Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Oral Microbiol ; 12(1): 1808750, 2020 Aug 24.
Article in English | MEDLINE | ID: mdl-32944158

ABSTRACT

BACKGROUND:  Porphyromonas gingivalis and Treponema denticola are proteolytic periodontopathogens that co-localize in polymicrobial subgingival plaque biofilms, display in vitro growth symbiosis and synergistic virulence in animal models of disease. These symbioses are underpinned by a range of metabolic interactions including cooperative hydrolysis of glycine-containing peptides to produce free glycine, which T. denticola uses as a major energy and carbon source. OBJECTIVE:  To characterize the P. gingivalis gene products essential for these interactions. Methods: The P. gingivalis transcriptome exposed to cell-free T. denticola conditioned medium was determined using RNA-seq. P. gingivalis proteases potentially involved in hydrolysis of glycine-containing peptides were identified using a bioinformatics approach. RESULTS:  One hundred and thirty-twogenes displayed differential expression, with the pattern of gene expression consistent with succinate cross-feeding from T. denticola to P. gingivalis and metabolic shifts in the P. gingivalis folate-mediated one carbon superpathway. Interestingly, no P. gingivalis proteases were significantly up-regulated. Three P. gingivalis proteases were identified as candidates and inactivated to determine their role in the release of free glycine. P. gingivalis PG0753 and PG1788 but not PG1605 are involved in the hydrolysis of glycine-containing peptides, making free glycine available for T. denticola utilization. CONCLUSION:  Collectively these metabolic interactions help to partition resources and engage synergistic interactions between these two species.

2.
Article in English | MEDLINE | ID: mdl-31921707

ABSTRACT

Chronic periodontitis has a polymicrobial biofilm etiology and interactions between key oral bacterial species, such as Porphyromonas gingivalis and Treponema denticola contribute to disease progression. P. gingivalis and T. denticola are co-localized in subgingival plaque and have been previously shown to exhibit strong synergy in growth, biofilm formation and virulence in an animal model of disease. The motility of T. denticola, although not considered as a classic virulence factor, may be involved in synergistic biofilm development between P. gingivalis and T. denticola. We determined the role of T. denticola motility in polymicrobial biofilm development using an optimized transformation protocol to produce two T. denticola mutants targeting the motility machinery. These deletion mutants were non-motile and lacked the gene encoding the flagellar hook protein of the periplasmic flagella (ΔflgE) or a component of the stator motor that drives the flagella (ΔmotB). The specificity of these gene deletions was determined by whole genome sequencing. Quantitative proteomic analyses of mutant strains revealed that the specific inactivation of the motility-associated gene, motB, had effects beyond motility. There were 64 and 326 proteins that changed in abundance in the ΔflgE and ΔmotB mutants, respectively. In the ΔflgE mutant, motility-associated proteins showed the most significant change in abundance confirming the phenotype change for the mutant was related to motility. However, the inactivation of motB as well as stopping motility also upregulated cellular stress responses in the mutant indicating pleiotropic effects of the mutation. T. denticola wild-type and P. gingivalis displayed synergistic biofilm development with a 2-fold higher biomass of the dual-species biofilms than the sum of the monospecies biofilms. Inactivation of T. denticola flgE and motB reduced this synergy. A 5-fold reduction in dual-species biofilm biomass was found with the motility-specific ΔflgE mutant suggesting that T. denticola periplasmic flagella are essential in synergistic biofilm formation with P. gingivalis.


Subject(s)
Bacterial Proteins/genetics , Biofilms/growth & development , Porphyromonas gingivalis/growth & development , Treponema denticola/genetics , Animals , Chronic Periodontitis/microbiology , Gene Deletion , Genome, Bacterial/genetics , Humans , Locomotion/genetics , Locomotion/physiology , Microbial Interactions/physiology , Treponema denticola/growth & development , Treponema denticola/pathogenicity , Virulence Factors/genetics , Virulence Factors/metabolism , Whole Genome Sequencing
3.
PLoS One ; 11(10): e0164313, 2016.
Article in English | MEDLINE | ID: mdl-27711252

ABSTRACT

Porphyromonas gingivalis utilises the Bacteroidetes-specific type IX secretion system (T9SS) to export proteins across the outer membrane (OM), including virulence factors such as the gingipains. The secreted proteins have a conserved carboxy-terminal domain essential for type IX secretion that is cleaved upon export. In P. gingivalis the T9SS substrates undergo glycosylation with anionic lipopolysaccharide (A-LPS) and are attached to the OM. In this study, comparative analyses of 24 Bacteroidetes genomes identified ten putative novel components of the T9SS in P. gingivalis, one of which was PG1058. Computer modelling of the PG1058 structure predicted a novel N- to C-terminal architecture comprising a tetratricopeptide repeat (TPR) domain, a ß-propeller domain, a carboxypeptidase regulatory domain-like fold (CRD) and an OmpA_C-like putative peptidoglycan binding domain. Inactivation of pg1058 in P. gingivalis resulted in loss of both colonial pigmentation and surface-associated proteolytic activity; a phenotype common to T9SS mutants. Immunoblot and LC-MS/MS analyses of subcellular fractions revealed T9SS substrates accumulated within the pg1058 mutant periplasm whilst whole-cell ELISA showed the Kgp gingipain was absent from the cell surface, confirming perturbed T9SS function. Immunoblot, TEM and whole-cell ELISA analyses indicated A-LPS was produced and present on the pg1058 mutant cell surface although it was not linked to T9SS substrate proteins. This indicated that PG1058 is crucial for export of T9SS substrates but not for the translocation of A-LPS. PG1058 is a predicted lipoprotein and was localised to the periplasmic side of the OM using whole-cell ELISA, immunoblot and LC-MS/MS analyses of subcellular fractions. The structural prediction and localisation of PG1058 suggests that it may have a role as an essential scaffold linking the periplasmic and OM components of the T9SS.


Subject(s)
Bacterial Proteins/chemistry , Lipid-Linked Proteins/chemistry , Porphyromonas gingivalis/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Lipid-Linked Proteins/genetics , Lipid-Linked Proteins/immunology , Lipid-Linked Proteins/metabolism , Lipopolysaccharides/metabolism , Molecular Sequence Data , Mutation , Peptide Hydrolases/metabolism , Phenotype , Porphyromonas gingivalis/genetics , Protein Domains , Tandem Mass Spectrometry
4.
Microb Pathog ; 94: 60-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26541672

ABSTRACT

Chronic periodontitis has a polymicrobial biofilm aetiology. Polymicrobial biofilms are complex, dynamic microbial communities formed by two or more bacterial species that are important for the persistence and proliferation of participating microbes in the environment. Interspecies adherence, which often involves bacterial surface-associated molecules, and communications are essential in the spatial and temporal development of a polymicrobial biofilm, which in turn is necessary for the overall fitness of a well-organized multispecies biofilm community. In the oral cavity, interactions between key oral bacterial species, including Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia, are essential for the progression of chronic periodontitis. In vivo, P. gingivalis and T. denticola are frequently found to co-exist in deep periodontal pockets and have been co-localized to the superficial layers of subgingival plaque as microcolony blooms adjacent to the pocket epithelium, suggesting possible interbacterial interactions that contribute towards disease. The motility and chemotactic ability of T. denticola, although not considered as classic virulence factors, are likely to be important in the synergistic biofilm formation with P. gingivalis. In vitro, P. gingivalis and T. denticola display a symbiotic relationship in nutrient utilization and growth promotion. Together these data suggest there is an intimate relationship between these two species that has evolved to enhance their survival and virulence.


Subject(s)
Dental Plaque/microbiology , Gingiva/microbiology , Porphyromonas gingivalis/physiology , Tannerella forsythia/growth & development , Treponema denticola/physiology , Adhesins, Bacterial/physiology , Biofilms/growth & development , Chemotaxis/physiology , Chronic Periodontitis/microbiology , Colony Count, Microbial , Humans , Microbial Interactions , Periodontal Pocket/microbiology , Symbiosis , Virulence
5.
NPJ Vaccines ; 1: 16022, 2016.
Article in English | MEDLINE | ID: mdl-29263860

ABSTRACT

Porphyromonas gingivalis infected mice with an established P. gingivalis-specific inflammatory immune response were protected from developing alveolar bone resorption by therapeutic vaccination with a chimera (KAS2-A1) immunogen targeting the major virulence factors of the bacterium, the gingipain proteinases. Protection was characterised by an antigen-specific IgG1 isotype antibody and Th2 cell response. Adoptive transfer of KAS2-A1-specific IgG1 or IgG2 expressing B cells confirmed that IgG1-mediated protection. Furthermore, parenteral or intraoral administration of KAS2-A1-specific polyclonal antibodies protected against the development of P. gingivalis-induced bone resorption. The KAS2-A1-specific antibodies neutralised the gingipains by inhibiting: proteolytic activity, binding to host cells/proteins and co-aggregation with other periodontal bacteria. Combining key gingipain sequences into a chimera vaccine produced an effective therapeutic intervention that protected against P. gingivalis-induced periodontitis.

6.
PLoS Pathog ; 10(3): e1003955, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603978

ABSTRACT

Porphyromonas gingivalis and Treponema denticola are strongly associated with chronic periodontitis. These bacteria have been co-localized in subgingival plaque and demonstrated to exhibit symbiosis in growth in vitro and synergistic virulence upon co-infection in animal models of disease. Here we show that during continuous co-culture a P. gingivalis:T. denticola cell ratio of 6∶1 was maintained with a respective increase of 54% and 30% in cell numbers when compared with mono-culture. Co-culture caused significant changes in global gene expression in both species with altered expression of 184 T. denticola and 134 P. gingivalis genes. P. gingivalis genes encoding a predicted thiamine biosynthesis pathway were up-regulated whilst genes involved in fatty acid biosynthesis were down-regulated. T. denticola genes encoding virulence factors including dentilisin and glycine catabolic pathways were significantly up-regulated during co-culture. Metabolic labeling using 13C-glycine showed that T. denticola rapidly metabolized this amino acid resulting in the production of acetate and lactate. P. gingivalis may be an important source of free glycine for T. denticola as mono-cultures of P. gingivalis and T. denticola were found to produce and consume free glycine, respectively; free glycine production by P. gingivalis was stimulated by T. denticola conditioned medium and glycine supplementation of T. denticola medium increased final cell density 1.7-fold. Collectively these data show P. gingivalis and T. denticola respond metabolically to the presence of each other with T. denticola displaying responses that help explain enhanced virulence of co-infections.


Subject(s)
Porphyromonas gingivalis/metabolism , Symbiosis/physiology , Treponema denticola/metabolism , Coculture Techniques , Coinfection , Microscopy, Electron, Scanning , Oligonucleotide Array Sequence Analysis , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/growth & development , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Treponema denticola/genetics , Treponema denticola/growth & development
7.
PLoS One ; 8(8): e71727, 2013.
Article in English | MEDLINE | ID: mdl-23990979

ABSTRACT

Chronic periodontitis has a polymicrobial biofilm aetiology and interactions between key bacterial species are strongly implicated as contributing to disease progression. Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia have all been implicated as playing roles in disease progression. P. gingivalis cell-surface-located protease/adhesins, the gingipains, have been suggested to be involved in its interactions with several other bacterial species. The aims of this study were to determine polymicrobial biofilm formation by P. gingivalis, T. denticola and T. forsythia, as well as the role of P. gingivalis gingipains in biofilm formation by using a gingipain null triple mutant. To determine homotypic and polymicrobial biofilm formation a flow cell system was employed and the biofilms imaged and quantified by fluorescent in situ hybridization using DNA species-specific probes and confocal scanning laser microscopy imaging. Of the three species, only P. gingivalis and T. denticola formed mature, homotypic biofilms, and a strong synergy was observed between P. gingivalis and T. denticola in polymicrobial biofilm formation. This synergy was demonstrated by significant increases in biovolume, average biofilm thickness and maximum biofilm thickness of both species. In addition there was a morphological change of T. denticola in polymicrobial biofilms when compared with homotypic biofilms, suggesting reduced motility in homotypic biofilms. P. gingivalis gingipains were shown to play an essential role in synergistic polymicrobial biofilm formation with T. denticola.


Subject(s)
Biofilms/growth & development , Porphyromonas gingivalis/physiology , Treponema denticola/physiology , Treponema/physiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Chronic Periodontitis/microbiology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Gingipain Cysteine Endopeptidases , In Situ Hybridization, Fluorescence , Microscopy, Confocal , Microscopy, Electron, Scanning , Mutation , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/ultrastructure , Species Specificity , Treponema/genetics , Treponema/ultrastructure , Treponema denticola/genetics , Treponema denticola/ultrastructure
8.
PLoS One ; 8(6): e65447, 2013.
Article in English | MEDLINE | ID: mdl-23762374

ABSTRACT

Porphyromonas gingivalis is a major pathogen associated with chronic periodontitis. The organism's cell-surface cysteine proteinases, the Arg-specific proteinases (RgpA, RgpB) and the Lys-specific proteinase (Kgp), which are known as gingipains have been implicated as major virulence factors. All three gingipain precursors contain a propeptide of around 200 amino acids in length that is removed during maturation. The aim of this study was to characterize the inhibitory potential of the Kgp and RgpB propeptides against the mature cognate enzymes. Mature Kgp was obtained from P. gingivalis mutant ECR368, which produces a recombinant Kgp with an ABM1 motif deleted from the catalytic domain (rKgp) that enables the otherwise membrane bound enzyme to dissociate from adhesins and be released. Mature RgpB was obtained from P. gingivalis HG66. Recombinant propeptides of Kgp and RgpB were produced in Escherichia coli and purified using nickel-affinity chromatography. The Kgp and RgpB propeptides displayed non-competitive inhibition kinetics with K(i) values of 2.04 µM and 12 nM, respectively. Both propeptides exhibited selectivity towards their cognate proteinase. The specificity of both propeptides was demonstrated by their inability to inhibit caspase-3, a closely related cysteine protease, and papain that also has a relatively long propeptide. Both propeptides at 100 mg/L caused a 50% reduction of P. gingivalis growth in a protein-based medium. In summary, this study demonstrates that gingipain propeptides are capable of inhibiting their mature cognate proteinases.


Subject(s)
Adhesins, Bacterial/chemistry , Cysteine Endopeptidases/chemistry , Hemagglutinins/chemistry , Peptide Fragments/pharmacology , Porphyromonas gingivalis/physiology , Protein Precursors/physiology , Recombinant Proteins/pharmacology , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Caspase 3/chemistry , Caspase 3/metabolism , Catalytic Domain , Chromatography, Liquid , Cysteine Endopeptidases/metabolism , Gingipain Cysteine Endopeptidases , Hemagglutinins/metabolism , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
FEBS Lett ; 587(9): 1275-80, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23499434

ABSTRACT

Arg-gingipain B (RgpB), a major virulence factor secreted by the periodontal pathogen Porphyromonas gingivalis is an Arg-specific cysteine proteinase. By monitoring proteolytic cleavage of a human salivary peptide histatin 5 using MALDI-TOF MS, RgpB purified from P. gingivalis HG66 was found to shift from a dominant Arg-X to dominant Lys-X activity, both in vitro and in vivo, upon reversible cysteine oxidation. Native PAGE analysis revealed the association of novel Lys-X activity with a reversible state change of the oxidized enzyme. The redox-regulated Lys-X activity of RgpB may provide a survival advantage to P. gingivalis against the oxidative host defence.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Porphyromonas gingivalis/enzymology , Adhesins, Bacterial/chemistry , Amino Acid Sequence , Catalytic Domain , Cysteine Endopeptidases/chemistry , Gingipain Cysteine Endopeptidases , Histatins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Porphyromonas gingivalis/cytology , Solubility , Substrate Specificity
10.
J Biol Chem ; 287(29): 24605-17, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22593568

ABSTRACT

Protein substrates of a novel secretion system of Porphyromonas gingivalis contain a conserved C-terminal domain (CTD) of ∼70-80 amino acid residues that is essential for their secretion and attachment to the cell surface. The CTD itself has not been detected in mature substrates, suggesting that it may be removed by a novel signal peptidase. More than 10 proteins have been shown to be essential for the proper functioning of the secretion system, and one of these, PG0026, is a predicted cysteine proteinase that also contains a CTD, suggesting that it may be a secreted component of the secretion system and a candidate for being the CTD signal peptidase. A PG0026 deletion mutant was constructed along with a PG0026C690A targeted mutant encoding an altered catalytic Cys residue. Analysis of clarified culture fluid fractions by SDS-PAGE and mass spectrometry revealed that the CTD was released intact into the surrounding medium in the wild type strain, but not in the PG0026 mutant strains. Western blot experiments revealed that the maturation of a model substrate was stalled at the CTD-removal step specifically in the PG0026 mutants, and whole cell ELISA experiments demonstrated partial secretion of substrates to the cell surface. The CTD was also shown to be accessible at the cell surface in the PG0026 mutants, suggesting that the CTD was secreted but could not be cleaved. The data indicate that PG0026 is responsible for the cleavage of the CTD signal after substrates are secreted across the OM.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Amino Acid Sequence , Blotting, Western , Computational Biology , Cryoelectron Microscopy , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Mass Spectrometry , Membrane Proteins/genetics , Microscopy, Electron, Scanning , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Deletion/genetics , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Virulence Factors/genetics
11.
J Bacteriol ; 193(1): 132-42, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20971915

ABSTRACT

Porphyromonas gingivalis, a periodontal pathogen, expresses a group of surface proteins with a common C-terminal domain (CTD) that are exported by a novel secretion system to the surface, where they are covalently attached. Using RgpB as a model CTD protein, we have produced a series of site-directed mutations in the CTD sequence at conserved residues and at residues that may be modified and, hence, surface attached. The mutant RgpB proteins were expressed in a P. gingivalis host lacking functional RgpB and RgpA Arg-specific proteases. The RgpB mutants produced were Y674F, Y674F Y718F, T675Q S679Q T682Q T684Q, T693Q, F695A, D696A, N698A, G699P, G716P, T724Q, T728Q T730Q, and K732Q and a protein with a deletion of residues 692 to 702 (Δ692-702). The mutants were characterized for cell-associated Arg-specific protease activity and for cellular distribution using anti-Rgp antibodies and Western blotting of culture fractions. All the mutants exhibited cell-associated Arg-specific activity similar to that of the positive control except for the D696A and Δ692-702 mutants. For all mutants, except D696A and Δ692-702, the RgpB proteins were found modified and attached to the cell surface, which was the same profile found in the positive-control strain. Only trace amounts of the precursor form of the Δ692-702 mutant were detected in the outer membrane, with none detected in the periplasm or culture fluid although cell transcript levels were normal. The results suggest that residues 692 to 702 of the CTD, in particular, residue D696, have an important role in the attachment of RgpB at the cell surface and that without attachment secretion does not occur.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Gene Expression Regulation, Bacterial/physiology , Porphyromonas gingivalis/metabolism , Adhesins, Bacterial/genetics , Amino Acid Sequence , Amino Acid Substitution , Bacterial Adhesion/physiology , Cysteine Endopeptidases/genetics , Gingipain Cysteine Endopeptidases , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Porphyromonas gingivalis/genetics
12.
Microbiology (Reading) ; 156(Pt 3): 774-788, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007650

ABSTRACT

Treponema denticola is an oral spirochaete that has been strongly associated with chronic periodontitis. The bacterium exists as part of a dense biofilm (subgingival dental plaque) accreted to the tooth. To determine T. denticola gene products important for persistence as a biofilm we developed a continuous-culture biofilm model and conducted a genome-wide transcriptomic analysis of biofilm and planktonic cells. A total of 126 genes were differentially expressed with a fold change of 1.5 or greater. This analysis identified the upregulation of putative prophage genes in the T. denticola 35405 genome. Intact bacteriophage particles were isolated from T. denticola and circular phage DNA was detected by PCR analysis. This represents the first, to our knowledge, functional bacteriophage isolated from T. denticola, which we have designated varphitd1. In biofilm cells there was also an upregulation of genes encoding several virulence factors, toxin-antitoxin systems and a family of putative transposases. Together, these data indicate that there is a higher potential for genetic mobility in T. denticola when growing as a biofilm and that these systems are important for the biofilm persistence and therefore virulence of this bacterium.


Subject(s)
Antitoxins/metabolism , Biofilms , Genome, Bacterial , Prophages/isolation & purification , Transposases/metabolism , Treponema denticola/genetics , Antitoxins/genetics , Computational Biology , DNA, Bacterial/genetics , DNA, Viral/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Viral , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Prophages/genetics , Prophages/ultrastructure , Proteome/metabolism , RNA, Bacterial/genetics , Transposases/genetics , Treponema denticola/enzymology , Treponema denticola/virology
13.
BMC Microbiol ; 9: 18, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-19175941

ABSTRACT

BACKGROUND: Porphyromonas gingivalis in subgingival dental plaque, as part of a mature biofilm, has been strongly implicated in the onset and progression of chronic periodontitis. In this study using DNA microarray we compared the global gene expression of a P. gingivalis biofilm with that of its planktonic counterpart grown in the same continuous culture. RESULTS: Approximately 18% (377 genes, at 1.5 fold or more, P-value < 0.01) of the P. gingivalis genome was differentially expressed when the bacterium was grown as a biofilm. Genes that were down-regulated in biofilm cells, relative to planktonic cells, included those involved in cell envelope biogenesis, DNA replication, energy production and biosynthesis of cofactors, prosthetic groups and carriers. A number of genes encoding transport and binding proteins were up-regulated in P. gingivalis biofilm cells. Several genes predicted to encode proteins involved in signal transduction and transcriptional regulation were differentially regulated and may be important in the regulation of biofilm growth. CONCLUSION: This study analyzing global gene expression provides insight into the adaptive response of P. gingivalis to biofilm growth, in particular showing a down regulation of genes involved in growth and metabolic activity.


Subject(s)
Biofilms , Gene Expression Profiling , Porphyromonas gingivalis/genetics , Gene Expression Regulation, Bacterial , Oligonucleotide Array Sequence Analysis , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/metabolism , RNA, Bacterial/genetics
14.
J Bacteriol ; 191(3): 1044-55, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19028886

ABSTRACT

Porphyromonas gingivalis is an anaerobic, asaccharolytic, gram-negative bacterium that has essential requirements for both iron and protoporphyrin IX, which it preferentially obtains as heme. A combination of large-scale quantitative proteomic analysis using stable isotope labeling strategies and mass spectrometry, together with transcriptomic analysis using custom-made DNA microarrays, was used to identify changes in P. gingivalis W50 protein and transcript abundances on changing from heme-excess to heme-limited continuous culture. This approach identified 160 genes and 70 proteins that were differentially regulated by heme availability, with broad agreement between the transcriptomic and proteomic data. A change in abundance of the enzymes of the aspartate and glutamate catabolic pathways was observed with heme limitation, which was reflected in organic acid end product levels of the culture fluid. These results demonstrate a shift from an energy-efficient anaerobic respiration to a less efficient process upon heme limitation. Heme limitation also resulted in an increase in abundance of a protein, PG1374, which we have demonstrated, by insertional inactivation, to have a role in epithelial cell invasion. The greater abundance of a number of transcripts/proteins linked to invasion of host cells, the oxidative stress response, iron/heme transport, and virulence of the bacterium indicates that there is a broad response of P. gingivalis to heme availability.


Subject(s)
Heme/pharmacology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromatography, Liquid , Culture Media/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Mass Spectrometry , Mutation , Oligonucleotide Array Sequence Analysis , Porphyromonas gingivalis/growth & development , Proteomics/methods , Transcription, Genetic/drug effects
15.
Infect Immun ; 75(3): 1436-42, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17220315

ABSTRACT

The contributions of three proteinase genes (rgpA, rgpB, and kgp) to the virulence of Porphyromonas gingivalis W50 were investigated in the murine periodontitis model. Mice were orally inoculated with eight doses (1 x 10(10) cells per dose) of rgpA, rgpB, kgp, rgpA rgpB, or rgpA rgpB kgp isogenic mutants, and the level of alveolar bone loss, immune response induced, and number of bacterial cells per half maxilla were compared with those of animals inoculated with wild-type P. gingivalis. The kgp, rgpB, rgpA rgpB, and rgpA rgpB kgp isogenic mutants induced significantly (P < 0.05) less bone loss than the rgpA isogenic mutant and the wild type did, and the virulence of the rgpA isogenic mutant and the wild type were not significantly different. Mice inoculated with the wild type or the rgpA isogenic mutant exhibited significantly (P < 0.01) more P. gingivalis cells per half maxilla than mice inoculated with rgpB, kgp, rgpA rgpB, and rgpA rgpB kgp isogenic mutants or nonchallenged mice did, as determined using real-time PCR. A significant positive correlation was found between the number of P. gingivalis cells detected per half maxilla and the amount of alveolar bone loss induced. Enzyme-linked immunosorbent assay results showed that each isogenic mutant and the wild type induced a predominant P. gingivalis antigen-specific immunoglobulin G3 (IgG3) response. Furthermore, the kgp and rgpA rgpB kgp isogenic mutants induced significantly (P < 0.05) lower IgG3 antibody responses than the responses induced by the wild type or the rgpA, rgpB, and rgpA rgpB isogenic mutants. The results suggest that the order in which the proteinases contribute to the virulence of P. gingivalis in the murine periodontitis model is Kgp > or = RgpB >> RgpA.


Subject(s)
Adhesins, Bacterial/physiology , Bacteroidaceae Infections/microbiology , Cysteine Endopeptidases/physiology , Periodontitis/microbiology , Porphyromonas gingivalis/pathogenicity , Alveolar Bone Loss/microbiology , Animals , Bacteroidaceae Infections/metabolism , Disease Models, Animal , Gingipain Cysteine Endopeptidases , Mice , Periodontitis/metabolism , Porphyromonas gingivalis/genetics , Virulence
16.
J Bacteriol ; 188(17): 6376-86, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16923905

ABSTRACT

Porphyromonas gingivalis produces outer membrane-attached proteins that include the virulence-associated proteinases RgpA and RgpB (Arg-gingipains) and Kgp (Lys-gingipain). We analyzed the P. gingivalis outer membrane proteome and identified numerous proteins with C-terminal domains similar in sequence to those of RgpB, RgpA, and Kgp, indicating that these domains may have a common function. Using RgpB as a model to investigate the role of the C-terminal domain, we expressed RgpB as a full-length zymogen (recombinant RgpB [rRgpB]), with a catalytic Cys244Ala mutation [rRgpB(C244A)], or with the C-terminal 72 amino acids deleted (rRgpB435) in an Arg-gingipain P. gingivalis mutant (YH522AB) and an Arg- and Lys-gingipain mutant (YH522KAB). rRgpB was catalytically active and located predominantly attached to the outer membrane of both background strains. rRgpB(C244A) was inactive and outer membrane attached, with a typical attachment profile for both background strains according to sodium dodecyl sulfate-polyacrylamide gel electrophoresis, but in YH522KAB, the prodomain was not removed. Thus, in vivo, RgpB export and membrane attachment are independent of the proteolytic activity of RgpA, RgpB, or Kgp. However, for maturation involving proteolytic processing of RgpB, the proteolytic activity of RgpB, RgpA, or Kgp is required. The C-terminally-truncated rRgpB435 was not attached to the outer membrane and was located as largely inactive, discrete 71-kDa and 48-kDa isoforms in the culture supernatant and the periplasm. These results suggest that the C-terminal domain is essential for outer membrane attachment and may be involved in a coordinated process of export and attachment to the cell surface.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Cysteine Endopeptidases/metabolism , Hemagglutinins/metabolism , Porphyromonas gingivalis/physiology , Protein Processing, Post-Translational , Adhesins, Bacterial/genetics , Amino Acid Sequence , Bacterial Adhesion , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Gingipain Cysteine Endopeptidases , Hemagglutinins/chemistry , Hemagglutinins/genetics , Molecular Sequence Data , Molecular Weight , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/physiology , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
17.
Vaccine ; 24(42-43): 6542-54, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-16839648

ABSTRACT

Porphyromonas gingivalis is a pathogen associated with chronic periodontitis, an inflammatory disease of the supporting tissues of the teeth. A major virulence factor for P. gingivalis is the RgpA-Kgp proteinase-adhesin complex. We have prepared the following recombinant proteins corresponding to domains/regions of the RgpA-Kgp complex; rRgpA(cat), rRgpA(A1), rRgpA(A2), rRgpA(A4), rRgpA(A1)(784-1009), rRgpA(A1)(911-1009), rKgp(A1) and rKgp(A1)(759-989). The ability of each recombinant protein to attenuate P. gingivalis infection, when used as a vaccine, in the murine lesion model was determined. All of the recombinant adhesin domains were found to significantly attenuate P. gingivalis infection with the most effective being rKgp(A1) and rKgp(A1)(759-989) followed by rRgpA(A1), rRgpA(A1)(784-1009) and rRgpA(A1)(911-1009). The predominant antibody isotype was IgG1. The A1 adhesins, which gave the best protection, contain specific motifs implicated in binding to host tissue. Immunisation with rRgpA(cat) had no effect on P. gingivalis infection. As well as detecting the Kgp(A1) adhesin in a Western blot, the rKgp(A1) and rKgp(A1)(759-989) antisera were also immunoreactive with the A1 and A3 adhesins of RgpA and HagA. Flow cytometric analysis indicated that rKgp(A1) and rRgpA(A1) antisera recognised native antigen on P. gingivalis whole cells. Furthermore, the rKgp(A1) and rKgp(A1)(759-989) antisera exhibited a similar immunoreactive profile with outer membrane preparations of all P. gingivalis serotypes and clinical isolates tested. The recombinant A1 adhesin therefore has potential in the development of a P. gingivalis vaccine.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Vaccines/immunology , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/prevention & control , Porphyromonas gingivalis/immunology , Animals , Bacterial Outer Membrane Proteins/immunology , Blotting, Western , Catalysis , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunization , Mice , Mice, Inbred BALB C , Peptide Hydrolases/immunology , Peptide Mapping , Vaccines, Synthetic/immunology
18.
J Bacteriol ; 188(7): 2454-62, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16547032

ABSTRACT

Porphyromonas gingivalis is an anaerobic microorganism that inhabits the oral cavity, where oxidative stress represents a constant challenge. A putative transcriptional regulator associated with oxidative stress, an oxyR homologue, is known from the P. gingivalis W83 genome sequence. We used microarrays to characterize the response of P. gingivalis to H2O2 and examine the role of oxyR in the regulation of this response. Most organisms in which oxyR has been investigated are facultative anaerobes or aerobes. In contrast to the OxyR-regulated response of these microorganisms to H2O2, the main feature of the response in P. gingivalis was a concerted up-regulation of insertion sequence elements related to IS1 transposases. Common OxyR-regulated genes such as dps and ahpFC were not positively regulated in P. gingivalis in response to H2O2. However, their expression was dependent on the presence of a functional OxyR, as revealed by microarray comparison of an oxyR mutant to the wild type. Phenotypic characterization of the oxyR mutant showed that OxyR plays a role in both the resistance to H2O2 and the aerotolerance of P. gingivalis. Escherichia coli and other bacteria with more complex respiratory requirements use OxyR for regulating resistance to H2O2 and use a separate regulator for aerotolerance. In P. gingivalis, the presence of a single protein combining the two functions might be related to the comparatively smaller genome size of this anaerobic microorganism. In conclusion, these results suggest that OxyR does not act as a sensor of H2O2 in P. gingivalis but constitutively activates transcription of oxidative-stress-related genes under anaerobic growth.


Subject(s)
Mouth/microbiology , Porphyromonas gingivalis/metabolism , Anaerobiosis , Antioxidants/metabolism , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Hydrogen Peroxide , Molecular Sequence Data , Mutagenesis, Insertional , Porphyromonas gingivalis/genetics
19.
J Biol Chem ; 277(26): 23433-40, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-11976326

ABSTRACT

In a search for a basic carboxypeptidase that might work in concert with the major virulence factors, the Arg- and Lys-specific cysteine endoproteinases of Porphyromonas gingivalis, a novel 69.8-kDa metallocarboxypeptidase CPG70 was purified to apparent homogeneity from the culture fluid of P. gingivalis HG66. Carboxypeptidase activity was measured by matrix-assisted laser desorption ionization-mass spectrometry using peptide substrates derived from a tryptic digest of hemoglobin. CPG70 exhibited activity with peptides containing C-terminal Lys and Arg residues. The k(cat)/K(m) values for the hydrolysis of the synthetic dipeptides FA-Ala-Lys and FA-Ala-Arg by CPG70 were 99 and 56 mm(-1)s(-1), respectively. The enzyme activity was strongly inhibited by the Arg analog (2-guanidinoethylmercapto)succinic acid and 1,10-phenanthroline. High resolution inductively coupled plasma-mass spectrometry demonstrated that 1 mol of CPG70 was associated with 0.6 mol of zinc, 0.2 mol of nickel, and 0.2 mol of copper. A search of the P. gingivalis W83 genomic data base (TIGR) with the N-terminal amino acid sequence determined for CPG70 revealed that the enzyme is an N- and C-terminally truncated form of a predicted 91.5-kDa protein (PG0232). Analysis of the deduced amino acid sequence of the full-length protein revealed an N-terminal signal sequence followed by a pro-segment, a metallocarboxypeptidase catalytic domain, three tandem polycystic kidney disease domains, and an 88-residue C-terminal segment. The catalytic domain exhibited the highest sequence identity with the duck metallocarboxypeptidase D domain II. Insertional inactivation of the gene encoding CPG70 resulted in a P. gingivalis isogenic mutant that was avirulent in the murine lesion model under the conditions tested.


Subject(s)
Carboxypeptidases/isolation & purification , Polycystic Kidney Diseases/etiology , Porphyromonas gingivalis/enzymology , Amino Acid Sequence , Animals , Carboxypeptidases/chemistry , Carboxypeptidases/toxicity , Catalytic Domain , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Porphyromonas gingivalis/pathogenicity
20.
Infect Immun ; 70(5): 2480-6, 2002 May.
Article in English | MEDLINE | ID: mdl-11953385

ABSTRACT

A major virulence factor of Porphyromonas gingivalis is the extracellular noncovalently associated complexes of Arg-X- and Lys-X-specific cysteine proteinases and adhesins designated the RgpA-Kgp complexes. In this study we investigated the ability of RgpA-Kgp as an immunogen to protect against P. gingivalis-induced periodontal bone loss in the rat. Specific-pathogen-free Sprague-Dawley rats were immunized with either formalin-killed whole P. gingivalis ATCC 33277 cells with incomplete Freund's adjuvant, RgpA-Kgp with incomplete Freund's adjuvant, or incomplete Freund's adjuvant alone. The animals were then challenged by oral inoculation with live P. gingivalis ATCC 33277 cells. Marked periodontal bone loss was observed in animals immunized with incomplete Freund's adjuvant alone; this bone loss was significantly (P < 0.05) greater than that detected in animals immunized with formalin-killed whole cells or RgpA-Kgp or in unchallenged animals. There was no significant difference in periodontal bone loss between animals immunized with formalin-killed whole cells and those immunized with RgpA-Kgp. The bone loss in these animals was also not significantly different from that in unchallenged animals. DNA probe analysis of subgingival plaque samples showed that 100% of the animals immunized with incomplete Freund's adjuvant alone and challenged with P. gingivalis ATCC 33277 were positive for the bacterium. However, P. gingivalis ATCC 33277 could not be detected in subgingival plaque samples from animals immunized with formalin-killed whole cells or with RgpA-Kgp. Immunization with formalin-killed whole cells or RgpA-Kgp induced a high-titer serum immunoglobulin G2a response. Western blot analysis of RgpA-Kgp using pooled protective antisera taken from rats immunized with RgpA-Kgp revealed immunodominant bands at 44, 39, and 27 kDa. In conclusion, immunization with RgpA-Kgp restricted colonization by P. gingivalis and periodontal bone loss in the rat.


Subject(s)
Alveolar Bone Loss/prevention & control , Bacterial Vaccines/immunology , Cysteine Endopeptidases/immunology , Hemagglutinins/immunology , Periodontitis/complications , Porphyromonas gingivalis/immunology , Adhesins, Bacterial , Animals , Antibodies, Bacterial/biosynthesis , Gingipain Cysteine Endopeptidases , Gingival Crevicular Fluid/microbiology , Immunization , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin G/classification , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...