1
|
Cherian C, Jannet Vennila J, Sharan L. Marine bromophenols as an effective inhibitor of virulent proteins (peptidyl arginine deiminase, gingipain R and hemagglutinin A) in Porphyromas gingivalis. Arch Oral Biol 2019; 100:119-128. [PMID: 30826505 DOI: 10.1016/j.archoralbio.2019.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Porphyromonas gingivalis, is one of the major oral pathogen that produce virulent proteins which mediate periodontal tissue inflammation and infection. Marine algae have recently gained popularity for its bioactive molecules and their oral applications. Marine bromophenols (MBs) is abundant in red algae which are reported to have wide medicinal properties. The current research primarily focuses to elucidate the bioactivity of MBs against the virulent proteins produced by P. gingivalis. MATERIALS AND METHODS Potent MBs which effectively binds and inhibits the virulent proteins peptidyl arginine deiminase (PPAD), gingipain R (Rgp) and hemagglutinin A (HgA) was identified through molecular docking and molecular simulation approach. MBs were extracted from Kappaphycus alvarezii (KAB), marine red algae found in India. The efficacy of this MB was studied against P. gingivalis by employing antibacterial activity assays, gingipain assay, hemagglutination inhibition assay (HIA) and mRNA expression analysis (q RT PCR). RESULTS MBs with benzene, methyl and glycosyl substitutions demonstrated significant docking score, with good stability and pharmacokinetic properties. In addition to the antibacterial activities against P. gingivalis, KAB was also found to inhibit the gingipain and hemagglutination activities. Exposure of KAB to the virulent genes in P. gingivalis resulted in low mRNA levels of these genes, which suggested the down regulation functions induced by the MBs. CONCLUSION Biochemical investigations revealed that KAB is a potent natural metabolite that can inhibit and control the virulent proteins produced by P. gingivalis. This study recommends future research to direct towards applicability of MBs in commercial dental products.
Collapse
Affiliation(s)
| | | | - Leena Sharan
- Karunya Institute of Technology & Sciences, India
| |
Collapse
|
2
|
Belibasakis G, Thurnheer T, Bostanci N. Porphyromonas gingivalis: a heartful oral pathogen? Virulence 2014; 5:463-4. [PMID: 24759693 PMCID: PMC4063808 DOI: 10.4161/viru.28930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/16/2014] [Indexed: 12/17/2022] Open
Affiliation(s)
- Georgios Belibasakis
- Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich, Switzerland
| | - Thomas Thurnheer
- Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich, Switzerland
| | - Nagihan Bostanci
- Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich, Switzerland
| |
Collapse
|
3
|
Nagano K. FimA Fimbriae of the Periodontal Disease-associated Bacterium Porphyromonas gingivalis. YAKUGAKU ZASSHI 2013; 133:963-74. [DOI: 10.1248/yakushi.13-00177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Keiji Nagano
- Department of Microbiology, School of Dentistry, Aichi Gakuin University
| |
Collapse
|
4
|
Sojar HT, Smith DF. Porphyromonas gingivalis fimbriae carbohydrate specificity assessment by glycomics. ACTA ACUST UNITED AC 2012; 66:83-7. [PMID: 22577836 DOI: 10.1111/j.1574-695x.2012.00989.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/01/2012] [Accepted: 05/07/2012] [Indexed: 11/26/2022]
Abstract
Porphyromonas gingivalis is a major pathogen in adult periodontitis. Fimbriae play an important role in the initial interaction between the bacteria and the host. Our earlier studies suggested that the oligosaccharide moiety of lactoferrin is involved in the interaction with fimbriae. Porphyromonas gingivalis fimbriae bound strongly to albumin-fucosylamide (albumin-1-amido-1-deoxy-L-fucose) and to a lesser extent to albumin-N-acetyl-D-galactosamine (albumin-p-aminophenyl-N-acetyl-β-D-galactosaminide, but failed to bind bovine serum albumin. In this study we explored the glycan array to determine the carbohydrate-binding specificity of P. gingivalis fimbriae. Purified fimbriae bind to glycans which have a Lewis(x), Galβ1-4(Fucα1-3) GlcNAcβ structure in common. Interestingly, all glycans have a terminal fucose moiety and if fucose is removed, the fimbriae fail to bind. This is the first study that suggests that fucose is important for P. gingivalis fimbriae binding.
Collapse
Affiliation(s)
- Hakimuddin T Sojar
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, USA.
| | | |
Collapse
|
5
|
Yamagishi VTK, Torneck CD, Friedman S, Huang GTJ, Glogauer M. Blockade of TLR2 inhibits Porphyromonas gingivalis suppression of mineralized matrix formation by human dental pulp stem cells. J Endod 2011; 37:812-8. [PMID: 21787495 DOI: 10.1016/j.joen.2011.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 03/17/2011] [Accepted: 03/18/2011] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Human dental pulp stem/progenitor cells (hDPSC) can differentiate into odontoblast-like cells and express dentin sialophosphoprotein (DSPP) and osteocalcin (OCN); thus, they may be used to regenerate dentin. However, residual bacterial components in the root canal may suppress this activity. PURPOSE This study investigated the effect of a Porphyromonas gingivalis component on the expression of DSPP and OCN by stimulated hDPSCs and the influence of blockade of TLR2-mediated P. gingivalis host recognition. METHODS Stimulated hDPSCs were exposed to varying concentrations of P. gingivalis lipopolysaccharide (LPS), and the expression of DSPP and OCN was measured. Similar groups of stimulated hDPSCs were exposed to TLR2 blocking agents before exposure to LPS. RESULTS hDPSCs exposed to 5, 10, and 20 μg/mL LPS exhibited a dose-dependent reduction in the expression of DSPP (3.19 ± 0.18, 2.60 ± 0.49, and 1.15 ± 0.29, respectively) and OCN (3.51 ± 1.18, 2.60 ± 0.67 and 1.66 ± 0.89, respectively). The expression of DSPP and OCN after exposure to 20 μg/mL of LPS was significantly lower than measured for unexposed stimulated cells (analysis of variance and post hoc Tukey test, P < .05). The blockade of TLR2 using an extra- and intracellular agent affected DSPP (4.67 ± 0.97 and 5.29 ± 1.66, respectively) and OCN (5.25 ± 1.69 and 5.82 ± 2.38, respectively) expression at levels comparable to stimulated cells unexposed to 20 μg/mL LPS (6.32 ± 2.47 and 4.70 ± 1.60 for DSPP and OCN, respectively). CONCLUSIONS The suppressing effect of P. gingivalis on mineralized matrix formation by hDPSCs is confirmed, and this suppression can be moderated by TLR2 blockade.
Collapse
|
6
|
Zhou J, Zhang J, Chao J. Porphyromonas gingivalis promotes monocyte migration by activating MMP-9. J Periodontal Res 2011; 47:236-42. [PMID: 22035412 DOI: 10.1111/j.1600-0765.2011.01427.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE The migration of monocytes into the local environment is crucial for their maturation into macrophages or osteoclasts in the pathogenesis of periodontal disease. The objective of this study was to investigate the role and mechanisms mediated by Porphyromonas gingivalis in promoting the migration of monocytes by regulating MMP-9 and TIMP-1 expression. MATERIAL AND METHODS Human THP1 monocytes were treated with culture supernatant derived from P. gingivalis (ATCC 33277) for 24 h. Zymography, western blot analysis and quantitative PCRs were performed to analyse protein and mRNA levels of MMP-9. Protein and mRNA levels of TIMP-1 from monocytes treated with or without P. gingivalis were determined as well. Transwell migration assay was carried out to analyse the effect of P. gingivalis on the migration of human peripheral blood CD14-positive monocytes. An MMP inhibitor (GM6001) and a proteinase inhibitor (leupeptin) were used to determine the role of MMP-9 in P. gingivalis supernatant- and lipopolysaccharide-induced monocyte migration. RESULTS In zymography and western blot, an 82 kDa band of active MMP-9 emerged in P. gingivalis-treated monocyte culture media in a dose-dependent manner, in addition to the MMP-9 proenzyme (92 kDa) band expressed in control cell culture media. P. gingivalis supernatant increased both the protein and the mRNA levels of MMP-9 and TIMP-1. P. gingivalis supernatant, but not its lipopolysaccharide, increased the migratory ability of CD14-positive monocytes. The increased migratory ability of P. gingivalis-treated monocytes was partly inhibited by leupeptin (200 μg/mL) and completely antagonized by the MMP inhibitor GM6001 (100 nm). Lipopolysaccharide of P. gingivalis increased protein and mRNA levels of MMP-9 in monocytes, but had no effect on the migratory ability or MMP-9 activation. CONCLUSION P. gingivalis supernatant increased the migratory ability of monocytes, in part, by increasing activation and expression of MMP-9.
Collapse
Affiliation(s)
- J Zhou
- Department of Pediatric Dentistry and Orthodontics, Medical University of South Carolina, Charleston, SC 29401, USA.
| | | | | |
Collapse
|
7
|
|
8
|
Sakai E, Naito M, Sato K, Hotokezaka H, Kadowaki T, Kamaguchi A, Yamamoto K, Okamoto K, Nakayama K. Construction of recombinant hemagglutinin derived from the gingipain-encoding gene of Porphyromonas gingivalis, identification of its target protein on erythrocytes, and inhibition of hemagglutination by an interdomain regional peptide. J Bacteriol 2007; 189:3977-86. [PMID: 17384191 PMCID: PMC1913415 DOI: 10.1128/jb.01691-06] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gingivalis, an anaerobic gram-negative bacterium associated with chronic periodontitis, can agglutinate human erythrocytes. In general, hemagglutination can be considered the ability to adhere to host cells; however, P. gingivalis-mediated hemagglutination has special significance because heme markedly accelerates growth of this bacterium. Although a number of studies have indicated that a major hemagglutinin of P. gingivalis is intragenically encoded by rgpA, kgp, and hagA, direct evidence has not been obtained. We demonstrated in this study that recombinant HGP44(720-1081), a fully processed HGP44 domain protein, had hemagglutinating activity but that an unprocessed form, HGP44(720-1138), did not. A peptide corresponding to residues 1083 to 1102, which was included in HGP44(720-1138) but not in HGP44(720-1081), could bind HGP44(720-1081) in a dose-dependent manner and effectively inhibited HGP44(720-1081)-mediated hemagglutination, indicating that the interdomain regional amino acid sequence may function as an intramolecular suppressor of hemagglutinating activity. Analyses by solid-phase binding and chemical cross-linking suggested that HGP44 interacted with glycophorin A on the erythrocyte membrane. Glycophorin A and, more effectively, asialoglycophorin, which were added exogenously, inhibited HGP44(720-1081)-mediated hemagglutination. Treatment of erythrocytes with RgpB proteinase resulted in degradation of glycophorin A on the membrane and a decrease in HGP44(720-1081)-mediated hemagglutination. Surface plasmon resonance detection analysis revealed that HGP44(720-1081) could bind to asialoglycophorin with a dissociation constant of 3.0 x 10(-7) M. These results indicate that the target of HGP44 on the erythrocyte membrane appears to be glycophorin A.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Sakamoto 1-7-1, Nagasaki 852-8588, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Sahingur SE, Boehm TK, Sojar HT, Sharma A, De Nardin E. Fibrinogen-neutrophil interactions in response to fMLP and Porphyromonas gingivalis fimbrial peptides. Immunol Invest 2006; 35:63-74. [PMID: 16531330 DOI: 10.1080/08820130500496811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Porphyromonas gingivalis (P.g) is the primary bacterial agent in many forms of chronic periodontitis. Since polymorphonuclear leukocytes (PMNs) are first-line responders to P.g.- induced inflammation, and fibrinogen is important for in vivo PMN in this disease, we have studied the effect of N-formyl-methionyl-leucyl-phenylalanine (fMLP) (an inflammatory stimulus), P.g. fimbriae and fimbrial peptides (based on FimA, the main structural protein of P.g. fimbriae) on PMN-fibrinogen interactions. Freshly isolated human PMNs were allowed to react with FITC-Fibrinogen and various fimbrial peptides (denoted as FimA followed by amino acid number within whole FimA protein), and FITC-Fibrinogen binding was measured using flow cytometry. Freshly isolated neutrophils were also challenged with Fibrinogen and/or fimbrial peptides to measure IL-8 secretion using ELISA. Our studies show that fibrinogen binding to PMNs is enhanced (p < 0.01) in response to fMLP as well as fimbrial peptides (FimA 61-80) containing the motif LTTE (p < 0.01) in a dose dependent manner but not in response to peptides without that motif. We also observed that fMLP and FimA 61-80 have an additive effect on fibrinogen binding to PMNs (p < 0.05), and fMLP and FimA 171-185 significantly inhibit fMLP-induced fibrinogen binding (p < 0.01). To determine of the role of inflammatory cytokines, we examined IL-8 release from PMNs in response to combinations of P. gingivalis fimbriae, fMLP and fibrinogen. In all cases, IL-8 release increased in a dose-dependent manner (p < 0.05). fMLP-fibrinogen effect on IL-8 release from PMNs was synergistic while fimbriae-fibrinogen effect was additive. In summary, PMN priming by fimbrial peptides facilitates fibrinogen-PMN interaction and may increase inflammation.
Collapse
Affiliation(s)
- S E Sahingur
- Department of Oral Biology, University at Buffalo, Buffalo, New York 14214, USA
| | | | | | | | | |
Collapse
|
10
|
Madianos PN, Bobetsis YA, Kinane DF. Generation of inflammatory stimuli: how bacteria set up inflammatory responses in the gingiva. J Clin Periodontol 2005; 32 Suppl 6:57-71. [PMID: 16128830 DOI: 10.1111/j.1600-051x.2005.00821.x] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES The primary aetiologic factor of periodontal disease is the bacterial biofilm. Gram-positive and gram-negative bacteria possess a plethora of structural or secreted components that may cause direct destruction to periodontal tissues or stimulate host cells to activate a wide range of inflammatory responses. These responses are intended to eliminate the microbial challenge, but may often cause further tissue damage. METHODS This review has been divided into three parts: (a) bacterial virulence factors, which includes basic information on bacterial virulence factors, and the principle inflammatory responses that host cells elicit against these factors, (b) main receptors and signalling pathways, which includes basic information about the main receptors that interact with the bacterial virulence factors, the nature of these interactions, and the activated signalling pathways that lead to inflammatory responses, and (c) initiation of inflammation, which includes a model by which the virulence factors may interact with host cells and lead to inflammatory responses in the gingiva. FINDINGS AND CONCLUSIONS Bacterial components/virulence factors may be involved in modulating inflammatory responses and include: lipopolysaccharides (LPS), peptidoglycans, lipotechoic acids, fimbriae, proteases, heat-shock proteins, formyl-methionyl peptides, and toxins. Potential host cell receptors involved in recognizing bacterial components and initiating signalling pathways that lead to inflammatory responses include: Toll-like receptors (TLRs), CD14, nucleotide-binding oligomerization domain proteins (Nod) and G-protein-coupled receptors, including formyl-methionyl peptide receptors and protease-activated receptors. Of the above bacterial and host molecules, evidence from experimental animal studies implicate LPS, fimbriae, proteases, TLRs, and CD14 in periodontal tissue or alveolar bone destruction. However, evidence verifying the involvement of any of the above molecules in periodontal tissue destruction in humans does not exist.
Collapse
Affiliation(s)
- P N Madianos
- Department of Periodontology, School of Dentistry, University of Athens, Athens, Greece.
| | | | | |
Collapse
|
11
|
Haraldsson G, Meurman JH, Könönen E, Holbrook WP. Properties of hemagglutination by Prevotella melaninogenica. Anaerobe 2005; 11:285-9. [PMID: 16701585 DOI: 10.1016/j.anaerobe.2005.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2004] [Revised: 02/16/2005] [Accepted: 02/25/2005] [Indexed: 11/30/2022]
Abstract
Although Prevotella melaninogenica belongs to the commensal oral microbiota, some strains possess putative virulence factors. For example, we have previously described fimbriated, hemagglutinating strains of P. melaninogenica, isolated from patients with periodontal disease. The aim of this investigation was to compare some chemical and physical properties of hemagglutination (HA) of P. melaninogenica with those of other pigmented gram-negative anaerobes. HA of 13 P. melaninogenica strains proved to be considerably weaker than that of the major periodontal pathogen, Porphyromonas gingivalis. Vigorous shaking reduced HA of shaken cells but the shaken supernatant had the same hemagglutinating activity as non-shaken cells. The hemagglutinating agent on P. melaninogenica seemed to be a protein, which can be separated from the cell and binds to lactose-, galactose-, and raffinose-containing carbohydrates on the erythrocytes. Adherence to epithelial cells did not differ significantly between the hemagglutinating and non-hemagglutinating strains of P. melaninogenica. Although P. melaninogenica is able to agglutinate erythrocytes, this potential virulence factor is of a considerably lower magnitude than that of major periodontal pathogens.
Collapse
|
12
|
Sztukowska M, Sroka A, Bugno M, Banbula A, Takahashi Y, Pike RN, Genco CA, Travis J, Potempa J. The C-terminal domains of the gingipain K polyprotein are necessary for assembly of the active enzyme and expression of associated activities. Mol Microbiol 2005; 54:1393-408. [PMID: 15554977 DOI: 10.1111/j.1365-2958.2004.04357.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The Porphyromonas gingivalis lysine-specific cysteine protease (gingipain K, Kgp) is expressed as a large precursor protein consisting of a leader sequence, a pro-fragment, a catalytic domain with a C-terminal IgG-like subdomain (IgSF) and a large haemagglutinin/adhesion (HA) domain. In order to directly study the role of these non-catalytic domains in pro-Kgp processing and maturation in P. gingivalis, the wild-type form of the gene was replaced with deletion variants encoding C-terminally truncated proteins, including KgpDeltaHA3/4 (Delta1292-1732 aa), KgpDeltaHA2-4 (Delta1157-1732 aa), KgpDeltaHA1-4 (Delta738-1732 aa), KgpDeltaC-term/HA (Delta681-1732 aa) and KgpDeltaIg/C-term/HA (602-1732 aa). Northern blot and reverse transcription polymerase chain reaction (RT-PCR) analysis revealed that all truncated variants of the kgp gene were transcribed in P. gingivalis. Despite high levels of kgpDeltaC-term/HA and kgpDeltaIg/C-term/HA transcripts, no Kgp-specific antigen was detected in cultures of these mutants as determined by Western blot analysis with monoclonal antibodies specific for the Kgp catalytic domain. Furthermore, only barely measurable amounts of Kgp-specific activity were detected in these two mutants. The remaining mutants expressed significant Kgp activity, however, at lower levels when compared with the parental strain. The decreased activity most probably resulted from altered folding and/or hindered secretion of the protein. The kgp gene truncation was also demonstrated to alter the distribution of the gingipain protein between membrane-associated and -secreted forms. While both gingipain K activity and the protein were cell membrane-associated in the parental strain, the mutants released significant amounts of both protein and activity into the media. Taken together, these results suggest that the C-terminal HA domains of Kgp are not only essential for full expression of gingipain activity, but also for proper processing of the multiprotein complex assembly on the P. gingivalis outer membrane. Moreover, our results indicate that the immunoglobulin-like subdomain is indispensable for proper folding and expression of the gingipains.
Collapse
Affiliation(s)
- Maryta Sztukowska
- Department of Microbiology, Faculty of Biotechnology, Jagiellonian University, Kraków, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Sojar HT, Sharma A, Genco RJ. Porphyromonas gingivalis fimbriae binds to neoglycoproteins: evidence for a lectin-like interaction. Biochimie 2004; 86:245-9. [PMID: 15194226 DOI: 10.1016/j.biochi.2004.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Accepted: 04/26/2004] [Indexed: 11/17/2022]
Abstract
Porphyromonas gingivalis is a likely major pathogen in adult periodontitis. Fimbriae in particular have been suggested as playing an important role in facilitating the initial interaction between the bacteria and the host and triggers host responses. Murakami et al. [Biochem. Biophys. Res. Commun. 192 (1993) 826] have shown that fimbriae of P. gingivalis strongly induced TNF-alpha gene expression in macrophages and expression of TNF-alpha was inhibited by N-acetyl-D-galactosamine, but not inhibited by other sugars. Studies by Sojar et al. [FEBS Lett. 422 (1998) 205] suggested that the oligosaccharide moiety of lactoferrin is involved in the interaction of P. gingivalis fimbriae and human lactoferrin. In the present study, purified fimbriae from P. gingivalis and neoglycoproteins were used to assess lectin-like interaction of fimbriae. In dot blot and overlay assays, iodinated purified P. gingivalis fimbriae as well as biotinylated purified P. gingivalis fimbriae bound strongly to albumin-fucosylamide (albumin-1-amido-1-deoxy-L-fucose) and by lesser extent to albumin-N-acetyl-D-galactosamine (albumin-p-aminophenyl-N-acetyl-beta-D-galactosaminide). However, fimbriae failed to bind carbohydrate free bovine serum albumin, which was used in preparation of the neoglycoproteins. These results suggests that P. gingivalis fimbriae bind to glycoconjugates through lectin-like interaction with carbohydrate. This protein-carbohydrate interactions may be important for triggering events in these cells, which mediate the host response of this pathogen.
Collapse
Affiliation(s)
- Hakimuddin T Sojar
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, 213 Foster Hall, 3435 Main Street, Buffalo, NY 14214-3092, USA.
| | | | | |
Collapse
|
14
|
Khlgatian M, Nassar H, Chou HH, Gibson FC, Genco CA. Fimbria-dependent activation of cell adhesion molecule expression in Porphyromonas gingivalis-infected endothelial cells. Infect Immun 2002; 70:257-67. [PMID: 11748191 PMCID: PMC127610 DOI: 10.1128/iai.70.1.257-267.2002] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gingivalis is an oral pathogen that has recently been associated with chronic inflammatory diseases such as atherosclerosis. The strength of the epidemiological associations of P. gingivalis with atherosclerosis can be increased by the demonstration that P. gingivalis can initiate and sustain growth in human vascular cells. We previously established that P. gingivalis can invade aortic, heart, and human umbilical vein endothelial cells (HUVEC), that fimbriae are required for invasion of endothelial cells, and that fimbrillin peptides can induce the expression of the chemokines interleukin 8 and monocyte chemotactic protein. In this study, we examined the expression of surface-associated cell adhesion molecules on endothelial cells in response to P. gingivalis infection by fluorescence-activated cell sorting FACS analysis and confocal microscopy. Coculture of HUVEC with P. gingivalis strain 381 or A7436 resulted in the induction in the expression of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1) and P- and E-selectins, which was maximal at 48 h postinfection. In contrast, we did not observe induction of ICAM-1, VCAM-1, or P- or E-selectin expression in HUVEC cultured with the noninvasive P. gingivalis fimA mutant DPG3 or when P. gingivalis was incubated with fimbrillin peptide-specific anti-sera prior to the addition to HUVEC. Furthermore, the addition of a peptide corresponding to the N-terminal domain of fimbrillin to HUVEC resulted in an increase in ICAM-1, VCAM-1, and P- and E-selectins, which was maximal at 48 h and similar to that observed for live P. gingivalis. Treatment of P. gingivalis-infected HUVEC with cytochalsin D, which prevented P. gingivalis invasion, also resulted in the inhibition of ICAM-1, VCAM-1, or P- and E-selectin expression. Taken together, these results indicate that active P. gingivalis invasion of HUVEC mediated via the major fimbriae stimulates surface-associated cell adhesion molecule expression. Stimulation of adhesion molecules involved in the recruitment of leukocytes to sites of inflammation by P. gingivalis may play a role in the pathogenesis of systemic inflammatory diseases associated with this microorganism, including atherosclerosis.
Collapse
Affiliation(s)
- Mary Khlgatian
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
15
|
Nassar H, Chou HH, Khlgatian M, Gibson FC, Van Dyke TE, Genco CA. Role for fimbriae and lysine-specific cysteine proteinase gingipain K in expression of interleukin-8 and monocyte chemoattractant protein in Porphyromonas gingivalis-infected endothelial cells. Infect Immun 2002; 70:268-76. [PMID: 11748192 PMCID: PMC127609 DOI: 10.1128/iai.70.1.268-276.2002] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent cross-sectional and prospective epidemiological studies have demonstrated an association between periodontal disease and atherosclerosis and human coronary heart disease. Previously, we have established that the periodontal pathogen Porphyromonas gingivalis is capable of invading aortic, heart, and human umbilical vein endothelial cells (HUVEC). Since atherosclerosis is a chronic inflammatory response initiated at the vascular wall, interactions of P. gingivalis with endothelial cells and the subsequent host cell response to infection may be important in the pathogenesis of atherosclerosis. In this study we examined the consequences of P. gingivalis infection of HUVEC on the expression of the chemokines interleukin-8 (IL-8) and monocyte chemotactic protein 1 (MCP-1). HUVEC were found to constitutively produce low levels of IL-8 and MCP-1. The addition of P. gingivalis fimbrillin-specific peptides, lipopolysaccharides (LPS), or heat-killed whole cell preparations to HUVEC stimulated modest IL-8 and MCP-1 responses. In contrast, coculture of HUVEC with live P. gingivalis strain A7436, 33277, or 381 abolished the IL-8 and MCP-1 responses. Inhibition of IL-8 and MCP-1 production was not dependent on bacterial adherence since similar results were obtained with the nonadherent P. gingivalis fimA mutant DPG3 or when P. gingivalis was preincubated with fimbrillin peptide antisera prior to the addition to HUVEC. Furthermore, treatment of P. gingivalis-infected HUVEC with cytochalsin D, which prevented P. gingivalis invasion, also abolished the constitutive IL-8 and MCP-1 responses. Treatment of HUVEC with E. coli LPS stimulated robust IL-8 and MCP-1 responses that were abolished when stimulated cells were cocultured with live P. gingivalis. Analysis of P. gingivalis-infected HUVEC cultures by an RNase protection assay revealed an increase in the IL-8 transcript relative to uninfected HUVEC. Pretreatment of P. gingivalis with protease inhibitors prior to the addition to HUVEC prevented the inhibition of IL-8 and MCP-1 production in P. gingivalis-infected HUVEC, indicating that the inhibition was proteolytically mediated. Coculture of HUVEC with a P. gingivalis mutant deficient in lysine-specific cysteine proteinase (gingipain K [Kgp]) resulted in an increase in both IL-8 transcription and protein expression relative to that observed in HUVEC cocultured with the P. gingivalis wild-type strain. These results indicate that P. gingivalis can temporally modulate the chemokine response in endothelial cells through both fimbriae and gingipain-mediated mechanisms.
Collapse
Affiliation(s)
- Hamdy Nassar
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | |
Collapse
|
16
|
Wu H, Fives-Taylor PM. Molecular strategies for fimbrial expression and assembly. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2001; 12:101-15. [PMID: 11345521 DOI: 10.1177/10454411010120020101] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fimbriae or pili are long, filamentous, multimeric macromolecules found on the bacterial cell surface. Bacteria express a diverse array of fimbriae or pili that are involved in bacterial adherence and invasion. Fimbriae can be categorized based on their modes of expression and assembly. Type I fimbriae and P pili are distributed peritrichously and translocated to the cell surface by a chaperone/usher pathway. Type 4 pili are located at the pole of the cell and assembled via the type II secretion system. Curli fimbriae are coiled surface structures assembled by an extracellular nucleation/precipitation pathway. Fimbriae of oral gram-negative and gram-positive bacteria have not been well-studied as compared with the fimbriae of enteric pathogens. Oral pathogens, such as Eikenella corrodens, Actinobacillus actinomycetemcomitans, and Porphyromonas gingivalis, possess fimbriae that have been implicated in bacterial adhesion and invasion. These fimbriae are potential virulence factors in oral infectious processes. A. actinomycetemcomitans and E. corrodens have Type 4-like fimbriae, whereas P. gingivalis displays a unique type of fimbriae. To date, fimbriae of the oral primary colonizers, Actinomyces naeslundii and Streptococcus parasanguis, represent the only fimbriae characterized for any gram-positive bacteria. The putative major fimbrial subunits, FimA and FimP of A. naeslundii and Fap1 of S. parasanguis, contain a signal sequence and cell-wall-sorting signal. The presence of extensive dipeptide repeats in Fap1 makes it unique among fimbrial molecules. Based on experimental data, a nucleation/precipitation pathway is proposed for fimbrial biogenesis of both S. parasanguis and A. naeslundii, although we cannot rule out an alternative covalent linkage model. The model systems described in this review served as a framework for hypotheses for how the known molecular factors of fimbriae on oral bacteria may be expressed and assembled.
Collapse
Affiliation(s)
- H Wu
- Department of Medicine, University of Vermont, Burlington 05405, USA
| | | |
Collapse
|
17
|
Takahashi Y, Yoshimoto H, Kato D, Hamada N, Arai M, Umemoto T. Reduced fimbria-associated activities of Porphyromonas gingivalis induced by recombinant fimbrial expression. FEMS Microbiol Lett 2001; 195:217-22. [PMID: 11179655 DOI: 10.1111/j.1574-6968.2001.tb10524.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The adhesion properties of the recombinant fimbriae (r-fimbriae) recovered from a YH522 transformant of Porphyromonas gingivalis which harbors a chimeric plasmid, pYHF2, containing the fimA gene of strain 381 were compared with those of the endogenous fimA fimbriae of strain 33277. The adhesion level of the r-fimbriae to Actinomyces viscosus was clearly lower than that of the endogenous fimbriae. In addition, the r-fimbriae were shown to lack some minor components detectable in the endogenous fimbriae. The plasmid pYHF2 prepared from the YH522 transformant was then transformed into six different P. gingivalis strains and the resultant pYHF2-containing strains were examined for their fimbrial expression. In spite of the presence of a considerable diversity in the expression level of the r-fimbriae among these transformants, it was evident that the strains expressing higher levels of the r-fimbriae exhibited a greater decrease in adhesion activity to other bacteria and to oral epithelial cells, as well as in self-aggregation.
Collapse
Affiliation(s)
- Y Takahashi
- Department of Oral Microbiology, Kanagawa Dental College, 82 Inaoka-cho, Yokosuka 238-8580, Japan
| | | | | | | | | | | |
Collapse
|
18
|
Lamont RJ, Jenkinson HF. Subgingival colonization by Porphyromonas gingivalis. ORAL MICROBIOLOGY AND IMMUNOLOGY 2000; 15:341-9. [PMID: 11154429 DOI: 10.1034/j.1399-302x.2000.150601.x] [Citation(s) in RCA: 174] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Porphyromonas gingivalis, a gram-negative anaerobe, is a major causative agent in the initiation and progression of severe forms of periodontal disease. In order to cause periodontal disease, P. gingivalis must colonize the subgingival region, a process that involves several distinct steps and multiple gene products. The organism must first navigate within the oral fluids in order to reach the hard or soft tissues of the mouth. Retention and growth of bacteria on these surfaces is facilitated by a repertoire of adhesins including fimbriae, hemagglutinins and proteinases. Once established subgingivally, P. gingivalis cells participate in intercellular communication networks with other oral prokaryotic cells and with eukaryotic cells. The establishment of these multiple interactive interfaces can lead to biofilm formation, invasion of root dentin and internalization within gingival epithelial cells. The resulting bacterial and host cellular locations, products and fate contribute to the success of P. gingivalis in colonizing the periodontal region.
Collapse
Affiliation(s)
- R J Lamont
- Department of Oral Biology, Box 357132, University of Washington, Seattle, WA 98195-7132, USA
| | | |
Collapse
|
19
|
Sojar HT, Han Y, Hamada N, Sharma A, Genco RJ. Role of the amino-terminal region of Porphyromonas gingivalis fimbriae in adherence to epithelial cells. Infect Immun 1999; 67:6173-6. [PMID: 10531284 PMCID: PMC97010 DOI: 10.1128/iai.67.11.6173-6176.1999] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gingivalis fimbriae elicit many responses in eukaryotic cells, including mitogenicity, cytokine production, epithelial cell invasion, and cellular immune response. Specific domains of the major fimbrial protein (FimA) have been shown to be important in triggering some of these functions. The goal of the present study was to identify the domain(s) of P. gingivalis FimA responsible for specific interaction with human mucosal epithelial cells. Fimbriated P. gingivalis strains have been shown to bind to buccal epithelial cells, whereas nonfimbriated strains bind at low levels or not at all. This and other studies provide evidence that FimA mediates the adherence of P. gingivalis to oral epithelial cells. To determine the specific region(s) of P. gingivalis FimA involved in epithelial cell binding, specific antipeptide antibodies were used to inhibit the binding of iodinated purified fimbriae as well as the binding of P. gingivalis cells to epithelial cells. Antibodies directed against peptides 49 to 68 (VVMANTAGAMELVGKTLAEVK) and 69 to 90 (ALTTELTAENQEAAGLIMTAEP) were found to highly inhibit both the binding of fimbriae and the binding of P. gingivalis cells to epithelial cells. The antibody against FimA peptides 69 to 90 also reacted with P. gingivalis fimbriae in immunogold labeling and immunoblot analysis, thereby indicating that this peptide domain is exposed on the surface of fimbriae. Our results suggest that the amino-terminal domain corresponding to amino acid residues 49 to 90 of the fimbrillin protein is a major epithelial cell binding domain of P. gingivalis fimbriae.
Collapse
Affiliation(s)
- H T Sojar
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214, USA.
| | | | | | | | | |
Collapse
|
20
|
Deshpande RG, Khan MB. Purification and characterization of hemolysin from Porphyromonas gingivalis A7436. FEMS Microbiol Lett 1999; 176:387-94. [PMID: 10427721 DOI: 10.1111/j.1574-6968.1999.tb13688.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Porphyromonas gingivalis, a periodontal pathogen, has the ability to lyse erythrocytes. The hemolytic activity of P. gingivalis A7436 was purified as a 45-kDa protein from the culture supernatant of a 3-days old culture using nickel-nitrilotriacetic acid chromatography. Erythrocytes treated with purified P. gingivalis hemolysin showed the presence of pores and extracellular debris by scanning electron microscopy. Active immunization of mice with 15 micrograms hemolysin induced neutralizing antibodies to hemolysin. Heating at 60 degrees C and treatment with trypsin and dithiothreitol abolished hemolytic activity, while incubation with the protease inhibitor Na-p-tosyl-L-lysine chloromethyl ketone caused no effect. We report here for the first time purification of a hemolysin from P. gingivalis A7436. The amino acid sequence of an internal peptide of hemolysin showed sequence similarity with fimbrillin from P. gingivalis HG564. However, the amino acid composition of purified hemolysin was different from that of P. gingivalis fimbrillin. Also, the ability to lyse but not agglutinate erythrocytes and to bind to nickel-nitrilotriacetic acid differentiates P. gingivalis hemolysin from fimbrillin.
Collapse
Affiliation(s)
- R G Deshpande
- Department of Microbiology and Immunology, Morehouse School of Medicine, Atlanta, GA 30310-1495, USA.
| | | |
Collapse
|
21
|
Shi Y, Ratnayake DB, Okamoto K, Abe N, Yamamoto K, Nakayama K. Genetic analyses of proteolysis, hemoglobin binding, and hemagglutination of Porphyromonas gingivalis. Construction of mutants with a combination of rgpA, rgpB, kgp, and hagA. J Biol Chem 1999; 274:17955-60. [PMID: 10364243 DOI: 10.1074/jbc.274.25.17955] [Citation(s) in RCA: 270] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Porphyromonas gingivalis produces arginine-specific cysteine proteinase (Arg-gingipain, RGP) and lysine-specific cysteine proteinase (Lys-gingipain, KGP) in the extracellular and cell-associated forms. Two separate genes (rgpA and rgpB) and a single gene (kgp) have been found to encode RGP and KGP, respectively. We constructed rgpA rgpB kgp triple mutants by homologous recombination with cloned rgp and kgp DNA interrupted by drug resistance gene markers. The triple mutants showed no RGP or KGP activity in either cell extracts or culture supernatants. The culture supernatants of the triple mutants grown in a rich medium had no proteolytic activity toward bovine serum albumin or gelatin derived from human type I collagen. Moreover, the mutants did not grow in a defined medium containing bovine serum albumin as the sole carbon/energy source. These results indicate that the proteolytic activity of P. gingivalis toward bovine serum albumin and gelatin derived from human type I collagen appears to be attributable to RGP and KGP. The hemagglutinin gene hagA of P. gingivalis possesses the adhesin domain regions responsible for hemagglutination and hemoglobin binding that are also located in the C-terminal regions of rgpA and kgp. A rgpA kgp hagA triple mutant constructed in this study exhibited no hemagglutination using sheep erythrocytes or hemoglobin binding activity, as determined by a solid-phase binding assay with horseradish peroxidase-conjugated human hemoglobin, indicating that the adhesin domains seem to be particularly important for P. gingivalis cells to agglutinate erythrocytes and bind hemoglobin, leading to heme acquisition.
Collapse
Affiliation(s)
- Y Shi
- Department of Microbiology, Faculty of Dentistry, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Affiliation(s)
- S C Holt
- Department of Microbiology, University of Texas Health Science Center at San Antonio, Graduate School of Biomedical Sciences, USA
| | | | | | | |
Collapse
|
23
|
Olsen I, Shah HN, Gharbia SE. Taxonomy and biochemical characteristics of Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis. Periodontol 2000 1999; 20:14-52. [PMID: 10522221 DOI: 10.1111/j.1600-0757.1999.tb00156.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- I Olsen
- Department of Oral Biology, Dental Faculty, University of Oslo, Norway
| | | | | |
Collapse
|
24
|
Lamont RJ, Jenkinson HF. Life below the gum line: pathogenic mechanisms of Porphyromonas gingivalis. Microbiol Mol Biol Rev 1998; 62:1244-63. [PMID: 9841671 PMCID: PMC98945 DOI: 10.1128/mmbr.62.4.1244-1263.1998] [Citation(s) in RCA: 772] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porphyromonas gingivalis, a gram-negative anaerobe, is a major etiological agent in the initiation and progression of severe forms of periodontal disease. An opportunistic pathogen, P. gingivalis can also exist in commensal harmony with the host, with disease episodes ensuing from a shift in the ecological balance within the complex periodontal microenvironment. Colonization of the subgingival region is facilitated by the ability to adhere to available substrates such as adsorbed salivary molecules, matrix proteins, epithelial cells, and bacteria that are already established as a biofilm on tooth and epithelial surfaces. Binding to all of these substrates may be mediated by various regions of P. gingivalis fimbrillin, the structural subunit of the major fimbriae. P. gingivalis is an asaccharolytic organism, with a requirement for hemin (as a source of iron) and peptides for growth. At least three hemagglutinins and five proteinases are produced to satisfy these requirements. The hemagglutinin and proteinase genes contain extensive regions of highly conserved sequences, with posttranslational processing of proteinase gene products contributing to the formation of multimeric surface protein-adhesin complexes. Many of the virulence properties of P. gingivalis appear to be consequent to its adaptations to obtain hemin and peptides. Thus, hemagglutinins participate in adherence interactions with host cells, while proteinases contribute to inactivation of the effector molecules of the immune response and to tissue destruction. In addition to direct assault on the periodontal tissues, P. gingivalis can modulate eucaryotic cell signal transduction pathways, directing its uptake by gingival epithelial cells. Within this privileged site, P. gingivalis can replicate and impinge upon components of the innate host defense. Although a variety of surface molecules stimulate production of cytokines and other participants in the immune response, P. gingivalis may also undertake a stealth role whereby pivotal immune mediators are selectively inactivated. In keeping with its strict metabolic requirements, regulation of gene expression in P. gingivalis can be controlled at the transcriptional level. Finally, although periodontal disease is localized to the tissues surrounding the tooth, evidence is accumulating that infection with P. gingivalis may predispose to more serious systemic conditions such as cardiovascular disease and to delivery of preterm infants.
Collapse
Affiliation(s)
- R J Lamont
- Department of Oral Biology, University of Washington, Seattle, Washington 98195, USA.
| | | |
Collapse
|
25
|
Baqui AA, Meiller TF, Turng BF, Kelley JI, Falkler WA. Functional changes in THP-1 human monocytic cells after stimulation with lipopolysaccharide of oral microorganisms and granulocyte macrophage colony stimulating factor. Immunopharmacol Immunotoxicol 1998; 20:493-518. [PMID: 9805230 DOI: 10.3109/08923979809031512] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A human THP-1 monocyte cell line culture system has been utilized to observe the effect of granulocyte macrophage colony stimulating factor (GM-CSF) supplementation with lipopolysaccharide (LPS) of oral microorganisms to stimulate monocyte/macrophage functional activity. LPS of oral microorganisms, Fusobacterium nucleatum and Porphyromonas gingivalis was produced by phenol-water extraction and characterized. The phagocytosis assay was performed using F1TC labeled Saccharomyces yeast particles. Phagocytic functional activity was observed in 10-11% of resting THP-1 cells. Treatment of THP-1 cells with LPS of F. nucleatum or P. gingivalis increased the phagocytic activity of THP-1 cells 2-3 fold. GM-CSF significantly increased phagocytosis either alone or when supplemented with LPS of F. nucleatum or P. gingivalis. A chemotaxis assay was performed using a 48 well chemotaxis chamber. Chemotactic functional activity of THP-1 cells was increased 2-fold after 4 days of treatment with GM-CSF. Stimulation of THP-1 cells with LPS of F. nucleatum or P. gingivalis significantly reduced the chemotactic activity indicating the maturation towards a fixed macrophage. There were functional variations (chemotaxis and phagocytosis) in THP-1 cells in response to LPS of oral microorganisms following stimulation with GM-CSF.
Collapse
Affiliation(s)
- A A Baqui
- Department of Oral Medicine, Dental School, University of Maryland, Baltimore 21201, USA
| | | | | | | | | |
Collapse
|
26
|
Hamada S, Amano A, Kimura S, Nakagawa I, Kawabata S, Morisaki I. The importance of fimbriae in the virulence and ecology of some oral bacteria. ORAL MICROBIOLOGY AND IMMUNOLOGY 1998; 13:129-38. [PMID: 10093527 DOI: 10.1111/j.1399-302x.1998.tb00724.x] [Citation(s) in RCA: 131] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cumulative evidence indicates that bacterial adherence to mucosal and tooth surfaces as well as bacterial coaggregation are essential steps for colonization of various oral bacterial species. Bacterial fimbriae have been shown to play an important role in the interaction between bacteria and host cells or among bacterial cells. The properties of fimbriae from selected species of oral bacteria are discussed in terms of virulence traits and ecological significance. Among others, Porphyromonas gingivalis fimbriae have been most extensively studied. The fimbrial structure is composed of 41-kDa fimbrillin proteins. DNA sequencing of the fimbrillin gene (fimA) from nine strains of P. gingivalis suggests intraspecies variation in the structure of fimA, while retaining common immunochemical specificities. P. gingivalis fimbriae exhibit a wide variety of biological activities including immunogenicity, binding to various host proteins, stimulation of cytokine production and promotion of bone resorption, Actinobacillus actinomycetemcomitans also possesses fimbriae; however, little is known concerning their chemical, genetical, and biological properties. Fimbriae of Prevotella intermedia are shown to induce hemagglutination reaction, while those of Prevotella loescheii are found to cause coaggregation with other bacteria, i.e., Actinomyces viscosus and sanguis streptococci. Fimbriae from gram-positive oral bacteria such as oral Actinomyces and sanguis streptococci are described. These fimbriae may participate in coaggregation, binding to saliva-coated hydroxyapatite or glycoprotein of the surface layer of oral epithelial cells. Taken together, fimbriae are key components in cell-to-surface and cell-to-cell adherence of oral bacteria and pathogenesis of some oral and systemic diseases.
Collapse
Affiliation(s)
- S Hamada
- Department of Oral Microbiology, Osaka University Faculty of Dentistry, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Ellen RP, Lépine G, Nghiem PM. In vitro models that support adhesion specificity in biofilms of oral bacteria. Adv Dent Res 1997; 11:33-42. [PMID: 9524440 DOI: 10.1177/08959374970110011401] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Adhesion to adsorbed pellicles and interspecies co-adhesion to form plaque biofilms involve selective interactions of bacterial adhesins with specific receptors. Our laboratory has devised in vitro assays for co-adhesion between Actinomyces naeslundii and Streptococcus oralis or Porphyromonas gingivalis on saliva-coated mineral and hexadecane droplet substrata. P. gingivalis structures significant for co-adhesion with A. naeslundii include surface vesicles and fimbriae. A family of arginine-specific cysteine proteinases in vesicles may be involved in adherence to bacteria, to host cells, and to matrix proteins. New research from several laboratories has found that such proteinases are processed from genes encoding polyproteins containing both proteinase and hemagglutinin domains. In addition to enzyme-substrate recognition, bacterial adhesion is often determined by specific protein-peptide and lectincarbohydrate recognition. A. naeslundii--salivary prolinerich protein, S. gordonii--salivary alpha-amylase, and Treponema denticola--matrix protein recognition are examples of the former. Co-adhesion of A. naeslundii and S. oralis is an example of the latter. Lactose can selectively desorb A. naeslundii cells from mixed biofilms with S. oralis, a demonstration of the significance of specificity. Although non-specific forces are probably secondary to stereochemical fit in determining the selective range of surfaces that bacteria have evolved to recognize and bind, they probably help stabilize non-covalent bonds within aligned, complementary domains.
Collapse
Affiliation(s)
- R P Ellen
- Department of Periodontics, University of Toronto, Faculty of Dentistry, ON, Canada
| | | | | |
Collapse
|
28
|
Ogawa T, Ogo H, Kinoshita A. Antagonistic effect of synthetic peptides corresponding to the binding regions within fimbrial subunit protein from Porphyromonas gingivalis to human gingival fibroblasts. Vaccine 1997; 15:230-6. [PMID: 9066043 DOI: 10.1016/s0264-410x(96)00127-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Specific binding region within fimbrial subunit protein (fimbrilin) from Porphyromonas gingivalis strain 381 was studied in cultured human gingival fibroblasts. Fluorescent micrographs visualised FITC-labelled fimbriae of P. gingivalis specifically bound to normal human fibroblast cell line (Gin-1) along the cell surface. Flow cytometric analysis also revealed the binding of FITC-labelled fimbriae to Gin-1 cells. Synthetic peptides composed of residues 1-20 (AFGVGDDESKVAKLTVMVYN) of the fimbrilin from P. gingivalis, FP381 (1-20), FP381 (69-80; ALTTELTAENQE) and FP381 (171-181; DA-NYLTGSLTT) definitely inhibited P. gingivalis fimbria-binding to Gin-1 cells by enzyme-linked immunosorbent assay (ELISA). Furthermore, based on the Scatchard plot analysis of the binding of 125I-labelled P. gingivalis fimbriae to Gin-1 cells, the apparent dissociation constant (Kd) was calculated as 15.9 pM, and the number of binding sites (Rt) was estimated as 150 sites/cell. Binding studies of 125I-labelled FP381(171-181) also revealed the presence of a non-interacting, single class of affinity binding sites: the apparent Kd and Rt were 29.2 nM and 18440 sites/cell on Gin-1 cells, respectively. These results demonstrate that specific binding regions on P. gingivalis fimbriae to human gingival fibroblasts are present, and certain corresponding peptides clearly inhibited the binding of P. gingivalis fimbriae to human gingival fibroblasts.
Collapse
Affiliation(s)
- T Ogawa
- Department of Oral Microbiology, Osaka University Faculty of Dentistry, Japan
| | | | | |
Collapse
|
29
|
Abstract
Adherence to a surface is a key element for colonization of the human oral cavity by the more than 500 bacterial taxa recorded from oral samples. Three surfaces are available: teeth, epithelial mucosa, and the nascent surface created as each new bacterial cell binds to existing dental plaque. Oral bacteria exhibit specificity for their respective colonization sites. Such specificity is directed by adhesin-receptor cognate pairs on genetically distinct cells. Colonization is successful when adherent cells grow and metabolically participate in the oral bacterial community. The potential roles of adherence-relevant molecules are discussed in the context of the dynamic nature of the oral econiche.
Collapse
Affiliation(s)
- C J Whittaker
- Laboratory of Microbial Ecology, National Institute of Dental Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
30
|
Lépine G, Ellen RP, Progulske-Fox A. Construction and preliminary characterization of three hemagglutinin mutants of Porphyromonas gingivalis. Infect Immun 1996; 64:1467-72. [PMID: 8606121 PMCID: PMC173946 DOI: 10.1128/iai.64.4.1467-1472.1996] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Targeted insertional mutagenesis was used to construct hagA, hagB, and hagC hemagglutinin mutants of Porphyromonas gingivalis. pJRD215-derived plasmids containing tetA(Q)2 and portions of the targeted genes were conjugated into P. gingivalis. Interruption of the three loci was confirmed by Southern hybridization, sequencing, reverse transcription-PCR, and microtiter hemagglutination assays. No significant differences in hydrophobicity or coadherence to Actinomyces viscosus were detected between the mutants and the wild-type strain.
Collapse
Affiliation(s)
- G Lépine
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
31
|
Chandad F, Mayrand D, Grenier D, Hinode D, Mouton C. Selection and phenotypic characterization of nonhemagglutinating mutants of Porphyromonas gingivalis. Infect Immun 1996; 64:952-8. [PMID: 8641806 PMCID: PMC173862 DOI: 10.1128/iai.64.3.952-958.1996] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
To further investigate the relationship between fimbriae and the hemagglutinating adhesin HA-Ag2 of Porphyromonas gingivalis, three spontaneous mutants of the type strain ATCC 33277 were selected by a hemadsorption procedure. They were characterized for hemagglutination, trypsin-like and lectin-binding activities, and hydrophobicity and for the presence of fimbriae. The presence of the 42-kDa (the fimbrilin subunit) and the 43- and 49-kDa (the HA-Ag2 components) polypeptides was investigated by immunoblotting using polyclonal and monoclonal antibodies directed to fimbriae and to the hemagglutinating adhesin HA-Ag2. Cells from two of the three mutants (M1 and M2) exhibited no or little hemagglutination activity and very low trypsin-like activity and did not show the 43- and 49-kDa polypeptides. Abnormal fimbriation in M1 was deduced from the following observations of cells grown for 18 h: absence of the 42-kDa polypeptide and of a 14-kDa polypeptide and no fimbriae visible on electron micrographs. While the cells of mutant M2, irrespective of the age of the culture, were found to lack the 43- and 49-kDa polypeptides and hemagglutination activity, the supernatants of cultures grown for 72 h had high hemagglutination and trypsin-like activities and revealed the presence of the 42-, 43-, and 49-kDa polypeptides. This suggests that M2 may be missing some molecules which anchor the components to the cell surface. Mutant M3 showed levels of activities similar to those of the parental strain but lacked the 43-kDa polypeptide. Other pleiotropic effects observed for the mutants included loss of dark pigmentation and lower hydrophobicity. The data from this study fuel an emerging consensus whereby fimbriation, hemagglutination, and proteolytic activities, as well as other functions in P. gingivalis, are intricate.
Collapse
Affiliation(s)
- F Chandad
- Groupe de Recherche en Ecologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada
| | | | | | | | | |
Collapse
|
32
|
Chandad F, Mouton C. Antigenic, structural, and functional relationships between fimbriae and the hemagglutinating adhesin HA-Ag2 of Porphyromonas gingivalis. Infect Immun 1995; 63:4755-63. [PMID: 7591132 PMCID: PMC173681 DOI: 10.1128/iai.63.12.4755-4763.1995] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
While the adhesive properties of Porphyromonas gingivalis are known to allow colonization of the subgingival tissues, the roles of fimbriae and adhesin molecules in hemagglutination remain unclear. The purpose of this study was to analyze the antigenic, structural, and functional relationships of these two components. Five populations of monoclonal antibodies were produced against (i) the hemagglutinating adhesin HA-Ag2 resolved by crossed immunoelectrophoresis (CIE), (ii) native fimbriae, and (iii) each of the three immunoprecipitates, Ag8a, Ag8b, and Ag8c, that define fimbriae by CIE. The tests used for characterization of the monoclonal antibodies included immunoblot reactivity, inhibition of hemagglutination, capacity to dissociate immunoprecipitates by CIE, localization of recognized epitopes by immunoelectron microscopy, and epitope mapping by competition enzyme-linked immunosorbent assay. The results from the different immunochemical tests clearly showed a close antigenic relationship between fimbriae and the hemagglutinating adhesin HA-Ag2. We were able to establish that the epitopic domain H1 of HA-Ag2 is hemagglutinin specific and that domain F2 is fimbria specific. Our data indicate that the polymeric structural unit of fimbriae must be complexed to HA-Ag2, the adhesin, to confer hemagglutination activity to the bacterial cells.
Collapse
Affiliation(s)
- F Chandad
- Groupe de Recherche en Ecologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, Canada
| | | |
Collapse
|
33
|
Yasuda K, Ogawa T, Shimauchi H. Identification of Porphyromonas gingivalis by an ELISA using a cellulose acetate paper point. METHODS IN MICROBIOLOGY 1995. [DOI: 10.1016/0167-7012(95)00049-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Hamada S, Fujiwara T, Morishima S, Takahashi I, Nakagawa I, Kimura S, Ogawa T. Molecular and immunological characterization of the fimbriae of Porphyromonas gingivalis. Microbiol Immunol 1994; 38:921-30. [PMID: 7723684 DOI: 10.1111/j.1348-0421.1994.tb02148.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- S Hamada
- Department of Oral Microbiology, Osaka University Faculty of Dentistry, Japan
| | | | | | | | | | | | | |
Collapse
|