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








Base de dados
Intervalo de ano de publicação
1.
Lab Chip ; 24(14): 3326-3346, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38874483

RESUMO

Periodontal disease (PD), a chronic inflammatory disorder that damages the tooth and its supporting components, is a common global oral health problem. Understanding the intricacies of these disorders, from gingivitis to severe PD, is critical for efficient treatment, diagnosis, and prevention in dental care. Periodontal biosensors and biomarkers are critical in improving oral health diagnostic skills. Clinicians may accomplish early identification, tailored therapy, and efficient tracking of periodontal diseases by using these technologies, ushering in a new age of accurate oral healthcare. Traditional periodontitis diagnostic methods frequently rely on physical probing and visual examinations, necessitating the development of point-of-care (POC) devices. As periodontal disorders necessitate more precise and rapid diagnosis, incorporating novel innovations in biosensors and biomarkers becomes increasingly crucial. These innovations improve our capacity to diagnose, monitor, and adapt periodontal therapies, bringing in the next phase of customized and effective dental healthcare. The review discusses the characteristics and stages of PD, clinical treatment techniques, prominent biomarkers and infection-associated factors that may be employed to determine PD, biomedical sensing, and POC appliances that have been created so far to diagnose stages of PD and its progression profile, as well as predicting future developments in this field.


Assuntos
Biomarcadores , Doenças Periodontais , Sistemas Automatizados de Assistência Junto ao Leito , Humanos , Doenças Periodontais/diagnóstico , Biomarcadores/análise , Técnicas Biossensoriais/instrumentação
2.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38731952

RESUMO

Porphyromonas gingivalis (Pg), a Gram-negative oral pathogen, promotes and accelerates periodontitis-associated gut disorders. Intestinal epithelial barrier dysfunction is crucial in the pathogenesis of intestinal and systemic diseases. In this study, we sought to elucidate the protective role of cinnamaldehyde (CNM, an activator of Nrf2) against P. gingivalis (W83) and Pg-derived lipopolysaccharide (Pg-LPS) induced intestinal epithelial barrier dysfunction via antioxidative mechanisms in IEC-6 cells. IEC-6 (ATCC, CRL-1592) cells were pretreated with or without CNM (100 µM), in the presence or absence of P. gingivalis (strain W83, 109 MOI) or Pg-LPS (1, 10, and 100 µg/mL), respectively, between 0-72 h time points by adopting a co-culture method. Intestinal barrier function, cytokine secretion, and intestinal oxidative stress protein markers were analyzed. P. gingivalis or Pg-LPS significantly (p < 0.05) increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels expressing oxidative stress damage. Pg-LPS, as well as Pg alone, induces inflammatory cytokines via TLR-4 signaling. Furthermore, infection reduced Nrf2 and NAD(P)H quinone dehydrogenase 1 (NQO1). Interestingly, inducible nitric oxide synthase (iNOS) protein expression significantly (p < 0.05) increased with Pg-LPS or Pg infection, with elevated levels of nitric oxide (NO). CNM treatment suppressed both Pg- and Pg-LPS-induced intestinal oxidative stress damage by reducing ROS, MDA, and NO production. Furthermore, CNM treatment significantly upregulated the expression of tight junction proteins via increasing the phosphorylation levels of PI3K/Akt/Nrf2 suppressing inflammatory cytokines. CNM protected against Pg infection-induced intestinal epithelial barrier dysfunction by activating the PI3K/Akt-mediated Nrf2 signaling pathway in IEC-6 cells.


Assuntos
Acroleína , Mucosa Intestinal , Fator 2 Relacionado a NF-E2 , Óxido Nítrico , Fosfatidilinositol 3-Quinases , Porphyromonas gingivalis , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Fator 2 Relacionado a NF-E2/metabolismo , Acroleína/análogos & derivados , Acroleína/farmacologia , Animais , Transdução de Sinais/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Porphyromonas gingivalis/patogenicidade , Fosfatidilinositol 3-Quinases/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Óxido Nítrico/metabolismo , Linhagem Celular , Lipopolissacarídeos , Estresse Oxidativo/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Citocinas/metabolismo
3.
J Alzheimers Dis Rep ; 6(1): 577-587, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275415

RESUMO

Background: Cleavage of the amyloid-ß protein precursor (AßPP) mediated by host secretase enzymes, releases several fragments including amyloid-ß (Aß40 and Aß42). Objective: To determine if Porphyromonas gingivalis conditioned medium cleaved AßPP to release Aß40 and Aß42. Methods: The SH-SY5Y cell line was challenged, in vitro, with P. gingivalis (Pg381) conditioned medium in the presence/absence of cytokines. The cells and their supernatants were assessed for AßPP cleavage fragments by immunoblotting and transmission electron microscopy. Results: Western blotting of the cell lysates with the anti-AßPP C-terminal antibody demonstrated variable molecular weight bands corresponding to full length and fragmented AßPP in lanes treated with the following factors: Tryptic soy broth (TSB), Pg381, IL-6, Pg381 + IL-1ß, and Pg381 + TNF-α. The low molecular weight bands corresponding to the C99 dimerized fragment were observed in the Pg381 and interlukin-6 (IL-6) treated groups and were significantly more intense in the presence of Pg381 with either IL-6 or TNF-α. Bands corresponding to the dimerized C83 fragment were observed with cells treated with TNF-α alone and with Pg381 combined with IL-1ß or IL-6 or TNF-α. The anti-Aß antibody detected statistically significant Aß40 and Aß42, levels when these two Aß species were pooled across test samples and compared to the untreated group. Electron microscopic examination of the supernatants demonstrated insoluble Aß40 and Aß42. Conclusion: These observations strongly imply that AßPP is an infection responsive protein cleaved via the amyloidogenic pathway on exposure to conditioned medium and in the presence of pro-inflammatory mediators.

4.
J Alzheimers Dis ; 82(3): 1259-1275, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151813

RESUMO

BACKGROUND: Periodontal disease(s) and metabolic illnesses negatively impact the quality of life and, eventually mental health. OBJECTIVE: This study investigated the effect of Porphyromonas gingivalis (W83) oral infection on the development of Alzheimer's disease (AD) pathophysiology in a wild-type obese, diabetic (db/db) mouse model. METHODS: The db/db mice were either orally infected with P. gingivalis and Fusobacterium nucleatum or sham infected for 16 weeks. The presence of amyloid-ß (Aß) and neurofibrillary tangles (NFTs) were assessed using a silver impregnation technique and subsequently by immunohistochemistry for tau and neuroinflammation. The mRNA abundance of a panel of 184 genes was performed using quantitative real-time PCR, and the differentially expressed genes were analyzed by Ingenuity Pathway Analysis. RESULTS: While no Aß plaques and NFTs were evident by silver impregnation, immunohistochemistry (glial cell markers) of the P. gingivalis-infected mice tissue sections exhibited neuroinflammation in the form of reactive microglia and astrocytes. Anti-tau immunopositivity, in addition to cells, was prominent in thickened axons of hippocampal CA neurons. The mRNA abundance of crucial genes in the insulin signaling pathway (INSR, IGF1, IRS, IDE, PIK3R, SGK1, GYS, GSK3B, AKT1) were upregulated, potentially exacerbating insulin resistance in the brain by P. gingivalis oral infection. Increased mRNA abundance of several kinases, membrane receptors, transcription factors, and pro-inflammatory mediators indicated hyperactivation of intracellular cascades with potential for tau phosphorylation and Aß release in the same infection group. CONCLUSION: P. gingivalis W83 infection of db/db mice provides a disease co-morbidity model with the potential to reproduce AD pathophysiology with induced periodontal disease.


Assuntos
Doença de Alzheimer/fisiopatologia , Infecções por Bacteroidaceae/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Obesidade/fisiopatologia , Porphyromonas gingivalis , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Animais , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/psicologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/psicologia , Camundongos , Camundongos Transgênicos , Obesidade/genética , Obesidade/psicologia
5.
J Cell Physiol ; 236(8): 5715-5724, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33400284

RESUMO

Periodontal ligament fibroblasts (PdLFs) are an elongated cell type in the periodontium with matrix and bone regulatory functions which become abnormal in periodontal disease (PD). Here we found that the normally elongated and oriented PdLF nucleus becomes rounded and loses orientation in a mouse model of PD. Using in vitro micropatterning of cultured primary PdLF cell shape, we show that PdLF elongation correlates with nuclear elongation and the presence of thicker, contractile F-actin fibers. The rounded nuclei in mouse PD models in vivo are, therefore, indicative of reduced actomyosin tension. Inhibiting actomyosin contractility by inhibiting myosin light chain kinase, Rho kinase or myosin ATPase activity, in cultured PdLFs each consistently reduced messenger RNA levels of bone regulatory protein osteoprotegerin (OPG). Infection of cultured PdLFs with two different types of periodontal bacteria (Porphyromonas gingivalis and Fusobacterium nucleatum) failed to recapitulate the observed nuclear rounding in vivo, upregulated nonmuscle myosin II phosphorylation and downregulated OPG. Collectively, our results add support to the hypothesis that PdLF contractility becomes decreased and contributes to disease progression in PD.


Assuntos
Actomiosina/metabolismo , Fibroblastos/metabolismo , Osteoprotegerina/metabolismo , Ligamento Periodontal/efeitos dos fármacos , Animais , Citocinas/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ligamento Periodontal/metabolismo , Porphyromonas gingivalis/metabolismo
6.
J Periodontol ; 92(7): 54-65, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33128253

RESUMO

BACKGROUND: Periodontal disease (PD) is known to be associated with endothelial dysfunction in patients with coronary artery and/or cardiovascular disease. In our study, we sought to explore the virulence of P. gingivalis (Pg) affecting glycogen synthase kinase 3 beta (GSK-3ß)/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/tetrahydrobiopterin (BH4 )/ nitric oxide synthase (NOS) expression in primary human aortic endothelial cells (pHAECs). METHODS: pHAECs were infected for 48 hours with Pg in vitro using the Human oxygen-Bacteria anaerobic coculture technique. Cell viability was determined, and target gene expression changes were evaluated by quantitative real-time polymerase chain reaction at the end of each incubation period. RESULTS: Pg impaired pHAEC viability 24 hours post-infection. Pg infection reduced mRNA expression levels of endothelial NOS (eNOS), Nrf2, and Phase II enzymes (heme oxygenase-1, catalase, superoxide dismutase-1) in a time-dependent manner. Significant (P <0.05) increase in the inflammatory markers (interleukin [IL]-1ß, IL-6, and tumor necrosis factor-α) were observed in the medium as well as in the infected cells. Interestingly, inducible NOS mRNA levels showed a significant (P <0.05) increase at 12 hours and 24 hours and were reduced at later time points. BH4 (cofactor of eNOS) biosynthesis enzyme dihydrofolate reductase (DHFR, salvage pathway) mRNA levels showed a significant (P <0.05) decrease, while mRNA levels of GSK-3ß were elevated. CONCLUSIONS: These results suggest that periodontal bacterial infection may cause significant changes in the endothelial GSK-3ß/BH4 /eNOS/Nrf2 pathways, which may lead to impaired vascular relaxation. Greater understanding of the factors that adversely affect endothelial cell function could contribute to the development of new therapeutic compounds to treat PD-induced vascular diseases.


Assuntos
Óxido Nítrico , Porphyromonas gingivalis , Células Endoteliais , Endotélio Vascular , Glicogênio Sintase Quinase 3 beta , Humanos , Fator 2 Relacionado a NF-E2
7.
J Alzheimers Dis Rep ; 4(1): 501-511, 2020 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-33532698

RESUMO

Porphyromonas gingivalis (P. gingivalis) is one of the several important bacterial pathogens associated with the sporadic Alzheimer's disease (AD). Different serotypes are either capsulated or are non-capsulated. It has been demonstrated that P. gingivalis (non-capsulated) can reproduce the neurodegenerative AD-like changes in vitro, and a capsular P. gingivalis (strain W83) could reproduce the cardinal hallmark lesions of AD in a wild-type mouse model. All P. gingivalis forms express proteolytically active proteases that enable cleavage of the amyloid-ß protin precursor (AßPP) and tau resulting in the formation of amyloid-ß and neurofibrillary tangles. Tau is an established substrate for gingipains, which can cleave tau into various peptides. Some of the P. gingivalis fragmented tau protein peptides contain "VQIINK" and "VQIVYK" hexapeptide motifs which map to the flanking regions of the microtubule binding domains and are also found in paired helical filaments that form NFTs. P. gingivalis can induce peripheral inflammation in periodontitis and can also initiate signaling pathways that activate kinases, which in turn, phosphorylate neuronal tau. Periodontal disease related inflammation has metabolic implications for an individual's peripheral and brain health as patients suffering from generalized periodontitis often have related co-morbidities and are "at risk" of developing AD. The aim here is to discuss the role of P. gingivalis behind such associations with the backdrop of huge efforts to test P. gingivalis virulence factors clinically (GAIN Trial: Phase 2/3 Study of COR388 in Subjects with AD) with inhibitors, which may lead to an intervention by reducing the pathogenic bacterial load.

8.
J Oral Microbiol ; 13(1): 1849493, 2020 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-33391626

RESUMO

Background: Evidence suggest periodontal bacterial infection can contribute to oral cancer initiation and progression. Aim: To investigate the effects of periodontal bacteria on oral cancer cell behavior using a cell-based system and a mouse carcinogenesis model. Methods: Oral cancer cell lines were polyinfected with four periodontal bacteria. Cytokine levels and relative changes in oncogene mRNA expression were determined post-infection. Oral tumours in mice induced by 4-nitroquinoline-1-oxide (4NQO) were compared with and without administrating periodontal bacteria. Results: Polyinfected oral cancer cells had upregulated MMP1, MMP9, and IL-8. The expression of cell survival markers MYC, JAK1, and STAT3 and epithelial-mesenchymal transition markers ZEB1 and TGF-ß were also significantly elevated. Monoinfections showed F. nucleatum alone had comparable or greater effects than the four bacteria together. Fusobacterial culture supernatant, primarily LPS, was sufficient to induce IL-8 secretion, demonstrating that direct contact of live Fusobacteria with cancer cells might not be required to exert changes in cancer cell behaviour. In the 4NQO-induced oral tumour model, mice infected with bacteria developed significantly larger and more numerous lesions compared to those not infected. Conclusion: This study demonstrated that Fusobacteria could potentially enhance cancer cell invasiveness, survival, and EMT when presented in the oral tumour microenvironment. Abbreviations: 4NQO, 4-nitroquinoline-1-oxide; ELISA, enzyme-linked immunosorbent assay; EMT, epithelial-mesenchymal transition; IL-8, interleukin-8; JAK1, Janus kinase 1; LPS, lipopolysaccharide; MMP, matrix metalloproteinase; OSCCs, oral squamous cell carcinomas; PK, proteinase K; PMB, Polymyxin B; qRT-PCR, quantitative real-time polymerase chain reaction; STAT3, signal transducer and activator of transcription 3; TGF-ß, transforming growth factor beta; ZEB1, zinc finger E-Box binding homeobox 1.

9.
Pathog Dis ; 76(8)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30351354

RESUMO

Plasma membrane-associated Toll-like receptor (TLR2 and TLR4) signaling contributes to oral microbe infection-induced periodontitis and atherosclerosis. We recently reported that either TLR2 or TLR4 receptor deficiency alters recognition of a consortium of oral pathogens, modifying host responses in periodontitis and atherosclerosis. We evaluated the effects of combined TLR2-/-TLR4-/- double knockout mice on innate immune signaling and induction of periodontitis and atherosclerosis after polybacterial infection with Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Fusobacterium nucleatum in a mouse model. Multispecies infections established gingival colonization in all TLR2-/-TLR4-/- mice and induced production of bacterial-specific IgG antibodies. In combined TLR2-/-TLR4-/- deficiency there was, however, reduced alveolar bone resorption and mild gingival inflammation with minimal migration of junctional epithelium and infiltration of inflammatory cells. This indicates a central role for TLR2 and TLR4 in periodontitis. Atherosclerotic plaque progression was markedly reduced in infected TLR2-/-TLR4-/- mice or in heterozygotes indicating a profound effect on plaque growth. However, bacterial genomic DNA was detected in multiple organs in TLR2-/-TLR4-/- mice indicating an intravascular dissemination from gingival tissue to heart, aorta, kidney and lungs. TRL2 and TLR4 were dispensable for systemic spread after polybacterial infections but TLR2 and 4 deficiency markedly reduces atherosclerosis induced by oral bacteria.


Assuntos
Aterosclerose/patologia , Infecções Bacterianas/patologia , Coinfecção/patologia , Imunidade Inata , Periodontite/patologia , Receptor 2 Toll-Like/deficiência , Receptor 4 Toll-Like/deficiência , Estruturas Animais/microbiologia , Animais , Aterosclerose/imunologia , Infecções Bacterianas/imunologia , Coinfecção/imunologia , Feminino , Fusobacterium nucleatum/imunologia , Masculino , Camundongos Knockout , Periodontite/imunologia , Porphyromonas gingivalis/imunologia , Transdução de Sinais , Tannerella forsythia/imunologia , Treponema denticola/imunologia
10.
Open Biol ; 8(9)2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-30209038

RESUMO

The periodontium is a structurally and functionally complex tissue that facilitates the anchorage of teeth in jaws. The periodontium consists of various cell types including stem cells, fibroblasts and epithelial cells. Cells of the periodontium are constantly exposed to mechanical stresses generated by biological processes such as the chewing motions of teeth, by flows generated by tongue motions and by forces generated by implants. Mechanical stresses modulate the function of cells in the periodontium, and may play a significant role in the development of periodontal disease. Here, we review the literature on the effect of mechanical forces on periodontal cells in health and disease with an emphasis on molecular and cellular mechanisms.


Assuntos
Mecanotransdução Celular , Periodonto/citologia , Proliferação de Células , Células Epiteliais/citologia , Fibroblastos/citologia , Humanos , Células-Tronco/citologia
11.
Pathog Dis ; 75(1)2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28104616

RESUMO

Periodontal disease (PD) and atherosclerotic vascular disease (ASVD) are both chronic inflammatory diseases with a polymicrobial etiology and have been epidemiologically associated. The purpose is to examine whether periodontal bacteria that infect the periodontium can also infect vascular tissues and enhance pre-existing early aortic atherosclerotic lesions in LDLRnull mice. Mice were orally infected with intermediate bacterial colonizer Fusobacterium nucleatum for the first 12 weeks followed by late bacterial colonizers (Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia) for the remaining 12 weeks mimicking the human oral microbiota ecological colonization. Genomic DNA from all four bacterial was detected in gingival plaque by PCR, consistently demonstrating infection of mouse gingival surfaces. Infected mice had significant levels of IgG and IgM antibodies, alveolar bone resorption, and showed apical migration of junctional epithelium revealing the induction of PD. These results support the ability of oral bacteria to cause PD in mice. Detection of bacterial genomic DNA in systemic organs indicates hematogenous dissemination from the gingival pockets. Bacterial infection did not alter serum lipid fractions or serum amyloid A levels and did not induce aortic atherosclerotic plaque. This is the first study examining the causal role of periodontal bacteria in induction of ASVD in LDLRnull mice.


Assuntos
Aterosclerose/genética , Aterosclerose/microbiologia , Interações Hospedeiro-Patógeno , Doenças Periodontais/genética , Doenças Periodontais/microbiologia , Receptores de LDL/deficiência , Perda do Osso Alveolar/etiologia , Perda do Osso Alveolar/patologia , Animais , Anticorpos Antibacterianos/imunologia , Aterosclerose/patologia , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Placa Dentária/microbiologia , Placa Dentária/patologia , Gengiva/metabolismo , Gengiva/microbiologia , Gengiva/patologia , Lipídeos/sangue , Masculino , Camundongos , Camundongos Knockout , Doenças Periodontais/patologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
12.
PLoS One ; 10(11): e0143291, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26619277

RESUMO

Periodontal disease (PD) develops from a synergy of complex subgingival oral microbiome, and is linked to systemic inflammatory atherosclerotic vascular disease (ASVD). To investigate how a polybacterial microbiome infection influences atherosclerotic plaque progression, we infected the oral cavity of ApoE null mice with a polybacterial consortium of 4 well-characterized periodontal pathogens, Porphyromonas gingivalis, Treponema denticola, Tannerealla forsythia and Fusobacterium nucleatum, that have been identified in human atherosclerotic plaque by DNA screening. We assessed periodontal disease characteristics, hematogenous dissemination of bacteria, peripheral T cell response, serum inflammatory cytokines, atherosclerosis risk factors, atherosclerotic plaque development, and alteration of aortic gene expression. Polybacterial infections have established gingival colonization in ApoE null hyperlipidemic mice and displayed invasive characteristics with hematogenous dissemination into cardiovascular tissues such as the heart and aorta. Polybacterial infection induced significantly higher levels of serum risk factors oxidized LDL (p < 0.05), nitric oxide (p < 0.01), altered lipid profiles (cholesterol, triglycerides, Chylomicrons, VLDL) (p < 0.05) as well as accelerated aortic plaque formation in ApoE null mice (p < 0.05). Periodontal microbiome infection is associated with significant decreases in Apoa1, Apob, Birc3, Fga, FgB genes that are associated with atherosclerosis. Periodontal infection for 12 weeks had modified levels of inflammatory molecules, with decreased Fas ligand, IL-13, SDF-1 and increased chemokine RANTES. In contrast, 24 weeks of infection induced new changes in other inflammatory molecules with reduced KC, MCSF, enhancing GM-CSF, IFNγ, IL-1ß, IL-13, IL-4, IL-13, lymphotactin, RANTES, and also an increase in select inflammatory molecules. This study demonstrates unique differences in the host immune response to a polybacterial periodontal infection with atherosclerotic lesion progression in a mouse model.


Assuntos
Periodontite Agressiva/microbiologia , Apolipoproteínas E/genética , Aterosclerose/etiologia , Microbiota , Periodontite Agressiva/complicações , Animais , Quimiocinas/genética , Quimiocinas/metabolismo , Fusobacterium nucleatum/isolamento & purificação , Inflamação/etiologia , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Camundongos , Boca/microbiologia , Porphyromonas gingivalis/isolamento & purificação , Linfócitos T/metabolismo , Treponema denticola/isolamento & purificação
13.
Infect Immun ; 83(12): 4582-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26371120

RESUMO

The American Heart Association supports an association between periodontal diseases and atherosclerosis but not a causal association. This study explores the use of the integrin ß6(-/-) mouse model to study the causality. We investigated the ability of a polymicrobial consortium of Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia, and Fusobacterium nucleatum to colonize the periodontium and induce local and systemic inflammatory responses. Polymicrobially infected Itgß6(-/-) mice demonstrate greater susceptibility to gingival colonization/infection, with severe gingival inflammation, apical migration of the junctional epithelium, periodontal pocket formation, alveolar bone resorption, osteoclast activation, bacterial invasion of the gingiva, a greater propensity for the bacteria to disseminate hematogenously, and a strong splenic T cell cytokine response. Levels of atherosclerosis risk factors, including serum nitric oxide, oxidized low-density lipoprotein, serum amyloid A, and lipid peroxidation, were significantly altered by polybacterial infection, demonstrating an enhanced potential for atherosclerotic plaque progression. Aortic gene expression revealed significant alterations in specific Toll-like receptor (TLR) and nucleotide-binding domain- and leucine-rich-repeat-containing receptor (NLR) pathway genes in response to periodontal bacterial infection. Histomorphometry of the aorta demonstrated larger atherosclerotic plaques in Itgß6(-/-) mice than in wild-type (WT) mice but no significant difference in atherosclerotic plaque size between mice with polybacterial infection and mice with sham infection. Fluorescence in situ hybridization demonstrated active invasion of the aortic adventitial layer by P. gingivalis. Our observations suggest that polybacterial infection elicits distinct aortic TLR and inflammasome signaling and significantly increases local aortic oxidative stress. These results are the first to demonstrate the mechanism of the host aortic inflammatory response induced by polymicrobial infection with well-characterized periodontal pathogens.


Assuntos
Túnica Adventícia/patologia , Antígenos de Neoplasias/imunologia , Aorta/patologia , Aterosclerose/complicações , Integrinas/imunologia , Periodontite/complicações , Placa Aterosclerótica/complicações , Túnica Adventícia/imunologia , Túnica Adventícia/microbiologia , Animais , Antígenos de Neoplasias/genética , Aorta/imunologia , Aorta/microbiologia , Aterosclerose/imunologia , Aterosclerose/microbiologia , Aterosclerose/patologia , Bacteroidetes/crescimento & desenvolvimento , Bacteroidetes/imunologia , Bacteroidetes/patogenicidade , Reabsorção Óssea , Modelos Animais de Doenças , Fusobacterium nucleatum/crescimento & desenvolvimento , Fusobacterium nucleatum/imunologia , Fusobacterium nucleatum/patogenicidade , Expressão Gênica , Gengiva/imunologia , Gengiva/microbiologia , Gengiva/patologia , Hibridização in Situ Fluorescente , Inflamassomos , Integrinas/deficiência , Integrinas/genética , Lipoproteínas LDL/genética , Lipoproteínas LDL/imunologia , Camundongos , Camundongos Knockout , Consórcios Microbianos , Periodontite/imunologia , Periodontite/microbiologia , Periodontite/patologia , Periodonto/imunologia , Periodonto/microbiologia , Periodonto/patologia , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/microbiologia , Placa Aterosclerótica/patologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/patogenicidade , Treponema denticola/crescimento & desenvolvimento , Treponema denticola/imunologia , Treponema denticola/patogenicidade
14.
PLoS One ; 10(6): e0129795, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26079509

RESUMO

The American Heart Association supports an association between periodontal disease (PD) and atherosclerotic vascular disease (ASVD) but does not as of yet support a causal relationship. Recently, we have shown that major periodontal pathogens Porphyromonas gingivalis and Treponema denticola are causally associated with acceleration of aortic atherosclerosis in ApoEnull hyperlipidemic mice. The aim of this study was to determine if oral infection with another significant periodontal pathogen Fusobacterium nucleatum can accelerate aortic inflammation and atherosclerosis in the aortic artery of ApoEnull mice. ApoEnull mice (n = 23) were orally infected with F. nucleatum ATCC 49256 and euthanized at 12 and 24 weeks. Periodontal disease assessments including F. nucleatum oral colonization, gingival inflammation, immune response, intrabony defects, and alveolar bone resorption were evaluated. Systemic organs were evaluated for infection, aortic sections were examined for atherosclerosis, and inflammatory markers were measured. Chronic oral infection established F. nucleatum colonization in the oral cavity, induced significant humoral IgG (P=0.0001) and IgM (P=0.001) antibody response (12 and 24 weeks), and resulted in significant (P=0.0001) alveolar bone resorption and intrabony defects. F. nucleatum genomic DNA was detected in systemic organs (heart, aorta, liver, kidney, lung) indicating bacteremia. Aortic atherosclerotic plaque area was measured and showed a local inflammatory infiltrate revealed the presence of F4/80+ macrophages and CD3+ T cells. Vascular inflammation was detected by enhanced systemic cytokines (CD30L, IL-4, IL-12), oxidized LDL and serum amyloid A, as well as altered serum lipid profile (cholesterol, triglycerides, chylomicrons, VLDL, LDL, HDL), in infected mice and altered aortic gene expression in infected mice. Despite evidence for systemic infection in several organs and modulation of known atherosclerosis risk factors, aortic atherosclerotic lesions were significantly reduced after F. nucleatum infection suggesting a potential protective function for this member of the oral microbiota.


Assuntos
Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Fusobacterium nucleatum/fisiologia , Deleção de Genes , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/microbiologia , Animais , Aorta/patologia , Biomarcadores/metabolismo , Citocinas/metabolismo , Progressão da Doença , Doenças da Gengiva/microbiologia , Imunidade Humoral , Inflamação/metabolismo , Masculino , Camundongos , Boca/microbiologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Fatores de Risco
15.
Pathog Dis ; 73(3)2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25663343

RESUMO

Tannerella forsythia is a Gram-negative anaerobic organism that inhabits the subgingival cavity and initiates connective tissue destruction and alveolar bone resorption in periodontal disease (PD). PD is a chronic immunoinflammatory disease and has been linked to several systemic diseases including atherosclerosis. This study evaluated the effects of a chronic oral infection with T. forsythia ATCC 43037 on the induction of PD, inflammatory markers and atherosclerosis risk factors in hyperlipidemic ApoE(null) mice. Mice were orally infected for 12 and 24 weeks prior to euthanasia. Bacterial colonization of the oral cavity and bacteremia was confirmed via isolation of genomic DNA from oral plaque and tissues. Oral infection elicited significantly elevated levels of serum IgG and IgM antibodies and alveolar bone resorption compared to control mice. Tannerella forsythia-infected mice had increased serum amyloid A, and significantly reduced serum nitric oxide when compared to controls. Tannerella forsythia chronic infection also significantly increased serum lipoproteins suggesting altered cholesterol metabolism and potential for aortic inflammation. Despite enhanced acute phase reactants and altered lipid profiles, T. forsythia infection was associated with decreased aortic plaque. This study investigates the potential of a known periodontal bacterial pathogen found in atherosclerotic plaque in humans to accelerate atherosclerosis in hyperlipdemic mice.


Assuntos
Aterosclerose/microbiologia , Bacteroidetes/imunologia , Infecções por Bactérias Gram-Negativas/complicações , Infecções por Bactérias Gram-Negativas/patologia , Inflamação/microbiologia , Doenças Periodontais/complicações , Doenças Periodontais/patologia , Perda do Osso Alveolar/etiologia , Perda do Osso Alveolar/patologia , Animais , Anticorpos Antibacterianos/sangue , Aterosclerose/patologia , Bacteriemia/microbiologia , Doença Crônica , Infecções por Bactérias Gram-Negativas/imunologia , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Inflamação/patologia , Lipoproteínas/sangue , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico/sangue , Doenças Periodontais/imunologia , Fatores de Risco , Proteína Amiloide A Sérica/análise
16.
J Periodontol ; 86(1): 137-45, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25101489

RESUMO

BACKGROUND: Periodontitis is a chronic, polymicrobial inflammatory disease that degrades connective tissue and alveolar bone and results in tooth loss. Oxidative stress has been linked to the onset of periodontal tissue breakdown and systemic inflammation, and the success of antiresorptive treatments will rely on how effectively they can ameliorate periodontal disease-induced oxidative stress during oral infection. METHODS: Rats were infected with polybacterial inoculum consisting of Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia, as an oral lavage every other week for 12 weeks. Daily subcutaneous injections of enoxacin, bis-enoxacin, alendronate, or doxycycline were administered for 6 weeks after 6 weeks of polybacterial infection in rats. The serum levels of oxidative stress parameters and antioxidant enzymes, including glutathione peroxidase, superoxide dismutase, and catalase, were evaluated in each of the infected, treated, and sham-infected rats. RESULTS: Rats infected with the periodontal pathogens displayed a five-fold increase in the oxidative stress index compared with controls as a result of increased levels of serum oxidants and decreases in total antioxidant activity. The overall decrease in antioxidant activity occurred despite increases in three important antioxidant enzymes, suggesting an imbalance between antioxidant macromolecules/small molecules production and antioxidant enzyme levels. Surprisingly, the bone-targeted antiresorptives bis-enoxacin and alendronate inhibited increases in oxidative stress caused by periodontitis. Bis-enoxacin, which has both antiresorptive and antibiotic activities, was more effective than alendronate, which acts only as an antiresorptive. CONCLUSION: To the best of the authors' knowledge, this is the first study to demonstrate that the increased oxidative stress induced by periodontal infection in rats can be ameliorated by bone-targeted antiresorptives.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Estresse Oxidativo/fisiologia , Periodontite/metabolismo , Alendronato/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Antioxidantes/análise , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroides/metabolismo , Catalase/sangue , Coinfecção/microbiologia , Doxiciclina/uso terapêutico , Enoxacino/uso terapêutico , Feminino , Sequestradores de Radicais Livres/sangue , Glutationa Peroxidase/sangue , Infecções por Bactérias Gram-Negativas/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Oxidantes/sangue , Estresse Oxidativo/efeitos dos fármacos , Periodontite/microbiologia , Periodontite/prevenção & controle , Porphyromonas gingivalis/fisiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/sangue , Treponema denticola/fisiologia
17.
PLoS One ; 9(5): e97811, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24836175

RESUMO

Atherosclerotic vascular disease is a leading cause of myocardial infarction and cerebrovascular accident, and independent associations with periodontal disease (PD) are reported. PD is caused by polymicrobial infections and aggressive immune responses. Genomic DNA of Porphyromonas gingivalis, the best-studied bacterial pathogen associated with severe PD, is detected within atherosclerotic plaque. We examined causal relationships between chronic P. gingivalis oral infection, PD, and atherosclerosis in hyperlipidemic ApoEnull mice. ApoEnull mice (n = 24) were orally infected with P. gingivalis for 12 and 24 weeks. PD was assessed by standard clinical measurements while the aorta was examined for atherosclerotic lesions and inflammatory markers by array. Systemic inflammatory markers serum amyloid A, nitric oxide, and oxidized low-density lipoprotein were analyzed. P. gingivalis infection elicited specific antibodies and alveolar bone loss. Fluorescent in situ hybridization detected viable P. gingivalis within oral epithelium and aorta, and genomic DNA was detected within systemic organs. Aortic plaque area was significantly increased in P. gingivalis-infected mice at 24 weeks (P<0.01). Aortic RNA and protein arrays indicated a strong Th2 response. Chronic oral infection with P. gingivalis results in a specific immune response, significant increases in oral bone resorption, aortic inflammation, viable bacteria in oral epithelium and aorta, and plaque development.


Assuntos
Aorta/microbiologia , Aterosclerose/etiologia , Infecções por Bacteroidaceae/complicações , Boca/microbiologia , Periodontite/complicações , Porphyromonas gingivalis , Animais , Aterosclerose/imunologia , Aterosclerose/microbiologia , Infecções por Bacteroidaceae/imunologia , Doença Crônica , Masculino , Camundongos , Periodontite/imunologia , Periodontite/microbiologia , Porphyromonas gingivalis/imunologia , Fatores de Risco , Transcriptoma
18.
PLoS One ; 9(3): e92119, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24638087

RESUMO

Periodontal diseases are multifactorial, caused by polymicrobial subgingival pathogens, including Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia. Chronic periodontal infection results in inflammation, destruction of connective tissues, periodontal ligament, and alveolar bone resorption, and ultimately tooth loss. Enoxacin and a bisphosphonate derivative of enoxacin (bis-enoxacin) inhibit osteoclast formation and bone resorption and also contain antibiotic properties. Our study proposes that enoxacin and/or bis-enoxacin may be useful in reducing alveolar bone resorption and possibly bacterial colonization. Rats were infected with 10(9) cells of polymicrobial inoculum consisting of P. gingivalis, T. denticola, and T. forsythia, as an oral lavage every other week for twelve weeks. Daily subcutaneous injections of enoxacin (5 mg/kg/day), bis-enoxacin (5, 25 mg/kg/day), alendronate (1, 10 mg/kg/day), or doxycycline (5 mg/day) were administered after 6 weeks of polymicrobial infection. Periodontal disease parameters, including bacterial colonization/infection, immune response, inflammation, alveolar bone resorption, and systemic spread, were assessed post-euthanasia. All three periodontal pathogens colonized the rat oral cavity during polymicrobial infection. Polymicrobial infection induced an increase in total alveolar bone resorption, intrabony defects, and gingival inflammation. Treatment with bis-enoxacin significantly decreased alveolar bone resorption more effectively than either alendronate or doxycycline. Histologic examination revealed that treatment with bis-enoxacin and enoxacin reduced gingival inflammation and decreased apical migration of junctional epithelium. These data support the hypothesis that bis-enoxacin and enoxacin may be useful for the treatment of periodontal disease.


Assuntos
Perda do Osso Alveolar/tratamento farmacológico , Perda do Osso Alveolar/etiologia , Enoxacino/uso terapêutico , Periodontite/induzido quimicamente , Periodontite/complicações , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/microbiologia , Animais , Contagem de Colônia Microbiana , DNA Bacteriano/genética , Placa Dentária/sangue , Placa Dentária/complicações , Placa Dentária/imunologia , Placa Dentária/microbiologia , Enoxacino/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade Humoral/efeitos dos fármacos , Imunidade Humoral/imunologia , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Mandíbula/efeitos dos fármacos , Mandíbula/microbiologia , Mandíbula/patologia , Periodontite/imunologia , Periodontite/microbiologia , Periodonto/efeitos dos fármacos , Periodonto/microbiologia , Periodonto/patologia , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Treponema/efeitos dos fármacos , Treponema/crescimento & desenvolvimento
19.
Infect Immun ; 82(5): 1959-67, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24566627

RESUMO

Treponema denticola is a predominantly subgingival oral spirochete closely associated with periodontal disease and has been detected in atherosclerosis. This study was designed to evaluate causative links between periodontal disease induced by chronic oral T. denticola infection and atherosclerosis in hyperlipidemic ApoE(-/-) mice. ApoE(-/-) mice (n = 24) were orally infected with T. denticola ATCC 35404 and were euthanized after 12 and 24 weeks. T. denticola genomic DNA was detected in oral plaque samples, indicating colonization of the oral cavity. Infection elicited significantly (P = 0.0172) higher IgG antibody levels and enhanced intrabony defects than sham infection. T. denticola-infected mice had higher levels of horizontal alveolar bone resorption than sham-infected mice and an associated significant increase in aortic plaque area (P ≤ 0.05). Increased atherosclerotic plaque correlated with reduced serum nitric oxide (NO) levels and increased serum-oxidized low-density lipoprotein (LDL) levels compared to those of sham-infected mice. T. denticola infection altered the expression of genes known to be involved in atherosclerotic development, including the leukocyte/endothelial cell adhesion gene (Thbs4), the connective tissue growth factor gene (Ctgf), and the selectin-E gene (Sele). Fluorescent in situ hybridization (FISH) revealed T. denticola clusters in both gingival and aortic tissue of infected mice. This is the first study examining the potential causative role of chronic T. denticola periodontal infection and vascular atherosclerosis in vivo in hyperlipidemic ApoE(-/-) mice. T. denticola is closely associated with periodontal disease and the rapid progression of atheroma in ApoE(-/-) mice. These studies confirm a causal link for active oral T. denticola infection with both atheroma and periodontal disease.


Assuntos
Aorta/microbiologia , Apolipoproteínas E/metabolismo , Aterosclerose/etiologia , Infecções por Bactérias Gram-Negativas/complicações , Doenças Periodontais/etiologia , Treponema denticola/fisiologia , Animais , Anticorpos Antibacterianos/sangue , Apolipoproteínas E/genética , Aterosclerose/microbiologia , Reabsorção Óssea/microbiologia , Gengivite/complicações , Gengivite/microbiologia , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Knockout , Doenças Periodontais/microbiologia , Fatores de Risco
20.
PLoS One ; 8(2): e57178, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23451182

RESUMO

Periodontal disease (PD) and atherosclerosis are both polymicrobial and multifactorial and although observational studies supported the association, the causative relationship between these two diseases is not yet established. Polymicrobial infection-induced periodontal disease is postulated to accelerate atherosclerotic plaque growth by enhancing atherosclerotic risk factors of orally infected Apolipoprotein E deficient (ApoE(null)) mice. At 16 weeks of infection, samples of blood, mandible, maxilla, aorta, heart, spleen, and liver were collected, analyzed for bacterial genomic DNA, immune response, inflammation, alveolar bone loss, serum inflammatory marker, atherosclerosis risk factors, and aortic atherosclerosis. PCR analysis of polymicrobial-infected (Porphyromonas gingivalis [P. gingivalis], Treponema denticola [T. denticola], and Tannerella forsythia [T. forsythia]) mice resulted in detection of bacterial genomic DNA in oral plaque samples indicating colonization of the oral cavity by all three species. Fluorescent in situ hybridization detected P. gingivalis and T. denticola within gingival tissues of infected mice and morphometric analysis showed an increase in palatal alveolar bone loss (p<0.0001) and intrabony defects suggesting development of periodontal disease in this model. Polymicrobial-infected mice also showed an increase in aortic plaque area (p<0.05) with macrophage accumulation, enhanced serum amyloid A, and increased serum cholesterol and triglycerides. A systemic infection was indicated by the detection of bacterial genomic DNA in the aorta and liver of infected mice and elevated levels of bacterial specific IgG antibodies (p<0.0001). This study was a unique effort to understand the effects of a polymicrobial infection with P. gingivalis, T. denticola and T. forsythia on periodontal disease and associated atherosclerosis in ApoE(null) mice.


Assuntos
Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/microbiologia , Infecções Bacterianas/microbiologia , Hiperlipidemias/microbiologia , Doenças Periodontais/microbiologia , Animais , Aterosclerose/complicações , Infecções Bacterianas/complicações , Sequência de Bases , Primers do DNA , DNA Bacteriano/análise , Placa Dentária/microbiologia , Hiperlipidemias/complicações , Hibridização in Situ Fluorescente , Camundongos , Camundongos Knockout , Doenças Periodontais/complicações , Reação em Cadeia da Polimerase , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/isolamento & purificação , Treponema denticola/genética , Treponema denticola/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA