Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
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
2.
Crit Rev Microbiol ; 46(2): 213-236, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32267781

ABSTRACT

Porphyromonas gingivalis (P. gingivalis), a Gram-negative facultative anaerobe of the oral cavity, is associated with the onset of various adverse pregnancy outcomes. P. gingivalis is linked with the development of preeclampsia, preterm labour, spontaneous abortion, gestational diabetes, foetal growth restriction, and misconception. The unique virulence factors, surface adhesions, enzymes of P. gingivalis can directly injure and alter the morphology, microbiome the foetal and maternal tissues. P. gingivalis can even exaggerate the production of cytokines, free radicals and acute-phase proteins in the uterine compartment that increases the risk of myometrial contraction and onset of preterm labour. Although evidence confirms the presence of P. gingivalis in the amniotic fluid and placenta of women with poor pregnancy outcomes, the intricate molecular mechanisms by which P. gingivalis initiates various antenatal and postnatal maternal and foetal complications are not well explained in the literature. Therefore, the present review aims to comprehensively summarise and highlight the recent and unique molecular pathogenic mechanisms of P. gingivalis associated with adverse pregnancy outcomes.


Subject(s)
Bacteroidaceae Infections/physiopathology , Porphyromonas gingivalis/physiology , Pregnancy Complications/physiopathology , Pregnancy Outcome , Animals , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Mouth/microbiology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/isolation & purification , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy Complications/microbiology
3.
Scand J Gastroenterol ; 54(12): 1419-1425, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31765575

ABSTRACT

Objective: Diarrhea-predominant irritable bowel syndrome (IBS-D) is the main subtype of IBS, a chronic functional gastrointestinal disorder. Small intestinal bacterial overgrowth (SIBO), which is characterized by dysbiosis of the bowel, causes gastrointestinal symptoms quite similar to IBS-D. However, whether SIBO correlates with IBS-D and its further mechanism remain unknown.Materials and Methods: The study included 60 IBS-D patients that fulfilled Rome IV criteria and 60 healthy controls. All subjects were undergoing a lactose breath test (LBT) to diagnose SIBO. IBS-D patients were further assigned to negative SIBO (SIBO-) subgroup and positive SIBO (SIBO+) subgroup to analyze the scores of symptoms and differences in the fecal microbiota.Results: The prevalence of SIBO in IBS-D patients was higher than that in healthy controls (51.7% vs. 16.7%, p ≤ .001). In addition, IBS-SSS in SIBO+ subgroup was significantly higher than SIBO- subgroup (p = .015). The 16S rRNA analyses showed that composition and abundance of fecal microbiota were obviously different between the two subgroups. There was a remarkable increase in Prevotella in IBS-D patients, especially in IBS-D SIBO+ sufferers. Meanwhile, there were a moderately positive correlation of the abundance of Prevotella (rho = 0.458, p ≤ .001) with IBS-SSS.Conclusion: SIBO is associated with IBS-D, which may be related to alteration in the intestinal microbiota. These findings suggest the potent role of Prevotella in gastrointestinal symptoms between SIBO and IBS-D, thus provide a novel insight into the connection between SIBO and IBS-D.


Subject(s)
Bacteroidaceae Infections , Diarrhea/microbiology , Intestine, Small/microbiology , Irritable Bowel Syndrome , Prevotella/isolation & purification , Adult , Bacteroidaceae Infections/diagnosis , Bacteroidaceae Infections/epidemiology , Bacteroidaceae Infections/physiopathology , Breath Tests/methods , China/epidemiology , Correlation of Data , Dysbiosis/microbiology , Dysbiosis/physiopathology , Feces/microbiology , Female , Humans , Irritable Bowel Syndrome/diagnosis , Irritable Bowel Syndrome/epidemiology , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/physiopathology , Male , Prevalence
4.
Article in English | MEDLINE | ID: mdl-31380305

ABSTRACT

Periodontitis is a common intraoral infection and is inextricably linked to systemic diseases. Recently, the regulation between host immunologic response and periodontal pathogens has become a hotspot to explain the mechanism of periodontitis and related systemic diseases. Since Porphyromonas gingivalis (P. gingivalis) was proved as critical periodontal pathogen above all, researches focusing on the mechanism of its virulence factors have received extensive attention. Studies have shown that in the development of periodontitis, in addition to the direct release of virulent factors by periodontal pathogens to destroy periodontal tissues, over-low or over-high intrinsic immune and inflammatory response mediated by Toll-like receptors (TLRs) can lead to more lasting destruction of periodontal tissues. It is very necessary to sort out how various cytopathic factors of P. gingivalis mediate inflammation and immune responses between the host through TLRs so as to help precisely prevent, diagnose, and treat periodontitis in clinic. This review summarizes the role of three most widely studied pathogenic factors produced by P. gingivalis (lipopolysaccharide, gingipains, pili) and their interactions with TLRs at the cellular and molecular level in the progress of periodontitis.


Subject(s)
Fimbriae, Bacterial/metabolism , Gingipain Cysteine Endopeptidases/metabolism , Host-Pathogen Interactions , Lipopolysaccharides/metabolism , Porphyromonas gingivalis/pathogenicity , Toll-Like Receptors/metabolism , Virulence Factors/metabolism , Animals , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Bacteroidaceae Infections/physiopathology , Disease Models, Animal , Humans , Periodontitis/microbiology , Periodontitis/pathology , Periodontitis/physiopathology
5.
Virulence ; 9(1): 845-859, 2018 12 31.
Article in English | MEDLINE | ID: mdl-29616874

ABSTRACT

Porphyromonas gingivalis, an opportunistic pathogen usurps gingival epithelial cells (GECs) as primary intracellular niche for its colonization in the oral mucosa. However, the precise characterization of the intracellular trafficking and fate of P. gingivalis in GECs remains incomplete. Therefore, we employed high-resolution three-dimensional-transmission-electron-microscopy to determine the subcellular location of P. gingivalis in human primary GECs upon invasion. Serial sections of infected-GECs and their tomographic reconstruction depicted ER-rich-double-membrane autophagosomal-vacuoles harboring P. gingivalis. Western-blotting and fluorescence confocal microscopy showed that P. gingivalis significantly induces LC3-lipidation in a time-dependent-manner and co-localizes with LC3, ER-lumen-protein Bip, or ER-tracker, which are major components of the phagophore membrane. Furthermore, GECs that were infected with FMN-green-fluorescent transformant-strain (PgFbFP) and selectively permeabilized by digitonin showed rapidly increasing large numbers of double-membrane-vacuolar-P. gingivalis over 24 hours of infection with a low-ratio of cytosolically free-bacteria. Moreover, inhibition of autophagy using 3-methyladenine or ATG5 siRNA significantly reduced the viability of intracellular P. gingivalis in GECs as determined by an antibiotic-protection-assay. Lysosomal marker, LAMP-1, showed a low-degree colocalization with P. gingivalis (∼20%). PgFbFP was used to investigate the fate of vacuolar- versus cytosolic-P. gingivalis by their association with ubiquitin-binding-adaptor-proteins, NDP52 and p62. Only cytosolic-P. gingivalis had a significant association with both markers, which suggests cytosolically-free bacteria are likely destined to the lysosomal-degradation pathway whereas the vacuolar-P. gingivalis survives. Therefore, the results reveal a novel mechanism for P. gingivalis survival in GECs by harnessing host autophagy machinery to establish a successful replicative niche and persistence in the oral mucosa.


Subject(s)
Autophagosomes/microbiology , Bacteroidaceae Infections/microbiology , Endoplasmic Reticulum/microbiology , Epithelial Cells/microbiology , Porphyromonas gingivalis/physiology , Autophagy , Bacteroidaceae Infections/physiopathology , Gingiva/cytology , Gingiva/microbiology , Humans , Microbial Viability , Porphyromonas gingivalis/growth & development
6.
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
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(3): 315-319, 2017 Mar.
Article in Chinese | MEDLINE | ID: mdl-28274308

ABSTRACT

Objective To investigate the impact of lipopolysaccharide of Porphyromonas gingivalis (Pg-LPS) on the autophagy of human gingival fibroblasts (HGFs). Methods Firstly, HGFs was stimulated with 10 µg/mL Pg-LPS for 12 hours or 24 hours. Rapamycin was used as a positive control. The expression of LC3B was detected by Western blotting and the distribution of autophagosomes was observed by indirect immunofluorescence staining. At the same time, mitochondrial ROS (mtROS) was labeled by MitoSOX Red. The levels of mtROS and mitochondrial autophagy were measured in HGFs after treated with Pg-LPS. Then, T-butyl-4 (BHA), N-acetylcysteine (NAC) and coenzyme Q10 (CoQ10) were used separately to block the ROS and the expression of LC3B in Pg-LPS-treated HGFs was tested by Western blotting. Results After treatment with Pg-LPS, the ratio of LC3BII/LC3BI and the number of autophagic cells significantly increased, and the increase in the 24-hour treatment group was the more obvious than that in the 12-hour treatment group. The mtROS production and mitochondrial autophagy were significantly promoted after Pg-LPS treatment. In addition, CoQ10 effectively reduced Pg-LPS-induced autophagy of HGFs. Conclusion Pg-LPS can induce the autophagy of HGFs by raising mtROS production, and autophagy is involved in the degradation of damaged mitochondria to maintain cellular homeostasis.


Subject(s)
Autophagy , Bacteroidaceae Infections/physiopathology , Fibroblasts/cytology , Gingiva/cytology , Lipopolysaccharides/metabolism , Porphyromonas gingivalis/metabolism , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Fibroblasts/microbiology , Gingiva/metabolism , Gingiva/microbiology , Humans , Mitochondria/metabolism , Porphyromonas gingivalis/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
8.
Beijing Da Xue Xue Bao Yi Xue Ban ; 49(1): 31-5, 2017 02 18.
Article in Chinese | MEDLINE | ID: mdl-28203000

ABSTRACT

OBJECTIVE: To compare the extent and time course of alveolar bone loss and osteoclast activation in two murine models of periodontal disease: molar ligation and Porphyromonas gingivalis (P. gingivalis) oral inoculation. METHODS: A split-mouth design was applied to two groups of mice (C57BL6, 6-8 weeks old, n=24 in both groups), resulting in four treatment groups: (1) Control group: unligated upper right 2nd molars receiving CMC only, (2)Ligature group: ligation of a 9-0 suture around the upper left 2nd molar, (3) P. gingivalis group: unligated upper right 2nd molar receiving P. gingivalis challenge only, (4)Ligature+P.gingivalis group: ligation of the upper left 2nd molar in combination with oral inoculation with 109 colony-forming units(CFU) P. gingivalis. Alveolar bone loss was measured as the cementoenamel junction and alveolar bone crest (CEJ-ABC) distance. In the study, 48 C57BL6 mice were designed and treated as described above, and osteoclasts were counted on histological sections following tartrate-resistant acid phosphatase (TRAP) staining and counts were normalized to alveolar bone surface distance. Then 36 C57BL6 mice were investigated, of which 30 were ligated a 9-0 silk ligature around the 2nd molar in the left maxillary quadrant and 6 were not ligated. After ligation for 1 week, the ligatures in 12 mice were taken off for either 1 week or 2 weeks. The CEJ-ABC distance of the 6 mice without ligation was baseline. The CEJ-ABC distances were measured and analyzed. The data were analyzed with one-way ANOVA. RESULTS: Molar ligation induced marked alveolar bone loss after 3, 6, 9 and 12 weeks [(0.16±0.04) mm, (0.16±0.02) mm, (0.18±0.03) mm, (0.17±0.02) mm], vs. corresponding controls [(0.09±0.03)mm,(0.10±0.01)mm,(0.12±0.04)mm,(0.12±0.01)mm] and P. gingivalis group [(0.09±0.03)mm, (0.12±0.01)mm,(0.12±0.02)mm,(0.10±0.01)mm], P<0.05. Combined treatment with molar ligation and P. gingivalis did not further increase the CEJ-ABC distance. Evidence for osteoclast activation was found one day after molar ligation, and TRAP-positive cell numbers peaked on day 3 (12±4 vs. control 2±2, P<0.01). After taking off ligature following ligation for 2 weeks, it showed significantly regrowth of alveolar bone compared with that before removal of the ligature on day 7 [(0.07±0.02)mm vs. (0.13±0.01)mm, P<0.01]. CONCLUSION: Molar ligation is a rapid and effective way to induce periodontal bone loss in mice. Osteoclast activation occurs within 24 hours of ligature placement, and the extent of bone loss well exceeds that of the P.gingivalis-induced bone loss. Removing ligature after periodontal disease might help bone regeneration by regrowth of the alveolar bone.


Subject(s)
Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Alveolar Bone Loss/physiopathology , Bacteroidaceae Infections/physiopathology , Constriction, Pathologic/physiopathology , Porphyromonas gingivalis/physiology , Tooth Cervix/microbiology , Tooth Cervix/pathology , Tooth Cervix/physiopathology , Animals , Bacteroidaceae Infections/chemically induced , Disease Models, Animal , Ligation/adverse effects , Ligation/methods , Mice , Mice, Inbred C57BL , Molar/microbiology , Molar/pathology , Molar/physiopathology , Osteoclasts/physiology
9.
Clin Oral Implants Res ; 28(6): 727-733, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27194177

ABSTRACT

OBJECTIVES: This study aimed to evaluate the dynamics of newly bone formation and dimensional change in diseased extraction sockets using Bio-Oss® Collagen with or without a collagen membrane. MATERIAL AND METHODS: In six beagle dogs, right and left 3rd and 4th mandibular premolars were hemisected and the distal roots were removed. Combined endodontic-periodontic lesions were induced in all sites using black silk, collagen sponge, endodontic files, and application of Porphyromonas gingivalis. After 4 months, among 4 premolars, three teeth were randomly selected per dog and allocated to the following experimental groups: Control group (no treatment but debridement), Test 1 group (only Bio-Oss® Collagen graft), and Test 2 group (Bio-Oss® Collagen graft with a collagen membrane). After 7 months from the baseline, the beagle dogs were sacrificed for histomorphometric and Micro-CT analysis. RESULTS: The vertical distance between buccal and lingual crests in the Control group (2.22 ± 0.26 mm) and Test 2 group (1.80 ± 0.16 mm) was significantly different. The socket of the Test 2 group (27.04 ± 5.25%) was occupied by a greater quantity of bone graft compared to the Test 1 group (18.49 ± 2.11%). CONCLUSION: Ridge preservation in diseased extraction sockets could compensate for buccal bone resorption by contact osteogenesis surrounding the bone graft particles at the bucco-coronal area during socket healing, and the application of a collagen membrane at the entrance of the socket is useful for preserving graft material at the coronal part of the socket.


Subject(s)
Bacteroidaceae Infections/physiopathology , Bone Regeneration , Bone Substitutes , Collagen , Minerals , Tooth Socket/physiology , Wound Healing , Animals , Chronic Disease , Disease Models, Animal , Dogs , Inflammation , Membranes, Artificial , Porphyromonas gingivalis , Tooth Extraction , Tooth Socket/cytology , Tooth Socket/diagnostic imaging , Tooth Socket/microbiology , X-Ray Microtomography
10.
Microb Pathog ; 92: 36-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26724741

ABSTRACT

The functional modulation of vascular endothelial cells associated with stroke and periodontal disease has not yet been clarified. The objective of this study is to analyze the vascular endothelial function of periodontitis and stroke animal models. We examined endothelial function and gingival blood flow in oral microcirculation in vivo and measured the isometric tension in vitro of the aorta in animal models for lifestyle-related diseases, such as periodontitis and stroke. Gingival reactive hyperemia (GRH) was measured using laser Doppler flowmetry. Wistar Kyoto rats (WKY) were used as control animals; Porphyromonas gingivalis (P. gingivalis) infected WKY (WKY + Pg) as the periodontitis model; stroke-prone spontaneously hypertensive rat (SHRSP) as the stroke model; and a final group consisting of P. gingivalis infected SHRSP (SHRSP + Pg). Furthermore, for each group, the relaxation of descending aortic ring preparations was measured using a force transducer. The GRH was estimated by maximum response (peak), time taken for the maximum response to fall to one half (T1/2), and increased total amount of blood flow (mass). The relative change in T1/2 and mass increased in SHRSP + Pg compared to WKY. However, mass significantly increased in WKY (758.59 ± 88.21 ml/min/100 g s to 1755.55 ± 226.10 ml/min/100 g s) and SHRSP (1214.87 ± 141.61 ml/min/100 g s to 2674.32 ± 675.48 ml/min/100 g s) after treatment with acetylcholine. In addition, T1/2 and mass significantly increased in WKY + Pg (624.18 ± 96.36 ml/min/100 g s to 2629.90 ± 612.01 ml/min/100 g s) and SHRSP + Pg (1116.36 ± 206.24 ml/min/100 g s to 1952.76 ± 217.39 ml/min/100 g s) after treatment with nitroglycerin. Furthermore, the endothelium-dependent relaxation of ring preparations, evoked by acetylcholine, was attenuated in SHRSP compared with WKY, but not in SHRSP + Pg. This attenuation effect in SHRSP could be prevented by superoxide dismutase pretreatment. Our results suggest altered endothelial function may occur in gingival tissue in animal models experiencing both periodontitis and stroke. Therefore, these results indicate the disruption of vascular function in oral microcirculation may be caused by the interaction between the oxidative stress induced by periodontitis and nitric oxide in periodontitis, similar to the interactions present in stroke cases.


Subject(s)
Aorta/physiopathology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/physiopathology , Microcirculation , Periodontitis/microbiology , Periodontitis/physiopathology , Porphyromonas gingivalis , Stroke/etiology , Animals , Blood Pressure , Disease Models, Animal , Hyperemia/etiology , Male , Rats , Rats, Inbred SHR , Regional Blood Flow , Stroke/physiopathology
11.
BMC Oral Health ; 14: 89, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25027664

ABSTRACT

BACKGROUND: Porphyromonas gingivalis has been shown to invade osteoblasts and inhibit their differentiation and mineralization in vitro. However, it is unclear if P. gingivalis can invade osteoblasts in vivo and how this would affect alveolar osteoblast/osteoclast dynamics. This study aims to answer these questions using a periodontitis mouse model under repetitive P. gingivalis inoculations. METHODS: For 3-month-old BALB/cByJ female mice, 10(9) CFU of P. gingivalis were inoculated onto the gingival margin of maxillary molars 4 times at 2-day intervals. After 2 weeks, another 4 inoculations at 2-day intervals were applied. Calcein was injected 7 and 2 days before sacrificing animals to label the newly formed bone. Four weeks after final inoculation, mice were sacrificed and maxilla collected. Immunohistochemistry, micro-CT, and bone histomorphometry were performed on the specimens. Sham infection with only vehicle was the control. RESULTS: P. gingivalis was found to invade gingival epithelia, periodontal ligament fibroblasts, and alveolar osteoblasts. Micro-CT showed alveolar bone resorption and significant reduction of bone mineral density and content in the infected mice compared to the controls. Bone histomorphometry showed a decrease in osteoblasts, an increase in osteoclasts and bone resorption, and a surprisingly increased osteoblastic bone formation in the infected mice compared to the controls. CONCLUSIONS: P. gingivalis invades alveolar osteoblasts in the periodontitis mouse model and cause alveolar bone loss. Although P. gingivalis appears to suppress osteoblast pool and enhance osteoclastic bone resorption, the bone formation capacity is temporarily elevated in the infected mice, possibly via some anti-microbial compensational mechanisms.


Subject(s)
Alveolar Bone Loss/microbiology , Bacteroidaceae Infections/physiopathology , Osteoblasts/microbiology , Osteoclasts/microbiology , Osteogenesis/physiology , Periodontitis/microbiology , Porphyromonas gingivalis/physiology , Alveolar Bone Loss/pathology , Alveolar Process/microbiology , Alveolar Process/pathology , Animals , Bone Density/physiology , Cell Count , Disease Models, Animal , Epithelium/microbiology , Female , Fibroblasts/microbiology , Fluoresceins , Fluorescent Dyes , Gingiva/microbiology , Maxilla/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Periodontal Ligament/microbiology , Periodontal Ligament/pathology , X-Ray Microtomography/methods
12.
PLoS Pathog ; 9(9): e1003627, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068934

ABSTRACT

Rheumatoid arthritis and periodontitis are two prevalent chronic inflammatory diseases in humans and are associated with each other both clinically and epidemiologically. Recent findings suggest a causative link between periodontal infection and rheumatoid arthritis via bacteria-dependent induction of a pathogenic autoimmune response to citrullinated epitopes. Here we showed that infection with viable periodontal pathogen Porphyromonas gingivalis strain W83 exacerbated collagen-induced arthritis (CIA) in a mouse model, as manifested by earlier onset, accelerated progression and enhanced severity of the disease, including significantly increased bone and cartilage destruction. The ability of P. gingivalis to augment CIA was dependent on the expression of a unique P. gingivalis peptidylarginine deiminase (PPAD), which converts arginine residues in proteins to citrulline. Infection with wild type P. gingivalis was responsible for significantly increased levels of autoantibodies to collagen type II and citrullinated epitopes as a PPAD-null mutant did not elicit similar host response. High level of citrullinated proteins was also detected at the site of infection with wild-type P. gingivalis. Together, these results suggest bacterial PAD as the mechanistic link between P. gingivalis periodontal infection and rheumatoid arthritis.


Subject(s)
Arthritis/microbiology , Bacterial Proteins/metabolism , Bacteroidaceae Infections/microbiology , Disease Models, Animal , Hydrolases/metabolism , Periodontitis/microbiology , Porphyromonas gingivalis/enzymology , Animals , Arthritis/immunology , Arthritis/pathology , Arthritis/physiopathology , Autoantibodies/analysis , Bacterial Proteins/genetics , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Bacteroidaceae Infections/physiopathology , Bone Resorption/etiology , Citrulline/metabolism , Disease Progression , Gene Deletion , Hydrolases/genetics , Joints/immunology , Joints/metabolism , Joints/microbiology , Joints/pathology , Male , Mice, Inbred DBA , Neutrophil Infiltration , Periodontitis/immunology , Periodontitis/metabolism , Periodontitis/pathology , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/isolation & purification , Prevotella intermedia/enzymology , Prevotella intermedia/immunology , Prevotella intermedia/isolation & purification , Protein Processing, Post-Translational , Protein-Arginine Deiminases , Severity of Illness Index
13.
Thromb Res ; 132(1): e8-14, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23706960

ABSTRACT

INTRODUCTION: A close association exists between oral health and cardiovascular disease. Periodontal disease induces early vascular changes while oral pathogens have been detected in sub gingival and atheromatous plaques. We examined the interrelationship between Periodontal disease, oral bacteria, surrogate sub-clinical markers and coronary artery disease (CAD) in a representative Asian Indian cohort. MATERIALS AND METHODS: 532 Gingivitis cases and 282 Periodontitis cases were assessed for early peripheral vascular changes, namely pulse wave velocity (PWV), arterial stiffness index (ASI) and ankle brachial index (ABI) using computerized oscillometry method. Relative quantitation (RQ) of Porphyromonas gingivalis (Pg) was estimated in saliva samples of 54 Periodontitis, 25 Gingivitis and 51 CAD cases (38 also had oral disease) by Taqman assay by amplifying pathogen-specific gene targets, 16srRNA and IktA, respectively, and 16s universal bacterial rRNA as endogenous control. RESULTS: PWV and ASI were elevated in Periodontitis compared to Gingivitis cases (p<0.0001) and in those with diabetes and hypertension. Cases with Periodontitis showed higher mean expression of Pg than Gingivitis (0.37±0.05 versus 0.15±0.04, p<0.0001), while CAD patients with oral disease (N=38) showed lower mean Pg expression than those without oral disease (N=13) (0.712±0.119 versus 1.526±0.257, p=0.008). Higher Pg expression was recorded in subjects with diabetes and hypertension. CONCLUSION: Oral disease induces early changes in the peripheral blood vessels. Further, common presence of Pg in subjects with oral disease, in those with established cardiovascular risk factors and in patients with symptomatic CAD reflects the importance of oral hygiene in the development of Coronary Artery Disease in Asian Indians.


Subject(s)
Coronary Artery Disease/epidemiology , Mouth/microbiology , Periodontal Diseases/complications , Periodontal Diseases/microbiology , Porphyromonas gingivalis/isolation & purification , Vascular Resistance , Adult , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/physiopathology , Female , Gingivitis/complications , Gingivitis/microbiology , Gingivitis/physiopathology , Humans , India/epidemiology , Male , Middle Aged , Periodontal Diseases/physiopathology , Risk Factors
14.
Cell Microbiol ; 15(6): 961-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23241000

ABSTRACT

Ligation of P2X7 receptors with a 'danger signal', extracellular ATP (eATP), has recently been shown to result in production of intracellular reactive-oxygen-species (ROS) in macrophages. We show that primary gingival epithelial cells (GECs) produce sustained, robust cellular ROS upon stimulation by eATP. The induction of ROS was mediated by P2X7 receptor signalling coupled with NADPH-oxidase activation, as determined by pharmacological inhibition and RNA interference. Furthermore, Porphyromonas gingivalis, an oral opportunistic pathogen, upregulated the antioxidant glutathione response, modulated eATP-induced cytosolic and mitochondrial ROS generated through P2X7 /NADPH-oxidase interactome, and subsequently blocked oxidative stress in GECs via temporal secretion of a P. gingivalis effector, nucleoside-diphosphate-kinase (Ndk). An ndk-deficient P. gingivalis mutant lacked the ability to inhibit ROS production and persist intracellularly following eATP stimulation. Treatment with recombinant Ndk significantly diminished eATP-evoked ROS production. P. gingivalis infection elicited a strong, time-dependent increase in anti-oxidativemitochondrial UCP2 levels, whereas ndk-deficient mutant did not cause any change. The results reveal a novel signalling cascade that is tightly coupled with eATP signalling and ROS regulation. Ndk by P. gingivalis counteracts these antimicrobial signalling activities by secreting Ndk, thus contributing to successful persistence of the pathogen.


Subject(s)
Adenosine Triphosphate/metabolism , NADPH Oxidases/metabolism , Nucleoside-Diphosphate Kinase/metabolism , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/pathogenicity , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2X7/metabolism , Signal Transduction/physiology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/pathology , Bacteroidaceae Infections/physiopathology , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gingiva/metabolism , Gingiva/microbiology , Gingiva/pathology , Humans , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mutation/genetics , Nucleoside-Diphosphate Kinase/genetics , Oxidative Stress/physiology , Porphyromonas gingivalis/physiology , Uncoupling Protein 2
15.
FEMS Immunol Med Microbiol ; 62(1): 91-100, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21314733

ABSTRACT

Prevotella intermedia is an important periodontal pathogen that induces various inflammatory and immune responses. In this study, we investigated the effects of P. intermedia on the plasminogen system in human periodontal ligament (hPDL) cells and explored the signaling pathways involved. Using semi-quantitative reverse transcription (RT)-PCR and quantitative real-time RT-qPCR, we demonstrated that P. intermedia challenge increased tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor (PAI)-2 expression in a concentration- and time-dependent manner, but exerted no influence on urokinase-type plasminogen activator and PAI-1mRNA expression in hPDL cells. Prevotella intermedia stimulation also enhanced tPA protein secretion as confirmed by enzyme-linked immunosorbent assay. Western blot results revealed that P. intermedia treatment increased phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 kinase (p38). ERK, JNK and protein kinase C inhibitors significantly attenuated the P. intermedia-induced tPA and PAI-2 expression. Furthermore, p38 and phosphatidylinositol 3-kinase inhibitors markedly decreased PAI-2 expression, whereas they showed no or little inhibition on tPA expression. In contrast, inhibition of protein kinase A greatly enhanced the upregulatory effect of P. intermedia on tPA and PAI-2 expression. Our results suggest that P. intermedia may contribute to periodontal tissue destruction by upregulating tPA and PAI-2 expression in hPDL cells via multiple signaling pathways.


Subject(s)
Bacteroidaceae Infections/microbiology , Periodontal Ligament/microbiology , Periodontal Ligament/physiopathology , Plasminogen Activator Inhibitor 2/metabolism , Prevotella intermedia/pathogenicity , Signal Transduction , Tissue Plasminogen Activator/metabolism , Bacteroidaceae Infections/physiopathology , Cells, Cultured/microbiology , Humans , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Periodontitis/microbiology , Periodontitis/physiopathology , Plasminogen Activator Inhibitor 2/genetics , Signal Transduction/physiology , Tissue Plasminogen Activator/genetics , Up-Regulation
16.
Eur J Oral Sci ; 118(6): 582-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21083619

ABSTRACT

The gingival epithelium plays an important role in the protection of oral tissues from microbial challenge. Oral keratinocytes form various cellular contacts, including tight junctions, and thus are able to create an epithelial barrier. A measurable indicator of barrier function in vitro is the transepithelial electrical resistance (TER). Porphyromonas gingivalis is recognized as a major aetiologic agent of periodontal disease and exhibits a variety of virulence factors. The aim of the study was to investigate the effect, in vitro, of infection with P. gingivalis on gingival barriers composed of primary and immortalized human keratinocytes. Primary and immortalized human gingival keratinocytes were infected with different strains of P. gingivalis. The impact of the bacterial challenge on the barrier was analysed by measuring the TER. The destructive effects of gingipains were blocked by specific enzyme inhibitors. After an initial increase of about 20-30% in infected wells, the TER decreased to zero. Gingipain inhibitors delayed the destruction of the barrier by 12 ± 4 h. In all cases, the loss of TER was accelerated if the system was infected from the basolateral side. A distinct effect of P. gingivalis on the epithelial barrier function of three-dimensional cultured epithelial cell models was demonstrated, which can partly be attributed to the activity of gingipains.


Subject(s)
Bacteroidaceae Infections/physiopathology , Gingiva/microbiology , Keratinocytes/microbiology , Porphyromonas gingivalis/pathogenicity , Adhesins, Bacterial/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Cell Culture Techniques , Cell Line , Cells, Cultured , Claudin-1 , Claudins , Cysteine Endopeptidases/drug effects , Dipeptides/pharmacology , Electric Impedance , Enzyme Inhibitors/pharmacology , Epithelium/microbiology , Epithelium/pathology , Gingipain Cysteine Endopeptidases , Gingiva/pathology , Humans , Keratinocytes/pathology , Ketones/pharmacology , Membrane Proteins/analysis , Occludin , Porphyromonas gingivalis/enzymology , Tight Junctions/microbiology , Tight Junctions/physiology , Time Factors
17.
Jpn J Infect Dis ; 63(6): 427-32, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21099094

ABSTRACT

Periodontitis is a chronic inflammatory disease caused by the infection of periodontopathic bacteria in dental plaque. However, an individual's susceptibility to this disease appears to be associated with multiple genetic factors, as seen in the case of leprosy. In order to gain a better understanding of the pathophysiology of periodontal disease in subjects with leprosy, we investigated the clinical features of periodontitis and the immunological responses against periodontopathic bacteria in 382 subjects with a history of leprosy and 451 age-matched control subjects. The prevalence of periodontitis and the degree of periodontal pocket depth were found to be significantly higher in leprosy patients than in age-matched controls. Furthermore, a comparison of the clinical parameters of lepromatous leprosy (L-lep) and tuberculoid leprosy (T-lep) patients showed that the probing pocket depth of L-lep patients with periodontal disease was significantly higher than that for T-lep patients. In contrast, serum IgG titers against Porphyromonas gingivalis in L-lep patients were significantly lower than in T-lep patients. These results imply that L-lep patients show more severe periodontal disease than T-lep patients or age-matched control subjects, and that low humoral immunity against P. gingivalis might be one of the genetic factors determining periodontal disease susceptibility in leprosy patients.


Subject(s)
Leprosy, Lepromatous/complications , Leprosy, Tuberculoid/complications , Periodontitis/immunology , Periodontitis/physiopathology , Adult , Aged , Aged, 80 and over , Antibodies, Bacterial/blood , Bacteroidaceae Infections/epidemiology , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/physiopathology , Case-Control Studies , Female , Humans , Japan/epidemiology , Leprosy, Lepromatous/epidemiology , Leprosy, Lepromatous/microbiology , Leprosy, Tuberculoid/epidemiology , Leprosy, Tuberculoid/microbiology , Male , Middle Aged , Periodontal Pocket , Periodontitis/epidemiology , Periodontitis/microbiology , Porphyromonas gingivalis/immunology , Prevalence , Severity of Illness Index
18.
Int Immunopharmacol ; 10(7): 744-50, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20399284

ABSTRACT

Periodontitis is a multifactorial polymicrobial infection characterized by a destructive inflammatory process affecting tooth-supporting tissues and resulting in periodontal pocket formation, alveolar bone resorption and, eventually, tooth loss. The continuous challenge of host immune and resident cells by periodontopathogens and their virulence factors, such as lipopolysaccharide (LPS), results in enhanced and uncontrolled secretion of cytokines. The latter directly or indirectly participate in tissue destruction and bone resorption. Metronidazole (MTZ) is a widely used antimicrobial agent. The immunomodulatory effects of antibiotics might influence the degree of the local response to infection on the human periodontal ligament cell (HPLC). HPLCs play a role in the immune response of the oral cavity. In addition, HPLC can produce cytokines that increase the inflammatory response and that supply for normal communication. MTZ has also been proposed in the field of periodontal therapy either with a systemic administration or with local biodegradable sustained-release agents. The local administration of MTZ in the form of gel significantly reduces the systemic side effects. The aim of the present study, was to simulate the in vivo conditions occurring in diseased periodontal sites, and to evaluate the effects of MTZ on the viability of isolated HPLCs. The ability of MTZ to modulate the release of interleukin (IL)-1beta, IL-6, IL-8, IL-12 and tumor necrosis factor alpha (TNF-alpha) in HPLC, treated or not with LPS of Porphyromonas gingivalis was also evaluated. The results obtained showed that MTZ had no cytotoxic effect on HPLC and was able to inhibit the production of pro-inflammatory cytokines analyzed. The ability of MTZ to determine immunomodulatory effects could provide possible therapeutic applications in the field of periodontal research.


Subject(s)
Anti-Infective Agents/pharmacology , Bacteroidaceae Infections/drug therapy , Metronidazole/pharmacology , Periodontal Ligament/drug effects , Porphyromonas gingivalis/immunology , Adult , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Bacteroidaceae Infections/physiopathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Male , Periodontal Ligament/metabolism , Periodontal Ligament/pathology , Periodontitis , Porphyromonas gingivalis/pathogenicity
20.
Neurol Med Chir (Tokyo) ; 49(8): 370-3, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19707005

ABSTRACT

A 67-year-old man presented with a rare case of cavernous sinus thrombophlebitis (CST) caused by Porphyromonas gingivalis with abscess formation extending to the orbital cavity. Neuroimaging demonstrated a cystic lesion in the right cavernous sinus that was hyperintense on diffusion-weighted imaging. The patient was successfully treated with surgical drainage and antibiotic administration. CST is rare and often has a fulminant progression with high rates of morbidity and mortality. The differential diagnosis of cavernous sinus lesions should include CST. Early recognition and differentiation from other diseases with aggressive medical and possible surgical intervention are necessary to reduce mortality and long-term sequelae. Diffusion-weighted imaging is useful for the early recognition and differentiation of CST from other diseases.


Subject(s)
Bacteroidaceae Infections/pathology , Cavernous Sinus Thrombosis/microbiology , Cavernous Sinus Thrombosis/pathology , Empyema, Subdural/microbiology , Empyema, Subdural/pathology , Porphyromonas gingivalis , Aged , Anti-Bacterial Agents/therapeutic use , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/physiopathology , Cavernous Sinus/microbiology , Cavernous Sinus/pathology , Cavernous Sinus/physiopathology , Cavernous Sinus Thrombosis/physiopathology , Diagnosis, Differential , Diffusion Magnetic Resonance Imaging , Disease Progression , Drainage/methods , Empyema, Subdural/physiopathology , Exophthalmos/etiology , Humans , Male , Neurosurgical Procedures , Orbit/pathology , Orbit/physiopathology , Orbit/surgery , Osteotomy/methods , Pain/etiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...