Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Immunity ; 57(4): 859-875.e11, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38513665

RESUMO

At mucosal surfaces, epithelial cells provide a structural barrier and an immune defense system. However, dysregulated epithelial responses can contribute to disease states. Here, we demonstrated that epithelial cell-intrinsic production of interleukin-23 (IL-23) triggers an inflammatory loop in the prevalent oral disease periodontitis. Epithelial IL-23 expression localized to areas proximal to the disease-associated microbiome and was evident in experimental models and patients with common and genetic forms of disease. Mechanistically, flagellated microbial species of the periodontitis microbiome triggered epithelial IL-23 induction in a TLR5 receptor-dependent manner. Therefore, unlike other Th17-driven diseases, non-hematopoietic-cell-derived IL-23 served as an initiator of pathogenic inflammation in periodontitis. Beyond periodontitis, analysis of publicly available datasets revealed the expression of epithelial IL-23 in settings of infection, malignancy, and autoimmunity, suggesting a broader role for epithelial-intrinsic IL-23 in human disease. Collectively, this work highlights an important role for the barrier epithelium in the induction of IL-23-mediated inflammation.


Assuntos
Interleucina-23 , Periodontite , Humanos , Células Epiteliais , Inflamação , Receptor 5 Toll-Like/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(36): e2400528121, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39186644

RESUMO

Many chronic inflammatory diseases are attributed to disturbances in host-microbe interactions, which drive immune-mediated tissue damage. Depending on the anatomic setting, a chronic inflammatory disease can exert unique local and systemic influences, which provide an exceptional opportunity for understanding disease mechanism and testing therapeutic interventions. The oral cavity is an easily accessible environment that allows for protective interventions aiming at modulating the immune response to control disease processes driven by a breakdown of host-microbe homeostasis. Periodontal disease (PD) is a prevalent condition in which quantitative and qualitative changes of the oral microbiota (dysbiosis) trigger nonresolving chronic inflammation, progressive bone loss, and ultimately tooth loss. Here, we demonstrate the therapeutic benefit of local sustained delivery of the myeloid-recruiting chemokine (C-C motif) ligand 2 (CCL2) in murine ligature-induced PD using clinically relevant models as a preventive, interventional, or reparative therapy. Local delivery of CCL2 into the periodontium inhibited bone loss and accelerated bone gain that could be ascribed to reduced osteoclasts numbers. CCL2 treatment up-regulated M2-macrophage and downregulated proinflammatory and pro-osteoclastic markers. Furthermore, single-cell ribonucleic acid (RNA) sequencing indicated that CCL2 therapy reversed disease-associated transcriptomic profiles of murine gingival macrophages via inhibiting the triggering receptor expressed on myeloid cells-1 (TREM-1) signaling in classically activated macrophages and inducing protein kinase A (PKA) signaling in infiltrating macrophages. Finally, 16S ribosomal ribonucleic acid (rRNA) sequencing showed mitigation of microbial dysbiosis in the periodontium that correlated with a reduction in microbial load in CCL2-treated mice. This study reveals a novel protective effect of CCL2 local delivery in PD as a model for chronic inflammatory diseases caused by a disturbance in host-microbe homeostasis.


Assuntos
Quimiocina CCL2 , Homeostase , Animais , Camundongos , Quimiocina CCL2/metabolismo , Doenças Periodontais/microbiologia , Doenças Periodontais/imunologia , Doenças Periodontais/terapia , Disbiose/imunologia , Disbiose/microbiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Osteoclastos/metabolismo , Periodontite/microbiologia , Periodontite/imunologia
3.
Immunity ; 46(1): 133-147, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28087239

RESUMO

Immuno-surveillance networks operating at barrier sites are tuned by local tissue cues to ensure effective immunity. Site-specific commensal bacteria provide key signals ensuring host defense in the skin and gut. However, how the oral microbiome and tissue-specific signals balance immunity and regulation at the gingiva, a key oral barrier, remains minimally explored. In contrast to the skin and gut, we demonstrate that gingiva-resident T helper 17 (Th17) cells developed via a commensal colonization-independent mechanism. Accumulation of Th17 cells at the gingiva was driven in response to the physiological barrier damage that occurs during mastication. Physiological mechanical damage, via induction of interleukin 6 (IL-6) from epithelial cells, tailored effector T cell function, promoting increases in gingival Th17 cell numbers. These data highlight that diverse tissue-specific mechanisms govern education of Th17 cell responses and demonstrate that mechanical damage helps define the immune tone of this important oral barrier.


Assuntos
Gengiva/imunologia , Imunidade nas Mucosas/imunologia , Vigilância Imunológica/imunologia , Mucosa Bucal/imunologia , Células Th17/imunologia , Animais , Citometria de Fluxo , Gengiva/microbiologia , Humanos , Mastigação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota , Mucosa Bucal/microbiologia , Reação em Cadeia da Polimerase em Tempo Real
4.
Periodontol 2000 ; 85(1): 82-89, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33226731

RESUMO

The microbial communities that inhabit the gingival crevice are responsible for the pathological processes that affect the periodontium. The changes in composition and function of subgingival bacteria as disease develops have been extensively studied. Subgingival communities, however, also contain fungi, Archaea, and viruses, which could contribute to the dysbiotic processes associated with periodontal diseases. High-throughput DNA sequencing has facilitated a better understanding of the mycobiome, archaeome, and virome. However, the number of studies available on the nonbacterial components of the subgingival microbiome remains limited in comparison with publications focusing on bacteria. Difficulties in characterizing fungal, archaeal, and viral populations arise from the small portion of the total metagenome mass they occupy and lack of comprehensive reference genome databases. In addition, specialized approaches potentially introducing bias are required to enrich for viral particles, while harsh methods of cell lysis are needed to recover nuclei acids from certain fungi. While the characterization of the subgingival diversity of fungi, Archaea and viruses is incomplete, emerging evidence suggests that they could contribute in different ways to subgingival dysbiosis. Certain fungi, such as Candida albicans are suggested to facilitate colonization of bacterial pathogens. Methanogenic Archaea are associated with periodontitis severity and are thought to partner synergistically with bacterial fermenters, while viruses may affect immune responses or shape microbial communities in ways incompletely understood. This review describes the manner in which omics approaches have improved our understanding of the diversity of fungi, Archaea, and viruses within subgingival communities. Further characterization of these understudied components of the subgingival microbiome is required, together with mechanistic studies to unravel their ecological role and potential contributions to dysbiosis.


Assuntos
Gengiva , Microbiota , Vírus , Archaea/genética , Bactérias/genética , DNA , Fungos , Gengiva/microbiologia , Humanos
5.
Periodontol 2000 ; 86(1): 57-78, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33690899

RESUMO

The subgingival crevice harbors diverse microbial communities. Shifts in the composition of these communities occur with the development of gingivitis and periodontitis, which are considered as successive stages of periodontal health deterioration. It is not clear, however, to what extent health- and gingivitis-associated microbiota are protective, or whether these communities facilitate the successive growth of periodontitis-associated taxa. To further our understanding of the dynamics of the microbial stimuli that trigger disruptions in periodontal homeostasis, we reviewed the available literature with the aim of defining specific microbial signatures associated with different stages of periodontal dysbiosis. Although several studies have evaluated the subgingival communities present in different periodontal conditions, we found limited evidence for the direct comparison of communities in health, gingivitis, and periodontitis. Therefore, we aimed to better define subgingival microbiome shifts by merging and reanalyzing, using unified bioinformatic processing strategies, publicly available 16S ribosomal RNA gene amplicon datasets of periodontal health, gingivitis, and periodontitis. Despite inherent methodological differences across studies, distinct community structures were found for health, gingivitis, and periodontitis, demonstrating the specific associations between gingival tissue status and the subgingival microbiome. Consistent with the concept that periodontal dysbiosis is the result of a process of microbial succession without replacement, more species were detected in disease than in health. However, gingivitis-associated communities were more diverse than those from subjects with periodontitis, suggesting that certain species ultimately become dominant as dysbiosis progresses. We identified the bacterial species associated with each periodontal condition and prevalent species that do not change in abundance from one state to another (core species), and we also outlined species co-occurrence patterns via network analysis. Most periodontitis-associated species were rarely detected in health but were frequently detected, albeit in low abundance, in gingivitis, which suggests that gingivitis and periodontitis are a continuum. Overall, we provide a framework of subgingival microbiome shifts, which can be used to generate hypotheses with respect to community assembly processes and the emergence of periodontal dysbiosis.


Assuntos
Gengivite , Microbiota , Periodontite , Disbiose , Humanos , RNA Ribossômico 16S/genética
6.
Periodontol 2000 ; 83(1): 14-25, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32385883

RESUMO

The last decade has witnessed unparalleled advances in our understanding of the complexity of the oral microbiome and the compositional changes that occur in subgingival biofilms in the transition from health to gingivitis and to destructive periodontal disease. The traditional view, which has held sway for the last 2 decades, that disease is characterized by the outgrowth of a consortium, or consortia, of a limited number of potentially pathogenic organisms, has given way to an alternative paradigm. In this new view, the microbiological changes associated with disease represent whole-scale alterations to the overall microbial population structure and to the functional properties of the entire community. Thus, and in common with other microbially mediated diseases of the gastrointestinal tract, the normally balanced, symbiotic, and generally benign commensal microbiome of the tooth-associated biofilm undergoes dysbiosis to a potentially deleterious microbiota. Coincident with progress in defining the microbiology of these diseases, there have been equally important advances in our understanding of the inflammatory systems of the periodontal tissues, their control, and how inflammation may contribute both to the development of dysbiosis and, in a deregulated state, the destructive disease process. One can therefore speculate that the inflammatory response and the periodontal microbiome are in a bidirectional balance in oral health and a bidirectional imbalance in periodontitis. However, despite these clear insights into both sides of the host/microbe balance in periodontal disease, there remain several unresolved issues concerning the role of the microbiota in disease. These include, but are not limited to, the factors which determine progression from gingivitis to periodontitis in a proportion of the population, whether dysbiosis causes disease or results from disease, and the molecular details of the microbial stimulus responsible for driving the destructive inflammatory response. Further progress in resolving these issues may provide significant benefit to diagnosis, treatment, and prevention.


Assuntos
Microbiota , Doenças Periodontais , Periodontite , Disbiose , Humanos , Periodonto
7.
J Calif Dent Assoc ; 44(7): 421-35, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27514154

RESUMO

High-throughput 16S rRNA gene sequencing has allowed the characterization of subgingival microbiome shifts from health to periodontitis identifying health-associated, periodontitis-associated and core species, which preserve their proportions from health to disease. The development of gingivitis is also characterized by distinct shifts. Microbiome shifts resemble microbial successions and result from interspecies interactions and community adaptation to the changing environment as inflammation ensues. Gingivitis-associated and core species are proposed as likely mediators of microbiome transitions.


Assuntos
Gengiva/microbiologia , Microbiota/fisiologia , Doenças Periodontais/microbiologia , Genes Bacterianos/genética , Gengivite/microbiologia , Humanos , Interações Microbianas/fisiologia , RNA Bacteriano/análise , RNA Ribossômico 16S/análise , Transcriptoma/genética
8.
BMC Nephrol ; 16: 80, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26055269

RESUMO

BACKGROUND: Evidence supports high prevalence of periodontitis in patients with chronic kidney disease. Several renal factors have been proposed as possible modifiers of periodontitis pathogenesis in this population. In this cross sectional study, we investigated whether distinct microbial profiles in renal patients could explain high periodontitis prevalence. METHODS: We characterized the subgingival microbiome in 14 End Stage Renal Disease (ESRD) and 13 control individuals with chronic periodontitis with similar demographic and clinical parameters. Medical, demographic and periodontal parameters were recorded. Subgingival biofilm samples were collected from the deepest pocket in two different quadrants and characterized via 454-pyrosequencing of the 16S rRNA gene. RESULTS: We found 874 species-level operational taxonomic units (OTU) across samples. Renal and control groups did not differ in the individual proportions of periodontitis-associated taxa. However, in principal coordinate plots of distance among samples based on OTU prevalence, some renal patients clustered apart from controls, with the microbial communities of these outlier subjects showing less diversity. Univariate correlation analysis showed a significant negative correlation between dialysis vintage and community diversity. CONCLUSIONS: Within the study limitations, dialysis vintage was associated with a less diverse periodontal microbial community in ESRD suggesting the need for further research.


Assuntos
Periodontite Crônica/microbiologia , Disbiose/microbiologia , Falência Renal Crônica/microbiologia , Microbiota/genética , Periodonto/microbiologia , RNA Bacteriano/análise , RNA Ribossômico 16S/análise , Adulto , Idoso , Estudos de Casos e Controles , Periodontite Crônica/complicações , Estudos Transversais , Disbiose/complicações , Feminino , Humanos , Falência Renal Crônica/complicações , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Diálise Renal , Análise de Sequência de RNA , Fatores de Tempo
9.
J Periodontol ; 95(7): 650-661, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38476115

RESUMO

BACKGROUND: The aim of the present study was to evaluate the subgingival microbiome in patients with grade C molar-incisor pattern periodontitis (C-MIP) affecting the primary or permanent dentitions. METHODS: DNA was isolated from subgingival biofilm samples from diseased and healthy sites from 45 C-MIP patients and subjected to phylogenetic microarray analysis. C-MIP sites were compared between children affected in the primary to those affected in the permanent dentitions. Within-subject differences between C-MIP-affected sites and dentition-matched healthy sites were also evaluated. RESULTS: C-MIP sites of subjects affected in the primary dentition showed partially overlapping but distinct microbial communities from C-MIP permanent dentition sites (p < 0.05). Differences were due to increased levels in primary C-MIP sites of certain species of the genera Capnocytophaga and Leptotrichia, while C-MIP permanent dentition sites showed higher prevalence of Filifactor alocis. Aggregatibacter actinomycetemcomitans (Aa) was among species seen in high prevalence and levels in both primary and permanent C-MIP sites. Moreover, both permanent and primary C-MIP sites showed distinct microbial communities when compared to dentition-matched healthy sites in the same subject (p < 0.01). CONCLUSIONS: Primary and permanent teeth with C-MIP showed a dysbiotic microbiome, with children affected in the primary dentition showing a distinct profile from those affected in the permanent dentition. However, Aa was enriched in both primary and permanent diseased sites, confirming that this microorganism is implicated in C-MIP in both dentitions.


Assuntos
Aggregatibacter actinomycetemcomitans , Biofilmes , Dentição Permanente , Microbiota , Periodontite , Dente Decíduo , Humanos , Dente Decíduo/microbiologia , Masculino , Feminino , Criança , Periodontite/microbiologia , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Adolescente , Capnocytophaga/isolamento & purificação , Leptotrichia , Gengiva/microbiologia , Estudos de Casos e Controles , DNA Bacteriano/análise , Pré-Escolar
10.
Sci Rep ; 14(1): 6222, 2024 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-38485747

RESUMO

Porphyromonas gingivalis, a Gram-negative anaerobic bacterium commonly found in human subgingival plaque, is a major etiologic agent for periodontitis and has been associated with multiple systemic pathologies. Many P. gingivalis strains have been identified and different strains possess different virulence factors. Current oral microbiome approaches (16S or shotgun) have been unable to differentiate P. gingivalis strains. This study presents a new approach that aims to improve the accuracy of strain identification, using a detection method based on sequencing of the intergenic spacer region (ISR) which is variable between P. gingivalis strains. Our approach uses two-step PCR to amplify only the P. gingivalis ISR region. Samples are then sequenced with an Illumina sequencer and mapped to specific strains. Our approach was validated by examining subgingival plaque from 153 participants with and without periodontal disease. We identified the avirulent strain ATCC33277/381 as the most abundant strain across all sample types. The W83/W50 strain was significantly enriched in periodontitis, with 13% of participants harboring that strain. Overall, this approach can have significant implications not only for the diagnosis and treatment of periodontal disease but also for other diseases where P. gingivalis or its toxins have been implicated, such as Alzheimer's disease.


Assuntos
Periodontite , Porphyromonas gingivalis , Humanos , Porphyromonas gingivalis/genética , Composição de Bases , Análise de Sequência de DNA , RNA Ribossômico 16S/genética , Filogenia , Periodontite/microbiologia
11.
Cell Rep ; 42(2): 112120, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36774550

RESUMO

Intestinal colonization of the oral bacterium Haemophilus parainfluenzae has been associated with Crohn's disease (CD) severity and progression. This study examines the role of periodontal disease (PD) as a modifier for colonization of H. parainfluenzae in patients with CD and explores the mechanisms behind H. parainfluenzae-mediated intestinal inflammation. Fifty subjects with and without CD were evaluated for the presence of PD, and their oral and fecal microbiomes were characterized. PD is associated with increased levels of H. parainfluenzae strains in subjects with CD. Oral inoculation of H. parainfluenzae elicits strain-dependent intestinal inflammation in murine models of inflammatory bowel disease, which is associated with increased intestinal interferon-γ (IFN-γ)+ CD4+ T cells and disruption of the host hypusination pathway. In summary, this study establishes a strain-specific pathogenic role of H. parainfluenzae in intestinal inflammation and highlights the potential effect of PD on intestinal colonization by pathogenic H. parainfluenzae strains in patients with CD.


Assuntos
Doença de Crohn , Doenças Periodontais , Humanos , Animais , Camundongos , Haemophilus parainfluenzae , Doença de Crohn/complicações , Doença de Crohn/metabolismo , Inflamação
12.
J Exp Med ; 220(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37261457

RESUMO

Neutrophil infiltration is a hallmark of periodontitis, a prevalent oral inflammatory condition in which Th17-driven mucosal inflammation leads to destruction of tooth-supporting bone. Herein, we document that neutrophil extracellular traps (NETs) are early triggers of pathogenic inflammation in periodontitis. In an established animal model, we demonstrate that neutrophils infiltrate the gingival oral mucosa at early time points after disease induction and expel NETs to trigger mucosal inflammation and bone destruction in vivo. Investigating mechanisms by which NETs drive inflammatory bone loss, we find that extracellular histones, a major component of NETs, trigger upregulation of IL-17/Th17 responses, and bone destruction. Importantly, human findings corroborate our experimental work. We document significantly increased levels of NET complexes and extracellular histones bearing classic NET-associated posttranslational modifications, in blood and local lesions of severe periodontitis patients, in the absence of confounding disease. Our findings suggest a feed-forward loop in which NETs trigger IL-17 immunity to promote immunopathology in a prevalent human inflammatory disease.


Assuntos
Armadilhas Extracelulares , Periodontite , Animais , Humanos , Histonas , Interleucina-17 , Inflamação/patologia , Periodontite/patologia , Neutrófilos/patologia
13.
Infect Immun ; 80(2): 620-32, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22104105

RESUMO

Candida albicans is a commensal colonizer of the gastrointestinal tract of humans, where it coexists with highly diverse bacterial communities. It is not clear whether this interaction limits or promotes the potential of C. albicans to become an opportunistic pathogen. Here we investigate the interaction between C. albicans and three species of streptococci from the viridans group, which are ubiquitous and abundant oral commensal bacteria. The ability of C. albicans to form biofilms with Streptococcus oralis, Streptococcus sanguinis, or Streptococcus gordonii was investigated using flow cell devices that allow abiotic biofilm formation under salivary flow. In addition, we designed a novel flow cell system that allows mucosal biofilm formation under conditions that mimic the environment in the oral and esophageal mucosae. It was observed that C. albicans and streptococci formed a synergistic partnership where C. albicans promoted the ability of streptococci to form biofilms on abiotic surfaces or on the surface of an oral mucosa analogue. The increased ability of streptococci to form biofilms in the presence of C. albicans could not be explained by a growth-stimulatory effect since the streptococci were unaffected in their growth in planktonic coculture with C. albicans. Conversely, the presence of streptococci increased the ability of C. albicans to invade organotypic models of the oral and esophageal mucosae under conditions of salivary flow. Moreover, characterization of mucosal invasion by the biofilm microorganisms suggested that the esophageal mucosa is more permissive to invasion than the oral mucosa. In summary, C. albicans and commensal oral streptococci display a synergistic interaction with implications for the pathogenic potential of C. albicans in the upper gastrointestinal tract.


Assuntos
Candida albicans/fisiologia , Estreptococos Viridans/fisiologia , Técnicas Bacteriológicas , Biofilmes , Técnicas de Cocultura , Esôfago , Humanos , Modelos Biológicos , Mucosa Bucal/microbiologia , Mucosa Bucal/fisiologia , Saliva , Especificidade da Espécie , Estreptococos Viridans/classificação
14.
Anaerobe ; 18(4): 459-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22771792

RESUMO

Periodontitis results from an ecological shift in the composition of subgingival biofilms. Subgingival community maturation is modulated by inter-organismal interactions and the relationship of communities with the host. In an effort to better understand this process, we evaluated biofilm formation, with oral commensal species, by three strains of the subgingivally prevalent microorganism Fusobacterium nucleatum and four strains of the periodontopathogen Porphyromonas gingivalis. We also tested the effect of serum, which resembles gingival exudates, on subgingival biofilms. Biofilms were allowed to develop in flow cells using salivary medium. We found that although not all strains of F. nucleatum were able to grow in mono-species biofilms, forming a community with health-associated partners Actinomyces oris and Veillonella parvula promoted biofilm growth of all F. nucleatum strains. Strains of P. gingivalis also showed variable ability to form mono-species biofilms. P. gingivalis W50 and W83 did not form biofilms, while ATCC 33277 and 381 formed biofilm structures, but only strain ATCC 33277 grew over time. Unlike the enhanced growth of F. nucleatum with the two health-associated species, no strain of P. gingivalis grew in three-species communities with A. oris and V. parvula. However, addition of F. nucleatum facilitated growth of P. gingivalis ATCC 33277 with health-associated partners. Importantly, serum negatively affected the adhesion of F. nucleatum, while it favored biofilm growth by P. gingivalis. This work highlights strain specificity in subgingival biofilm formation. Environmental factors such as serum alter the colonization patterns of oral microorganisms and could impact subgingival biofilms by selectively promoting pathogenic species.


Assuntos
Biofilmes/efeitos dos fármacos , Fusobacterium nucleatum/crescimento & desenvolvimento , Porphyromonas gingivalis/crescimento & desenvolvimento , Soro/metabolismo , Actinomyces/crescimento & desenvolvimento , Aderência Bacteriana , Técnicas Bacteriológicas , Biota , Meios de Cultura/metabolismo , Humanos , Interações Microbianas , Viabilidade Microbiana , Saliva/metabolismo , Especificidade da Espécie
15.
J Am Dent Assoc ; 153(10): 979-988, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36038399

RESUMO

BACKGROUND: In the absence of a full spectrum of evidence-based guidelines for the appropriate use of antimicrobial agents, dentists, including periodontists, remain a highly frequent antibiotic prescribing group. With the goal of understanding antibiotic prescribing practices, the authors surveyed a convenience sample of dental practitioners and periodontists to identify differences between the 2 cohorts and assess the factors that affect prescribing practices. METHODS: An institutional review board-approved 15-item survey was developed to capture antibiotic prescribing practices addressing the main research question, factors affecting systemic antibiotic prescription patterns, and prescription timing. The authors collaborated with the American Dental Association (ADA) and the American Academy of Periodontology (AAP) for survey dissemination. Responses were summarized using descriptive statistics. Multivariable models were developed to identify antibiotic prescription predictors. RESULTS: Overall, 32.4% of the participants prescribed systemic antibiotics with scaling and root planing. When comparing the 2 groups, the authors found that 46.4% and 18.7% of the AAP and ADA members, respectively, prescribed systemic antibiotics with scaling and root planing (P = .0001). The authors found a significant difference between the AAP and ADA groups in prescription timing (P = .01). The multivariable model revealed that practitioner sex (P = .03), AAP membership (P = .0001), and years of practitioner experience (P = .04) predicted antibiotic prescription practices. The geographic location, practice setting, and occupation type did not predict antibiotic prescription patterns. CONCLUSION: The authors found a lack of clarity related to prescription timing, factors determining prescription patterns, and selection of patient population who would benefit more from antibiotics. PRACTICAL IMPLICATIONS: This study confirmed a lack of clarity related to antibiotic prescription patterns in combination with nonsurgical periodontal treatment.


Assuntos
Antibacterianos , Odontólogos , Antibacterianos/uso terapêutico , Assistência Odontológica , Humanos , Padrões de Prática Odontológica , Prescrições , Papel Profissional
16.
J Periodontol ; 93(11): 1635-1648, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35533310

RESUMO

BACKGROUND: This study investigated the association between menopausal hormone therapy (HT) use and the subgingival microbiome, for which published information is limited. METHODS: This cross-sectional study included 1270 postmenopausal women, aged 53-81 years, who completed clinical examinations. Detailed information on HT use (type, delivery mode, duration) was obtained from questionnaires. HT use was categorized into three groups (never, former, current). 16S rRNA sequencing was performed on subgingival plaque samples obtained during dental examinations. Operational taxonomic units were centered log2-ratio (CLR) transformed to account for the compositional data structure. Analysis of variance was used to compare mean microbial relative abundances across HT categories with Benjamini-Hochberg correction. RESULTS: Significantly higher alpha diversity (Shannon Index) and beta diversity (Aitchison distance) was observed in never compared with current HT users (p < 0.05, each). Of the total 245 microbial taxa identified, 18 taxa differed significantly among the three HT groups, 11 of which were higher in current users and seven of which were lower in current users as compared with never users (p < 0.05, each). Differences in relative abundance between never and current HT users were materially unchanged after adjustment for age, body mass index, and oral hygiene. CONCLUSIONS: Relative abundance of several subgingival bacteria differed significantly between never and current HT users in a cohort of postmenopausal women. Additional studies are needed to determine the extent that these relationships might account for the previously reported inverse association between HT use and periodontal disease in older women.


Assuntos
Terapia de Reposição de Estrogênios , Menopausa , Microbiota , Feminino , Humanos , Bactérias , Estudos Transversais , RNA Ribossômico 16S/genética
17.
ISME J ; 15(5): 1490-1504, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33372193

RESUMO

Recent studies describe in detail the shifts in composition of human-associated polymicrobial communities from health to disease. However, the specific processes that drive the colonization and overgrowth of pathogens within these communities remain incompletely understood. We used in vitro culture systems and a disease-relevant mouse model to show that population size, which determines the availability of an endogenous diffusible small molecule, limits the growth, colonization, and in vivo virulence of the human oral pathogen Porphyromonas gingivalis. This bacterial pathogen overcomes the requirement for an endogenous cue by utilizing a cell-density dependent, growth-promoting, soluble molecule provided by the symbiotic early colonizer Veillonella parvula, but not produced by other commensals tested. Our work shows that exchange of cell-density-dependent diffusible cues between specific early and late colonizing species in a polymicrobial community drives microbial successions, pathogen colonization and disease development, representing a target process for manipulation of the microbiome towards the healthy state.


Assuntos
Biofilmes , Veillonella , Animais , Camundongos , Porphyromonas gingivalis , Virulência
18.
Curr Oral Health Rep ; 7(1): 12-21, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33344104

RESUMO

PURPOSE OF REVIEW: This review summarizes mechanisms by which Porphyromonas gingivalis interacts with community members and the host so that it can persist in the periodontium under inflammatory conditions that drive periodontal disease. RECENT FINDINGS: Recent advances indicate that, in great part, the pathogenicity of P. gingivalis is dependent upon its ability to establish residence in the subgingival environment and to subvert innate immunity in a manner that uncouples the nutritionally favorable (for the bacteria) inflammatory response from antimicrobial pathways. While the initial establishment of P. gingivalis is dependent upon interactions with early colonizing bacteria, the immune subversion strategies of P. gingivalis in turn benefit co-habiting species. SUMMARY: Specific interspecies interactions and subversion of the host response contribute to the emergence and persistence of dysbiotic communities and are thus targets of therapeutic approaches for the treatment of periodontitis.

19.
Front Cell Infect Microbiol ; 10: 588155, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117738

RESUMO

Efforts to map gingival tissue proteomes and microbiomes have been hampered by lack of sufficient tissue extraction methods. The pressure cycling technology (PCT) is an emerging platform for reproducible tissue homogenisation and improved sequence retrieval coverage. Therefore, we employed PCT to characterise the proteome and microbiome profiles in healthy and diseased gingival tissue. Healthy and diseased contralateral gingival tissue samples (total n = 10) were collected from five systemically healthy individuals (51.6 ± 4.3 years) with generalised chronic periodontitis. The tissues were then lysed and digested using a Barocycler, proteins were prepared and submitted for mass spectrometric analysis and microbiome DNA for 16S rRNA profiling analysis. Overall, 1,366 human proteins were quantified (false discovery rate 0.22%), of which 69 proteins were differentially expressed (≥2 peptides and p < 0.05, 62 up, 7 down) in periodontally diseased sites, compared to healthy sites. These were primarily extracellular or vesicle-associated proteins, with functions in molecular transport. On the microbiome level, 362 species-level operational taxonomic units were identified. Of those, 14 predominant species accounted for >80% of the total relative abundance, whereas 11 proved to be significantly different between healthy and diseased sites. Among them, Treponema sp. HMT253 and Fusobacterium naviforme and were associated with disease sites and strongly interacted (r > 0.7) with 30 and 6 up-regulated proteins, respectively. Healthy-site associated strains Streptococcus vestibularis, Veillonella dispar, Selenomonas sp. HMT478 and Leptotrichia sp. HMT417 showed strong negative interactions (r < -0.7) with 31, 21, 9, and 18 up-regulated proteins, respectively. In contrast the down-regulated proteins did not show strong interactions with the regulated bacteria. The present study identified the proteomic and intra-tissue microbiome profile of human gingiva by employing a PCT-assisted workflow. This is the first report demonstrating the feasibility to analyse full proteome profiles of gingival tissues in both healthy and disease sites, while deciphering the tissue site-specific microbiome signatures.


Assuntos
Microbiota , Proteoma , Fusobacterium , Gengiva , Humanos , Proteômica , RNA Ribossômico 16S/genética , Streptococcus , Veillonella
20.
J Fungi (Basel) ; 5(2)2019 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-31200520

RESUMO

Oral candidiasis is a common side effect of cancer chemotherapy. To better understand predisposing factors, we followed forty-five subjects who received 5-fluorouracil- or doxorubicin-based treatment, during one chemotherapy cycle. Subjects were evaluated at baseline, prior to the first infusion, and at three additional visits within a two-week window. We assessed the demographic, medical and oral health parameters, neutrophil surveillance, and characterized the salivary bacteriome and mycobiome communities through amplicon high throughput sequencing. Twenty percent of all subjects developed oral candidiasis. Using multivariate statistics, we identified smoking, amount of dental plaque, low bacteriome and mycobiome alpha-diversity, and the proportions of specific bacterial and fungal taxa as baseline predictors of oral candidiasis development during the treatment cycle. All subjects who developed oral candidiasis had baseline microbiome communities dominated by Candida and enriched in aciduric bacteria. Longitudinally, oral candidiasis was associated with a decrease in salivary flow prior to lesion development, and occurred simultaneously or before oral mucositis. Candidiasis was also longitudinally associated with a decrease in peripheral neutrophils but increased the neutrophil killing capacity of Candida albicans. Oral candidiasis was not found to be associated with mycobiome structure shifts during the cycle but was the result of an increase in Candida load, with C. albicans and Candida dubliniensis being the most abundant species comprising the salivary mycobiome of the affected subjects. In conclusion, we identified a set of clinical and microbiome baseline factors associated with susceptibility to oral candidiasis, which might be useful tools in identifying at risk individuals, prior to chemotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA