Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 11: 571049, 2020.
Article in English | MEDLINE | ID: mdl-33193352

ABSTRACT

Objectives: Rheumatoid arthritis is an autoimmune disease with multifactorial etiopathogenesis. Among the environmental factors, mucosal infections and the inducing pathobionts are gaining increasing attention. We here set out to explore the gut-joint-axis and the impact of Clostridioides difficile infection on subsequent arthritis. Methods: We combined C. difficile infection in DBA/1J × B10.Q F1 mice with collagen induced arthritis (CIA). Mice were infected via oral gavage and infection was monitored by weight loss, colonic histology, and antibodies against bacteria. Scoring of arthritis was performed macroscopically. Intestinal microbiomes were analyzed and immune responses were monitored via quantification of transcription factor-specific mRNA isolated from the inguinal and mesenteric lymph nodes. Results: Infection with C. difficile VPI 10463 resulted in significant weight loss and severe colitis yet accelerated the reversal towards the original microbiome after antibiotic treatment. Spontaneous clearance of VPI 10463 infection reduced the incidence of subsequent CIA and led to mesenteric Treg and Th2 polarization. However, this attenuating effect was abrogated if VPI 10463 was eradicated via vancomycin followed by fecal microbiota transplantation. Moreover, VPI 10463 infection following the onset of CIA lacked therapeutic potential. Conclusion: Our results demonstrate that infection with C. difficile VPI10463 induced an inflammation of the gut that protected from subsequent arthritis development in mice. Both, microbial changes to the gut and immune cell mobilization and/or polarization may have contributed to arthritis protection. The prospect of potential therapeutic benefits resulting from C. difficile infections or some byproduct thereof call for further experiments that help elucidate exact mechanisms.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Clostridioides difficile/physiology , Clostridium Infections/immunology , Mesentery/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Disease Models, Animal , Disease Resistance , Fecal Microbiota Transplantation , Feces/microbiology , Gastrointestinal Microbiome , Humans , Lymphocyte Activation , Mice , Mice, Inbred DBA
2.
Sci Rep ; 8(1): 15129, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30310087

ABSTRACT

Increasing evidence supports the association of periodontitis with rheumatoid arthritis. Even though a prominent role has been postulated for Porphyromonas gingivalis, many bacterial species contribute to the pathogenesis of periodontal disease. We therefore investigated the impact of Porphyromonas gingivalis as well as other major pathobionts on the development of both, periodontitis and arthritis in the mouse. Pathobionts used - either alone or in combination - were Porphyromonas gingivalis, Fusobacterium nucleatum and Aggregatibacter actinomycetemcomintans. Periodontitis was induced via oral gavage in SKG, DBA/1 and F1 (DBA/1 × B10.Q) mice and collagen-induced arthritis was provoked via immunization and boost with bovine collagen type II. Alveolar bone loss was quantified via micro computed tomography, arthritis was evaluated macroscopically and histologically and serum antibodies were assessed. Among the strains tested, only F1 mice were susceptible to P. gingivalis induced periodontitis and showed significant alveolar bone loss. Bone loss was paralleled by antibody titers against P. gingivalis. Of note, mice inoculated with the mix of all three pathobionts showed less alveolar bone loss than mice inoculated with P. gingivalis alone. However, oral inoculation with either F. nucleatum or A. actinomycetemcomintans alone accelerated subsequent arthritis onset and progression. This is the first report of a triple oral inoculation of pathobionts combined with collagen-induced arthritis in the mouse. In this interplay and this particular genetic setting, F. nucleatum and A. actinomycetemcomitans exerted a protective impact on P. gingivalis induced alveolar bone loss. By themselves they did not induce periodontitis yet accelerated arthritis onset and progression.


Subject(s)
Actinobacteria , Alveolar Bone Loss/etiology , Alveolar Bone Loss/pathology , Arthritis/etiology , Arthritis/pathology , Fusobacterium nucleatum , Porphyromonas gingivalis , Actinobacteria/physiology , Alveolar Bone Loss/metabolism , Animals , Antibodies, Bacterial/immunology , Arthritis/metabolism , Arthritis, Experimental , Disease Models, Animal , Disease Progression , Disease Susceptibility , Fusobacterium nucleatum/physiology , Mice , Periodontitis/etiology , Periodontitis/pathology , Porphyromonas gingivalis/physiology
3.
J Innate Immun ; 10(4): 264-278, 2018.
Article in English | MEDLINE | ID: mdl-29860256

ABSTRACT

Periodontitis (PD) is a widespread chronic inflammatory disease in the human population. Porphyromonas gingivalis is associated with PD and can citrullinate host proteins via P. gingivalis peptidyl arginine deiminase (PPAD). Here, we hypothesized that infection of human dental follicle stem cells (hDFSCs) with P. gingivalis and subsequent interaction with neutrophils will alter the neutrophil phenotype. To test this hypothesis, we established and analyzed a triple-culture system of neutrophils and hDFSCs primed with P. gingivalis. Mitogen-activated pathway blocking reagents were applied to gain insight into stem cell signaling after infection. Naïve hDFSCs do not influence the neutrophil phenotype. However, infection of hDFSCs with P. gingivalis prolongs the survival of neutrophils and increases their migration. These phenotypic changes depend on direct cellular contacts and PPAD expression by P. gingivalis. Active JNK and ERK pathways in primed hDFSCs are essential for the phenotypic changes in neutrophils. Collectively, our results confirm that P. gingivalis modifies hDFSCs, thereby causing an immune imbalance.


Subject(s)
Bacterial Proteins/metabolism , Bacteroidaceae Infections/immunology , Dental Sac/pathology , Neutrophils/physiology , Periodontitis/immunology , Porphyromonas gingivalis/physiology , Protein-Arginine Deiminases/metabolism , Stem Cells/physiology , Cells, Cultured , Coculture Techniques , Humans , Immunomodulation , MAP Kinase Kinase 4/metabolism , Neutrophil Activation , Signal Transduction , Stem Cells/microbiology
4.
Front Microbiol ; 9: 53, 2018.
Article in English | MEDLINE | ID: mdl-29441048

ABSTRACT

A wide range of bacterial species are harbored in the oral cavity, with the resulting complex network of interactions between the microbiome and host contributing to physiological as well as pathological conditions at both local and systemic levels. Bacterial communities inhabit the oral cavity as primary niches in a symbiotic manner and form dental biofilm in a stepwise process. However, excessive formation of biofilm in combination with a corresponding deregulated immune response leads to intra-oral diseases, such as dental caries, gingivitis, and periodontitis. Moreover, oral commensal bacteria, which are classified as so-called "pathobionts" according to a now widely accepted terminology, were recently shown to be present in extra-oral lesions with distinct bacterial species found to be involved in the onset of various pathophysiological conditions, including cancer, atherosclerosis, chronic infective endocarditis, and rheumatoid arthritis. The present review focuses on oral pathobionts as commensal and healthy members of oral biofilms that can turn into initiators of disease. We will shed light on the processes involved in dental biofilm formation and also provide an overview of the interactions of P. gingivalis, as one of the most prominent oral pathobionts, with host cells, including epithelial cells, phagocytes, and dental stem cells present in dental tissues. Notably, a previously unknown interaction of P. gingivalis bacteria with human stem cells that has impact on human immune response is discussed. In addition to this very specific interaction, the present review summarizes current knowledge regarding the immunomodulatory effect of P. gingivalis and other oral pathobionts, members of the oral microbiome, that pave the way for systemic and chronic diseases, thereby showing a link between periodontitis and rheumatoid arthritis.

5.
Front Biosci (Schol Ed) ; 9(1): 180-193, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814584

ABSTRACT

The regeneration of periodontal tissues still remains a challenge in periodontology. The aim of the present study was to examine the regenerative potential of a) different collagen support versus blank, b) different collagen support +/- a growth factor cocktail (GF) and c) a collagen powder versus collagen powder + periodontal ligament stem cells (PDLSCs) comparatively in a large animal model. The stem cells (SC) were isolated from extracted teeth of 15 adult miniature pigs. A total of 60 class II furcation defects were treated with the materials named above. Concluding, a histological evaluation followed. A significant increase in regeneration was observed in all treatment groups. The new attachment formation reached a maximum of 77 percent. In the control group a new attachment formation of 13 percent was observed. The study shows that all implanted materials improved periodontal regeneration, though there were no significant differences between the experimental groups. Within the limitations of this study, it can be assumed that the lack of significant differences is due to the complexity of the clinical setting.


Subject(s)
Periodontal Ligament/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Cementogenesis , Collagen , Furcation Defects/therapy , Intercellular Signaling Peptides and Proteins/pharmacology , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Random Allocation , Regeneration/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Swine , Swine, Miniature , Tissue Scaffolds
6.
Sci Rep ; 6: 39096, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27974831

ABSTRACT

Periodontitis is characterized by inflammation associated with the colonization of different oral pathogens. We here aimed to investigate how bacteria and host cells shape their environment in order to limit inflammation and tissue damage in the presence of the pathogen. Human dental follicle stem cells (hDFSCs) were co-cultured with gram-negative P. intermedia and T. forsythia and were quantified for adherence and internalization as well as migration and interleukin secretion. To delineate hDFSC-specific effects, gingival epithelial cells (Ca9-22) were used as controls. Direct effects of hDFSCs on neutrophils (PMN) after interaction with bacteria were analyzed via chemotactic attraction, phagocytic activity and NET formation. We show that P. intermedia and T. forsythia adhere to and internalize into hDFSCs. This infection decreased the migratory capacity of the hDFSCs by 50%, did not disturb hDFSC differentiation potential and provoked an increase in IL-6 and IL-8 secretion while leaving IL-10 levels unaltered. These environmental modulations correlated with reduced PMN chemotaxis, phagocytic activity and NET formation. Our results suggest that P. intermedia and T. forsythia infected hDFSCs maintain their stem cell functionality, reduce PMN-induced tissue and bone degradation via suppression of PMN-activity, and at the same time allow for the survival of the oral pathogens.


Subject(s)
Dental Sac/cytology , Neutrophils/cytology , Periodontitis/microbiology , Prevotella intermedia/pathogenicity , Stem Cells/cytology , Tannerella forsythia/pathogenicity , Bacterial Adhesion , Cell Differentiation , Cell Line , Cell Movement , Dental Sac/immunology , Dental Sac/microbiology , Female , Gingiva/cytology , Humans , Interleukins/metabolism , Male , Periodontitis/immunology , Prevotella intermedia/immunology , Stem Cells/immunology , Stem Cells/microbiology , Tannerella forsythia/immunology
7.
J Cell Mol Med ; 19(9): 2067-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26058313

ABSTRACT

Mesenchymal stem cells (MSCs) are widely recognized as critical players in tissue regeneration. New insights into stem cell biology provide evidence that MSCs may also contribute to host defence and inflammation. In case of tissue injury or inflammatory diseases, e.g. periodontitis, stem cells are mobilized towards the site of damage, thus coming in close proximity to bacteria and bacterial components. Specifically, in the oral cavity, complex ecosystems of commensal bacteria live in a mutually beneficial state with the host. However, the formation of polymicrobial biofilm communities with pathogenic properties may trigger an inadequate host inflammatory-immune response, leading to the disruption of tissue homoeostasis and development of disease. Because of their unique characteristics, MSCs are suggested as crucial regulators of tissue regeneration even under such harsh environmental conditions. The heterogeneous effects of bacteria on MSCs across studies imply the complexity underlying the interactions between stem cells and bacteria. Hence, a better understanding of stem cell behaviour at sites of inflammation appears to be a key strategy in developing new approaches for in situ tissue regeneration. Here, we review the literature on the effects of oral bacteria on cell proliferation, differentiation capacity and immunomodulation of dental-derived MSCs.


Subject(s)
Bacteria/metabolism , Mouth/microbiology , Regeneration , Stem Cells/cytology , Cell Differentiation , Humans , Immunomodulation
8.
PLoS One ; 9(11): e110616, 2014.
Article in English | MEDLINE | ID: mdl-25369260

ABSTRACT

BACKGROUND: In patients with periodontitis, it is highly likely that local (progenitor) cells encounter pathogenic bacteria. The purpose of this in vitro study was to elucidate how human dental follicle stem cells (hDFSC) react towards a direct challenge with anaerobic periodontal pathogens under their natural oxygen-free atmosphere. HDFSC were compared to human bone marrow mesenchymal stem cells (hBMSC) and differentiated primary human gingival fibroblasts (hGiF), as well as permanent gingival carcinoma cells (Ca9-22). METHODOLOGY/PRINCIPAL FINDINGS: The different cell types were investigated in a co-culture system with Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum). The viability of the cells and pathogens under anaerobic conditions, as well as interactions in terms of adherence and internalization, were examined. Additionally, the release of pro-inflammatory interleukin-8 (IL-8) and anti-inflammatory interleukin-10 (IL-10) was quantified via enzyme-linked immunosorbent assay. The bacteria adhered less efficiently to hDFSC compared to Ca9-22 (P. gingivalis: 0.18% adherence to hDFSC; 3.1% adherence to Ca9-22). Similar results were observed for host cell internalization (F. nucleatum: 0.002% internalization into hDFSC; 0.09% internalization into Ca9-22). Statistically significantly less IL-8 was secreted from hDFSC after stimulation with F. nucleatum and P. gingivalis in comparison with hGiF (F. nucleatum: 2080.0 pg/ml--hGiF; 19.7 pg/ml--hDFSC). The IL-10 response of the differentiated cells was found to be low in relation to their pro-inflammatory IL-8 response. CONCLUSIONS/SIGNIFICANCE: The results indicate that dental stem cells are less prone to interactions with pathogenic bacteria than differentiated cells in an anaerobic environment. Moreover, during bacterial challenge, the stem cell immune response seems to be more towards an anti-inflammatory reaction. For a potential future therapeutic use of hDFSC, these findings support the idea of a save application.


Subject(s)
Dental Sac/cytology , Porphyromonas gingivalis/physiology , Stem Cells/microbiology , Bacterial Adhesion/physiology , Bone Marrow Cells/cytology , Cell Survival , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-10/analysis , Interleukin-8/analysis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/microbiology , Microscopy, Fluorescence , Porphyromonas gingivalis/growth & development , Stem Cells/cytology , Stem Cells/metabolism
9.
PLoS One ; 8(11): e78226, 2013.
Article in English | MEDLINE | ID: mdl-24223777

ABSTRACT

BACKGROUND: Human mesenchymal stem cells (hMSCs) are multipotent by nature and are originally isolated from bone marrow. In light of a future application of hMSCs in the oral cavity, a body compartment with varying oxygen partial pressures and an omnipresence of different bacterial species i.e. periodontitis pathogens, we performed this study to gain information about the behavior of hMSC in an anaerobic system and the response in interaction with oral bacterial pathogens. METHODOLOGY/PRINCIPAL FINDINGS: We established a model system with oral pathogenic bacterial species and eukaryotic cells cultured in anaerobic conditions. The facultative anaerobe bacteria Fusobacterium nucleatum, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans were studied. Their effects on hMSCs and primary as well as permanent gingival epithelial cells (Ca9-22, HGPEC) were comparatively analyzed. We show that hMSCs cope with anoxic conditions, since 40% vital cells remain after 72 h of anaerobic culture. The Ca9-22 and HGPEC cells are significantly more sensitive to lack of oxygen. All bacterial species reveal a comparatively low adherence to and internalization into hMSCs (0.2% and 0.01% of the initial inoculum, respectively). In comparison, the Ca9-22 and HGPEC cells present better targets for bacterial adherence and internalization. The production of the pro-inflammatory chemokine IL-8 is higher in both gingival epithelial cell lines compared to hMSCs and Fusobacterium nucleatum induce a time-dependent cytokine secretion in both cell lines. Porphyromonas gingivalis is less effective in stimulating secretion of IL-8 in the co-cultivation experiments. CONCLUSIONS/SIGNIFICANCE: HMSCs are suitable for use in anoxic regions of the oral cavity. The interaction with local pathogenic bacteria does not result in massive pro-inflammatory cytokine responses. The test system established in this study allowed further investigation of parameters prior to set up of oral hMSC in vivo studies.


Subject(s)
Aggregatibacter actinomycetemcomitans/physiology , Fusobacterium nucleatum/physiology , Mesenchymal Stem Cells/immunology , Porphyromonas gingivalis/physiology , Aggregatibacter actinomycetemcomitans/pathogenicity , Anaerobiosis , Bacterial Adhesion , Cell Hypoxia , Cell Line , Coculture Techniques , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Fusobacterium nucleatum/pathogenicity , Gingiva/cytology , Gingiva/immunology , Gingiva/microbiology , Humans , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/microbiology , Porphyromonas gingivalis/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...