Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
2.
J Dent Res ; 99(6): 604-612, 2020 06.
Article in English | MEDLINE | ID: mdl-32091956

ABSTRACT

Oral cancer, predominantly oral squamous cell carcinoma (OSCC), is the eighth-most common cancer worldwide, with a 5-y survival rate <50%. There are numerous risk factors for oral cancer, among which periodontal disease is gaining increasing recognition. The creation of a sustained dysbiotic proinflammatory environment by periodontal bacteria may serve to functionally link periodontal disease and oral cancer. Moreover, traditional periodontal pathogens, such as Porphyromonas gingivalis, Fusobacterium nucleatum, and Treponema denticola, are among the species most frequently identified as being enriched in OSCC, and they possess a number of oncogenic properties. These organisms share the ability to attach and invade oral epithelial cells, and from there each undergoes its own unique molecular dialogue with the host epithelium, which ultimately converges on acquired phenotypes associated with cancer, including inhibition of apoptosis, increased proliferation, and activation of epithelial-to-mesenchymal transition leading to increased migration of epithelial cells. Additionally, emerging properties of structured bacterial communities may increase oncogenic potential, and consortia of P. gingivalis and F. nucleatum are synergistically pathogenic within in vivo oral cancer models. Interestingly, however, some species of oral streptococci can antagonize the phenotypes induced by P. gingivalis, indicating functionally specialized roles for bacteria in oncogenic communities. Transcriptomic data support the concept that functional, rather than compositional, properties of oral bacterial communities have more relevance to cancer development. Collectively, the evidence is consistent with a modified polymicrobial synergy and dysbiosis model for bacterial involvement in OSCC, with driver mutations generating a conducive microenvironment on the epithelial boundary, which becomes further dysbiotic by the synergistic action of bacterial communities.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Fusobacterium nucleatum , Humans , Porphyromonas gingivalis , Tumor Microenvironment
3.
J Dent Res ; 98(12): 1308-1314, 2019 11.
Article in English | MEDLINE | ID: mdl-31356756

ABSTRACT

Oral supra- and subgingival biofilms are complex communities in which hundreds of bacteria, viruses, and fungi reside and interact. In these social environments, microbes compete and cooperate for resources, such as living space and nutrients. The metabolic activities of bacteria can transform their microenvironment and dynamically influence the fitness and growth of cohabitating organisms. Biofilm communities are temporally and spatially organized largely due to cell-to-cell communication, which promotes synergistic interactions. Metabolic interactions maintain biofilm homeostasis through mutualistic cross-feeding, metabolic syntrophy, and cross-respiration. These interactions include reciprocal metabolite exchanges that promote the growth of physiologically compatible bacteria, processive catabolism of complex substrates, and unidirectional interactions that are globally important for the polymicrobial community. Additionally, oral bacterial interactions can lead to detoxification of oxidative compounds, which will provide protection to the community at large. It has also been established that specific organisms provide terminal electron acceptors to partner species that result in a shift from fermentation to respiration, thus increasing ATP yields and improving fitness. Indeed, many interspecies relationships are multidimensional, and the net outcome can be spatially and temporally dependent. Cross-kingdom interactions also occur as oral yeast are antagonistic to some oral bacteria, while numerous mutualistic interactions contribute to yeast-bacterial colonization, fitness in the oral community, and the pathogenesis of caries. Consideration of this social environment reveals behaviors and phenotypes that are not apparent through the study of microbes in isolation. Here, we provide a comprehensive overview of the metabolic interactions that shape the oral microbial community.


Subject(s)
Bacteria/metabolism , Biofilms , Microbial Interactions , Microbiota , Mouth/microbiology , Humans , Signal Transduction , Yeasts/metabolism
4.
Mol Oral Microbiol ; 33(5): 364-377, 2018 10.
Article in English | MEDLINE | ID: mdl-29939498

ABSTRACT

Acinetobacter baumannii is a nosocomial, opportunistic pathogen that causes several serious conditions including meningitis, septicemia, endocarditis, and pneumonia. It can be found in the oral biofilm, which may be a reservoir for pneumonia and chronic obstructive pulmonary disease. Subgingival colonization by A. baumannii is associated with chronic and aggressive periodontitis as well as refractory periodontal disease. Porphyromonas gingivalis, a keystone periodontal pathogen localized to subgingival plaque, is also implicated in several chronic conditions including aspiration pneumonia. Although both bacteria are found together in subgingival plaque and can cause multiple polymicrobial infections, nothing is known about the interactions between these two important human pathogens. In this study, we used RNA sequencing to understand the transcriptional response of both species as they adapt to heterotypic communities. Among the differentially regulated genes were those encoding a number of important virulence factors for both species including adhesion, biofilm formation, and protein secretion. Additionally, the presence of A. baumannii increased the abundance of P. gingivalis in model dual-species communities. Collectively these results suggest that both P. gingivalis and A. baumannii adapt to each other and have synergistic potential for increased pathogenicity. In identifying the mechanisms that promote pathogenicity and refractory disease, novel approaches to mitigate polymicrobial synergistic interactions may be developed to treat or prevent associated diseases.


Subject(s)
Acinetobacter baumannii/genetics , Adhesins, Bacterial/physiology , Biofilms/growth & development , Microbial Interactions , Porphyromonas gingivalis/genetics , Acinetobacter baumannii/pathogenicity , Adhesins, Bacterial/genetics , Dental Plaque/microbiology , Gene Expression Profiling , Humans , Porphyromonas gingivalis/pathogenicity , Sequence Analysis, RNA , Virulence Factors/metabolism
6.
Infect Immun ; 85(7)2017 07.
Article in English | MEDLINE | ID: mdl-28438978

ABSTRACT

Peptoanaerobacter stomatis is a newly appreciated taxon associated with periodontal diseases; however, little is known about the organism's pathogenic potential or its interaction with the host immune response. Neutrophils are the most abundant innate immune cell present in the gingival tissue and function to constrain the oral microbial challenge. However, some periodontal pathogens have developed strategies to evade phagocytosis and killing by neutrophils. Therefore, to begin to understand the role of P. stomatis in periodontitis, we studied its interactions with human neutrophils. Our data showed that after 30 min of incubation, neutrophils failed to engulf P. stomatis efficiently; however, when P. stomatis was internalized, it was promptly eradicated. P. stomatis challenge induced a robust intracellular respiratory burst; however, this response did not contribute to bacterial killing. Minimal superoxide release was observed by direct bacterial challenge; however, P. stomatis significantly increased N-formyl-methionyl-leucyl phenylalanine (fMLF)-stimulated superoxide release to an extent similar to that of cells primed with tumor necrosis factor alpha (TNF-α). When neutrophils were challenged with P. stomatis, 52% of the bacterium-containing phagosomes were enriched for the specific granule marker lactoferrin and 82% with the azurophil granule marker elastase. P. stomatis challenge stimulated exocytosis of the four neutrophil granule subtypes. Moreover, P. stomatis susceptibility to extracellular killing could be attributed to the exocytosis of antimicrobial components present in neutrophil granules. Priming neutrophils for an enhanced respiratory burst together with promoting granule content release could contribute to the chronic inflammation and tissue destruction that characterize periodontal diseases.


Subject(s)
Clostridiales/immunology , Cytoplasmic Granules/metabolism , Exocytosis , Neutrophils/immunology , Humans , Respiratory Burst , Superoxides/metabolism
7.
Mol Oral Microbiol ; 32(4): 314-323, 2017 08.
Article in English | MEDLINE | ID: mdl-27498608

ABSTRACT

The development of synergistically pathogenic communities of Porphyromonas gingivalis and Streptococcus gordonii is controlled by a tyrosine-phosphorylation-dependent signaling pathway in P. gingivalis. The Ptk1 bacterial tyrosine (BY) kinase of P. gingivalis is required for maximal community development and for the production of extracellular polysaccharide. We show that the consensus BY kinase Walker A and B domains, the RK cluster, and the YC domain of Ptk1 are necessary for autophosphorylation and for substrate phosphorylation. Mass spectrometry showed that six tyrosine residues in a 16-amino-acid C-terminal region were phosphorylated in recombinant (r) Ptk1. Complementation of a ptk1 mutant with the wild-type ptk1 allele in trans restored community development between P. gingivalis and S. gordonii, and extracellular polysaccharide production by P. gingivalis. In contrast, complementation of Δptk1 with ptk1 containing a mutation in the Walker A domain failed to restore community development or extracellular polysaccharide production. rPtk1 was capable of phosphorylating the tyrosine phosphatase Ltp1 and the transcriptional regulator CdhR, both of which are involved in the development of P. gingivalis communities with S. gordonii.


Subject(s)
Porphyromonas gingivalis/enzymology , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Biofilms/growth & development , Gene Expression Regulation, Bacterial , Microbial Interactions , Mutation , Phosphorylation , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Streptococcus gordonii/genetics , Streptococcus gordonii/physiology , Structure-Activity Relationship
8.
J Dent Res ; 95(11): 1291-7, 2016 10.
Article in English | MEDLINE | ID: mdl-27323953

ABSTRACT

Fimbriae are protein-based filamentous appendages that protrude from the bacterial cell surface and facilitate host adhesion. Two types of fimbriae, FimA and Mfa1, of the periodontal pathogen Porphyromonas gingivalis are responsible for adherence to other bacteria and to host cells in the oral cavity. Both fimbrial forms are composed of 5 proteins, but there is limited information about their polymerization mechanisms. Here, the authors evaluated the function of Mfa5, one of the Mfa1 fimbrial accessory proteins. Using mfa5 gene disruption and complementation studies, the authors revealed that Mfa5 affects the incorporation of other accessory proteins, Mfa3 and Mfa4, into fibers and the expression of fimbriae on the cell surface. Mfa5 is predicted to have a C-terminal domain (CTD) that uses the type IX secretion system (T9SS), which is limited to this organism and related Bacteroidetes species, for translocation across the outer membrane. To determine the relationship between the putative Mfa5 CTD and the T9SS, mutants were constructed with in-frame deletion of the CTD and deletion of porU, a C-terminal signal peptidase linked to T9SS-mediated secretion. The ∆CTD-expressing strain presented a similar phenotype to the mfa5 disruption mutant with reduced expression of fimbriae lacking all accessory proteins. The ∆porU mutants and the ∆CTD-expressing strain showed intracellular accumulation of Mfa5. These results indicate that Mfa5 function requires T9SS-mediated translocation across the outer membrane, which is dependent on the CTD, and subsequent incorporation into fibers. These findings suggest the presence of a novel polymerization mechanism of the P. gingivalis fimbriae.


Subject(s)
Fimbriae Proteins/physiology , Porphyromonas gingivalis/physiology , Bacterial Adhesion/physiology , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Fimbriae Proteins/genetics , Fimbriae Proteins/isolation & purification , Gene Expression Regulation, Bacterial/physiology , Mutation/genetics , Porphyromonas gingivalis/genetics
9.
Mol Oral Microbiol ; 31(4): 354-64, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26358096

ABSTRACT

Porphyromonas gingivalis is a Gram-negative anaerobe and keystone periodontal pathogen. A mariner transposon insertion mutant library has recently been used to define 463 genes as putatively essential for the in vitro growth of P. gingivalis ATCC 33277 in planktonic culture (Library 1). We have independently generated a transposon insertion mutant library (Library 2) for the same P. gingivalis strain and herein compare genes that are putatively essential for in vitro growth in complex media, as defined by both libraries. In all, 281 genes (61%) identified by Library 1 were common to Library 2. Many of these common genes are involved in fundamentally important metabolic pathways, notably pyrimidine cycling as well as lipopolysaccharide, peptidoglycan, pantothenate and coenzyme A biosynthesis, and nicotinate and nicotinamide metabolism. Also in common are genes encoding heat-shock protein homologues, sigma factors, enzymes with proteolytic activity, and the majority of sec-related protein export genes. In addition to facilitating a better understanding of critical physiological processes, transposon-sequencing technology has the potential to identify novel strategies for the control of P. gingivalis infections. Those genes defined as essential by two independently generated TnSeq mutant libraries are likely to represent particularly attractive therapeutic targets.


Subject(s)
Bacterial Proteins/genetics , DNA Transposable Elements , Gene Library , Genes, Bacterial , Heat-Shock Proteins/genetics , Porphyromonas gingivalis/genetics , Sigma Factor/genetics , Chromosome Mapping/methods , Genes, Essential , High-Throughput Nucleotide Sequencing/methods , Lipopolysaccharides/biosynthesis , Mutagenesis, Insertional , Mutation , Periodontal Diseases/microbiology , Porphyromonas gingivalis/growth & development , Pyrimidines/metabolism
10.
Mol Oral Microbiol ; 31(2): 136-61, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26042999

ABSTRACT

Recent studies have shown that the transcriptional landscape of the pleiomorphic fungus Candida albicans is highly dependent upon growth conditions. Here using a dual RNA-seq approach we identified 299 C. albicans and 72 Streptococcus gordonii genes that were either upregulated or downregulated specifically as a result of co-culturing these human oral cavity microorganisms. Seventy-five C. albicans genes involved in responses to chemical stimuli, regulation, homeostasis, protein modification and cell cycle were significantly (P ≤ 0.05) upregulated, whereas 36 genes mainly involved in transport and translation were downregulated. Upregulation of filamentation-associated TEC1 and FGR42 genes, and of ALS1 adhesin gene, concurred with previous evidence that the C. albicans yeast to hypha transition is promoted by S. gordonii. Increased expression of genes required for arginine biosynthesis in C. albicans was potentially indicative of a novel oxidative stress response. The transcriptional response of S. gordonii to C. albicans was less dramatic, with only eight S. gordonii genes significantly (P ≤ 0.05) upregulated at least two-fold (glpK, rplO, celB, rplN, rplB, rpsE, ciaR and gat). The expression patterns suggest that signals from S. gordonii cause a positive filamentation response in C. albicans, whereas S. gordonii appears to be transcriptionally less influenced by C. albicans.


Subject(s)
Candida albicans/genetics , Mouth/microbiology , Streptococcus gordonii/genetics , Adhesins, Bacterial/genetics , Bacterial Adhesion/genetics , Biofilms , Candida albicans/physiology , Candida albicans/ultrastructure , DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Fungal , Microbial Interactions , Streptococcus gordonii/physiology , Streptococcus gordonii/ultrastructure , Transcription Factors/genetics , Transcriptome
11.
Mol Oral Microbiol ; 30(1): 2-15, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25052812

ABSTRACT

Actinetobacter baumannii is an important nosocomial pathogen that can cause a wide range of serious conditions including pneumonia, meningitis, necrotizing fasciitis and sepsis. It is also a major cause of wound infections in military personnel injured during the conflicts in Afghanistan and Iraq, leading to its popular nickname of 'Iraqibacter'. Contributing to its success in clinical settings is resistance to environmental stresses such as desiccation and disinfectants. Moreover, in recent years there has been a dramatic increase in the number of A. baumannii strains with resistance to multiple antibiotic classes. Acinetobacter baumannii is an inhabitant of oral biofilms, which can act as a reservoir for pneumonia and chronic obstructive pulmonary disease. Subgingival colonization by A. baumannii increases the risk of refractory periodontitis. Pathogenesis of the organism involves adherence, biofilm formation and iron acquisition. In addition, A. baumannii can induce apoptotic cell death in epithelial cells and kill hyphal forms of Candida albicans. Virulence factors that have been identified include pili, the outer membrane protein OmpA, phospholipases and extracellular polysaccharide. Acinetobacter baumannii can sense blue light through a blue-light sensing using flavin (BLUF) domain protein, BlsA. The resulting conformational change in BlsA leads to changes in gene expression, including virulence genes.


Subject(s)
Acinetobacter Infections/microbiology , Acinetobacter baumannii , Cross Infection/microbiology , Mouth/microbiology , Periodontitis/microbiology , Virulence Factors/physiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Acinetobacter baumannii/physiology , Apoptosis , Bacterial Adhesion , Biofilms/growth & development , Drug Resistance, Bacterial , Humans , Iron/metabolism , Light , Microbial Interactions , Virulence/genetics , Virulence/physiology
13.
Mol Oral Microbiol ; 29(5): 185-93, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24899524

ABSTRACT

Porphyromonas gingivalis is one of the main etiological organisms in periodontal disease. On oral surfaces P. gingivalis is a component of multispecies biofilm communities and can modify the pathogenic potential of the community as a whole. Accumulation of P. gingivalis in communities is facilitated by interspecies binding and communication with the antecedent colonizer Streptococcus gordonii. In this study we screened a library of small molecules to identify structures that could serve as lead compounds for the development of inhibitors of P. gingivalis community development. Three small molecules were identified that effectively inhibited accumulation of P. gingivalis on a substratum of S. gordonii. The structures of the small molecules are derived from the marine alkaloids oroidin and bromoageliferin and contain a 2-aminoimidazole or 2-aminobenzimidazole moiety. The most active compounds reduced expression of mfa1 and fimA in P. gingivalis, genes encoding the minor and major fimbrial subunits, respectively. These fimbrial adhesins are necessary for P. gingivalis co-adhesion with S. gordonii. These results demonstrate the potential for a small molecular inhibitor-based approach to the prevention of diseases associated with P. gingivalis.


Subject(s)
Bacterial Adhesion/drug effects , Biofilms/drug effects , Porphyromonas gingivalis/drug effects , Small Molecule Libraries , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/drug effects , Benzimidazoles/pharmacology , Carbon-Sulfur Lyases/drug effects , Fimbriae Proteins/antagonists & inhibitors , Humans , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Imidazoles/pharmacology , Microbial Interactions , Microscopy, Confocal/methods , Pili, Sex/drug effects , Porphyromonas gingivalis/physiology , Pyrroles/pharmacology , Streptococcus gordonii/physiology
14.
Mol Oral Microbiol ; 28(6): 467-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24118823

ABSTRACT

The fimbriae of Porphyromonas gingivalis, the causative agent of periodontitis, have been implicated in various aspects of pathogenicity, such as colonization, adhesion and aggregation. Porphyromonas gingivalis ATCC 33277 has two adhesins comprised of the FimA and Mfa1 fimbriae. We characterized the PGN0289 (Mfa3) protein, which is one of the three accessory proteins of Mfa1 fimbriae in P. gingivalis. The Mfa3 protein was present in two different sizes, 40 and 43 kDa, in the cell. The 43-kDa and 40-kDa Mfa3 were detected largely in the inner membrane and the outer membrane, respectively. Purified Mfa1 fimbriae contained the 40-kDa Mfa3 alone. Furthermore, the 40-kDa Mfa3 started with the Ala(44) residue of the deduced amino acid sequence, indicating that the N-terminal region of the nascent protein expressed from the mfa3 gene is processed in the transport step from the inner membrane into fimbriae. Immuno-electron microscopy revealed that Mfa3 localized at the tip of the fimbrial shaft. Interestingly, deletion of the mfa3 gene resulted in the absence of other accessory proteins, PGN0290 and PGN0291, in the purified Mfa1 fimbriae, suggesting that Mfa3 is required for integration of PGN0290 and PGN0291 into fimbriae. A double mutant of mfa3 and fimA genes (phenotype Mfa1 plus, FimA minus) showed increased auto-aggregation and biofilm formation similar to a double mutant of mfa1 and fimA genes (phenotype Mfa1(-) , FimA(-) ). These findings suggest that the tip protein Mfa3 of the Mfa1 fimbriae may function in the integration of accessory proteins and in the colonization of P. gingivalis.


Subject(s)
Bacterial Proteins/analysis , Fimbriae Proteins/analysis , Fimbriae Proteins/physiology , Fimbriae, Bacterial/physiology , Porphyromonas gingivalis/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biofilms/growth & development , Fimbriae Proteins/chemistry , Fimbriae Proteins/genetics , Fimbriae, Bacterial/chemistry , Mutation , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/ultrastructure
15.
J Periodontal Res ; 48(6): 713-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23441958

ABSTRACT

BACKGROUND AND OBJECTIVE: Gingival keratinocytes are used in model systems to investigate the interaction between periodontal bacteria and the epithelium in the initial stages of the periodontal disease process. Primary gingival epithelial cells (GECs) have a finite lifespan in culture before they enter senescence and cease to replicate, while epithelial cells immortalized with viral proteins can exhibit chromosomal rearrangements. The aim of this study was to generate a telomerase immortalized human gingival epithelial cell line and compare its in vitro behaviour to that of human GECs. MATERIAL AND METHODS: Human primary GECs were immortalized with a bmi1/hTERT combination to prevent cell cycle triggers of senescence and telomere shortening. The resultant cell-line, telomerase immortalized gingival keratinocytes (TIGKs), were compared to GECs for cell morphology, karyotype, growth and cytokeratin expression, and further characterized for replicative lifespan, expression of toll-like receptors and invasion by P. gingivalis. RESULTS: TIGKs showed morphologies, karyotype, proliferation rates and expression of characteristic cytokeratin proteins comparable to GECs. TIGKs underwent 36 passages without signs of senescence and expressed transcripts for toll-like receptors 1-6, 8 and 9. A subpopulation of cells underwent stratification after extended time in culture. The cytokeratin profiles of TIGK monolayers were consistent with basal cells. When allowed to stratify, cytokeratin profiles of TIGKs were consistent with suprabasal cells of the junctional epithelium. Further, TIGKs were comparable to GECs in previously reported levels and kinetics of invasion by wild-type P. gingivalis and an invasion defective ΔserB mutant. CONCLUSION: Results confirm bmi1/hTERT immortalization of primary GECs generated a robust cell line with similar characteristics to the parental cell type. TIGKs represent a valuable model system for the study of oral bacteria interactions with host gingival cells.


Subject(s)
Gingiva/cytology , Adult , Cell Count , Cell Culture Techniques , Cell Line, Transformed , Cell Proliferation , Cell Shape/physiology , Cellular Senescence/physiology , Epithelial Attachment/cytology , Epithelial Cells/cytology , Epithelial Cells/microbiology , Gingiva/microbiology , Humans , Karyotype , Keratin-13/metabolism , Keratin-14/metabolism , Keratin-19/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Polycomb Repressive Complex 1/genetics , Porphyromonas gingivalis/physiology , Retroviridae/genetics , Telomerase/genetics , Telomere/physiology , Toll-Like Receptors/metabolism , Transduction, Genetic , Zinc Fingers/genetics
16.
Mol Oral Microbiol ; 28(1): 70-81, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23194602

ABSTRACT

The periodontal pathogen Porphyromonas gingivalis experiences a number of environmental conditions in the oral cavity, and must monitor and respond to a variety of environmental cues. However, the organism possesses only five full two-component systems, one of which is the hybrid system GppX. To investigate the regulon controlled by GppX we performed RNA-Seq on a ΔGppX mutant. Fifty-three genes were upregulated and 37 genes were downregulated in the ΔGppX mutant. Pathway analyses revealed no systemic function for GppX under nutrient-replete conditions; however, over 40% of the differentially abundant genes were annotated as encoding hypothetical proteins indicating a novel role for GppX. Abundance of small RNA was, in general, not affected by the absence of GppX. To further define the role of GppX with respect to regulation of a hypothetical protein observed with the greatest significant relative abundance change relative to a wild-type control, PGN_0151, we constructed a series of strains in which the ΔgppX mutation was complemented with a GppX protein containing specific domain and phosphotransfer mutations. The transmembrane domains, the DNA-binding domain and the phosphotransfer residues were all required for regulation of PGN_0151. In addition, binding of GppX to the PGN_0151 promoter regions was confirmed by an electrophoretic mobility shift assay. Both the ΔGppX mutant and a ΔPGN_0151 mutant were deficient in monospecies biofilm formation, suggesting a role for the GppX-PGN_0151 regulon in colonization and survival of the organism.


Subject(s)
Bacterial Proteins/genetics , Mutation/genetics , Porphyromonas gingivalis/genetics , Regulon/genetics , Alleles , AraC Transcription Factor/genetics , Aspartic Acid/genetics , Bacteriological Techniques , Biofilms/growth & development , Gene Expression Profiling , Gene Expression Regulation, Bacterial/genetics , Genes, Bacterial/genetics , Helix-Turn-Helix Motifs/genetics , Histidine Kinase , Humans , Membrane Proteins/genetics , Porphyromonas gingivalis/physiology , Promoter Regions, Genetic/genetics , Protein Kinases/genetics , Sequence Analysis, RNA , Signal Transduction/genetics
17.
Mol Oral Microbiol ; 28(2): 83-101, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23253299

ABSTRACT

Establishment of a community is considered to be essential for microbial growth and survival in the human oral cavity. Biofilm communities have increased resilience to physical forces, antimicrobial agents and nutritional variations. Specific cell-to-cell adherence processes, mediated by adhesin-receptor pairings on respective microbial surfaces, are able to direct community development. These interactions co-localize species in mutually beneficial relationships, such as streptococci, veillonellae, Porphyromonas gingivalis and Candida albicans. In transition from the planktonic mode of growth to a biofilm community, microorganisms undergo major transcriptional and proteomic changes. These occur in response to sensing of diffusible signals, such as autoinducer molecules, and to contact with host tissues or other microbial cells. Underpinning many of these processes are intracellular phosphorylation events that regulate a large number of microbial interactions relevant to community formation and development.


Subject(s)
Biofilms , Microbial Consortia/physiology , Mouth/microbiology , Candida albicans/physiology , Humans , Microbial Interactions/physiology , Microbial Viability , Porphyromonas gingivalis/physiology , Proteome/physiology , Quorum Sensing/physiology , Streptococcus/physiology , Transcriptome/physiology
18.
Mol Oral Microbiol ; 27(6): 409-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23134607

ABSTRACT

Recent advancements in the periodontal research field are consistent with a new model of pathogenesis according to which periodontitis is initiated by a synergistic and dysbiotic microbial community rather than by select 'periopathogens', such as the 'red complex'. In this polymicrobial synergy, different members or specific gene combinations within the community fulfill distinct roles that converge to shape and stabilize a disease-provoking microbiota. One of the core requirements for a potentially pathogenic community to arise involves the capacity of certain species, termed 'keystone pathogens', to modulate the host response in ways that impair immune surveillance and tip the balance from homeostasis to dysbiosis. Keystone pathogens also elevate the virulence of the entire microbial community through interactive communication with accessory pathogens. Other important core functions for pathogenicity require the expression of diverse molecules (e.g. appropriate adhesins, cognate receptors, proteolytic enzymes and proinflammatory surface structures/ligands), which in combination act as community virulence factors to nutritionally sustain a heterotypic, compatible and proinflammatory microbial community that elicits a non-resolving and tissue-destructive host response. On the basis of the fundamental concepts underlying this model of periodontal pathogenesis, that is, polymicrobial synergy and dysbiosis, we term it the PSD model.


Subject(s)
Microbial Consortia/physiology , Periodontitis/microbiology , Symbiosis/physiology , Bacteria/immunology , Bacteria/pathogenicity , Host-Pathogen Interactions/physiology , Humans , Immune Evasion/immunology , Immune Evasion/physiology , Inflammation Mediators/immunology , Periodontitis/immunology , Virulence/physiology
19.
Mol Oral Microbiol ; 26(6): 365-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22053964

ABSTRACT

An association between the gram-positive anaerobe Filifactor alocis and periodontal disease has recently emerged; however, possible pathogenic mechanisms have not been investigated. In this study we examined the responses of primary cultures of gingival epithelial cells (GECs) to infection with F. alocis. Secretion of the pro-inflammatory cytokines interleukin-1ß, interleukin-6 and tumor necrosis factor-α from GECs was stimulated by F. alocis infection. F. alocis also induced apoptosis in GECs through pathways that involved caspase-3 but not caspase-9. Apoptosis was coincident with inhibition of mitogen-activated protein kinase kinase (MEK) activation. These results show that F. alocis has characteristics in common with established periodontal pathogens and has the potential to contribute to periodontal tissue destruction.


Subject(s)
Fusobacterium/pathogenicity , Gingiva/microbiology , Apoptosis/immunology , Blotting, Western , Caspase 3/analysis , Caspase 9/analysis , Cells, Cultured , Coculture Techniques , Epithelial Cells/immunology , Epithelial Cells/microbiology , Flow Cytometry , Fusobacterium/immunology , Fusobacterium Infections/immunology , Gingiva/immunology , Humans , Inflammation Mediators/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , MAP Kinase Signaling System/immunology , Microscopy, Confocal , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/analysis
20.
Mol Oral Microbiol ; 26(5): 303-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21896157

ABSTRACT

Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia are consistently associated with adult periodontitis. This study sought to document the host transcriptome to a P. gingivalis, T. denticola, and T.forsythia challenge as a polymicrobial infection using a murine calvarial model of acute inflammation and bone resorption. Mice were infected with P. gingivalis, T. denticola, and T. forsythia over the calvaria, after which the soft tissues and calvarial bones were excised. A Murine GeneChip(®) array analysis of transcript profiles showed that 6997 genes were differentially expressed in calvarial bones (P < 0.05) and 1544 genes were differentially transcribed in the inflamed tissues after the polymicrobial infection. Of these genes, 4476 and 1035 genes in the infected bone and tissues were differentially expressed by upregulation. Biological pathways significantly impacted by the polymicrobial infection in calvarial bone included leukocyte transendothelial migration (LTM), cell adhesion molecules, adherens junction, major histocompatibility complex antigen, extracellular matrix-receptor interaction, and antigen processing and presentation resulting in inflammatory/cytokine/chemokine transcripts stimulation in bone and soft tissue. Intense inflammation and increased activated osteoclasts were observed in calvarias compared with sham-infected controls. Quantitative real-time RT-PCR analysis confirmed that the mRNA level of selected genes corresponded with the microarray expression. The polymicrobial infection regulated several LTM and extracellular membrane pathway genes in a manner distinct from mono-infection with P. gingivalis, T. denticola, or T. forsythia. To our knowledge, this is the first definition of the polymicrobially induced transcriptome in calvarial bone and soft tissue in response to periodontal pathogens.


Subject(s)
Alveolar Bone Loss/microbiology , Bacteroides/genetics , Chronic Periodontitis/microbiology , Coinfection/microbiology , Mouth Mucosa/microbiology , Porphyromonas gingivalis/genetics , Transcriptome/genetics , Treponema denticola/genetics , Adherens Junctions , Alveolar Bone Loss/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Physiological Phenomena/genetics , Chemotaxis, Leukocyte/genetics , Collagen Type III/genetics , Extracellular Matrix Proteins/genetics , Female , Gene Expression Profiling , Host-Pathogen Interactions/genetics , Mice , Mice, Inbred BALB C , Microarray Analysis , Real-Time Polymerase Chain Reaction , Skull/microbiology , Transendothelial and Transepithelial Migration/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...