Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(6): 107384, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762177

ABSTRACT

Antimicrobial resistance poses a serious threat to human health worldwide and its incidence continues to increase owing to the overuse of antibiotics and other factors. Macrolide antibiotics such as erythromycin (EM) have immunomodulatory effects in addition to their antibacterial activity. Long-term, low-dose administration of macrolides has shown clinical benefits in treating non-infectious inflammatory respiratory diseases. However, this practice may also increase the emergence of drug-resistant bacteria. In this study, we synthesized a series of EM derivatives, and screened them for two criteria: (i) lack of antibacterial activity and (ii) ability to suppress tumor necrosis factor-α (TNF-α) production in THP-1 cells stimulated with lipopolysaccharide. Among the 37 synthesized derivatives, we identified a novel 12-membered ring macrolide EM982 that lacked antibacterial activity against Staphylococcus aureus and suppressed the production of TNF-α and other cytokines. The effects of EM982 on Toll-like receptor 4 (TLR4) signaling were analyzed using a reporter assay and Western blotting. The reporter assay showed that EM982 suppressed the activation of transcription factors, NF-κB and/or activator protein 1 (AP-1), in HEK293 cells expressing human TLR4. Western blotting showed that EM982 inhibited the phosphorylation of both IκB kinase (IKK) ß and IκBα, which function upstream of NF-κB, whereas it did not affect the phosphorylation of p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and c-Jun N-terminal kinase, which act upstream of AP-1. These results suggest that EM982 suppresses cytokine production by inhibiting phosphorylation of IKKß and IκBα, resulting in the inactivation of NF-κB.


Subject(s)
Cytokines , I-kappa B Kinase , NF-KappaB Inhibitor alpha , Humans , I-kappa B Kinase/metabolism , Phosphorylation/drug effects , NF-KappaB Inhibitor alpha/metabolism , Cytokines/metabolism , Erythromycin/pharmacology , Erythromycin/chemistry , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Macrolides/pharmacology , Macrolides/chemistry , NF-kappa B/metabolism , Signal Transduction/drug effects , Staphylococcus aureus/drug effects , Toll-Like Receptor 4/metabolism
2.
J Biol Chem ; 299(6): 104760, 2023 06.
Article in English | MEDLINE | ID: mdl-37119853

ABSTRACT

Pneumococcus is the main cause of bacterial pneumonia. Pneumococcal infection has been shown to cause elastase, an intracellular host defense factor, to leak from neutrophils. However, when neutrophil elastase (NE) leaks extracellularly, it can degrade host cell surface proteins such as epidermal growth factor receptor (EGFR) and potentially disrupt the alveolar epithelial barrier. In this study, we hypothesized that NE degrades the extracellular domain (ECD) of EGFR in alveolar epithelial cells and inhibits alveolar epithelial repair. Using SDS-PAGE, we showed that NE degraded the recombinant EGFR ECD and its ligand epidermal growth factor, and that the degradation of these proteins was counteracted by NE inhibitors. Furthermore, we confirmed the degradation by NE of EGFR expressed in alveolar epithelial cells in vitro. We showed that intracellular uptake of epidermal growth factor and EGFR signaling was downregulated in alveolar epithelial cells exposed to NE and found that cell proliferation was inhibited in these cells These negative effects of NE on cell proliferation were abolished by NE inhibitors. Finally, we confirmed the degradation of EGFR by NE in vivo. Fragments of EGFR ECD were detected in bronchoalveolar lavage fluid from pneumococcal pneumonia mice, and the percentage of cells positive for a cell proliferation marker Ki67 in lung tissue was reduced. In contrast, administration of an NE inhibitor decreased EGFR fragments in bronchoalveolar lavage fluid and increased the percentage of Ki67-positive cells. These findings suggest that degradation of EGFR by NE could inhibit the repair of alveolar epithelium and cause severe pneumonia.


Subject(s)
ErbB Receptors , Leukocyte Elastase , Pneumonia, Pneumococcal , Animals , Mice , Bronchoalveolar Lavage Fluid , Epithelial Cells/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Ki-67 Antigen/metabolism , Leukocyte Elastase/metabolism , Lung/metabolism , Pneumonia, Pneumococcal/metabolism , Proteinase Inhibitory Proteins, Secretory/metabolism
3.
J Immunol ; 204(5): 1214-1224, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31980574

ABSTRACT

Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits ß2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPß binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPß binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.


Subject(s)
Calcium-Binding Proteins/immunology , Cell Adhesion Molecules/immunology , Dehydroepiandrosterone/pharmacology , Leukocytes/immunology , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/immunology , CD18 Antigens/immunology , Cell Adhesion/immunology , Endothelium, Vascular/immunology , Female , Gene Expression Regulation/immunology , Leukocytes/cytology , Mice , Phosphatidylinositol 3-Kinases/immunology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, trkA/immunology
4.
Int J Mol Sci ; 23(10)2022 May 16.
Article in English | MEDLINE | ID: mdl-35628348

ABSTRACT

Periodontitis is one of the most common oral diseases resulting in gingival inflammation and tooth loss. Growing evidence indicates that it results from dysbiosis of the oral microbiome, which interferes with the host immune system, leading to bone destruction. Immune cells activate periodontal ligament cells to express the receptor activator of nuclear factor kappa-B (NF-κB) ligand (RANKL) and promote osteoclast activity. Osteocytes have active roles in periodontitis progression in the bone matrix. Local proteins are involved in bone regeneration through functional immunological plasticity. Here, we discuss the current knowledge of cellular and molecular mechanisms in periodontitis, the roles of local proteins, and promising synthetic compounds generating a periodontal regeneration effect. It is anticipated that this may lead to a better perception of periodontitis pathophysiology.


Subject(s)
Periodontitis , Humans , Immune System/metabolism , NF-kappa B , Osteoclasts/metabolism , Osteocytes/metabolism , Periodontitis/drug therapy , Periodontitis/metabolism
5.
J Biol Chem ; 295(21): 7261-7273, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32280065

ABSTRACT

The integrin-binding secreted protein developmental endothelial locus-1 (DEL-1) is involved in the regulation of both the initiation and resolution of inflammation in different diseases, including periodontitis, an oral disorder characterized by inflammatory bone loss. Here, using a mouse model of bone regeneration and in vitro cell-based mechanistic studies, we investigated whether and how DEL-1 can promote alveolar bone regeneration during resolution of experimental periodontitis. Compared with WT mice, mice lacking DEL-1 or expressing a DEL-1 variant with an Asp-to-Glu substitution in the RGD motif ("RGE point mutant"), which does not interact with RGD-dependent integrins, exhibited defective bone regeneration. Local administration of DEL-1 or of its N-terminal segment containing the integrin-binding RGD motif, but not of the RGE point mutant, reversed the defective bone regeneration in the DEL-1-deficient mice. Moreover, DEL-1 (but not the RGE point mutant) promoted osteogenic differentiation of MC3T3-E1 osteoprogenitor cells or of primary calvarial osteoblastic cells in a ß3 integrin-dependent manner. The ability of DEL-1 to promote in vitro osteogenesis, indicated by induction of osteogenic genes such as the master transcription factor Runt-related transcription factor-2 (Runx2) and by mineralized nodule formation, depended on its capacity to induce the phosphorylation of focal adhesion kinase (FAK) and of extracellular signal-regulated kinase 1/2 (ERK1/2). We conclude that DEL-1 can activate a ß3 integrin-FAK-ERK1/2-RUNX2 pathway in osteoprogenitors and promote new bone formation in mice. These findings suggest that DEL-1 may be therapeutically exploited to restore bone lost due to periodontitis and perhaps other osteolytic conditions.


Subject(s)
Bone Regeneration , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation , MAP Kinase Signaling System , Osteoblasts/metabolism , Osteogenesis , Animals , Calcium-Binding Proteins/genetics , Cell Adhesion Molecules/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Osteoblasts/cytology
6.
Microbiol Immunol ; 64(7): 493-501, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32320095

ABSTRACT

Streptococcus mutans is the main pathogen of dental caries and adheres to the tooth surface via soluble and insoluble glucans produced by the bacterial glucosyltransferase enzyme. Thus, the S. mutans glucosyltransferase is an important virulence factor for this cariogenic bacterium. Sulfated vizantin effectively inhibits biofilm formation by S. mutans without affecting its growth. In this study, less S. mutans biofilm formation occurred on hydroxyapatite discs coated with sulfated vizantin than on noncoated discs. Sulfated vizantin showed no cytotoxicity against the human gingival cell line Ca9-22. Sulfated vizantin dose-dependently inhibited the extracellular release of cell-free glucosyltransferase from S. mutans and enhanced the accumulation of cell-associated glucosyltransferase, compared with that observed with untreated bacteria. Sulfated vizantin disrupted the localization balance between cell-associated glucosyltransferase and cell-free glucosyltransferase, resulting in inhibited biofilm maturation. These results indicate that sulfated vizantin can potentially serve as a novel agent for preventing dental caries.


Subject(s)
Biofilms/drug effects , Biofilms/growth & development , Glycolipids/pharmacology , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Trehalose/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Bacterial Proteins/metabolism , Cell Line , Dental Caries/microbiology , Dental Caries/prevention & control , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Humans , Sulfates/chemistry , Trehalose/pharmacology , Virulence Factors/metabolism
7.
Microbiol Immunol ; 63(6): 213-222, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31106894

ABSTRACT

Hinokitiol, a component of the essential oil isolated from Cupressaceae, possesses antibacterial and antifungal activities and has been used in oral care products. In this study, the antibacterial activities of hinokitiol toward various oral, nasal and nasopharyngeal pathogenic bacteria, including Streptococcus mutans, Streptococcus sobrinus, Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, Prevotella intermedia, Fusobacterium nucleatum, methicillin-resistant and -susceptible Staphylococcus aureus, antibiotic-resistant and -susceptible Streptococcus pneumoniae, and Streptococcus pyogenes were examined. Growth of all these bacterial strains was significantly inhibited by hinokitiol, minimal inhibitory concentrations of hinokitiol against S. mutans, S. sobrinus, P. gingivalis, P. intermedia, A. actinomycetemcomitans, F. nucleatum, methicillin-resistant S. aureus, methicillin-susceptible S. aureus, antibiotic-resistant S. pneumoniae isolates, antibiotic-susceptible S. pneumoniae, and S. pyogenes being 0.3, 1.0, 1.0, 30, 0.5, 50, 50, 30, 0.3-1.0, 0.5, and 0.3 µg/mL, respectively. Additionally, with the exception of P. gingivalis, hinokitiol exerted bactericidal effects against all bacterial strains 1 hr after exposure. Hinokitiol did not display any significant cytotoxicity toward the human gingival epithelial cell line Ca9-22, pharyngeal epithelial cell line Detroit 562, human umbilical vein endothelial cells, or human gingival fibroblasts, with the exception of treatment with 500 µg/mL hinokitiol, which decreased numbers of viable Ca9-22 cells and gingival fibroblasts by 13% and 12%, respectively. These results suggest that hinokitiol exhibits antibacterial activity against a broad spectrum of pathogenic bacteria and has low cytotoxicity towards human epithelial cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Monoterpenes/pharmacology , Mouth/microbiology , Tropolone/analogs & derivatives , Aggregatibacter actinomycetemcomitans/drug effects , Bacteria/classification , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Epithelial Cells/drug effects , Fusobacterium nucleatum/drug effects , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Porphyromonas gingivalis/drug effects , Prevotella intermedia/drug effects , Staphylococcus aureus/drug effects , Streptococcus mutans/drug effects , Streptococcus pneumoniae/drug effects , Streptococcus pyogenes/drug effects , Streptococcus sobrinus/drug effects , Tropolone/pharmacology
8.
Microbiol Immunol ; 63(3-4): 100-110, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30817027

ABSTRACT

Aggregatibacter actinomycetemcomitans is considered to be associated with periodontitis. Leukotoxin (LtxA), which destroys leukocytes in humans, is one of this bacterium's major virulence factors. Amounts of neutrophil elastase (NE), which is normally localized in the cytoplasm of neutrophils, are reportedly increased in the saliva of patients with periodontitis. However, the mechanism by which NE is released from human neutrophils and the role of NE in periodontitis is unclear. In the present study, it was hypothesized that LtxA induces NE release from human neutrophils, which subsequently causes the breakdown of periodontal tissues. LtxA-treatment did not induce significant cytotoxicity against human gingival epithelial cells (HGECs) or human gingival fibroblasts (HGFs). However, it did induce significant cytotoxicity against human neutrophils, leading to NE release. Furthermore, NE and the supernatant from LtxA-treated human neutrophils induced detachment and death of HGECs and HGFs, these effects being inhibited by administration of an NE inhibitor, sivelestat. The present results suggest that LtxA mediates human neutrophil lysis and induces the subsequent release of NE, which eventually results in detachment and death of HGECs and HGFs. Thus, LtxA-induced release of NE could cause breakdown of periodontal tissue and thereby exacerbate periodontitis.


Subject(s)
Aggregatibacter actinomycetemcomitans/metabolism , Epithelial Cells/pathology , Exotoxins/metabolism , Fibroblasts/pathology , Gingiva/microbiology , Leukocyte Elastase/metabolism , Neutrophils/pathology , Periodontitis/microbiology , Aggregatibacter actinomycetemcomitans/pathogenicity , Cell Death/physiology , Cell Line , Epithelial Cells/microbiology , Fibroblasts/microbiology , Gingiva/cytology , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Leukocyte Elastase/antagonists & inhibitors , Neutrophils/microbiology , Sulfonamides/pharmacology , Virulence Factors/metabolism
9.
J Infect Chemother ; 25(3): 229-232, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30279114

ABSTRACT

Increase in antimicrobial resistance (AMR) among pathogenic bacteria is a serious threat to public health. Surveillance studies to monitor shifting trends in resistance are important and guide the selection of appropriate antimicrobial agents for a particular organism. Furthermore, these studies help in dissemination of accurate information regarding AMR to the public. In this study, we investigated the antimicrobial susceptibility patterns of Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis clinical isolates from outpatient children with acute otitis media in Japan from 2014 to 2017. A total of 8693 strains (2415 of S. pneumoniae, 3657 of H. influenzae, and 2621 of M. catarrhalis) were clinically isolated, and their antimicrobial susceptibilities to benzylpenicillin (PCG), ampicillin (ABPC), amoxicillin-clavulanic (AMPC/CVA), azithromycin (AZM), ceftriaxone (CTRX), and levofloxacin (LVFX) were investigated. Based on the minimum inhibitory concentration (MIC) breakpoints, the average proportion of S. pneumoniae isolates non-susceptible to PCG and AZM was 38.2% and 82.0% respectively. The average proportion of H. influenzae isolates non-susceptible to ABPC, CVA/AMPC, and CTRX was 61.9%, 43.5%, and 49.4%, respectively. The high prevalence of these resistant organisms is attributed to frequent use of antibiotic agents in Japan. Moreover, the proportion of LVFX-non-susceptible H. influenzae isolates increased in this four-year study. Here, we report updates regarding the AMR trends amongst the major pathogens that cause acute otitis media in Japan. Continuing surveillance of antimicrobial susceptibility and application of control measures against further transmission are required to decrease the emergence of resistant strains.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/microbiology , Haemophilus influenzae/drug effects , Moraxella catarrhalis/drug effects , Otitis Media/microbiology , Streptococcus pneumoniae/drug effects , Adolescent , Bacterial Infections/epidemiology , Child , Child, Preschool , Cohort Studies , Drug Resistance, Bacterial , Humans , Infant , Infant, Newborn , Microbial Sensitivity Tests , Otitis Media/epidemiology
10.
J Proteome Res ; 17(9): 3153-3175, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30111112

ABSTRACT

Periodontitis is a prevalent chronic inflammatory disease associated with dysbiosis. Although complement inhibition has been successfully used to treat periodontitis in animal models, studies globally analyzing inflamed tissue proteins to glean insight into possible mechanisms of action are missing. Using quantitative shotgun proteomics, we aimed to investigate differences in composition of inflammatory gingival tissue exudate ("gingival crevicular fluid"; GCF), before and after local administration of an inhibitor of the central complement component, C3, in nonhuman primates. The C3 inhibitor, Cp40 (also known as AMY-101) was administered locally in the maxillary gingival tissue of cynomolgus monkeys with established periodontitis, either once a week (1×-treatment; n = 5 animals) or three times per week (3×-treatment; n = 10 animals), for 6 weeks followed by another 6 weeks of observation in the absence of treatment. 45 GCF samples were processed for FASP digestion and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. Data were processed using the ProgenesisQI software. The statistical significance of differences between the groups was determined by RM-ANOVA, and a protein expression change was considered as a true regulation at >2-fold and p < 0.05. The human orthologues were subjected to Gene Ontology analyses using PANTHER. Data are available via ProteomeXchange with identifier PXD009502. 573 proteins with >2 peptides were longitudinally quantified. Both 3× and 1× administration of Cp40 resulted in significant down-regulation of dozens of proteins during the 6-week course of treatment as compared to baseline. Following drug withdrawal at 6 weeks, more than 50% of the down-regulated proteins showed increased levels at week 12. The top scored pathway was "complement activation, alternative pathway", and several proteins involved in this pathway were down-regulated at 6 weeks. We mapped the proteomic fingerprint changes in local tissue exudate of cynomolgus monkey periodontitis in response to C3 inhibition and identified the alternative pathway of complement activation and leukocyte degranulation as main targets, which are thus likely to play significant roles in periodontal disease pathogenesis. Label-free quantitative proteomics strategies utilizing GCF are powerful tools for the identification of treatment targets and providing insights into disease mechanisms.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Complement C3/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Gingival Crevicular Fluid/chemistry , Peptides, Cyclic/pharmacology , Periodontitis/drug therapy , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Chromatography, Liquid , Complement C3/genetics , Complement Pathway, Alternative/genetics , Disease Models, Animal , Drug Administration Schedule , Gene Expression Regulation , Gene Ontology , Gingiva/drug effects , Gingiva/immunology , Gingiva/pathology , Gingival Crevicular Fluid/drug effects , Gingival Crevicular Fluid/immunology , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/pathology , Macaca fascicularis , Molecular Sequence Annotation , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Proteome/classification , Proteome/genetics , Proteome/immunology , Tandem Mass Spectrometry
11.
Article in English | MEDLINE | ID: mdl-30181369

ABSTRACT

Streptococcus pneumoniae is a leading cause of community-acquired pneumonia. Over the past 2 decades, macrolide resistance among S. pneumoniae organisms has been increasing steadily and has escalated at an alarming rate worldwide. However, the use of macrolides in the treatment of community-acquired pneumonia has been reported to be effective regardless of the antibiotic susceptibility of the causative pneumococci. Although previous studies suggested that sub-MICs of macrolides inhibit the production of the pneumococcal pore-forming toxin pneumolysin by macrolide-resistant S. pneumoniae (MRSP), the underlying mechanisms of the inhibitory effect have not been fully elucidated. Here, we show that the release of pneumococcal autolysin, which promotes cell lysis and the release of pneumolysin, was inhibited by treatment with azithromycin and erythromycin, whereas replenishing with recombinant autolysin restored the release of pneumolysin from MRSP. Additionally, macrolides significantly downregulated ply transcription followed by a slight decrease of the intracellular pneumolysin level. These findings suggest the mechanisms involved in the inhibition of pneumolysin in MRSP, which may provide an additional explanation for the benefits of macrolides on the outcome of treatment for pneumococcal diseases.


Subject(s)
Drug Resistance, Bacterial/drug effects , Macrolides/pharmacology , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/metabolism , Streptolysins/metabolism , Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Bacterial Proteins/metabolism , Community-Acquired Infections/drug therapy , Community-Acquired Infections/microbiology , Humans , Microbial Sensitivity Tests/methods , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology
12.
Cell Immunol ; 325: 14-22, 2018 03.
Article in English | MEDLINE | ID: mdl-29366563

ABSTRACT

Streptococcus pneumoniae is a leading cause of bacterial pneumonia. Our previous study suggested that S. pneumoniae autolysis-dependently releases intracellular pneumolysin, which subsequently leads to lung injury. In this study, we hypothesized that pneumococcal autolysis induces the leakage of additional intracellular molecules that could increase the pathogenicity of S. pneumoniae. Liquid chromatography tandem-mass spectrometry analysis identified that chaperone protein DnaK, elongation factor Tu (EF-Tu), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were released with pneumococcal DNA by autolysis. We demonstrated that recombinant (r) DnaK, rEF-Tu, and rGAPDH induced significantly higher levels of interleukin-6 and tumor necrosis factor production in peritoneal macrophages and THP-1-derived macrophage-like cells via toll-like receptor 4. Furthermore, the DNA-binding activity of these proteins was confirmed by surface plasmon resonance assay. We demonstrated that pneumococcal DnaK, EF-Tu, and GAPDH induced the production of proinflammatory cytokines in macrophages, and might cause host tissue damage and affect the development of pneumococcal diseases.


Subject(s)
Autolysis/metabolism , DNA-Binding Proteins/metabolism , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins , Chromatography, Liquid/methods , Cytokines/metabolism , DNA-Binding Proteins/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Molecular Chaperones/metabolism , Peptide Elongation Factor Tu/metabolism , Pneumococcal Infections/genetics , Streptococcus pneumoniae/genetics , THP-1 Cells , Tandem Mass Spectrometry/methods , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
13.
Microbiol Immunol ; 62(5): 310-316, 2018 May.
Article in English | MEDLINE | ID: mdl-29577412

ABSTRACT

Vizantin is an insoluble adjuvant that activates macrophages and lymphocytes. Recently, 2,2',3,3',4,4'-hexasulfated-vizantin (sulfated vizantin), which enables solubilization of vizantin, was developed by the present team. Sulfated vizantin was found to enhance bactericidal activity against multi-drug resistant Pseudomonas aeruginosa in RAW264.7 cells. In addition, spread of P. aeruginosa was inhibited in RAW264.7 cells treated with sulfated vizantin. When only sulfated vizantin and P. aeruginosa were incubated, sulfated vizantin did not affect growth of P. aeruginosa. Formation of DNA-based extracellular traps (ETs), a novel defense mechanism in several types of innate immune cells, helps to eliminate pathogens. In the present study, ET-forming macrophages constituted the majority of immune cells. Sulfated vizantin induced ET formation in RAW264.7 cells, whereas a Ca-chelating reagent, EDTA, and T-type calcium channel blocker, tetrandrine, inhibited ET formation and attenuated inhibition of spread of P. aeruginosa in sulfated vizantin-treated cells. Thus, sulfated vizantin induces ET formation in phagocytic cells in a Ca-dependent manner, thus preventing spread of P. aeruginosa. Hence, sulfated vizantin may be useful in the management of infectious diseases.


Subject(s)
Extracellular Traps/drug effects , Glycolipids/pharmacology , Macrophages/drug effects , Macrophages/immunology , Trehalose/analogs & derivatives , Animals , Anti-Bacterial Agents/pharmacology , Benzylisoquinolines/pharmacology , Calcium/metabolism , Dimethylformamide/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Edetic Acid/pharmacology , Macrophages/physiology , Mice , Nifedipine/pharmacology , Phagocytosis/drug effects , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/immunology , RAW 264.7 Cells/drug effects , Sulfates/chemistry , Trehalose/pharmacology
14.
J Immunol ; 195(4): 1427-35, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26150532

ABSTRACT

B-lineage cells (B lymphocytes and plasma cells) predominate in the inflammatory infiltrate of human chronic periodontitis. However, their role in disease pathogenesis and the factors responsible for their persistence in chronic lesions are poorly understood. In this regard, two cytokines of the TNF ligand superfamily, a proliferation-inducing ligand (APRIL) and B-lymphocyte stimulator (BLyS), are important for the survival, proliferation, and maturation of B cells. Thus, we hypothesized that APRIL and/or BLyS are upregulated in periodontitis and contribute to induction of periodontal bone loss. This hypothesis was addressed in both human and mouse experimental systems. We show that, relative to healthy controls, the expression of APRIL and BLyS mRNA and protein was upregulated in natural and experimental periodontitis in humans and mice, respectively. The elevated expression of these cytokines correlated with increased numbers of B cells/plasma cells in both species. Moreover, APRIL and BLyS partially colocalized with κ L chain-expressing B-lineage cells at the epithelial-connective tissue interface. Ligature-induced periodontitis resulted in significantly less bone loss in B cell-deficient mice compared with wild-type controls. Ab-mediated neutralization of APRIL or BLyS diminished the number of B cells in the gingival tissue and inhibited bone loss in wild-type, but not in B cell-deficient, mice. In conclusion, B cells and specific cytokines involved in their growth and differentiation contribute to periodontal bone loss. Moreover, APRIL and BLyS have been identified as potential therapeutic targets in periodontitis.


Subject(s)
Alveolar Bone Loss/metabolism , B-Cell Activating Factor/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Periodontitis/immunology , Periodontitis/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Adult , Aged , Alveolar Bone Loss/genetics , Animals , B-Cell Activating Factor/genetics , Case-Control Studies , Disease Models, Animal , Female , Gene Expression , Humans , Immunoglobulin kappa-Chains/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Periodontitis/genetics , Periodontitis/pathology , Plasma Cells/immunology , Plasma Cells/metabolism , RNA, Messenger/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
15.
J Clin Periodontol ; 44(5): 472-483, 2017 May.
Article in English | MEDLINE | ID: mdl-28207941

ABSTRACT

AIM: We have previously shown that the secreted glycoprotein milk fat globule epidermal growth factor 8 (MFG-E8) has anti-inflammatory and anti-osteoclastogenic properties. Our objective was to investigate the potential of MFG-E8 as a diagnostic or therapeutic agent in periodontitis. MATERIALS AND METHODS: Periodontitis was induced in non-human primates (NHPs) by placing ligatures around posterior teeth on both halves of the mandible for a split-mouth design: one side was treated with MFG-E8-Fc and the other with Fc control. Disease was assessed by clinical periodontal examinations, radiographic analysis of bone loss, and analysis of cytokine mRNA expression in gingival biopsy samples. Gingival crevicular fluid (GCF) was collected from human healthy volunteers or subjects with gingivitis, chronic moderate periodontitis, or chronic severe periodontitis. Additionally, GCF was collected from a subset of severe periodontitis patients following scaling and root planing (SRP) and after pocket reduction surgery. GCF was analysed to quantify MFG-E8 and periodontitis-relevant cytokines using multiplex assays. RESULTS: In NHPs, sites treated with MFG-E8-Fc exhibited significantly less ligature-induced periodontal inflammation and bone loss than Fc control-treated sites. In humans, the GCF levels of MFG-E8 were significantly higher in health than in periodontitis, whereas the reverse was true for the proinflammatory cytokines tested. Consistently, MFG-E8 was elevated in GCF after both non-surgical (SRP) and surgical periodontal treatment of periodontitis patients. CONCLUSION: MFG-E8 is, in principle, a novel therapeutic agent and biomarker of periodontitis.


Subject(s)
Antigens, Surface/therapeutic use , Chronic Periodontitis/diagnosis , Chronic Periodontitis/therapy , Gingival Crevicular Fluid/metabolism , Milk Proteins/therapeutic use , Animals , Antigens, Surface/metabolism , Biomarkers/metabolism , Chronic Periodontitis/metabolism , Disease Models, Animal , Female , Gingivitis/diagnosis , Gingivitis/metabolism , Gingivitis/therapy , Humans , Macaca fascicularis , Milk Proteins/metabolism
16.
Semin Immunol ; 25(1): 65-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23684627

ABSTRACT

Complement plays a key role in immunity and inflammation through direct effects on immune cells or via crosstalk and regulation of other host signaling pathways. Deregulation of these finely balanced complement activities can link infection to inflammatory tissue damage. Periodontitis is a polymicrobial community-induced chronic inflammatory disease that can destroy the tooth-supporting tissues. In this review, we summarize and discuss evidence that complement is involved in the dysbiotic transformation of the periodontal microbiota and in the inflammatory process that leads to the destruction of periodontal bone. Recent insights into the mechanisms of complement involvement in periodontitis have additionally provided likely targets for therapeutic intervention against this oral disease.


Subject(s)
Complement System Proteins/immunology , Dysbiosis/immunology , Host-Pathogen Interactions , Periodontitis/immunology , Periodontium/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Dysbiosis/drug therapy , Homeostasis/drug effects , Humans , Microbiota/drug effects , Microbiota/immunology , Periodontitis/prevention & control , Periodontium/drug effects , Periodontium/microbiology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Signal Transduction/immunology
17.
J Immunol ; 192(12): 6020-7, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24808362

ABSTRACT

Chronic periodontitis is induced by a dysbiotic microbiota and leads to inflammatory destruction of tooth-supporting connective tissue and bone. The third component of complement, C3, is a point of convergence of distinct complement activation mechanisms, but its involvement in periodontitis was not previously addressed. We investigated this question using two animal species models, namely, C3-deficient or wild-type mice and nonhuman primates (NHPs) locally treated with a potent C3 inhibitor (the compstatin analog Cp40) or an inactive peptide control. In mice, C3 was required for maximal periodontal inflammation and bone loss, and for the sustenance of the dysbiotic microbiota. The effect of C3 on the microbiota was therefore different from that reported for the C5a receptor, which is required for the initial induction of dysbiosis. C3-dependent bone loss was demonstrated in distinct models, including Porphyromonas gingivalis-induced periodontitis, ligature-induced periodontitis, and aging-associated periodontitis. Importantly, local treatment of NHPs with Cp40 inhibited ligature-induced periodontal inflammation and bone loss, which correlated with lower gingival crevicular fluid levels of proinflammatory mediators (e.g., IL-17 and RANKL) and decreased osteoclastogenesis in bone biopsy specimens, as compared with control treatment. To our knowledge, this is the first time, for any disease, that complement inhibition in NHPs was shown to inhibit inflammatory processes that lead to osteoclastogenesis and bone loss. These data strongly support the feasibility of C3-targeted intervention for the treatment of human periodontitis.


Subject(s)
Bacteroidaceae Infections , Bone Resorption , Complement C3 , Periodontitis , Porphyromonas gingivalis/immunology , Pyridones/pharmacology , Animals , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Bone Resorption/drug therapy , Bone Resorption/genetics , Bone Resorption/immunology , Bone Resorption/pathology , Complement C3/antagonists & inhibitors , Complement C3/genetics , Complement C3/immunology , Disease Models, Animal , Female , Humans , Inflammation Mediators/immunology , Macaca fascicularis , Male , Mice , Osteoclasts/immunology , Osteoclasts/pathology , Peptides, Cyclic/pharmacology , Periodontitis/drug therapy , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology
18.
J Clin Periodontol ; 43(3): 238-49, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26728318

ABSTRACT

AIM: Human periodontitis is associated with overactivation of complement, which is triggered by different mechanisms converging on C3, the central hub of the system. We assessed whether the C3 inhibitor Cp40 inhibits naturally occurring periodontitis in non-human primates (NHPs). MATERIALS AND METHODS: Non-human primates with chronic periodontitis were intra-gingivally injected with Cp40 either once (5 animals) or three times (10 animals) weekly for 6 weeks followed by a 6-week follow-up period. Clinical periodontal examinations and collection of gingival crevicular fluid and biopsies of gingiva and bone were performed at baseline and during the study. A one-way repeated-measures anova was used for data analysis. RESULTS: Whether administered once or three times weekly, Cp40 caused a significant reduction in clinical indices that measure periodontal inflammation (gingival index and bleeding on probing), tissue destruction (probing pocket depth and clinical attachment level) or tooth mobility. These clinical changes were associated with significantly reduced levels of pro-inflammatory mediators and decreased numbers of osteoclasts in bone biopsies. The protective effects of Cp40 persisted, albeit at reduced efficacy, for at least 6 weeks following drug discontinuation. CONCLUSION: Cp40 inhibits pre-existing chronic periodontal inflammation and osteoclastogenesis in NHPs, suggesting a novel adjunctive anti-inflammatory therapy for treating human periodontitis.


Subject(s)
Periodontitis/drug therapy , Animals , Complement C3 , Gingival Crevicular Fluid , Macaca fascicularis , Male , Peptides , Periodontal Attachment Loss/drug therapy , Periodontal Index , Periodontal Pocket/drug therapy
19.
Adv Exp Med Biol ; 865: 57-74, 2015.
Article in English | MEDLINE | ID: mdl-26306443

ABSTRACT

The complement system is a network of interacting fluid-phase and cell surface-associated molecules that trigger, amplify, and regulate immune and inflammatory signaling pathways. Dysregulation of this finely balanced network can destabilize host-microbe homeostasis and cause inflammatory tissue damage. Evidence from clinical and animal model-based studies suggests that complement is implicated in the pathogenesis of periodontitis, a polymicrobial community-induced chronic inflammatory disease that destroys the tooth-supporting tissues. This review discusses molecular mechanisms of complement involvement in the dysbiotic transformation of the periodontal microbiome and the resulting destructive inflammation, culminating in loss of periodontal bone support. These mechanistic studies have additionally identified potential therapeutic targets. In this regard, interventional studies in preclinical models have provided proof-of-concept for using complement inhibitors for the treatment of human periodontitis.


Subject(s)
Bacteroidaceae Infections/drug therapy , Complement Inactivating Agents/therapeutic use , Complement System Proteins/metabolism , Dysbiosis/drug therapy , Periodontitis/drug therapy , Receptors, Complement/antagonists & inhibitors , Animals , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Complement Activation/drug effects , Disease Models, Animal , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/pathology , Host-Pathogen Interactions/drug effects , Humans , Macaca fascicularis , Mice , Peptides, Cyclic/therapeutic use , Periodontitis/immunology , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/pathogenicity , Pyridones/therapeutic use , Receptors, Complement/immunology , Receptors, Complement/metabolism
20.
STAR Protoc ; 5(3): 103162, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38935507

ABSTRACT

Periodontal ligament cells (PDLCs) and macrophages in bone marrow cells have been widely used to investigate novel therapeutic agents to treat periodontitis. Here, we present a protocol for collecting primary mouse PDLCs and bone marrow cells. We detail steps for culturing and differentiation for both cell types and review data analysis for in vitro experiments using primary PDLCs and bone marrow cells. This protocol can be used to explore the impact of novel therapeutic agents using in vitro experiments. For complete details on the use and execution of this protocol, please refer to Sirisereephap et al.1.

SELECTION OF CITATIONS
SEARCH DETAIL