Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Oral Microbiol ; 37(6): 256-265, 2022 12.
Article in English | MEDLINE | ID: mdl-36189827

ABSTRACT

OBJECTIVES: We have previously characterized the main osteoimmunological events that occur during ligature periodontitis. This study aims to determine the polymicrobial community shifts that occur during disease development. METHODS: Periodontitis was induced in C57BL/6 mice using the ligature-induced periodontitis model. Healthy oral mucosa swabs and ligatures were collected every 3 days from 0 to 18 days post-ligature placement. Biofilm samples were evaluated by 16SrRNA gene sequencing (Illumina MiSeq) and QIIME. Time-course changes were determined by relative abundance, diversity, and rank analyses (PERMANOVA, Bonferroni-adjusted). RESULTS: Microbial differences between health and periodontal inflammation were observed at all phylogenic levels. An evident microbial community shift occurred in 25 genera during the advancement of "gingivitis" (3-6 days) to periodontitis (9-18 days). From day 0 to 18, dramatic changes were identified in Streptococcus levels, with an overall decrease (54.04%-0.02%) as well an overall increase of Enterococcus and Lactobacillus (23.7%-73.1% and 10.1%-70.2%, respectively). Alpha-diversity decreased to its lowest at 3 days, followed by an increase in diversity as disease advancement. Beta-diversity increased after ligature placement, indicating that bone loss develops in response to a greater microbial variability (p = 0.001). Levels of facultative and strict anaerobic bacteria augmented over the course of disease progression, with a total of eight species significantly different during the 18-day period. CONCLUSION: The data supports that murine gingival inflammation and alveolar bone loss develop in response to microbiome shifts. Bacterial diversity increased during progression to bone loss. These findings further support the utilization of the periodontitis ligature model for microbial shift analysis under different experimental conditions.


Subject(s)
Alveolar Bone Loss , Periodontitis , Mice , Animals , Dysbiosis , Mice, Inbred C57BL , Periodontitis/microbiology , Alveolar Bone Loss/microbiology , Inflammation , Biofilms , Disease Models, Animal
2.
Int J Mol Sci ; 23(13)2022 Jun 26.
Article in English | MEDLINE | ID: mdl-35806105

ABSTRACT

This study aimed to evaluate the effects of hesperidin (HE) on in vitro osteoclastogenesis and dietary supplementation on mouse periodontal disease and femoral bone phenotype. RAW 264.7 cells were stimulated with RANKL in the presence or absence of HE (1, 100 or 500 µM) for 5 days, and evaluated by TRAP, TUNEL and Western Blot (WB) analyses. In vivo, C57BL/6 mice were given HE via oral gavage (125, 250 and 500 mg/kg) for 4 weeks. A sterile silk ligature was placed between the first and second right maxillary molars for 10 days and microcomputed tomography (µCT), histopathological and immunohistochemical evaluation were performed. Femoral bones subjected or not to dietary HE (500 mg/kg) for 6 and 12 weeks were evaluated using µCT. In vitro, HE 500 µM reduced formation of RANKL-stimulated TRAP-positive(+) multinucleated cells (500 µM) as well as c-Fos and NFATc1 protein expression (p < 0.05), markers of osteoclasts. In vivo, dietary HE 500 mg/kg increased the alveolar bone resorption in ligated teeth (p < 0.05) and resulted in a significant increase in TRAP+ cells (p < 0.05). Gingival inflammatory infiltrate was greater in the HE 500 mg/kg group even in the absence of ligature. In femurs, HE 500 mg/kg protected trabecular and cortical bone mass at 6 weeks of treatment. In conclusion, HE impaired in vitro osteoclastogenesis, but on the contrary, oral administration of a high concentration of dietary HE increased osteoclast numbers and promoted inflammation-induced alveolar bone loss. However, HE at 500 mg/kg can promote a bone-sparing effect on skeletal bone under physiological conditions.


Subject(s)
Alveolar Bone Loss , Bone Resorption , Hesperidin , Alveolar Bone Loss/pathology , Animals , Bone Resorption/metabolism , Cell Differentiation , Hesperidin/pharmacology , Homeostasis , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism , X-Ray Microtomography
3.
J Periodontol ; 93(9): 1366-1377, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35404474

ABSTRACT

BACKGROUND: Periodontal destruction can be the result of different known and yet-to-be-discovered biological pathways. Recent human genetic association studies have implicated interferon-gamma inducible protein 16 (IFI16) and absent in melanoma 2 (AIM2) with high periodontal interleukin (IL)-1ß levels and more destructive disease, but mechanistic evidence is lacking. Here, we sought to experimentally validate these observational associations and better understand IFI16 and AIM2's roles in periodontitis. METHODS: Periodontitis was induced in Ifi204-/- (IFI16 murine homolog) and Aim2-/- mice using the ligature model. Chimeric mice were created to identify the main source cells of Ifi204 in the periodontium. IFI16-silenced human endothelial cells were treated with periodontal pathogens in vitro. Periodontal tissues from Ifi204-/- mice were evaluated for alveolar bone (micro-CT), cell inflammatory infiltration (MPO+ staining), Il1b (qRT-PCR), and osteoclast numbers (cathepsin K+ staining). RESULTS: Ifi204-deficient mice> exhibited >20% higher alveolar bone loss than wild-type (WT) (P < 0.05), while no significant difference was found in Aim2-/- mice. Ifi204's effect on bone loss was primarily mediated by a nonbone marrow source and was independent of Aim2. Ifi204-deficient mice had greater neutrophil/macrophage trafficking into gingival tissues regardless of periodontitis development compared to WT. In human endothelial cells, IFI16 decreased the chemokine response to periodontal pathogens. In murine periodontitis, Ifi204 depletion elevated gingival Il1b and increased osteoclast numbers at diseased sites (P < 0.05). CONCLUSIONS: These findings support IFI16's role as a novel regulator of inflammatory cell trafficking to the periodontium that protects against bone loss and offers potential targets for the development of new periodontal disease biomarkers and therapeutics.


Subject(s)
Alveolar Bone Loss , Nuclear Proteins , Periodontitis , Phosphoproteins , Alveolar Bone Loss/genetics , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/prevention & control , Animals , Biomarkers/metabolism , Cathepsin K , Disease Models, Animal , Endothelial Cells/metabolism , Interferon-gamma/metabolism , Interferons/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Periodontitis/genetics , Periodontitis/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism
4.
Cell ; 182(2): 447-462.e14, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32758418

ABSTRACT

The precise mechanism by which oral infection contributes to the pathogenesis of extra-oral diseases remains unclear. Here, we report that periodontal inflammation exacerbates gut inflammation in vivo. Periodontitis leads to expansion of oral pathobionts, including Klebsiella and Enterobacter species, in the oral cavity. Amassed oral pathobionts are ingested and translocate to the gut, where they activate the inflammasome in colonic mononuclear phagocytes, triggering inflammation. In parallel, periodontitis results in generation of oral pathobiont-reactive Th17 cells in the oral cavity. Oral pathobiont-reactive Th17 cells are imprinted with gut tropism and migrate to the inflamed gut. When in the gut, Th17 cells of oral origin can be activated by translocated oral pathobionts and cause development of colitis, but they are not activated by gut-resident microbes. Thus, oral inflammation, such as periodontitis, exacerbates gut inflammation by supplying the gut with both colitogenic pathobionts and pathogenic T cells.


Subject(s)
Colitis/pathology , Enterobacter/physiology , Gastrointestinal Microbiome , Klebsiella/physiology , Mouth/microbiology , Animals , Colitis/microbiology , Colon/microbiology , Colon/pathology , Disease Models, Animal , Enterobacter/isolation & purification , Female , Inflammasomes/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-1beta/metabolism , Klebsiella/isolation & purification , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodontitis/microbiology , Periodontitis/pathology , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
5.
J Immunol ; 204(8): 2177-2191, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32169848

ABSTRACT

Emerging evidence suggests comprehensive immune profiling represents a highly promising, yet insufficiently tapped approach to identify potentially prognostic signatures for periodontitis. In this report, we agnostically identified a periodontitis-associated inflammatory expression network with multiple biomarkers identified within gingival crevicular fluid samples from study participants by applying principal component analysis. We identified an IL-17-dominated trait that is associated with periodontal disease and is inversely modified by the level of IL-10. IL-10 mitigated chemokine CXCL5 and CXCL1 expressions in IL-17-stimulated peripheral blood monocytic cells and peripheral blood monocytic cell-derived macrophages. Il10-deficient mice presented more bone loss, which was associated with more Il17 and IL-17-mediated chemokine and cytokine expression at the transcriptional levels in comparison with control wild-type mice in both the Porphyromonas gingivalis-induced experimental murine periodontitis and ligature-induced alveolar bone-loss models. The dampening effect of IL-10 on the excessive signaling of IL-17 appeared to be mediated by innate immune cells populations rather than by gingival epithelial cells, which are the major cell target for IL-17 signaling. Additionally, elevated IL-17 response in Il10-deficient mice specifically elicited an M1-skewing macrophage phenotype in the gingiva that was associated with the advanced bone loss in the ligature model. In summary, IL-17 dominated an inflammatory network characteristic of periodontitis, and IL-10 dampens this excessive IL-17-mediated periodontitis trait.


Subject(s)
Inflammation/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Periodontitis/immunology , Animals , Cells, Cultured , Gingival Crevicular Fluid/immunology , Humans , Interleukin-10/deficiency , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Principal Component Analysis
6.
Periodontol 2000 ; 82(1): 93-114, 2020 02.
Article in English | MEDLINE | ID: mdl-31850638

ABSTRACT

Inflammasomes are a group of multimolecular intracellular complexes assembled around several innate immune proteins. Recognition of a diverse range of microbial, stress and damage signals by inflammasomes results in direct activation of caspase-1, which subsequently induces the only known form of secretion of active interleukin-1ß and interleukin-18. Although the importance of interleukin-1ß in the periodontium is not questioned, the impact of inflammasomes in periodontal disease and its potential for therapeutics in periodontology is still in its very early stages. Increasing evidence in preclinical models and human data strongly implicate the involvement of inflammasomes in a number of inflammatory, autoinflammatory and autoimmune disorders. Here we review: (a) the currently known inflammasome functions, (b) clinical/preclinical data supporting inflammasome involvement in the context of periodontal and comorbid diseases and (c) potential therapies targeting inflammasomes. To clarify further the inflammasome involvement in periodontitis, we present analyses of data from a large clinical study (n = 5809) that measured the gingival crevicular fluid-interleukin-1ß and grouped the participants based on current periodontal disease classifications. We review data on 4910 European-Americans that correlate 16 polymorphisms in the interleukin-1B region with high gingival crevicular fluid-interleukin-1ß levels. We show that inflammasome components are increased in diseased periodontal tissues and that the caspase-1 inhibitor, VX-765, inhibits ~50% of alveolar bone loss in experimental periodontitis. The literature review further supports that although patients clinically present with the same phenotype, the disease that develops probably has different underlying biological pathways. The current data indicate that inflammasomes have a role in periodontal disease pathogenesis. Understanding the contribution of different inflammasomes to disease development and distinct patient susceptibility will probably translate into improved, personalized therapies.


Subject(s)
Inflammasomes , Periodontal Diseases , Caspase 1 , Gingival Crevicular Fluid , Humans , NLR Family, Pyrin Domain-Containing 3 Protein
7.
Mol Pharm ; 16(9): 4017-4023, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31361146

ABSTRACT

The in vivo antibacterial activity of NO-releasing hyperbranched polymers was evaluated against Porphyromonas gingivalis, a key oral pathogen associated with periodontitis, using a murine subcutaneous chamber model. Escalating doses of NO-releasing polymers (1.5, 7.5, and 37.5 mg/kg) were administered into a P. gingivalis-infected chamber once a day for 3 days. Chamber fluids were collected on day 4, with microbiological evaluation indicating a dose-dependent bactericidal action. In particular, NO-releasing polymers at 37.5 mg/kg (1170 µg of NO/kg) achieved complete bacterial eradication (>6-log reduction in bacterial viability), demonstrating greater efficacy than amoxicillin (∼4-log reduction in bacterial viability), a commonly used antibiotic. Time-kill assays further revealed that largest dose (37.5 mg/kg; 1170 µg of NO/kg) resulted in ∼3-log killing of P. gingivalis after only a single dose. Based on these results, the potential clinical utility of NO-releasing hyperbranched polymers appears promising, particularly for oral health applications.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacteroidaceae Infections/drug therapy , Nitric Oxide/chemistry , Nitric Oxide/therapeutic use , Periodontitis/drug therapy , Polymers/chemistry , Porphyromonas gingivalis/drug effects , Amoxicillin/therapeutic use , Animals , Bacteroidaceae Infections/microbiology , Disease Models, Animal , Epoxy Compounds/chemistry , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microbial Viability/drug effects , Periodontitis/microbiology , Polyamines/chemistry , Treatment Outcome
8.
Nat Protoc ; 13(10): 2247-2267, 2018 10.
Article in English | MEDLINE | ID: mdl-30218100

ABSTRACT

Periodontal disease (PD) is a common dental disease associated with the interaction between dysbiotic oral microbiota and host immunity. It is a prevalent disease, resulting in loss of gingival tissue, periodontal ligament, cementum and alveolar bone. PD is a major form of tooth loss in the adult population. Experimental animal models have enabled the study of PD pathogenesis and are used to test new therapeutic approaches for treating the disease. The ligature-induced periodontitis model has several advantages as compared with other models, including rapid disease induction, predictable bone loss and the capacity to study periodontal tissue and alveolar bone regeneration because the model is established within the periodontal apparatus. Although mice are the most convenient and versatile animal models used in research, ligature-induced periodontitis has been more frequently used in large animals. This is mostly due to the technical challenges involved in consistently placing ligatures around murine teeth. To reduce the technical challenge associated with the traditional ligature model, we previously developed a simplified method to easily install a bacterially retentive ligature between two molars for inducing periodontitis. In this protocol, we provide detailed instructions for placement of the ligature and demonstrate how the model can be used to evaluate gingival tissue inflammation and alveolar bone loss over a period of 18 d after ligature placement. This model can also be used on germ-free mice to investigate the role of human oral bacteria in periodontitis in vivo. In conclusion, this protocol enables the mechanistic study of the pathogenesis of periodontitis in vivo.


Subject(s)
Disease Models, Animal , Periodontitis/pathology , Animals , Bacteriological Techniques/methods , Female , Humans , Mice , Mice, Inbred C57BL , Periodontitis/etiology , Periodontitis/microbiology
9.
Nat Commun ; 9(1): 3686, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30206230

ABSTRACT

There is no agnostic GWAS evidence for the genetic control of IL-1ß expression in periodontal disease. Here we report a GWAS for "high" gingival crevicular fluid IL-1ß expression among 4910 European-American adults and identify association signals in the IL37 locus. rs3811046 at this locus (p = 3.3 × 10-22) is associated with severe chronic periodontitis (OR = 1.50; 95% CI = 1.12-2.00), 10-year incident tooth loss (≥3 teeth: RR = 1.33; 95% CI = 1.09-1.62) and aggressive periodontitis (OR = 1.12; 95% CI = 1.01-1.26) in an independent sample of 4927 German/Dutch adults. The minor allele at rs3811046 is associated with increased expression of IL-1ß in periodontal tissue. In RAW macrophages, PBMCs and transgenic mice, the IL37 variant increases expression of IL-1ß and IL-6, inducing more severe periodontal disease, while IL-37 protein production is impaired and shows reduced cleavage by caspase-1. A second variant in the IL37 locus (rs2708943, p = 4.2 × 10-7) associates with attenuated IL37 mRNA expression. Overall, we demonstrate that IL37 variants modulate the inflammatory cascade in periodontal disease.


Subject(s)
Genetic Variation , Genome-Wide Association Study , Gingival Crevicular Fluid/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-1/genetics , Interleukin-1beta/metabolism , Periodontium/pathology , Amino Acid Sequence , Animals , Chronic Periodontitis/blood , Chronic Periodontitis/genetics , Chronic Periodontitis/pathology , Disease Models, Animal , Female , Genetic Loci , HEK293 Cells , Haplotypes/genetics , Humans , Inflammation/blood , Interleukin-1/metabolism , Interleukin-1beta/blood , Interleukin-1beta/genetics , Leukocytes, Mononuclear/metabolism , Mice, Transgenic , Polymorphism, Single Nucleotide/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stroke/genetics , Tooth Loss/genetics
10.
J Periodontol ; 88(7): 663-672, 2017 07.
Article in English | MEDLINE | ID: mdl-28387608

ABSTRACT

BACKGROUND: The single nucleotide polymorphism (SNP) context of a previously identified periodontitis-associated locus is investigated, and its association with microbial, biologic, and periodontal disease clinical parameters is examined. METHODS: A 200-kb spanning region of 1q12 previously highlighted in a genome-wide association scan among 4,766 European American individuals (SNP rs1633266) was annotated. Two haplotype blocks were selected. Association of these polymorphisms with data on microbial plaque composition, gingival crevicular fluid (GCF)-interleukin (IL)-1ß levels, and clinical parameters of periodontal disease were examined. Descriptive analysis of IFI16 and AIM2 protein expression in gingival tissues from healthy individuals (n = 2) and individuals with chronic periodontitis (n = 2) was done via immunohistochemistry. RESULTS: The highlighted locus is a 100-kb region containing the interferon γ-inducible protein 16 (IFI16) and absent in melanoma 2 (AIM2) genes. Two haplotype blocks, rs6940 and rs1057028, were significantly associated with increased extent bleeding on probing and levels of microorganisms Porphyromonas gingivalis, Tannerella forsythia, and Campylobacter rectus (P ≤0.05). Haplotype block rs1057028 was also significantly associated with pathogens Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans, increased GCF-IL-1ß levels, and extent of probing depth ≥4 mm (P ≤0.05). Prevalence of severe periodontitis (biofilm-gingival interface P3 classification) was positively associated with haplotype block rs1057028. Similar trends were observed for haplotype block rs1057028. IFI16 and AIM2 protein expression was observed in multiple cell types of gingival tissues, including inflammatory cells. CONCLUSION: This study found IFI16 and AIM2 SNPs associated with higher levels of periodontal microorganisms and an increased percentage of periodontal disease clinical parameters, suggesting the need for functional studies and additional fine-mapping of variants in the 1q12-locus.


Subject(s)
Chronic Periodontitis/genetics , DNA-Binding Proteins/genetics , Gingiva/metabolism , Nuclear Proteins/genetics , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , Aged , Aggregatibacter actinomycetemcomitans/isolation & purification , Chronic Periodontitis/metabolism , Chronic Periodontitis/microbiology , DNA-Binding Proteins/metabolism , Dental Plaque/microbiology , Female , Fusobacterium nucleatum/isolation & purification , Genome-Wide Association Study , Gingiva/microbiology , Humans , Male , Middle Aged , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , White People
11.
PLoS One ; 11(7): e0158866, 2016.
Article in English | MEDLINE | ID: mdl-27409077

ABSTRACT

Strain NI1060 is an oral bacterium responsible for periodontitis in a murine ligature-induced disease model. To better understand its pathogenicity, we have determined the complete sequence of its 2,553,982 bp genome. Although closely related to Pasteurella pneumotropica, a pneumonia-associated rodent commensal based on its 16S rRNA, the NI1060 genomic content suggests that they are different species thriving on different energy sources via alternative metabolic pathways. Genomic and phylogenetic analyses showed that strain NI1060 is distinct from the genera currently described in the family Pasteurellaceae, and is likely to represent a novel species. In addition, we found putative virulence genes involved in lipooligosaccharide synthesis, adhesins and bacteriotoxic proteins. These genes are potentially important for host adaption and for the induction of dysbiosis through bacterial competition and pathogenicity. Importantly, strain NI1060 strongly stimulates Nod1, an innate immune receptor, but is defective in two peptidoglycan recycling genes due to a frameshift mutation. The in-depth analysis of its genome thus provides critical insights for the development of NI1060 as a prime model system for infectious disease.


Subject(s)
DNA, Bacterial/genetics , Genome, Bacterial/genetics , Pasteurellaceae/classification , Pasteurellaceae/genetics , Periodontitis/microbiology , Base Sequence , Lipopolysaccharides/chemistry , Pasteurellaceae/pathogenicity , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Virulence Factors
12.
Adv Healthc Mater ; 5(6): 676-87, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26820240

ABSTRACT

Scaffold design incorporating multiscale cues for clinically relevant, aligned tissue regeneration has potential to improve structural and functional integrity of multitissue interfaces. The objective of this preclinical study is to develop poly(ε-caprolactone) (PCL) scaffolds with mesoscale and microscale architectural cues specific to human ligament progenitor cells and assess their ability to form aligned bone-ligament-cementum complexes in vivo. PCL scaffolds are designed to integrate a 3D printed bone region with a micropatterned PCL thin film consisting of grooved pillars. The patterned film region is seeded with human ligament cells, fibroblasts transduced with bone morphogenetic protein-7 genes seeded within the bone region, and a tooth dentin segment positioned on the ligament region prior to subcutaneous implantation into a murine model. Results indicate increased tissue alignment in vivo using micropatterned PCL films, compared to random-porous PCL. At week 6, 30 µm groove depth significantly enhances oriented collagen fiber thickness, overall cell alignment, and nuclear elongation relative to 10 µm groove depth. This study demonstrates for the first time that scaffolds with combined hierarchical mesoscale and microscale features can align cells in vivo for oral tissue repair with potential for improving the regenerative response of other bone-ligament complexes.


Subject(s)
Polyesters/chemistry , Printing, Three-Dimensional , Tissue Scaffolds , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Regeneration/physiology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cells, Cultured , Collagen/chemistry , Disease Models, Animal , Humans , Mice , Microscopy, Fluorescence , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Periodontal Ligament/transplantation , X-Ray Microtomography
13.
Mol Oral Microbiol ; 31(3): 243-258, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26177212

ABSTRACT

Periodontitis is a polymicrobial inflammatory disease that results from the interaction between the oral microbiota and the host immunity. Although the innate immune response is important for disease initiation and progression, the innate immune receptors that recognize both classical and putative periodontal pathogens that elicit an immune response have not been elucidated. By using the Human Oral Microbe Identification Microarray (HOMIM), we identified multiple predominant oral bacterial species in human plaque biofilm that strongly associate with severe periodontitis. Ten of the identified species were evaluated in greater depth, six being classical pathogens and four putative novel pathogens. Using human peripheral blood monocytes (HPBM) and murine bone-marrow-derived macrophages (BMDM) from wild-type (WT) and Toll-like receptor (TLR)-specific and MyD88 knockouts (KOs), we demonstrated that heat-killed Campylobacter concisus, Campylobacter rectus, Selenomonas infelix, Porphyromonas endodontalis, Porphyromonas gingivalis, and Tannerella forsythia mediate high immunostimulatory activity. Campylobacter concisus, C. rectus, and S. infelix exhibited robust TLR4 stimulatory activity. Studies using mesothelial cells from WT and NOD1-specific KOs and NOD2-expressing human embryonic kidney cells demonstrated that Eubacterium saphenum, Eubacterium nodatum and Filifactor alocis exhibit robust NOD1 stimulatory activity, and that Porphyromonas endodontalis and Parvimonas micra have the highest NOD2 stimulatory activity. These studies allowed us to provide important evidence on newly identified putative pathogens in periodontal disease pathogenesis showing that these bacteria exhibit different immunostimulatory activity via TLR4, NOD1, and NOD2 (Clinicaltrials.gov NCT01154855).


Subject(s)
Dental Plaque/microbiology , Immunization , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Periodontal Diseases/immunology , Periodontal Diseases/microbiology , Toll-Like Receptor 4/immunology , Animals , Biofilms , Campylobacter rectus/immunology , Campylobacter rectus/isolation & purification , Campylobacter rectus/pathogenicity , Dental Plaque/immunology , Female , Humans , Macrophages/immunology , Male , Mice , Mice, Knockout , Monocytes , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Periodontal Diseases/physiopathology , Porphyromonas/immunology , Porphyromonas/isolation & purification , Porphyromonas/pathogenicity , Porphyromonas endodontalis/immunology , Porphyromonas endodontalis/isolation & purification , Porphyromonas endodontalis/pathogenicity , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/isolation & purification , Tannerella forsythia/immunology , Tannerella forsythia/isolation & purification , Tannerella forsythia/pathogenicity
14.
Trends Microbiol ; 22(3): 157-63, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24433922

ABSTRACT

Periodontitis is a common dental disease which results in irreversible alveolar bone loss around teeth, and subsequent tooth loss. Previous studies have focused on bacteria that damage the host and the roles of commensals to facilitate their colonization. Although some immune responses targeting oral bacteria protect the host from alveolar bone loss, recent studies show that particular host defense responses to oral bacteria can induce alveolar bone loss. Host-damaging and immunostimulatory oral bacteria cooperatively induce bone loss by inducing gingival damage followed by immunostimulation. In mouse models of experimental periodontitis induced by either Porphyromonas gingivalis or ligature, γ-proteobacteria accumulate and stimulate host immune responses to induce host damage. Here we review the differential roles of individual bacterial groups in promoting bone loss through the induction of host damage and immunostimulation.


Subject(s)
Alveolar Bone Loss/immunology , Alveolar Bone Loss/microbiology , Bacteria/immunology , Mouth/microbiology , Periodontitis/immunology , Periodontitis/microbiology , Alveolar Bone Loss/pathology , Animals , Disease Models, Animal , Mice , Periodontitis/complications , Periodontitis/pathology
15.
Cell Host Microbe ; 13(5): 595-601, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23684310

ABSTRACT

Periodontitis is a common disease that is characterized by resorption of the alveolar bone and mediated by commensal bacteria that trigger host immune responses and bone destruction through unidentified mechanisms. We report that Nod1, an innate intracellular host receptor for bacterial peptidoglycan-related molecules, is critical for commensal-induced periodontitis in a mouse model. Mice lacking Nod1 exhibit reduced bone resorption as well as impaired recruitment of neutrophils to gingival tissues and osteoclasts to the alveolar bone, which mediate tissue and bone destruction. Further analysis showed that accumulation of a Nod1-stimulating commensal bacterium, NI1060, at gingival sites was sufficient to induce neutrophil recruitment and bone resorption. Genomic sequencing revealed that NI1060 is a mouse-specific bacterium that is related to bacteria associated with the development of aggressive periodontitis in humans. These findings provide insight into commensal-host interactions contributing to periodontitis and identify a potential target for preventing this common oral disease.


Subject(s)
Alveolar Bone Loss/pathology , Bacteria/pathogenicity , Host-Pathogen Interactions , Mouth/microbiology , Nod1 Signaling Adaptor Protein/metabolism , Periodontitis/pathology , Signal Transduction , Animals , Mice , Periodontitis/complications
16.
J Immunol ; 189(6): 3085-91, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22888139

ABSTRACT

Clostridium difficile is a Gram-positive obligate anaerobic pathogen that causes pseudomembranous colitis in antibiotic-treated individuals. Commensal bacteria are known to have a significant role in the intestinal accumulation of C. difficile after antibiotic treatment, but little is known about how they affect host immunity during C. difficile infection. In this article, we report that C. difficile infection results in translocation of commensals across the intestinal epithelial barrier that is critical for neutrophil recruitment through the induction of an IL-1ß-mediated positive-feedback loop. Mice lacking ASC, an essential mediator of IL-1ß and IL-18 processing and secretion, were highly susceptible to C. difficile infection. ASC(-/-) mice exhibited enhanced translocation of commensals to multiple organs after C. difficile infection. Notably, ASC(-/-) mice exhibited impaired CXCL1 production and neutrophil influx into intestinal tissues in response to C. difficile infection. The impairment in neutrophil recruitment resulted in reduced production of IL-1ß and CXCL1 but not IL-18. Importantly, translocated commensals were required for ASC/Nlrp3-dependent IL-1ß secretion by neutrophils. Mice lacking IL-1ß were deficient in inducing CXCL1 secretion, suggesting that IL-1ß is the dominant inducer of ASC-mediated CXCL1 production during C. difficile infection. These results indicate that translocated commensals play a crucial role in CXCL1-dependent recruitment of neutrophils to the intestine through an IL-1ß/NLRP3/ASC-mediated positive-feedback mechanism that is important for host survival and clearance of translocated commensals during C. difficile infection.


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Enterocolitis, Pseudomembranous/prevention & control , Interleukin-1beta/physiology , Symbiosis/immunology , Up-Regulation/immunology , Animals , Biological Transport, Active/genetics , Biological Transport, Active/immunology , Cell Communication/immunology , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Enterocolitis, Pseudomembranous/pathology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Survival Analysis , Up-Regulation/genetics
17.
Biochem Biophys Res Commun ; 391(4): 1579-84, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19968964

ABSTRACT

Type-I ribosome-inactivating protein-trichosanthin (TCS) exhibits selective cytotoxicity toward different types of cells. It is believed that the cytotoxicity results from the inhibition of ribosomes to decrease protein synthesis, thereby indicating that there are specific mechanisms for TCS entry into target cells to reach the ribosomes. Low-density lipoprotein (LDL) receptor-related protein 1 (LRP1) is a large scavenger receptor that is responsible for the binding and endocytosis of diverse biological ligands on the cell surface. In this study, we demonstrated that 2 choriocarcinoma cell lines can significantly bind and internalize TCS. In contrast, Hela cell line displayed no obvious TCS binding and endocytosis. Furthermore LRP1 gene silencing in JAR and BeWo cell lines blocked TCS binding; TCS could also interact with LRP1.The results of our study established that LRP1 was a major receptor for phagocytosis of TCS in JAR and BeWo cell lines and might be the molecular basis of TCS abortificient and anti-choriocarcinoma activity.


Subject(s)
Abortifacient Agents, Nonsteroidal/metabolism , Antineoplastic Agents, Phytogenic/metabolism , Choriocarcinoma/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Trichosanthin/metabolism , Abortifacient Agents, Nonsteroidal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Endocytosis , HeLa Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Phagocytosis , RNA, Small Interfering/genetics , Trichosanthin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...