Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
J Oral Biosci ; 66(2): 365-372, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38579987

ABSTRACT

OBJECTIVES: Periodontitis, commonly associated with Porphyromonas gingivalis (Pg), involves intricate alterations of oral intercellular interactions, in which extracellular vesicles (EVs) play a pivotal role. The understanding of the miRNA profiles in the EVs derived from Pg-infected cells (Pg-EVs) remains incomplete despite acknowledging their importance in intercellular communication during periodontitis. Therefore, our objective was to identify and characterize the miRNAs enriched in Pg-EVs. METHODS: Microarray analysis was conducted to examine the miRNA profiles in the EVs derived from Pg-infected THP-1 cells. We compared the identified miRNAs with those upregulated in the EVs after stimulation with LPS. Additionally, we explored how inhibiting TLR signaling during Pg infection affects the transcription of specific miRNAs. We investigated the unique sequence motifs specific to the miRNAs concentrated in Pg-EVs. RESULTS: The levels of eleven miRNAs, including miR-155, were increased in Pg-EVs compared with those elevated after LPS stimulation. The Pg-induced miR-155 upregulation via TLR2 but not TLR4 signaling suggests the influence of TLR signaling on the miRNA composition of EVs. Furthermore, the miRNAs upregulated in Pg-EVs contained AGAGGG and GRGGSGC sequence motifs. CONCLUSIONS: Our findings demonstrate that Pg-induced alterations in EV-containing miRNA composition occur in a TLR4-independent manner. Notably, the concentrated miRNAs in Pg-EVs harbor specific motifs with a high G + C content within their sequences. The upregulation of specific miRNAs in EVs under infectious conditions suggests the influence of both innate immune receptor signals and miRNA sequence characteristics.


Subject(s)
Extracellular Vesicles , MicroRNAs , Porphyromonas gingivalis , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/genetics , Humans , Signal Transduction , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/genetics , Lipopolysaccharides/pharmacology , Periodontitis/microbiology , Periodontitis/metabolism , Periodontitis/genetics , Up-Regulation , Microarray Analysis
2.
Int J Mol Sci ; 25(8)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38674094

ABSTRACT

Porphyromonas gingivalis (Pg) and its gingipain proteases contribute to Alzheimer's disease (AD) pathogenesis through yet unclear mechanisms. Cellular secretion of small extracellular vesicles or exosomes (EXO) increases with aging as part of the senescence-associated secretory phenotype (SASP). We have shown that EXO isolated from Pg-infected dendritic cells contain gingipains and other Pg antigens and transmit senescence to bystander gingival cells, inducing alveolar bone loss in mice in vivo. Here, EXO were isolated from the gingiva of mice and humans with/without periodontitis (PD) to determine their ability to penetrate the blood-brain barrier (BBB) in vitro and in vivo. PD was induced by Pg oral gavage for 6 weeks in C57B6 mice. EXO isolated from the gingiva or brain of donor Pg-infected (PD EXO) or control animals (Con EXO) were characterized by NTA, Western blot, and TEM. Gingival PD EXO or Con EXO were labeled and injected into the gingiva of uninfected WT mouse model. EXO biodistribution in brains was tracked by an in vivo imaging system (IVIS) and confocal microscopy. The effect of human PD EXO on BBB integrity and permeability was examined using TEER and FITC dextran assays in a human in vitro 3D model of the BBB. Pg antigens (RGP and Mfa-1) were detected in EXO derived from gingival and brain tissues of donor Pg-infected mice. Orally injected PD EXO from donor mice penetrated the brains of recipient uninfected mice and colocalized with hippocampal microglial cells. IL-1ß and IL-6 were expressed in human PD EXO and not in Con EXO. Human PD EXO promoted BBB permeability and penetrated the BBB in vitro. This is the first demonstration that microbial-induced EXO in the oral cavity can disseminate, cross the BBB, and may contribute to AD pathogenesis.


Subject(s)
Blood-Brain Barrier , Extracellular Vesicles , Gingiva , Periodontitis , Porphyromonas gingivalis , Blood-Brain Barrier/metabolism , Animals , Humans , Mice , Extracellular Vesicles/metabolism , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Periodontitis/microbiology , Periodontitis/metabolism , Periodontitis/pathology , Gingiva/metabolism , Gingiva/microbiology , Mice, Inbred C57BL , Male , Exosomes/metabolism , Female , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/metabolism
3.
ACS Infect Dis ; 10(4): 1152-1161, 2024 04 12.
Article in English | MEDLINE | ID: mdl-38442009

ABSTRACT

Periodontitis, a chronic infectious disease in periodontal tissues, is characterized by an imbalance of alveolar bone resorption and remodeling, which eventually results in tooth loosening and even tooth loss. The etiology of periodontitis is polymicrobial synergy and dysbiosis, in which Porphyromonas gingivalis (P. gingivalis) is one of the primary pathogens responsible for periodontitis progression. The interplay of EphrinB2/EphB4 is crucial for osteoblast-osteoclast communication during bone remodeling and healing. This study investigates the mechanism of EphB4/EphrinB2 transduction modulating osteogenesis inhibition and bone resorption in periodontitis induced by P. gingivalis. An in vivo model of chronic periodontitis provoked by P. gingivalis was constructed, the inflammation and bone resorption were evaluated. The expression of EphB4 and EphrinB2 proteins in periodontal tissues was detected, which was also evaluated, respectively, in osteoblasts and osteoclasts infected with P. gingivalis in vitro. Then, a simulated coculture model of osteoblasts and osteoclasts was established to activate the forward and reverse pathways of EphB4/EphrinB2 with P. gingivalis infection. This study showed that P. gingivalis infection promoted alveolar bone resorption in rats and enhanced EphB4 and EphrinB2 expression in periodontal tissues. EphB4 and molecules associated with osteogenesis in osteoblasts infected with P. gingivalis were inhibited, while EphrinB2 and osteoclast differentiation-related markers in osteoclasts were activated. In conclusion, this study suggested that EphB4/EphrinB2 proteins were involved in alveolar bone remodeling in the process of periodontitis induced by P. gingivalis infection. Moreover, attenuated EphB4/EphrinB2 with P. gingivalis infection weakened osteoblast activity and enhanced osteoclast activity.


Subject(s)
Bone Resorption , Periodontitis , Receptor, EphB2 , Receptor, EphB4 , Animals , Rats , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/microbiology , Osteoclasts/metabolism , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Signal Transduction , Receptor, EphB2/metabolism , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology
4.
Cell Prolif ; 57(6): e13609, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38351596

ABSTRACT

The association between Porphyromonas gingivalis infection and oral squamous cell carcinoma (OSCC) has been established by numerous epidemiological studies. However, the underlying mechanism specific to this connection remains unclear. By bioinformatical analysis, we identified ZFP36 as a potentially significant co-expressed gene in both the OSCC gene database and the persistent infection model of P. gingivalis. To further investigate the role of ZFP36, we established a cell model that human immortalized oral epithelial cells (HIOECs) that were sustainedly infected by P. gingivalis (MOI = 1) for a duration of 30 weeks. Our findings indicated that sustained infection with P. gingivalis inhibited the expression of ZFP36 protein and induced changes in the biological behaviour of HIOECs. The mechanism investigation demonstrated the potential role of ZFP36 in regulating the cancer-related biological behaviour of HIOECs. Subsequent studies revealed that highly expressed CCAT1 could serve as a molecular scaffold in the formation of the ZFP36/CCAT1/MK2 complex. This complex formation enhanced the binding abundance of MK2 and ZFP36, thereby promoting the inhibition of ZFP36 protein phosphorylation. To summarize, low expression of ZFP36 protein under persistent P. gingivalis infection enhances the cancer-related biological behaviour of HIOECs.


Subject(s)
Bacteroidaceae Infections , Epithelial Cells , Porphyromonas gingivalis , Tristetraprolin , Humans , Porphyromonas gingivalis/pathogenicity , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/metabolism , Tristetraprolin/metabolism , Tristetraprolin/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/microbiology , Mouth Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Phosphorylation
5.
Can J Physiol Pharmacol ; 100(7): 679-688, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35442801

ABSTRACT

Endothelin-1 (ET-1), the most potent vasoconstrictor identified to date, contributes to cerebrovascular dysfunction and brain ET-1 levels were shown to be related to Alzheimer's disease and related dementias (ADRD) progression. ET-1 also contributes to neuroinflammation, especially in infections of the central nervous system. Recent studies causally linked chronic periodontal infection with an opportunistic anaerobic bacterium Porphyromonas gingivalis (Coykendall et al.) Shah & Collins to AD development. Thus, the goal of the study was to determine the impact of P. gingivalis infection on the ET system and cell senescence in brain microvascular endothelial cells. Cells were infected with a multiplicity of infection 50 P. gingivalis with and without extracellular ATP-induced oxidative stress for 24 h. Cell lysates were collected for analysis of endothelin A receptor (ETA)/endothelin B receptor (ETB) receptor as well as senescence markers. ET-1 levels in cell culture media were measured with enzyme-linked immunosorbent assay. P. gingivalis infection increased ET-1 (pg/mL) secretion, as well as the ETA receptor expression, whereas decreased lamin A/C expression compared to control. Tight junction protein claudin-5 was also decreased under these conditions. ETA or ETB receptor blockade during infection did not affect ET-1 secretion or the expression of cell senescence markers. Current findings suggest that P. gingivalis infection may compromise endothelial integrity and activate the ET system.


Subject(s)
Bacteroidaceae Infections , Endothelial Cells , Porphyromonas gingivalis , Bacteroidaceae Infections/metabolism , Base Composition , Brain/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelin-1/metabolism , Endothelins , Phylogeny , Porphyromonas gingivalis/metabolism , RNA, Ribosomal, 16S , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Receptors, Endothelin/metabolism , Sequence Analysis, DNA
6.
Bioengineered ; 13(2): 2336-2345, 2022 02.
Article in English | MEDLINE | ID: mdl-35034548

ABSTRACT

Long non-coding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) is a novel pro-inflammatory factor in severe human diseases. Since inflammatory plays important roles in periodontitis progression, we aimed to explore the role of NEAT1 in chronic periodontitis (CP) in vitro. We established a periodontitis cell model was established by Porphyromonas gingivalis lipopolysaccharide (Pg-LPS)-induced periodontal ligament cells (PDLCs). Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to detect the expression of NEAT1, microRNA (miR)-200c-3p, and tumor necrosis factor receptor-associated factor 6 (TRAF6). Cell viability, inflammatory factors, and protein expression of Bcl-2, Bax, and TRAF6 were analyzed by MTT, enzyme-linked immunosorbent assay, and Western blot. The target relationships among NEAT1, miR-200c-3p, and TRAF6 were predicted by the StarBase/TargetScan software, and further validated by dual-luciferase reporter assay. In this research, NEAT1 is up-regulated in CP tissues and periodontitis model group. Silencing of NEAT1 and over-expression of miR-200c-3p enhanced cell viability and repressed apoptosis in the periodontitis model group. NEAT1 targets miR-200c-3p, and miR-200c-3p further targets TRAF6. MiR-200c-3p inhibitor or over-expression of TRAF6 reversed the promoting effect of NEAT1 knockdown on cell viability, and the inhibiting effects on inflammatory cytokines and cell apoptosis. Consequently, the silencing of NEAT1 inhibits inflammation and apoptosis via targeting miR-200c-3p/TRAF6 axis, thereby contributing to alleviate the progression of CP. This finding could provide an underlying target for the treatment of CP.


Subject(s)
Bacteroidaceae Infections/metabolism , Chronic Periodontitis/metabolism , Models, Biological , Periodontal Ligament/metabolism , Porphyromonas gingivalis/metabolism , RNA, Long Noncoding/metabolism , Bacteroidaceae Infections/microbiology , Chronic Periodontitis/microbiology , Female , Humans , Male , Periodontal Ligament/microbiology , RNA, Long Noncoding/genetics
7.
Biomed Res Int ; 2022: 9770899, 2022.
Article in English | MEDLINE | ID: mdl-35028318

ABSTRACT

Periodontitis is a chronic inflammatory disease caused by periodontopathogenic bacteria that form biofilms in periodontal pockets. The gingival epithelium acts as the first physical barrier in fighting attacks by periodontopathogenic pathogens, such as the primary etiological agent Porphyromonas gingivalis, and various exogenous chemicals, as well as regulates the local innate immune responses. Therefore, the development of novel oral care products to inhibit inflammatory reactions caused by bacterial infection and protect the gingival epithelium is necessary. Juncus effusus L. has generally been used as an indigenous medicine, such as a diuretic, an antipyretic, and an analgesic, in ancient practice. In this study, we examined the effects of a water extract from J. effusus L. on the inhibition of the inflammatory reaction elicited by bacterial infection and protection of the oral epithelium by chemical irritation. Pretreatment of oral epithelial cells with the water extract from J. effusus L. significantly reduced P. gingivalis or its lipopolysaccharide- (LPS-) mediated production of chemokines (interleukin-8 and C-C-chemokine ligand20) in a concentration-dependent manner with comparable to or greater effects than epigallocatechin gallate and protected oral epithelial cells from injury by chemical irritants, cetylpyridinium chloride, and benzethonium chloride. Moreover, the water extract from J. effusus L. in the presence of antimicrobial agents or antifibrinolytics already used as ingredients in mouthwash could significantly reduce the production of chemokines from P. gingivalis LPS-stimulated oral epithelial cells in a concentration-dependent manner. These findings suggest that the water extract from J. effusus L. is potentially useful for oral care to prevent oral infections, such as periodontal infections, and maintain oral epithelial function.


Subject(s)
Anti-Inflammatory Agents , Keratinocytes/metabolism , Magnoliopsida/chemistry , Mouth Mucosa/metabolism , Plant Extracts , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/prevention & control , Cell Line, Transformed , Humans , Keratinocytes/pathology , Mouth Mucosa/pathology , Periodontitis/metabolism , Periodontitis/pathology , Periodontitis/prevention & control , Plant Extracts/chemistry , Plant Extracts/pharmacology , Porphyromonas gingivalis/metabolism
8.
Gerodontology ; 39(2): 139-147, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33599317

ABSTRACT

OBJECTIVE: This paper describes the effect of Porphyromonas gingivalis (P gingivalis) lipopolysaccharide (LPS) on the expression of interleukin-6 (IL-6) and C-C motif chemokine ligand 2 (CCL2) in cultured hCMEC/D3 human brain microvascular endothelial cells. BACKGROUND: P gingivalis is one of the important pathogens in periodontitis, and periodontitis is a risk factor for brain disorders including cerebrovascular diseases and Alzheimer's disease. However, the mechanisms underlying the pathogenesis of P gingivalis-mediated brain diseases are incompletely understood. Effects of P gingivalis LPS on brain endothelial cells are not known well. METHODS: The hCMEC/D3 human brain microvascular endothelial cells were cultured and treated with P gingivalis LPS. The expression of IL-6 and CCL2 mRNA and protein was examined using quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. Effect of inhibitors of Toll-like receptor (TLR) 2, TLR4, nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) was also investigated. Phosphorylation of NF-κB p65, p38 MAPK and JNK was examined using Western blotting. RESULTS: P gingivalis LPS-induced mRNA and protein expression of IL-6 and CCL2 in hCMEC/D3 cells in a concentration-dependent manner at the concentration of 0.5-50 µg/mL. Maximal mRNA expression of IL-6 and CCL2 was found 2 and 4 hours after stimulation, respectively. Induction of IL-6 and CCL2 by P gingivalis LPS was almost completely inhibited by pretreatment of cells with TLR4 inhibitor but not by TLR2 inhibitor. Treatment of cells with P gingivalis LPS for up to 2 hours induced phosphorylation of NF-κB p65, p38 MAPK and JNK. IL-6 induction was decreased by pretreatment of cells with NF-κB inhibitor SN50 or p38 MAPK inhibitor SB203580, while CCL2 induction was reduced by SN50 or JNK inhibitor SP600125. CONCLUSIONS: IL-6 and CCL2 produced upon P gingivalis LPS stimulation may contribute to the inflammatory reactions in brain endothelial cells and subsequent neurological disorders such as cerebrovascular and Alzheimer's diseases.


Subject(s)
Bacteroidaceae Infections/metabolism , Brain/cytology , Chemokine CCL2/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Porphyromonas gingivalis , Bacteroidaceae Infections/immunology , Cells, Cultured , Chemokines/metabolism , Endothelial Cells/metabolism , Humans , Ligands , NF-kappa B/metabolism , Periodontitis/complications , RNA, Messenger/genetics , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Pathol Oncol Res ; 27: 1609976, 2021.
Article in English | MEDLINE | ID: mdl-34955686

ABSTRACT

Purpose: The present study focused on exploring the associations of Porphyromonas gingivalis (P. gingivalis) infection and low Beclin1 expression with clinicopathological parameters and survival of esophageal squamous cell carcinoma (ESCC) patients, so as to illustrate its clinical significance and prognostic value. Methods: Immunohistochemistry (IHC) was used to detect P. gingivalis infection status and Beclin1 expression in 370 ESCC patients. The chi-square test was adopted to illustrate the relationship between categorical variables, and Cohen's kappa coefficient was used for correlation analysis. Kaplan-Meier survival curves with the log-rank test were used to analyse the correlation of P. gingivalis infection and low Beclin1 expression with survival time. The effects of P. gingivalis infection and Beclin1 downregulation on the proliferation, migration and antiapoptotic abilities of ESCC cells in vitro were detected by Cell Counting Kit-8, wound healing and flow cytometry assays. For P. gingivalis infection of ESCC cells, cell culture medium was replaced with antibiotic-free medium when the density of ESCC cells was 70-80%, cells were inoculated with P. gingivalis at a multiplicity of infection (MOI) of 10. Result: P. gingivalis infection was negatively correlated with Beclin1 expression in ESCC tissues, and P. gingivalis infection and low Beclin1 expression were associated with differentiation status, tumor invasion depth, lymph node metastasis, clinical stage and prognosis in ESCC patients. In vitro experiments confirmed that P. gingivalis infection and Beclin1 downregulation potentiate the proliferation, migration and antiapoptotic abilities of ESCC cells (KYSE150 and KYSE30). Our results provide evidence that P. gingivalis infection and low Beclin1 expression were associated with the development and progression of ESCC. Conclusion: Long-term smoking and alcohol consumption causes poor oral and esophageal microenvironments and ESCC patients with these features were more susceptible to P. gingivalis infection and persistent colonization, and exhibited lower Beclin1 expression, worse prognosis and more advanced clinicopathological features. Our findings indicate that effectively eliminating P. gingivalis colonization and restoring Beclin1 expression in ESCC patients may contribute to preventation and targeted treatment, and yield new insights into the aetiological research on ESCC.


Subject(s)
Bacteroidaceae Infections/microbiology , Beclin-1/metabolism , Esophageal Neoplasms/microbiology , Esophageal Squamous Cell Carcinoma/microbiology , Porphyromonas gingivalis/isolation & purification , Apoptosis , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/mortality , Bacteroidaceae Infections/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Male , Middle Aged , Prognosis
10.
Int J Mol Sci ; 22(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34769513

ABSTRACT

The non-enzymatic addition of glucose (glycation) to circulatory and tissue proteins is a ubiquitous pathophysiological consequence of hyperglycemia in diabetes. Given the high incidence of periodontitis and diabetes and the emerging link between these conditions, it is of crucial importance to define the basic virulence mechanisms employed by periodontopathogens such as Porphyromonas gingivalis in mediating the disease process. The aim of this study was to determine whether glycated proteins are more easily utilized by P. gingivalis to stimulate growth and promote the pathogenic potential of this bacterium. We analyzed the properties of three commonly encountered proteins in the periodontal environment that are known to become glycated and that may serve as either protein substrates or easily accessible heme sources. In vitro glycated proteins were characterized using colorimetric assays, mass spectrometry, far- and near-UV circular dichroism and UV-visible spectroscopic analyses and SDS-PAGE. The interaction of glycated hemoglobin, serum albumin and type one collagen with P. gingivalis cells or HmuY protein was examined using spectroscopic methods, SDS-PAGE and co-culturing P. gingivalis with human keratinocytes. We found that glycation increases the ability of P. gingivalis to acquire heme from hemoglobin, mostly due to heme sequestration by the HmuY hemophore-like protein. We also found an increase in biofilm formation on glycated collagen-coated abiotic surfaces. We conclude that glycation might promote the virulence of P. gingivalis by making heme more available from hemoglobin and facilitating bacterial biofilm formation, thus increasing P. gingivalis pathogenic potential in vivo.


Subject(s)
Bacteroidaceae Infections/metabolism , Diabetes Complications/physiopathology , Erythrocytes/metabolism , Heme/metabolism , Hemoglobins/metabolism , Periodontitis/microbiology , Porphyromonas gingivalis/pathogenicity , Animals , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Glycosylation , Hemeproteins/chemistry , Hemoglobins/chemistry , Horses , Periodontitis/pathology , Porphyromonas gingivalis/isolation & purification , Porphyromonas gingivalis/metabolism
11.
Anaerobe ; 72: 102458, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34547426

ABSTRACT

Porphyromonas gingivalis inhibits the release of CXCL8 by gingival epithelial cells and reduces their proliferation. We previously reported that Bifidocaterium sp. and Lactobacillus sp. immunomodulate gingival epithelial cells response to this periodontal pathogen, but their effects on re-epithelialization properties are still unknown. Herein we explored these activities of potential probiotics on gingival epithelial cells and clarified their mechanisms. The immortalized OBA-9 lineage was used to perform in vitro scratches. Twelve clinical isolates and commercially available strains of Bifidobacterium sp. and Lactobacillus sp. were screened. L. casei 324 m and B. pseudolongum 1191A were selected to perform mechanistic assays with P. gingivalis W83 infection and the following parameters were measured: percentage of re-epithelialization by DAPI immunofluorescence area measurement; cell number by Trypan Blue exclusion assay; CXCL8 regulation by ELISA and RT-qPCR; and expression of CXCL8 cognate receptors-CXCR1 and CXCR2 by Flow Cytometry. Complementary mechanistic assays were performed with CXCL8, in the presence or absence of the CXCR1/CXCR2 inhibitor-reparixin. L. casei 324 m and B. pseudolongum 1191A enhanced re-epithelialization/cell proliferation as well as inhibited the harmful effects of P. gingivalis W83 on these activities through an increase in the expression and release of CXCL8 and in the number of cells positive for CXCR1/CXCR2. Further, we revealed that the beneficial effects of these potential probiotics were dependent on activation of the CXCL8-CXCR1/CXCR2 axis. The current findings indicate that these potential probiotics strains may improve wound healing in the context of the periodontal tissues by a CXCL8 dependent mechanism.


Subject(s)
Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Host-Pathogen Interactions , Microbial Interactions , Porphyromonas gingivalis , Probiotics/administration & dosage , Re-Epithelialization , Biomarkers , Cell Line , Gene Expression Regulation , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Wound Healing
12.
J Alzheimers Dis ; 83(2): 665-681, 2021.
Article in English | MEDLINE | ID: mdl-34334391

ABSTRACT

BACKGROUND: Studies have reported that synaptic failure occurs before the Alzheimer's disease (AD) onset. The systemic Porphyromonas gingivalis (P. gingivalis) infection is involved in memory decline. We previously showed that leptomeningeal cells, covering the brain, activate glial cells by releasing IL-1ß in response to systemic inflammation. OBJECTIVE: In the present study, we focused on the impact of leptomeningeal cells on neurons during systemic P. gingivalis infection. METHODS: The responses of leptomeningeal cells and cortical neurons to systemic P. gingivalis infection were examined in 15-month-old mice. The mechanism of IL-1ß production by P. gingivalis infected leptomeningeal cells was examined, and primary cortical neurons were treated with P. gingivalis infected leptomeningeal cells condition medium (Pg LCM). RESULTS: Systemic P. gingivalis infection increased the expression of IL-1ß in leptomeninges and reduced the synaptophysin (SYP) expression in leptomeninges proximity cortex in mice. Leptomeningeal cells phagocytosed P. gingivalis resulting in lysosomal rupture and cathepsin B (CatB) leakage. Leaked CatB mediated NLRP3 inflammasome activation inducing IL-1ß secretion in leptomeningeal cells. Pg LCM decreased the expression of synaptic molecules, including SYP, which was inhibited by an IL-1 receptor antagonist pre-treatment. CONCLUSION: These observations demonstrate that P. gingivalis infection is involved in synaptic failure by inducing CatB/NLRP3 inflammasome-mediated IL-1ß production in leptomeningeal cells. The periodontal bacteria-induced synaptic damage may accelerate the onset and cognitive decline of AD.


Subject(s)
Bacteroidaceae Infections/metabolism , Meninges , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Porphyromonas gingivalis/metabolism , Animals , Cathepsin B/metabolism , Female , Humans , Inflammation/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism
13.
Innate Immun ; 27(4): 324-342, 2021 05.
Article in English | MEDLINE | ID: mdl-34018827

ABSTRACT

Periodontitis is induced by periodontal dysbiosis characterized by the predominance of anaerobic species. TLRs constitute the classical pathway for cell activation by infection. Interestingly, the Toll/IL-1 receptor homology domain adapters initiate signaling events, leading to the activation of the expression of the genes involved in the host immune response. The aim of this study was to evaluate the effects of Porphyromonas gingivalis on the expression and protein-protein interactions among five TIR adapters (MAL, MyD88, TRIF, TRAM and SARM) in gingival epithelial cells and endothelial cells. It was observed that P. gingivalis is able to modulate the signaling cascades activated through its recognition by TLR4/2 in gingival epithelial cells and endothelial cells. Indeed, MAL-MyD88 protein-protein interactions associated with TLR4 was the main pathway activated by P. gingivalis infection. When transient siRNA inhibition was performed, cell viability, inflammation, and cell death induced by infection decreased and such deleterious effects were almost absent when MAL or TRAM were targeted. This study emphasizes the role of such TIR adapter proteins in P. gingivalis elicited inflammation and the precise evaluation of TIR adapter protein interactions may pave the way for future therapeutics in both periodontitis and systemic disease with a P. gingivalis involvement, such as atherothrombosis.


Subject(s)
Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Gingivitis/metabolism , Gingivitis/microbiology , Porphyromonas gingivalis , Receptors, Interleukin-1/genetics , Toll-Like Receptors/genetics , Adolescent , Adult , Aged , Cell Survival , Dysbiosis , Female , Gene Expression Regulation , Humans , Male , Middle Aged , RNA, Small Interfering/pharmacology , Receptors, Interleukin-1/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptors/metabolism , Young Adult
14.
J Mol Neurosci ; 71(1): 89-100, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32557144

ABSTRACT

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.


Subject(s)
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.
J Neurochem ; 158(3): 724-736, 2021 08.
Article in English | MEDLINE | ID: mdl-32441775

ABSTRACT

Cerebrovascular-related amyloidogenesis is found in over 80% of Alzheimer's disease (AD) cases, and amyloid ß (Aß) generation is increased in the peripheral macrophages during infection of Porphyromonas gingivalis (P. gingivalis), a causal bacterium for periodontitis. In this study, we focused on receptor for advanced glycation end products (RAGE), the key molecule involves in Aß influx after P. gingivalis infection to test our hypothesis that Aß transportation from periphery into the brain, known as "Aß influx," is enhanced by P. gingivalis infection. Using cultured hCMEC/D3 cell line, in comparison to uninfected cells, directly infection with P. gingivalis (multiplicity of infection, MOI = 5) significantly increased a time-dependent RAGE expression resulting in a dramatic increase in Aß influx in the hCMEC/D3 cells; the P. gingivalis-up-regulated RAGE expression was significantly decreased by NF-κB and Cathepsin B (CatB)-specific inhibitors, and the P.gingivalis-increased IκBα degradation was significantly decreased by CatB-specific inhibitor. Furthermore, the P. gingivalis-increased Aß influx was significantly reduced by RAGE-specific inhibitor. Using 15-month-old mice (C57BL/6JJmsSlc, female), in comparison to non-infection mice, systemic P. gingivalis infection for three consecutive weeks (1 × 108  CFU/mouse, every 3 days, intraperitoneally) significantly increased the RAGE expression in the CD31-positive endothelial cells and the Aß loads around the CD31-positive cells in the mice's brains. The RAGE expression in the CD31-positive cells was positively correlated with the Aß loads. These observations demonstrate that the up-regulated RAGE expression in cerebral endothelial cells mediates the Aß influx after P. gingivalis infection, and CatB plays a critical role in regulating the NF-κB/RAGE expression. Cover Image for this issue: https://doi.org/10.1111/jnc.15073.


Subject(s)
Amyloid beta-Peptides/metabolism , Bacteroidaceae Infections/metabolism , Cerebral Cortex/metabolism , Endothelial Cells/metabolism , Peptide Fragments/metabolism , Porphyromonas gingivalis , Receptor for Advanced Glycation End Products/biosynthesis , Animals , Cerebral Cortex/microbiology , Cerebrovascular Circulation/physiology , Cerebrovascular Disorders/metabolism , Cerebrovascular Disorders/microbiology , Endothelial Cells/microbiology , Female , Mice , Mice, Inbred C57BL , Up-Regulation/physiology
16.
FEBS J ; 288(5): 1479-1495, 2021 03.
Article in English | MEDLINE | ID: mdl-32681704

ABSTRACT

Several studies have shown a clear association between periodontal disease and increased risk of cardiovascular disease. Porphyromonas gingivalis (Pg), a key oral pathogen, and its cell surface-expressed gingipains, induce oedema in a zebrafish larvae infection model although the mechanism of these vascular effects is unknown. Here, we aimed to determine whether Pg-induced vascular damage is mediated by gingipains. In vitro, human endothelial cells from different vascular beds were invaded by wild-type (W83) but not gingipain-deficient (ΔK/R-ab) Pg. W83 infection resulted in increased endothelial permeability as well as decreased cell surface abundance of endothelial adhesion molecules PECAM-1 and VE-cadherin compared to infection with ΔK/R-ab. In agreement, when transgenic zebrafish larvae expressing fluorescently labelled PECAM-1 or VE-cadherin were systemically infected with W83 or ΔK/R-ab, a significant reduction in adhesion molecule fluorescence was observed specifically in endothelium proximal to W83 bacteria through a gingipain-dependent mechanism. Furthermore, this was associated with increased vascular permeability in vivo when assessed by dextran leakage microangiography. These data are the first to show that Pg directly mediates vascular damage in vivo by degrading PECAM-1 and VE-cadherin. Our data provide a molecular mechanism by which Pg might contribute to cardiovascular disease.


Subject(s)
Bacteroidaceae Infections/etiology , Cardiomegaly/etiology , Edema/etiology , Endothelial Cells/drug effects , Gingipain Cysteine Endopeptidases/toxicity , Porphyromonas gingivalis/pathogenicity , Animals , Animals, Genetically Modified , Antigens, CD/genetics , Antigens, CD/metabolism , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/pathology , Cadherins/genetics , Cadherins/metabolism , Capillary Permeability/drug effects , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Edema/genetics , Edema/metabolism , Edema/pathology , Embryo, Nonmammalian , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fluorescein Angiography , Gene Expression/drug effects , Genes, Reporter , Gingipain Cysteine Endopeptidases/biosynthesis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Larva/drug effects , Larva/microbiology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/metabolism , Primary Cell Culture , Signal Transduction , Zebrafish
17.
Arch Oral Biol ; 121: 104963, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33157496

ABSTRACT

OBJECTIVE: The purpose of the present study was to explore the sequential changes in the cellular metabolism in gingival fibroblasts (GFs) in response toPorphyromonas gingvalis (P. gingivalis) ATCC33277 infection. DESIGN: GFs were treated withP. gingivalis at the MOI of 50 for 4, 24 and 48 h to mimic the early, medium, and late phase in the bacterial infection. LDH assay and cell counting kit-8 were utilized to explore cell death and proliferation. Real-time PCR was utilized to explore the gene transcription of pro-inflammatory genes. The relative levels of biomolecules in GFs were measured by gas chromatography-mass spectrometry. Principal component analysis and orthogonal partial least-squares-discriminant analysis were performed to visualize the metabolic difference among experimental groups. In addition, pathway analysis was conducted regarding differential metabolites in GFs. RESULTS: P. gingivalis infection triggered significant gene transcription of IL-1ß, IL 6, MCP 1, and MMP 1 in GFs. In addition, P. gingivalis stimulated cell proliferation of GFs at MOI of 10, 50 and 250. Moreover, P. gingivalis triggered significant cell death at higher MOI. 69, 173 and 148 metabolites were qualitatively detected at 4, 24 and 48 h after P. gingivalis infection respectively in GFs, showing a sequential change of different phase. Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that ATP-binding cassette transporters, glutathione, purine and pyrimidine metabolism was significantly altered in different phase. CONCLUSIONS: Human GFs may sequentially rewire metabolomics to shape the inflammatory responses and support the proliferation of host cells during P. gingivalis infection.


Subject(s)
Bacteroidaceae Infections/metabolism , Fibroblasts/metabolism , Fibroblasts/microbiology , Gingiva/cytology , Porphyromonas gingivalis/pathogenicity , Cells, Cultured , Humans , Inflammation , Metabolome , Real-Time Polymerase Chain Reaction
18.
FASEB J ; 35(2): e21171, 2021 02.
Article in English | MEDLINE | ID: mdl-33197074

ABSTRACT

Skeletal muscles have a high metabolic capacity, which play key roles in glucose metabolism. Although periodontal disease increases the risk of metabolic syndrome, the relationship between periodontal bacterial infection and skeletal muscle metabolic dysfunction is unclear. We found that anti-Porphyromonas gingivalis (Pg) antibody titers positively correlated with intramuscular adipose tissue content (IMAC), fasting blood glucose, and HOMA-IR in metabolic syndrome patients. In C57BL/6J mice fed a high-fat diet, recipients of oral Pg (HFPg) had impaired glucose tolerance, insulin resistance, and higher IMAC compared to recipients of saline (HFco). The soleus muscle in HFPg mice exhibited fat infiltration and lower glucose uptake with higher Tnfa expression and lower insulin signaling than in HFco mice. Gene set enrichment analysis showed that TNFα signaling via NFκB gene set was enriched in the soleus muscle of HFPg mice. Moreover, TNF-α also decreased glucose uptake in C2C12 myoblast cells in vitro. Based on 16S rRNA sequencing, Pg administration altered the gut microbiome, particularly by decreasing the abundance of genus Turicibacter. Microbial network of the gut microbiome was dramatically changed by Pg administration. Our findings suggest that infection with Pg is a risk factor for metabolic syndrome and skeletal muscle metabolic dysfunction via gut microbiome alteration.


Subject(s)
Bacteroidaceae Infections/metabolism , Blood Glucose/metabolism , Gastrointestinal Microbiome/genetics , Metabolic Syndrome/blood , Muscle, Skeletal/metabolism , Periodontal Diseases/blood , Porphyromonas gingivalis/metabolism , Adipose Tissue/metabolism , Adult , Aged , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacteroidaceae Infections/microbiology , Cell Line, Transformed , Diet, High-Fat , Feces/microbiology , Female , Glucose Intolerance/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Insulin Resistance , Japan/epidemiology , Male , Metabolic Syndrome/complications , Metabolic Syndrome/epidemiology , Metabolic Syndrome/microbiology , Mice , Mice, Inbred C57BL , Middle Aged , Myoblasts/metabolism , Periodontal Diseases/complications , Periodontal Diseases/epidemiology , Periodontal Diseases/microbiology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/immunology , RNA, Ribosomal, 16S/genetics
19.
Int J Immunopathol Pharmacol ; 34: 2058738420974893, 2020.
Article in English | MEDLINE | ID: mdl-33259259

ABSTRACT

Periodontitis is an inflammatory disease caused by host immune response, resulting in a loss of periodontium and alveolar bone. Immune cells, such as T cells and macrophages, play a critical role in the periodontitis onset. Halofuginone, a natural quinazolinone alkaloid, has been shown to possess anti-fibrosis, anti-cancer, and immunomodulatory properties. However, the effect of halofuginone on periodontitis has never been reported. In this study, a ligature-induced mice model of periodontitis was applied to investigate the potential beneficial effect of halofuginone on periodontitis. We demonstrated that the administration of halofuginone significantly reduced the expression levels of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) in vivo, and markedly suppressed immune cell infiltration into the infected sites. Furthermore, we also observed that halofuginone treatment blocked the T-helper 17 (Th17) cell differentiation in vivo and in vitro. We demonstrated for the first time that halofuginone alleviated the onset of periodontitis through reducing immune responses.


Subject(s)
Bacteroidaceae Infections/drug therapy , Chronic Periodontitis/drug therapy , Gingiva/drug effects , Immunologic Factors/pharmacology , Piperidines/pharmacology , Quinazolinones/pharmacology , Animals , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Cell Differentiation/drug effects , Cells, Cultured , Chronic Periodontitis/immunology , Chronic Periodontitis/metabolism , Chronic Periodontitis/microbiology , Cytokines/metabolism , Disease Models, Animal , Gingiva/immunology , Gingiva/metabolism , Gingiva/microbiology , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Porphyromonas gingivalis/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
20.
Front Immunol ; 11: 588036, 2020.
Article in English | MEDLINE | ID: mdl-33240277

ABSTRACT

Periodontal disease is a disease of tooth-supporting tissues. It is a chronic disease with inflammatory nature and infectious etiology produced by a dysbiotic subgingival microbiota that colonizes the gingivodental sulcus. Among several periodontal bacteria, Porphyromonas gingivalis (P. gingivalis) highlights as a keystone pathogen. Previous reports have implied that chronic inflammatory response and measurable bone resorption are observed in young mice, even after a short period of periodontal infection with P. gingivalis, which has been considered as a suitable model of experimental periodontitis. Also, encapsulated P. gingivalis strains are more virulent than capsular-defective mutants, causing an increased immune response, augmented osteoclastic activity, and accrued alveolar bone resorption in these rodent experimental models of periodontitis. Recently, P. gingivalis has been associated with Alzheimer's disease (AD) pathogenesis, either by worsening brain pathology in AD-transgenic mice or by inducing memory impairment and age-dependent neuroinflammation middle-aged wild type animals. We hypothesized here that the more virulent encapsulated P. gingivalis strains could trigger the appearance of brain AD-markers, neuroinflammation, and cognitive decline even in young rats subjected to a short periodontal infection exposure, due to their higher capacity of activating brain inflammatory responses. To test this hypothesis, we periodontally inoculated 4-week-old male Sprague-Dawley rats with K1, K2, or K4 P. gingivalis serotypes and the K1-isogenic non-encapsulated mutant (GPA), used as a control. 45-days after periodontal inoculations with P. gingivalis serotypes, rat´s spatial memory was evaluated for six consecutive days in the Oasis maze task. Following functional testing, the animals were sacrificed, and various tissues were removed to analyze alveolar bone resorption, cytokine production, and detect AD-specific biomarkers. Strikingly, only K1 or K2 P. gingivalis-infected rats displayed memory deficits, increased alveolar bone resorption, pro-inflammatory cytokine production, changes in astrocytic morphology, increased Aß1-42 levels, and Tau hyperphosphorylation in the hippocampus. None of these effects were observed in rats infected with the non-encapsulated bacterial strains. Based on these results, we propose that the bacterial virulence factors constituted by capsular polysaccharides play a central role in activating innate immunity and inflammation in the AD-like pathology triggered by P. gingivalis in young rats subjected to an acute experimental infection episode.


Subject(s)
Alzheimer Disease , Bacteroidaceae Infections , Periodontitis , Porphyromonas gingivalis , Animals , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Bone Resorption , Cytokines/immunology , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/microbiology , Learning , Lipid Peroxidation , Male , Periodontitis/immunology , Periodontitis/metabolism , Periodontitis/microbiology , Rats, Sprague-Dawley , Serogroup , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...