Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Lab Chip ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874483

ABSTRACT

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.

2.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731952

ABSTRACT

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.


Subject(s)
Acrolein , Intestinal Mucosa , NF-E2-Related Factor 2 , Nitric Oxide , Phosphatidylinositol 3-Kinases , Porphyromonas gingivalis , Proto-Oncogene Proteins c-akt , Signal Transduction , NF-E2-Related Factor 2/metabolism , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Porphyromonas gingivalis/pathogenicity , Phosphatidylinositol 3-Kinases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Nitric Oxide/metabolism , Cell Line , Lipopolysaccharides , Oxidative Stress/drug effects , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Toll-Like Receptor 4/metabolism , Reactive Oxygen Species/metabolism , Cytokines/metabolism
3.
Biophys J ; 122(18): 3630-3645, 2023 09 19.
Article in English | MEDLINE | ID: mdl-36617192

ABSTRACT

Epithelial cells lining a gland and cells grown in a soft extracellular matrix polarize with apical proteins exposed to the lumen and basal proteins in contact with the extracellular matrix. Alterations to polarity, including an apical-out polarity, occur in human cancers. Although some aberrant polarity states may result from altered protein trafficking, recent observations of an extraordinary tissue-level inside-out unfolding suggest an alternative pathway for altered polarity. Because mechanical alterations are common in human cancer, including an upregulation of RhoA-mediated actomyosin tension in acinar epithelia, we explored whether perturbing mechanical homeostasis could cause apical-out eversion. Acinar eversion was robustly induced by direct activation of RhoA in normal and tumor epithelial acini, or indirect activation of RhoA through blockage of ß1-integrins, disruption of the LINC complex, oncogenic Ras activation, or Rac1 inhibition. Furthermore, laser ablation of a portion of the untreated acinus was sufficient to induce eversion. Analyses of acini revealed high curvature and low phosphorylated myosin in the apical cell surfaces relative to the basal surfaces. A vertex-based mathematical model that balances tension at cell-cell interfaces revealed a fivefold greater basal cell surface tension relative to the apical cell surface tension. The model suggests that the difference in surface energy between the apical and basal surfaces is the driving force for acinar eversion. Our findings raise the possibility that a loss of mechanical homeostasis may cause apical-out polarity states in human cancers.


Subject(s)
Epithelial Cells , Extracellular Matrix , Humans , Cell Membrane/metabolism , Integrin beta1/metabolism , Cell Polarity/physiology
4.
J Alzheimers Dis Rep ; 6(1): 577-587, 2022.
Article in English | MEDLINE | ID: mdl-36275415

ABSTRACT

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.

5.
J Alzheimers Dis ; 82(3): 1259-1275, 2021.
Article in English | MEDLINE | ID: mdl-34151813

ABSTRACT

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.


Subject(s)
Alzheimer Disease/physiopathology , Bacteroidaceae Infections/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Obesity/physiopathology , Porphyromonas gingivalis , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Animals , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/psychology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/psychology , Mice , Mice, Transgenic , Obesity/genetics , Obesity/psychology
6.
J Cell Physiol ; 236(8): 5715-5724, 2021 08.
Article in English | MEDLINE | ID: mdl-33400284

ABSTRACT

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.


Subject(s)
Actomyosin/metabolism , Fibroblasts/metabolism , Osteoprotegerin/metabolism , Periodontal Ligament/drug effects , Animals , Cytokines/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Periodontal Ligament/metabolism , Porphyromonas gingivalis/metabolism
7.
J Periodontol ; 92(7): 54-65, 2021 07.
Article in English | MEDLINE | ID: mdl-33128253

ABSTRACT

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.


Subject(s)
Nitric Oxide , Porphyromonas gingivalis , Endothelial Cells , Endothelium, Vascular , Glycogen Synthase Kinase 3 beta , Humans , NF-E2-Related Factor 2
8.
J Alzheimers Dis Rep ; 4(1): 501-511, 2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33532698

ABSTRACT

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.

9.
J Oral Microbiol ; 13(1): 1849493, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33391626

ABSTRACT

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.

10.
Pathog Dis ; 76(8)2018 11 01.
Article in English | MEDLINE | ID: mdl-30351354

ABSTRACT

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.


Subject(s)
Atherosclerosis/pathology , Bacterial Infections/pathology , Coinfection/pathology , Immunity, Innate , Periodontitis/pathology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , Animal Structures/microbiology , Animals , Atherosclerosis/immunology , Bacterial Infections/immunology , Coinfection/immunology , Female , Fusobacterium nucleatum/immunology , Male , Mice, Knockout , Periodontitis/immunology , Porphyromonas gingivalis/immunology , Signal Transduction , Tannerella forsythia/immunology , Treponema denticola/immunology
11.
Open Biol ; 8(9)2018 08 16.
Article in English | MEDLINE | ID: mdl-30209038

ABSTRACT

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.


Subject(s)
Mechanotransduction, Cellular , Periodontium/cytology , Cell Proliferation , Epithelial Cells/cytology , Fibroblasts/cytology , Humans , Stem Cells/cytology
12.
J Alzheimers Dis ; 60(2): 359-369, 2017.
Article in English | MEDLINE | ID: mdl-28800332

ABSTRACT

The polymicrobial dysbiotic subgingival biofilm microbes associated with periodontal disease appear to contribute to developing pathologies in distal body sites, including the brain. This study examined oxidative stress, in the form of increased protein carbonylation and oxidative protein damage, in the tumor necrosis factor-α (TNF-α) transgenic mouse that models inflammatory TNF-α excess during bacterial infection; and in the apolipoprotein knockout (ApoE-/-) mouse brains, following Porphyromonas gingivalis gingival monoinfection. Following 2,4-dinitrophenylhydrazine derivatization, carbonyl groups were detected in frontal lobe brain tissue lysates by immunoblotting and immunohistochemical analysis of fixed tissue sections from the frontotemporal lobe and the hippocampus. Immunoblot analysis confirmed the presence of variable carbonyl content and oxidative protein damage in all lysates, with TNF-α transgenic blots exhibiting increased protein carbonyl content, with consistently prominent bands at 25 kDa (p = 0.0001), 43 kDa, and 68 kDa, over wild-type mice. Compared to sham-infected ApoE-/- mouse blots, P. gingivalis-infected brain tissue blots demonstrated the greatest detectable protein carbonyl content overall, with numerous prominent bands at 25 kDa (p = 0.001) and 43 kDa (p = 0.0001) and an exclusive band to this group between 30-43 kDa* (p = 0.0001). In addition, marked immunostaining was detected exclusively in the microvasculature in P. gingivalis-infected hippocampal tissue sections, compared to sham-infected, wild-type, and TNF-α transgenic mice. This study revealed that the hippocampal microvascular structure of P. gingivalis-infected ApoE-/- mice possesses elevated oxidative stress levels, resulting in the associated tight junction proteins being susceptible to increased oxidative/proteolytic degradation, leading to a loss of functional integrity.


Subject(s)
Apolipoproteins E/deficiency , Bacteroidaceae Infections/physiopathology , Microvessels/pathology , Oxidative Stress/genetics , Porphyromonas gingivalis/pathogenicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Apolipoproteins E/genetics , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/virology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/virology , Phenylhydrazines/metabolism , Protein Carbonylation/genetics , Protein Carbonylation/physiology , Tumor Necrosis Factor-alpha/genetics
13.
Int J Mol Sci ; 18(4)2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28425970

ABSTRACT

Altered microRNA expression is implicated in cardiovascular diseases. Our objective was to determine microRNA signatures in thoracic aortic aneurysms (TAAs) and abdominal aortic aneurysms (AAAs) compared with control non-aneurysmal aortic specimens. We evaluated the expression of fifteen selected microRNA in human TAA and AAA operative specimens compared to controls. We observed significant upregulation of miR-221 and downregulation of miR-1 and -133 in TAA specimens. In contrast, upregulation of miR-146a and downregulation of miR-145 and -331-3p were found only for AAA specimens. Upregulation of miR-126 and -486-5p and downregulation of miR-30c-2*, -155, and -204 were observed in specimens of TAAs and AAAs. The data reveal microRNA expression signatures unique to aneurysm location and common to both thoracic and abdominal pathologies. Thus, changes in miR-1, -29a, -133a, and -221 are involved in TAAs and miR-145, -146, and -331-3p impact AAAs. This work validates prior studies on microRNA expression in aneurysmal diseases.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Thoracic/genetics , Genetic Predisposition to Disease , MicroRNAs/genetics , Aged , Aortic Aneurysm, Abdominal/diagnosis , Aortic Aneurysm, Thoracic/diagnosis , Case-Control Studies , Cluster Analysis , Computational Biology/methods , Female , Gene Expression Profiling , Genetic Association Studies , Humans , Male , Middle Aged , Risk Factors , Transcriptome
14.
J Oral Microbiol ; 9(1): 1270602, 2017.
Article in English | MEDLINE | ID: mdl-28326151

ABSTRACT

This study explored the origin of age-related granules in the apolipoprotein E gene knockout (ApoE-/-) B6 background mice brains following chronic gingival infection with Porphyromonas gingivalis for 24 weeks. Intracerebral localization of P. gingivalis was detected by fluorescence in situ hybridization (FISH) and its protease by immunohistochemistry. The age-related granules were observed by periodic acid-Schiff (PAS), silver impregnation, and immunostaining. FISH showed intracerebral dissemination of P. gingivalis cells (p = 0.001). PAS and silver impregnation demonstrated the presence of larger inclusions restricted to the CA1, CA2, and dentate gyrus sectors of the hippocampus. A specific monoclonal antibody to bacterial peptidoglycan detected clusters of granules with variable sizes in mice brains infected with P. gingivalis (p = 0.004), and also highlighted areas of diffuse punctate staining equating to physical tissue damage. Mouse immunoglobulin G was observed in the capillaries of the cerebral parenchyma of all P. gingivalis-infected brains (p = 0.001), and on pyramidal neurons in some severely affected mice, compared with the sham-infected mice. Gingipains was also observed in microvessels of the hippocampus in the infected mice. This study supports the possibility of early appearance of age-related granules in ApoE-/- mice following inflammation-mediated tissue injury, accompanied by loss of cerebral blood-brain barrier integrity.

15.
Pathog Dis ; 75(1)2017 01 01.
Article in English | MEDLINE | ID: mdl-28104616

ABSTRACT

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.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/microbiology , Host-Pathogen Interactions , Periodontal Diseases/genetics , Periodontal Diseases/microbiology , Receptors, LDL/deficiency , Alveolar Bone Loss/etiology , Alveolar Bone Loss/pathology , Animals , Antibodies, Bacterial/immunology , Atherosclerosis/pathology , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Infections/pathology , Dental Plaque/microbiology , Dental Plaque/pathology , Gingiva/metabolism , Gingiva/microbiology , Gingiva/pathology , Lipids/blood , Male , Mice , Mice, Knockout , Periodontal Diseases/pathology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
16.
J Periodontol ; 88(2): e32-e41, 2017 02.
Article in English | MEDLINE | ID: mdl-27786620

ABSTRACT

BACKGROUND: Peri-implantitis is a complex polymicrobial biofilm-induced inflammatory osteolytic gingival infection that results in orofacial implant failures. To the best knowledge of the authors, there are no preclinical in vivo studies in implant dentistry that have investigated the inflammatory response to known microbial biofilms observed in humans. The aim of this study is to develop a novel peri-implant rat model using an established model of polymicrobial periodontitis. METHODS: Wistar rats were used for the study of experimental peri-implantitis. One month after extraction of maxillary first molars, a titanium mini-implant was inserted. Two months after implant healing, implants were uncovered, and abutment fixing was done using cyanoacrylate to prevent abutment loosening. Rats were separated into two groups (group A: polymicrobial-infected and group B: sham-infected). One week after healing of abutments, rats were infected with Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia for 12 weeks. Bacterial colonization, bone resorption, and implant inflammation were evaluated by polymerase chain reaction (PCR), microcomputed tomography, and histology, respectively. RESULTS: Three rats with four implants in the infection group and two rats with three implants in the sham-infection group were analyzed. PCR analysis revealed presence of bacterial genomic DNA, and infection elicited significant immunoglobulin (Ig)G and IgM antibody responses, indicating bacterial colonization/infection around implants. Infection induced an enhanced mean distance from implant platform to the first bone-to-implant contact, extensive peri-implantitis with advanced bone resorption, and extensive inflammation with granulation tissue and polymorphonuclear leukocytes. CONCLUSIONS: To the best knowledge of the authors, this is the first study to develop a novel rat model of polymicrobial peri-implantitis. With modifications to improve implant retention it could offer significant advantages for studies of initiation and progression of peri-implantitis.


Subject(s)
Peri-Implantitis/microbiology , Animals , Biofilms , Dental Implantation, Endosseous , Disease Models, Animal , Polymerase Chain Reaction , Porphyromonas gingivalis/pathogenicity , Rats , Rats, Wistar , Tannerella forsythia/pathogenicity , Treponema denticola/pathogenicity , X-Ray Microtomography
17.
Arthritis Res Ther ; 18(1): 161, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27405639

ABSTRACT

BACKGROUND: It has been previously hypothesized that oral microbes may be an etiological link between rheumatoid arthritis (RA) and periodontal disease. However, the mechanistic basis of this association is incompletely understood. Here, we investigated the role of periodontal bacteria in induction of joint inflammation in collagen-induced arthritis (CIA) in B10.RIII mice. METHODS: CIA-prone B10.RIII mice were infected orally with a polybacterial mixture of Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia for 24 weeks before induction of CIA. The ability of polybacterial mixture to colonize the periodontium and induce systemic response, horizontal alveolar bone resorption in infected B10.RIII mice was investigated. Arthritis incidence, severity of joint inflammation, pannus formation, skeletal damage, hematogenous dissemination of the infection, matrix metalloproteinase 3 (MMP3) levels, and interleukin-17 expression levels were evaluated. RESULTS: B10.RIII mice had gingival colonization with all three bacteria, higher levels of anti-bacterial immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies, significant alveolar bone resorption, and hematogenous dissemination of P. gingivalis to synovial joints. Infected B10.RIII mice had more severe arthritis, and higher serum matrix metalloproteinase 3 levels and activity. Histopathological analysis showed increased inflammatory cell infiltration, destruction of articular cartilage, erosions, and pannus formation. Additionally, involved joints showed had expression levels of interleukin-17. CONCLUSION: These findings demonstrate that physical presence of periodontal bacteria in synovial joints of B10.RIII mice with collagen-induced arthritis is associated with arthritis exacerbation, and support the hypothesis that oral bacteria, specifically P. gingivalis, play a significant role in augmenting autoimmune arthritis due to their intravascular dissemination to the joints.


Subject(s)
Arthritis, Experimental/microbiology , Arthritis, Experimental/pathology , Periodontitis/complications , Synovial Membrane/microbiology , Animals , Mice , Periodontitis/microbiology , Periodontium/microbiology
18.
J Alzheimers Dis ; 51(4): 935-48, 2016.
Article in English | MEDLINE | ID: mdl-26923007

ABSTRACT

The primary goal of advancement in clinical services is to provide a health care system that enhances an individual's quality of life. Incidence of diabetes mellitus, cardiovascular disease, and associated dementia coupled with the advancing age of the population, have led to an increase in the worldwide challenge to the healthcare system. In order to overcome these challenges, prior knowledge of common, reliable risk factors and their effectors is essential. Oral health constitutes one such relatively unexplored but indispensable risk factor for aforementioned co-morbidities, in the form of poor oral hygiene and tooth loss during aging. Behavioral traits such as low education, smoking, poor diet, neglect of oral health, lack of exercise, and hypertension are few of the risk factors that are shared commonly among these conditions. In addition, common genetic susceptibility traits such as the apolipoprotein E gene, together with an individual's lifestyle can also influence the development of co-morbidities such as periodontitis, atherosclerosis/stroke, diabetes, and Alzheimer's disease. This review specifically addresses the susceptibility of apolipoprotein E gene allele 4 as the plausible commonality for the etiology of co-morbidities that eventually result from periodontal diseases and ultimately progress to dementia.


Subject(s)
Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Periodontal Diseases/epidemiology , Periodontal Diseases/genetics , Aging/genetics , Animals , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Comorbidity , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Humans , Mice
19.
Anaerobe ; 38: 70-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26481834

ABSTRACT

Periodontal disease initiated by subgingival pathogens is linked with diminished secretion of saliva, and implies pathogenic bacteria dissemination to or affects secondary sites such as the salivary glands. MicroRNAs activated in response to bacteria may modulate immune responses against pathogens. Therefore, Sprague-Dawley rats were infected by oral lavage consisting of polymicrobial inocula, namely Porphyromonas gingivalis, Tannerella forsythia, and Treponema denticola, or sham-infected for 12 weeks (n = 6). We quantified inflammatory miRNA expression levels of miRNA-132, miR-146a, and miR-155 at secondary sites to the primary infection of the gingiva, including submandibular salivary glands, lacrimal glands, and pancreas. The presence of bacteria was detected in situ at secondary sites. Infected rat gingiva showed increased relative expression of miR-155. In contrast, miRNA-155 expression was decreased in submandibular salivary glands, along with positive identification of P. gingivalis in 2/6 and T. denticola in 1/6 rat salivary glands. Furthermore, miRNA-132 and miRNA-146a were significantly decreased in the pancreas of infected rats. This study is the first to show primary periodontal infections can alter miRNA profiles in secondary sites such as the salivary gland and pancreas. Whether these alterations contribute to pathologies of salivary glands in Sjögren's syndrome or of pancreas in diabetes warrants further investigation.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Periodontal Diseases/genetics , Periodontal Diseases/microbiology , Salivary Glands/metabolism , Salivary Glands/microbiology , Animals , Disease Models, Animal , Female , Organ Specificity/genetics , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/isolation & purification , RNA, Ribosomal, 16S/genetics , Rats , Treponema denticola/genetics , Treponema denticola/isolation & purification
20.
PLoS One ; 10(11): e0143291, 2015.
Article in English | MEDLINE | ID: mdl-26619277

ABSTRACT

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.


Subject(s)
Aggressive Periodontitis/microbiology , Apolipoproteins E/genetics , Atherosclerosis/etiology , Microbiota , Aggressive Periodontitis/complications , Animals , Chemokines/genetics , Chemokines/metabolism , Fusobacterium nucleatum/isolation & purification , Inflammation/etiology , Interleukins/genetics , Interleukins/metabolism , Male , Mice , Mouth/microbiology , Porphyromonas gingivalis/isolation & purification , T-Lymphocytes/metabolism , Treponema denticola/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...