ABSTRACT
Periodontitis is proposed as a risk factor for preterm delivery, fetal growth restriction, and preeclampsia with severe consequences for maternal and neonatal health, but the biological mechanisms involved are elusive. Porphyromonas gingivalis gain access to the placental bed and impair trophoblast cell function, as assessed in murine and human pregnancy, suggesting a pathogenic role in adverse pregnancy and neonatal outcomes. P. gingivalis releases outer membrane vesicles (P. gingivalis OMV) during growth that spread to distant tissues and are internalized in host cells as described in metabolic, neurological, and vascular systemic diseases. Here we tested the hypothesis that P. gingivalis OMV internalized in trophoblast cells disrupt their metabolism leading to trophoblast and placenta dysfunction and adverse pregnancy outcomes. An in vitro design with human trophoblast cells incubated with P. gingivalis OMV was used together with ex vivo and in vivo approaches in pregnant mice treated with P. gingivalis OMV. P. gingivalis OMV modulated human trophoblast cell metabolism by reducing glycolytic pathways and decreasing total reactive oxygen species with sustained mitochondrial activity. Metabolic changes induced by P. gingivalis OMV did not compromise cell viability; instead, it turned trophoblast cells into a metabolic resting state where central functions such as migration and invasion were reduced. The effects of P. gingivalis OMV on human trophoblast cells were corroborated ex vivo in mouse whole placenta and in vivo in pregnant mice: P. gingivalis OMV reduced glycolytic pathways in the placenta and led to lower placental and fetal weight gain in vivo with reduced placental expression of the glucose transporter GLUT1. The present results point to OMV as a key component of P. gingivalis involved in adverse pregnancy outcomes, and even more, unveil a metabolic cue in the deleterious effect of P. gingivalis OMV on trophoblast cells and mouse pregnancy, providing new clues to understand pathogenic mechanisms in pregnancy complications and other systemic diseases.
Subject(s)
Periodontitis , Porphyromonas gingivalis , Pregnancy , Female , Mice , Animals , Humans , Porphyromonas gingivalis/metabolism , Trophoblasts/pathology , Pregnancy Outcome , Placenta/pathology , Periodontitis/pathologyABSTRACT
AIM: Angiogenesis contributes to the development of apical periodontitis, periodontitis, and other oral pathologies; however, it remains unclear how this process is triggered. The aim was to evaluate whether lipopolysaccharide (LPS) from Porphyromonas endodontalis and Porphyromonas gingivalis induced angiogenesis-related effects in vitro via TLR2 and TLR4. METHODOLOGY: Porphyromonas endodontalis LPS (ATCC 35406 and clinical isolate) was purified with TRIzol, whereas P. gingivalis LPS was obtained commercially. The effects of the different LPS (24 h) in endothelial cell migration were analysed by Transwell assays, following quantification in an optical microscope (40×). The effects of LPS on FAK Y397 phosphorylation were assessed by Western blotting. Angiogenesis in vitro was determined in an endothelial tube formation assay (14 h) in Matrigel in the absence or presence of either LPS. IL-6 and VEGF-A levels were determined in cell supernatants, following 24 h treatment with LPS, and measured in multiplex bead immunoassay. The involvement of TLR2 and TLR4 was assessed with blocking antibodies. The statistical analysis was performed using STATA 12® (StataCorp LP). RESULTS: The results revealed that P. endodontalis LPS, but not P. gingivalis LPS, stimulated endothelial cell migration. Pre-treatment with anti-TLR2 and anti-TLR4 antibodies prevented P. endodontalis LPS-induced cell migration. P. endodontalis LPS promoted FAK phosphorylation on Y397, as observed by an increased p-FAK/FAK ratio. Both P. gingivalis and P. endodontalis LPS (ATCC 35406) induced endothelial tube formation in a TLR-2 and -4-dependent manner, as shown by using blocking antibodies, however, only TLR2 blocking decreased tube formation induced by P. endodontalis (clinical isolate). Moreover, all LPS induced IL-6 and VEGF-A synthesis in endothelial cells. TLR2 and TLR4 were required for IL-6 induction by P. endodontalis LPS (ATCC 35406), while only TLR4 was involved in IL-6 secretion by the other LPS. Finally, VEGF-A synthesis did not require TLR signalling. CONCLUSION: Porphyromonas endodontalis and P. gingivalis LPS induced angiogenesis via TLR2 and TLR4. Collectively, these data contribute to understanding the role of LPS from Porphyromonas spp. in angiogenesis and TLR involvement.
Subject(s)
Lipopolysaccharides , Toll-Like Receptor 2 , Lipopolysaccharides/pharmacology , Toll-Like Receptor 2/metabolism , Porphyromonas gingivalis/metabolism , Porphyromonas endodontalis/metabolism , Vascular Endothelial Growth Factor A , Endothelial Cells/metabolism , Antibodies, Blocking , Interleukin-6 , Toll-Like Receptor 4/metabolismABSTRACT
OBJECTIVE: To characterize the inhibitory activity of a novel bacteriocin produced by Staphylococcus epidermidis against this periodontal pathogen. DESIGN: The bacteriocin activity was evaluated by the agar diffusion method over a lawn of P. gingivalis ATCC 33277. The bacteriocin was purified by Reverse Phase-High Performance Liquid Chromatography (RP-HPLC) and Matrix Assisted Laser Desorption Ionization -Time of Flight Mass Spectrometry (MALDI-TOF-MS). In addition, the bacteriocin host specificity, production on different media cultures and susceptibility to enzymes, pH, and heat treatment were determined. RESULTS: The bacteriocin BAC 14990 was selective to P. gingivalis, suggesting a narrow activity range. The production during the growth curve indicated that S. epidermidis had a continued production of this antimicrobial, showing the highest concentration in the stationary phase. The purification of BAC 14990 showed that bacteriocin had a molecular mass of 5795 Da. BAC 14990 was partially resistant to the treatment with proteinase K and papain, however, was fully susceptible to amylase treatment indicating the presence of sugar residues in the protein, suggesting a conjugated type of bacteriocin. Also, this diffusible inhibitory substance was heat and pH treatment resistant. CONCLUSIONS: The results indicate the isolation of a new staphylococcal complex bacteriocin that is able to eliminate a Gram-negative bacterium. These results could contribute to the development of treatments directed against pathogens in mixed communities, as is the case with oral diseases.
Subject(s)
Bacteriocins , Bacteriocins/pharmacology , Bacteriocins/metabolism , Staphylococcus epidermidis/metabolism , Porphyromonas gingivalis/metabolism , Anti-Bacterial Agents/therapeutic use , Spectrometry, Mass, Matrix-Assisted Laser Desorption-IonizationABSTRACT
OBJECTIVE: To investigate the involvement of IL-6/STAT3 signaling pathway activation in macrophage polarization and bone destruction related to apical periodontitis (AP) stimulated by Porphyromonas gingivalis. METHODOLOGY: Macrophage polarization, IL-6/STAT3 expression, and the presence of P. gingivalis were detected in human AP tissues via RT-qPCR, western blotting, and immunohistochemistry staining. Murine bone marrow derived macrophages were isolated and cultured with P. gingivalis W83 in vitro, and levels of macrophage IL-6 expression, STAT3 phosphorylation, and macrophage polarization with or without the selective STAT3 phosphorylation inhibitor Stattic (5 µM) were detected via ELISA, western blotting, RT-qPCR, and flow cytometry, respectively. P. gingivalis-induced murine AP models were constructed, and bone destruction and macrophage polarization in the apical region were evaluated. Transwell co-culture systems were used to investigate the effects of macrophages infected with P. gingivalis on osteogenesis and osteoclastogenesis. RESULTS: P. gingivalis was detected in human AP tissues that highly expressed IL-6/STAT3, and the M1 subtype of macrophages was more abundant in these tissues. P. gingivalis infection induced IL-6 expression, STAT3 phosphorylation, and M1 polarization of macrophages, while 5 µM of Stattic partially abolished these activation effects. Systemic STAT3 blockade via oral administration of Stattic at a dose of 25 mg kg-1 alleviated murine periapical bone resorption and apical infiltration of M1 macrophages induced by P. gingivalis infection in vivo. Furthermore, macrophages infected with P. gingivalis promoted bone destruction via secretion of IL-6, TNF-α, and RANKL, which hinder pre-osteoblast expression of Runx2 and accelerate pre-osteoclast expression of NFAT2. CONCLUSIONS: The activation of IL-6/STAT3 signaling pathway is involved in mediating macrophages M1 polarization in the P. gingivalis induced apical inflammatory context and may also be intimately involved in the bone loss caused by P. gingivalis infection, directing the M1 macrophage infiltration during the progression of AP.
Subject(s)
Periapical Periodontitis , Porphyromonas gingivalis , Mice , Humans , Animals , Porphyromonas gingivalis/metabolism , Interleukin-6 , Macrophages , Periapical Periodontitis/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacologyABSTRACT
Apical Lesions of Endodontic Origin (ALEO) are initiated by polymicrobial endodontic canal infection. Porphyromonas gingivalis (Pg) and Porphyromonas endodontalis (Pe) lipopolysaccharides (LPS) can induce a pro-inflammatory macrophage response through their recognition by TLR2 and TLR4. However, polarization responses induced by Pg and/or Pe LPS in macrophages are not fully understood. We aimed to characterize the polarization profiles of macrophages differentiated from THP-1 cells following Pg and/or Pe LPS stimulation from reference strain and clinical isolates. A modified LPS purification protocol was implemented and the electrophoretic LPS profiles were characterized. THP-1 human monocytes differentiated to macrophages were stimulated with Pg and Pe LPS. Polarization profiles were characterized through cell surface markers and secreted cytokines levels after 24 h of stimulation. TLR2 and TLR4 cell surfaces and transcriptional levels were determined after 24 or 2 h of LPS stimulation, respectively. LPS from Pg induced a predominant M1 profile in macrophages evidenced by changes in the expression of the surface marker CD64 and pro-inflammatory cytokine profiles, TNF-α, IL-1ß, IL-6, and IL-12. Pe LPS was unable to induce a significant response. TLR2 and TLR4 expressions were neither modified by Pg or Pe LPS. Pg LPS, but not Pe LPS, induced a macrophage M1 Profile.
Subject(s)
Lipopolysaccharides , Porphyromonas gingivalis , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Porphyromonas endodontalis/metabolism , Porphyromonas gingivalis/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolismABSTRACT
OBJECTIVE: The purpose of this study is to investigate the pathogenic role of PPARα in periodontal antigen treated gingival cells in vitro and in experimental periodontitis in vivo . METHODOLOGY: Gingival fibroblasts, gingival epithelial cells and splenocytes were isolated from C57BL/6J wild type (WT) mice and treated with fixed P. gingivalis at for 48 hours. The mRNA levels of PPARs, TNFα, IL-1ß and IL-10 were detected by Real-time quantitative PCR. Silk ligatures after being soaked in the P.gingivalis suspension were tied around both maxillary second molars of WT mice or PPARα knock-out (KO) mice for two weeks. PPARα agonist fenofibrate and vehicle control were injected into the different side of the palatal gingiva on days 3, 6, and 9. At day 14, bone resorption and gingival mRNA expression levels of PPARs, TNFα, IL-1ß and IL-10 were measured by micro-computed tomography and RT-qPCR respectively. RESULTS: P. gingivalis treatment downregulated the expression of PPARα, but not PPARß or PPARγ, and increased the expression of TNF-α and IL-1ß in Gingival fibroblasts, gingival epithelial cells and splenocytes from WT mice. Gingival mRNA levels of PPARα were significantly decreased in experimental periodontitis in WT mice. The bone loss of PPARα KO mice in experimental periodontitis was significantly higher than WT mice and was not reduced by fenofibrate treatment. Gingival TNFα protein expressions were significantly increased by P. gingivalis associated ligation and decreased by fenofibrate treatment in WT mice but not in PPARα KO mice. CONCLUSION: This study suggests that PPARα plays an essential role in periodontitis.
Subject(s)
Alveolar Bone Loss , Fenofibrate , PPAR alpha , Periodontitis , Alveolar Bone Loss/pathology , Animals , Fenofibrate/pharmacology , Gingiva/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/metabolism , Periodontitis/metabolism , Periodontitis/pathology , Porphyromonas gingivalis/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , X-Ray MicrotomographyABSTRACT
OBJECTIVE: The aim was to determine the changes of inflammatory mediator expression in human macrophages stimulated with outer membrane vesicles purified from Porphyromonas gingivalis. DESIGN: outer membrane vesicles purified by ultracentrifugation from ATCC 33277 and W83 P. gingivalis strains were used for stimulating human macrophages and determine their inflammatory mediator expression changes. U937 monocyte cells line were differentiated into macrophages and stimulated with outer membrane vesicles for 30 min and six hours. In Independent experiments, the outer membrane vesicles and viable bacteria control were pre-treated with the gingipain inhibitors KYT-1 and KYT-36 (Arg-gingipain and Lys-gingipain, respectively) or Polymyxin-B to block the lipopolysaccharide activity to evaluate the secretion changes of immune mediators IL-1ß, IL-6, TNF-α, IL-8, MCP-1, MIP-1α and RANTES by flow cytometry. A factorial ANOVA was used to analyze the data. RESULTS: The outer membrane vesicles of P. gingivalis ATCC 33277 displayed higher Arg-gingipain activity than those obtained from the P. gingivalis W83 strain (0.6 U/µg vs. 0.46 U/µg). Although the outer membrane vesicles of P. gingivalis stimulated the production of cytokines and chemokines, specific Arg-gingipain and Lys-gingipain inhibition induced significant increases in IL-1ß, IL-6, IL-8, MCP-1, and RANTES levels, and this induction was significantly greater at 6 h compared to 30 min (*p < 0.05). On the contrary, TNF-α secretion decreased when gingipains were blocked. CONCLUSIONS: outer membrane vesicles may play a dual role during P. gingivalis infection based on their ability to induce changes in the immune responses of human macrophages, probably via gingipain-dependent events.
Subject(s)
Chemokine CCL5 , Porphyromonas gingivalis , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/pharmacology , Chemokine CCL5/metabolism , Cysteine Endopeptidases/metabolism , Cytokines/metabolism , Gingipain Cysteine Endopeptidases , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Macrophages/metabolism , Porphyromonas gingivalis/metabolism , Tumor Necrosis Factor-alpha/metabolism , U937 CellsABSTRACT
The role of Porphyromonas gingivalis (P. gingivalis) or its virulence factors, including lipopolysaccharide (LPS) not only has been related with periodontitis but also with endothelial dysfunction, a key mechanism involved in the genesis of atherosclerosis and hypertension that involving systemic inflammatory markers as angiotensin II (Ang II) and cytokines. This study compares the effect of repeated and unique exposures of P. gingivalis W83 LPS and live bacteria on the production and expression of inflammatory mediators and vasoconstrictor molecules with Ang II. Human coronary artery endothelial cells (HCAEC) were stimulated with purified LPS of P. gingivalis (1.0, 3.5 or 7.0 µg/mL) or serial dilutions of live bacteria (MOI 1: 100 - 1:0,1) at a single or repeated exposure for a time of 24 h. mRNA expression levels of AGTR1, AGTR2, IL-8, IL-1ß and MCP-1 were determined by RT-qPCR, and IL-6, MCP-1, IL-8, IL-1ß and GM-CSF levels were measured by flow cytometry, ELISA determined Ang II levels. Live bacteria in a single dose increased mRNA levels of AGTR1, and repeated doses increased mRNA levels of IL-8 and IL-1ß (p < 0.05). Repeated exposure of live-P. gingivalis induced significant production IL-6, MCP-1 and GM-CSF (p < 0.05). Moreover, these MCP-1, IL-6 and GM-CSF levels were greater than in cells treated with single exposure (p < 0.05), The expression of AGTR1 and production of Ang II induced by live-P. gingivalis W83 showed a vasomotor effect of whole bacteria in HCAEC more than LPS. In conclusion, the findings of this study suggest that repeated exposure of P. gingivalis in HCAEC induces the activation of proinflammatory and vasoconstrictor molecules that lead to endothelial dysfunction being a key mechanism of the onset and progression of arterial hypertension and atherosclerosis.
Subject(s)
Angiotensin II/metabolism , Atherosclerosis/metabolism , Hypertension/metabolism , Periodontitis/etiology , Porphyromonas gingivalis/metabolism , Angiotensin II/genetics , Atherosclerosis/microbiology , Atherosclerosis/pathology , Chemokine CCL2/genetics , Coronary Vessels/metabolism , Coronary Vessels/microbiology , Coronary Vessels/pathology , Endothelial Cells/drug effects , Endothelial Cells/microbiology , Flow Cytometry , Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Hypertension/microbiology , Hypertension/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interleukin-6/genetics , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Periodontitis/genetics , Periodontitis/metabolism , Periodontitis/microbiology , Porphyromonas gingivalis/pathogenicity , RNA, Messenger/genetics , Receptor, Angiotensin, Type 1/geneticsABSTRACT
Porphyromonas gingivalis has been extensively associated with both the onset and progression of periodontitis. We previously isolated and characterized two P. gingivalis strains, one from a patient exhibiting severe chronic periodontitis (CP3) and another from a periodontally healthy individual (H3). We previously showed that CP3 and H3 exhibit differences in virulence since H3 showed a lower resistance to cationic peptides compared with CP3, and a lower ability to induce proliferation in gingival epithelial cells. Here, we aimed to determine whether differences in virulence between these two strains are associated with the presence or absence of specific genes encoding virulence factors. We sequenced the whole genomes of both P. gingivalis CP3 and H3 and conducted a comparative analysis regarding P. gingivalis virulence genetic determinants. To do so, we performed a homology search of predicted protein sequences in CP3 and H3 genomes against the most characterized virulence genes for P. gingivalis available in the literature. In addition, we performed a genomic comparison of CP3 and H3 with all the 62 genomes of P. gingivalis found in NCBI's RefSeq database. This approach allowed us to determine the evolutionary relationships of CP3 and H3 with other virulent and avirulent strains; and additionally, to detect variability in presence/absence of virulence genes among P. gingivalis genomes. Our results show genetic variability in the hemagglutinin genes. While CP3 possesses one copy of hagA and two of hagC, H3 has no hagA and only one copy of hagC. Experimentally, this finding is related to lower in vitro hemmaglutination ability of H3 compared to CP3. Moreover, while CP3 encodes a gene for a major fimbrium subunit FimA type 4 (CP3_00160), H3 possess a FimA type 1 (H3_01400). Such genetic differences are in agreement with both lower biofilm formation ability and less intracellular invasion to oral epithelial cells exhibited by H3, compared with the virulent strain CP3. Therefore, here we provide new results on the genome sequences, comparative genomics analyses, and phenotypic analyses of two P. gingivalis strains. The genomics comparison of these two strains with the other 62 genomes included in the analysis provided relevant results regarding genetic determinants and their association with P. gingivalis virulence.
Subject(s)
Chronic Periodontitis/pathology , Gene Expression Regulation, Bacterial , Genome, Bacterial , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/pathogenicity , Virulence Factors/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Case-Control Studies , Cell Line , Chronic Periodontitis/microbiology , Epithelial Cells/microbiology , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Gene Ontology , Genetic Variation , Genomics , Gingiva/microbiology , Humans , Lectins/genetics , Lectins/metabolism , Molecular Sequence Annotation , Phenotype , Phylogeny , Porphyromonas gingivalis/classification , Porphyromonas gingivalis/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Analysis, DNA , Virulence , Virulence Factors/metabolismABSTRACT
This study aimed at evaluating the transcriptional profile of apoptosis-related genes after in vitro stimulation of peripheral blood mononuclear cells (PBMCs) derived from individuals with periodontitis (P) and healthy nonperiodontitis (NP) control subjects with P. gingivalis HmuY protein. PBMCs from the P and NP groups were stimulated with HmuY P. gingivalis protein, and the expression of genes related to apoptosis was assessed by custom real-time polymerase chain reaction array (Custom RT2 PCR Array). Compared with the NP group, the P group showed low relative levels of apoptosis-related gene expression, downregulated for FAS, FAS ligand, TNFSF10 (TRAIL), BAK1, CASP9, and APAF1 after P. gingivalis HmuY protein stimulation. Furthermore, the P group exhibited low levels of relative gene expression, downregulated for CASP7 when the cells were not stimulated. Our data suggest that P. gingivalis HmuY protein might participate differently in the modulation of the intrinsic and extrinsic apoptosis pathways.
Subject(s)
Apoptosis/physiology , Bacterial Proteins/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Apoptosis/genetics , Bacterial Proteins/genetics , Humans , Real-Time Polymerase Chain ReactionABSTRACT
BACKGROUND AND OBJECTIVE: Although previous studies revealed the potential use of probiotics in the control of periodontitis, little is known about their interactions with gingival epithelial cells (GECs). Since GECs comprise the first defense in the subgingival microenvironment, the aim of this study was to evaluate the effect of probiotic lactobacilli and bifidobacteria strains on OBA-9 cells challenged with Porphyromonas gingivalis. METHODS: Immortalized human GECs (OBA-9) were challenged with live P. gingivalis (strains W83 and ATCC33277) and co-infected with one of 12 tested probiotic strains at a multiplicity of infection (MOI) of 1:1000 for 2 hours. Bacterial adhesion and invasion were determined by antibiotic exclusion analysis and CFU counting. OBA-9 viability was assessed by MTT assay, and levels of inflammatory mediators (TNF-α, IL-1ß, and CXCL8) in the supernatants were determined by ELISA. The expression of genes encoding Toll-like receptors (TLR2, TLR4) was evaluated by RT-qPCR. RESULTS: Both strains of P. gingivalis were able to adhere and invade OBA-9 cells, with significant loss in cell viability, increase in the levels of TNF-α and IL-1ß, and upregulation of TLR4. However, co-infection with probiotics attenuated these effects in P. gingivalis challenged GECs. Most probiotics maintained OBA-9 viability and reduced pathogens adhesion and invasion. Furthermore, probiotics were able to adhere to GECs, which was enhanced for most strains in the presence of P. gingivalis. The synthesis of IL-1ß and TNF-α by P. gingivalis in challenged GECs was reduced in co-culture with most of the tested probiotics, whereas the secretion of CXCL8 increased, and TLR4 was downregulated. CONCLUSION: Probiotics can alter the interaction of GECs with P. gingivalis by modulating the pathogen's ability to adhere and invade these cells, as well as by regulating the innate immune response. Such properties are strain-specific and may indicate the most efficient probiotics to control periodontitis.
Subject(s)
Antibiosis/immunology , Bifidobacterium/physiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gingiva/cytology , Gingiva/immunology , Immunity, Innate , Lactobacillus/physiology , Periodontitis/prevention & control , Periodontitis/therapy , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/pathogenicity , Probiotics , Cells, Cultured , Cellular Microenvironment/immunology , Humans , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Periodontitis/immunology , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolismABSTRACT
BACKGROUND: Porphyromonas gingivalis (Pg) is a major periodontal pathogen that contains immunostimulatory components. Periodontal ligament mesenchymal stem cells (PDLMSCs) are responsible for regeneration of the periodontium that is lost due to periodontitis. Pathologic factors within the microenvironment that impair resident PDLMSCs are not well understood. The present study investigates in vitro the effects of Pg protein extract (PgPE) on biologic properties of CD105-enriched PDL progenitor cell populations (PDL-CD105+). METHODS: Five populations of PDL-CD105+ cells were exposed to PgPE and assessed for cell viability, apoptosis, and proinflammatory gene expression (interleukin-1ß [IL-1ß], tumor necrosis factor-alpha [TNF-α], and IL-6) by quantitative reverse transcription polymerase chain reaction, IL-6 immunostaining, activation of IL-6/signal transducer and activator of transcription (STAT) 3 signaling pathway, and osteogenic differentiation potential. RESULTS: PgPE treatment (2 µg/mL) did not affect cell viability or survival but induced a significant increase in IL-1ß, TNF-α, and IL-6 messenger RNA (mRNA) expression and positive staining for IL-6. A total of 29 genes from the IL-6/STAT3 pathway were upregulated on PgPE stimulation. These genes are related to biologic processes involved in the control of cell survival (B-cell lymphoma 2 [BCL2]), cell proliferation (hepatocytehepatocyte growth factor), cytokine-mediated signaling pathway (suppressor of cytokine signaling 3, C-X-C ligand 8 [CXCL8]), and response to stress (CXCL8, mitogen-activated protein kinase 3, BCL2-associated X protein, and BCL2). Additionally, PgPE treatment caused an increase in alkaline phosphatase mRNA expression in PDL-CD105+ cells after 7 days of osteogenic induction, although mineral nodule formation was comparable to the control group. CONCLUSIONS: These results suggest that the inflammatory profile induced by PgPE treatment in PDL-CD105+ cells did not affect cell viability, apoptosis, or osteogenic differentiation, perhaps due to increased expression of genes involved in the control of cell proliferation and protection against cell death.
Subject(s)
Bacterial Proteins/pharmacology , Cell Differentiation , Mesenchymal Stem Cells/physiology , Osteogenesis , Periodontal Ligament/growth & development , Porphyromonas gingivalis/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cellular Microenvironment , Female , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/microbiology , Osteogenesis/drug effects , Periodontal Ligament/microbiology , Young AdultABSTRACT
ABSTRACT Objective Halitosis can be caused by microorganisms that produce volatile sulphur compounds (VSCs), which colonize the surface of the tongue and subgingival sites. Studies have reported that the use of natural products can reduce the bacterial load and, consequently, the development of halitosis. The aim of this study was to evaluate the antimicrobial activity of the essential oil of Melaleuca alternifolia on the growth and volatile sulphur compound (VSC) production of oral bacteria compared with chlorhexidine. Material and Methods The effects of these substances were evaluated by the Minimum Inhibitory Concentration (MIC) and Minimum Bactericidal Concentration (MBC) in planktonic cultures of Porphyromonas gingivalis and Porphyromonas endodontalis. In addition, gas chromatography analyses were performed to measure the concentration of VSCs from bacterial cultures and to characterize M. alternifolia oil components. Results The MIC and MBC values were as follows: M. alternifolia - P. gingivalis (MIC and MBC=0.007%), P. endodontalis (MIC and MBC=0.007%=0.5%); chlorhexidine - P. gingivalis and P. endodontalis (MIC and MBC=1.5 mg/mL). M. alternifolia significantly reduced the growth and production of hydrogen sulfide (H2S) by P. gingivalis (p<0.05, ANOVA-Dunnet) and the H2S and methyl mercaptan (CH3SH) levels of P. endodontalis (p<0.05, ANOVA-Dunnet). Chlorhexidine reduced the growth of both microorganisms without altering the production of VSC in P. endodontalis. For P. gingivalis, the production of H2S and CH3SH decreased (p<0.05, ANOVA-Dunnet). Conclusion M. alternifolia can reduce bacterial growth and VSCs production and could be used as an alternative to chlorhexidine.
Subject(s)
Sulfur Compounds/metabolism , Porphyromonas gingivalis/drug effects , Tea Tree Oil/pharmacology , Melaleuca/chemistry , Porphyromonas endodontalis/drug effects , Anti-Bacterial Agents/pharmacology , Sulfur Compounds/analysis , Time Factors , Microbial Sensitivity Tests , Cells, Cultured , Reproducibility of Results , Analysis of Variance , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/metabolism , Porphyromonas endodontalis/growth & development , Porphyromonas endodontalis/metabolism , Bacterial Load/drug effects , Halitosis/metabolism , Halitosis/microbiology , Halitosis/prevention & control , Gas Chromatography-Mass SpectrometryABSTRACT
OBJECTIVE: Halitosis can be caused by microorganisms that produce volatile sulphur compounds (VSCs), which colonize the surface of the tongue and subgingival sites. Studies have reported that the use of natural products can reduce the bacterial load and, consequently, the development of halitosis. The aim of this study was to evaluate the antimicrobial activity of the essential oil of Melaleuca alternifolia on the growth and volatile sulphur compound (VSC) production of oral bacteria compared with chlorhexidine. MATERIAL AND METHODS: The effects of these substances were evaluated by the Minimum Inhibitory Concentration (MIC) and Minimum Bactericidal Concentration (MBC) in planktonic cultures of Porphyromonas gingivalis and Porphyromonas endodontalis. In addition, gas chromatography analyses were performed to measure the concentration of VSCs from bacterial cultures and to characterize M. alternifolia oil components. RESULTS: The MIC and MBC values were as follows: M. alternifolia - P. gingivalis (MIC and MBC=0.007%), P. endodontalis (MIC and MBC=0.007%=0.5%); chlorhexidine - P. gingivalis and P. endodontalis (MIC and MBC=1.5 mg/mL). M. alternifolia significantly reduced the growth and production of hydrogen sulfide (H2S) by P. gingivalis (p<0.05, ANOVA-Dunnet) and the H2S and methyl mercaptan (CH3SH) levels of P. endodontalis (p<0.05, ANOVA-Dunnet). Chlorhexidine reduced the growth of both microorganisms without altering the production of VSC in P. endodontalis. For P. gingivalis, the production of H2S and CH3SH decreased (p<0.05, ANOVA-Dunnet). CONCLUSION: M. alternifolia can reduce bacterial growth and VSCs production and could be used as an alternative to chlorhexidine.
Subject(s)
Anti-Bacterial Agents/pharmacology , Melaleuca/chemistry , Porphyromonas endodontalis/drug effects , Porphyromonas gingivalis/drug effects , Sulfur Compounds/metabolism , Tea Tree Oil/pharmacology , Analysis of Variance , Bacterial Load/drug effects , Cells, Cultured , Gas Chromatography-Mass Spectrometry , Halitosis/metabolism , Halitosis/microbiology , Halitosis/prevention & control , Microbial Sensitivity Tests , Porphyromonas endodontalis/growth & development , Porphyromonas endodontalis/metabolism , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/metabolism , Reproducibility of Results , Sulfur Compounds/analysis , Time FactorsABSTRACT
Pitanga, Eugenia uniflora L., is a tropical fruit, which may be consumed as juice. While beneficial health effects of Eugenia uniflora L. leaf extracts have extensively been studied, limited data are available on an anti-inflammatory potential of pitanga juice. The aim of the presented study was to investigate anti-inflammatory properties of pitanga juice with regards to a prevention of inflammation-related periodontal diseases. For this purpose, six healthy volunteers swirled pitanga juice, containing 35% pitanga pulp, for 10 min. Thereafter, oral gum epithelial cells were harvested using a sterile brush and stimulated with lipopolysaccharides from Porphyromonas gingivalis (PG-LPS) for 6 h. Furthermore, human gingival fibroblasts (HGF-1) were used to elucidate the anti-inflammatory potential of pitanga juice constituents, cyanidin-3-glucoside and oxidoselina-1,3,7(11)-trien-8-one, in juice representative concentrations of 119 µg ml(-1) and 30 µg ml(-1), respectively. For the first time, an anti-inflammatory impact of pitanga juice on gingival epithelial cells was shown by means of an attenuation of IL-8 release by 55 ± 8.2% and 52 ± 11% in non-stimulated and PG-LPS-stimulated cells, respectively. In addition, both cyanidin-3-glucoside and oxidoselina-1,3,7(11)-trien-8-one reduced the LPS-stimulated CXCL8 mRNA expression by 50 ± 15% and 37 ± 18% and IL-8 release by 52 ± 9.9% and 45 ± 3.7% in HGF-1 cells, when concomitantly incubated with 10 µg ml(-1)PG-LPS for 6 h, revealing an anti-inflammatory potential of the volatile compound oxidoselina-1,3,7(11)-trien-8-one for the first time.
Subject(s)
Anti-Inflammatory Agents/pharmacology , Beverages/analysis , Epithelial Cells/drug effects , Fruit/chemistry , Gingiva/drug effects , Syzygium/chemistry , Anthocyanins/pharmacology , Anti-Inflammatory Agents/chemistry , Cell Line , Fibroblasts/drug effects , Gingiva/metabolism , Glucosides/pharmacology , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Lipopolysaccharides/adverse effects , Porphyromonas gingivalis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Volatile Organic Compounds/chemistry , Volatile Organic Compounds/pharmacologyABSTRACT
The aim of this study was to clone, express the gene of Hgp44 in adhesin domains of gingipains from Porphyromonas gingivalis and purify the protein. Furthermore, the effect of Hgp44 from P. gingivalis on inducing HUVECs to secrete IL-6 and IL-8 was evaluated. The Hgp44 gene fragment was amplified by polymerase chain reaction, and then inserted into the cloning vector pMD18-T and linked with a prokaryotic expression vector pET22b to construct the recombinant expression plasmid pET22b-Hgp44. Fusion protein expression was induced by IPTG, and it was purified by immobilized metal-chelating affinity chromatography (IMAC) using an Ni(2+) matrix column. HUVECs were cultured in vitro and different concentrations of Hgp44 were added to confluent HUVEC monolayers and incubated for 2, 8 and 24 h. We extracted the supernatants and then used ELISA kits to test the changes in IL-6 and IL-8 levels. Finally, a 1100-bp fragment was successfully amplified, and the expression of the fusion protein was examined by SDS-PAGE and Western blot analysis, and the data showed that the protein was 44 kDa in size and expressed mostly in the form of inclusion bodies. The purification of the fusion protein was achieved using Ni(2+) affinity chromatography. About 3.5 mg/L fusion protein was obtained. Hgp44 could induce HUVECs to secrete IL-6 and IL-8 levels, which were remarkably increased. In a word, Hgp44 was successfully expressed in a prokaryotic expression system and purified by IMAC using the Ni(2+) matrix column. The effect of Hgp44 in inducing HUVECs to secrete IL-6 and IL-8 was demonstrated.
Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/metabolism , Bacterial Proteins/pharmacology , Cells, Cultured , Gene Expression , Genetic Vectors/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacologyABSTRACT
BACKGROUND AND OBJECTIVE: Antimicrobial peptides, such as beta-defensins, secreted by gingival epithelial cells, are thought to play a major role in preventing periodontal diseases. In the present study, we investigated the ability of green tea polyphenols to induce human beta-defensin (hBD) secretion in gingival epithelial cells and to protect hBDs from proteolytic degradation by Porphyromonas gingivalis. MATERIAL AND METHODS: Gingival epithelial cells were treated with various amounts (25-200 µg/mL) of green tea extract or epigallocatechin-3-gallate (EGCG). The secretion of hBD1 and hBD2 was measured using ELISAs, and gene expression was quantified by real-time PCR. The treatments were also carried out in the presence of specific kinase inhibitors to identify the signaling pathways involved in hBD secretion. The ability of green tea extract and EGCG to prevent hBD degradation by proteases of P. gingivalis present in a bacterial culture supernatant was evaluated by ELISA. RESULTS: The secretion of hBD1 and hBD2 was up-regulated, in a dose-dependent manner, following the stimulation of gingival epithelial cells with a green tea extract or EGCG. Expression of the hBD gene in gingival epithelial cells treated with green tea polyphenols was also increased. EGCG-induced secretion of hBD1 and hBD2 appeared to involve extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase. Lastly, green tea extract and EGCG prevented the degradation of recombinant hBD1 and hBD2 by a culture supernatant of P. gingivalis. CONCLUSION: Green tea extract and EGCG, through their ability to induce hBD secretion by epithelial cells and to protect hBDs from proteolytic degradation by P. gingivalis, have the potential to strengthen the epithelial antimicrobial barrier. Future clinical studies will indicate whether these polyphenols represent a valuable therapeutic agent for treating/preventing periodontal diseases.
Subject(s)
Camellia sinensis , Catechin/analogs & derivatives , Plant Extracts/pharmacology , Porphyromonas gingivalis/drug effects , beta-Defensins/drug effects , Butadienes/pharmacology , Catechin/pharmacology , Cell Line , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gingiva/drug effects , Gingiva/metabolism , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nitriles/pharmacology , Porphyromonas gingivalis/metabolism , Proteolysis/drug effects , Pyridines/pharmacology , Up-Regulation , beta-Defensins/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitorsABSTRACT
Protease-activated receptor 2 (PAR2) is implicated in the pathogenesis of chronic inflammatory diseases, including periodontitis; it can be activated by gingipain and produced by Porphyromonas gingivalis and by neutrophil protease 3 (P3). PAR2 activation plays a relevant role in inflammatory processes by inducing the release of important inflammatory mediators associated with periodontal breakdown. The effects of periodontal treatment on PAR2 expression and its association with levels of proinflammatory mediators and activating proteases were investigated in chronic periodontitis patients. Positive staining for PAR2 was observed in gingival crevicular fluid cells and was reflective of tissue destruction. Overexpression of PAR2 was positively associated with inflammatory clinical parameters and with the levels of interleukin-6 (IL-6), IL-8, tumor necrosis factor alpha, matrix metalloprotease 2 (MMP-2), MMP-8, hepatocyte growth factor, and vascular endothelial growth factor. Elevated levels of gingipain and P3 and decreased levels of dentilisin and the protease inhibitors secretory leukocyte protease inhibitor and elafin were also associated with PAR2 overexpression. Healthy periodontal sites from individuals with chronic periodontitis showed diminished expression of PAR2 mRNA and the PAR2 protein (P < 0.05). Furthermore, periodontal treatment resulted in decreased PAR2 expression and correlated with decreased expression of inflammatory mediators and activating proteases. We concluded that periodontal treatment resulted in decreased levels of proteases and that proinflammatory mediators are associated with decreased PAR2 expression, suggesting that PAR2 expression is influenced by the presence of periodontal infection and is not a constitutive characteristic favoring periodontal inflammation.
Subject(s)
Gingival Crevicular Fluid/cytology , Periodontitis/metabolism , Periodontitis/therapy , Receptor, PAR-2/metabolism , Adhesins, Bacterial/metabolism , Adult , Bacterial Proteins , Chymotrypsin/metabolism , Cysteine Endopeptidases/metabolism , Elafin/metabolism , Female , Gingipain Cysteine Endopeptidases , Hepatocyte Growth Factor/metabolism , Humans , Inflammation/immunology , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 8/metabolism , Middle Aged , Myeloblastin/metabolism , Peptide Hydrolases , Periodontal Pocket/immunology , Periodontitis/immunology , Porphyromonas gingivalis/metabolism , RNA, Messenger/biosynthesis , Receptor, PAR-2/biosynthesis , Receptor, PAR-2/genetics , Secretory Leukocyte Peptidase Inhibitor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Young AdultABSTRACT
OBJECTIVE: The aim of this study was to compare the production of the chemokines CCL3 and CXCL12 by cultured dental pulp fibroblasts from permanent (PDPF) and deciduous (DDPF) teeth under stimulation by Porphyromonas gingivalis LPS (PgLPS). MATERIAL AND METHODS: Primary culture of fibroblasts from permanent (n=3) and deciduous (n=2) teeth were established using an explant technique. After the fourth passage, fibroblasts were stimulated by increasing concentrations of PgLPS (0-10 µg/mL) at 1, 6 and 24 h. The cells were tested for viability through MTT assay, and production of the chemokines CCL3 and CXCL12 was determined through ELISA. Comparisons among samples were performed using One-way ANOVA for MTT assay and Two-way ANOVA for ELISA results. RESULTS: Cell viability was not affected by the antigen after 24 h of stimulation. PgLPS induced the production of CCL3 by dental pulp fibroblasts at similar levels for both permanent and deciduous pulp fibroblasts. Production of CXCL12, however, was significantly higher for PDPF than DDPF at 1 and 6 h. PgLPS, in turn, downregulated the production of CXCL12 by PDPF but not by DDPF. CONCLUSION: These data suggest that dental pulp fibroblasts from permanent and deciduous teeth may present a differential behavior under PgLPS stimulation.