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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 204(8): 2177-2191, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32169848

RESUMO

Emerging evidence suggests comprehensive immune profiling represents a highly promising, yet insufficiently tapped approach to identify potentially prognostic signatures for periodontitis. In this report, we agnostically identified a periodontitis-associated inflammatory expression network with multiple biomarkers identified within gingival crevicular fluid samples from study participants by applying principal component analysis. We identified an IL-17-dominated trait that is associated with periodontal disease and is inversely modified by the level of IL-10. IL-10 mitigated chemokine CXCL5 and CXCL1 expressions in IL-17-stimulated peripheral blood monocytic cells and peripheral blood monocytic cell-derived macrophages. Il10-deficient mice presented more bone loss, which was associated with more Il17 and IL-17-mediated chemokine and cytokine expression at the transcriptional levels in comparison with control wild-type mice in both the Porphyromonas gingivalis-induced experimental murine periodontitis and ligature-induced alveolar bone-loss models. The dampening effect of IL-10 on the excessive signaling of IL-17 appeared to be mediated by innate immune cells populations rather than by gingival epithelial cells, which are the major cell target for IL-17 signaling. Additionally, elevated IL-17 response in Il10-deficient mice specifically elicited an M1-skewing macrophage phenotype in the gingiva that was associated with the advanced bone loss in the ligature model. In summary, IL-17 dominated an inflammatory network characteristic of periodontitis, and IL-10 dampens this excessive IL-17-mediated periodontitis trait.


Assuntos
Inflamação/imunologia , Interleucina-10/imunologia , Interleucina-17/imunologia , Periodontite/imunologia , Animais , Células Cultivadas , Líquido do Sulco Gengival/imunologia , Humanos , Interleucina-10/deficiência , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Componente Principal
2.
Int Endod J ; 55(10): 1042-1052, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35869806

RESUMO

AIM: The aim of this study was to evaluate the IFI16 and IFN-α/ß receptors expression during the genesis and development of experimental apical periodontitis (AP) in mice teeth. METHODOLOGY: Apical periodontitis was induced in the lower first molars of 40 C57BL/6 mice. They were divided according to the experimental periods 2, 7, 14, 21 and 42 days (n = 8 per group). Five animals were used as a control group (without AP). Specimens were submitted to histological processing for description of the inflammatory process, immunostaining for the presence/absence and localization of IFI16 and IFN-α/ß receptors (qualitative and semi-quantitative analysis) and tartrate-resistant acid phosphatase (TRAP) histoenzimology. RESULTS: The results showed a gradual development of AP over the experimental times. The expression of IFI16 was noticeably more exacerbated in the experimental early period (day 2) whilst the lowest expression was observed in the control group (p = .02). For IFN-α/ß receptors, a higher intensity staining was observed 42 days after AP induction, that was statistically different from the control group (p = .02). In addition, the number of TRAP-positive cells was higher on the later periods (days 21 and 42; p < .001). CONCLUSION: IFI16 protein expression was highest during the early periods after AP induction in mice teeth, whilst IFN-α/ß receptor expression was highest after AP became established.


Assuntos
Interferon gama , Proteínas Nucleares/metabolismo , Periodontite Periapical , Fosfoproteínas/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Dente Molar/patologia , Osteoclastos/metabolismo , Periodontite Periapical/patologia
3.
Periodontol 2000 ; 85(1): 126-160, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33226693

RESUMO

Since 2010, next-generation sequencing platforms have laid the foundation to an exciting phase of discovery in oral microbiology as it relates to oral and systemic health and disease. Next-generation sequencing has allowed large-scale oral microbial surveys, based on informative marker genes, such as 16S ribosomal RNA, community gene inventories (metagenomics), and functional analyses (metatranscriptomics), to be undertaken. More specifically, the availability of next-generation sequencing has also paved the way for studying, in greater depth and breadth, the effect of systemic factors on the periodontal microbiome. It was natural to investigate systemic diseases, such as diabetes, in such studies, along with systemic conditions or states, , pregnancy, menopause, stress, rheumatoid arthritis, and systemic lupus erythematosus. In addition, in recent years, the relevance of systemic "variables" (ie, factors that are not necessarily diseases or conditions, but may modulate the periodontal microbiome) has been explored in detail. These include ethnicity and genetics. In the present manuscript, we describe and elaborate on the new and confirmatory findings unveiled by next-generation sequencing as it pertains to systemic factors that may shape the periodontal microbiome. We also explore the systemic and mechanistic basis for such modulation and highlight the importance of those relationships in the management and treatment of patients.


Assuntos
Artrite Reumatoide , Microbiota , Feminino , Humanos , Metagenômica , Gravidez , RNA Ribossômico 16S/genética
4.
Periodontol 2000 ; 82(1): 93-114, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31850638

RESUMO

Inflammasomes are a group of multimolecular intracellular complexes assembled around several innate immune proteins. Recognition of a diverse range of microbial, stress and damage signals by inflammasomes results in direct activation of caspase-1, which subsequently induces the only known form of secretion of active interleukin-1ß and interleukin-18. Although the importance of interleukin-1ß in the periodontium is not questioned, the impact of inflammasomes in periodontal disease and its potential for therapeutics in periodontology is still in its very early stages. Increasing evidence in preclinical models and human data strongly implicate the involvement of inflammasomes in a number of inflammatory, autoinflammatory and autoimmune disorders. Here we review: (a) the currently known inflammasome functions, (b) clinical/preclinical data supporting inflammasome involvement in the context of periodontal and comorbid diseases and (c) potential therapies targeting inflammasomes. To clarify further the inflammasome involvement in periodontitis, we present analyses of data from a large clinical study (n = 5809) that measured the gingival crevicular fluid-interleukin-1ß and grouped the participants based on current periodontal disease classifications. We review data on 4910 European-Americans that correlate 16 polymorphisms in the interleukin-1B region with high gingival crevicular fluid-interleukin-1ß levels. We show that inflammasome components are increased in diseased periodontal tissues and that the caspase-1 inhibitor, VX-765, inhibits ~50% of alveolar bone loss in experimental periodontitis. The literature review further supports that although patients clinically present with the same phenotype, the disease that develops probably has different underlying biological pathways. The current data indicate that inflammasomes have a role in periodontal disease pathogenesis. Understanding the contribution of different inflammasomes to disease development and distinct patient susceptibility will probably translate into improved, personalized therapies.


Assuntos
Inflamassomos , Doenças Periodontais , Caspase 1 , Líquido do Sulco Gengival , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR
5.
Mol Pharm ; 16(9): 4017-4023, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31361146

RESUMO

The in vivo antibacterial activity of NO-releasing hyperbranched polymers was evaluated against Porphyromonas gingivalis, a key oral pathogen associated with periodontitis, using a murine subcutaneous chamber model. Escalating doses of NO-releasing polymers (1.5, 7.5, and 37.5 mg/kg) were administered into a P. gingivalis-infected chamber once a day for 3 days. Chamber fluids were collected on day 4, with microbiological evaluation indicating a dose-dependent bactericidal action. In particular, NO-releasing polymers at 37.5 mg/kg (1170 µg of NO/kg) achieved complete bacterial eradication (>6-log reduction in bacterial viability), demonstrating greater efficacy than amoxicillin (∼4-log reduction in bacterial viability), a commonly used antibiotic. Time-kill assays further revealed that largest dose (37.5 mg/kg; 1170 µg of NO/kg) resulted in ∼3-log killing of P. gingivalis after only a single dose. Based on these results, the potential clinical utility of NO-releasing hyperbranched polymers appears promising, particularly for oral health applications.


Assuntos
Antibacterianos/química , Antibacterianos/uso terapêutico , Infecções por Bacteroidaceae/tratamento farmacológico , Óxido Nítrico/química , Óxido Nítrico/uso terapêutico , Periodontite/tratamento farmacológico , Polímeros/química , Porphyromonas gingivalis/efeitos dos fármacos , Amoxicilina/uso terapêutico , Animais , Infecções por Bacteroidaceae/microbiologia , Modelos Animais de Doenças , Compostos de Epóxi/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Periodontite/microbiologia , Poliaminas/química , Resultado do Tratamento
6.
Adv Exp Med Biol ; 1197: 179-189, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31732942

RESUMO

The genetic basis of oral health has long been theorized, but little information exists on the heritable variance in common oral and dental disease traits explained by the human genome. We sought to add to the evidence base of heritability of oral and dental traits using high-density genotype data in a well-characterized community-based cohort of middle-age adults. We used genome-wide association (GWAS) data combined with clinical and biomarker information in the Dental Atherosclerosis Risk In Communities (ARIC) cohort. Genotypes comprised SNPs directly typed on the Affymetrix Genome-Wide Human SNP Array 6.0 chip with minor allele frequency of >5% (n = 656,292) or were imputed using HapMap II-CEU (n = 2,104,905). We investigated 30 traits including "global" [e.g., number of natural teeth (NT) and incident tooth loss], clinically defined (e.g., dental caries via the DMFS index, periodontitis via the CDC/AAP and WW17 classifications), and biologically informed (e.g., subgingival pathogen colonization and "complex" traits). Heritability (i.e., variance explained; h2) was calculated using Visscher's Genome-wide Complex Trait Analysis (GCTA), using a random-effects mixed linear model and restricted maximum likelihood (REML) regression adjusting for ancestry (10 principal components), age, and sex. Heritability estimates were modest for clinical traits-NT = 0.11 (se = 0.07), severe chronic periodontitis (CDC/AAP) = 0.22 (se = 0.19), WW17 Stage 4 vs. 1/2 = 0.15 (se = 0.11). "High gingival index" and "high red complex colonization" had h2 > 0.50, while a periodontal complex trait defined by high IL-1ß GCF expression and Aggregatibacter actinomycetemcomitans subgingival colonization had the highest h2 = 0.72 (se = 0.32). Our results indicate that all GWAS SNPs explain modest levels of the observed variance in clinical oral and dental measures. Subgingival bacterial colonization and complex phenotypes encompassing both bacterial colonization and local inflammatory response had the highest heritability, suggesting that these biologically informed traits capture aspects of the disease process and are promising targets for genomics investigations, according to the notion of precision oral health.


Assuntos
Cárie Dentária , Estudo de Associação Genômica Ampla , Fenótipo , Cárie Dentária/genética , Cárie Dentária/microbiologia , Genótipo , Humanos , Polimorfismo de Nucleotídeo Único/genética
7.
Hum Mol Genet ; 25(10): 2113-2129, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962152

RESUMO

Genome-wide association studies (GWAS) of chronic periodontitis (CP) defined by clinical criteria alone have had modest success to-date. Here, we refine the CP phenotype by supplementing clinical data with biological intermediates of microbial burden (levels of eight periodontal pathogens) and local inflammatory response (gingival crevicular fluid IL-1ß) and derive periodontal complex traits (PCTs) via principal component analysis. PCTs were carried forward to GWAS (∼2.5 million markers) to identify PCT-associated loci among 975 European American adult participants of the Dental ARIC study. We sought to validate these findings for CP in the larger ARIC cohort (n = 821 participants with severe CP, 2031-moderate CP, 1914-healthy/mild disease) and an independent German sample including 717 aggressive periodontitis cases and 4210 controls. We identified six PCTs with distinct microbial community/IL-1ß structures, although with overlapping clinical presentations. PCT1 was characterized by a uniformly high pathogen load, whereas PCT3 and PCT5 were dominated by Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis, respectively. We detected genome-wide significant signals for PCT1 (CLEC19A, TRA, GGTA2P, TM9SF2, IFI16, RBMS3), PCT4 (HPVC1) and PCT5 (SLC15A4, PKP2, SNRPN). Overall, the highlighted loci included genes associated with immune response and epithelial barrier function. With the exception of associations of BEGAIN with severe and UBE3D with moderate CP, no other loci were associated with CP in ARIC or aggressive periodontitis in the German sample. Although not associated with current clinically determined periodontal disease taxonomies, upon replication and mechanistic validation these candidate loci may highlight dysbiotic microbial community structures and altered inflammatory/immune responses underlying biological sub-types of CP.


Assuntos
Periodontite Crônica/genética , Estudo de Associação Genômica Ampla , Proteínas do Tecido Nervoso/genética , Doenças Periodontais/genética , Ubiquitina-Proteína Ligases/genética , Periodontite Crônica/microbiologia , Periodontite Crônica/patologia , Feminino , Alemanha , Líquido do Sulco Gengival/microbiologia , Humanos , Inflamação/genética , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1beta/genética , Masculino , Doenças Periodontais/microbiologia , Doenças Periodontais/patologia , Fenótipo , Porphyromonas gingivalis/patogenicidade , Análise de Componente Principal , Proteínas Associadas SAP90-PSD95
8.
Am J Pathol ; 179(5): 2431-42, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21939632

RESUMO

Proteoglycan 4 (PRG4), a critical protective factor in articular joints, is implicated in hematopoietic progenitor cell expansion and megakaryopoiesis. PRG4 loss-of-function mutations result in camptodactyly-arthropathy-coxa vara-pericarditis (CACP) syndrome, which is characterized primarily by precocious joint failure. PRG4 was identified as a novel parathyroid hormone (PTH) responsiveness gene in osteoblastic cells in bone, and was investigated as a potential mediator of PTH actions on hematopoiesis. Sixteen-week-old Prg4(-/-) mutant and Prg4(+/+) wild-type mice were treated daily with intermittent PTH (residues 1-34) or vehicle for 6 weeks. At 22 weeks of age, Prg4 mutant mice had increased peripheral blood neutrophils and decreased marrow B220(+) (B-lymphocytic) cells, which were normalized by PTH. The PTH-induced increase in marrow Lin(-)Sca-1(+)c-Kit(+) (hematopoietic progenitor) cells was blunted in mutant mice. Basal and PTH-stimulated stromal cell-derived factor-1 (SDF-1) was decreased in mutant mice, suggesting SDF-1 as a candidate regulator of proteoglycan 4 actions on hematopoiesis in vivo. PTH stimulation of IL-6 mRNA was greater in mutant than in wild-type calvaria and bone marrow, suggesting a compensatory mechanism in the PTH-induced increase in marrow hematopoietic progenitor cells. In summary, proteoglycan 4 is a novel PTH-responsive factor regulating immune cells and PTH actions on marrow hematopoietic progenitor cells.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Hormônio Paratireóideo/farmacologia , Proteoglicanas/metabolismo , Animais , Medula Óssea/metabolismo , Quimiocina CXCL12/metabolismo , Hematopoese/fisiologia , Interleucina-6/metabolismo , Linfócitos/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Trombopoetina/metabolismo
9.
Mol Oral Microbiol ; 37(6): 256-265, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36189827

RESUMO

OBJECTIVES: We have previously characterized the main osteoimmunological events that occur during ligature periodontitis. This study aims to determine the polymicrobial community shifts that occur during disease development. METHODS: Periodontitis was induced in C57BL/6 mice using the ligature-induced periodontitis model. Healthy oral mucosa swabs and ligatures were collected every 3 days from 0 to 18 days post-ligature placement. Biofilm samples were evaluated by 16SrRNA gene sequencing (Illumina MiSeq) and QIIME. Time-course changes were determined by relative abundance, diversity, and rank analyses (PERMANOVA, Bonferroni-adjusted). RESULTS: Microbial differences between health and periodontal inflammation were observed at all phylogenic levels. An evident microbial community shift occurred in 25 genera during the advancement of "gingivitis" (3-6 days) to periodontitis (9-18 days). From day 0 to 18, dramatic changes were identified in Streptococcus levels, with an overall decrease (54.04%-0.02%) as well an overall increase of Enterococcus and Lactobacillus (23.7%-73.1% and 10.1%-70.2%, respectively). Alpha-diversity decreased to its lowest at 3 days, followed by an increase in diversity as disease advancement. Beta-diversity increased after ligature placement, indicating that bone loss develops in response to a greater microbial variability (p = 0.001). Levels of facultative and strict anaerobic bacteria augmented over the course of disease progression, with a total of eight species significantly different during the 18-day period. CONCLUSION: The data supports that murine gingival inflammation and alveolar bone loss develop in response to microbiome shifts. Bacterial diversity increased during progression to bone loss. These findings further support the utilization of the periodontitis ligature model for microbial shift analysis under different experimental conditions.


Assuntos
Perda do Osso Alveolar , Periodontite , Camundongos , Animais , Disbiose , Camundongos Endogâmicos C57BL , Periodontite/microbiologia , Perda do Osso Alveolar/microbiologia , Inflamação , Biofilmes , Modelos Animais de Doenças
10.
J Periodontol ; 93(9): 1366-1377, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35404474

RESUMO

BACKGROUND: Periodontal destruction can be the result of different known and yet-to-be-discovered biological pathways. Recent human genetic association studies have implicated interferon-gamma inducible protein 16 (IFI16) and absent in melanoma 2 (AIM2) with high periodontal interleukin (IL)-1ß levels and more destructive disease, but mechanistic evidence is lacking. Here, we sought to experimentally validate these observational associations and better understand IFI16 and AIM2's roles in periodontitis. METHODS: Periodontitis was induced in Ifi204-/- (IFI16 murine homolog) and Aim2-/- mice using the ligature model. Chimeric mice were created to identify the main source cells of Ifi204 in the periodontium. IFI16-silenced human endothelial cells were treated with periodontal pathogens in vitro. Periodontal tissues from Ifi204-/- mice were evaluated for alveolar bone (micro-CT), cell inflammatory infiltration (MPO+ staining), Il1b (qRT-PCR), and osteoclast numbers (cathepsin K+ staining). RESULTS: Ifi204-deficient mice> exhibited >20% higher alveolar bone loss than wild-type (WT) (P < 0.05), while no significant difference was found in Aim2-/- mice. Ifi204's effect on bone loss was primarily mediated by a nonbone marrow source and was independent of Aim2. Ifi204-deficient mice had greater neutrophil/macrophage trafficking into gingival tissues regardless of periodontitis development compared to WT. In human endothelial cells, IFI16 decreased the chemokine response to periodontal pathogens. In murine periodontitis, Ifi204 depletion elevated gingival Il1b and increased osteoclast numbers at diseased sites (P < 0.05). CONCLUSIONS: These findings support IFI16's role as a novel regulator of inflammatory cell trafficking to the periodontium that protects against bone loss and offers potential targets for the development of new periodontal disease biomarkers and therapeutics.


Assuntos
Perda do Osso Alveolar , Proteínas Nucleares , Periodontite , Fosfoproteínas , Perda do Osso Alveolar/genética , Perda do Osso Alveolar/metabolismo , Perda do Osso Alveolar/prevenção & controle , Animais , Biomarcadores/metabolismo , Catepsina K , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Interferon gama/metabolismo , Interferons/metabolismo , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Periodontite/genética , Periodontite/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo
11.
Nat Med ; 27(5): 892-903, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33767405

RESUMO

Despite signs of infection-including taste loss, dry mouth and mucosal lesions such as ulcerations, enanthema and macules-the involvement of the oral cavity in coronavirus disease 2019 (COVID-19) is poorly understood. To address this, we generated and analyzed two single-cell RNA sequencing datasets of the human minor salivary glands and gingiva (9 samples, 13,824 cells), identifying 50 cell clusters. Using integrated cell normalization and annotation, we classified 34 unique cell subpopulations between glands and gingiva. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral entry factors such as ACE2 and TMPRSS members were broadly enriched in epithelial cells of the glands and oral mucosae. Using orthogonal RNA and protein expression assessments, we confirmed SARS-CoV-2 infection in the glands and mucosae. Saliva from SARS-CoV-2-infected individuals harbored epithelial cells exhibiting ACE2 and TMPRSS expression and sustained SARS-CoV-2 infection. Acellular and cellular salivary fractions from asymptomatic individuals were found to transmit SARS-CoV-2 ex vivo. Matched nasopharyngeal and saliva samples displayed distinct viral shedding dynamics, and salivary viral burden correlated with COVID-19 symptoms, including taste loss. Upon recovery, this asymptomatic cohort exhibited sustained salivary IgG antibodies against SARS-CoV-2. Collectively, these data show that the oral cavity is an important site for SARS-CoV-2 infection and implicate saliva as a potential route of SARS-CoV-2 transmission.


Assuntos
COVID-19/virologia , Boca/virologia , SARS-CoV-2/isolamento & purificação , Saliva/virologia , Enzima de Conversão de Angiotensina 2/análise , Infecções Assintomáticas , COVID-19/etiologia , Humanos , Serina Endopeptidases/análise , Distúrbios do Paladar/etiologia , Distúrbios do Paladar/virologia , Replicação Viral
12.
J Periodontol ; 91 Suppl 1: S6-S11, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32533779

RESUMO

A genome-wide association study of ≈2.5 million markers identified unique biologically informed periodontal complex traits with distinct microbial communities and interleukin-1ß (IL-1ß) levels. Each trait was associated with different single nucleotide polymorphisms. These variants include genes associated with immune responses, microbial colonization, and the epithelial barrier function. The specific set of variants leads to individual biological paths that converge into an overlapping clinical phenotype of periodontal tissue destruction. This concept suggests that periodontal disease is a group of distinct conditions. We identified polymorphisms in inflammasome genes interferon gamma inducible protein 16 (IFI16) and absent in melanoma 2 (AIM2) that were associated with increased severity of periodontal disease. Inflammasomes respond to pathogen or tissue "danger" signals and assemble into multiprotein "machineries" that are essential for the cleavage of proinflammatory mediator IL-1ß into an active form. Thus, understanding how variants of IFI16 and AIM2 contribute to periodontal disease pathogenesis may lead to treatment options that address individual biological variations and precision therapies for oral health.


Assuntos
Inflamassomos , Doenças Periodontais , Caspase 1 , Estudo de Associação Genômica Ampla , Humanos , Inflamassomos/genética , Interleucina-1beta/genética , Doenças Periodontais/genética , Polimorfismo de Nucleotídeo Único/genética
13.
J Periodontol ; 88(7): 663-672, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28387608

RESUMO

BACKGROUND: The single nucleotide polymorphism (SNP) context of a previously identified periodontitis-associated locus is investigated, and its association with microbial, biologic, and periodontal disease clinical parameters is examined. METHODS: A 200-kb spanning region of 1q12 previously highlighted in a genome-wide association scan among 4,766 European American individuals (SNP rs1633266) was annotated. Two haplotype blocks were selected. Association of these polymorphisms with data on microbial plaque composition, gingival crevicular fluid (GCF)-interleukin (IL)-1ß levels, and clinical parameters of periodontal disease were examined. Descriptive analysis of IFI16 and AIM2 protein expression in gingival tissues from healthy individuals (n = 2) and individuals with chronic periodontitis (n = 2) was done via immunohistochemistry. RESULTS: The highlighted locus is a 100-kb region containing the interferon γ-inducible protein 16 (IFI16) and absent in melanoma 2 (AIM2) genes. Two haplotype blocks, rs6940 and rs1057028, were significantly associated with increased extent bleeding on probing and levels of microorganisms Porphyromonas gingivalis, Tannerella forsythia, and Campylobacter rectus (P ≤0.05). Haplotype block rs1057028 was also significantly associated with pathogens Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans, increased GCF-IL-1ß levels, and extent of probing depth ≥4 mm (P ≤0.05). Prevalence of severe periodontitis (biofilm-gingival interface P3 classification) was positively associated with haplotype block rs1057028. Similar trends were observed for haplotype block rs1057028. IFI16 and AIM2 protein expression was observed in multiple cell types of gingival tissues, including inflammatory cells. CONCLUSION: This study found IFI16 and AIM2 SNPs associated with higher levels of periodontal microorganisms and an increased percentage of periodontal disease clinical parameters, suggesting the need for functional studies and additional fine-mapping of variants in the 1q12-locus.


Assuntos
Periodontite Crônica/genética , Proteínas de Ligação a DNA/genética , Gengiva/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas/genética , Polimorfismo de Nucleotídeo Único , Idoso , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Periodontite Crônica/metabolismo , Periodontite Crônica/microbiologia , Proteínas de Ligação a DNA/metabolismo , Placa Dentária/microbiologia , Feminino , Fusobacterium nucleatum/isolamento & purificação , Estudo de Associação Genômica Ampla , Gengiva/microbiologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , População Branca
14.
J Periodontol ; 87(1): 27-35, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26430927

RESUMO

BACKGROUND: This study measures microbial composition changes during biofilm overgrowth and subsequent removal among patients with various states of periodontal disease. METHODS: In this prospective cohort study, 175 participants with various periodontal states (five biofilm-gingival interface [BGI] groups) abstained from oral hygiene while using an acrylic stent. At day 21, participants reinstituted oral hygiene and were followed for 4 weeks. Clinical parameters were recorded, and subgingival plaque samples were analyzed at baseline, peak of induction (day 21), and resolution using 16S rRNA probes (human oral microbe identification microarray [HOMIM]). Using the change score (peak at induction minus baseline) for bleeding on probing and probing depth (PD), the patients were separated into high and low clinical responders. RESULTS: At baseline, synergistetes were more abundant in moderate and severe periodontitis (BGI-P2 and -P3) compared to mild periodontitis (BGI-P1), health (BGI-H), and gingivitis (BGI-G) (P = 0.005). Overall, at day 21 there was an increase in HOMIM scores of firmicutes (P ≤0.001), fusobacteria (P = 0.003), proteobacteria (P ≤0.001), synergistetes (P = 0.04), and bacteroidetes (P ≤0.001). At resolution, these phyla returned to baseline, except for synergistetes. Levels of synergistetes were significantly higher at day 21 (P ≤0.0001) and resolution (P = 0.0002) for high clinical responders compared to low responders. CONCLUSION: The association of synergistetes as a baseline predictor of incident PD increase, as well as the higher levels at day 21, indicates a pathogenic role for these organisms in disease progression in addition to the previously characterized fusobacteria, proteobacteria, firmicutes, and bacteroidetes.


Assuntos
Biofilmes , Placa Dentária , Humanos , Índice Periodontal , Bolsa Periodontal , Periodontite , Estudos Prospectivos , RNA Ribossômico 16S
15.
J Periodontol ; 85(12): 1770-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25079398

RESUMO

BACKGROUND: The purpose of this study is to determine whether baseline salivary inflammatory biomarkers could discriminate between different clinical levels of disease and/or detect clinical changes over a 3-week stent-induced biofilm overgrowth (SIBO) period. METHODS: A total of 168 participants were enrolled in a 21-day experimental gingivitis investigation and grouped according to clinical measures of periodontal status of health and diseased individuals representing each of five biofilm gingival interface (BGI) periodontal groups: 1) health, all probing depth (PD) <3 mm and bleeding on probing (BOP) <10%; 2) gingivitis, all PD <3 mm and BOP ≥10%; 3) periodontitis (P)1, ≥1 site with PD >3 mm and BOP ≤10%; 4) P2, ≥1 site with PD >3 mm and BOP >10% but ≤50%; and 5) P3, ≥1 site with PD >3 mm and BOP >50%. Stents were used to prevent plaque removal during brushing over one maxillary and one mandibular posterior dental sextant for 21 days. Clinical periodontal parameters and unstimulated saliva were collected at screening, baseline, and each week during SIBO. Saliva samples were assessed for levels of 13 different biomarkers by multiplex immunoassay. RESULTS: Higher salivary levels of interleukin (IL)-1ß, matrix metalloproteinase (MMP)-3, MMP-8, MMP-9, and neutrophil gelatinase-associated lipocalin (NGAL) were found in diseased groups compared with the healthy group at baseline. Conversely, higher IL-1 receptor antagonist (ra) levels were found in healthy patients at baseline. In addition, during SIBO, MMP-1, tissue inhibitor of metalloproteinase (TIMP)-1, and TIMP-2 levels increased across all participant groups. A stepwise linear regression model using all salivary biomarkers demonstrated that, at baseline, increased IL-1ra (P = 0.004) and IL-6 (P = 0.009) were significantly associated with change in PDs during SIBO. CONCLUSIONS: In summary, this investigation supports salivary levels of IL-1ra and IL-6 as potential indicators for PD changes during induced gingival inflammation. In addition, participants from the BGI-P3 group (severe periodontitis) demonstrated elevated baseline levels of IL-1ß, MMP-3, MMP-8, MMP-9, and NGAL compared with the other study groups, strengthening the relevance of participants' biologic phenotype on expression of salivary biomarkers.


Assuntos
Biofilmes/crescimento & desenvolvimento , Biomarcadores/análise , Mediadores da Inflamação/análise , Saliva/química , Proteínas de Fase Aguda/análise , Adulto , Idoso , Estudos de Coortes , Placa Dentária/microbiologia , Feminino , Gengiva/metabolismo , Gengivite/microbiologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1/análise , Interleucina-1beta/análise , Interleucina-6/análise , Lipocalina-2 , Lipocalinas/análise , Masculino , Metaloproteinase 1 da Matriz/análise , Metaloproteinase 3 da Matriz/análise , Metaloproteinase 8 da Matriz/análise , Metaloproteinase 9 da Matriz/análise , Pessoa de Meia-Idade , Periodontite/classificação , Periodontite/microbiologia , Estudos Prospectivos , Proteínas Proto-Oncogênicas/análise , Inibidor Tecidual de Metaloproteinase-1/análise , Inibidor Tecidual de Metaloproteinase-2/análise , Adulto Jovem
16.
J Periodontol ; 84(5): 694-703, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22769442

RESUMO

BACKGROUND: Periostin is a matricellular protein essential for tissue integrity and maturation and is believed to have a key function as a modulator of periodontal ligament (PDL) homeostasis. The aim of this study is to evaluate whether periodontal disease-associated pathogen-related virulence factors (endotoxins/lipopolysaccharides [LPS]) and proinflammatory cytokines alter the expression of periostin in PDL cells. METHODS: Human PDL cultures were exposed to inflammatory mediators (tumor necrosis factor-α [TNF-α]), bacterial virulence factors (Porphyromonas gingivalis LPS) or a combination in a biomechanically challenged environment. Culture conditions were applied for 24 hours, 4 days, and 7 days. Periostin and TGF-ß inducible gene clone H3 (ßIGH3) mRNA expression from cell lysates were analyzed. Periostin and ßIGH3 proteins were also detected and semiquantified in both cell lysates and cell culture supernatants by Western blot. In addition, periostin localization by immunofluorescence was performed. Analysis of variance and Fisher tests were used to define the statistical differences among groups (P <0.05). RESULTS: In a mechanically challenged environment, periostin protein was more efficiently incorporated into the matrix compared to the non-loaded controls (higher levels of periostin in the supernatant in the non-loaded group). Interestingly, chronic exposure to proinflammatory cytokines and/or microbial virulence factors significantly decreased periostin protein levels in the loaded cultures. There was greater variability on ßIGH3 levels, and no particular pattern was clearly evident. CONCLUSIONS: Inflammatory mediators (TNF-α) and bacterial virulence factors (P. gingivalis LPS) decrease periostin expression in human PDL fibroblasts. These results support a potential mechanism by which periostin alterations could act as a contributing factor during periodontal disease progression.


Assuntos
Moléculas de Adesão Celular/biossíntese , Interações Hospedeiro-Patógeno , Lipopolissacarídeos/farmacologia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Porphyromonas gingivalis , Fator de Necrose Tumoral alfa/farmacologia , Adulto , Análise de Variância , Western Blotting , Células Cultivadas , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Ligamento Periodontal/citologia , Fator de Crescimento Transformador beta/metabolismo
17.
Cell Host Microbe ; 13(5): 595-601, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23684310

RESUMO

Periodontitis is a common disease that is characterized by resorption of the alveolar bone and mediated by commensal bacteria that trigger host immune responses and bone destruction through unidentified mechanisms. We report that Nod1, an innate intracellular host receptor for bacterial peptidoglycan-related molecules, is critical for commensal-induced periodontitis in a mouse model. Mice lacking Nod1 exhibit reduced bone resorption as well as impaired recruitment of neutrophils to gingival tissues and osteoclasts to the alveolar bone, which mediate tissue and bone destruction. Further analysis showed that accumulation of a Nod1-stimulating commensal bacterium, NI1060, at gingival sites was sufficient to induce neutrophil recruitment and bone resorption. Genomic sequencing revealed that NI1060 is a mouse-specific bacterium that is related to bacteria associated with the development of aggressive periodontitis in humans. These findings provide insight into commensal-host interactions contributing to periodontitis and identify a potential target for preventing this common oral disease.


Assuntos
Perda do Osso Alveolar/patologia , Bactérias/patogenicidade , Interações Hospedeiro-Patógeno , Boca/microbiologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Periodontite/patologia , Transdução de Sinais , Animais , Camundongos , Periodontite/complicações
18.
Methods Mol Biol ; 887: 135-48, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22566053

RESUMO

In humans, microbially induced inflammatory periodontal diseases are the primary initiators that disrupt the functional and structural integrity of the periodontium (i.e., the alveolar bone, the periodontal ligament, and the cementum). The reestablishment of its original structure, properties, and function constitutes a significant challenge in the development of new therapies to regenerate tooth-supporting defects. Preclinical models represent an important in vivo tool to critically evaluate and analyze the key aspects of novel regenerative therapies, including (1) safety, (2) effectiveness, (3) practicality, and (4) functional and structural stability over time. Therefore, these models provide foundational data that supports the clinical validation and the development of novel innovative regenerative periodontal technologies. Steps are provided on the use of the root fenestration animal model for the proper evaluation of periodontal outcome measures using the following parameters: descriptive histology, histomorphometry, immunostaining techniques, three-dimensional imaging, electron microscopy, gene expression analyses, and safety assessments. These methods will prepare investigators and assist them in identifying the key end points that can then be adapted to later stage human clinical trials.


Assuntos
Regeneração Tecidual Guiada Periodontal/métodos , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Animais , Regeneração Óssea/fisiologia , Humanos , Ratos
19.
Head Face Med ; 7: 13, 2011 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-21767401

RESUMO

BACKGROUND: Extracellular matrix proteins are key factors that influence the regenerative capacity of tissues. The objective of the present study was to evaluate the effects of enamel matrix derivative (EMD), TGF-ß1, and the combination of both factors (EMD+TGF-ß1) on human osteoblastic cell cultures. METHODS: Cells were obtained from alveolar bone of three adult patients using enzymatic digestion. Effects of EMD, TGF-ß1, or a combination of both were analyzed on cell proliferation, bone sialoprotein (BSP), osteopontin (OPN) and alkaline phosphatase (ALP) immunodetection, total protein synthesis, ALP activity and bone-like nodule formation. RESULTS: All treatments significantly increased cell proliferation compared to the control group at 24 h and 4 days. At day 7, EMD group showed higher cell proliferation compared to TGF-ß1, EMD + TGF-ß1 and the control group. OPN was detected in the majority of the cells for all groups, whereas fluorescence intensities for ALP labeling were greater in the control than in treated groups; BSP was not detected in all groups. All treatments decreased ALP levels at 7 and 14 days and bone-like nodule formation at 21 days compared to the control group. CONCLUSIONS: The exposure of human osteoblastic cells to EMD, TGF-ß1 and the combination of factors in vitro supports the development of a less differentiated phenotype, with enhanced proliferative activity and total cell number, and reduced ALP activity levels and matrix mineralization.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteínas do Esmalte Dentário/farmacologia , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Fosfatase Alcalina/metabolismo , Processo Alveolar/citologia , Bromodesoxiuridina , Células Cultivadas , Imunofluorescência , Humanos , Osteoblastos/metabolismo , Osteopontina/metabolismo , Sialoglicoproteínas/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo
20.
J Clin Periodontol ; 34(6): 514-22, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17509092

RESUMO

AIM: The objective of this study was to evaluate the effects of enamel matrix derivative (EMD), transforming growth factor-beta1 (TGF-beta1), and a combination of both factors (EMD+TGF-beta1) on periodontal ligament (PDL) fibroblasts. MATERIAL AND METHODS: Human PDL fibroblasts were obtained from three adult patients with a clinically healthy periodontium, using the explant technique. The effects of EMD, TGF-beta1, or a combination of both were analysed on PDL cell proliferation, adhesion, wound healing, and total protein synthesis, and on alkaline phosphatase (ALP) activity and bone-like nodule formation. RESULTS: Treatment with EMD for 4, 7, and 10 days increased cell proliferation significantly compared with the negative control (p<0.05). At day 10, EMD and EMD+TGF-beta1 showed a higher cell proliferation compared with TGF-beta1 (p<0.01). Cell adhesion was significantly up-regulated by TGF-beta1 compared with EMD and EMD+TGF-beta1 (p<0.01). EMD enhanced in vitro wound healing of PDL cells compared with the other treatments. Total protein synthesis was significantly increased in PDL cells cultured with EMD compared with PDL cells treated with TGF-beta1 or EMD+TGF-beta1 (p<0.05). EMD induced ALP activity in PDL fibroblasts, which was associated with an increase of bone-like nodules. CONCLUSION: These findings support the hypothesis that EMD and TGF-beta1 may play an important role in periodontal regeneration. EMD induced PDL fibroblast proliferation and migration, total protein synthesis, ALP activity, and mineralization, while TGF-beta1 increased cellular adhesion. However, the combination of both factors did not positively alter PDL fibroblast behaviour.


Assuntos
Materiais Biocompatíveis/farmacologia , Proteínas do Esmalte Dentário/farmacologia , Fibroblastos/efeitos dos fármacos , Ligamento Periodontal/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Adulto , Fosfatase Alcalina/efeitos dos fármacos , Antimetabólitos , Materiais Biocompatíveis/administração & dosagem , Bromodesoxiuridina , Calcificação Fisiológica/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas do Esmalte Dentário/administração & dosagem , Combinação de Medicamentos , Fibroblastos/citologia , Humanos , Ligamento Periodontal/citologia , Proteínas/efeitos dos fármacos , Fatores de Tempo , Fator de Crescimento Transformador beta1/administração & dosagem , Regulação para Cima/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA