Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 239
1.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article En | MEDLINE | ID: mdl-38892314

GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.


Apolipoproteins E , Atherosclerosis , Periodontal Diseases , Porphyromonas gingivalis , Animals , Mice , Apolipoproteins E/deficiency , Periodontal Diseases/microbiology , Periodontal Diseases/prevention & control , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Atherosclerosis/microbiology , Telomerase/metabolism , Peptide Fragments/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Alzheimer Disease/microbiology , Periodontitis/microbiology , Periodontitis/prevention & control , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/prevention & control , Disease Models, Animal , Mice, Inbred C57BL , Male , Humans
2.
Int J Mol Sci ; 25(10)2024 May 07.
Article En | MEDLINE | ID: mdl-38791123

Periodontitis is linked to the onset and progression of oral squamous cell carcinoma (OSCC), an epidemiologically frequent and clinically aggressive malignancy. In this context, Fusobacterium (F.) nucleatum and Porphyromonas (P.) gingivalis, two bacteria that cause periodontitis, are found in OSCC tissues as well as in oral premalignant lesions, where they exert pro-tumorigenic activities. Since the two bacteria are present also in endodontic diseases, playing a role in their pathogenesis, here we analyze the literature searching for information on the impact that endodontic infection by P. gingivalis or F. nucleatum could have on cellular and molecular events involved in oral carcinogenesis. Results from the reviewed papers indicate that infection by P. gingivalis and/or F. nucleatum triggers the production of inflammatory cytokines and growth factors in dental pulp cells or periodontal cells, affecting the survival, proliferation, invasion, and differentiation of OSCC cells. In addition, the two bacteria and the cytokines they induce halt the differentiation and stimulate the proliferation and invasion of stem cells populating the dental pulp or the periodontium. Although most of the literature confutes the possibility that bacteria-induced endodontic inflammatory diseases could impact on oral carcinogenesis, the papers we have analyzed and discussed herein recommend further investigations on this topic.


Fusobacterium Infections , Fusobacterium nucleatum , Mouth Neoplasms , Porphyromonas gingivalis , Humans , Porphyromonas gingivalis/pathogenicity , Fusobacterium nucleatum/pathogenicity , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Fusobacterium Infections/microbiology , Fusobacterium Infections/complications , Carcinogenesis , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/complications , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Periodontitis/microbiology , Animals , Cytokines/metabolism
5.
J Clin Periodontol ; 51(7): 818-839, 2024 Jul.
Article En | MEDLINE | ID: mdl-38414291

AIM: Blood-brain barrier (BBB) disorder is one of the early findings in cognitive impairments. We have recently found that Porphyromonas gingivalis bacteraemia can cause cognitive impairment and increased BBB permeability. This study aimed to find out the possible key virulence factors of P. gingivalis contributing to the pathological process. MATERIALS AND METHODS: C57/BL6 mice were infected with P. gingivalis or gingipains or P. gingivalis lipopolysaccharide (P. gingivalis LPS group) by tail vein injection for 8 weeks. The cognitive behaviour changes in mice, the histopathological changes in the hippocampus and cerebral cortex, the alternations of BBB permeability, and the changes in Mfsd2a and Cav-1 levels were measured. The mechanisms of Ddx3x-induced regulation on Mfsd2a by arginine-specific gingipain A (RgpA) in BMECs were explored. RESULTS: P. gingivalis and gingipains significantly promoted mice cognitive impairment, pathological changes in the hippocampus and cerebral cortex, increased BBB permeability, inhibited Mfsd2a expression and up-regulated Cav-1 expression. After RgpA stimulation, the permeability of the BBB model in vitro increased, and the Ddx3x/Mfsd2a/Cav-1 regulatory axis was activated. CONCLUSIONS: Gingipains may be one of the key virulence factors of P. gingivalis to impair cognition and enhance BBB permeability by the Ddx3x/Mfsd2a/Cav-1 axis.


Blood-Brain Barrier , Gingipain Cysteine Endopeptidases , Mice, Inbred C57BL , Porphyromonas gingivalis , Virulence Factors , Animals , Porphyromonas gingivalis/pathogenicity , Blood-Brain Barrier/microbiology , Mice , Virulence Factors/metabolism , Adhesins, Bacterial/metabolism , Male , Disease Models, Animal , Permeability , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/complications
7.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article En | MEDLINE | ID: mdl-34884507

Streptococcus pneumoniae is an important causative organism of respiratory tract infections. Although periodontal bacteria have been shown to influence respiratory infections such as aspiration pneumonia, the synergistic effect of S. pneumoniae and Porphyromonas gingivalis, a periodontopathic bacterium, on pneumococcal infections is unclear. To investigate whether P. gingivalis accelerates pneumococcal infections, we tested the effects of inoculating P. gingivalis culture supernatant (PgSup) into S. pneumoniae-infected mice. Mice were intratracheally injected with S. pneumoniae and PgSup to induce pneumonia, and lung histopathological sections and the absolute number and frequency of neutrophils and macrophages in the lung were analyzed. Proinflammatory cytokine/chemokine expression was examined by qPCR and ELISA. Inflammatory cell infiltration was observed in S. pneumoniae-infected mice and S. pnemoniae and PgSup mixed-infected mice, and mixed-infected mice showed more pronounced inflammation in lung. The ratios of monocytes/macrophages and neutrophils were not significantly different between the lungs of S. pneumoniae-infected mice and those of mixed-infected mice. PgSup synergistically increased TNF-α expression/production and IL-17 production compared with S. pneumoniae infection alone. We demonstrated that PgSup enhanced inflammation in pneumonia caused by S. pneumoniae, suggesting that virulence factors produced by P. gingivalis are involved in the exacerbation of respiratory tract infections such as aspiration pneumonia.


Bacteroidaceae Infections/complications , Inflammation/pathology , Lung/pathology , Neutrophil Infiltration/immunology , Pneumonia, Pneumococcal/pathology , Porphyromonas gingivalis/physiology , Streptococcus pneumoniae/physiology , Animals , Bacteroidaceae Infections/microbiology , Chemokines/metabolism , Cytokines/metabolism , Inflammation/etiology , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/epidemiology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology
8.
Environ Toxicol Pharmacol ; 88: 103755, 2021 Nov.
Article En | MEDLINE | ID: mdl-34662732

Periodontitis is a most prevalent and infectious multifactorial inflammatory disease and is characterized by the progressive destruction of the tooth-supporting tissues. Porphyromonas gingivalis, a Gram­negative oral anaerobe, mainly causes periodontitis and it is one of the most important risk factors responsible for aggravation of existing systemic diseases. Several experimental and clinical studies have shown the positive association between periodontitis and different forms of liver disease. Periodontal diseases increase the prevalence of non-alcoholic fatty liver diseases and cirrhosis. Infected periodontium and pathogens in the periodontal microenvironments release pathogen-associated molecular patterns such as peptidoglycan, lipopolysaccharides, gingipain, fimbria, bacterial DNA, etc, and damage-associated molecular patterns such as interleukins-1α, ß, - 8, and galectin-3, etc. These virulence factors and cytokines enter the bloodstream, disseminate into the whole body, and induce a variety of systemic pathological effects, including liver diseases (steatosis and fibrosis). Maintaining oral hygiene by scaling and root planning significantly improves liver damage in patients with periodontitis. Dentists and physicians should have more awareness in understanding the bidirectional nature of the relationship between oral and systemic diseases. Importantly, periodontitis condition aggravates simple fatty liver into fibrotic disease and therefore, the aim of this review is to understand the possible link between periodontitis and liver diseases.


Bacteroidaceae Infections/complications , Endotoxins/adverse effects , Liver Diseases/etiology , Periodontitis/complications , Porphyromonas gingivalis , Animals , Humans , Life Style , Oral Hygiene
9.
Biochem Biophys Res Commun ; 576: 80-85, 2021 10 22.
Article En | MEDLINE | ID: mdl-34482027

Epidemiological data have shown that periodontal bacterial infection, periodontitis, and oral squamous cell carcinoma have close relationship on the disease progress and risk. However, the specific role of periodontal microbes and their mechanism in the development of oral squamous cell carcinoma is not yet clear. In our previous work, metagenomic Illumina Mi-seq analysis was used to identify tstructure and abundance of periodontital microbiome. Accoding to the results, we used Porphyromonas.spp. and Fusobacterium.spp. as the periodontitis positive microbiota; Neisseria.spp and Corynebacterium.spp as periodontitis negative microbiota (their average relative abundance were >5%). These representative strains of the above genus were used to infect OSCC cells to explore their effect on tumor cell biology behavior, and detect the expression level of the gene in related to inflammation, migration, invasion and cell cycle. We find that periodontitis positive correlated microbiota had a promoting effect on the development of oral squamous cell carcinoma in vitro by regulating mRNA and protein expression of IL-6, IL-8, MMP-9 and Cyclin-D1. Periodontitis negative correlated microbiota had suppression effect on the development of oral squamous cell carcinoma in vitro analysis.


Head and Neck Neoplasms/microbiology , Microbiota , Periodontitis/microbiology , Squamous Cell Carcinoma of Head and Neck/microbiology , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Cell Line, Tumor , Cell Movement/physiology , Corynebacterium/genetics , Corynebacterium/isolation & purification , Fusobacterium Infections/complications , Fusobacterium Infections/microbiology , Fusobacterium Infections/pathology , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/isolation & purification , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Neisseria sicca/genetics , Neisseria sicca/isolation & purification , Neisseriaceae Infections/complications , Neisseriaceae Infections/microbiology , Neisseriaceae Infections/pathology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/isolation & purification , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
10.
BMB Rep ; 54(6): 323-328, 2021 06.
Article En | MEDLINE | ID: mdl-34078528

Periodontal diseases have been reported to have a multidirectional association with metabolic disorders. We sought to investigate the correlation between periodontitis and diabetes or fatty liver disease using HFD-fed obese mice inoculated with P. gingivalis. Body weight, alveolar bone loss, serological biochemistry, and glucose level were determined to evaluate the pathophysiology of periodontitis and diabetes. For the evaluation of fatty liver disease, hepatic nonalcoholic steatohepatitis (NASH) was assessed by scoring steatosis, inflammation, hepatocyte ballooning and the crucial signaling pathways involved in liver metabolism were analyzed. The C-reactive protein (CRP) level and NASH score in P. gingivalis-infected obese mice were significantly elevated. Particularly, the extensive lobular inflammation was observed in the liver of obese mice infected with P. gingivalis. Moreover, the expression of metabolic regulatory factors, including peroxisome proliferator-activated receptor γ (Pparγ) and the fatty acid transporter Cd36, was up-regulated in the liver of P. gingivalis-infected obese mice. However, inoculation of P. gingivalis had no significant influence on glucose homeostasis, insulin resistance, and hepatic mTOR/AMPK signaling. In conclusion, our results indicate that P. gingivalis can induce the progression of fatty liver disease in HFD-fed mice through the upregulation of CD36-PPARγ axis. [BMB Reports 2021; 54(6): 323-328].


Bacteroidaceae Infections/complications , CD36 Antigens/metabolism , Inflammation/pathology , Non-alcoholic Fatty Liver Disease/pathology , Obesity/physiopathology , PPAR gamma/metabolism , Porphyromonas gingivalis/physiology , Animals , Bacteroidaceae Infections/microbiology , CD36 Antigens/genetics , Diet, High-Fat , Disease Progression , Inflammation/metabolism , Inflammation/microbiology , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/microbiology , PPAR gamma/genetics
11.
Cancer Res ; 81(10): 2745-2759, 2021 05 15.
Article En | MEDLINE | ID: mdl-34003774

Porphyromonas gingivalis (P. gingivalis) is a keystone periodontal pathogen associated with various digestive cancers. However, whether P. gingivalis can promote colorectal cancer and the underlying mechanism associated with such promotion remains unclear. In this study, we found that P. gingivalis was enriched in human feces and tissue samples from patients with colorectal cancer compared with those from patients with colorectal adenoma or healthy subjects. Cohort studies demonstrated that P. gingivalis infection was associated with poor prognosis in colorectal cancer. P. gingivalis increased tumor counts and tumor volume in the ApcMin/+ mouse model and increased tumor growth in orthotopic rectal and subcutaneous carcinoma models. Furthermore, orthotopic tumors from mice exposed to P. gingivalis exhibited tumor-infiltrating myeloid cell recruitment and a proinflammatory signature. P. gingivalis promoted colorectal cancer via NLRP3 inflammasome activation in vitro and in vivo. NLRP3 chimeric mice harboring orthotopic tumors showed that the effect of NLRP3 on P. gingivalis pathogenesis was mediated by hematopoietic sources. Collectively, these data suggest that P. gingivalis contributes to colorectal cancer neoplasia progression by activating the hematopoietic NLRP3 inflammasome. SIGNIFICANCE: This study demonstrates that the periodontal pathogen P. gingivalis can promote colorectal tumorigenesis by recruiting myeloid cells and creating a proinflammatory tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/10/2745/F1.large.jpg.


Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neoplastic Stem Cells/pathology , Porphyromonas gingivalis/pathogenicity , Animals , Apoptosis , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Myeloid Cells/pathology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/microbiology , Prognosis , Survival Rate , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
12.
Dis Mon ; 67(9): 101166, 2021 Sep.
Article En | MEDLINE | ID: mdl-33663798

OBJECTIVES: The effect of antiretroviral therapy (ART) on the oral pathogenic microbes in human immunodeficiency virus-1 seropositive patients remains relatively unexplored. Thus, the present study assessed the effect of ART on the sub-gingival levels of 3 pathogenic microbes. MATERIALS AND METHODS: The study groups consisted of 60 human immunodeficiency virus-1 seropositive patients divided into 3 groups of 20 each. Group 1 had periodontitis and did not start with the ART. Group 2 had periodontitis and started with ART (Tenofovir Disoproxil Fumarate 300 mg + Lamivudine 300 mg + Efavirenz 600 mg) at least 6 months before the study. Group 3 with normal periodontium, and have not started ART. The sub-gingival loads of Cytomegalovirus, Epstein-Barr virus, and the Porphyromonas gingivalis levels were assessed, along with the CD4 counts. RESULTS: The cytomegalovirus load was highest in group 1, followed by groups 2, and 3 (p-value of 0.271). The Epstein-Barr load was highest for group 2, followed by group 3, and 1 (p-value of 0.022). The P.gingivalis load was highest in group 2, followed by groups 1 and 3, (p-value of 0.028). The Epstein-Barr and Cytomegalovirus counts were significantly higher (p-value < 0.02) when the CD4 counts were less than 500 cells/cu3. CONCLUSION: ART did not cause any significant reduction in the sub-gingival levels of any of the 3 examined microbes. Given the lack of any significant effect on the sub-gingival microbial loads by the ART, human immunodeficiency virus patients may require additional anti-microbial agents and regular mechanical plaque removal to maintain their periodontal status.


Antiretroviral Therapy, Highly Active , Cytomegalovirus/growth & development , HIV Infections , HIV-1/growth & development , Herpesvirus 4, Human/growth & development , Periodontitis , Porphyromonas gingivalis/growth & development , Adult , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , CD4 Lymphocyte Count , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Female , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/virology , Humans , Male , Periodontitis/complications , Periodontitis/microbiology , Periodontitis/virology , Periodontium/drug effects , Periodontium/microbiology , Periodontium/pathology , Periodontium/virology
14.
J Mol Neurosci ; 71(1): 89-100, 2021 Jan.
Article En | MEDLINE | ID: mdl-32557144

Hyperphosphorylated tau is the main component of neurofibrillary tangles and involved in the pathogenesis of Alzheimer's disease (AD). Increasing evidences suggest close associations between Porphyromonas gingivalis (P. gingivalis) and AD, but the relationship between P. gingivalis and tau hyperphosphorylation is still unclear. In this study, we investigated whether peripheral infection with P. gingivalis caused tau hyperphosphorylation by using wild Sprague-Dawley (SD) rats and HT-22 cells. The rats were injected with P. gingivalis suspension or phosphate-buffered saline 3 times per week. After 4 weeks or 12 weeks, the rats were sacrificed for analyzing systemic inflammation, neuroinflammation, and tau hyperphosphorylation. The results showed that the severity of phosphorylated tau at the AD-related sites Thr181 and Thr231 and the number of activated astrocytes were notably greater in the hippocampus of rats with P. gingivalis injection. And the levels of the inflammatory cytokines interleukin (IL)-1ß and IL-6 and tumor necrosis factor-α in serum and hippocampus were also increased in the rats with P. gingivalis injection. In addition, the activity of protein phosphatase 2A (PP2A) was significantly inhibited in the hippocampus of rats with P. gingivalis injection. In vitro, IL-1ß induced tau hyperphosphorylation by inhibiting the activity of PP2A in HT-22 cells and application of the PP2A promoter efficiently attenuated IL-1ß-induced tau hyperphosphorylation in HT-22 cells. These results indicated that P. gingivalis could induce tau hyperphosphorylation via, in part, attenuating the activity of PP2A through triggering systemic inflammation and neuroinflammation in wild-type SD rats.


Alzheimer Disease/microbiology , Bacteroidaceae Infections/metabolism , Porphyromonas gingivalis/pathogenicity , Protein Processing, Post-Translational , tau Proteins/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , Astrocytes/metabolism , Bacteremia/metabolism , Bacteroidaceae Infections/complications , Bacteroidaceae Infections/microbiology , Cell Line , Cytokines/analysis , Cytokines/blood , Disease Models, Animal , Enzyme Activation , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Inflammation , Male , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation , Phosphothreonine/metabolism , Porphyromonas gingivalis/physiology , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Rats , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood
15.
Phytomedicine ; 81: 153420, 2021 Jan.
Article En | MEDLINE | ID: mdl-33278781

BACKGROUND: Periodontitis is a common oral disease characterized as inflammation on gingival tissue and alveolar bone resorption. Spirulina maxima has been reported to have anti-oxidative and anti-inflammatory effects on gastric ulcers. However, its effects on gingival inflammation and alveolar bone resorption of periodontitis have not been studied. PURPOSE: This study was designed to investigate the effects of S. maxima on the P. gingivalis-induced periodontitis and to elucidate its mechanism. METHODS: The phycocyanin contents in S. maxima were identified by high-performance liquid chromatography. 8-week old SD rats were induced periodontitis by inoculation with P. gingivalis for 14 days. The rats were then orally treated with S. maxima 100, 200, 400 mg/kg, or indomethacin (IND, positive control) 5 mg/kg for an additional 14 days. Inflammatory responses, expressions of collagenases in gingival tissue, osteoclast formation and activation, alveolar bone resorption, osteogenesis-related markers, and BMP2/Smad signaling in alveolar bone were measured. RESULTS: Pro-inflammatory cytokines such as TNF-α, IL-1ß, IL-6, and inflammatory transcription factor NF-κB were decreased in gingival tissue by S. maxima administration. Also, myeloperoxidase (MPO) activity and matrix metalloproteinase (MMPs) expression were decreased by S. maxima administration. Conversely, S. maxima increased IL-4, anti-inflammatory cytokine from Th2 cells. The osteoprotegerin (OPG) / receptor activator of NF-κB ligand (RANKL) expression ratio, which represents osteoclast-osteoblast balance, was increased in S. maxima-treated groups. The alveolar bone loss and the number of TRAP-positive osteoclast cells were also declined in S. maxima-treated groups while the osteoblasts count was increased. Besides, in S. maxima-treated groups, the osteogenesis-related factors were promoted and BMP-2/Smad pathway was up-regulated in a periodontitis condition. CONCLUSION: S. maxima reduces periodontitis induced by P. gingivalis through anti-inflammatory effect and resultant reduction in bone loss, suggesting that S. maxima might be a potential agent for treating periodontitis.


Alveolar Bone Loss/drug therapy , Bacteroidaceae Infections/complications , Periodontitis/drug therapy , Periodontitis/microbiology , Porphyromonas gingivalis , Spirulina/chemistry , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/microbiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacteroidaceae Infections/drug therapy , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/drug effects , Male , Osteoblasts/drug effects , Osteoclasts/drug effects , Periodontitis/metabolism , Phycocyanin/analysis , Rats, Sprague-Dawley , Smad Proteins/metabolism
16.
PLoS One ; 15(10): e0241205, 2020.
Article En | MEDLINE | ID: mdl-33112888

To assess the influence of periodontal disease on cerebral hemorrhage and its clinical course, we examined the association of the serum IgG titer of periodontal pathogens with hemorrhage growth and 3-month outcome. We consecutively enrolled 115 patients with acute cerebral hemorrhage (44 females, aged 71.3 ± 13.1 years) and used ELISA to evaluate the serum IgG titers of 9 periodontal pathogens: Porphyromonas gingivalis, Aggregatibacter (A.) actinomycetemcomitans, Prevotella intermedia, Prevotella nigrescens, Fusobacterium (F.) nucleatum, Treponema denticola, Tannerella forsythensis, Campylobacter rectus, and Eikenella corrodens. Significant hematoma growth was defined as an increase in the volume of >33% or an absolute increase in the volume of >12.5 mL. A poor outcome was defined as a 3 or higher on the modified Rankin Scale. We observed hemorrhage growth in 13 patients (11.3%). Multivariate analysis revealed that increased IgG titers of A. actinomycetemcomitans independently predicted the elevated hemorrhage growth (odds ratio 5.26, 95% confidence interval 1.52-18.25, p = 0.01). Notably, augmented IgG titers of F. nucleatum but not A. actinomycetemcomitans led to a poorer 3-month outcome (odds ratio 7.86, 95% confidence interval 1.08-57.08, p = 0.04). Thus, we demonstrate that elevated serum IgG titers of A. actinomycetemcomitans are an independent factor for predicting cerebral hemorrhage growth and that high serum IgG titers of F. nucleatum may predict a poor outcome in patients with this disease. Together, these novel data reveal how systemic periodontal pathogens may affect stroke patients, and, should, therefore, be taken into consideration in the management and treatment of these individuals.


Antibodies, Bacterial/blood , Bacteroidaceae Infections/complications , Bacteroidaceae/immunology , Cerebral Hemorrhage/pathology , Immunoglobulin G/blood , Periodontal Diseases/microbiology , Aged , Bacteroidaceae/classification , Bacteroidaceae/pathogenicity , Bacteroidaceae Infections/microbiology , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/etiology , Female , Humans , Male , Periodontal Diseases/epidemiology , Periodontal Diseases/immunology , Prognosis
17.
BMJ Case Rep ; 13(9)2020 Sep 07.
Article En | MEDLINE | ID: mdl-32900736

Prevotella genus comprises of obligate anaerobic, gram-negative bacteria that are commensal organisms of oral cavity, gut and vaginal mucosa. Although many Prevotella species have well-established pathogenicity with respect to pulmonary infections, rarely has Prevotella pleuritidis been isolated as a cause of lung abscess. We present a rare case of left lower lobe lung abscess due to P. pleuritidis identified using next-generation sequencing of microbial cell-free DNA testing. A brief review of the literature regarding Prevotella species pulmonary infections, use of next-generation cell-free DNA testing early in the evaluation, antibiotic susceptibility and resistance is also a part of this report.


Bacteroidaceae Infections/complications , Lung Abscess/microbiology , Prevotella , Adult , Humans , Male
18.
Sci Rep ; 10(1): 14745, 2020 09 08.
Article En | MEDLINE | ID: mdl-32901057

The control of inflammation and infection is crucial for periodontal wound healing and regeneration. M101, an oxygen carrier derived from Arenicola marina, was tested for its anti-inflammatory and anti-infectious potential based on its anti-oxidative and tissue oxygenation properties. In vitro, no cytotoxicity was observed in oral epithelial cells (EC) treated with M101. M101 (1 g/L) reduced significantly the gene expression of pro-inflammatory markers such as TNF-α, NF-κΒ and RANKL in P. gingivalis-LPS stimulated and P. gingivalis-infected EC. The proteome array revealed significant down-regulation of pro-inflammatory cytokines (IL-1ß and IL-8) and chemokine ligands (RANTES and IP-10), and upregulation of pro-healing mediators (PDGF-BB, TGF-ß1, IL-10, IL-2, IL-4, IL-11 and IL-15) and, extracellular and immune modulators (TIMP-2, M-CSF and ICAM-1). M101 significantly increased the gene expression of Resolvin-E1 receptor. Furthermore, M101 treatment reduced P. gingivalis biofilm growth over glass surface, observed with live/dead analysis and by decreased P. gingivalis 16 s rRNA expression (51.7%) (p < 0.05). In mice, M101 reduced the clinical abscess size (50.2%) in P. gingivalis-induced calvarial lesion concomitant with a decreased inflammatory score evaluated through histomorphometric analysis, thus, improving soft tissue and bone healing response. Therefore, M101 may be a novel therapeutic agent that could be beneficial in the management of P. gingivalis associated diseases.


Anti-Inflammatory Agents/pharmacology , Bacteroidaceae Infections/complications , Brain Abscess/drug therapy , Inflammation/drug therapy , Oxygen/pharmacology , Polychaeta/chemistry , Skull/drug effects , Animals , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/microbiology , Brain Abscess/microbiology , Brain Abscess/pathology , Gingiva/chemistry , Gingiva/microbiology , Humans , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/isolation & purification , Signal Transduction , Skull/microbiology , Skull/pathology
19.
Sci Rep ; 10(1): 14606, 2020 09 03.
Article En | MEDLINE | ID: mdl-32884071

Porphyromonas gingivalis is a periodontal pathogen implicated in a range of pregnancy disorders that involve impaired spiral artery remodeling (ISAR) with or without fetal growth restriction (FGR). Using a rodent periodontitis model, we assessed the ability of P. gingivalis to produce ISAR and FGR in Sprague Dawley (SD) and Wistar (WIS) rats. Both infected SD and WIS rats developed ISAR, but only WIS rats developed FGR despite both rat strains having equivalent microbial loads within the placenta. Neither maternal systemic inflammation nor placental (fetal) inflammation was a feature of FGR in WIS rats. Unique to infected WIS rats, was loss of trophoblast cell density within the junctional zone of the placenta that was not present in SD tissues. In addition, infected WIS rats had a higher proportion of junctional zone trophoblast cells positive for cytoplasmic high temperature requirement A1 (Htra1), a marker of cellular oxidative stress. Our results show a novel phenomenon present in P. gingivalis-induced FGR, with relevance to human disease since dysregulation of placental Htra1 and placental oxidative stress are features of preeclamptic placentas and preeclampsia with FGR.


Arteries/pathology , Bacteroidaceae Infections/complications , Fetal Growth Retardation/etiology , Porphyromonas gingivalis/pathogenicity , Trophoblasts/pathology , Vascular Remodeling , Animals , Arteries/microbiology , Bacteroidaceae Infections/microbiology , Female , Fetal Growth Retardation/pathology , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Rats, Wistar , Trophoblasts/microbiology
20.
PLoS Biol ; 18(9): e3000825, 2020 09.
Article En | MEDLINE | ID: mdl-32886690

Microbial dysbiosis in the upper digestive tract is linked to an increased risk of esophageal squamous cell carcinoma (ESCC). Overabundance of Porphyromonas gingivalis is associated with shorter survival of ESCC patients. We investigated the molecular mechanisms driving aggressive progression of ESCC by P. gingivalis. Intracellular invasion of P. gingivalis potentiated proliferation, migration, invasion, and metastasis abilities of ESCC cells via transforming growth factor-ß (TGFß)-dependent Drosophila mothers against decapentaplegic homologs (Smads)/Yes-associated protein (YAP)/Transcriptional coactivator with PDZ-binding motif (TAZ) activation. Smads/YAP/TAZ/TEA domain transcription factor1 (TEAD1) complex formation was essential to initiate downstream target gene expression, inducing an epithelial-mesenchymal transition (EMT) and stemness features. Furthermore, P. gingivalis augmented secretion and bioactivity of TGFß through glycoprotein A repetitions predominant (GARP) up-regulation. Accordingly, disruption of either the GARP/TGFß axis or its activated Smads/YAP/TAZ complex abrogated the tumor-promoting role of P. gingivalis. P. gingivalis signature genes based on its activated effector molecules can efficiently distinguish ESCC patients into low- and high-risk groups. Targeting P. gingivalis or its activated effectors may provide novel insights into clinical management of ESCC.


Bacteroidaceae Infections/complications , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Porphyromonas gingivalis/physiology , Transforming Growth Factor beta/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Animals , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/mortality , Bacteroidaceae Infections/pathology , Cells, Cultured , Disease Progression , Drosophila , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/microbiology , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/microbiology , Esophageal Squamous Cell Carcinoma/mortality , Female , Follow-Up Studies , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Signal Transduction/physiology , Smad Proteins/metabolism , Survival Analysis , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , YAP-Signaling Proteins
...