tumorigenesis and extended the life expectancy in tumor-bearing mice. In addition, nisin has antimicrobial effects on key periodontal pathogens. Thus, the purpose of this study was to test the hypothesis that key periodontal pathogens (Porphyromonas gingivalis, Treponema denticola, and Fusobacterium nucleatum) promote oral cancer via specific host-bacterial interactions, and that bacteriocin/nisin therapy may modulate these responses. All three periodontal pathogens enhanced oral squamous cell carcinoma (OSCC) cell migration, invasion, tumorsphere formation, and tumorigenesis in vivo, without significantly affecting cell proliferation or apoptosis. In contrast, oral commensal bacteria did not affect OSCC cell migration. Pathogen-enhanced OSCC cell migration was mediated via integrin alpha V and FAK activation, since stably blocking alpha V or FAK expression abrogated these effects. Nisin inhibited these pathogen-mediated processes. Further, Treponema denticola induced TLR2 and 4 and MyD88 expression. Stable suppression of MyD88 significantly inhibited Treponema denticola-induced FAK activation and abrogated pathogen-induced migration. Together, these data demonstrate that periodontal pathogens contribute to a highly aggressive cancer phenotype via crosstalk between TLR/MyD88 and integrin/FAK signaling. Nisin can modulate these pathogen-mediated effects, and thus has therapeutic potential as an antimicrobial and anti-tumorigenic agent.


Asunto(s)
Infecciones por Bacteroidaceae/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Periodontitis/tratamiento farmacológico , Porphyromonas gingivalis/efectos de los fármacos , Probióticos/farmacología , Animales , Apoptosis , Infecciones por Bacteroidaceae/metabolismo , Infecciones por Bacteroidaceae/microbiología , Infecciones por Bacteroidaceae/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/microbiología , Carcinoma de Células Escamosas/patología , Movimiento Celular , Proliferación Celular , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Integrinas/genética , Integrinas/metabolismo , Ratones , Ratones Desnudos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/microbiología , Neoplasias de la Boca/patología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Periodontitis/metabolismo , Periodontitis/microbiología , Periodontitis/patología , Porphyromonas gingivalis/patogenicidad , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Periodontal Res ; 57(1): 131-141, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34839547

RESUMEN

INTRODUCTION: The functional interplay between cementum of the root and alveolar bone of the socket is tuned by a uniquely positioned 70-80 µm wide fibrous and lubricious ligament in a dentoalveolar joint (DAJ). In this study, structural and biomechanical properties of the DAJ, periodontal ligament space (PDL-space also known as the joint space), alveolar bone of the socket, and cementum of the tooth root that govern the biomechanics of a lipopolysaccharide (LPS)-affected DAJ were mapped both in space and time. METHODS: The hemi-maxillae from 20 rats (4 control at 6 weeks of age, 4 control and 4 LPS-affected at 12 weeks of age, 4 control and 4 LPS-affected at 16 weeks of age) were investigated using a hybrid technique; micro-X-ray computed tomography (5 µm resolution) in combination with biomechanical testing in situ. Temporal variations in bone and cementum volume fractions were evaluated. Trends in mineral apposition rates (MAR) in additional six Sprague Dawley rats (3 controls, 3 LPS-affected) were revealed by transforming spatial fluorochrome signals to functional growth rates (linearity factor - RW) of bone, dentin, and cementum using a fast Fourier transform on fluorochrome signals from 100-µm hemi-maxillae sections. RESULTS: An overall change in LPS-affected DAJ biomechanics (a 2.5-4.5X increase in tooth displacement and 2X tooth rotation at 6 weeks, no increase in displacement and a 7X increase in rotation at 12 weeks; 27% increase in bone effective strain at 6 weeks and 11% at 12 weeks relative to control) was associated with structural changes in the coronal regions of the DAJ (15% increase in PDL-space from 0 to 6 weeks but only 5% from 6 to 12 weeks compared to control). A significant increase (p < 0.05) in PDL-space between ligated and age-matched control was observed. The bone fraction of ligated at 12 weeks was significantly lower than its age-matched control, and no significant differences (p > 0.05) between groups were observed at 6 weeks. Cementum in the apical regions grew faster but nonlinearly (11% and 20% increase in cementum fraction (CF) at 6 and 12 weeks) compared to control. Alveolar bone revealed site-specific nonlinear growth with an overall increase in MAR (108.5 µm/week to 126.7 µm/week after LPS treatment) compared to dentin (28.3 µm/week in control vs. 26.1 µm/week in LPS-affected) and cementum (126.5 µm/week in control vs. 119.9 µm/week in LPS-affected). A significant increase in CF (p < 0.05) in ligated specimens was observed at 6 weeks of age. CONCLUSIONS: Anatomy-specific responses of cementum and bone to the mechano-chemo stimuli, and their collective temporal contribution to observed changes in PDL-space were perpetuated by altered tooth movement. Data highlight the "resilience" of DAJ function through the predominance of nonlinear growth response of cementum, changes in PDL-space, and bone architecture. Despite the significant differences in bone and cementum architectures, data provided insights into the reactionary effects of cementum as a built-in compensatory mechanism to reestablish functional competence of the DAJ. The spatial shifts in architectures of alveolar bone and cementum, and consequently ligament space, highlight adaptations farther away from the site of insult, which also is another novel insight from this study. These adaptations when correlated within the context of joint function (biomechanics) illustrate that they are indeed necessary to sustain DAJ function albeit being pathological.


Asunto(s)
Cemento Dental , Lipopolisacáridos , Animales , Maxilar , Ligamento Periodontal/diagnóstico por imagen , Ratas , Ratas Sprague-Dawley
9.
Clin Oral Investig ; 26(3): 2465-2478, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34622310

RESUMEN

OBJECTIVES: The objective of this study was to examine the association between the oral microbiome and pregnancy outcomes, specifically healthy or preterm low birth weight (PLBW) in individuals with and without periodontal disease (PD). MATERIAL AND METHODS: In this prospective clinical trial, we recruited 186 pregnant women, 17 of whom exhibited PD and delivered PLBW infants (PD-PLBW group). Of the remaining women, 155 presented PD and delivered healthy infants; 18 of these subjects with similar periodontal condition and age matched to the PD-PLBW group, and they became the PD-HD group. From the total group, 11 women exhibited healthy gingiva and had a healthy delivery (HD) and healthy infants (H-HD group), and 3 exhibited healthy gingiva and delivered PLBW infants (H-PLBW group). Periodontal parameters were recorded, and subgingival plaque and serum were collected during 26-28 gestational weeks. For the plaque samples, microbial abundance and diversity were accessed by 16S rRNA sequencing. RESULTS: Women with PD showed an enrichment in the genus Porphyromonas, Treponema, and Filifactor, whereas women with healthy gingiva showed an enrichment in Streptococcus, Actinomyces, and Corynebacterium, independently of the birth status. Although no significant difference was found in the beta diversity between the 4 groups, women that had PLBW infants presented a significantly lower abundance of the genus Neisseria, independently of PD status. CONCLUSION: Lower levels of Neisseria align with preterm low birth weight in pregnant women, whereas a higher abundance of Treponema, Porphyromonas, Fretibacterium, and Filifactor and a lower abundance of Streptococcus may contribute to periodontal disease during pregnancy. CLINICAL RELEVANCE: The oral commensal Neisseria have potential in the prediction of PLBW.


Asunto(s)
Microbiota , Nacimiento Prematuro , Femenino , Humanos , Lactante , Recién Nacido de Bajo Peso , Recién Nacido , Neisseria , Embarazo , Resultado del Embarazo , ARN Ribosómico 16S
10.
Int J Mol Sci ; 23(16)2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-36012597

RESUMEN

The oral cavity is one of the environments on the human body with the highest concentrations of microorganisms that coexist harmoniously and maintain homeostasis related to oral health. Several local factors can shift the microbiome to a pathogenic state of dysbiosis. Existing treatments for infections caused by changes in the oral cavity aim to control biofilm dysbiosis and restore microbial balance. Studies have used probiotics as treatments for oral diseases, due to their ability to reduce the pathogenicity of the microbiota and immunoinflammatory changes. This review investigates the role of the probiotic Bifidobacterium animalis subsp. lactis (B. lactis) HN019 in oral health, and its mechanism of action in pre-clinical and clinical studies. This probiotic strain is a lactic acid bacterium that is safe for human consumption. It mediates bacterial co-aggregation with pathogens and modulates the immune response. Studies using B. lactis HN019 in periodontitis and peri-implant mucositis have shown it to be a potential adjuvant treatment with beneficial microbiological and immunological effects. Studies evaluating its oral effects and mechanism of action show that this probiotic strain has the potential to be used in several dental applications because of its benefit to the host.


Asunto(s)
Bifidobacterium animalis , Periodontitis , Probióticos , Bacterias , Biopelículas , Disbiosis/terapia , Humanos , Periodontitis/terapia , Probióticos/farmacología
11.
Periodontol 2000 ; 87(1): 11-16, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463994

RESUMEN

The landscape in dentistry is changing as emerging studies continue to reveal that periodontal health impacts systemic health, and vice versa. Population studies, clinical studies, and in vitro animal studies underscore the critical importance of oral health to systemic health. These inextricable relationships come to the forefront as oral diseases, such as periodontal disease, take root. Special populations bring to bear the multimodal relationships between oral and systemic health. Specifically, periodontal disease has been associated with diabetes, metabolic syndrome, obesity, eating disorders, liver disease, cardiovascular disease, Alzheimer disease, rheumatoid arthritis, adverse pregnancy outcomes, and cancer. Although bidirectional relationships are recognized, the potential for multiple comorbidities, relationships, and connections (multimodal relationships) also exists. Proposed mechanisms that mediate this connection between oral and systemic health include predisposing and precipitating factors, such as genetic factors (gene polymorphisms), environmental factors (stress, habits-such as smoking and high-fat diets/consumption of highly processed foods), medications, microbial dysbiosis and bacteremias/viremias/microbemias, and an altered host immune response. Thus, in a susceptible host, these predisposing and precipitating factors trigger the onset of periodontal disease and systemic disease/conditions. Further, high-throughput sequencing technologies are shedding light on the dark matter that comprises the oral microbiome. This has resulted in better characterization of the oral microbial dysbiosis, including putative bacterial periodontopathogens and shifts in oral virome composition during disease. Multiple laboratory and clinical studies have illustrated that both eukaryotic and prokaryotic viruses within subgingival plaque and periodontal tissues affect periodontal inflammation, putative periodontopathogens, and the host immune response. Although the association between herpesviruses and periodontitis and the degree to which these viruses directly aggravate periodontal tissue damage remain unclear, the benefits to periodontal health found from prolonged administration of antivirals in immunocompromised or immunodeficient individuals demonstrates that specific populations are possibly more susceptible to viral periodontopathogens. Thus, it may be important to further examine the implications of viral pathogen involvement in periodontitis and perhaps it is time to embrace the viral dark matter within the periodontal environment to fully comprehend the pathogenesis and systemic implications of periodontitis. Emerging data from the coronavirus disease 2019 pandemic further underscores the inextricable connection between oral and systemic health, with high levels of the severe acute respiratory syndrome coronavirus 2 angiotensin-converting enzyme 2 receptor noted on oral tissues (tongue) and an allostatic load or overload paradigm of chronic stress likely contributing to rapid breakdown of oral/dental, periodontal, and peri-implant tissues. These associations exist within a framework of viremias/bacteremias/microbemias, systemic inflammation, and/or disturbances of the immune system in a susceptible host. A thorough review of systemic and oral diseases and conditions and their mechanistic, predisposing, and precipitating factors are paramount to better addressing the oral and systemic health and needs of our patients.


Asunto(s)
COVID-19 , Enfermedades Periodontales , Animales , Disbiosis , Femenino , Humanos , Salud Bucal , Embarazo , SARS-CoV-2
12.
Periodontol 2000 ; 87(1): 276-281, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463984

RESUMEN

Because of hormonal and immunologic changes, there are significant changes in the oral microbiome that emerge during pregnancy. Recent evidence further suggests that there is an association between the presence of periodontal disease and a pregnancy-associated oral dysbiosis. Although this oral dysbiosis and pathogenic periodontal bacteria are considered to be associated with adverse pregnancy outcomes, it is still not clear how an oral dysbiosis during pregnancy can modulate oral diseases and birth outcomes. To develop preventive or therapeutic interventions, it is critical to understand the oral microbiome changes that emerge during pregnancy and their association with adverse pregnancy outcomes. In the present review, we summarize the current literature on normal changes in the oral microbiome that occur during pregnancy; the pathogenic changes in the oral microbiome believed to occur in association with adverse pregnancy outcomes; and the association between the placental microbiome and the oral microbiome.


Asunto(s)
Microbiota , Enfermedades Periodontales , Disbiosis , Femenino , Humanos , Placenta , Embarazo , Resultado del Embarazo
13.
Periodontol 2000 ; 87(1): 254-267, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463979

RESUMEN

Dysbiosis of the oral microbiome is associated with a variety of oral and systemic diseases, including periodontal disease. Oral dysbiosis in periodontal disease leads to an exacerbated host immune response that induces progressive periodontal tissue destruction and ultimately tooth loss. To counter the disease-associated dysbiosis of the oral cavity, strategies have been proposed to reestablish a "healthy" microbiome via the use of probiotics. This study reviews the literature on the use of probiotics for modifying the oral microbial composition toward a beneficial state that might alleviate disease progression. Four in vitro and 10 preclinical studies were included in the analysis, and these studies explored the effects of probiotics on cultured biofilm growth and bacterial gene expressions, as well as modulation of the host response to inflammation. The current molecular findings on probiotics provide fundamental evidence for further clinical research for the use of probiotics in periodontal therapy. They also point out an important caveat: Changing the biofilm composition might alter the normal oral flora that is beneficial and/or critical for oral health.


Asunto(s)
Microbiota , Enfermedades Periodontales , Probióticos , Disbiosis , Humanos , Enfermedades Periodontales/terapia , Probióticos/uso terapéutico
14.
Periodontol 2000 ; 87(1): 204-240, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463983

RESUMEN

Periodontal disease, a chronic inflammatory disease of the periodontal tissues, is not only a major cause of tooth loss, but it is also known to exacerbate/be associated with various metabolic disorders, such as obesity, diabetes, dyslipidemia, and cardiovascular disease. Recently, growing evidence has suggested that periodontal disease has adverse effects on the pathophysiology of liver disease. In particular, nonalcoholic fatty liver disease, a hepatic manifestation of metabolic syndrome, has been associated with periodontal disease. Nonalcoholic fatty liver disease is characterized by hepatic fat deposition in the absence of a habitual drinking history, viral infections, or autoimmune diseases. A subset of nonalcoholic fatty liver diseases can develop into more severe and progressive forms, namely nonalcoholic steatohepatitis. The latter can lead to cirrhosis and hepatocellular carcinoma, which are end-stage liver diseases. Extensive research has provided plausible mechanisms to explain how periodontal disease can negatively affect nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, namely via hematogenous or enteral routes. During periodontitis, the liver is under constant exposure to various pathogenic factors that diffuse systemically from the oral cavity, such as bacteria and their by-products, inflammatory cytokines, and reactive oxygen species, and these can be involved in disease promotion of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Also, gut microbiome dysbiosis induced by enteral translocation of periodontopathic bacteria may impair gut wall barrier function and promote the transfer of hepatotoxins and enterobacteria to the liver through the enterohepatic circulation. Moreover, in a population with metabolic syndrome, the interaction between periodontitis and systemic conditions related to insulin resistance further strengthens the association with nonalcoholic fatty liver disease. However, most of the pathologic links between periodontitis and nonalcoholic fatty liver disease in humans are provided by epidemiologic observational studies, with the causal relationship not yet being established. Several systematic and meta-analysis studies also show conflicting results. In addition, the effect of periodontal treatment on nonalcoholic fatty liver disease has hardly been studied. Despite these limitations, the global burden of periodontal disease combined with the recent nonalcoholic fatty liver disease epidemic has important clinical and public health implications. Emerging evidence suggests an association between periodontal disease and liver diseases, and thus we propose the term periodontal disease-related nonalcoholic fatty liver disease or periodontal disease-related nonalcoholic steatohepatitis. Continued efforts in this area will pave the way for new diagnostic and therapeutic approaches based on a periodontologic viewpoint to address this life-threatening liver disease.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Enfermedades Periodontales , Humanos , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedades Periodontales/complicaciones
15.
Periodontol 2000 ; 87(1): 282-298, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463988

RESUMEN

Mediators of the initiation, development, and recurrence of periodontitis include the oral microbiome embedded in subgingival plaque and the host immune response to a dysbiosis within this dynamic and complex microbial community. Although mediators have been studied extensively, researchers in the field have been unable to fully ascribe certain clinical presentations of periodontitis to their nature. Emergence of high-throughput sequencing technologies has resulted in better characterization of the microbial oral dysbiosis that extends beyond the extensively studied putative bacterial periodontopathogens to a shift in the oral virome composition during disease conditions. Although the biological dark matter inserted by retroviruses was once believed to be nonfunctional, research has revealed that it encodes historical viral-eukaryotic interactions and influences host development. The objective of this review is to evaluate the proposed association of herpesviruses to the etiology and pathogenesis of periodontal disease and survey the highly abundant prokaryotic viruses to delineate their potential roles in biofilm dynamics, as well as their interactions with putative bacterial periodontopathogens and eukaryotic cells. The findings suggest that potential novel periodontal therapies targeting or utilizing the oral virome can alleviate certain clinical presentations of periodontitis. Perhaps it is time to embrace the viral dark matter within the periodontal environment to fully comprehend the pathogenesis and systemic implications of periodontitis.


Asunto(s)
Microbiota , Periodontitis , Virus , Disbiosis , Humanos , Viroma
16.
Periodontol 2000 ; 87(1): 17-31, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463986

RESUMEN

People with eating disorders suffer from a mental disorder that negatively affects their physical and/or mental health. The three most frequent eating disorders are binge eating disorder, bulimia nervosa, and anorexia nervosa. Environmental and genetic factors are involved in the pathogenesis of eating disorders in vulnerable persons. Although treatment varies among different types of eating disorders, nutrition, medical care combined with psychotherapy and medications are standard of care. The aim of this review is to give an overview of the oral health impact of eating disorders with a special emphasis on the periodontium. Oral health professionals have a unique role to play in the early diagnosis of eating disorders because of the important impact that eating disorders have on the oral cavity. In vomiting-associated eating disorders, the risk of erosive tooth wear is mainly localized to the palatal surfaces of the incisors. Emerging evidence also indicates a high frequency of gingivitis and gingival recessions associated with compulsive toothbrushing. A holistic approach, including oral health and functional rehabilitation, should be promoted by physicians, psychiatrists, and dentists for people with eating disorders.


Asunto(s)
Anorexia Nerviosa , Bulimia Nerviosa , Trastornos de Alimentación y de la Ingestión de Alimentos , Gingivitis , Bulimia Nerviosa/complicaciones , Bulimia Nerviosa/terapia , Trastornos de Alimentación y de la Ingestión de Alimentos/complicaciones , Trastornos de Alimentación y de la Ingestión de Alimentos/terapia , Humanos , Salud Bucal
17.
Periodontol 2000 ; 87(1): 107-131, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463991

RESUMEN

States of oral health and disease reflect the compositional and functional capacities of, as well as the interspecies interactions within, the oral microbiota. The oral cavity exists as a highly dynamic microbial environment that harbors many distinct substrata and microenvironments that house diverse microbial communities. Specific to the oral cavity, the nonshedding dental surfaces facilitate the development of highly complex polymicrobial biofilm communities, characterized not only by the distinct microbes comprising them, but cumulatively by their activities. Adding to this complexity, the oral cavity faces near-constant environmental challenges, including those from host diet, salivary flow, masticatory forces, and introduction of exogenous microbes. The composition of the oral microbiome is shaped throughout life by factors including host genetics, maternal transmission, as well as environmental factors, such as dietary habits, oral hygiene practice, medications, and systemic factors. This dynamic ecosystem presents opportunities for oral microbial dysbiosis and the development of dental and periodontal diseases. The application of both in vitro and culture-independent approaches has broadened the mechanistic understandings of complex polymicrobial communities within the oral cavity, as well as the environmental, local, and systemic underpinnings that influence the dynamics of the oral microbiome. Here, we review the present knowledge and current understanding of microbial communities within the oral cavity and the influences and challenges upon this system that encourage homeostasis or provoke microbiome perturbation, and thus contribute to states of oral health or disease.


Asunto(s)
Microbiota , Enfermedades Periodontales , Disbiosis , Humanos , Boca , Salud Bucal
18.
Periodontol 2000 ; 87(1): 43-49, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463995

RESUMEN

Because the US population is living to an older age, the number of individuals with cognitive impairment and periodontitis is increasing, as both conditions/diseases increase with age. Dental informed consent best practices for dental/periodontal treatment of individuals with cognitive impairment have not been explored, yet warrant consideration, because complex dental treatments to address periodontal needs/edentulism raise challenges for informed consent in the elderly with cognitive impairment. The purpose of this review is to help practitioners better understand this topic and develop best practices in dentistry for informed consent of patients with cognitive impairment that need extensive dental treatment, including surgical and implant therapy.


Asunto(s)
Disfunción Cognitiva , Periodontitis , Anciano , Humanos , Consentimiento Informado
19.
Periodontol 2000 ; 87(1): 94-106, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463997

RESUMEN

Chronic stress is a relevant disease to periodontal practice, encompassing 25%-28% of the US population (American Psychological Association 2015). While it is well established that chronic psychologic stress can have significant deleterious systemic effects, only in recent decades have we begun to explore the biochemical, microbial, and physiologic impacts of chronic stress diseases on oral tissues. Currently, chronic stress is classified as a "risk indicator" for periodontal disease. However, as the evidence in this field matures with additional clinically controlled trials, more homogeneous data collection methods, and a better grasp of the biologic underpinnings of stress-mediated dysbiosis, emerging evidence suggests that chronic stress and related diseases (depression, anxiety) may be significant contributing factors in periodontal/peri-implant disease progression and inconsistent wound healing following periodontal-related therapeutics. Ideal solutions for these patients include classification of the disease process and de-escalation of chronic stress conditions through coping strategies. This paper also summarizes periodontal/implant-related therapeutic approaches to ensure predictable results for this specific patient subpopulation.


Asunto(s)
Implantes Dentales , Periimplantitis , Enfermedades Periodontales , Disbiosis , Humanos , Enfermedades Periodontales/terapia , Cicatrización de Heridas
20.
Periodontol 2000 ; 87(1): 157-165, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34463978

RESUMEN

Periodontal health in the elderly is influenced by numerous factors, including systemic conditions, patient compliance, age-associated changes, and restorative procedures. The numerous comorbidities seen in the elderly necessitate individualized approaches for treatment planning. In this paper, we review how age, comorbidities, oral hygiene, and restorative dental procedures collectively influence the treatment and management of the periodontium in the elderly. The elderly population is predicted to double in 30 years, which will have an economic impact the dental profession needs to plan for. Preventative and noninvasive treatment, supportive periodontal therapy, and patient-specific maintenance plans are imperative to maintaining oral health in the older population. Multiple coexisting changes, including xerostomia, altered wound healing, altered bone physiology, altered microbiome, and diminished plaque control, can add complexity to periodontal management. Considerations of the patient's general health, the selected periodontal treatment plan, and the selected completed restorative procedures need to be considered. The influence of caries, fixed prosthodontics, partial dentures, shortened dental arch, and implant therapy can have unintended impacts on periodontal health in the elderly. Adverse periodontal outcomes in the elderly can be minimized by carefully assessing the patient's medical history, impact of medications, functional needs, properly finishing and contouring restorations to avoid plaque accumulation, and designing restorations to allow access for hygiene. Partial dentures can be a source of plaque accumulation leading to periodontal disease, caries, and recession around abutment teeth. A shortened dental arch should be considered as a functional and cost-effective alternative to partial dentures. With dental implants, the patient's tissue phenotype, keratinized tissue quantity, risk of peri-implantitis, and patient access for maintaining adequate oral hygiene are all important to consider. Implant risk-assessment tools show promise by providing a systematic approach for early diagnosis to avoid future complications.


Asunto(s)
Implantes Dentales , Periimplantitis , Enfermedades Periodontales , Anciano , Humanos , Salud Bucal , Higiene Bucal , Enfermedades Periodontales/terapia , Periodoncio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA