1001
|
Koide M, Kobayashi Y, Ninomiya T, Nakamura M, Yasuda H, Arai Y, Okahashi N, Yoshinari N, Takahashi N, Udagawa N. Osteoprotegerin-deficient male mice as a model for severe alveolar bone loss: comparison with RANKL-overexpressing transgenic male mice. Endocrinology 2013; 154:773-82. [PMID: 23291450 DOI: 10.1210/en.2012-1928] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Periodontitis, an inflammatory disease of periodontal tissues, is characterized by excessive alveolar bone resorption. An increase in the receptor activator of nuclear factor-κB ligand (RANKL) to osteoprotegerin (OPG) ratio is thought to reflect the severity of periodontitis. Here, we examined alveolar bone loss in OPG-deficient (OPG(-/-)) mice and RANKL-overexpressing transgenic (RANKL-Tg) mice. Alveolar bone loss in OPG(-/-) mice at 12 weeks was significantly higher than that in RANKL-Tg mice. OPG(-/-) but not RANKL-Tg mice exhibited severe bone resorption especially in cortical areas of the alveolar bone. An increased number of osteoclasts was observed in the cortical areas in OPG(-/-) but not in RANKL-Tg mice. Immunohistochemical analyses showed many OPG-positive signals in osteocytes but not osteoblasts. OPG-positive osteocytes in the cortical area of alveolar bones and long bones were abundant in both wild-type and RANKL-Tg mice. This suggests the resorption in cortical bone areas to be prevented by OPG produced locally. To test the usefulness of OPG(-/-) mice as an animal model for screening drugs to prevent alveolar bone loss, we administered an antimouse RANKL antibody or risedronate, a bisphosphonate, to OPG(-/-) mice. They suppressed alveolar bone resorption effectively. OPG(-/-) mice are useful for screening therapeutic agents against alveolar bone loss.
Collapse
Affiliation(s)
- Masanori Koide
- Institute for Oral Science, Matsumoto Dental University, Nagano 399-0781, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1002
|
|
1003
|
Nowak M, Krämer B, Haupt M, Papapanou PN, Kebschull J, Hoffmann P, Schmidt-Wolf IG, Jepsen S, Brossart P, Perner S, Kebschull M. Activation of invariant NK T cells in periodontitis lesions. THE JOURNAL OF IMMUNOLOGY 2013; 190:2282-91. [PMID: 23365081 DOI: 10.4049/jimmunol.1201215] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Periodontitis is one of the most prevalent human inflammatory diseases. The major clinical phenotypes of this polymicrobial, biofilm-mediated disease are chronic and aggressive periodontitis, the latter being characterized by a rapid course of destruction that is generally attributed to an altered immune-inflammatory response against periodontal pathogens. Still, the biological basis for the pathophysiological distinction of the two disease categories has not been well documented yet. Type I NKT cells are a lymphocyte subset with important roles in regulating immune responses to either tolerance or immunity, including immune responses against bacterial pathogens. In this study, we delineate the mechanisms of NKT cell activation in periodontal infections. We show an infiltration of type I NKT cells in aggressive, but not chronic, periodontitis lesions in vivo. Murine dendritic cells infected with aggressive periodontitis-associated Aggregatibacter actinomycetemcomitans triggered a type I IFN response followed by type I NKT cell activation. In contrast, infection with Porphyromonas gingivalis, a principal pathogen in chronic periodontitis, did not induce NKT cell activation. This difference could be explained by the absence of a type I IFN response to P. gingivalis infection. We found these IFNs to be critical for NKT cell activation. Our study provides a conceivable biological distinction between the two periodontitis subforms and identifies factors required for the activation of the immune system in response to periodontal bacteria.
Collapse
Affiliation(s)
- Michael Nowak
- Department of Prostate Cancer Research, Institute of Pathology, University of Bonn, Bonn 53127, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1004
|
Kebschull M, Haupt M, Jepsen S, Deschner J, Nickenig G, Werner N. Mobilization of endothelial progenitors by recurrent bacteremias with a periodontal pathogen. PLoS One 2013; 8:e54860. [PMID: 23355901 PMCID: PMC3552864 DOI: 10.1371/journal.pone.0054860] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 12/19/2012] [Indexed: 11/19/2022] Open
Abstract
Background Periodontal infections are independent risk factors for atherosclerosis. However, the exact mechanisms underlying this link are yet unclear. Here, we evaluate the in vivo effects of bacteremia with a periodontal pathogen on endothelial progenitors, bone marrow-derived cells capable of endothelial regeneration, and delineate the critical pathways for these effects. Methods 12-week old C57bl6 wildtype or toll-like receptor (TLR)-2 deficient mice were repeatedly intravenously challenged with 109 live P. gingivalis 381 or vehicle. Numbers of Sca1+/flk1+ progenitors, circulating angiogenic cells, CFU-Hill, and late-outgrowth EPC were measured by FACS/culture. Endothelial function was assessed using isolated organ baths, reendothelization was measured in a carotid injury model. RANKL/osteoprotegerin levels were assessed by ELISA/qPCR. Results In wildtype mice challenged with intravenous P.gingivalis, numbers of Sca1+/flk1+ progenitors, CAC, CFU-Hill, and late-outgrowth EPC were strongly increased in peripheral circulation and spleen, whereas Sca1+/flk1+ progenitor numbers in bone marrow decreased. Circulating EPCs were functional, as indicated by improved endothelial function and improved reendothelization in infected mice. The osteoprotegerin/RANKL ratio was increased after P. gingivalis challenge in the bone marrow niche of wildtype mice and late-outgrowth EPC in vitro. Conversely, in mice deficient in TLR2, no increase in progenitor mobilization or osteoprotegerin/RANKL ratio was detected. Conclusion Recurrent transient bacteremias, a feature of periodontitis, increase peripheral EPC counts and decrease EPC pools in the bone marrow, thereby possibly reducing overall endothelial regeneration capacity, conceivably explaining pro-atherogenic properties of periodontal infections. These effects are seemingly mediated by toll-like receptor (TLR)-2.
Collapse
Affiliation(s)
- Moritz Kebschull
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
- Department of Internal Medicine II, University of Bonn, Bonn, Germany
| | - Manuela Haupt
- Department of Internal Medicine II, University of Bonn, Bonn, Germany
| | - Søren Jepsen
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| | - James Deschner
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, University of Bonn, Bonn, Germany
| | - Nikos Werner
- Department of Internal Medicine II, University of Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
1005
|
Shin J, Kho SA, Choi YS, Kim YC, Rhyu IC, Choi Y. Antibody and T cell responses to Fusobacterium nucleatum and Treponema denticola in health and chronic periodontitis. PLoS One 2013; 8:e53703. [PMID: 23335969 PMCID: PMC3546045 DOI: 10.1371/journal.pone.0053703] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 12/03/2012] [Indexed: 12/14/2022] Open
Abstract
The characteristics of the T cell response to the members of oral flora are poorly understood. We characterized the antibody and T cell responses to FadA and Td92, adhesins from Fusobacterium nucleatum, an oral commensal, and Treponema denticola, a periodontal pathogen, respectively. Peripheral blood and saliva were obtained from healthy individuals and patients with untreated chronic periodontitis (CP, n = 11 paris) and after successful treatment of the disease (n = 9). The levels of antigen-specific antibody were measured by ELISA. In plasma, IgG1 was the most abundant isotype of Ab for both Ags, followed by IgA and then IgG4. The levels of FadA-specific salivary IgA (sIgA) were higher than Td92-specific sIgA and the FadA-specific IgA levels observed in plasma. However, the periodontal health status of the individuals did not affect the levels of FadA- or Td92-specific antibody. Even healthy individuals contained FadA- and Td92-specific CD4+ T cells, as determined by the detection of intracytoplasmic CD154 after short-term in vitro stimulation of peripheral blood mononuclear cells (PBMCs) with the antigens. Patients with CP tended to possess increased numbers of FadA- and Td92-specific CD4+ T cells but reduced numbers of Td92-specific Foxp3+CD4+ Tregs than the healthy subjects. Both FadA and Td92 induced the production of IFNγ and IL-10 but inhibited the secretion of IL-4 by PBMCs. In conclusion, F. nucleatum induced Th3 (sIgA)- and Th1 (IFNγ and IgG1)-dominant immune responses, whereas T. denticola induced a Th1 (IFNγ and IgG1)-dominant response. This IFNγ-dominant cytokine response was impaired in CP patients, and the Td92-induced IFNγ levels were negatively associated with periodontal destruction in patients. These findings may provide new insights into the homeostatic interaction between the immune system and oral bacteria and the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Jieun Shin
- Department of Immunology and Molecular Microbiology, BK21 CLS, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Sang-A Kho
- Department of Periodontology, BK21 CLS, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Yun S. Choi
- Department of Immunology and Molecular Microbiology, BK21 CLS, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Yong C. Kim
- Department of Immunology and Molecular Microbiology, BK21 CLS, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - In-Chul Rhyu
- Department of Periodontology, BK21 CLS, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Youngnim Choi
- Department of Immunology and Molecular Microbiology, BK21 CLS, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
- * E-mail:
| |
Collapse
|
1006
|
Li X, Lan HY, Huang XR, Zhang C, Jin LJ. Expression profile of macrophage migration-inhibitory factor in human gingiva and reconstituted human gingival epithelia stimulated by Porphyromonas gingivalis lipopolysaccharide. J Periodontal Res 2013; 48:527-32. [PMID: 23298274 DOI: 10.1111/jre.12035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2012] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Macrophage migration-inhibitory factor (MIF) plays crucial roles in the recruitment and activation of macrophages as well as in helping to kill bacteria. This study investigated the expression profile of MIF in human gingiva under different periodontal conditions and its expression patterns induced by Porphyromonas gingivalis lipopolysaccharide (LPS) in gingival epithelia. MATERIAL AND METHODS Gingival tissue samples were collected from deep pockets and clinically healthy sites of 22 nonsmoking subjects with chronic periodontitis. The expression of MIF mRNA and protein was evaluated using real-time PCR and immunohistochemistry, respectively. The in vitro study analyzed the effects of P. gingivalis LPS on the expression of MIF in a reconstituted human gingival epithelia (RHGE) model. RESULTS In gingival epithelia, MIF protein was diffusely expressed from the basal layer to the granular and spinous layers; whereas, in the underlying connective tissues, MIF was observed around the dilated blood vessels in the deep-pocket tissues. A significantly lower level of expression of MIF mRNA and an increased level of expression of MIF protein were found in deep-pocket tissues compared with clinically healthy tissues. Expression of MIF mRNA in the RHGE model was significantly down-regulated by P. gingivalis LPS. CONCLUSION The present study suggests that MIF expression may be related to periodontal conditions and that its expression profile could be modulated by P. gingivalis LPS. MIF may play a role in periodontal pathogenesis.
Collapse
Affiliation(s)
- X Li
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
1007
|
Does propolis help to maintain oral health? EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:351062. [PMID: 23365605 PMCID: PMC3556426 DOI: 10.1155/2013/351062] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 12/22/2012] [Indexed: 12/19/2022]
Abstract
Propolis, known also as bee glue, is a wax-cum-resin substance which is created out of a mix of buds from some trees with the substance secreted from bee's glands. Its diverse chemical content is responsible for its many precious salubrious properties. It was used in medicine already in ancient Egypt. Its multiple applications during the centuries have been studied and described in details. The purpose of this study is to present the possible use of propolis in treatment of various diseases of oral cavity in their dental aspect. The paper presents properties and possible applications of bee glue depending on dental specialities. An overview of publications which appeared during the recent years will allow the reader to follow all the possibilities to apply propolis in contemporary dentistry.
Collapse
|
1008
|
Scheres N, Crielaard W. Gingival fibroblast responsiveness is differentially affected by Porphyromonas gingivalis: implications for the pathogenesis of periodontitis. Mol Oral Microbiol 2012; 28:204-18. [PMID: 23279858 DOI: 10.1111/omi.12016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2012] [Indexed: 12/26/2022]
Abstract
In periodontitis, tissue damage results mainly from aberrant host responses to oral microorganisms. Fibroblasts can play an important role in this. Gingival fibroblasts do not develop tolerance against the lipopolysaccharide of Porphyromonas gingivalis, a keystone pathogen in periodontitis, which may partly explain the persistence of inflammation. However, besides lipopolysaccharide, live P. gingivalis possess numerous virulence traits to impair host-responses. We hypothesized that fibroblast-responsiveness to a bacterial challenge could be affected by live P. gingivalis. We investigated if inflammatory responses of gingival fibroblasts to P. gingivalis were altered, when the fibroblasts had encountered P. gingivalis previously. On consecutive days, primary human gingival fibroblasts were challenged twice for 6 h with live P. gingivalis, or fibroblasts were preincubated for 24 h with a lower concentration of live P. gingivalis and re-challenged for 6 h with a higher concentration. As the P. gingivalis capsule and proteases are involved in modulating host responses, we used encapsulated P. gingivalis W83 and a non-encapsulated mutant, and P. gingivalis ATCC33277 and a lys-gingipain and arg-gingipain mutant, to challenge fibroblasts. With all P. gingivalis-strains, interleukin-8 and monocyte chemoattractant protein-1 responses to the second challenge were less strong in fibroblasts that had been challenged with P. gingivalis before. These lower responses might correspond with higher interleukin-1 receptor agonist expression. Fibroblast responses to a second challenge were not influenced by 24 h preincubation. Reduced chemokine responses after consecutive potent P. gingivalis challenges indicate that gingival fibroblast responsiveness is affected by a previous bacterial encounter. In periodontitis, such reduced chemokine responses may impair chemotaxis and clearance of oral microorganisms, thereby leading to prolonged inflammatory responses and tissue damage.
Collapse
Affiliation(s)
- N Scheres
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam, VU University Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
1009
|
Abstract
Periodontitis is a highly prevalent, biofilm-mediated chronic inflammatory disease that results in the loss of the tooth-supporting tissues. It features two major clinical entities: chronic periodontitis, which is more common, and aggressive periodontitis, which usually has an early onset and a rapid progression. Natural killer (NK) cells are a distinct subgroup of lymphocytes that play a major role in the ability of the innate immune system to steer immune responses. NK cells are abundant in periodontitis lesions, and NK cell activation has been causally linked to periodontal tissue destruction. However, the exact mechanisms of their activation and their role in the pathophysiology of periodontitis are elusive. Here, we show that the predominant NK cell-activating molecule in periodontitis is CD2-like receptor activating cytotoxic cells (CRACC). We show that CRACC induction was significantly more pronounced in aggressive than chronic periodontitis and correlated positively with periodontal disease severity, subgingival levels of specific periodontal pathogens, and NK cell activation in vivo. We delineate how Aggregatibacter actinomycetemcomitans, an oral pathogen that is causally associated with aggressive periodontitis, indirectly induces CRACC on NK cells via activation of dendritic cells and subsequent interleukin 12 (IL-12) signaling. In contrast, we demonstrate that fimbriae from Porphyromonas gingivalis, a principal pathogen in chronic periodontitis, actively attenuate CRACC induction on NK cells. Our data suggest an involvement of CRACC-mediated NK cell activation in periodontal tissue destruction and point to a plausible distinction in the pathobiology of aggressive and chronic periodontitis that may help explain the accelerated tissue destruction in aggressive periodontitis.
Collapse
|
1010
|
Luo W, Wang CY, Jin L. Baicalin downregulates Porphyromonas gingivalis lipopolysaccharide-upregulated IL-6 and IL-8 expression in human oral keratinocytes by negative regulation of TLR signaling. PLoS One 2012; 7:e51008. [PMID: 23239998 PMCID: PMC3519831 DOI: 10.1371/journal.pone.0051008] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 10/31/2012] [Indexed: 11/18/2022] Open
Abstract
Periodontal (gum) disease is one of the main global oral health burdens and severe periodontal disease (periodontitis) is a leading cause of tooth loss in adults globally. It also increases the risk of cardiovascular disease and diabetes mellitus. Porphyromonas gingivalis lipopolysaccharide (LPS) is a key virulent attribute that significantly contributes to periodontal pathogenesis. Baicalin is a flavonoid from Scutellaria radix, an herb commonly used in traditional Chinese medicine for treating inflammatory diseases. The present study examined the modulatory effect of baicalin on P. gingivalis LPS-induced expression of IL-6 and IL-8 in human oral keratinocytes (HOKs). Cells were pre-treated with baicalin (0–80 µM) for 24 h, and subsequently treated with P. gingivalis LPS at 10 µg/ml with or without baicalin for 3 h. IL-6 and IL-8 transcripts and proteins were detected by real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. The expression of nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) proteins was analyzed by western blot. A panel of genes related to toll-like receptor (TLR) signaling was examined by PCR array. We found that baicalin significantly downregulated P. gingivalis LPS-stimulated expression of IL-6 and IL-8, and inhibited P. gingivalis LPS-activated NF-κB, p38 MAPK and JNK. Furthermore, baicalin markedly downregulated P. gingivalis LPS-induced expression of genes associated with TLR signaling. In conclusion, the present study shows that baicalin may significantly downregulate P. gingivalis LPS-upregulated expression of IL-6 and IL-8 in HOKs via negative regulation of TLR signaling.
Collapse
Affiliation(s)
- Wei Luo
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Cun-Yu Wang
- University of California Los Angeles, School of Dentistry, Los Angeles, California, United States of America
| | - Lijian Jin
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
- * E-mail:
| |
Collapse
|
1011
|
Moutsopoulos NM, Kling HM, Angelov N, Jin W, Palmer RJ, Nares S, Osorio M, Wahl SM. Porphyromonas gingivalis promotes Th17 inducing pathways in chronic periodontitis. J Autoimmun 2012; 39:294-303. [PMID: 22560973 PMCID: PMC3416947 DOI: 10.1016/j.jaut.2012.03.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 03/10/2012] [Accepted: 03/25/2012] [Indexed: 01/05/2023]
Abstract
In periodontitis, a common chronic inflammatory condition, gram-negative-rich bacterial biofilms trigger, in susceptible individuals, perpetuating inflammation that results in extensive tissue damage of tooth supporting structures. To delineate immune cell-dependent mechanisms whereby bacterial challenge drives persistent destructive inflammation in periodontitis and other inflammatory diseases, we studied involved tissues ex vivo and investigated host cell responses to the periodontal pathogen Porphyromonas gingivalis, in vitro. Diseased lesions were populated by abundant Th17 cells, linked to infection, chronic inflammation/autoimmunity and tissue pathology. In vitro, P. gingivalis, particularly the more virulent strain W83, stimulated myeloid antigen presenting cells (APC) to drive Th17 polarization. Supernatants from myeloid APC exposed to P. gingivalis were capable of enhancing Th17 but not Th1 polarization. P. gingivalis favored the generation of Th17 responses by stimulating the production of Th17 related cytokines IL-1β, IL-6 and IL-23, but not Th1 related IL-12. By inducing NFκB activation, P. gingivalis promoted IL-1β, IL-6 and IL-12p40 production, but not IRF3 phosphorylation, connected to generation of the IL-12p35 chain, ultimately restricting formation of the intact IL-12 molecule. Promotion of Th17 lineage responses was also aided by P. gingivalis proteases, which appeared to differentially degrade pivotal cytokines. In this regard, IL-12 was largely degraded by P. gingivalis, whereas IL-1β was more resistant to proteolysis. Our data unveil multiple pathways by which P. gingivalis may orchestrate chronic inflammation, providing insights into interventional strategies.
Collapse
Affiliation(s)
- Niki M Moutsopoulos
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4352, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1012
|
Abstract
The human mouth harbours one of the most diverse microbiomes in the human body, including viruses, fungi, protozoa, archaea and bacteria. The bacteria are responsible for the two commonest bacterial diseases of man: dental caries (tooth decay) and the periodontal (gum) diseases. Archaea are restricted to a small number of species of methanogens while around 1000 bacterial species have been found, with representatives from the phyla Actinobacteria, Bacteroidetes, Firmicutes, Proteobacteria, Spirochaetes, Synergistetes and Tenericutes and the uncultured divisions GN02, SR1 and TM7. Around half of oral bacteria are as yet uncultured and culture-independent methods have been successfully used to comprehensively describe the oral bacterial community. The human oral microbiome database (HOMD, www.homd.org) provides a comprehensive resource consisting of descriptions of oral bacterial taxa, a 16S rRNA identification tool and a repository of oral bacterial genome sequences. Individuals' oral microbiomes are highly specific at the species level, although overall the human oral microbiome shows few geographical differences. Although caries and periodontitis are clearly bacterial diseases, they are not infectious diseases in the classical sense because they result from a complex interaction between the commensal microbiota, host susceptibility and environmental factors such as diet and smoking. Periodontitis, in particular, appears to result from an inappropriate inflammatory reaction to the normal microbiota, exacerbated by the presence of some disease-associated bacterial species. In functional terms, there appears to considerable redundancy among the oral microbiota and a focus on functional rather than phylogenetic diversity may be required in order to fully understand host-microbiome interactions.
Collapse
Affiliation(s)
- William G Wade
- King's College London Dental Institute, Microbiology Unit, Floor 17, Tower Wing, Guy's Campus, London SE1 9RT, UK.
| |
Collapse
|
1013
|
Abstract
The major strides accomplished in elucidating the pathophysiology of rheumatoid arthritis (RA) have translated into therapeutic breakthroughs in clinical practice. However, currently available treatments work only for as long as they are taken. The development of curative treatments will probably require a better understanding of the earliest phases of RA and perhaps the identification of the etiological factors, which are probably numerous. These objectives are being pursued in studies of preclinical RA. The literature review presented herein indicates that the immunological conflict probably originates outside the joints, at mucous membrane sites and, more specifically, in the upper aerodigestive tract. The preclinical phase of RA can last for many years, and some patients probably never progress to arthritis. An immunological conflict develops then spins out of control, causing increases in autoantibody titers and subsequently in levels of serum markers for inflammation, before the development of the first joint symptoms. Improved knowledge of the preclinical phase, together with information from genetic markers, will allow the identification of profiles associated with susceptibility to RA and perhaps, in the future, the development of preventive strategies.
Collapse
|
1014
|
Peyyala R, Ebersole JL. Multispecies biofilms and host responses: "discriminating the trees from the forest". Cytokine 2012; 61:15-25. [PMID: 23141757 DOI: 10.1016/j.cyto.2012.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 09/28/2012] [Accepted: 10/04/2012] [Indexed: 02/07/2023]
Abstract
Periodontal diseases reflect a tissue destructive process of the hard and soft tissues of the periodontium that are initiated by the accumulation of multispecies bacterial biofilms in the subgingival sulcus. This accumulation, in both quantity and quality of bacteria, results in a chronic immunoinflammatory response of the host to control this noxious challenge, leading to collateral damage of the tissues. As knowledge of the characteristics of the host-bacterial interactions in the oral cavity has expanded, new knowledge has become available on the complexity of the microbial challenge and the repertoire of host responses to this challenge. Recent results from the Human Microbiome Project continue to extend the array of taxa, genera, and species of bacteria that inhabit the multiple niches in the oral cavity; however, there is rather sparse information regarding variations in how host cells discriminate commensal from pathogenic species, as well as how the host response is affected by the three-dimensional architecture and interbacterial interactions that occur in the oral biofilms. This review provides some insights into these processes by including existing literature on the biology of nonoral bacterial biofilms, and the more recent literature just beginning to document how the oral cavity responds to multispecies biofilms.
Collapse
Affiliation(s)
- R Peyyala
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY 40536, United States
| | | |
Collapse
|
1015
|
Ding PH, Wang CY, Darveau RP, Jin LJ. Nuclear factor-κB and p38 mitogen-activated protein kinase signaling pathways are critically involved in Porphyromonas gingivalis lipopolysaccharide induction of lipopolysaccharide-binding protein expression in human oral keratinocytes. Mol Oral Microbiol 2012. [PMID: 23194012 DOI: 10.1111/omi.12010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Lipopolysaccharide (LPS) -binding protein (LBP) plays a crucial role in innate host response to bacterial challenge. Porphyromonas gingivalis is a keystone pathogen in periodontal disease and the shift of P. gingivalis LPS lipid A structure from penta-acylated (LPS(1690)) to tetra-acylated (LPS(1435/1449)) isoform may significantly contribute to periodontal pathogenesis. We recently demonstrated that LBP is expressed in human gingiva and contributes to periodontal homeostasis. Furthermore, different isoforms of P. gingivalis LPS differently modulate the immuno-inflammatory response, and P. gingivalis LPS(1690) induces LBP expression in human oral keratinocytes (HOKs). This study further examined the signaling mechanisms of P. gingivalis LPS(1690) -induced and Escherichia coli LPS-induced LBP expression in HOKs. Both P. gingivalis LPS(1690) and E. coli LPS were potent inducers of LBP expression in HOKs. The former activated phosphorylation of IκBα, p65, p38 mitogen-activated protein kinase (MAPK) and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), whereas the latter phosphorylated IκBα, p38 MAPK and SAPK/JNK. A nuclear translocation of NF-κB transcription factor was confirmed upon stimulation by both forms of LPS. Further blocking assay showed that P. gingivalis LPS(1690) induction of LBP was through NF-κB and p38 MPAK pathways, whereas E. coli LPS-induced LBP expression was mediated by NF-κB, p38 MPAK and JNK pathways. This study demonstrates that NF-κB and p38 MAPK signaling pathways are involved in P. gingivalis LPS(1690) induction of LBP expression in HOKs. The current findings could enhance the understanding of the molecular mechanisms of innate defense in maintenance of periodontal homeostasis.
Collapse
Affiliation(s)
- P-H Ding
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | | | | | | |
Collapse
|
1016
|
Klein BA, Tenorio EL, Lazinski DW, Camilli A, Duncan MJ, Hu LT. Identification of essential genes of the periodontal pathogen Porphyromonas gingivalis. BMC Genomics 2012; 13:578. [PMID: 23114059 PMCID: PMC3547785 DOI: 10.1186/1471-2164-13-578] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/24/2012] [Indexed: 01/09/2023] Open
Abstract
Background Porphyromonas gingivalis is a Gram-negative anaerobic bacterium associated with periodontal disease onset and progression. Genetic tools for the manipulation of bacterial genomes allow for in-depth mechanistic studies of metabolism, physiology, interspecies and host-pathogen interactions. Analysis of the essential genes, protein-coding sequences necessary for survival of P. gingivalis by transposon mutagenesis has not previously been attempted due to the limitations of available transposon systems for the organism. We adapted a Mariner transposon system for mutagenesis of P. gingivalis and created an insertion mutant library. By analyzing the location of insertions using massively-parallel sequencing technology we used this mutant library to define genes essential for P. gingivalis survival under in vitro conditions. Results In mutagenesis experiments we identified 463 genes in P. gingivalis strain ATCC 33277 that are putatively essential for viability in vitro. Comparing the 463 P. gingivalis essential genes with previous essential gene studies, 364 of the 463 are homologues to essential genes in other species; 339 are shared with more than one other species. Twenty-five genes are known to be essential in P. gingivalis and B. thetaiotaomicron only. Significant enrichment of essential genes within Cluster of Orthologous Groups ‘D’ (cell division), ‘I’ (lipid transport and metabolism) and ‘J’ (translation/ribosome) were identified. Previously, the P. gingivalis core genome was shown to encode 1,476 proteins out of a possible 1,909; 434 of 463 essential genes are contained within the core genome. Thus, for the species P. gingivalis twenty-two, seventy-seven and twenty-three percent of the genome respectively are devoted to essential, core and accessory functions. Conclusions A Mariner transposon system can be adapted to create mutant libraries in P. gingivalis amenable to analysis by next-generation sequencing technologies. In silico analysis of genes essential for in vitro growth demonstrates that although the majority are homologous across bacterial species as a whole, species and strain-specific subsets are apparent. Understanding the putative essential genes of P. gingivalis will provide insights into metabolic pathways and niche adaptations as well as clinical therapeutic strategies.
Collapse
Affiliation(s)
- Brian A Klein
- Department of Molecular Biology and Microbiology, Tufts University Sackler School of Biomedical Sciences, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
1017
|
Kverka M, Tlaskalova-Hogenova H. Two faces of microbiota in inflammatory and autoimmune diseases: triggers and drugs. APMIS 2012; 121:403-21. [DOI: 10.1111/apm.12007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/13/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Miloslav Kverka
- Department of Immunology and Gnotobiology, Institute of Microbiology; Academy of Sciences of the Czech Republic; Prague; Czech Republic
| | - Helena Tlaskalova-Hogenova
- Department of Immunology and Gnotobiology, Institute of Microbiology; Academy of Sciences of the Czech Republic; Prague; Czech Republic
| |
Collapse
|
1018
|
Abe T, Hosur KB, Hajishengallis E, Reis ES, Ricklin D, Lambris JD, Hajishengallis G. Local complement-targeted intervention in periodontitis: proof-of-concept using a C5a receptor (CD88) antagonist. THE JOURNAL OF IMMUNOLOGY 2012; 189:5442-8. [PMID: 23089394 DOI: 10.4049/jimmunol.1202339] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
When excessively activated or deregulated, complement becomes a major link between infection and inflammatory pathology including periodontitis. This oral inflammatory disease is associated with a dysbiotic microbiota, leads to the destruction of bone and other tooth-supporting structures, and exerts an adverse impact on systemic health. We have previously shown that mice deficient either in complement C5a receptor (C5aR; CD88) or TLR2 are highly and similarly resistant to periodontitis, suggesting that a cross-talk between the two receptors may be involved in the disease process. In this paper, we show that C5aR and TLR2 indeed synergize for maximal inflammatory responses in the periodontal tissue and uncover a novel pharmacological target to abrogate periodontitis. Using two different mouse models of periodontitis, we show that local treatments with a C5aR antagonist inhibited periodontal inflammation through downregulation of TNF, IL-1β, IL-6, and IL-17 and further protected against bone loss, regardless of the presence of TLR2. These findings not only reveal a crucial cooperation between C5aR and TLR2 in periodontal inflammation but also provide proof-of-concept for local targeting of C5aR as a powerful candidate for the treatment of human periodontitis.
Collapse
Affiliation(s)
- Toshiharu Abe
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
1019
|
Gursoy UK, Könönen E, Huumonen S, Tervahartiala T, Pussinen PJ, Suominen AL, Sorsa T. Salivary type I collagen degradation end-products and related matrix metalloproteinases in periodontitis. J Clin Periodontol 2012; 40:18-25. [PMID: 23078613 DOI: 10.1111/jcpe.12020] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/29/2012] [Accepted: 09/09/2012] [Indexed: 11/28/2022]
Abstract
AIM Type I collagen degradation end-products and related matrix metalloproteinases (MMPs) were examined aiming to detect potential markers of periodontitis in saliva, with high sensitivity and specificity. MATERIALS AND METHODS The salivary concentrations of MMP-8, MMP-9 and MMP-13, tartrate-resistant acid phosphatase serum type 5b, C-terminal cross-linked telopeptide of type I collagen (CTx), N-terminal cross-linked telopeptide of type I collagen (NTx) and cross-linked carboxyterminal telopeptide of type I collagen were analysed in 230 subjects. Oral health examination included panoramic radiography. RESULTS The concentrations of MMP-8, MMP-9 and MMP-13 in saliva were higher in subjects with generalized periodontitis than in controls. Of the tested salivary markers, MMP-8 was the only marker capable of differentiating subjects with severe alveolar bone loss from those with slight bone loss (p < 0.001). The association between the salivary MMP-8 levels and periodontitis remained significant after the adjustment with age, gender and smoking. In addition, significant correlations were found between the tested markers and periodontal parameters. CONCLUSION Enzymes and end-products of type I collagen degradation have different associations with each other and with periodontal status that may reflect their roles in the cascade leading to alveolar bone loss. MMP-8 is a strong biomarker candidate for detecting alveolar bone destruction.
Collapse
Affiliation(s)
- Ulvi K Gursoy
- Institute of Dentistry, University of Turku, Turku, Finland.
| | | | | | | | | | | | | |
Collapse
|
1020
|
Ghosh SK, Yohannes E, Bebek G, Weinberg A, Jiang B, Willard B, Chance MR, Kinter MT, McCormick TS. Proteomic and bioinformatic profile of primary human oral epithelial cells. J Proteome Res 2012; 11:5492-502. [PMID: 23035736 DOI: 10.1021/pr3007254] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Wounding of the oral mucosa occurs frequently in a highly septic environment. Remarkably, these wounds heal quickly and the oral cavity, for the most part, remains healthy. Deciphering the normal human oral epithelial cell (NHOEC) proteome is critical for understanding the mechanism(s) of protection elicited when the mucosal barrier is intact, as well as when it is breached. Combining 2D gel electrophoresis with shotgun proteomics resulted in identification of 1662 NHOEC proteins. Proteome annotations were performed based on protein classes, molecular functions, disease association and membership in canonical and metabolic signaling pathways. Comparing the NHOEC proteome with a database of innate immunity-relevant interactions (InnateDB) identified 64 common proteins associated with innate immunity. Comparison with published salivary proteomes revealed that 738/1662 NHOEC proteins were common, suggesting that significant numbers of salivary proteins are of epithelial origin. Gene ontology analysis showed similarities in the distributions of NHOEC and saliva proteomes with regard to biological processes, and molecular functions. We also assessed the interindividual variability of the NHOEC proteome and observed it to be comparable with other primary cells. The baseline proteome described in this study should serve as a resource for proteome studies of the oral mucosa, especially in relation to disease processes.
Collapse
Affiliation(s)
- Santosh K Ghosh
- Department of Biological Sciences, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
1021
|
Glycyrrhetinic acid inhibits Porphyromonas gingivalis lipopolysaccharide-induced vascular permeability via the suppression of interleukin-8. Inflamm Res 2012; 62:145-54. [DOI: 10.1007/s00011-012-0560-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 09/18/2012] [Accepted: 09/20/2012] [Indexed: 10/27/2022] Open
|
1022
|
Hajishengallis G, Lamont RJ. Beyond the red complex and into more complexity: the polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology. Mol Oral Microbiol 2012; 27:409-19. [PMID: 23134607 DOI: 10.1111/j.2041-1014.2012.00663.x] [Citation(s) in RCA: 767] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2012] [Indexed: 12/11/2022]
Abstract
Recent advancements in the periodontal research field are consistent with a new model of pathogenesis according to which periodontitis is initiated by a synergistic and dysbiotic microbial community rather than by select 'periopathogens', such as the 'red complex'. In this polymicrobial synergy, different members or specific gene combinations within the community fulfill distinct roles that converge to shape and stabilize a disease-provoking microbiota. One of the core requirements for a potentially pathogenic community to arise involves the capacity of certain species, termed 'keystone pathogens', to modulate the host response in ways that impair immune surveillance and tip the balance from homeostasis to dysbiosis. Keystone pathogens also elevate the virulence of the entire microbial community through interactive communication with accessory pathogens. Other important core functions for pathogenicity require the expression of diverse molecules (e.g. appropriate adhesins, cognate receptors, proteolytic enzymes and proinflammatory surface structures/ligands), which in combination act as community virulence factors to nutritionally sustain a heterotypic, compatible and proinflammatory microbial community that elicits a non-resolving and tissue-destructive host response. On the basis of the fundamental concepts underlying this model of periodontal pathogenesis, that is, polymicrobial synergy and dysbiosis, we term it the PSD model.
Collapse
Affiliation(s)
- G Hajishengallis
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
1023
|
Abstract
Recent studies have highlighted the importance of the human microbiome in health and disease. However, for the most part the mechanisms by which the microbiome mediates disease, or protection from it, remain poorly understood. The keystone-pathogen hypothesis holds that certain low-abundance microbial pathogens can orchestrate inflammatory disease by remodelling a normally benign microbiota into a dysbiotic one. In this Opinion article, we critically assess the available literature that supports this hypothesis, which may provide a novel conceptual basis for the development of targeted diagnostics and treatments for complex dysbiotic diseases.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
1024
|
Fine DH, Sreenivasan PK, McKiernan M, Tischio-Bereski D, Furgang D. Whole mouth antimicrobial effects after oral hygiene: comparison of three dentifrice formulations. J Clin Periodontol 2012; 39:1056-64. [DOI: 10.1111/j.1600-051x.2012.01938.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2012] [Indexed: 12/13/2022]
Affiliation(s)
- Daniel H. Fine
- Department of Oral Biology, University of Medicine and Dentistry of New Jersey; New Jersey Dental School, - UMDNJ; Newark; NJ; USA
| | | | - Marie McKiernan
- Department of Oral Biology, University of Medicine and Dentistry of New Jersey; New Jersey Dental School, - UMDNJ; Newark; NJ; USA
| | - Debra Tischio-Bereski
- Department of Oral Biology, University of Medicine and Dentistry of New Jersey; New Jersey Dental School, - UMDNJ; Newark; NJ; USA
| | - David Furgang
- Department of Oral Biology, University of Medicine and Dentistry of New Jersey; New Jersey Dental School, - UMDNJ; Newark; NJ; USA
| |
Collapse
|
1025
|
Paino A, Lohermaa E, Sormunen R, Tuominen H, Korhonen J, Pöllänen MT, Ihalin R. Interleukin-1β is internalised by viable Aggregatibacter actinomycetemcomitans biofilm and locates to the outer edges of nucleoids. Cytokine 2012; 60:565-74. [PMID: 22898394 DOI: 10.1016/j.cyto.2012.07.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/15/2012] [Accepted: 07/17/2012] [Indexed: 10/28/2022]
Abstract
The opportunistic pathogen Aggregatibacter actinomycetemcomitans causes periodontitis, which is a biofilm infection that destroys tooth-supportive tissues. Interleukin (IL)-1β, a central proinflammatory cytokine of periodontitis, is an essential first line cytokine for local inflammation that modulates the cell proliferation and anti-pathogen response of human gingival keratinocytes. Previously, we demonstrated that A. actinomycetemcomitans biofilms bind IL-1β; however, whether this binding is an active process is not known. In this study, we showed for the first time with immuno-electron microscopy that viable bacterial biofilm cells internalised IL-1β when co-cultured with an organotypic mucosa. Decreased biofilm viability hindered the ability of biofilm to sequester IL-1β and caused IL-1β leakage into the culture medium. In some A. actinomycetemcomitans cells, intracellular IL-1β localized to the outer edges of the nucleoids. We identified the DNA-binding protein HU as an IL-1β interacting protein with mass spectroscopy and showed the interaction of recombinant HU and IL-1βin vitro using enzyme-linked immunosorbent assay (ELISA). Close contact with a viable A. actinomycetemcomitans biofilm decreased the proliferation and apoptosis of human gingival keratinocytes as demonstrated using Ki-67 and the terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) staining, respectively. Our results suggest that viable A. actinomycetemcomitans biofilms may disturb the critical first steps of local inflammation in periodontitis by binding and internalising IL-1β. The interaction of IL-1β with conserved HU provides a potential mechanism for shaping bacterial gene expression.
Collapse
Affiliation(s)
- Annamari Paino
- Department of Biochemistry and Food Chemistry, University of Turku, FI-20014 Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
1026
|
Saiki K, Konishi K. Strategies for targeting the gingipain secretion system of Porphyromonas gingivalis. J Oral Biosci 2012. [DOI: 10.1016/j.job.2012.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
1027
|
Nagano K, Abiko Y, Yoshida Y, Yoshimura F. Porphyromonas gingivalis FimA fimbriae: Roles of the fim gene cluster in the fimbrial assembly and antigenic heterogeneity among fimA genotypes. J Oral Biosci 2012. [DOI: 10.1016/j.job.2012.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
1028
|
Zijnge V, Kieselbach T, Oscarsson J. Proteomics of protein secretion by Aggregatibacter actinomycetemcomitans. PLoS One 2012; 7:e41662. [PMID: 22848560 PMCID: PMC3405016 DOI: 10.1371/journal.pone.0041662] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 06/25/2012] [Indexed: 01/18/2023] Open
Abstract
The extracellular proteome (secretome) of periodontitis-associated bacteria may constitute a major link between periodontitis and systemic diseases. To obtain an overview of the virulence potential of Aggregatibacter actinomycetemcomitans, an oral and systemic human pathogen implicated in aggressive periodontitis, we used a combined LC-MS/MS and bioinformatics approach to characterize the secretome and protein secretion pathways of the rough-colony serotype a strain D7S. LC-MS/MS revealed 179 proteins secreted during biofilm growth. Further to confirming the release of established virulence factors (e.g. cytolethal distending toxin [CDT], and leukotoxin [LtxA]), we identified additional putative virulence determinants in the secretome. These included DegQ, fHbp, LppC, Macrophage infectivity protein (MIP), NlpB, Pcp, PotD, TolB, and TolC. This finding indicates that the number of extracellular virulence-related proteins is much larger than previously demonstrated, which was also supported by in silico analysis of the strain D7S genome. Moreover, our LC-MS/MS and in silico data revealed that at least Type I, II, and V secretion are actively used to excrete proteins directly into the extracellular space, or via two-step pathways involving the Sec/Tat systems for transport across the inner membrane, and outer membrane factors, secretins and auto-transporters, respectively for delivery across the outer membrane. Taken together, our results provide a molecular basis for further elucidating the role of A. actinomycetemcomitans in periodontal and systemic diseases.
Collapse
Affiliation(s)
- Vincent Zijnge
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| | | | - Jan Oscarsson
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
1029
|
Rangé H, Léger T, Huchon C, Ciangura C, Diallo D, Poitou C, Meilhac O, Bouchard P, Chaussain C. Salivary proteome modifications associated with periodontitis in obese patients. J Clin Periodontol 2012; 39:799-806. [PMID: 22780105 DOI: 10.1111/j.1600-051x.2012.01913.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2012] [Indexed: 12/20/2022]
Abstract
AIM To identify changes in the salivary protein/peptide profiles by differential proteomics in obese patients with or without periodontitis. MATERIAL AND METHODS Periodontal examinations and whole saliva samples were obtained from 38 obese patients (mean age: 45.1 ± 7.3 years, mean BMI: 49.3 ± 9 kg/m(2) ) including 13 periodontitis and 25 non-periodontitis subjects, and 19 healthy controls (mean age: 44.2 ± 6.4 years, mean BMI: 21.5 ± 2.1 kg/m(2) ). Surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) mass spectrometry (MS) was used to compare the whole saliva polypeptide profiles. RESULTS The SELDI-TOF-MS analysis detected eight putative markers. Six of them were increased and identified in obese subjects versus controls (albumin, α and β haemoglobin chains, α-defensins 1, 2 and 3). Alpha-defensins were less abundant in saliva of periodontitis obese patients (36.47 ± 19.84 μA) versus non-periodontitis obese patients (43.44 ± 30.34 μA), whereas α-defensins were more abundant in obese patients (40.99 ± 26.66 μA) versus controls (27.1 ± 23.98 μA). CONCLUSIONS Periodontal status modifies the salivary proteome in obese patients. Alpha-defensins may play a role in gingival inflammation, and be involved in the higher susceptibility of obese patients to periodontal diseases.
Collapse
Affiliation(s)
- Hélène Rangé
- Department of Periodontology, Service of Odontology, Garancière Rothschild Hospital, AP-HP, Paris 7-Denis Diderot University, U.F.R. of Odontology, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
1030
|
McIntosh ML, Hajishengallis G. Inhibition of Porphyromonas gingivalis-induced periodontal bone loss by CXCR4 antagonist treatment. Mol Oral Microbiol 2012; 27:449-57. [PMID: 23134610 DOI: 10.1111/j.2041-1014.2012.00657.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Microbial pathogens have evolved mechanisms to proactively manipulate innate immunity, thereby improving their fitness in mammalian hosts. We have previously shown that Porphyromonas gingivalis exploits CXC-chemokine receptor-4 (CXCR4) to instigate a subversive crosstalk with Toll-like receptor 2 that inhibits leukocyte killing of this periodontal pathogen. However, whether CXCR4 plays a role in periodontal disease pathogenesis has not been previously addressed. Here, we hypothesized that CXCR4 is required for P. gingivalis virulence in the periodontium and that treatment with AMD3100, a potent CXCR4 antagonist, would inhibit P. gingivalis-induced periodontitis. Indeed, mice given AMD3100 via osmotic minipumps became resistant to induction of periodontal bone loss following oral inoculation with P. gingivalis. AMD3100 appeared to act in an antimicrobial manner, because mice treated with AMD3100 were protected against P. gingivalis colonization and the associated elevation of the total microbiota counts in the periodontal tissue. Moreover, even when administered 2 weeks after infection, AMD3100 halted the progression of P. gingivalis-induced periodontal bone loss. Therefore, AMD3100 can act in both preventive and therapeutic ways and CXCR4 antagonism could be a promising novel approach to treat human periodontitis.
Collapse
Affiliation(s)
- M L McIntosh
- Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, Louisville, KY, USA
| | | |
Collapse
|
1031
|
Divaris K, Monda K, North K, Olshan A, Lange E, Moss K, Barros S, Beck J, Offenbacher S. Genome-wide association study of periodontal pathogen colonization. J Dent Res 2012; 91:21S-28S. [PMID: 22699663 PMCID: PMC3383103 DOI: 10.1177/0022034512447951] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pathological shifts of the human microbiome are characteristic of many diseases, including chronic periodontitis. To date, there is limited evidence on host genetic risk loci associated with periodontal pathogen colonization. We conducted a genome-wide association (GWA) study among 1,020 white participants of the Atherosclerosis Risk in Communities Study, whose periodontal diagnosis ranged from healthy to severe chronic periodontitis, and for whom "checkerboard" DNA-DNA hybridization quantification of 8 periodontal pathogens was performed. We examined 3 traits: "high red" and "high orange" bacterial complexes, and "high" Aggregatibacter actinomycetemcomitans (Aa) colonization. Genotyping was performed on the Affymetrix 6.0 platform. Imputation to 2.5 million markers was based on HapMap II-CEU, and a multiple-test correction was applied (genome-wide threshold of p < 5 × 10(-8)). We detected no genome-wide significant signals. However, 13 loci, including KCNK1, FBXO38, UHRF2, IL33, RUNX2, TRPS1, CAMTA1, and VAMP3, provided suggestive evidence (p < 5 × 10(-6)) of association. All associations reported for "red" and "orange" complex microbiota, but not for Aa, had the same effect direction in a second sample of 123 African-American participants. None of these polymorphisms was associated with periodontitis diagnosis. Investigations replicating these findings may lead to an improved understanding of the complex nature of host-microbiome interactions that characterizes states of health and disease.
Collapse
Affiliation(s)
- K. Divaris
- Department of Pediatric Dentistry, School of Dentistry, University of North Carolina-Chapel Hill, 228 Brauer Hall, CB#7450, NC 27599, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, NC, USA
| | - K.L. Monda
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, NC, USA
- The Center for Observational Research, Amgen Inc., Thousand Oaks, CA, USA
| | - K.E. North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, NC, USA
- Carolina Center for Genome Sciences, University of North Carolina-Chapel Hill, NC, USA
| | - A.F. Olshan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, NC, USA
| | - E.M. Lange
- Department of Genetics, School of Medicine, University of North Carolina-Chapel Hill, NC, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, NC, USA
| | - K. Moss
- Department of Dental Ecology, School of Dentistry, University of North Carolina-Chapel Hill, NC, USA
| | - S.P. Barros
- Department of Periodontology, School of Dentistry, University of North Carolina-Chapel Hill, NC, USA
| | - J.D. Beck
- Department of Dental Ecology, School of Dentistry, University of North Carolina-Chapel Hill, NC, USA
| | - S. Offenbacher
- Department of Periodontology, School of Dentistry, University of North Carolina-Chapel Hill, NC, USA
| |
Collapse
|
1032
|
Marquis A, Genovese S, Epifano F, Grenier D. The plant coumarins auraptene and lacinartin as potential multifunctional therapeutic agents for treating periodontal disease. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 12:80. [PMID: 22742512 PMCID: PMC3489859 DOI: 10.1186/1472-6882-12-80] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/28/2012] [Indexed: 12/16/2022]
Abstract
BACKGROUND Periodontal diseases are bacterial infections leading to chronic inflammation disorders that are frequently observed in adults. In the present study, we evaluated the effect of auraptene and lacinartin, two natural oxyprenylated coumarins, on the growth, adherence properties, and collagenase activity of Porphyromonas gingivalis. We also investigated the capacity of these compounds to reduce cytokine and matrix metalloproteinase (MMP) secretion by lipopolysaccharide (LPS)-stimulated macrophages and to inhibit MMP-9 activity. METHODS Microplate dilution assays were performed to determine the effect of auraptene and lacinartin on P. gingivalis growth as well as biofilm formation stained with crystal violet. Adhesion of FITC-labeled P. gingivalis to oral epithelial cells was monitored by fluorometry. The effects of auraptene and lacinartin on LPS-induced cytokine and MMP secretion by macrophages were determined by immunological assays. Fluorogenic assays were used to evaluate the capacity of the two coumarins to inhibit the activity of P. gingivalis collagenase and MMP-9. RESULTS Only lacinartin completely inhibited P. gingivalis growth in a complex culture medium. However, under iron-limiting conditions, auraptene and lacinartin both inhibited the growth of P. gingivalis. Lacinartin also inhibited biofilm formation by P. gingivalis and promoted biofilm desorption. Both compounds prevented the adherence of P. gingivalis to oral epithelial cells, dose-dependently reduced the secretion of cytokines (IL-8 and TNF-α) and MMP-8 and MMP-9 by LPS-stimulated macrophages, and inhibited MMP-9 activity. Lacinartin also inhibited P. gingivalis collagenase activity. CONCLUSIONS By acting on multiple targets, including pathogenic bacteria, tissue-destructive enzymes, and the host inflammatory response, auraptene and lacinartin may be promising natural compounds for preventing and treating periodontal diseases.
Collapse
|
1033
|
Ding PH, Wang CY, Darveau RP, Jin L. Porphyromonas gingivalis LPS stimulates the expression of LPS-binding protein in human oral keratinocytes in vitro. Innate Immun 2012; 19:66-75. [PMID: 22736337 DOI: 10.1177/1753425912450348] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
LPS-binding protein (LBP) functions as a crucial molecule in innate immune responses to bacterial challenge. Our study has shown the expression of LBP in human gingiva and its significant association with periodontal health and disease. Porphyromonas gingivalis is a key pathogen of periodontal disease. P. gingivalis LPS as a main virulence factor is strongly involved in periodontal pathogenesis and it displays a significant lipid A structural heterogeneity. Currently, it remains unknown whether, and to what extent, the lipid A structural heterogeneity of P. gingivalis LPS affects LBP expression. The present study investigated the expression profile of LBP in human oral keratinocytes (HOKs) stimulated by two isoforms of P. gingivalis LPS [tetra- (LPS(1435/1449)) and penta-acylated (LPS(1690))] and Escherichia coli LPS, and the involvement of TLRs in LBP expression. The results showed that the expression of LBP mRNA and peptide was significantly up-regulated by P. gingivalis LPS(1690) and E. coli LPS, while P. gingivalis LPS(1435/1449) did not affect LBP expression. Blocking assay and siRNA gene silencing revealed that P. gingivalis LPS(1690)-induced LBP expression was through both TLR2 and TLR4. This in vitro study demonstrates that P. gingivalis LPS with a lipid A structural heterogeneity differentially modulates LBP expression in HOKs.
Collapse
Affiliation(s)
- Pei-Hui Ding
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | |
Collapse
|
1034
|
Al-Hebshi NN, Shamsan AAA, Al-Ak'hali MS. Interleukin-1 Two-Locus Haplotype Is Strongly Associated with Severe Chronic Periodontitis among Yemenis. Mol Biol Int 2012; 2012:231309. [PMID: 22778957 PMCID: PMC3388377 DOI: 10.1155/2012/231309] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 04/30/2012] [Indexed: 12/15/2022] Open
Abstract
Aim. To assess IL-1A C[-889]T and IL-1B C[3954]T genotypes as well as haplotypes in relation to sever chronic periodontitis (SCP) among Yemenis. Materials and Methods. 40 cases with SCP and 40 sex- and age-matched controls were included; all were nonsmokers and free of systemic diseases. Genotyping at each locus was performed using an established PCR-RFLP assay. The Haploview and SimHap software were used to assess data for Hardy-Weinberg's equilibrium (HWE) and linkage disequilibrium (LD) and to obtain subject-level haplotypes. Multiple logistic regression was used to seek for associations in dominant, additive, and recessive models. Results. Mean plaque index (MPI) showed the strongest association with SCP (OR = 16). A significant LD was observed in the cases (D' = 0.80 and r(2) = 0.47). The genotype at each locus showed significant association with SCP in the recessive model (TT versus TC + CC) even after adjustment for MPI (OR = 6.29 & 461, resp.). The C-T haplotype conferred protection against SCP in a dominant manner (OR = 0.16). On the other hand, the T-T haplotype in double dose (recessive model) showed strong association with CP (OR = 15.6). Conclusions. IL-1 two-locus haplotype is associated with SCP in Yemenis. Haplotype-based analysis may be more suited for use in genetic association studies of periodontitis.
Collapse
Affiliation(s)
- Nezar Noor Al-Hebshi
- Molecular Research Laboratory, Faculty of Medical Sciences, University of Science and Technology, Sana'a, Yemen
| | | | | |
Collapse
|
1035
|
Kukita A, Ichigi Y, Takigawa I, Watanabe T, Kukita T, Miyamoto H. Infection of RANKL-primed RAW-D macrophages with Porphyromonas gingivalis promotes osteoclastogenesis in a TNF-α-independent manner. PLoS One 2012; 7:e38500. [PMID: 22723864 PMCID: PMC3377672 DOI: 10.1371/journal.pone.0038500] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/10/2012] [Indexed: 11/19/2022] Open
Abstract
Infection of macrophages with bacteria induces the production of pro-inflammatory cytokines including TNF-α. TNF-α directly stimulates osteoclast differentiation from bone marrow macrophages in vitro as well as indirectly via osteoblasts. Recently, it was reported that bacterial components such as LPS inhibited RANKL-induced osteoclastogenesis in early stages, but promoted osteoclast differentiation in late stages. However, the contribution to osteoclast differentiation of TNF-α produced by infected macrophages remains unclear. We show here that Porphyromonas gingivalis, one of the major pathogens in periodontitis, directly promotes osteoclastogenesis from RANKL-primed RAW-D (subclone of RAW264) mouse macrophages, and we show that TNF-α is not involved in the stimulatory effect on osteoclastogenesis. P. gingivalis infection of RANKL-primed RAW-D macrophages markedly stimulated osteoclastogenesis in a RANKL-independent manner. In the presence of the TLR4 inhibitor, polymyxin B, infection of RANKL-primed RAW-D cells with P. gingivalis also induced osteoclastogenesis, indicating that TLR4 is not involved. Infection of RAW-D cells with P. gingivalis stimulated the production of TNF-α, whereas the production of TNF-α by similarly infected RANKL-primed RAW-D cells was markedly down-regulated. In addition, infection of RANKL-primed macrophages with P. gingivalis induced osteoclastogenesis in the presence of neutralizing antibody against TNF-α. Inhibitors of NFATc1 and p38MAPK, but not of NF-κB signaling, significantly suppressed P. gingivalis-induced osteoclastogenesis from RANKL-primed macrophages. Moreover, re-treatment of RANKL-primed macrophages with RANKL stimulated osteoclastogenesis in the presence or absence of P. gingivalis infection, whereas re-treatment of RANKL-primed macrophages with TNF-α did not enhance osteoclastogenesis in the presence of live P. gingivalis. Thus, P. gingivalis infection of RANKL-primed macrophages promoted osteoclastogenesis in a TNF-α independent manner, and RANKL but not TNF-α was effective in inducing osteoclastogenesis from RANKL-primed RAW-D cells in the presence of P. gingivalis.
Collapse
Affiliation(s)
- Akiko Kukita
- Department of Microbiology, Faculty of Medicine, Saga University, Saga, Japan.
| | | | | | | | | | | |
Collapse
|
1036
|
Saha S, Tomaro-Duchesneau C, Tabrizian M, Prakash S. Probiotics as oral health biotherapeutics. Expert Opin Biol Ther 2012; 12:1207-20. [PMID: 22690730 DOI: 10.1517/14712598.2012.693474] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Oral health is affected by its resident microorganisms. Three prominent oral disorders are dental caries, gingivitis and periodontitis, with the oral microbiota playing a key role in the initiation/progression of all three. Understanding the microbiota and the diseases they may cause is critical to the development of new therapeutics. This review is focused on probiotics for the prevention and/or treatment of oral diseases. AREAS COVERED This review describes the oral ecosystem and its correlation with oral health/disease. The pathogenesis and current prevention/treatment strategies of periodontal diseases (PD) and dental caries (DC) are depicted. An introduction of probiotics is followed by an analysis of their role in PD and DC, and their potential role(s) in oral health. Finally, a discussion ensues on the future research directions and limitations of probiotics for oral health. EXPERT OPINION An effective oral probiotic formulation should contribute to the prevention/treatment of microbial diseases of the oral cavity. Understanding the oral microbiota's role in oral disease is important for the development of a therapeutic to prevent/treat dental diseases. However, investigations into clinical efficacy, delivery/dose optimization, mechanism(s) of action and other related parameters are yet to be fully explored. Keeping this in mind, investigations into oral probiotic therapies are proving promising.
Collapse
Affiliation(s)
- Shyamali Saha
- McGill University, Physiology and Artificial Cells and Organs Research Centre, Departments of Biomedical Engineering, Biomedical Technology and Cell Therapy, Research Laboratory, Faculty of Medicine, 3775 University Street, Montreal, Quebec, H3A 2B4, Canada
| | | | | | | |
Collapse
|
1037
|
Liu B, Faller LL, Klitgord N, Mazumdar V, Ghodsi M, Sommer DD, Gibbons TR, Treangen TJ, Chang YC, Li S, Stine OC, Hasturk H, Kasif S, Segrè D, Pop M, Amar S. Deep sequencing of the oral microbiome reveals signatures of periodontal disease. PLoS One 2012; 7:e37919. [PMID: 22675498 PMCID: PMC3366996 DOI: 10.1371/journal.pone.0037919] [Citation(s) in RCA: 269] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 04/30/2012] [Indexed: 11/18/2022] Open
Abstract
The oral microbiome, the complex ecosystem of microbes inhabiting the human mouth, harbors several thousands of bacterial types. The proliferation of pathogenic bacteria within the mouth gives rise to periodontitis, an inflammatory disease known to also constitute a risk factor for cardiovascular disease. While much is known about individual species associated with pathogenesis, the system-level mechanisms underlying the transition from health to disease are still poorly understood. Through the sequencing of the 16S rRNA gene and of whole community DNA we provide a glimpse at the global genetic, metabolic, and ecological changes associated with periodontitis in 15 subgingival plaque samples, four from each of two periodontitis patients, and the remaining samples from three healthy individuals. We also demonstrate the power of whole-metagenome sequencing approaches in characterizing the genomes of key players in the oral microbiome, including an unculturable TM7 organism. We reveal the disease microbiome to be enriched in virulence factors, and adapted to a parasitic lifestyle that takes advantage of the disrupted host homeostasis. Furthermore, diseased samples share a common structure that was not found in completely healthy samples, suggesting that the disease state may occupy a narrow region within the space of possible configurations of the oral microbiome. Our pilot study demonstrates the power of high-throughput sequencing as a tool for understanding the role of the oral microbiome in periodontal disease. Despite a modest level of sequencing (~2 lanes Illumina 76 bp PE) and high human DNA contamination (up to ~90%) we were able to partially reconstruct several oral microbes and to preliminarily characterize some systems-level differences between the healthy and diseased oral microbiomes.
Collapse
Affiliation(s)
- Bo Liu
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
- Department of Computer Science, University of Maryland, College Park, Maryland, United States of America
| | - Lina L. Faller
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Niels Klitgord
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Varun Mazumdar
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Mohammad Ghodsi
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
- Department of Computer Science, University of Maryland, College Park, Maryland, United States of America
| | - Daniel D. Sommer
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Theodore R. Gibbons
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
- Biological Sciences Graduate Program, University of Maryland, College Park, Maryland, United States of America
| | - Todd J. Treangen
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
- The McKusick-Nathans Institute for Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yi-Chien Chang
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Shan Li
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - O. Colin Stine
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Hatice Hasturk
- The Forysth Institute, Department of Periodontology, Cambridge, Massachusetts, United States of America
| | - Simon Kasif
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- Children’s Informatics Program, Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Boston, Massachusetts, United States of America
| | - Daniel Segrè
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- Department of Biology, Boston University, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Mihai Pop
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
- Department of Computer Science, University of Maryland, College Park, Maryland, United States of America
- Biological Sciences Graduate Program, University of Maryland, College Park, Maryland, United States of America
| | - Salomon Amar
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- Center for Anti-Inflammatory Therapeutics; Boston University Goldman School of Dental Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
1038
|
Coutinho-Silva R, Ojcius DM. Role of extracellular nucleotides in the immune response against intracellular bacteria and protozoan parasites. Microbes Infect 2012; 14:1271-7. [PMID: 22634346 DOI: 10.1016/j.micinf.2012.05.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/29/2012] [Accepted: 05/14/2012] [Indexed: 02/06/2023]
Abstract
Extracellular nucleotides are danger signals involved in recognition and control of intracellular pathogens. They are an important component of the innate immune response against intracellular pathogens, inducing the recruitment of inflammatory cells, stimulating secretion of cytokines, and producing inflammatory mediators such as reactive oxygen species (ROS) and nitric oxide (NO). In the case of extracellular ATP, some of the immune responses are mediated through activation of the NLRP3 inflammasome and secretion of the cytokine, interleukin-1β (IL-1β), through a mechanism dependent on ligation of the P2X7 receptor. Here we review the role of extracellular nucleotides as sensors of intracellular bacteria and protozoan parasites, and discuss how these pathogens manipulate purinergic signaling to diminish the immune response against infection.
Collapse
Affiliation(s)
- Robson Coutinho-Silva
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ 21941-902, Brazil.
| | | |
Collapse
|
1039
|
Bougas K, Ransjö M, Johansson A. Effects of Porphyromonas gingivalis surface-associated material on osteoclast formation. Odontology 2012; 101:140-9. [DOI: 10.1007/s10266-012-0068-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/03/2012] [Indexed: 10/28/2022]
|
1040
|
Bostanci N, Belibasakis GN. Doxycycline inhibits TREM-1 induction by Porphyromonas gingivalis. ACTA ACUST UNITED AC 2012; 66:37-44. [DOI: 10.1111/j.1574-695x.2012.00982.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 03/12/2012] [Accepted: 04/25/2012] [Indexed: 12/15/2022]
Affiliation(s)
- Nagihan Bostanci
- Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich; Switzerland
| | - Georgios N. Belibasakis
- Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich; Switzerland
| |
Collapse
|
1041
|
Glurich I, Acharya A, Shukla SK, Nycz GR, Brilliant MH. The oral-systemic personalized medicine model at Marshfield Clinic. Oral Dis 2012; 19:1-17. [PMID: 22458294 DOI: 10.1111/j.1601-0825.2012.01921.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Periodontal disease and diabetes, two diseases that have achieved epidemic status, share a bidirectional relationship driven by micro-inflammatory processes. The present review frames the current understanding of the pathological processes that appear to link these diseases and advances the hypothesis that reversal of the epidemic is possible through application of interdisciplinary intervention and advancement of oral-systemic personalized medicine. An overview of how Marshfield Clinic's unique clinical, informatics and bio-repository resources and infrastructures are being aligned to advance oral-systemic personalized medicine is presented as an interventional model with the potential to reverse the epidemic trends seen for these two chronic diseases over the past several decades. The overall vision is to engineer a transformational shift in paradigm from 'personalized medicine' to 'personalized health'.
Collapse
Affiliation(s)
- I Glurich
- Office of Scientific Writing and Publication, Marshfield, WI 54449, USA
| | | | | | | | | |
Collapse
|
1042
|
The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss. Nat Immunol 2012; 13:465-73. [PMID: 22447028 DOI: 10.1038/ni.2260] [Citation(s) in RCA: 333] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/10/2012] [Indexed: 12/14/2022]
Abstract
Aging is linked to greater susceptibility to chronic inflammatory diseases, several of which, including periodontitis, involve neutrophil-mediated tissue injury. Here we found that aging-associated periodontitis was accompanied by lower expression of Del-1, an endogenous inhibitor of neutrophil adhesion dependent on the integrin LFA-1, and by reciprocal higher expression of interleukin 17 (IL-17). Consistent with that, IL-17 inhibited gingival endothelial cell expression of Del-1, thereby promoting LFA-1-dependent recruitment of neutrophils. Young Del-1-deficient mice developed spontaneous periodontitis that featured excessive neutrophil infiltration and IL-17 expression; disease was prevented in mice doubly deficient in Del-1 and LFA-1 or in Del-1 and the IL-17 receptor. Locally administered Del-1 inhibited IL-17 production, neutrophil accumulation and bone loss. Therefore, Del-1 suppressed LFA-1-dependent recruitment of neutrophils and IL-17-triggered inflammatory pathology and may thus be a promising therapeutic agent for inflammatory diseases.
Collapse
|
1043
|
Honda K. Porphyromonas gingivalis sinks teeth into the oral microbiota and periodontal disease. Cell Host Microbe 2012; 10:423-5. [PMID: 22100158 DOI: 10.1016/j.chom.2011.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Periodontitis is linked to polymicrobial interactions and the presence of Porphyromonas gingivalis. In this issue of Cell Host & Microbe, Hajishengallis et al. (2011) demonstrate that P. gingivalis colonization in the oral cavity changes the composition of the oral commensal microbiota and accelerates microbiota-mediated bone-destructive periodontitis, indicating that this single, low-abundance species is a keystone in periodontal disease.
Collapse
Affiliation(s)
- Kenya Honda
- Department of Immunology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan.
| |
Collapse
|
1044
|
Nelson A, De Soyza A, Perry JD, Sutcliffe IC, Cummings SP. Polymicrobial challenges to Koch's postulates: ecological lessons from the bacterial vaginosis and cystic fibrosis microbiomes. Innate Immun 2012; 18:774-83. [PMID: 22377802 DOI: 10.1177/1753425912439910] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Koch's postulates have shaped our understanding of infectious diseases; however, one of the tangential consequences of them has been the emergence of a predominantly monomicrobial perspective concerning disease aetiology. This orthodoxy has been undermined by the growing recognition that some important infectious diseases have a polymicrobial aetiology. A significant new development in our understanding of polymicrobial infections is the recognition that they represent functional ecosystems and that to understand such systems and the outcome and impact of therapeutic interventions requires an understanding of how these communities arise and develop. Therefore, it is timely to explore what we can learn from other fields. In particular, ecological theory may inform our understanding of how polymicrobial communities assemble their structure and their dynamics over time. Such work may also offer insights into how such communities move from stable to unstable states, as well as the role of invasive pathogens in the progression of the disease. Ecological theory offers a theoretical framework around which testable hypotheses can be developed to clarify the polymicrobial nature and dynamics of such infections in the face of environmental change and therapeutic interventions.
Collapse
Affiliation(s)
- Andrew Nelson
- School of Applied Sciences, University of Northumbria, Newcastle-upon-Tyne, UK
| | | | | | | | | |
Collapse
|
1045
|
Miller DP, Bell JK, McDowell JV, Conrad DH, Burgner JW, Héroux A, Marconi RT. Structure of factor H-binding protein B (FhbB) of the periopathogen, Treponema denticola: insights into progression of periodontal disease. J Biol Chem 2012; 287:12715-22. [PMID: 22371503 DOI: 10.1074/jbc.m112.339721] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Periodontitis is the most common disease of microbial etiology in humans. Periopathogen survival is dependent upon evasion of complement-mediated destruction. Treponema denticola, an important contributor to periodontitis, evades killing by the alternative complement cascade by binding factor H (FH) to its surface. Bound FH is rapidly cleaved by the T. denticola protease, dentilisin. In this report, the structure of the T. denticola FH-binding protein, FhbB, was solved to 1.7 Å resolution. FhbB possesses a unique fold that imparts high thermostability. The kinetics of the FH/FhbB interaction were assessed using surface plasmon resonance. A K(D) value in the micromolar range (low affinity) was demonstrated, and rapid off kinetics were observed. Site-directed mutagenesis and sucrose octasulfate competition assays collectively indicate that the negatively charged face of FhbB binds within FH complement control protein module 7. This study provides significant new insight into the molecular basis of FH/FhbB interaction and advances our understanding of the role that T. denticola plays in the development and progression of periodontal disease.
Collapse
Affiliation(s)
- Daniel P Miller
- Department of Microbiology and Immunology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | | | | | | | |
Collapse
|
1046
|
Abstract
The oral microbial community represents the best-characterized consortium associated with the human host. There are strong correlations between the qualitative composition of the oral microbiota and clinically healthy or diseased states. However, additional studies are needed to elucidate the mechanisms that define these microbial/host relationships.
Collapse
|
1047
|
Kuramoto A, Yoshinaga Y, Kaneko T, Ukai T, Shiraishi C, Oshino K, Ichimura I, Hara Y. The formation of immune complexes is involved in the acute phase of periodontal destruction in rats. J Periodontal Res 2012; 47:455-62. [PMID: 22283745 DOI: 10.1111/j.1600-0765.2011.01453.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Loss of clinical attachment and alveolar bone destruction are major symptoms of periodontitis, caused by not only the destructive effect of periodontopathic bacteria but also the overactive response of the host immune system against periodontal pathogens. The details of the participation of the immune system in the onset and progression of periodontitis are unclear. In this study, we attempted to determine whether the host immune system, and in particular the formation of immune complexes, is involved in the periodontal destruction. MATERIAL AND METHODS We applied ovalbumin or lipopolysaccharide (LPS) as antigens and their specific immunoglobulin G (IgG) antibodies purified from rat serum to rat gingival sulcus alternately. Loss of attachment, alveolar bone destruction and the numbers of inflammatory cells infiltrating the periodontal tissue and osteoclasts on the alveolar bone surface were investigated histometrically. The formation of immune complex was confirmed by immunohistological staining of complement C1qB. RESULTS Loss of attachment and the presence of C1qB were observed histopathologically in both experimental groups. The group that had been treated with LPS and anti-LPS IgG showed greater loss of attachment. The number of inflammatory cells in the periodontal tissue was increased in both experimental groups, while osteoclasts at the alveolar bone crest were observed only in the group that had been treated with LPS and anti-LPS IgG. CONCLUSION In the present study, we showed that the formation of immune complex appears to be involved in the acute phase of periodontal destruction and that the biological activity of antigens is also important.
Collapse
Affiliation(s)
- A Kuramoto
- Department of Periodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
1048
|
Giannobile WV, McDevitt JT, Niedbala RS, Malamud D. Translational and clinical applications of salivary diagnostics. Adv Dent Res 2012; 23:375-80. [PMID: 21917748 DOI: 10.1177/0022034511420434] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
There have been significant advances in techniques for the detection of biomarker signals in the oral cavity (e.g., ELISAs for proteins, PCR for RNA and DNA) as well as the engineering and development of microfluidic approaches to make oral-based point-of-care (POC) methods for the diagnosis for both local and systemic conditions a reality. In this section, we focus on three such approaches, namely, periodontal disease management, early markers for systemic diseases, and salivary markers useful for pharmacogenomic studies. Novel approaches using non-invasive, salivary samples and user-friendly devices offer results that are as sensitive and specific as laboratory-based analyses using blood or urine.
Collapse
Affiliation(s)
- W V Giannobile
- Department of Periodontics, Michigan Center for Oral Health Research, University of Michigan School of Dentistry, Ann Arbor, USA
| | | | | | | |
Collapse
|
1049
|
Development of a modified gentamicin resistance cassette for genetic manipulation of the oral spirochete Treponema denticola. Appl Environ Microbiol 2012; 78:2059-62. [PMID: 22247130 DOI: 10.1128/aem.07461-11] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Herein, we report that a modified gentamicin cassette and a PCR-based method can be used for targeted mutagenesis of the oral spirochete Treponema denticola. This approach minimizes polar effects and spontaneous antibiotic resistance. Therefore, it can serve as a reliable tool for genetic manipulation of T. denticola.
Collapse
|
1050
|
Pathogenic microbes and community service through manipulation of innate immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 946:69-85. [PMID: 21948363 DOI: 10.1007/978-1-4614-0106-3_5] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The periodontal pathogen Porphyromonas gingivalis undermines major components of innate immunity, such as complement, Toll-like receptors (TLR), and their crosstalk pathways. At least in principle, these subversive activities could promote the adaptive fitness of the entire periodontal biofilm community. In this regard, the virulence factors responsible for complement and TLR exploitation (gingipain enzymes, atypical lipopolysaccharide molecules, and fimbriae) are released as components of readily diffusible membrane vesicles, which can thus become available to other biofilm organisms. This review summarizes important immune subversive tactics of P. gingivalis which might enable it to exert a supportive impact on the oral microbial community.
Collapse
|