Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Oral Microbiol ; 39(2): 40-46, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37459655

ABSTRACT

The oral organism Tannerella forsythia is auxotrophic for peptidoglycan amino sugar N-acetylmuramic acid (MurNAc). It survives in the oral cavity by scavenging MurNAc- and MurNAc-linked peptidoglycan fragments (muropeptides) secreted by co-habiting bacteria such as Fusobacterium nucleatum with which it forms synergistic biofilms. Muropeptides, MurNAc-l-Ala-d-isoGln (MDP, muramyl dipeptide) and d-γ-glutamyl-meso-DAP (iE-DAP dipeptide), are strong immunostimulatory molecules that activate nucleotide oligomerization domain (NOD)-like innate immune receptors and induce the expression of inflammatory cytokines and antimicrobial peptides. In this study, we utilized an in vitro T. forsythia-F. nucleatum co-culture model to determine if T. forsythia can selectively scavenge NOD ligands from the environment and impact NOD-mediated inflammation. The results showed that NOD-stimulatory molecules were secreted by F. nucleatum in the spent culture broth, which subsequently induced cytokine and antimicrobial peptide expression in oral epithelial cells. In the spent broth from T. forsythia-F. nucleatum co-cultures, the NOD-stimulatory activity was significantly reduced. These data indicated that F. nucleatum releases NOD2-stimulatory muropeptides in the environment, and T. forsythia can effectively scavenge the muropeptides released by co-habiting bacteria to dampen NOD-mediated host responses. This proof-of-principle study demonstrated that peptidoglycan scavenging by T. forsythia can impact the innate immunity of oral epithelium by dampening NOD activation.


Subject(s)
Fusobacterium nucleatum , Tannerella forsythia , Tannerella forsythia/metabolism , Fusobacterium nucleatum/physiology , Peptidoglycan , Mouth , Epithelial Cells/metabolism , Cytokines/metabolism
2.
Mol Oral Microbiol ; 37(3): 109-121, 2022 06.
Article in English | MEDLINE | ID: mdl-35576119

ABSTRACT

Interleukin-34 (IL-34) is a cytokine that supports the viability and differentiation of macrophages. An important cytokine for the development of epidermal immunity, IL-34, is present and plays a role in the immunity of the oral environment. IL-34 has been linked to inflammatory periodontal diseases, which involve innate phagocytes, including macrophages. Whether IL-34 can alter the ability of macrophages to effectively interact with oral microbes is currently unclear. Using macrophages derived from human blood monocytes with either the canonical cytokine colony-stimulating factor (CSF)1 or IL-34, we compared the ability of the macrophages to phagocytose, kill, and respond through the production of cytokines to the periodontal keystone pathogen Porphyromonas gingivalis. While macrophages derived from both cytokines were able to engulf the bacterium equally, IL-34-derived macrophages were much less capable of killing internalized P. gingivalis. Of the macrophage cell surface receptors known to interact with P. gingivalis, dendritic cell-specific intercellular adhesion molecule-grabbing nonintegrin was found to have the largest variation between IL-34- and CSF1-derived macrophages. We also found that upon interaction with P. gingivalis, IL-34-derived macrophages produced significantly less of the neutrophil chemotactic factor IL-8 than macrophages derived in the presence of CSF1. Mechanistically, we identified that the levels of IL-8 corresponded with P. gingivalis survival and dephosphorylation of the major transcription factor NF-κB p65. Overall, we found that macrophages differentiated in the presence of IL-34, a dominant cytokine in the oral gingiva, have a reduced ability to kill the keystone pathogen P. gingivalis and may be susceptible to specific bacteria-mediated cytokine modification.


Subject(s)
Interleukin-8 , Interleukins/immunology , Macrophages/immunology , Porphyromonas gingivalis , Bacteroidaceae Infections/immunology , Gingiva/immunology , Gingiva/microbiology , Gingival Diseases/immunology , Humans , NF-kappa B/metabolism , NF-kappa B/pharmacology , Porphyromonas gingivalis/metabolism
3.
Pathog Dis ; 80(1)2022 04 29.
Article in English | MEDLINE | ID: mdl-35404415

ABSTRACT

Tannerella forsythia is strongly implicated in the development of periodontitis, an inflammatory disease that destroys the bone and soft tissues supporting the tooth.  To date, the knowledge of the virulence attributes of T. forsythia species has mainly come from studies with a laboratory adapted strain (ATCC 43037). In this study, we focused on two T. forsythia clinical isolates, UB4 and UB20, in relation to their ability to activate macrophages. We found that these clinical isolates differentially induced proinflammatory cytokine expression in macrophages. Prominently, the expression of the chemokine protein IP-10 (CXCL10) was highly induced by UB20 as compared to UB4 and the laboratory strain ATCC 43037. Our study focused on the lipopolysaccharide component (LPS) of these strains and found that UB20 expressed a smooth-type LPS, unlike UB4 and ATCC 43037 each of which expressed a rough-type LPS. The LPS from UB20, via activation of TLR4, was found to be a highly potent inducer of IP-10 expression via signaling through STAT1 (signal transducer and activator of transcription-1). These data suggest that pathogenicity of T. forsythia species could be strain dependent and the LPS heterogeneity associated with the clinical strains might be responsible for their pathogenic potential and severity of periodontitis.


Subject(s)
Periodontitis , Tannerella forsythia , Chemokine CXCL10/genetics , Humans , Interferon-gamma , Lipopolysaccharides , Macrophages
4.
Mol Oral Microbiol ; 37(2): 42-52, 2022 04.
Article in English | MEDLINE | ID: mdl-34958712

ABSTRACT

Recent epidemiological studies have shown that inflammatory bowel disease is associated with periodontal disease. The oral-gut microbiota axis is a potential mechanism intersecting the two diseases. Porphyromonas gingivalis is currently considered a keystone oral pathogen involved in periodontal disease pathogenesis and disease progression. Recent studies have shown that oral ingestion of P. gingivalis leads to intestinal inflammation. However, the molecular underpinnings of P. gingivalis-mediated gut inflammation have remained elusive. In this study, we show that the oral administration of P. gingivalis indeed leads to ileal inflammation and alteration in gut microbiota with significant reduction in bacterial alpha diversity despite the absence of P. gingivalis in the lower gastrointestinal tract. Utilizing an antibiotic-conditioned mouse model, cecal microbiota transfer experiments were performed to demonstrate that P. gingivalis-induced dysbiotic gut microbiota is sufficient to reproduce gut pathology. Furthermore, we observed a significant expansion in small intestinal lamina propria IL9+ CD4+ T cells, which was negatively correlated with both bacterial and fungal alpha diversity, signifying that P. gingivalis-mediated intestinal inflammation may be due to the subsequent loss of gut microbial diversity. Finally, we detected changes in gene expression related to gut epithelial barrier function, showing the potential downstream effect of intestinal IL9+ CD4+ T-cell induction. This study for the first time showed the mechanism behind P. gingivalis-mediated intestinal inflammation where P. gingivalis indirectly induces intestinal IL9+ CD4+ T cells and inflammation by altering the gut microbiota. Understanding the mechanism of P. gingivalis-mediated intestinal inflammation may lead to the development of novel therapeutic approaches to alleviate the morbidity from inflammatory bowel disease patients with periodontal disease.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Periodontal Diseases , Animals , CD4-Positive T-Lymphocytes , Humans , Inflammation/pathology , Interleukin-9 , Mice , Periodontal Diseases/microbiology , Porphyromonas gingivalis/genetics , T-Lymphocytes
5.
Front Physiol ; 12: 722859, 2021.
Article in English | MEDLINE | ID: mdl-34594237

ABSTRACT

Periodontitis is a bacterially-induced inflammatory disease that leads to tooth loss. It results from the damaging effects of a dysregulated immune response, mediated largely by neutrophils, macrophages, T cells and B cells, on the tooth-supporting tissues including the alveolar bone. Specifically, infiltrating B cells at inflamed gingival sites with an ability to secrete RANKL and inflammatory cytokines are thought to play roles in alveolar bone resorption. However, the direct contribution of B cells in alveolar bone resorption has not been fully appreciated. In this study we sought to define the contribution of RANKL expressing B cells in periodontitis by employing a mouse model of pathogen-induced periodontitis that used conditional knockout mice with B cell-targeted RANKL deletion. Briefly, alveolar bone loss was assessed in the wild-type, B-cell deficient (Jh), or B-cell-RANKL deleted (RANKLΔB) mice orally infected with the periodontal pathogen Tannerella forsythia. The RANKLΔB mice were obtained by crossing Cd19-Cre knock-in mice with mice homozygous for conditional RANKL-flox allele (RANKLflox/flox). The alveolar bone resorption was determined by morphometric analysis and osteoclastic activity of the jaw bone. In addition, the bone resorptive potential of the activated effector B cells was assessed ex vivo. The data showed that the RANKL producing B cells increased significantly in the T. forsythia-infected wild-type mice compared to the sham-infected mice. Moreover, T. forsythia-infection induced higher alveolar bone loss in the wild-type and RANKLflox/flox mice compared to infection either in the B cell deficient (Jh) or the B-cell specific RANKL deletion (RANKLΔB) mice. These data established that the oral-pathogen activated B cells contribute significantly to alveolar bone resorption via RANKL production.

6.
PLoS One ; 12(3): e0173394, 2017.
Article in English | MEDLINE | ID: mdl-28264048

ABSTRACT

The oral pathogen Tannerella forsythia is implicated in the development of periodontitis, a common inflammatory disease that leads to the destruction of the gum and tooth supporting tissues, often leading to tooth loss. T. forsythia is a unique Gram-negative organism endowed with an elaborate protein O-glycosylation system that allows the bacterium to express a glycosylated surface (S)-layer comprising two high molecular weight glycoproteins modified with O-linked oligosaccharides. The T. forsythia S-layer has been implicated in the modulation of cytokine responses of antigen presenting cells, such as macrophages, that play a significant role during inflammation associated with periodontitis. The macrophage-inducible C-type lectin receptor (Mincle) is an FcRγ-coupled pathogen recognition receptor that recognizes a wide variety of sugar containing ligands from fungal and bacterial pathogens. In this study, we aimed to determine if Mincle might be involved in the recognition of T. forsythia S-layer and modulation of cytokine response of macrophages against the bacterium. Binding studies using recombinant Mincle-Fc fusion protein indicated a specific Ca2+-dependent binding of Mincle to T. forsythia S-layer. Subsequent experiments with Mincle-expressing and Mincle-knockdown macrophages revealed a role for Mincle/S-layer interaction in the induction of both pro- and anti-inflammatory cytokine secretion in macrophages stimulated with T. forsythia as well as its S-layer. Together, these studies revealed Mincle as an important macrophage receptor involved in the modulation of cytokine responses of macrophages against T. forsythia, and thus may play a critical role in orchestrating the host immune response against the bacterium.


Subject(s)
Lectins, C-Type/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Periodontitis/microbiology , Tannerella forsythia/immunology , Tannerella forsythia/metabolism , Cell Differentiation , Cell Line , Cytokines/metabolism , Glycosylation , Humans , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/immunology , Periodontitis/genetics , Periodontitis/immunology , Phagocytosis/immunology , Protein Binding , RNA, Small Interfering/genetics , Tannerella forsythia/pathogenicity
7.
PLoS One ; 9(9): e108030, 2014.
Article in English | MEDLINE | ID: mdl-25225799

ABSTRACT

Alveolar bone (tooth-supporting bone) erosion is a hallmark of periodontitis, an inflammatory disease that often leads to tooth loss. Periodontitis is caused by a select group of pathogens that form biofilms in subgingival crevices between the gums and teeth. It is well-recognized that the periodontal pathogen Porphyromonas gingivalis in these biofilms is responsible for modeling a microbial dysbiotic state, which then initiates an inflammatory response destructive to the periodontal tissues and bone. Eradication of this pathogen is thus critical for the treatment of periodontitis. Previous studies have shown that oral inoculation in mice with an attenuated strain of the periodontal pathogen Tannerella forsythia altered in O-glycan surface composition induces a Th17-linked mobilization of neutrophils to the gingival tissues. In this study, we sought to determine if immune priming with such a Th17-biasing strain would elicit a productive neutrophil response against P. gingivalis. Our data show that inoculation with a Th17-biasing T. forsythia strain is effective in blocking P. gingivalis-persistence and associated alveolar bone loss in mice. This work demonstrates the potential of O-glycan modified Tannerella strains or their O-glycan components for harnessing Th17-mediated immunity against periodontal and other mucosal pathogens.


Subject(s)
Alveolar Bone Loss/immunology , Bacteria/immunology , Bacteria/metabolism , Neutrophils/immunology , Periodontitis/immunology , Periodontitis/microbiology , Polysaccharides, Bacterial/immunology , Th17 Cells/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Neutrophil Infiltration/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
Front Microbiol ; 4: 310, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24146665

ABSTRACT

Protein modification with complex glycans is increasingly being recognized in many pathogenic and non-pathogenic bacteria, and is now thought to be central to the successful life-style of those species in their respective hosts. This review aims to convey current knowledge on the extent of protein glycosylation in periodontal pathogenic bacteria and its role in the modulation of the host immune responses. The available data show that surface glycans of periodontal bacteria orchestrate dendritic cell cytokine responses to drive T cell immunity in ways that facilitate bacterial persistence in the host and induce periodontal inflammation. In addition, surface glycans may help certain periodontal bacteria protect against serum complement attack or help them escape immune detection through glycomimicry. In this review we will focus mainly on the generalized surface-layer protein glycosylation system of the periodontal pathogen Tannerella forsythia in shaping innate and adaptive host immunity in the context of periodontal disease. In addition, we will also review the current state of knowledge of surface protein glycosylation and its potential for immune modulation in other periodontal pathogens.

9.
Immunol Invest ; 42(7): 519-31, 2013.
Article in English | MEDLINE | ID: mdl-24004056

ABSTRACT

Bone undergoes a continuous cycle of remodeling for maintenance and healing. For almost a decade it has been appreciated that the immune system is intricately linked to bone homeostasis. Both acute and chronic inflammatory responses have been shown to impact bone health. A common form of inflammatory disease that causes bone destruction is the chronic infectious disease known as periodontitis (PD). PD is a bacteria-driven inflammation of the tooth-supporting apparatus that leads to resorption of the alveolar (jaw) bone, often leading to tooth loss. At the host-bacteria interface, Toll-like receptors (TLRs) play an instructive role in the development of innate and T cell adaptive responses to oral bacteria. Specifically, it is becoming apparent that TLR2-mediated inflammatory responses represent the major arm of the host immune response during periodontitis, and form an important link between periodontal infection and ensuing periodontal bone loss. This review summarizes the role of TLR2-mediated interplay between immune cells and bone cells in a periodontal disease setting.


Subject(s)
Jaw , Periodontitis/etiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptors/metabolism , Alveolar Bone Loss/immunology , Alveolar Bone Loss/metabolism , Animals , Humans , Jaw/immunology , Jaw/metabolism , Jaw/microbiology , Jaw/pathology , Signal Transduction , Toll-Like Receptor 2/metabolism
10.
Biochem Biophys Res Commun ; 423(3): 577-82, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22695115

ABSTRACT

Pathogenesis of many bacterially-induced inflammatory diseases is driven by Toll-like receptor (TLR) mediated immune responses following recognition of bacterial factors by different TLRs. Periodontitis is a chronic inflammation of the tooth supporting apparatus often leading to tooth loss, and is caused by a Gram-negative bacterial consortium that includes Tannerella forsythia. This bacterium expresses a virulence factor, the BspA, which drives periodontal inflammation by activating TLR2. The N-terminal portion of the BspA protein comprises a leucine-rich repeat (LRR) domain previously shown to be involved in the binding and activation of TLR2. The objective of the current study was to identify specific epitopes in the LRR domain of BspA that interact with TLR2. Our results demonstrate that a sequence motif GC(S/T)GLXSIT is involved in mediating the interaction of BspA with TLR2. Thus, our study has identified a peptide motif that mediates the binding of a bacterial protein to TLR2 and highlights the promiscuous nature of TLR2 with respect to ligand binding. This work could provide a structural basis for designing peptidomimetics to modulate the activity of TLR2 in order to block bacterially-induced inflammation.


Subject(s)
Bacterial Proteins/metabolism , Bacteroidetes/metabolism , Membrane Proteins/metabolism , Toll-Like Receptor 2/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , HEK293 Cells , Humans , Leucine , Membrane Proteins/chemistry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Periodontitis/microbiology , Protein Interaction Maps , Protein Structure, Tertiary , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 2/genetics , Trypsin/chemistry
11.
Infect Immun ; 80(7): 2436-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22547549

ABSTRACT

Tannerella forsythia is strongly associated with chronic periodontitis, an inflammatory disease of the tooth-supporting tissues, leading to tooth loss. Fusobacterium nucleatum, an opportunistic pathogen, is thought to promote dental plaque formation by serving as a bridge bacterium between early- and late-colonizing species of the oral cavity. Previous studies have shown that F. nucleatum species synergize with T. forsythia during biofilm formation and pathogenesis. In the present study, we showed that coinfection of F. nucleatum and T. forsythia is more potent than infection with either species alone in inducing NF-κB activity and proinflammatory cytokine secretion in monocytic cells and primary murine macrophages. Moreover, in a murine model of periodontitis, mixed infection with the two species induces synergistic alveolar bone loss, characterized by bone loss which is greater than the additive alveolar bone losses induced by each species alone. Further, in comparison to the single-species infection, mixed infection caused significantly increased inflammatory cell infiltration in the gingivae and osteoclastic activity in the jaw bones. These data show that F. nucleatum subspecies and T. forsythia synergistically stimulate the host immune response and induce alveolar bone loss in a murine experimental periodontitis model.


Subject(s)
Alveolar Bone Loss/microbiology , Alveolar Bone Loss/pathology , Bacteroidetes/pathogenicity , Fusobacterium nucleatum/pathogenicity , Gram-Negative Bacterial Infections/pathology , Periodontitis/microbiology , Periodontitis/pathology , Animals , Coinfection/microbiology , Coinfection/pathology , Disease Models, Animal , Female , Gram-Negative Bacterial Infections/microbiology , Mice , Mice, Inbred BALB C
12.
J Immunol ; 187(1): 501-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632710

ABSTRACT

Periodontal disease (PD) is a chronic inflammation of the tooth-supporting soft tissue and alveolar bone due to infection by a select group of gram-negative microbes, which leads to tooth loss if untreated. Because mice deficient in CD4(+) cells are resistant to infection-induced alveolar bone loss, Th cells have been implicated in bone-destructive processes during PD. However, the extent to which different Th cell subtypes play roles in pathogenesis or host protection remains to be defined and is likely to vary depending on the dominant microorganism involved. By far, Porphyromonas gingivalis is the best-studied periodontal microbe in PD. Although the gram-negative anaerobe Tannerella forsythia is also a vital contributor to periodontal bone loss, almost nothing is known about immune responses to this organism. Previous studies from our laboratory revealed that T. forsythia induces periodontal bone loss in mice and that this bone loss depends on the bacterially expressed BspA protein. In this study, we showed that T. forsythia activates murine APCs primarily through TLR2-dependent signaling via BspA. Furthermore, T. forsythia infection causes a pronounced Th2 bias, evidenced by T cell expression of IL-5, but not IFN-γ or IL-17, in draining lymph nodes. Consistently, deficiencies in TLR2 or STAT6 result in resistance to T. forsythia-induced alveolar bone loss. Thus, TLR2 signaling and Th2 cells play pathogenic roles in T. forsythia-induced alveolar bone destruction.


Subject(s)
Alveolar Bone Loss/immunology , Flavobacteriaceae Infections/immunology , Flavobacteriaceae/immunology , Signal Transduction/immunology , Th2 Cells/immunology , Toll-Like Receptor 2/physiology , Alveolar Bone Loss/genetics , Alveolar Bone Loss/pathology , Animals , Flavobacteriaceae Infections/genetics , Flavobacteriaceae Infections/pathology , Inflammation Mediators/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Random Allocation , Signal Transduction/genetics , Th2 Cells/metabolism , Th2 Cells/pathology , Toll-Like Receptor 2/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...